Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Molecules ; 29(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38731597

ABSTRACT

Fibrosis is a ubiquitous pathology, and prior studies have indicated that various artemisinin (ART) derivatives (including artesunate (AS), artemether (AM), and dihydroartemisinin (DHA)) can reduce fibrosis in vitro and in vivo. The medicinal plant Artemisia annua L. is the natural source of ART and is widely used, especially in underdeveloped countries, to treat a variety of diseases including malaria. A. afra contains no ART but is also antimalarial. Using human dermal fibroblasts (CRL-2097), we compared the effects of A. annua and A. afra tea infusions, ART, AS, AM, DHA, and a liver metabolite of ART, deoxyART (dART), on fibroblast viability and expression of key fibrotic marker genes after 1 and 4 days of treatment. AS, DHA, and Artemisia teas reduced fibroblast viability 4 d post-treatment in up to 80% of their respective controls. After 4 d of treatment, AS DHA and Artemisia teas downregulated ACTA2 up to 10 fold while ART had no significant effect, and AM increased viability by 10%. MMP1 and MMP3 were upregulated by AS, 17.5 and 32.6 fold, respectively, and by DHA, 8 and 51.8 fold, respectively. ART had no effect, but A. annua and A. afra teas increased MMP3 5 and 16-fold, respectively. Although A. afra tea increased COL3A1 5 fold, MMP1 decreased >7 fold with no change in either transcript by A. annua tea. Although A. annua contains ART, it had a significantly greater anti-fibrotic effect than ART alone but was less effective than A. afra. Immunofluorescent staining for smooth-muscle α-actin (α-SMA) correlated well with the transcriptional responses of drug-treated fibroblasts. Together, proliferation, qPCR, and immunofluorescence results show that treatment with ART, AS, DHA, and the two Artemisia teas yield differing responses, including those related to fibrosis, in human dermal fibroblasts, with evidence also of remodeling of fibrotic ECM.


Subject(s)
Artemisia , Artemisinins , Fibroblasts , Fibrosis , Humans , Artemisinins/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Artemisia/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cell Survival/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 3/genetics , Actins/metabolism , Actins/genetics , Artesunate/pharmacology , Gene Expression Regulation/drug effects , Artemether/pharmacology , Skin/drug effects , Skin/metabolism , Skin/pathology
2.
Cell Biol Int ; 43(11): 1317-1322, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31441159

ABSTRACT

The anti-malaria drug artesunate and other chemical analogs of artemisinin have demonstrated cytostatic and cytotoxic effects in bacterial and cancer cells. Artemisinin-derived compounds have also been demonstrated to attenuate fibrosis in preclinical animal models, but the mechanisms by which this inhibition occurs are not well-understood. We investigated the effects of artesunate on the emergence of the myofibroblast, which is causally implicated in pro-fibrotic pathologies. CRL-2097 human dermal fibroblasts were analyzed for protein and transcript expression after treatment with artesunate to analyze fibroblast activation. Proliferation and apoptosis were also evaluated following treatment with artesunate in this cell line. Treatment of human dermal fibroblasts with artesunate antagonized fibroblast activation and pro-fibrotic extracellular matrix (ECM) deposition, both at basal culture conditions and when cultured in the presence of exogenous transforming growth factor-ß1 (TGF-ß1), a major pro-fibrotic cytokine. Artesunate-treated fibroblasts also demonstrated decreased proliferation and increased apoptosis. Transcript analysis by quantitative real-time polymerase chain reaction demonstrated that artesunate downregulated expression of pro-fibrotic genes including canonical myofibroblast markers, ECM genes, and several TGF-ß receptors and ligands, and upregulated expression of cell cycle inhibitors and matrix-metalloproteinases. Together, these data demonstrate that artesunate antagonizes fibroblast activation and decreases expression of pro-fibrotic genes, while also promoting myofibroblast apoptosis, suggesting that these mechanisms may be responsible in part for the anti-fibrotic effects of artesunate described previously.


Subject(s)
Artesunate/pharmacology , Myofibroblasts/metabolism , Skin/pathology , Transforming Growth Factor beta1/metabolism , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Extracellular Matrix/metabolism , Fibrosis , Humans , Myofibroblasts/cytology
3.
Cell Mol Life Sci ; 75(20): 3663-3681, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30027295

ABSTRACT

Fibrosis is a pathological form of aberrant tissue repair, the complications of which account for nearly half of all deaths in the industrialized world. All tissues are susceptible to fibrosis under particular pathological sets of conditions. Though each type of fibrosis has characteristics and hallmarks specific to that particular condition, there appear to be common factors underlying fibrotic diseases. One of these ubiquitous factors is the paradigm of the activated myofibroblast in the promotion of fibrotic phenotypes. Recent research has implicated metabolic byproducts of the amino acid tryptophan, namely serotonin and kynurenines, in the pathology or potential pharmacologic therapy of fibrosis, in part through their effects on development of myofibroblast phenotypes. Here, we review literature underlying what is known mechanistically about the effects of these compounds and their respective pathways on fibrosis. Pharmacologic administration of kynurenine improves scarring outcomes in vivo likely not only through its well-characterized immunosuppressive properties but also via its demonstrated antagonism of fibroblast activation and of collagen deposition. In contrast, serotonin directly promotes activation of fibroblasts via activation of canonical TGF-ß signaling, and overstimulation with serotonin leads to fibrotic outcomes in vivo. Recently discovered feedback inhibition between serotonin and kynurenine pathways also reveals more information about the cellular physiology of tryptophan metabolism and may also underlie possible paradigms for anti-fibrotic therapy. Together, understanding of the effects of tryptophan metabolism on modulation of fibrosis may lead to the development of new therapeutic avenues for treatment through exploitation of these effects.


Subject(s)
Fibroblasts/metabolism , Kynurenine/metabolism , Serotonin/metabolism , Tryptophan/metabolism , Cell Differentiation , Fibroblasts/cytology , Fibrosis , Humans , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
4.
Bioessays ; 38 Suppl 1: S33-44, 2016 07.
Article in English | MEDLINE | ID: mdl-27417120

ABSTRACT

The study of cellular senescence and proliferative lifespan is becoming increasingly important because of the promises of autologous cell therapy, the need for model systems for tissue disease and the implication of senescent cell phenotypes in organismal disease states such as sarcopenia, diabetes and various cancers, among others. Here, we explain the concepts of proliferative cellular lifespan and cellular senescence, and we present factors that have been shown to mediate cellular lifespan positively or negatively. We review much recent literature and present potential molecular mechanisms by which lifespan mediation occurs, drawing from the fields of telomere biology, metabolism, NAD(+) and sirtuin biology, growth factor signaling and oxygen and antioxidants. We conclude that cellular lifespan and senescence are complex concepts that are governed by multiple independent and interdependent pathways, and that greater understanding of these pathways, their interactions and their convergence upon specific cellular phenotypes may lead to viable therapies for tissue regeneration and treatment of age-related pathologies, which are caused by or exacerbated by senescent cells in vivo.


Subject(s)
Cell Proliferation , Cellular Senescence , Animals , Antioxidants , Eukaryota/metabolism , Eukaryota/physiology , Humans , Signal Transduction , Telomere
5.
J Cell Biochem ; 117(9): 2056-66, 2016 09.
Article in English | MEDLINE | ID: mdl-26851891

ABSTRACT

Identification of molecular mechanisms that regulate cellular replicative lifespan is needed to better understand the transition between a normal and a neoplastic cell phenotype. We have previously reported that low oxygen-mediated activity of FGF2 leads to an increase in cellular lifespan and acquisition of regeneration competence in human dermal fibroblasts (iRC cells). Though cells display a more plastic developmental phenotype, they remain non-tumorigenic when injected into SCID mice (Page et al. [2009] Cloning Stem Cells 11:417-426; Page et al. [2011] Eng Part A 17:2629-2640) allowing for investigation of mechanisms that regulate increased cellular lifespan in a non-tumorigenic system. Analysis of chromatin modification enzymes by qRT-PCR revealed a 13.3-fold upregulation of the arginine methyltransferase PRMT8 in iRC cells. Increased protein expression was confirmed in both iRC and human embryonic stem cells-the first demonstration of endogenous human PRMT8 expression outside the brain. Furthermore, iRC cells express a novel PRMT8 mRNA variant. Using siRNA-mediated knockdown we demonstrated that this novel variant was required for proliferation of human dermal fibroblasts (hDFs) and grade IV glioblastomas. PRMT8 upregulation in a non-tumorigenic system may offer a potential diagnostic biomarker and a therapeutic target for cells in pre-cancerous and cancerous states. J. Cell. Biochem. 117: 2056-2066, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Proliferation/physiology , Dermis/enzymology , Fibroblasts/enzymology , Gene Expression Regulation, Developmental/physiology , Membrane Proteins , Protein-Arginine N-Methyltransferases , Up-Regulation/physiology , Animals , Cell Line , Fibroblasts/transplantation , Heterografts , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, SCID , Protein-Arginine N-Methyltransferases/biosynthesis , Protein-Arginine N-Methyltransferases/genetics
6.
J Cell Biochem ; 117(4): 1000-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26403263

ABSTRACT

Deposition of collagen-based extracellular matrix by fibroblasts during wound healing leads to scar formation--a typical outcome of the healing process in soft tissue wounds. The process can, however, be skewed in favor of tissue regeneration by manipulation of wound environment. Low oxygen conditions and supplementation with FGF2 provide extracellular cues that drive wound fibroblasts towards a pro-regenerative phenotype. Under these conditions, fibroblasts dramatically alter expression of many genes among which the most significantly deregulated are extracellular matrix and adhesion molecules. Here we investigate the mechanism of a collagen I binding integrin α11 (ITGA11) deregulation in response to low oxygen-mediated FGF2 effects in dermal fibroblasts. Using RT-PCR, qRT-PCR, Western blotting, and immunocytochemistry, we describe significant down-regulation of ITGA11. Decrease in ITGA11 is associated with its loss from focal adhesions. We show that loss of ITGA11 requires FGF2 induced ERK1/2 activity and in the presence of FGF2, ITGA11 expression cannot be rescued by TGFß1, a potent activator of ITGA11. Our results indicate that FGF2 may be redirecting fibroblasts towards an anti-fibrotic phenotype by overriding TGFß1 mediated ITGA11 expression.


Subject(s)
Cicatrix/prevention & control , Fibroblast Growth Factors/pharmacology , Fibroblasts/drug effects , Integrin alpha Chains/genetics , Re-Epithelialization/drug effects , Transforming Growth Factor beta1/genetics , Cell Hypoxia , Cicatrix/genetics , Cicatrix/metabolism , Cicatrix/pathology , DNA Methylation/drug effects , Dermis/drug effects , Dermis/injuries , Dermis/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Focal Adhesions/drug effects , Gene Expression Regulation , Humans , Integrin alpha Chains/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Oxygen/pharmacology , Primary Cell Culture , Re-Epithelialization/genetics , Signal Transduction , Transforming Growth Factor beta1/metabolism
7.
BMC Genomics ; 14: 656, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24066673

ABSTRACT

BACKGROUND: Adult human fibroblasts grown in low oxygen and with FGF2 supplementation have the capacity to tip the healing outcome of skeletal muscle injury - by favoring regeneration response in vivo over scar formation. Here, we compare the transcriptomes of control adult human dermal fibroblasts and induced regeneration-competent (iRC) fibroblasts to identify transcriptional changes that may be related to their regeneration competence. RESULTS: We identified a unique gene-expression profile that characterizes FGF2-induced iRC fibroblast phenotype. Significantly differentially expressed genes due to FGF2 treatment were identified and analyzed to determine overrepresented Gene Ontology terms. Genes belonging to extracellular matrix components, adhesion molecules, matrix remodelling, cytoskeleton, and cytokines were determined to be affected by FGF2 treatment. CONCLUSIONS: Transcriptome analysis comparing control adult human fibroblasts with FGF2-treated fibroblasts identified functional groups of genes that reflect transcriptional changes potentially contributing to their regeneration competence. This comparative transcriptome analysis should contribute new insights into genes that characterize cells with greater regenerative potential.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Regeneration/genetics , Transcriptome/genetics , Adult , Cell Adhesion Molecules/metabolism , Cell Proliferation/drug effects , Cytokines/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Gene Ontology , Humans , Receptors, Cytokine/metabolism , Regeneration/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/drug effects , Wound Healing/drug effects , Wound Healing/genetics
8.
Histochem Cell Biol ; 139(1): 195-204, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23064788

ABSTRACT

Immunocytochemistry is a powerful tool for detection and visualization of specific molecules in living or fixed cells, their localization and their relative abundance. One of the most commonly used fluorescent DNA dyes in immunocytochemistry applications is 4',6-diamidino-2-phenylindole dihydrochloride, known as DAPI. DAPI binds strongly to DNA and is used extensively for visualizing cell nuclei. It is excited by UV light and emits characteristic blue fluorescence. Here, we report a phenomenon based on an apparent photoconversion of DAPI that results in detection of a DAPI signal using a standard filter set for detection of green emission due to blue excitation. When a sample stained with DAPI only was first imaged with the green filter set (FITC/GFP), only a weak cytoplasmic autofluorescence was observed. Next, we imaged the sample with a DAPI filter set, obtaining a strong nuclear DAPI signal as expected. Upon reimaging the same samples with a FITC/GFP filter set, robust nuclear fluorescence was observed. We conclude that excitation with UV results in a photoconversion of DAPI that leads to detection of DAPI due to excitation and emission in the FITC/GFP channel. This phenomenon can affect data interpretation and lead to false-positive results when used together with fluorochrome-labeled nuclear proteins detected with blue excitation and green emission. In order to avoid misinterpretations, extra precaution should be taken to prepare staining solutions with low DAPI concentration and DAPI (UV excitation) images should be acquired after all other higher wavelength images. Of various DNA dyes tested, Hoechst 33342 exhibited the lowest photoconversion while that for DAPI and Hoechst 33258 was much stronger. Different fixation methods did not substantially affect the strength of photoconversion. We also suggest avoiding the use of mounting medium with high glycerol concentrations since glycerol showed the strongest impact on photoconversion. This photoconversion effect cannot be avoided even when using narrow bandpass filter sets.


Subject(s)
Fluorescent Dyes/radiation effects , Immunohistochemistry/methods , Indoles/radiation effects , Photochemical Processes , Ultraviolet Rays , Benzimidazoles/chemistry , Benzimidazoles/radiation effects , Bisbenzimidazole/chemistry , Bisbenzimidazole/radiation effects , Carcinoma, Embryonal/metabolism , Cell Line, Tumor , False Positive Reactions , Fixatives/chemistry , Fluorescent Dyes/chemistry , Glycerol/chemistry , Humans , Indoles/chemistry , Male , Microscopy, Fluorescence , Reproducibility of Results , Testicular Neoplasms/pathology
9.
ACS Biomater Sci Eng ; 9(5): 2292-2300, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37126371

ABSTRACT

The edible nature of many plants makes leaves particularly useful as scaffolds for the development of cultured meat, where animal tissue is grown in the laboratory setting. Recently, we demonstrated that decellularized spinach leaves can serve as scaffolds to grow and differentiate cells for cultured meat products. However, conventional decellularization methods use solutions that are not considered safe for use in food, such as organic solvents (hexanes) and detergents (triton X-100 (TX100)). This study modified decellularization protocols to incorporate detergents that are regulated (REG) by the United States Food and Drug Administration (FDA) for use in food, such as Polysorbate 20 (PS20), and eliminates the use of hexanes for cuticle removal. Spinach leaves were decellularized with sodium dodecyl sulfate and then with either TX100 (control) or PS20. The average DNA content for TX100 samples and PS20 samples was similar (1.3 ± 0.07 vs 1.3 ± 0.05 ng/mg; TX100 vs PS20, p = ns). The importance of cuticle removal was tested by removing hexanes from the protocol. Groups that included the cuticle removal step exhibited an average reduction in DNA content of approximately 91.7%, and groups that omitted the cuticle removal step exhibited an average reduction of approximately 90.3% (p = ns), suggesting that the omission of the cuticle removal step did not impede decellularization. Lastly, primary bovine satellite cells (PBSCs) were cultured for 7 days (d) on the surface of spinach leaves decellularized using the REG protocol. After the 7 d incubation period, PBSCs grown on the surface of REG scaffolds had an average viability of approximately 97.4%. These observations suggest that the REG protocol described in this study is an effective decellularization method, more closely adhering to food safety guidelines, that could be implemented in lab grown meat and alternative protein products.


Subject(s)
Tissue Engineering , Tissue Scaffolds , Animals , Cattle , Tissue Engineering/methods , Detergents/pharmacology , Hexanes/pharmacology , Extracellular Matrix , Octoxynol/pharmacology , DNA/pharmacology
10.
Genesis ; 50(3): 286-99, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22223599

ABSTRACT

Using a combination of deep sequencing and bioinformatics approach, we for the first time identify miRNAs and their relative abundance in mature, metaphase II arrested eggs in Xenopus laevis. We characterize 115 miRNAs that have been described either in Xenopus tropicalis (85), X. laevis (9), or other vertebrate species (21) that also map to known Xenopus pre-miRNAs and to the X. tropicalis genome. In addition, 72 new X. laevis putative candidate miRNAs are identified based on mapping to X. tropicalis genome within regions that have the propensity to form hairpin loops. These data expand on the availability of genetic information in X. laevis and identify target miRNAs for future functional studies.


Subject(s)
MicroRNAs/genetics , Ovum/metabolism , Animals , Computational Biology/methods , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Metaphase , Xenopus , Xenopus laevis
11.
Acta Pharm Sin B ; 11(2): 322-339, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33643815

ABSTRACT

Fibrosis is a pathological reparative process that can occur in most organs and is responsible for nearly half of deaths in the developed world. Despite considerable research, few therapies have proven effective and been approved clinically for treatment of fibrosis. Artemisinin compounds are best known as antimalarial therapeutics, but they also demonstrate antiparasitic, antibacterial, anticancer, and anti-fibrotic effects. Here we summarize literature describing anti-fibrotic effects of artemisinin compounds in in vivo and in vitro models of tissue fibrosis, and we describe the likely mechanisms by which artemisinin compounds appear to inhibit cellular and tissue processes that lead to fibrosis. To consider alternative routes of administration of artemisinin for treatment of internal organ fibrosis, we also discuss the potential for more direct oral delivery of Artemisia plant material to enhance bioavailability and efficacy of artemisinin compared to administration of purified artemisinin drugs at comparable doses. It is our hope that greater understanding of the broad anti-fibrotic effects of artemisinin drugs will enable and promote their use as therapeutics for treatment of fibrotic diseases.

12.
Int J Mol Med ; 43(1): 325-335, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30365043

ABSTRACT

Fibroblast activation is a key step in the establishment of skin fibrosis induced by acute injury, and it is characterized by the differentiation of plastic resident tissue fibroblasts into contractile, extracellular matrix­secreting myofibroblasts. As fibroblast activation must be regulated in vivo, fibroblasts receive signals from the surrounding environment that initiate their fibrotic program. Thus, the present study investigated the effects of mitogen­activated protein kinase (MAPK) signaling pathways on fibroblast activation. It was demonstrated in primary human dermal fibroblasts that small molecule­mediated inhibition of extracellular signal­regulated kinase (ERK) and c­Jun N­terminal kinase (JNK) potentiated fibroblast activation, and that small molecule­mediated inhibition of p38 antagonized fibroblast activation. ERK and JNK inhibition cooperatively enhanced fibroblast activation mediated by treatment with exogenous transforming growth factor (TGF)­ß1, and p38 inhibition antagonized ERK inhibitor­mediated or JNK inhibitor­mediated fibroblast activation. Transcript analysis demonstrated that ERK and JNK inhibitor­mediated fibroblast activation was accompanied by distinct changes in the expression of TGF­ß­associated ligands and receptors, and that p38 inhibitor­mediated antagonism of fibroblast activation was accompanied by a distinct expression paradigm of TGF­ß­associated genes, including upregulation of betaglycan. ERK inhibitor­mediated and JNK inhibitor­mediated fibroblast activation was partially antagonized by small molecule­mediated inhibition of TGF­ß receptor (R)1, indicating that these mechanisms of fibroblast activation are partially dependent on TGF­ß/TGF­ßR signaling. These data collectively demonstrate and provide partial explanations of the varied effects and pathway dependencies of MAPK inhibitor­mediated effects on fibroblast activation.


Subject(s)
Dermis/pathology , Fibroblasts/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Transforming Growth Factor beta/metabolism , Cells, Cultured , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Fibrosis , Gene Expression Regulation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Ligands , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transcription, Genetic/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Tissue Eng Regen Med ; 12(3): 572-582, 2018 03.
Article in English | MEDLINE | ID: mdl-28513058

ABSTRACT

Smooth muscle cells (SMCs) are essential for tissue engineering strategies to fabricate organs such as blood vessels, the oesophagus and bladder, and to create disease models of these systems. In order for such therapies and models to be feasible, SMCs must be sourced effectively to enable production of large numbers of functional cells. In vitro, SMCs divide slowly and demonstrate short proliferative lifespans compared with other types of cells, including stem cells and fibroblasts, limiting the number of cells that can be derived from expansion in culture of a primary isolation. As such, it would be beneficial to better understand the factors underlying induction and maintenance of SMC phenotypes, in order to produce new sources of SMCs for tissue engineering and disease modelling. Here we report the ability of human dermal fibroblasts to display patterns of gene expression resembling contractile SMCs when cultured under conditions that are known to promote a contractile phenotype in SMCs, including culture on collagen IV, low-serum culture, TGF-ß1 treatment and hypoxia. These factors drive expression of the myogenic transcription factor myocardin, as well as expression of several of its gene targets that are known contributors to contractile phenotype in SMCs, including smooth muscle alpha actin, calponin, and myosin heavy chain. Our results suggest that culture conditions associated with culture of SMCs may be sufficient to induce myogenic gene expression patterns and potential myogenic function in non-muscle cells.


Subject(s)
Biomarkers/metabolism , Fibroblasts/metabolism , Muscle Contraction/drug effects , Muscle Development/drug effects , Myocytes, Smooth Muscle/metabolism , Oxygen/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Hypoxia/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dermis/cytology , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Humans , Male , Muscle Development/genetics , Myocytes, Smooth Muscle/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic/drug effects , Young Adult
14.
J Dermatol Sci ; 88(3): 339-348, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28899582

ABSTRACT

BACKGROUND: Previous human and animal studies have demonstrated the ability of exogenously administered basic fibroblast growth factor (FGF2) to act as an antifibrotic agent in the skin. Though the activity of FGF2 as an anti-scarring agent is well-established for fibrotic skin wounds, the mechanisms by which FGF2 exerts these actions are not entirely understood. Canonical FGF2 signaling proceeds in part via FGFR/MAPK pathways in human dermal fibroblasts, and FGF2 has been described to prevent or reverse the fibroblast-to-myofibroblast transition, which is driven by TGFß signaling and understood to be an important step in the formation of a fibrotic scar in vivo. Thus, we set out to investigate the antagonistic effects of FGF2 on TGFß signaling as well as the broader effects of MAPK inhibition on the TGFß-mediated induction of myofibroblast gene expression. OBJECTIVE: To better understand the effects of FGF2 signaling pathways on myofibroblastic gene expression and cell phenotypes. METHODS: Human dermal fibroblasts were cultured in vitro in the presence of FGF2, TGFß, and/or MAPK inhibitors, and the effects of these agents were investigated by molecular biology techniques including qRT-PCR, immunofluorescence, Western blot, and flow cytometry. RESULTS: FGF2 inhibited TGFß-mediated fibroblast activation, resulting in more rapidly proliferating, spindle-shaped cells, compared to the more slowly proliferating, flatter TGFß-treated cells. Treatment with FGF2 also attenuated TGFß-mediated increase in expression of myofibroblast markers smooth muscle α-actin, calponin, transgelin, connective tissue growth factor, ED-A fibronectin, and collagen I. FGF2-mediated antagonism of the TGFß-mediated fibroblast-to-myofibroblast transition was reversed by small molecule inhibition of ERK or JNK, and it was potentiated by inhibition of p38. MAPK inhibition was demonstrated to have qualitatively similar effects even in the absence of exogenous FGF2, and small molecule inhibition of p38 MAPK was sufficient to attenuate TGFß-mediated fibroblast activation. CONCLUSIONS: Inhibition of select MAPK signaling pathways can reverse or potentiate anti-fibrotic FGF2 effects on human dermal fibroblasts, as well as exert their effects independently of exogenous FGF2 supplementation.


Subject(s)
Cicatrix/drug therapy , Fibroblast Growth Factor 2/pharmacology , MAP Kinase Signaling System/drug effects , Myofibroblasts/drug effects , Skin/metabolism , Biomarkers/metabolism , Cell Line , Cell Proliferation/drug effects , Cicatrix/pathology , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/therapeutic use , Humans , MAP Kinase Signaling System/physiology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Protein Kinase Inhibitors/pharmacology , Receptors, Transforming Growth Factor beta , Skin/cytology , Skin/pathology , Transforming Growth Factor beta/metabolism , Wound Healing/physiology
15.
Cytokine Growth Factor Rev ; 38: 49-58, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28967471

ABSTRACT

Fibrosis is a pathological condition that is characterized by the replacement of dead or damaged tissue with a nonfunctional, mechanically aberrant scar, and fibrotic pathologies account for nearly half of all deaths worldwide. The causes of fibrosis differ somewhat from tissue to tissue and pathology to pathology, but in general some of the cellular and molecular mechanisms remain constant regardless of the specific pathology in question. One of the common mechanisms underlying fibroses is the paradigm of the activated fibroblast, termed the "myofibroblast," a differentiated mesenchymal cell with demonstrated contractile activity and a high rate of collagen deposition. Fibroblast growth factor 2 (FGF2), one of the members of the mammalian fibroblast growth factor family, is a cytokine with demonstrated antifibrotic activity in non-human animal, human, and in vitro models. FGF2 is highly pleiotropic and its receptors are present on many different cell types throughout the body, lending a great deal of variety to the potential mechanisms of FGF2 effects on fibrosis. However, recent reports demonstrate that a substantial contribution to the antifibrotic effects of FGF2 comes from the inhibitory effects of FGF2 on connective tissue fibroblasts, activated myofibroblasts, and myofibroblast progenitors. FGF2 demonstrates effects antagonistic towards fibroblast activation and towards mesenchymal transition of potential myofibroblast-forming cells, as well as promotes a gene expression paradigm more reminiscent of regenerative healing, such as that which occurs in the fetal wound healing response, than fibrotic resolution. With a better understanding of the mechanisms by which FGF2 alters the wound healing cascade and results in a shift away from scar formation and towards functional tissue regeneration, we may be able to further address the critical need of therapy for varied fibrotic pathologies across myriad tissue types.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Fibrosis/metabolism , Myofibroblasts/metabolism , Animals , Cell Differentiation , Fibrosis/genetics , Gene Expression , Humans , Myofibroblasts/cytology , Phenotype , Stem Cells/cytology
16.
Oncol Lett ; 13(3): 1983-1989, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28454353

ABSTRACT

Recent emphasis has been placed on the role of epigenetic regulators and epigenetic marks as biomarkers for cancer diagnosis and prognosis, and as therapeutic targets for treatment. One such class of regulators is the protein arginine methyltransferase (PRMT) family. The present study examined available curated data regarding the expression and alteration of one of the least studied PRMT family members, PRMT8, in various types of cancer and cancer cell lines. Publicly available cancer data on PRMT8 expression were examined using the Human Protein Atlas and the Kaplan-Meier Plotter, and reverse transcription-polymerase chain reaction was used to screen a selection of human cell lines for variant-specific PRMT8 expression. High levels of PRMT8 expression in breast, ovarian and cervical cancer was observed. Additionally, in patients with breast and ovarian cancer, high PRMT8 expression was correlated with increased patient survival, whereas in gastric cancer, high PRMT8 expression was correlated with decreased patient survival. The present study also investigated the expression of PRMT8 variant 2, a novel transcript variant recently identified in our laboratory, in various cancer cell lines. Variant-specific expression of PRMT8 in numerous distinct cancer cell lines derived from different tissues, including the expression of the novel PRMT8 variant 2 in U87MG glioblastoma cells was demonstrated. The present study proposes the possibility of PRMT8 as a cancer biomarker, based on the high level of PRMT8 expression in various types of cancer, particularly in tissues that would not normally be expected to express PRMT8, and on the correlation of PRMT8 and patient lifespan in several cancer types. Variant-specific expression of PRMT8 in diverse cancer cell lines suggests the possibility of alternate PRMT8 isoforms to have diverse effects on cancer cell phenotypes.

17.
Stem Cell Res ; 21: 106-116, 2017 05.
Article in English | MEDLINE | ID: mdl-28433654

ABSTRACT

Basic fibroblast growth factor (FGF2) is a highly pleiotropic member of a large family of growth factors with a broad range of activities, including mitogenesis and angiogenesis (Ornitz et al., 1996; Zhang et al., 2006), and it is known to be essential for maintenance of balance between survival, proliferation, and self-renewal in human pluripotent stem cells (Eiselleova et al., 2009; Zoumaro-Djayoon et al., 2011). A single FGF2 transcript can be translated into five FGF2 protein isoforms, an 18kDa low molecular weight (LMW) isoform and four larger high molecular weight (HMW) isoforms (Arese et al., 1999; Arnaud et al., 1999). As they are not generally secreted, high molecular weight (HMW) FGF2 isoforms have predominantly been investigated intracellularly; only a very limited number of studies have investigated their activity as extracellular factors. Here we report over-expression, isolation, and biological activity of all recombinant human FGF2 isoforms. We show that HMW FGF2 isoforms can support self-renewal of human embryonic stem cells (hESCs) in vitro. Exogenous supplementation with HMW FGF2 isoforms also activates the canonical FGFR/MAPK pathway and induces mitogenic activity in a manner similar to that of the 18kDa FGF2 isoform. Though all HMW isoforms, when supplemented exogenously, are able to recapitulate LMW FGF2 activity to some degree, it appears that certain isoforms tend to do so more poorly, demonstrating a lesser functional response by several measures. A better understanding of isoform-specific FGF2 effects will lead to a better understanding of developmental and pathological FGF2 signaling.


Subject(s)
Cell Self Renewal , Fibroblast Growth Factor 2/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , Base Sequence , Cell Line , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Cell Shape/drug effects , Dermis/cytology , Fibroblast Growth Factor 2/isolation & purification , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Mice , Mitogens/pharmacology , Molecular Weight , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism
18.
Acta Biomater ; 52: 9-20, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28017866

ABSTRACT

Wound infections, particularly of chronic wounds, pose a substantial challenge for designing antimicrobial dressings that are both effective against pathogens, and do not interfere with wound healing. Due to their broad-spectrum antimicrobial and immunomodulatory activities, naturally-occurring antimicrobial peptides (AMPs) are promising alternative treatments. However, their cytotoxicity at high concentrations and poor stability hinders their clinical use. To mitigate these undesirable properties, we investigated the effects of tethering human AMP cathelicidin LL37 to collagen, one of the main extracellular matrix proteins in wound sites, secreted by fibroblasts, and in commercially-available wound dressings. The active domain of human AMP cathelicidin, LL37, and two chimeric peptides containing LL37 fused to collagen binding domains (derived from collagenase - cCBD-LL37 or fibronectin - fCBD-LL37) were synthesized and adsorbed to PURACOL® type I collagen scaffolds. After 14days, 73%, 81% and 99% of LL37, cCBD-LL37 and fCBD-LL37, respectively, was retained on the scaffolds and demonstrated undiminished antimicrobial activity when challenged with both Gram-positive and Gram-negative bacterial strains. Loaded scaffolds were not cytotoxic to fibroblasts despite retaining peptides at concentrations 24 times higher than the reported cytotoxic concentrations in solution. These findings indicate that biopolymer-tethered AMPs may represent a viable alternative for preventing and treating wound infection while also supporting tissue repair. STATEMENT OF SIGNIFICANCE: Over 6.5million people annually in the United States suffer chronic wounds; many will become infected with antibiotic-resistant bacteria. Treatments used to prevent and fight infection are toxic and may hinder wound healing. AMPs are broad-spectrum antimicrobials that also promote healing; however, their instability and toxicity are major challenges. To overcome treatment gaps, we functionalized collagen scaffolds with chimeric antimicrobial peptides (AMPs) with collagen binding domains to create antimicrobial and non-cytotoxic scaffolds that may promote healing. This is the first report of CBD-mediated delivery of AMPs onto collagen scaffolds that demonstrates no cytotoxicity toward fibroblasts. This study also suggests that retention of antimicrobial activity is CBD-dependent, which provides foundations for fundamental studies of CBD-AMP mechanisms and clinical explorations.


Subject(s)
Bacterial Physiological Phenomena/drug effects , Bandages/microbiology , Cathelicidins/administration & dosage , Collagen/chemistry , Delayed-Action Preparations/administration & dosage , Fibroblasts/drug effects , Peptide Fragments/administration & dosage , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Cathelicidins/chemistry , Cell Survival/drug effects , Cells, Cultured , Delayed-Action Preparations/chemistry , Equipment Design , Extracellular Matrix Proteins/chemistry , Humans , Peptide Fragments/chemistry
19.
Biomaterials ; 125: 13-22, 2017 05.
Article in English | MEDLINE | ID: mdl-28222326

ABSTRACT

Despite significant advances in the fabrication of bioengineered scaffolds for tissue engineering, delivery of nutrients in complex engineered human tissues remains a challenge. By taking advantage of the similarities in the vascular structure of plant and animal tissues, we developed decellularized plant tissue as a prevascularized scaffold for tissue engineering applications. Perfusion-based decellularization was modified for different plant species, providing different geometries of scaffolding. After decellularization, plant scaffolds remained patent and able to transport microparticles. Plant scaffolds were recellularized with human endothelial cells that colonized the inner surfaces of plant vasculature. Human mesenchymal stem cells and human pluripotent stem cell derived cardiomyocytes adhered to the outer surfaces of plant scaffolds. Cardiomyocytes demonstrated contractile function and calcium handling capabilities over the course of 21 days. These data demonstrate the potential of decellularized plants as scaffolds for tissue engineering, which could ultimately provide a cost-efficient, "green" technology for regenerating large volume vascularized tissue mass.


Subject(s)
Perfusion/methods , Plant Leaves/chemistry , Plant Vascular Bundle/chemistry , Stem Cells/cytology , Stem Cells/physiology , Tissue Engineering/instrumentation , Tissue Scaffolds , Batch Cell Culture Techniques/instrumentation , Cell-Free System/chemistry , Cells, Cultured , Equipment Design , Extracellular Matrix/chemistry , Humans , Petroselinum/chemistry , Spinacia oleracea/chemistry , Tissue Engineering/methods
20.
Cell Reprogram ; 16(1): 18-28, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24405062

ABSTRACT

Direct reprogramming of a differentiated somatic cell into a developmentally more plastic cell would offer an alternative to applications in regenerative medicine that currently depend on either embryonic stem cells (ESCs), adult stem cells, or induced pluripotent stem cells (iPSCs). Here we report the potential of select Xenopus laevis egg extract fractions, in combination with exogenous fibroblast growth factor-2 (FGF2), to affect life span, morphology, gene expression, protein translation, and cellular localization of OCT4 and NANOG transcription factors, and the developmental potential of human dermal fibroblasts in vitro. A gradual change in morphology is accompanied by translation of embryonic transcription factors and their nuclear localization and a life span exceeding 60 population doublings. Cells acquire the ability to follow adipogenic, neuronal, and osteogenic differentiation under appropriate induction conditions in vitro. Analysis of active extract fractions reveals that Xenopus egg protein and RNAs as well as exogenously supplemented FGF2 are required and sufficient for induction and maintenance of this phenotypic change. Factors so far identified in the active fractions include FGF2 itself, transforming growth factor-ß, maskin, and nucleoplasmin. Identification of critical factors needed for reprogramming may allow for nonviral, chemically defined derivation of human-induced multipotent cells that can be maintained by exogenous FGF2.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Complex Mixtures/pharmacology , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/metabolism , Multipotent Stem Cells/metabolism , Oocytes/chemistry , Adult , Animals , Cell Line , Complex Mixtures/chemistry , Female , Fibroblasts/cytology , Homeodomain Proteins/metabolism , Humans , Male , Multipotent Stem Cells/cytology , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL