Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 800
Filter
1.
PLoS One ; 19(10): e0311927, 2024.
Article in English | MEDLINE | ID: mdl-39401205

ABSTRACT

Magnet-mediated gene therapy has gained considerable interest from researchers as a novel alternative for treating genetic disorders, particularly through the use of superparamagnetic iron oxide nanoparticles (NPs)-such as magnetite NPs (Fe3O4NPs)-as non-viral genetic vectors. Despite their commercial availability for specific genetic transfection, such as in microglia cell lines, many potential uses remain unexplored. Still, ethical concerns surrounding the use of human DNA often impede genetic research. Hence, this study examined DNA-coated Fe3O4NPs (DNA-Fe3O4NPs) as potential transfection vectors for human foreskin fibroblasts (HFFs) and A549 (lung cancer) cell lines, using banana (Musa sp.) as a low-cost, and bioethically unproblematic DNA source. Following coprecipitation synthesis, DNA-Fe3O4NP characterization revealed a ζ-potential of 40.65 ± 4.10 mV, indicating good colloidal stability in aqueous media, as well as a superparamagnetic regime, evidenced by the absence of hysteresis in their magnetization curves. Successful DNA coating on the NPs was confirmed through infrared spectra and surface analysis results, while magnetite content was verified via characteristic X-ray diffraction peaks. Transmission electron microscopy (TEM) determined the average size of the DNA-Fe3O4NPs to be 14.69 ± 5.22 nm. TEM micrographs also showed no morphological changes in the DNA-Fe3O4NPs over a 30-day period. Confocal microscopy of HFF and A549 lung cancer cell lines incubated with fluoresceinamine-labeled DNA-Fe3O4NPs demonstrated their internalization into both the cytoplasm and nucleus. Neither uncoated Fe3O4NPs nor DNA-Fe3O4NPs showed cytotoxicity to A549 lung cancer cells at 1-50 µg/mL and 25-100 µg/mL, respectively, after 24 h. HFFs also maintained viability at 1-10 µg/mL for both NP types. In conclusion, DNA-Fe3O4NPs were successfully internalized into cells and exhibited no cytotoxicity in both healthy and cancerous cells across a range of concentrations. These NPs, capable of binding to various types of DNA and RNA, hold promise for applications in gene therapy.


Subject(s)
DNA , Magnetite Nanoparticles , Musa , Humans , Magnetite Nanoparticles/chemistry , Musa/chemistry , A549 Cells , Fruit/chemistry , Fibroblasts/metabolism , Cell Survival/drug effects , Transfection , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Line, Tumor
2.
Oxid Med Cell Longev ; 2024: 2812290, 2024.
Article in English | MEDLINE | ID: mdl-39411276

ABSTRACT

Diabetes mellitus is associated with chronic wound-healing problems that significantly impact patients' quality of life and substantially increase expenditure on healthcare. Therefore, the identification of compounds that can aid healing is justified. Anredera cordifolia (Ten.) has been used in folk medicine for curative purposes; however, the causal mechanisms underlying its healing effects remain to be elucidated. In this study, the effect of the ethanolic extract of A. cordifolia was evaluated in an in vitro healing model using fibroblasts cultivated under normoglycemic and hyperglycemic environments. The extract was predominantly composed of phytol and exhibited genoprotective activity. Fibroblast migration attenuated the adverse effects of hyperglycemia, favoring cell proliferation. Collagen levels were significantly increased in ruptured fibroblasts under both standard and hyperglycemic environments. The phytogenomic effect of the extract on three genes related to extracellular matrix formation, maintenance, and degradation showed that A. cordifolia increased the expression of genes related to matrix synthesis and maintenance in both normoglycemic and hyperglycemic individuals. Furthermore, it reduced the expression of genes related to matrix degradation. Overall, this is the first study to demonstrate the effectiveness of A. cordifolia in wound healing, elucidating possible causal mechanisms that appear to be based on the genoprotective effect of this plant on the migratory and proliferative phases of the wound healing process; these effects are probably related to phytol, its main constituent.


Subject(s)
Cell Movement , Cell Proliferation , Fibroblasts , Hyperglycemia , Plant Extracts , Wound Healing , Cell Proliferation/drug effects , Cell Movement/drug effects , Plant Extracts/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Wound Healing/drug effects , Humans , Hyperglycemia/drug therapy , Ethanol/chemistry , Diabetes Mellitus/drug therapy
3.
Int J Mol Sci ; 25(17)2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39273169

ABSTRACT

Parkinson's disease (PD) is a multifactorial, chronic, and progressive neurodegenerative disorder inducing movement alterations as a result of the loss of dopaminergic (DAergic) neurons of the pars compacta in the substantia nigra and protein aggregates of alpha synuclein (α-Syn). Although its etiopathology agent has not yet been clearly established, environmental and genetic factors have been suggested as the major contributors to the disease. Mutations in the glucosidase beta acid 1 (GBA1) gene, which encodes the lysosomal glucosylceramidase (GCase) enzyme, are one of the major genetic risks for PD. We found that the GBA1 K198E fibroblasts but not WT fibroblasts showed reduced catalytic activity of heterozygous mutant GCase by -70% but its expression levels increased by 3.68-fold; increased the acidification of autophagy vacuoles (e.g., autophagosomes, lysosomes, and autolysosomes) by +1600%; augmented the expression of autophagosome protein Beclin-1 (+133%) and LC3-II (+750%), and lysosomal-autophagosome fusion protein LAMP-2 (+107%); increased the accumulation of lysosomes (+400%); decreased the mitochondrial membrane potential (∆Ψm) by -19% but the expression of Parkin protein remained unperturbed; increased the oxidized DJ-1Cys106-SOH by +900%, as evidence of oxidative stress; increased phosphorylated LRRK2 at Ser935 (+1050%) along with phosphorylated α-synuclein (α-Syn) at pathological residue Ser129 (+1200%); increased the executer apoptotic protein caspase 3 (cleaved caspase 3) by +733%. Although exposure of WT fibroblasts to environmental neutoxin rotenone (ROT, 1 µM) exacerbated the autophagy-lysosomal system, oxidative stress, and apoptosis markers, ROT moderately increased those markers in GBA1 K198E fibroblasts. We concluded that the K198E mutation endogenously primes skin fibroblasts toward autophagy dysfunction, OS, and apoptosis. Our findings suggest that the GBA1 K198E fibroblasts are biochemically and molecularly equivalent to the response of WT GBA1 fibroblasts exposed to ROT.


Subject(s)
Apoptosis , Autophagy , Fibroblasts , Glucosylceramidase , Mitochondria , Oxidative Stress , Glucosylceramidase/metabolism , Glucosylceramidase/genetics , Humans , Fibroblasts/metabolism , Autophagy/genetics , Mitochondria/metabolism , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Skin/metabolism , Skin/pathology , Lysosomes/metabolism , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mutation
4.
Int J Mol Sci ; 25(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273443

ABSTRACT

Vascular smooth muscle cells (SMCs) can transition between a quiescent contractile or "differentiated" phenotype and a "proliferative-dedifferentiated" phenotype in response to environmental cues, similar to what in occurs in the wound healing process observed in fibroblasts. When dysregulated, these processes contribute to the development of various lung and cardiovascular diseases such as Chronic Obstructive Pulmonary Disease (COPD). Long non-coding RNAs (lncRNAs) have emerged as key modulators of SMC differentiation and phenotypic changes. In this study, we examined the expression of lncRNAs in primary human pulmonary artery SMCs (hPASMCs) during cell-to-cell contact-induced SMC differentiation. We discovered a novel lncRNA, which we named Differentiation And Growth Arrest-Related lncRNA (DAGAR) that was significantly upregulated in the quiescent phenotype with respect to proliferative SMCs and in cell-cycle-arrested MRC5 lung fibroblasts. We demonstrated that DAGAR expression is essential for SMC quiescence and its knockdown hinders SMC differentiation. The treatment of quiescent SMCs with the pro-inflammatory cytokine Tumor Necrosis Factor (TNF), a known inducer of SMC dedifferentiation and proliferation, elicited DAGAR downregulation. Consistent with this, we observed diminished DAGAR expression in pulmonary arteries from COPD patients compared to non-smoker controls. Through pulldown experiments followed by mass spectrometry analysis, we identified several proteins that interact with DAGAR that are related to cell differentiation, the cell cycle, cytoskeleton organization, iron metabolism, and the N-6-Methyladenosine (m6A) machinery. In conclusion, our findings highlight DAGAR as a novel lncRNA that plays a crucial role in the regulation of cell proliferation and SMC differentiation. This paper underscores the potential significance of DAGAR in SMC and fibroblast physiology in health and disease.


Subject(s)
Cell Differentiation , Cell Proliferation , Fibroblasts , Myocytes, Smooth Muscle , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Fibroblasts/metabolism , Cell Differentiation/genetics , Myocytes, Smooth Muscle/metabolism , Cell Proliferation/genetics , Pulmonary Artery/metabolism , Pulmonary Artery/cytology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Cells, Cultured
5.
Lasers Med Sci ; 39(1): 225, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39207591

ABSTRACT

BACKGROUND: Low-level Laser Therapy (LLLT) has demonstrated its potential in promoting fiber matrix maturation, collagen synthesis, and fibroblast proliferation, contributing to tissue regeneration. Our study aimed to investigate the impact of LLLT on collagen type I synthesis, cell proliferation, and viability in human ligament fibroblasts derived from the Anterior Cruciate Ligament (ACL). METHODS: Tissue samples were obtained from individuals undergoing arthroscopic ACL reconstruction surgery. Primary human fibroblasts were isolated, and immunohistochemical assays confirmed their characteristics. LLLT at 850 nm was administered in three groups: Low dose (1.0 J/cm²), High dose (5.0 J/cm²), and Control (0.0 J/cm²). Cell viability was calculated using a membrane integrity assay, proliferation was determined by automated counting, and collagen type I concentration in cell culture was measured using an immunoassay. RESULTS: Fibroblasts showed decreased viability after low and high doses of LLLT, increased proliferation at the low dose, and increased collagen synthesis at the high dose on day 10 for both sexes after treatment. CONCLUSION: Our study demonstrated that LLLT may improve the early ligament healing process by increasing cell proliferation at the low dose and enhancing collagen type I synthesis at the high dose in human ligament fibroblasts.


Subject(s)
Anterior Cruciate Ligament , Cell Proliferation , Cell Survival , Collagen Type I , Fibroblasts , Low-Level Light Therapy , Wound Healing , Humans , Fibroblasts/radiation effects , Fibroblasts/metabolism , Low-Level Light Therapy/methods , Collagen Type I/metabolism , Cell Proliferation/radiation effects , Female , Male , Cell Survival/radiation effects , Wound Healing/radiation effects , Anterior Cruciate Ligament/radiation effects , Anterior Cruciate Ligament/surgery , Cells, Cultured , Adult
6.
Matrix Biol ; 133: 103-115, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39153517

ABSTRACT

Skeletal muscle fibrosis is defined as the excessive accumulation of extracellular matrix (ECM) components and is a hallmark of muscular dystrophies. Fibro-adipogenic progenitors (FAPs) are the main source of ECM, and thus have been strongly implicated in fibrogenesis. In skeletal muscle fibrotic models, including muscular dystrophies, FAPs undergo dysregulations in terms of proliferation, differentiation, and apoptosis, however few studies have explored the impact of FAPs migration. Here, we studied fibroblast and FAPs migration and identified lysophosphatidic acid (LPA), a signaling lipid central to skeletal muscle fibrogenesis, as a significant migration inductor. We identified LPA receptor 1 (LPA1) mediated signaling as crucial for this effect through a mechanism dependent on the Hippo pathway, another pathway implicated in fibrosis across diverse tissues. This cross-talk favors the activation of the Yes-associated protein 1 (YAP) and Transcriptional coactivator with PDZ-binding motif (TAZ), leading to increased expression of fibrosis-associated genes. This study reveals the role of YAP in LPA-mediated fibrotic responses as inhibition of YAP transcriptional coactivator activity hinders LPA-induced migration in fibroblasts and FAPs. Moreover, we found that FAPs derived from the mdx4cv mice, a murine model of Duchenne muscular dystrophy, display a heightened migratory phenotype due to enhanced LPA signaling compared to wild-type FAPs. Remarkably, we found that the inhibition of LPA1 or YAP transcriptional coactivator activity in mdx4cv FAPs reverts this phenotype. In summary, the identified LPA-LPA1-YAP pathway emerges as a critical driver of skeletal muscle FAPs migration and provides insights into potential novel targets to mitigate fibrosis in muscular dystrophies.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Movement , Fibroblasts , Fibrosis , Lysophospholipids , Muscle, Skeletal , Receptors, Lysophosphatidic Acid , Signal Transduction , YAP-Signaling Proteins , Lysophospholipids/metabolism , Animals , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Mice , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , Hippo Signaling Pathway , Mice, Inbred mdx , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Adipogenesis/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology
7.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38999961

ABSTRACT

Skin wound healing is coordinated by a delicate balance between proinflammatory and anti-inflammatory responses, which can be affected by opportunistic pathogens and metabolic or vascular diseases. Several antimicrobial peptides (AMPs) possess immunomodulatory properties, suggesting their potential to support skin wound healing. Here, we evaluated the proregenerative activity of three recently described AMPs (Clavanin A, Clavanin-MO, and Mastoparan-MO). Human primary dermal fibroblasts (hFibs) were used to determine peptide toxicity and their capacity to induce cell proliferation and migration. Furthermore, mRNA analysis was used to investigate the modulation of genes associated with skin regeneration. Subsequently, the regenerative potential of the peptides was further confirmed using an ex vivo organotypic model of human skin (hOSEC)-based lesion. Our results indicate that the three molecules evaluated in this study have regenerative potential at nontoxic doses (i.e., 200 µM for Clavanin-A and Clavanin-MO, and 6.25 µM for Mastoparan-MO). At these concentrations, all peptides promoted the proliferation and migration of hFibs during in vitro assays. Such processes were accompanied by gene expression signatures related to skin regenerative processes, including significantly higher KI67, HAS2 and CXCR4 mRNA levels induced by Clavanin A and Mastoparan-MO. Such findings translated into significantly accelerated wound healing promoted by both Clavanin A and Mastoparan-MO in hOSEC-based lesions. Overall, the data demonstrate the proregenerative properties of these peptides using human experimental skin models, with Mastoparan-MO and Clavanin A showing much greater potential for inducing wound healing compared to Clavanin-MO.


Subject(s)
Cell Movement , Cell Proliferation , Fibroblasts , Regeneration , Skin , Wound Healing , Humans , Wound Healing/drug effects , Skin/metabolism , Skin/drug effects , Cell Proliferation/drug effects , Cell Movement/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Regeneration/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , Antimicrobial Peptides/pharmacology , Cells, Cultured , Peptides/pharmacology
8.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000270

ABSTRACT

The combination of a polyphenol, quercetin, with dasatinib initiated clinical trials to evaluate the safety and efficacy of senolytics in idiopathic pulmonary fibrosis, a lung disease associated with the presence of senescent cells. Another approach to senotherapeutics consists of controlling inflammation related to cellular senescence or "inflammaging", which participates, among other processes, in establishing pulmonary fibrosis. We evaluate whether polyphenols such as caffeic acid, chlorogenic acid, epicatechin, gallic acid, quercetin, or resveratrol combined with different senotherapeutics such as metformin or rapamycin, and antifibrotic drugs such as nintedanib or pirfenidone, could present beneficial actions in an in vitro model of senescent MRC-5 lung fibroblasts. A senescent-associated secretory phenotype (SASP) was evaluated by the measurement of interleukin (IL)-6, IL-8, and IL-1ß. The senescent-associated ß-galactosidase (SA-ß-gal) activity and cellular proliferation were assessed. Fibrosis was evaluated using a Picrosirius red assay and the gene expression of fibrosis-related genes. Epithelial-mesenchymal transition (EMT) was assayed in the A549 cell line exposed to Transforming Growth Factor (TGF)-ß in vitro. The combination that demonstrated the best results was metformin and caffeic acid, by inhibiting IL-6 and IL-8 in senescent MRC-5 cells. Metformin and caffeic acid also restore cellular proliferation and reduce SA-ß-gal activity during senescence induction. The collagen production by senescent MRC-5 cells was inhibited by epicatechin alone or combined with drugs. Epicatechin and nintedanib were able to control EMT in A549 cells. In conclusion, caffeic acid and epicatechin can potentially increase the effectiveness of senotherapeutic drugs in controlling lung diseases whose pathophysiological component is the presence of senescent cells and fibrosis.


Subject(s)
Cellular Senescence , Fibroblasts , Lung , Polyphenols , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , Cellular Senescence/drug effects , Polyphenols/pharmacology , Lung/pathology , Lung/drug effects , Lung/metabolism , A549 Cells , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Metformin/pharmacology , Caffeic Acids/pharmacology , Indoles/pharmacology , Senotherapeutics/pharmacology , Cell Line , Senescence-Associated Secretory Phenotype/drug effects , Sirolimus/pharmacology , Interleukin-8/metabolism , Interleukin-8/genetics , Transforming Growth Factor beta/metabolism , Pyridones
9.
Int J Mol Sci ; 25(13)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39000409

ABSTRACT

Cardiac fibrosis is a severe outcome of Chagas disease (CD), caused by the protozoan Trypanosoma cruzi. Clinical evidence revealed a correlation between fibrosis levels with impaired cardiac performance in CD patients. Therefore, we sought to analyze the effect of inhibitors of TGF-ß (pirfenidone), p38-MAPK (losmapimod) and c-Jun (SP600125) on the modulation of collagen deposition in cardiac fibroblasts (CF) and in vivo models of T. cruzi chronic infection. Sirius Red/Fast Green dye was used to quantify both collagen expression and total protein amount, assessing cytotoxicity. The compounds were also used to treat C57/Bl6 mice chronically infected with T. cruzi, Brazil strain. We identified an anti-fibrotic effect in vitro for pirfenidone (TGF-ß inhibitor, IC50 114.3 µM), losmapimod (p38 inhibitor, IC50 17.6 µM) and SP600125 (c-Jun inhibitor, IC50 3.9 µM). This effect was independent of CF proliferation since these compounds do not affect T. cruzi-induced host cell multiplication as measured by BrdU incorporation. Assays of chronic infection of mice with T. cruzi have shown a reduction in heart collagen by pirfenidone. These results propose a novel approach to fibrosis therapy in CD, with the prospect of repurposing pirfenidone to prevent the onset of ECM accumulation in the hearts of the patients.


Subject(s)
Chagas Cardiomyopathy , Fibrosis , Mice, Inbred C57BL , Pyridones , Animals , Pyridones/pharmacology , Pyridones/therapeutic use , Chagas Cardiomyopathy/drug therapy , Chagas Cardiomyopathy/parasitology , Chagas Cardiomyopathy/metabolism , Chagas Cardiomyopathy/pathology , Mice , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/parasitology , Myocardium/pathology , Myocardium/metabolism , Collagen/metabolism , Trypanosoma cruzi/drug effects , Humans , Chronic Disease , Transforming Growth Factor beta/metabolism , Disease Models, Animal , p38 Mitogen-Activated Protein Kinases/metabolism , Male , Anthracenes
10.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062847

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) and their extracellular vesicles (MSC-EVs) have been described to have important roles in tissue regeneration, including tissue repair, control of inflammation, enhancing angiogenesis, and regulating extracellular matrix remodeling. MSC-EVs have many advantages for use in regeneration therapies such as facility for dosage, histocompatibility, and low immunogenicity, thus possessing a lower possibility of rejection. In this work, we address the potential activity of MSC-EVs isolated from adipose-derived MSCs (ADMSC-EVs) cultured on cross-linked dextran microcarriers, applied to test the scalability and reproducibility of EV production. Isolated ADMSC-EVs were added into cultured human dermal fibroblasts (NHDF-1), keratinocytes (HaCat), endothelial cells (HUVEC), and THP-1 cell-derived macrophages to evaluate cellular responses (i.e., cell proliferation, cell migration, angiogenesis induction, and macrophage phenotype-switching). ADMSC viability and phenotype were assessed during cell culture and isolated ADMSC-EVs were monitored by nanotracking particle analysis, electron microscopy, and immunophenotyping. We observed an enhancement of HaCat proliferation; NHDF-1 and HaCat migration; endothelial tube formation on HUVEC; and the expression of inflammatory cytokines in THP-1-derived macrophages. The increased expression of TGF-ß and IL-1ß was observed in M1 macrophages treated with higher doses of ADMSC-EVs. Hence, EVs from microcarrier-cultivated ADMSCs are shown to modulate cell behavior, being able to induce skin tissue related cells to migrate and proliferate as well as stimulate angiogenesis and cause balance between pro- and anti-inflammatory responses in macrophages. Based on these findings, we suggest that the isolation of EVs from ADMSC suspension cultures makes it possible to induce in vitro cellular responses of interest and obtain sufficient particle numbers for the development of in vivo concept tests for tissue regeneration studies.


Subject(s)
Cell Proliferation , Extracellular Vesicles , Macrophages , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Extracellular Vesicles/metabolism , Macrophages/metabolism , Macrophages/cytology , Cell Movement , THP-1 Cells , Fibroblasts/metabolism , Fibroblasts/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Cell Culture Techniques/methods , Cells, Cultured , Keratinocytes/metabolism , Keratinocytes/cytology , Cytokines/metabolism
11.
Food Chem ; 459: 140371, 2024 Nov 30.
Article in English | MEDLINE | ID: mdl-39002333

ABSTRACT

The increase of coarse particulate matter (PM10) due to industrialization and urban sprawl has been identified as a significant contributor to air pollution and a threat to human skin health and premature aging. The objective was to analyze the antioxidant effect of phenolic-enriched extracts (PHE) obtained from black bean (BB) and pinto bean (PB) varieties (Phaseolus vulgaris L.) and pure phenolic compounds (rutin, catechin, and gallic acid) in two human dermal fibroblasts cell lines exposed to PM10. Petunidin-3-O-glucoside was the most abundant anthocyanin, with 57 ± 0.9 mg/g dry extract (DE) in PHE-BB. Gallic acid was the prevalent phenolic acid with 8.2 ± 2.8 mg/g DE in PHE-BB (p < 0.05). Hs27 and Hs68 cell lines were exposed to PM10 (100 µg/mL) to induce oxidative stress; PHE-BB reduced it by 69% ± 12 and PHE-PB by 80% ± 5 relative to PM10 treatment (p < 0.05). Delphinidin-3-O-glucoside showed the highest binding affinity in adenosine monophosphate-activated protein kinase (AMPK) with -9.0 kcal/mol and quercetin-3-D-galactoside with -6.9 kcal/mol in sirtuin 1 (Sirt1). Rutin increased the expression of Sirt1 by 30% (p < 0.05) in the Hs27 cell line treated with PM10. Common bean extracts can potentially reduce oxidative stress induced by PM10 in human dermal fibroblasts.


Subject(s)
Fibroblasts , Phaseolus , Plant Extracts , Polycyclic Aromatic Hydrocarbons , Polyphenols , Reactive Oxygen Species , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , Plant Extracts/pharmacology , Plant Extracts/chemistry , Phaseolus/chemistry , Cell Line , Polyphenols/pharmacology , Polyphenols/chemistry , Reactive Oxygen Species/metabolism , Polycyclic Aromatic Hydrocarbons/pharmacology , Polycyclic Aromatic Hydrocarbons/chemistry , Antioxidants/pharmacology , Antioxidants/chemistry , Oxidative Stress/drug effects
12.
Cell Signal ; 120: 111241, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38825173

ABSTRACT

Cardiac fibroblasts (CF) are mesenchymal-type cells responsible for maintaining the homeostasis of the heart's extracellular matrix (ECM). Their dysfunction leads to excessive secretion of ECM proteins, tissue stiffening, impaired nutrient and oxygen exchange, and electrical abnormalities in the heart. Additionally, CF act as sentinel cells in the cardiac tissue microenvironment, responding to various stimuli that may affect heart function. Deleterious stimuli induce an inflammatory response in CF, increasing the secretion of cytokines such as IL-1ß and TNF-α and the expression of cell adhesion molecules like ICAM1 and VCAM1, initially promoting damage resolution by recruiting immune cells. However, constant harmful stimuli lead to a chronic inflammatory process and heart dysfunction. Therefore, it is necessary to study the mechanisms that govern CF inflammation. NFκB is a key regulator of the cardiac inflammatory process, making the search for mechanisms of NFκB regulation and CF inflammatory response crucial for developing new treatment options for cardiovascular diseases. SGK1, a serine-threonine protein kinase, is one of the regulators of NFκB and is involved in the fibrotic effects of angiotensin II and aldosterone, as well as in CF differentiation. However, its role in the CF inflammatory response is unknown. On the other hand, many bioactive natural products have demonstrated anti-inflammatory effects, but their role in CF inflammation is unknown. One such molecule is boldine, an alkaloid obtained from Boldo (Peumus boldus), a Chilean endemic tree with proven cytoprotective effects. However, its involvement in the regulation of SGK1 and CF inflammation is unknown. In this study, we evaluated the role of SGK1 and boldine in the inflammatory response in CF isolated from neonatal Sprague-Dawley rats. The involvement of SGK1 was analyzed using GSK650394, a specific SGK1 inhibitor. Our results demonstrate that SGK1 is crucial for LPS- and IFN-γ-induced inflammatory responses in CF (cytokine expression, cell adhesion molecule expression, and leukocyte adhesion). Furthermore, a conditioned medium (intracellular content of CF subject to freeze/thaw cycles) was used to simulate a sterile inflammation condition. The conditioned medium induced a potent inflammatory response in CF, which was completely prevented by the SGK1 inhibitor. Finally, our results indicate that boldine inhibits both SGK1 activation and the CF inflammatory response induced by LPS, IFN-γ, and CF-conditioned medium. Taken together, our results position SGK1 as an important regulator of the CF inflammatory response and boldine as a promising anti-inflammatory drug in the context of cardiovascular diseases.


Subject(s)
Aporphines , Fibroblasts , Immediate-Early Proteins , NF-kappa B , Protein Serine-Threonine Kinases , Signal Transduction , Animals , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Immediate-Early Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Signal Transduction/drug effects , Rats , Aporphines/pharmacology , Inflammation/metabolism , Inflammation/pathology , Myocardium/pathology , Myocardium/metabolism , Cells, Cultured , Rats, Sprague-Dawley
13.
Toxicol In Vitro ; 99: 105883, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936442

ABSTRACT

Melanoma is a type of tumor skin with high metastatic potential. Reconstructed human skin, development for pre-clinic assay, are make using primary human cells, but with same limitations. The aim this study was to characterize a cell culture model, with structure similar to human skin containing melanoma cells entirely from cell lines. Reconstructed skin with melanoma were development using human fibroblasts (MRC5), human epidermal keratinocytes (HaCat), and human melanoma (SK-MEL-28) embedded in collagen type I. The structure was characterized by hematoxylin-eosin stained, as well as points of melanoma cell invasion, which was associated with activity of MMPs (MMP-2 and MMP-9) by zymographic method. Then, the gene expression of the target molecular mechanisms involved in melanoma progression were evaluated. Here, the model development showed a region epidermis organized and separated from the dermis, with fibroblast cells confined and melanoma cells form delimited area invasion. MMP-2 and MMP-9 were identified during of cell culture and gene expression of BRAF, NRAS, and Vimentin was confirmed. The proposed model provides one more opportunity to study in vitro tumor biology of melanoma and also to allows the study of new drugs with more reliable results then whats we would find in vivo.


Subject(s)
Fibroblasts , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Melanoma , Skin Neoplasms , Humans , Melanoma/pathology , Melanoma/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Skin Neoplasms/pathology , Fibroblasts/metabolism , Fibroblasts/drug effects , Cell Line, Tumor , Skin/metabolism , Skin/pathology , Neoplasm Invasiveness , Keratinocytes/drug effects , Cell Line , Vimentin/metabolism , Vimentin/genetics
14.
Sci Prog ; 107(2): 368504241253695, 2024.
Article in English | MEDLINE | ID: mdl-38801654

ABSTRACT

The current emphasis within the cosmetic market on sustainable ingredients has heightened the exploration of new sources for natural, active components. Actinomycetota, recognized for producing pigments with bioactive potential, offer promising functional cosmetic ingredients. This study aimed to optimize pigment and antioxidant metabolite production from the Gordonia hongkongensis strain EUFUS-Z928 by implementing the Plackett-Burman experimental design and response surface methodology. Extracts derived from this strain exhibited no cytotoxic activity against human primary dermal fibroblast (HDFa, ATCC® PCS-201-012™, Primary Dermal Fibroblast; Normal, Human, Adult). Eight variables, including inoculum concentration, carbon and nitrogen source concentration, NaCl concentration, pH, incubation time, temperature, and stirring speed, were analyzed using the Plackett-Burman experimental design. Subsequently, factors significantly influencing pigment and antioxidant metabolite production, such as temperature, inoculum concentration, and agitation speed, were further optimized using response surface methodology and Box-Behnken design. The results demonstrated a substantial increase in absorbance (from 0.091 to 0.32), DPPH radical scavenging capacity (from 27.60% to 84.61%), and ABTS radical scavenging capacity (from 17.39% to 79.77%) compared to responses obtained in the isolation medium. The validation of the mathematical model accuracy exceeded 90% for all cases. Furthermore, liquid chromatography coupled with mass spectrometry (LC-MS) facilitated the identification of compounds potentially responsible for enhanced pigment production and antioxidant capacity in extracts derived from G. hongkongensis. Specifically, six carotenoids, red-orange pigments with inherent antioxidant capacity, were identified as the main enhanced compounds. This comprehensive approach effectively optimized the culture conditions and medium of a G. hongkongensis strain, resulting in enhanced carotenoid production and antioxidant capacity. Beyond identifying bioactive compounds and their potential cosmetic applications, this study offers insights into the broader industrial applicability of these extracts. It underscores the potential of G. hongkongensis and hints at the future utilization of other untapped sources of rare actinomycetes within the industry.


Subject(s)
Antioxidants , Carotenoids , Antioxidants/metabolism , Antioxidants/chemistry , Carotenoids/metabolism , Carotenoids/chemistry , Humans , Fibroblasts/metabolism , Fibroblasts/drug effects , Gordonia Bacterium/metabolism
15.
J Periodontal Res ; 59(3): 611-621, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38500269

ABSTRACT

BACKGROUND AND OBJECTIVE: Forkhead box-O 1 (FOXO1) is a transcription factor actively involved in oral wound healing at the epithelial barrier. However, less is known regarding the role of FOXO1 during the tissue repair response in the connective tissue compartment. This study explored the involvement of FOXO1 in the modulation of fibroblast activity related to wound healing. METHODS: Primary cultures of human gingival fibroblasts were obtained from four healthy young donors. Myofibroblastic differentiation, collagen gel contraction, cell migration, cell spreading, and integrin activation were evaluated in the presence or absence of a FOXO1 inhibitor (AS1842856). Variations in mRNA and proteins of interest were evaluated through qRT-PCR and western blot, respectively. Distribution of actin, α-smooth muscle actin, and ß1 integrin was evaluated using immunofluorescence. FOXO1 and TGF-ß1 expression in gingival wound healing was assessed by immunohistochemistry in gingival wounds performed in C57BL/6 mice. Images were analyzed using ImageJ/Fiji. ANOVA or Kruskal-Wallis test followed by Tukey's or Dunn's post-hoc test was performed. All data are expressed as mean ± SD. p < .05 was considered statistically significant. RESULTS: FOXO1 inhibition caused a decrease in the expression of the myofibroblastic marker α-SMA along with a reduction in fibronectin, type I collagen, TGF-ß1, and ß1 integrin mRNA level. The FOXO1 inhibitor also caused decreases in cell migration, cell spreading, collagen gel contraction, and ß1 integrin activation. FOXO1 and TGF-ß1 were prominently expressed in gingival wounds in fibroblastic cells located at the wound bed. CONCLUSION: The present study indicates that FOXO1 plays an important role in the modulation of several wound-healing functions in gingival fibroblast. Moreover, our findings reveal an important regulatory role for FOXO1 on the differentiation of gingival myofibroblasts, the regulation of cell migration, and collagen contraction, all these functions being critical during tissue repair and fibrosis.


Subject(s)
Actins , Cell Movement , Fibroblasts , Forkhead Box Protein O1 , Gingiva , Wound Healing , Humans , Gingiva/cytology , Gingiva/metabolism , Wound Healing/physiology , Fibroblasts/metabolism , Forkhead Box Protein O1/metabolism , Animals , Cells, Cultured , Cell Differentiation , Mice, Inbred C57BL , Transforming Growth Factor beta1/metabolism , Mice , Integrin beta1 , Myofibroblasts , Quinolones
16.
Biotechnol Bioeng ; 121(6): 1961-1972, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38555480

ABSTRACT

Oxygen is essential for tissue regeneration, playing a crucial role in several processes, including cell metabolism and immune response. Therefore, the delivery of oxygen to wounds is an active field of research, and recent studies have highlighted the potential use of photosynthetic biomaterials as alternative oxygenation approach. However, while plants have traditionally been used to enhance tissue regeneration, their potential to produce and deliver local oxygen to wounds has not yet been explored. Hence, in this work we studied the oxygen-releasing capacity of Marchantia polymorpha explants, showing their capacity to release oxygen under different illumination settings and temperatures. Moreover, co-culture experiments revealed that the presence of these explants had no adverse effects on the viability and morphology of fibroblasts in vitro, nor on the viability of zebrafish larvae in vivo. Furthermore, oxygraphy assays demonstrate that these explants could fulfill the oxygen metabolic requirements of zebrafish larvae and freshly isolated skin biopsies ex vivo. Finally, the biocompatibility of explants was confirmed through a human skin irritation test conducted in healthy volunteers following the ISO-10993-10-2010. This proof-of-concept study provides valuable scientific insights, proposing the potential use of freshly isolated plants as biocompatible low-cost oxygen delivery systems for wound healing and tissue regeneration.


Subject(s)
Bandages , Oxygen , Photosynthesis , Zebrafish , Animals , Oxygen/metabolism , Proof of Concept Study , Humans , Wound Healing/drug effects , Skin/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism
17.
Photobiomodul Photomed Laser Surg ; 42(2): 140-147, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38393824

ABSTRACT

Objective: The objective of this study is to evaluate the potential effects of photobiomodulation (PBM) on cell proliferation and extracellular matrix production of human fibroblasts (FN1) cultured in 2D. Background: Patients with healing difficulties suffer injuries that take time to recover. In addition, aging can be seen in our faces daily when we look in the mirror; in both situations, collagen production is reduced. Fibroblasts act in the beginning and at the end of the inflammation phase, signaling to immune agents, and platelets, and producing collagen, coordinating repair. PBM increases cell viability, proliferation, and mRNA production. Methods: Human fibroblasts were irradiated three times after cell seed (after 24, 48, and 72 h) using a gallium-aluminum arsenideGaAlAs low-level laser (LLL). Cell viability, proliferative response, synthesis of collagen types I and III, and soluble collagen production were analyzed. The statistical significance of differences between groups was determined using unpaired one-way analysis of variance (ANOVA) p < 0.05. Results: PBM increased significantly the number of fibroblasts, and the production of collagen types I (Col I) and III (Col III), after three sessions of LLL with 2.5 J per session, every 24 h, for 3 consecutive days; total energy delivered after 72 h is 7.5 J. Conclusions: This energy density of LLL increases fibroblast proliferation and collagen production in vitro without side effects.


Subject(s)
Low-Level Light Therapy , Humans , Collagen/metabolism , Extracellular Matrix/metabolism , Cell Proliferation , Fibroblasts/metabolism
18.
Cells ; 12(24)2023 12 08.
Article in English | MEDLINE | ID: mdl-38132110

ABSTRACT

The study of 3D cell culture has increased in recent years as a model that mimics the tumor microenvironment (TME), which is characterized by exhibiting cellular heterogeneity, allowing the modulation of different signaling pathways that enrich this microenvironment. The TME exhibits two main cell populations: cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAM). The aim of this study was to investigate 3D cell cultures of non-small cell lung cancer (NSCLC) alone and in combination with short-term cultured dermal fibroblasts (FDH) and with differentiated macrophages of the THP-1 cell line. Homotypic and heterotypic spheroids were morphologically characterized using light microscopy, immunofluorescence and transmission electron microscopy. Cell viability, cycle profiling and migration assay were performed, followed by the evaluation of the effects of some chemotherapeutic and potential compounds on homotypic and heterotypic spheroids. Both homotypic and heterotypic spheroids of NSCLC were generated with fibroblasts or macrophages. Heterotypic spheroids with fibroblast formed faster, while homotypic ones reached larger sizes. Different cell populations were identified based on spheroid zoning, and drug effects varied between spheroid types. Interestingly, heterotypic spheroids with fibroblasts showed similar responses to the treatment with different compounds, despite being smaller. Cellular viability analysis required multiple methods, since the responses varied depending on the spheroid type. Because of this, the complexity of the spheroid should be considered when analyzing compound effects. Overall, this study contributes to our understanding of the behavior and response of NSCLC cells in 3D microenvironments, providing valuable insights for future research and therapeutic development.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Spheroids, Cellular , Coculture Techniques , Tumor Microenvironment , Lung Neoplasms/pathology , Macrophages , Cell Culture Techniques , Fibroblasts/metabolism
19.
Front Immunol ; 14: 1243537, 2023.
Article in English | MEDLINE | ID: mdl-37860000

ABSTRACT

Introduction: Eotaxin-1/CCL11 is a pivotal chemokine crucial for eosinophil homing to the lungs of asthmatic patients. Recent studies also suggest that CCL11 is involved in the aging process, as it is upregulated in elderly, and correlated with shorter telomere length in leukocytes from asthmatic children. Despite its potential pro-aging effects, the precise contribution of CCL11 and the underlying mechanisms involved in the promotion of cellular senescence remains unclear. Therefore, the primary goal of this study was to explore the role of CCL11 on senescence development and the signaling pathways activated by this chemokine in lung fibroblasts. Methods: To investigate the targets potentially modulated by CCL11, we performed an in silico analysis using PseudoCell. We validated in vitro the activation of these targets in the human lung fibroblast cell line MRC-5 following rhCCL11 exposure. Finally, we performed differential gene expression analysis in human airway epithelial cells of asthmatic patients to assess CCL11 signaling and activation of additional senescent markers. Results: Our study revealed that eotaxin-1/CCL11 promote reactive oxygen secretion (ROS) production in lung fibroblasts, accompanied by increased activation of the DNA damage response (DDR) and p-TP53 and γH2AX. These modifications were accompanied by cellular senescence promotion and increased secretion of senescence-associated secretory phenotype inflammatory cytokines IL-6 and IL-8. Furthermore, our data show that airway epithelial lung cells from atopic asthmatic patients overexpress CCL11 along with aging markers such as CDKN2A (p16INK4a) and SERPINE1. Discussion: These findings provide new insights into the mechanisms underlying the pro-aging effects of CCL11 in the lungs of asthmatic patients. Understanding the role of CCL11 on senescence development may have important implications for the treatment of age-related lung diseases, such as asthma.


Subject(s)
Asthma , Lung , Child , Humans , Aged , Chemokine CCL11/metabolism , Reactive Oxygen Species/metabolism , Lung/metabolism , Asthma/metabolism , Cellular Senescence , Fibroblasts/metabolism
20.
Cell Signal ; 109: 110778, 2023 09.
Article in English | MEDLINE | ID: mdl-37343898

ABSTRACT

Cardiac fibroblasts (CFs) activation is a common response to most pathological conditions affecting the heart, characterized by increased cellular secretory capacity and increased expression of fibrotic markers, such as collagen I and smooth muscle actin type alpha (α-SMA). Fibrotic activation of CFs induces the increase in tissue protein content, with the consequent tissue stiffness, diastolic dysfunction, and heart failure. Therefore, the search for new mechanisms of CFs activation is important to find novel treatments for cardiac diseases characterized by fibrosis. In this regard, TGF-ß1, a cytokine with proinflammatory and fibrotic properties, is crucial in the CFs activation and the development of fibrotic diseases, whereas its molecular targets are not completely known. Serum and glucocorticoid-regulated kinase (SGK1) is a protein involved in various pathophysiological phenomena, especially cardiac and renal diseases that curse with fibrosis. Additionally, SGK1 phosphorylates and regulates the activity and expression of several targets, highlighting FoxO3a for its role in the regulation of oxidative stress and CFs activation induced by TGF-ß1. However, the regulation of SGK1 by TGF-ß1 and its role in CFs activation have not been studied. In this work, we evaluate the role of SGK1 in CFs isolated from neonatal Sprague-Dawley rats. The participation of SGK1 in the fibrotic activation of CFs induced by TGF-ß1 was analyzed, using an inhibitor or siRNA of SGK1. In addition, the role of SGK1 on the regulation of FoxO3a and oxidative stress induced by TGF-ß1 was analyzed. Our results indicate that TGF-ß1 increased both the activity and expression of SGK1 in CFs, requiring the activation of MAPKs, ERK1/2, p38 and JNK, while inhibition and silencing of SGK1 prevented TGF-ß1-induced fibrotic activation of CFs. In addition, SGK1 inhibition prevented FoxO3a inactivation and expression reduction, catalase and SOD2 expression decrease, and the increase of oxidative stress induced by TGF-ß1. Taken together, our results position SGK1 as an important regulator of CFs activation driven by TGF-ß1, at least in part, through the regulation of FoxO3a and oxidative stress.


Subject(s)
Myocardium , Transforming Growth Factor beta1 , Rats , Animals , Rats, Sprague-Dawley , Myocardium/metabolism , Transforming Growth Factor beta1/metabolism , Oxidative Stress , Fibroblasts/metabolism , Fibrosis
SELECTION OF CITATIONS
SEARCH DETAIL