Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Intervalo de año de publicación
1.
J Proteome Res ; 23(7): 2561-2575, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38810023

RESUMEN

Emergent advancements on the role of the intestinal microbiome for human health and disease necessitate well-defined intestinal cellular models to study and rapidly assess host, microbiome, and drug interactions. Differentiated Caco-2 cell line is commonly utilized as an epithelial model for drug permeability studies and has more recently been utilized for investigating host-microbiome interactions. However, its suitability to study such interactions remains to be characterized. Here, we employed multilevel proteomics to demonstrate that both spontaneous and butyrate-induced Caco-2 differentiations displayed similar protein and pathway changes, including the downregulation of proteins related to translation and proliferation and upregulation of functions implicated in host-microbiome interactions, such as cell adhesion, tight junction, extracellular vesicles, and responses to stimuli. Lysine acetylomics revealed that histone protein acetylation levels were decreased along with cell differentiation, while the acetylation in proteins associated with mitochondrial functions was increased. This study also demonstrates that, compared to spontaneous differentiation methods, butyrate-containing medium accelerates Caco-2 differentiation, with earlier upregulation of proteins related to host-microbiome interactions, suggesting its superiority for assay development using this intestinal model. Altogether, this multiomics study emphasizes the controlled progression of Caco-2 differentiation toward a specialized intestinal epithelial-like cell and establishes its suitability for investigating the host-microbiome interactions.


Asunto(s)
Butiratos , Diferenciación Celular , Proteómica , Humanos , Células CACO-2 , Proteómica/métodos , Butiratos/farmacología , Acetilación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/microbiología , Microbioma Gastrointestinal , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/microbiología , Proteoma/metabolismo , Proteoma/análisis
2.
Cancer Immunol Immunother ; 73(1): 11, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38231448

RESUMEN

The human CC chemokine receptor 8 (CCR8) is specifically expressed on tumor-infiltrating regulatory T cells (TITRs) and is a promising drug target for cancer immunotherapy. However, the role of CCR8 signaling in TITR biology and the effectiveness of CCR8 small molecule antagonists as TITR-targeting immunotherapy remain subjects of ongoing debate. In this work, we generated a novel cellular model of TITRs by culturing peripheral blood mononuclear cell-derived regulatory T cells in medium containing tumor cell-conditioned medium, CD3/CD28 activator, interleukin-2 and 1α,25-dihydroxyvitamin D3. This cellular model (named TITR mimics) highly and stably expressed a series of TITR signature molecules, including CCR8, FOXP3, CD30, CD39, CD134, CD137, TIGIT and Tim-3. Moreover, TITR mimics displayed robust in vitro immunosuppressive activity. To unravel the functional role of CCR8 in TITR mimics, a chemotaxis assay was performed showing strong and CCR8-specific migration toward CCL1, the natural chemokine agonist of CCR8. However, either stimulation (with CCL1) or blocking (with the small molecule antagonist NS-15) of CCR8 signaling did not affect the immunosuppressive activity, proliferation and survival of TITR mimics. Collectively, our work provides a method for the generation of TITR mimics in vitro, which can be used to study TITR biology and to evaluate drug candidates targeting TITRs. Furthermore, our findings suggest that CCR8 signaling primarily regulates migration of these cells.


Asunto(s)
Leucocitos Mononucleares , Neoplasias , Humanos , Receptores CCR8 , Linfocitos T Reguladores , Medios de Cultivo Condicionados
3.
BMC Cancer ; 24(1): 105, 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38342891

RESUMEN

BACKGROUND: Histiocytoses are rare disorders manifested by increased proliferation of pathogenic myeloid cells sharing histological features with macrophages or dendritic cells and accumulating in various organs, i.a., bone and skin. Pre-clinical in vitro models that could be used to determine molecular pathways of the disease are limited, hence research on histiocytoses is challenging. The current study compares cytophysiological features of progenitor, stromal-like cells derived from histiocytic lesions (sl-pHCs) of three pediatric patients with different histiocytoses types and outcomes. The characterized cells may find potential applications in drug testing. METHODS: Molecular phenotype of the cells, i.e. expression of CD1a and CD207 (langerin), was determined using flow cytometry. Cytogenetic analysis included GTG-banded metaphases and microarray (aCGH) evaluation. Furthermore, the morphology and ultrastructure of cells were evaluated using a confocal and scanning electron microscope. The microphotographs from the confocal imaging were used to reconstruct the mitochondrial network and its morphology. Basic cytophysiological parameters, such as viability, mitochondrial activity, and proliferation, were analyzed using multiple cellular assays, including Annexin V/7-AAD staining, mitopotential analysis, BrdU test, clonogenicity analysis, and distribution of cells within the cell cycle. Biomarkers potentially associated with histiocytoses progression were determined using RT-qPCR at mRNA, miRNA and lncRNA levels. Intracellular accumulation of histiocytosis-specific proteins was detected with Western blot. Cytotoxicyty and IC50 of vemurafenib and trametinib were determined with MTS assay. RESULTS: Obtained cellular models, i.e. RAB-1, HAN-1, and CHR-1, are heterogenic in terms of molecular phenotype and morphology. The cells express CD1a/CD207 markers characteristic for dendritic cells, but also show intracellular accumulation of markers characteristic for cells of mesenchymal origin, i.e. vimentin (VIM) and osteopontin (OPN). In subsequent cultures, cells remain viable and metabolically active, and the mitochondrial network is well developed, with some distinctive morphotypes noted in each cell line. Cell-specific transcriptome profile was noted, providing information on potential new biomarkers (non-coding RNAs) with diagnostic and prognostic features. The cells showed different sensitivity to vemurafenib and trametinib. CONCLUSION: Obtained and characterized cellular models of stromal-like cells derived from histiocytic lesions can be used for studies on histiocytosis biology and drug testing.


Asunto(s)
Histiocitosis de Células de Langerhans , Humanos , Niño , Histiocitosis de Células de Langerhans/tratamiento farmacológico , Histiocitosis de Células de Langerhans/genética , Histiocitosis de Células de Langerhans/diagnóstico , Vemurafenib , Macrófagos/metabolismo , Biomarcadores , Fenotipo , Antígenos CD , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo
4.
Cell Biochem Funct ; 42(2): e3932, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38332678

RESUMEN

Mucopolysaccharidosis type II (MPS II) is an inborn error of the metabolism resulting from several possible mutations in the gene coding for iduronate-2-sulfatase (IDS), which leads to a great clinical heterogeneity presented by these patients. Many studies demonstrate the involvement of oxidative stress in the pathogenesis of inborn errors of metabolism, and mitochondrial dysfunction and oxidative stress can be related since most of reactive oxygen species come from mitochondria. Cellular models have been used to study different diseases and are useful in biochemical research to investigate them in a new promising way. The aim of this study is to develop a heterozygous cellular model for MPS II and analyze parameters of oxidative stress and mitochondrial dysfunction and investigate the in vitro effect of genistein and coenzyme Q10 on these parameters for a better understanding of the pathophysiology of this disease. The HP18 cells (heterozygous c.261_266del6/c.259_261del3) showed almost null results in the activity of the IDS enzyme and presented accumulation of glycosaminoglycans (GAGs), allowing the characterization of this knockout cellular model by MPS II gene editing. An increase in the production of reactive species was demonstrated (p < .05 compared with WT vehicle group) and genistein at concentrations of 25 and 50 µm decreased in vitro its production (p < .05 compared with HP18 vehicle group), but there was no effect of coenzyme Q10 in this parameter. There was a tendency for lysosomal pH change in HP18 cells in comparison to WT group and none of the antioxidants tested demonstrated any effect on this parameter. There was no increase in the activity of the antioxidant enzymes superoxide dismutase and catalase and oxidative damage to DNA in HP18 cells in comparison to WT group and neither genistein nor coenzyme q10 had any effect on these parameters. Regarding mitochondrial membrane potential, genistein induced mitochondrial depolarization in both concentrations tested (p < .05 compared with HP18 vehicle group and compared with WT vehicle group) and incubation with coenzyme Q10 demonstrated no effect on this parameter. In conclusion, it is hypothesized that our cellular model could be compared with a milder MPS II phenotype, given that the accumulation of GAGs in lysosomes is not as expressive as another cellular model for MPS II presented in the literature. Therefore, it is reasonable to expect that there is no mitochondrial depolarization and no DNA damage, since there is less lysosomal impairment, as well as less redox imbalance.


Asunto(s)
Iduronato Sulfatasa , Enfermedades Mitocondriales , Mucopolisacaridosis II , Ubiquinona/análogos & derivados , Humanos , Mucopolisacaridosis II/tratamiento farmacológico , Mucopolisacaridosis II/genética , Genisteína/farmacología , Potencial de la Membrana Mitocondrial , Estrés Oxidativo , Iduronato Sulfatasa/metabolismo , Iduronato Sulfatasa/farmacología , Antioxidantes/farmacología , Antioxidantes/metabolismo
5.
Int J Biometeorol ; 68(8): 1675-1687, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38814473

RESUMEN

The present study was conducted to understand transcriptional response of skin fibroblast of yak (Bos grunniens) and cows of Bos indicus origin to hypoxia stress. Six primary fibroblast cell lines derived from three individuals each of Ladakhi yak (Bos grunniens) and Sahiwal cows (Bos indicus) were exposed to low oxygen concentration for a period of 24 h, 48 h and 72 h. The expression of 10 important genes known to regulate hypoxia response such as HIF1A, VEGFA, EPAS1, ATP1A1, GLUT1, HMOX1, ECE1, TNF-A, GPx and SOD were evaluated in fibroblast cells of Ladakhi yak (LAY-Fb) and Sahiwal cows (SAC-Fb) during pre- and post-hypoxia stress. A panel of 10 reference genes (GAPDH, RPL4, EEF1A1, RPS9, HPRT1, UXT, RPS23, B2M, RPS15, ACTB) were also evaluated for their expression stability to perform accurate normalization. The expression of HIF1A was significantly (p < 0.05) induced in both LAY-Fb (2.29-fold) and SAC-Fb (2.07-fold) after 24 h of hypoxia stress. The angiogenic (VEGFA), metabolic (GLUT1) and antioxidant genes (SOD and GPx) were also induced after 24 h of hypoxia stress. However, EPAS1 and ATP1A1 induced significantly (p < 0.05) after 48 h whereas, ECE1 expression induced significantly (p < 0.05) at 72 h after exposure to hypoxia. The TNF-alpha which is a pro-inflammatory gene induced significantly (p < 0.05) at 24 h in SAC-Fb and at 72 h in LAY-Fb. The induction of hypoxia associated genes indicated the utility of skin derived fibroblast as cellular model to evaluate transcriptome signatures post hypoxia stress in populations adapted to diverse altitudes.


Asunto(s)
Altitud , Fibroblastos , Hipoxia , Piel , Animales , Bovinos , Fibroblastos/metabolismo , Piel/metabolismo , Hipoxia/genética , Clima Tropical , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Estrés Fisiológico/genética
6.
Int J Mol Sci ; 25(5)2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38473862

RESUMEN

Hereditary spastic paraplegias (HSPs) comprise a family of degenerative diseases mostly hitting descending axons of corticospinal neurons. Depending on the gene and mutation involved, the disease could present as a pure form with limb spasticity, or a complex form associated with cerebellar and/or cortical signs such as ataxia, dysarthria, epilepsy, and intellectual disability. The progressive nature of HSPs invariably leads patients to require walking canes or wheelchairs over time. Despite several attempts to ameliorate the life quality of patients that have been tested, current therapeutical approaches are just symptomatic, as no cure is available. Progress in research in the last two decades has identified a vast number of genes involved in HSP etiology, using cellular and animal models generated on purpose. Although unanimously considered invaluable tools for basic research, those systems are rarely predictive for the establishment of a therapeutic approach. The advent of induced pluripotent stem (iPS) cells allowed instead the direct study of morphological and molecular properties of the patient's affected neurons generated upon in vitro differentiation. In this review, we revisited all the present literature recently published regarding the use of iPS cells to differentiate HSP patient-specific neurons. Most studies have defined patient-derived neurons as a reliable model to faithfully mimic HSP in vitro, discovering original findings through immunological and -omics approaches, and providing a platform to screen novel or repurposed drugs. Thereby, one of the biggest hopes of current HSP research regards the use of patient-derived iPS cells to expand basic knowledge on the disease, while simultaneously establishing new therapeutic treatments for both generalized and personalized approaches in daily medical practice.


Asunto(s)
Ataxia Cerebelosa , Células Madre Pluripotentes , Paraplejía Espástica Hereditaria , Animales , Humanos , Paraplejía Espástica Hereditaria/genética , Neuronas , Axones , Mutación
7.
J Theor Biol ; 556: 111312, 2023 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-36279960

RESUMEN

Cancer is one of the leading causes of mortality and morbidity among people worldwide. Cancer appears as solid tumors in many cases. Angiogenesis is the growth of blood vessels from the existing vasculature and is one of the imperative processes in tumor growth. Another vital phenomenon for formation and functionality of this vasculature network is lumen formation. The results of recent studies indicate the importance of blood pressure in this mechanism. Computational modeling can study these processes in different scales. Hence, wide varieties of these models have been proposed during recent years. In this research, a multi-scale model is developed for the angiogenesis process. In the extracellular scale, the growth factor concentration is calculated via the reaction diffusion equation. At the cellular scale, growth, migration, and the adhesion of endothelial cells are modeled by the Potts cellular model. At the intra-cellular scale by considering biochemical signals, a Boolean network model describes migration, division, or apoptosis of endothelial cells. A stochastic model developed for lumen formation via inverse membrane blebbing mechanism. A CFD simulation was also used to investigate the role of pulsated blood pressure in the inverse membrane blebbing mechanism. The lumen formation model shows stochastic behavior in blebs expansion and lumen expansion. Comparing the stochastic model's results with the CFD simulation also shows the vital role of pressure pulse and the topology of the blebs in bleb retraction.


Asunto(s)
Células Endoteliales , Humanos , Simulación por Computador , Morfogénesis , Neovascularización Fisiológica
8.
Int J Mol Sci ; 24(7)2023 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-37046993

RESUMEN

Lafora disease (LD) is a neurological disorder characterized by progressive myoclonus epilepsy. The hallmark of the disease is the presence of insoluble forms of glycogen (polyglucosan bodies, or PGBs) in the brain. The accumulation of PGBs is causative of the pathophysiological features of LD. However, despite the efforts made by different groups, the question of why PGBs accumulate in the brain is still unanswered. We have recently demonstrated that, in vivo, astrocytes accumulate most of the PGBs present in the brain, and this could lead to astrocyte dysfunction. To develop a deeper understanding of the defects present in LD astrocytes that lead to LD pathophysiology, we obtained pure primary cultures of astrocytes from LD mice from the postnatal stage under conditions that accumulate PGBs, the hallmark of LD. These cells serve as novel in vitro models for studying PGBs accumulation and related LD dysfunctions. In this sense, the metabolomics of LD astrocytes indicate that they accumulate metabolic intermediates of the upper part of the glycolytic pathway, probably as a consequence of enhanced glucose uptake. In addition, we also demonstrate the feasibility of using the model in the identification of different compounds that may reduce the accumulation of polyglucosan inclusions.


Asunto(s)
Enfermedad de Lafora , Ratones , Animales , Enfermedad de Lafora/metabolismo , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Glucanos/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo
9.
Dokl Biochem Biophys ; 510(1): 91-94, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37582869

RESUMEN

The study investigated the effect of GABA in various concentrations and D-GB-115 at a concentration of 10-7 M on the behavior of Paramecium caudatum. It was shown that GABA increases motor activity and changes the movement strategy of these protozoans, and the dose-effect relationship is domed, which can be explained by the presence of two types of GABA receptors in the outer membrane of paramecia: GABA-A and GABA-B. The active concentrations of GABA range from 10-3 to 10-13 M. The effect of pharmacological agents interacting with the GABA system on the behavior of ciliates (nembutal and D-GB-115) was studied.


Asunto(s)
Paramecium caudatum , Ácido gamma-Aminobutírico , Actividad Motora , Colecistoquinina
10.
Neurobiol Dis ; 162: 105576, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34871734

RESUMEN

There is ample pathological and biological evidence for endo-lysosomal dysfunction in Alzheimer's disease (AD) and emerging genetic studies repeatedly implicate endo-lysosomal genes as associated with increased AD risk. The endo-lysosomal network (ELN) is essential for all cell types of the central nervous system (CNS), yet each unique cell type utilizes cellular trafficking differently (see Fig. 1). Challenges ahead involve defining the role of AD associated genes in the functionality of the endo-lysosomal network (ELN) and understanding how this impacts the cellular dysfunction that occurs in AD. This is critical to the development of new therapeutics that will impact, and potentially reverse, early disease phenotypes. Here we review some early evidence of ELN dysfunction in AD pathogenesis and discuss the role of selected AD-associated risk genes in this pathway. In particular, we review genes that have been replicated in multiple genome-wide association studies(Andrews et al., 2020; Jansen et al., 2019; Kunkle et al., 2019; Lambert et al., 2013; Marioni et al., 2018) and reviewed in(Andrews et al., 2020) that have defined roles in the endo-lysosomal network. These genes include SORL1, an AD risk gene harboring both rare and common variants associated with AD risk and a role in trafficking cargo, including APP, through the ELN; BIN1, a regulator of clathrin-mediated endocytosis whose expression correlates with Tau pathology; CD2AP, an AD risk gene with roles in endosome morphology and recycling; PICALM, a clathrin-binding protein that mediates trafficking between the trans-Golgi network and endosomes; and Ephrin Receptors, a family of receptor tyrosine kinases with AD associations and interactions with other AD risk genes. Finally, we will discuss how human cellular models can elucidate cell-type specific differences in ELN dysfunction in AD and aid in therapeutic development.


Asunto(s)
Enfermedad de Alzheimer , Proteínas de Transporte de Membrana , Enfermedad de Alzheimer/genética , Endosomas/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/metabolismo , Lisosomas/metabolismo , Proteínas de Transporte de Membrana/genética , Fenotipo
11.
Int J Mol Sci ; 23(8)2022 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-35456986

RESUMEN

Cellular senescence is defined as irreversible cell cycle arrest caused by various processes that render viable cells non-functional, hampering normal tissue homeostasis. It has many endogenous and exogenous inducers, and is closely connected with age, age-related pathologies, DNA damage, degenerative disorders, tumor suppression and activation, wound healing, and tissue repair. However, the literature is replete with contradictory findings concerning its triggering mechanisms, specific biomarkers, and detection protocols. This may be partly due to the wide range of cellular and in vivo animal or human models of accelerated aging that have been used to study senescence and test senolytic drugs. This review summarizes recent findings concerning senescence, presents some widely used cellular and animal senescence models, and briefly describes the best-known senolytic agents.


Asunto(s)
Envejecimiento , Senescencia Celular , Envejecimiento/genética , Animales , Biomarcadores , Puntos de Control del Ciclo Celular , Senescencia Celular/genética , Daño del ADN
12.
Int J Mol Sci ; 23(6)2022 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-35328496

RESUMEN

Coronary in-stent restenosis is a late complication of angioplasty. It is a multifactorial process that involves vascular smooth muscle cells (VSMCs), endothelial cells, and inflammatory and genetic factors. In this study, the transcriptomic landscape of VSMCs' phenotypic switch process was assessed under stimuli resembling stent injury. Co-cultured contractile VSMCs and endothelial cells were exposed to a bare metal stent and platelet-derived growth factor (PDGF-BB) 20 ng/mL. Migratory capacity (wound healing assay), proliferative capacity, and cell cycle analysis of the VSMCs were performed. RNAseq analysis of contractile vs. proliferative VSMCs was performed. Gene differential expression (DE), identification of new long non-coding RNA candidates (lncRNAs), gene ontology (GO), and pathway enrichment (KEGG) were analyzed. A competing endogenous RNA network was constructed, and significant lncRNA-miRNA-mRNA axes were selected. VSMCs exposed to "stent injury" conditions showed morphologic changes, with proliferative and migratory capacities progressing from G0-G1 cell cycle phase to S and G2-M. RNAseq analysis showed DE of 1099, 509 and 64 differentially expressed mRNAs, lncRNAs, and miRNAs, respectively. GO analysis of DE genes showed significant enrichment in collagen and extracellular matrix organization, regulation of smooth muscle cell proliferation, and collagen biosynthetic process. The main upregulated nodes in the lncRNA-mediated ceRNA network were PVT1 and HIF1-AS2, with downregulation of ACTA2-AS1 and MIR663AHG. The PVT1 ceRNA axis appears to be an attractive target for in-stent restenosis diagnosis and treatment.


Asunto(s)
Reestenosis Coronaria , MicroARNs , ARN Largo no Codificante , Reestenosis Coronaria/genética , Células Endoteliales/metabolismo , Redes Reguladoras de Genes , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética
13.
Dokl Biochem Biophys ; 502(1): 36-39, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35275304

RESUMEN

The  effect  of  adrenaline  in  various  concentrations  and  dopamine  at  a  concentration  of 10-10 mol/mL on the behavior of Paramecium caudatum was studied. It is shown that adrenaline reduces motor activity and changes the movement strategy of these protozoans; a dose-dependent behavioral response on the drug concentration was observed. This effect can be explained by the presence of adrenaline receptors located on the surface of the cell membrane. To study the direct effect of adrenaline on alpha and beta adrenergic receptors, the effect of non-selective adrenoblockers nicergoline and timolol is considered in this paper. At the same time, dopamine at a concentration of 10-10 mol/mL does not have a significant effect on the nature and magnitude of motor activity during the entire registration time, since this organism does not have receptors for this mediator. The proposed method makes it possible to quickly and objectively assess the nature of the effects of various pharmaceuticals acting on the catecholamine system.


Asunto(s)
Paramecium caudatum , Catecolaminas , Epinefrina/farmacología , Células Eucariotas , Preparaciones Farmacéuticas
14.
J Cell Mol Med ; 25(14): 6634-6651, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34075722

RESUMEN

Available therapies aimed at treating age-related osteoporosis are still insufficient. Therefore, designing reliable in vitro model for the analysis of molecular mechanisms underlying senile osteoporosis is highly required. We have isolated and characterized progenitor cells isolated from bone marrow (BMSCs) of osteoporotic mice strain SAM/P6 (BMSCSAM/P6 ). The cytophysiology of BMSCSAM/P6 was for the first time compared with BMSCs isolated from healthy BALB/c mice (BMSCBALB/c ). Characterization of the cells included evaluation of their multipotency, morphology and determination of specific phenotype. Viability of BMSCs cultures was determined in reference to apoptosis profile, metabolic activity, oxidative stress, mitochondrial membrane potential and caspase activation. Additionally, expression of relevant biomarkers was determined with RT-qPCR. Obtained results indicated that BMSCSAM/P6 and BMSCBALB/c show the typical phenotype of mesenchymal stromal cells (CD44+, CD73+, CD90+) and do not express CD45. Further, BMSCSAM/P6 were characterized by deteriorated multipotency, decreased metabolic activity and increased apoptosis occurrence, accompanied by elevated oxidative stress and mitochondria depolarisation. The transcriptome analyses showed that BMSCSAM/P6 are distinguished by lowered expression of molecules crucial for proper osteogenesis, including Coll-1, Opg and Opn. However, the expression of Trap, DANCR1 and miR-124-3p was significantly up-regulated. Obtained results show that BMSCSAM/P6 present features of progenitor cells with disturbed metabolism and could serve as appropriate model for in vitro investigation of age-dependent osteoporosis.


Asunto(s)
Diferenciación Celular/genética , Células Madre Mesenquimatosas/inmunología , Osteogénesis/genética , Osteoporosis/genética , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/inmunología , Animales , Diferenciación Celular/inmunología , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/inmunología , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Osteoblastos/inmunología , Osteoblastos/metabolismo , Osteogénesis/inmunología , Osteoporosis/inmunología , Osteoporosis/patología , Células Madre/inmunología , Células Madre/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/inmunología
15.
Biochem Soc Trans ; 49(4): 1791-1802, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34415299

RESUMEN

Microglia are the major component of the innate immune system in the central nervous system. They promote the maintenance of brain homeostasis as well as support inflammatory processes that are often related to pathological conditions such as neurodegenerative diseases. Depending on the stimulus received, microglia cells dynamically change their phenotype releasing specific soluble factors and largely modify the cargo of their secreted extracellular vesicles (EVs). Despite the mechanisms at the basis of microglia actions have not been completely clarified, the recognized functions exerted by their EVs in patho-physiological conditions represent the proof of the crucial role of these organelles in tuning cell-to-cell communication, promoting either protective or harmful effects. Consistently, in vitro cell models to better elucidate microglia EV production and mechanisms of their release have been increased in the last years. In this review, the main microglial cellular models that have been developed and validated will be described and discussed, with particular focus on those used to produce and derive EVs. The advantages and disadvantages of their use will be evidenced too. Finally, given the wide interest in applying EVs in diagnosis and therapy too, the heterogeneity of available models for producing microglia EVs is here underlined, to prompt a cross-check or comparison among them.


Asunto(s)
Vesículas Extracelulares/metabolismo , Microglía/metabolismo , Modelos Biológicos , Animales , Línea Celular , Humanos , Sanguijuelas
16.
Biochem Biophys Res Commun ; 525(4): 968-973, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32173533

RESUMEN

Myogenic differentiation mechanisms are generally assessed using a murine cell line placed in low concentrations of an animal-derived serum. To more closely approximate in vivo pathophysiological conditions, recent studies have combined the use of human muscle cells with human serum. Nevertheless, the in vitro studies of the effects of a human microenvironment on the differentiation process of human myoblasts require the identification of the culture conditions that would provide an optimal and reproducible differentiation process of human muscle cells. We assessed the differentiation variability resulting from the use of human myoblasts and serums from healthy subjects by measuring the myotube diameter, fusion index and surface covered by myotubes. We showed the preserved cell-dependent variability of the differentiation response of myoblasts cultured in human serums compared to FBS. We found that using a pool of serums reduced the serum-dependent variability of the myogenic response compared to individual serums. We validated our methodology by showing the atrophying effect of pooled serums from COPD patients on healthy human myotubes. By replacing animal-derived tissues with human myoblasts and serums, and by validating the sensitivity of cultured human muscle cells to a pathological microenvironment, this human cell culture model offers a valuable tool for studying the role of the microenvironment in chronic disease.


Asunto(s)
Desarrollo de Músculos/efectos de los fármacos , Mioblastos/citología , Suero/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Persona de Mediana Edad , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Suero/metabolismo , Albúmina Sérica Bovina/farmacología
17.
J Transl Med ; 18(1): 439, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33213473

RESUMEN

BACKGROUND: Poly(ADP-ribose) polymerase inhibitors (PARPis) specifically target homologous recombination deficiency (HRD) cells and display good therapeutic effect in women with advanced-stage BRCA1/2-mutated breast and epithelial ovarian cancer (EOC). However, about 50% of high grade serous ovarian cancers (HGSOC) present with HRD due to epigenetic BRCA1 inactivation, as well as genetic/epigenetic inactivation(s) of other HR genes, a feature known as "BRCAness". Therefore, there is a potential for extending the use of PARPis to these patients if HR status can be identified. METHODS: We have developed a 3D (spheroid) functional assay to assess the sensitivity of two PARPis (niraparib and olaparib) in ascites-derived primary cell cultures (AsPCs) from HGSOC patients. A method for AsPCs preparation was established based on a matrix (agarose), allowing for easy isolation and successive propagation of monolayer and 3D AsPCs. Based on this method, we performed cytotoxicity assays on 42 AsPCs grown both as monolayers and spheroids. RESULTS: The response to PARPis treatment in monolayer AsPCs, was significantly higher, compared to 3D AsPCs, as 88% and 52% of the monolayer AsPCs displayed sensitivity to niraparib and olaparib respectively, while 66% of the 3D AsPCs were sensitive to niraparib and 38% to olaparib, the latter being more consistent with previous estimates of HRD (40%-60%) in EOC. Moreover, niraparib displayed a significantly stronger cytotoxic effect in both in 3D and monolayer AsPCs, which was confirmed by consecutive analyses of the HR pathway activity (γH2AX foci formation) in PARPis-sensitive and resistant AsPCs. Global gene expression comparison of 6 PARPi-resistant and 6 PARPi-sensitive 3D AsPCs was indicative for the predominant downregulation of numerous genes and networks with previously demonstrated roles in EOC chemoresistance, suggesting that the PARPis-sensitive AsPCs could display enhanced sensitivity to other chemotherapeutic drugs, commonly applied in cancer management. Microarray data validation identified 24 potential gene biomarkers associated with PARPis sensitivity. The differential expression of 7 selected biomarkers was consecutively confirmed by immunohistochemistry in matched EOC tumor samples. CONCLUSION: The application of this assay and the potential biomarkers with possible predictive significance to PARPis therapy of EOC patients now need testing in the setting of a clinical trial.


Asunto(s)
Neoplasias Ováricas , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Adenosina Difosfato Ribosa/uso terapéutico , Biomarcadores , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/genética , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico
18.
Int J Mol Sci ; 21(10)2020 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-32422974

RESUMEN

In the western world, colorectal cancer (CRC) is the third most common cause of cancer-related deaths. Survival is closely related to the stage of cancer at diagnosis striking the clinical need for biomarkers capable of early detection. To search for possible biological parameters for early diagnosis of CRC we evaluated protein expression for three CREC (acronym: Cab45, reticulocalbin, ERC-55, calumenin) proteins: reticulocalbin, calumenin, and ERC-55 in a cellular model consisting of a normal derived colon mucosa cell line, NCM460, and a primary adenocarcinoma cell line of the colon, SW480. Furthermore, this cellular model was analyzed by a top-down proteomic approach, 2-dimensional polyacrylamide gel electrophoresis (2D-PAGE) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) for novel putative diagnostic markers by identification of differentially expressed proteins between the two cell lines. A different colorectal carcinoma cell line, HCT 116, was used in a bottom-up proteomic approach with label-free quantification (LFQ) LC-MS/MS. The two cellular models gave sets of putative diagnostic CRC biomarkers. Various of these novel putative markers were verified with increased expression in CRC patient neoplastic tissue compared to the expression in a non-involved part of the colon, including reticulocalbin, calumenin, S100A6 and protein SET. Characterization of these novel identified biological features for CRC patients may have diagnostic potential and therapeutic relevance in this malignancy characterized by a still unmet clinical need.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Mucosa Intestinal/metabolismo , Proteoma/genética , Anciano , Anciano de 80 o más Años , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Neoplasias Colorrectales/patología , Proteínas de Unión al ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Células HCT116 , Chaperonas de Histonas/genética , Humanos , Masculino , Persona de Mediana Edad , Proteína A6 de Unión a Calcio de la Familia S100/genética
19.
Int J Mol Sci ; 21(9)2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32380730

RESUMEN

Gaucher disease (GD) is an autosomal recessive lysosomal storage disorder caused by mutations in the acid ß-glucosidase gene (GBA1). Besides causing GD, GBA1 mutations constitute the main genetic risk factor for developing Parkinson's disease. The molecular basis of neurological manifestations in GD remain elusive. However, neuroinflammation has been proposed as a key player in this process. We exploited CRISPR/Cas9 technology to edit GBA1 in the human monocytic THP-1 cell line to develop an isogenic GD model of monocytes and in glioblastoma U87 cell lines to generate an isogenic GD model of glial cells. Both edited (GBA1 mutant) cell lines presented low levels of mutant acid ß-glucosidase expression, less than 1% of residual activity and massive accumulation of substrate. Moreover, U87 GBA1 mutant cells showed that the mutant enzyme was retained in the ER and subjected to proteasomal degradation, triggering unfolded protein response (UPR). U87 GBA1 mutant cells displayed an increased production of interleukin-1ß, both with and without inflammosome activation, α-syn accumulation and a higher rate of cell death in comparison with wild-type cells. In conclusion, we developed reliable, isogenic, and easy-to-handle cellular models of GD obtained from commercially accessible cells to be employed in GD pathophysiology studies and high-throughput drug screenings.


Asunto(s)
Sistemas CRISPR-Cas , Enfermedad de Gaucher/genética , Edición Génica , Modelos Biológicos , Biomarcadores , Línea Celular , Susceptibilidad a Enfermedades , Estrés del Retículo Endoplásmico , Degradación Asociada con el Retículo Endoplásmico , Enfermedad de Gaucher/metabolismo , Enfermedad de Gaucher/patología , Expresión Génica , Glucosilceramidasa/genética , Humanos , Mediadores de Inflamación/metabolismo , Monocitos/metabolismo , Mutación , Respuesta de Proteína Desplegada
20.
J Neurochem ; 150(5): 566-576, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31265132

RESUMEN

Misfolding and aggregation of alpha-synuclein (α-synuclein) with concomitant cytotoxicity is a hallmark of Lewy body related disorders such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Although it plays a pivotal role in pathogenesis and disease progression, the function of α-synuclein and the molecular mechanisms underlying α-synuclein-induced neurotoxicity in these diseases are still elusive. Many in vitro and in vivo experimental models mimicking α-synuclein pathology such as oligomerization, toxicity and more recently neuronal propagation have been generated over the years. In particular, cellular models have been crucial for our comprehension of the pathogenic process of the disease and are beneficial for screening of molecules capable of modulating α-synuclein toxicity. Here, we review α-synuclein based cell culture models that reproduce some features of the neuronal populations affected in patients, from basic unicellular organisms to mammalian cell lines and primary neurons, to the cutting edge models of patient-specific cell lines. These reprogrammed cells known as induced pluripotent stem cells (iPSCs) have garnered attention because they closely reproduce the characteristics of neurons found in patients and provide a valuable tool for mechanistic studies. We also discuss how different cell models may constitute powerful tools for high-throughput screening of molecules capable of modulating α-synuclein toxicity and prevention of its propagation. This article is part of the Special Issue "Synuclein".


Asunto(s)
Agregación Patológica de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Reprogramación Celular , Dopamina/metabolismo , Evaluación Preclínica de Medicamentos , Células HEK293 , Humanos , Técnicas In Vitro , Células Madre Pluripotentes Inducidas/metabolismo , Cuerpos de Lewy/metabolismo , Modelos Neurológicos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Pliegue de Proteína , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Sinucleinopatías/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA