Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
medRxiv ; 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38496672

RESUMEN

The co-occurrence of insulin resistance (IR)-related metabolic conditions with neuropsychiatric disorders is a complex public health challenge. Evidence of the genetic links between these phenotypes is emerging, but little is currently known about the genomic regions and biological functions that are involved. To address this, we performed Local Analysis of [co]Variant Association (LAVA) using large-scale (N=9,725-933,970) genome-wide association studies (GWASs) results for three IR-related conditions (type 2 diabetes mellitus, obesity, and metabolic syndrome) and nine neuropsychiatric disorders. Subsequently, positional and expression quantitative trait locus (eQTL)-based gene mapping and downstream functional genomic analyses were performed on the significant loci. Patterns of negative and positive local genetic correlations (|rg|=0.21-1, pFDR<0.05) were identified at 109 unique genomic regions across all phenotype pairs. Local correlations emerged even in the absence of global genetic correlations between IR-related conditions and Alzheimer's disease, bipolar disorder, and Tourette's syndrome. Genes mapped to the correlated regions showed enrichment in biological pathways integral to immune-inflammatory function, vesicle trafficking, insulin signalling, oxygen transport, and lipid metabolism. Colocalisation analyses further prioritised 10 genetically correlated regions for likely harbouring shared causal variants, displaying high deleterious or regulatory potential. These variants were found within or in close proximity to genes, such as SLC39A8 and HLA-DRB1, that can be targeted by supplements and already known drugs, including omega-3/6 fatty acids, immunomodulatory, antihypertensive, and cholesterol-lowering drugs. Overall, our findings underscore the complex genetic landscape of IR-neuropsychiatric multimorbidity, advocating for an integrated disease model and offering novel insights for research and treatment strategies in this domain.

2.
Int J Biol Macromol ; 262(Pt 1): 129999, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38331080

RESUMEN

In recent years, chitosan (CS) has received much attention as a functional biopolymer for various applications, especially in the biomedical field. It is a natural polysaccharide created by the chemical deacetylation of chitin (CT) that is nontoxic, biocompatible, and biodegradable. This natural polymer is difficult to process; however, chemical modification of the CS backbone allows improved use of functional derivatives. CS and its derivatives are used to prepare hydrogels, membranes, scaffolds, fibers, foams, and sponges, primarily for regenerative medicine. Tissue engineering (TE), currently one of the fastest-growing fields in the life sciences, primarily aims to restore or replace lost or damaged organs and tissues using supports that, combined with cells and biomolecules, generate new tissue. In this sense, the growing interest in the application of biomaterials based on CS and some of its derivatives is justifiable. This review aims to summarize the most important recent advances in developing biomaterials based on CS and its derivatives and to study their synthesis, characterization, and applications in the biomedical field, especially in the TE area.


Asunto(s)
Quitosano , Quitosano/uso terapéutico , Quitosano/química , Ingeniería de Tejidos , Materiales Biocompatibles/uso terapéutico , Materiales Biocompatibles/química , Medicina Regenerativa , Andamios del Tejido
3.
J Inherit Metab Dis ; 47(3): 494-508, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38196161

RESUMEN

Proteostatic regulation of tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine biosynthesis, is crucial for maintaining proper brain neurotransmitter homeostasis. Variants of the TH gene are associated with tyrosine hydroxylase deficiency (THD), a rare disorder with a wide phenotypic spectrum and variable response to treatment, which affects protein stability and may lead to accelerated degradation, loss of TH function and catecholamine deficiency. In this study, we investigated the effects of the TH cofactor tetrahydrobiopterin (BH4) on the stability of TH in isolated protein and in DAn- differentiated from iPSCs from a human healthy subject, as well as from THD patients with the R233H variant in homozygosity (THDA) and R328W and T399M variants in heterozygosity (THDB). We report an increase in TH and dopamine levels, and an increase in the number of TH+ cells in control and THDA cells. To translate this in vitro effect, we treated with BH4 a knock-in THD mouse model with Th variant corresponding to R233H in patients. Importantly, treatment with BH4 significantly improved motor function in these mice, as demonstrated by increased latency on the rotarod test and improved horizontal activity (catalepsy). In conclusion, our study demonstrates the stabilizing effects of BH4 on TH protein levels and function in THD neurons and mice, rescuing disease phenotypes and improving motor outcomes. These findings highlight the therapeutic potential of BH4 as a treatment option for THDA patients with specific variants and provide insights into the modulation of TH stability and its implications for THD management.


Asunto(s)
Biopterinas , Modelos Animales de Enfermedad , Neuronas , Fenotipo , Tirosina 3-Monooxigenasa , Biopterinas/análogos & derivados , Animales , Humanos , Tirosina 3-Monooxigenasa/metabolismo , Ratones , Neuronas/metabolismo , Dopamina/metabolismo , Masculino , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/genética , Fenilcetonurias/metabolismo , Femenino , Técnicas de Sustitución del Gen
4.
J Inherit Metab Dis ; 47(3): 533-550, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38168036

RESUMEN

Several mouse models have been developed to study human defects of primary and secondary inherited monoamine neurotransmitter disorders (iMND). As the field continues to expand, current defects in corresponding mouse models include enzymes and a molecular co-chaperone involved in monoamine synthesis and metabolism (PAH, TH, PITX3, AADC, DBH, MAOA, DNAJC6), tetrahydrobiopterin (BH4) cofactor synthesis and recycling (adGTPCH1/DRD, arGTPCH1, PTPS, SR, DHPR), and vitamin B6 cofactor deficiency (ALDH7A1), as well as defective monoamine neurotransmitter packaging (VMAT1, VMAT2) and reuptake (DAT). No mouse models are available for human DNAJC12 co-chaperone and PNPO-B6 deficiencies, disorders associated with recessive variants that result in decreased stability and function of the aromatic amino acid hydroxylases and decreased neurotransmitter synthesis, respectively. More than one mutant mouse is available for some of these defects, which is invaluable as different variant-specific (knock-in) models may provide more insights into underlying mechanisms of disorders, while complete gene inactivation (knock-out) models often have limitations in terms of recapitulating complex human diseases. While these mouse models have common phenotypic traits also observed in patients, reflecting the defective homeostasis of the monoamine neurotransmitter pathways, they also present with disease-specific manifestations with toxic accumulation or deficiency of specific metabolites related to the specific gene affected. This review provides an overview of the currently available models and may give directions toward selecting existing models or generating new ones to investigate novel pathogenic mechanisms and precision therapies.


Asunto(s)
Modelos Animales de Enfermedad , Neurotransmisores , Animales , Ratones , Humanos , Neurotransmisores/metabolismo , Monoaminas Biogénicas/metabolismo
5.
FEBS J ; 291(3): 510-526, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37863644

RESUMEN

Hydroxymethylbilane synthase (HMBS), involved in haem biosynthesis, catalyses the head-to-tail coupling of four porphobilinogens (PBGs) via a dipyrromethane (DPM) cofactor. DPM is composed of two PBGs, and a hexapyrrole is built before the tetrapyrrolic 1-hydroxymethylbilane product is released. During this elongation, stable enzyme (E) intermediates are formed from the holoenzyme, with additional PBG substrates (S): ES, ES2 , ES3 and ES4 . Native PAGE and mass spectrometry of the acute intermittent porphyria (AIP)-associated HMBS variant p.Arg167Gln demonstrated an increased amount of ES3 . Kinetic parameters indicated catalytic dysfunction, however, the product release was not entirely prevented. Isolation and crystal structure analysis of the ES3 intermediate (PDB: 8PND) showed that a pentapyrrole was fully retained within the active site, revealing that polypyrrole elongation proceeds within the active site via a third interaction site, intermediate pyrrole site 3 (IPS3). The AIP-associated HMBS variant p.Arg195Cys, located on the opposite side to p.Arg167Gln in the active site, accumulated the ES4 intermediate in the presence of excess PBG, implying that product hydrolysis was obstructed. Arg167 is thus involved in all elongation steps and is a determinant for the rate of enzyme catalysis, whereas Arg195 is important for releasing the product. Moreover, by substituting residues in the vicinity of IPS3, our results indicate that a fully retained hexapyrrole could be hydrolysed in a novel site in proximity of the IPS3.


Asunto(s)
Hidroximetilbilano Sintasa , Porfiria Intermitente Aguda , Humanos , Hidroximetilbilano Sintasa/química , Hidroximetilbilano Sintasa/genética , Hidroximetilbilano Sintasa/metabolismo , Polímeros , Pirroles , Dominio Catalítico , Mutación
6.
Nat Prod Res ; : 1-10, 2023 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-37548293

RESUMEN

The investigation of natural alternatives to conventional fungicides is of imminent need. Mosiera bullata (Britton & P. Wilson) Bisse is a Cuban endemic plant species belonging to the Myrtaceae family. The objective of the present study was to perform a bioassay-guided fractionation to explore the potential of extracts and fractions from M. bullata leaves against a panel of fungal plant pathogens. The M. bullata total extract was confirmed to have good antifungal activity against R. oryzae (IC50 = 4.86 µg/mL) and moderate activity against F. oxysporum (IC50 = 352.40 µg/mL) and F. solani (IC50 = 427.38 µg/mL) and fungicidal effect against R. oryzae. Five compounds belonging to the class of phloroglucinol dimers were tentatively characterized by UHPLC-HRMS and reported for the first time in M. bullata and the genus Mosiera. These results suggest the potential of M. bullata total extract as a natural antifungal product for the control of diseases in agriculture.

7.
Neurosci Biobehav Rev ; 152: 105301, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37414376

RESUMEN

Potassium channels (K+-channels) selectively control the passive flow of potassium ions across biological membranes and thereby also regulate membrane excitability. Genetic variants affecting many of the human K+-channels are well known causes of Mendelian disorders within cardiology, neurology, and endocrinology. K+-channels are also primary targets of many natural toxins from poisonous organisms and drugs used within cardiology and metabolism. As genetic tools are improving and larger clinical samples are being investigated, the spectrum of clinical phenotypes implicated in K+-channels dysfunction is rapidly expanding, notably within immunology, neurosciences, and metabolism. K+-channels that previously were considered to be expressed in only a few organs and to have discrete physiological functions, have recently been found in multiple tissues and with new, unexpected functions. The pleiotropic functions and patterns of expression of K+-channels may provide additional therapeutic opportunities, along with new emerging challenges from off-target effects. Here we review the functions and therapeutic potential of K+-channels, with an emphasis on the nervous system, roles in neuropsychiatric disorders and their involvement in other organ systems and diseases.


Asunto(s)
Encefalopatías , Canales de Potasio , Humanos , Canales de Potasio/genética , Canales de Potasio/metabolismo , Encefalopatías/tratamiento farmacológico , Potasio/metabolismo
9.
Commun Biol ; 5(1): 1283, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36418492

RESUMEN

Vesicular monoamine transporter 2 (VMAT2) is responsible for packing monoamine neurotransmitters into synaptic vesicles for storage and subsequent neurotransmission. VMAT2 inhibitors are approved for symptomatic treatment of tardive dyskinesia and Huntington's chorea, but despite being much-studied inhibitors their exact binding site and mechanism behind binding and inhibition of monoamine transport are not known. Here we report the identification of several approved drugs, notably ß2-adrenergic agonists salmeterol, vilanterol and formoterol, ß2-adrenergic antagonist carvedilol and the atypical antipsychotic ziprasidone as inhibitors of rat VMAT2. Further, plausible binding modes of the established VMAT2 inhibitors reserpine and tetrabenazine and hit compounds salmeterol and ziprasidone were identified using molecular dynamics simulations and functional assays using VMAT2 wild-type and mutants. Our findings show VMAT2 as a potential off-target of treatments with several approved drugs in use today and can also provide important first steps in both drug repurposing and therapy development targeting VMAT2 function.


Asunto(s)
Antipsicóticos , Animales , Ratas , Agonistas Adrenérgicos , Antipsicóticos/farmacología , Piperazinas , Proteínas de Transporte Vesicular de Monoaminas/genética , Antagonistas Adrenérgicos/farmacología
10.
J Refract Surg ; 38(11): 698-707, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36367259

RESUMEN

PURPOSE: To compare two aspheric ablation profiles in myopic refractive surgery using different asphericity targets. METHODS: Patients underwent laser in situ keratomileusis (LASIK) with the WaveLight EX500 laser platform (Alcon, WaveLight Laser Technologie). Asymmetric surgery was performed, programming the wavefront-optimized (WFO) ablation profile in one eye and the custom-Q (CQ) profile in the contralateral eye. The patients were divided into two groups following a systematic randomization method. The Q-target programmed for the preoperative Q group was equal to the preoperative asphericity of the CQ profile, and for the -0.6 Q-target group, the Q-target was set to -0.6. RESULTS: The study included 100 patients (200 eyes). Both groups had comparable safety and efficacy indexes greater than 1. A similar oblate shift in postoperative asphericity was seen in both groups regardless of the ablation profile and programmed Q-target. Asphericity was 0.33 ± 0.34 and 0.35 ± 0.29 (P = .18) in the preoperative Q group and 0.26 ± 0.28 and 0.26 ± 0.27 (P = .89) in the -0.6 Q-target group for WFO and CQ, respectively. A lower spherical aberration was found with CQ compared to WFO when the Q-target was set to -0.6: 0.211 ± 0.121 versus 0.144 ± 0.114 (P < .01). However, no statistically significant differences were found when the preoperative Q-target was used. CONCLUSIONS: WFO and CQ treatments are similar in terms of refractive and visual outcomes. CQ offers greater control over the increase in positive spherical aberration after myopic refractive surgery, but it does not represent an advantage over WFO in the oblate shift in postoperative asphericity regardless of the Q-target programmed. [J Refract Surg. 2022;38(11):698-707.].


Asunto(s)
Queratomileusis por Láser In Situ , Miopía , Humanos , Láseres de Excímeros/uso terapéutico , Agudeza Visual , Miopía/cirugía , Queratomileusis por Láser In Situ/métodos , Refracción Ocular , Resultado del Tratamiento
11.
J Mol Biol ; 434(11): 167372, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35662461

RESUMEN

The identification of new drugs for novel therapeutic targets requires the screening of libraries containing tens of thousands of compounds. While experimental screenings are assisted by high-throughput technologies, in target-based biophysical assays, such as differential scanning fluorimetry (DSF), the analysis steps must be calculated manually, often combining several software packages. To simplify the determination of the melting temperature (Tm) of the target and the change induced by ligand binding (ΔTm), we developed the HTSDSF explorer, a versatile, all-in-one, user-friendly application suite. Implemented as a server-client application, in the primary screenings, HTSDSF explorer pre-analyzes and displays the Tm and ΔTm results interactively, thereby allowing the user to study hundreds of conditions and select the primary hits in minutes. This application also allows the determination of preliminary binding constants (KD) through a series of subsequent dose-response assays on the primary hits, thereby facilitating the ranking of validated hits and the advance of drug discovery efforts.


Asunto(s)
Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Uso de Internet , Relación Dosis-Respuesta a Droga , Fluorometría/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos
12.
J Leukoc Biol ; 112(1): 31-45, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35674096

RESUMEN

Leukemogenesis is proposed to result from the continuous interplay between inducive bone marrow (BM) microenvironments and malignant precursor cells. Recent findings point toward an abnormal production of proinflammatory mediators within the BM from acute lymphoblastic leukemia (ALL) patients, although the mechanism underlying this phenomenon is uncertain. Here, we have identified 3 miRNAs, miR-146a-5p, miR-181b-5p, and miR-199b-3p, as potential candidates for TLR8 ligation, which are overexpressed in ALL and show agonist functional binding. When purified from ALL exosomes, they demonstrated their capacity of inducing cytokine production by both, hematopoietic and stromal BM cells. Of note, the exposure of BM cells from ALL patients to the proinflammatory milieu resulting from these miRNAs agonist activity revealed the proliferation of normal progenitors, while poor effects were recorded in the leukemic counterpart. The unconventional roles of the tumor-secreted miRNAs as TLR8 agonist ligands may provide a novel mechanism contributing a tumor-microenvironment feedback loop by switching on proinflammatory pathways that further activate normal hematopoietic precursors and support ALL progression. Secreted B-ALL TLR8-agonist miRNAs are involved in the promotion of proinflammatory microenvironments that target normal hematopoietic cells. B-lineage ALL cells secrete exosomes containing miRNAs endowed with the ability of functionally binding TLR8 in hematopoietic and BM mesenchymal stromal cells. Upon TLR8 signaling, the activation of the NF-kB pathway induces secretion of proinflammatory cytokines that, in turn, promotes cell proliferation in early hematopoietic cell populations, driving a tumor-microenvironment-hematopoietic activation feedback loop that may reduce the normal hematopoietic stem and progenitor cell compartment and facilitate cancer progression.


Asunto(s)
MicroARNs , Leucemia-Linfoma Linfoblástico de Células Precursoras , Médula Ósea/patología , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Receptor Toll-Like 8/metabolismo , Microambiente Tumoral
13.
PLoS One ; 17(6): e0269281, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35671319

RESUMEN

Activity-regulated cytoskeleton-associated protein (Arc) is a multidomain protein of retroviral origin with a vital role in the regulation of synaptic plasticity and memory formation in mammals. However, the mechanistic and structural basis of Arc function is poorly understood. Arc has an N-terminal domain (NTD) involved in membrane binding and a C-terminal domain (CTD) that binds postsynaptic protein ligands. In addition, the NTD and CTD both function in Arc oligomerisation, including assembly of retrovirus-like capsids involved in intercellular signalling. To obtain new tools for studies on Arc structure and function, we produced and characterised six high-affinity anti-Arc nanobodies (Nb). The CTD of rat and human Arc were both crystallised in ternary complexes with two Nbs. One Nb bound deep into the stargazin-binding pocket of Arc CTD and suggested competitive binding with Arc ligand peptides. The crystallisation of the human Arc CTD in two different conformations, accompanied by SAXS data and molecular dynamics simulations, paints a dynamic picture of the mammalian Arc CTD. The collapsed conformation closely resembles Drosophila Arc in capsids, suggesting that we have trapped a capsid-like conformation of the human Arc CTD. Our data obtained with the help of anti-Arc Nbs suggest that structural dynamics of the CTD and dimerisation of the NTD may promote the formation of capsids. Taken together, the recombinant high-affinity anti-Arc Nbs are versatile tools that can be further developed for studying mammalian Arc structure and function, as well as mechanisms of Arc capsid formation, both in vitro and in vivo. For example, the Nbs could serve as a genetically encoded tools for inhibition of endogenous Arc interactions in the study of neuronal function and plasticity.


Asunto(s)
Anticuerpos de Dominio Único , Animales , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Drosophila/metabolismo , Mamíferos/metabolismo , Ratas , Dispersión del Ángulo Pequeño , Anticuerpos de Dominio Único/metabolismo , Difracción de Rayos X
14.
Nat Commun ; 13(1): 74, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013193

RESUMEN

Tyrosine hydroxylase (TH) catalyzes the rate-limiting step in the biosynthesis of dopamine (DA) and other catecholamines, and its dysfunction leads to DA deficiency and parkinsonisms. Inhibition by catecholamines and reactivation by S40 phosphorylation are key regulatory mechanisms of TH activity and conformational stability. We used Cryo-EM to determine the structures of full-length human TH without and with DA, and the structure of S40 phosphorylated TH, complemented with biophysical and biochemical characterizations and molecular dynamics simulations. TH presents a tetrameric structure with dimerized regulatory domains that are separated 15 Å from the catalytic domains. Upon DA binding, a 20-residue α-helix in the flexible N-terminal tail of the regulatory domain is fixed in the active site, blocking it, while S40-phosphorylation forces its egress. The structures reveal the molecular basis of the inhibitory and stabilizing effects of DA and its counteraction by S40-phosphorylation, key regulatory mechanisms for homeostasis of DA and TH.


Asunto(s)
Dopamina/farmacología , Inhibidores Enzimáticos/farmacología , Tirosina 3-Monooxigenasa/antagonistas & inhibidores , Tirosina 3-Monooxigenasa/química , Secuencia de Aminoácidos , Dominio Catalítico , Catecolaminas/metabolismo , Microscopía por Crioelectrón , Dopamina/química , Dopamina/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Modelos Moleculares , Fosforilación , Unión Proteica , Dominios Proteicos , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
15.
J Pers Med ; 11(11)2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34834538

RESUMEN

Dopa-responsive dystonia (DRD) is a rare movement disorder associated with defective dopamine synthesis. This impairment may be due to the fact of a deficiency in GTP cyclohydrolase I (GTPCHI, GCH1 gene), sepiapterin reductase (SR), tyrosine hydroxylase (TH), or 6-pyruvoyl tetrahydrobiopterin synthase (PTPS) enzyme functions. Mutations in GCH1 are most frequent, whereas fewer cases have been reported for individual SR-, PTP synthase-, and TH deficiencies. Although termed DRD, a subset of patients responds poorly to L-DOPA. As this is regularly observed in severe cases of TH deficiency (THD), there is an urgent demand for more adequate or personalized treatment options. TH is a key enzyme that catalyzes the rate-limiting step in catecholamine biosynthesis, and THD patients often present with complex and variable phenotypes, which results in frequent misdiagnosis and lack of appropriate treatment. In this expert opinion review, we focus on THD pathophysiology and ongoing efforts to develop novel therapeutics for this rare disorder. We also describe how different modeling approaches can be used to improve genotype to phenotype predictions and to develop in silico testing of treatment strategies. We further discuss the current status of mathematical modeling of catecholamine synthesis and how such models can be used together with biochemical data to improve treatment of DRD patients.

16.
Front Immunol ; 12: 746492, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34737747

RESUMEN

B-cell acute lymphoblastic leukemia (B-ALL) results from the expansion of malignant lymphoid precursors within the bone marrow (BM), where hematopoietic niches and microenvironmental signals provide leukemia-initiating cells (LICs) the conditions to survive, proliferate, initiate disease, and relapse. Normal and malignant lymphopoiesis are highly dependent on the BM microenvironment, particularly on CXCL12-abundant Reticular (CAR) cells, which provide a niche for maintenance of primitive cells. During B-ALL, leukemic cells hijack BM niches, creating a proinflammatory milieu incompetent to support normal hematopoiesis but favoring leukemic proliferation. Although the lack of a phenotypic stem cell hierarchy is apparent in B-ALL, LICs are a rare and quiescent population potentially responsible for chemoresistance and relapse. Here, we developed novel patient-derived leukemia spheroids (PDLS), an ex vivo avatar model, from mesenchymal stromal cells (MSCs) and primary B-ALL cells, to mimic specialized niche structures and cell-to-cell intercommunication promoting normal and malignant hematopoiesis in pediatric B-ALL. 3D MSC spheroids can recapitulate CAR niche-like hypoxic structures that produce high levels of CXCL10 and CXCL11. We found that PDLS were preferentially enriched with leukemia cells displaying functional properties of LICs, such as quiescence, low reactive oxygen species, drug resistance, high engraftment in immunodeficient mice, and long-term leukemogenesis. Moreover, the combination of PDLS and patient-derived xenografts confirmed a microenvironment-driven hierarchy in their leukemic potential. Importantly, transcriptional profiles of MSC derived from primary patient samples revealed two unique signatures (1), a CXCL12low inflammatory and leukemia expansion (ILE)-like niche, that likely supports leukemic burden, and (2) a CXCL11hi immune-suppressive and leukemia-initiating cell (SLIC)-like niche, where LICs are likely sustained. Interestingly, the CXCL11+ hypoxic zones were recapitulated within the PDLS that are capable of supporting LIC functions. Taken together, we have implemented a novel PDLS system that enriches and supports leukemia cells with stem cell features driven by CXCL11+ MSCs within hypoxic microenvironments capable of recapitulating key features, such as tumor reemergence after exposure to chemotherapy and tumor initiation. This system represents a unique opportunity for designing ex vivo personalized avatars for B-ALL patients to evaluate their own LIC pathobiology and drug sensitivity in the context of the tumor microenvironment.


Asunto(s)
Células Madre Neoplásicas/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Esferoides Celulares , Nicho de Células Madre , Células Tumorales Cultivadas , Animales , Médula Ósea/patología , Femenino , Xenoinjertos , Humanos , Células Madre Mesenquimatosas/patología , Ratones , Microambiente Tumoral
17.
Cancers (Basel) ; 13(12)2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-34208232

RESUMEN

Heat shock protein (Hsp) synthesis is upregulated in a wide range of cancers to provide the appropriate environment for tumor progression. The Hsp110 and Hsp70 families have been associated to cancer cell survival and resistance to chemotherapy. In this study, we explore the strategy of drug repurposing to find new Hsp70 and Hsp110 inhibitors that display toxicity against melanoma cancer cells. We found that the hits discovered using Apg2, a human representative of the Hsp110 family, as the initial target bind also to structural regions present in members of the Hsp70 family, and therefore inhibit the remodeling activity of the Hsp70 system. One of these compounds, the spasmolytic agent pinaverium bromide used for functional gastrointestinal disorders, inhibits the intracellular chaperone activity of the Hsp70 system and elicits its cytotoxic activity specifically in two melanoma cell lines by activating apoptosis. Docking and molecular dynamics simulations indicate that this compound interacts with regions located in the nucleotide-binding domain and the linker of the chaperones, modulating their ATPase activity. Thus, repurposing of pinaverium bromide for cancer treatment appears as a promising novel therapeutic approach.

18.
Am J Pathol ; 191(9): 1537-1549, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34139193

RESUMEN

Epithelial barrier impairment is a hallmark of several pathologic processes in the gut, including inflammatory bowel diseases. Several intracellular signals prevent apoptosis in intestinal epithelial cells. Herein, we show that in colonocytes, rictor/mammalian target of rapamycin complex 2 (mTORC2) signaling is a prosurvival stimulus. Mechanistically, mTORC2 activates Akt, which, in turn, inhibits apoptosis by phosphorylating B-cell lymphoma 2 (BCL2) associated agonist of cell death (Bad) and preventing caspase-3 activation. Nevertheless, during inflammation, rictor/mTORC2 signaling declines and Akt activity is reduced. Consequently, active caspase-3 increases in surface colonocytes undergoing apoptosis/anoikis and causes epithelial barrier breakdown. Likewise, Rictor ablation in intestinal epithelial cells interrupts mTORC2/Akt signaling and increases apoptosis/anoikis of surface colonocytes without affecting the crypt architecture. The increase in epithelial permeability induced by Rictor ablation produces a mild inflammatory response in the colonic mucosa, but minimally affects the development/establishment of colitis. The data identify a previously unknown mechanism by which rictor/mTORC2 signaling regulates apoptosis/anoikis in intestinal epithelial cells during colitis and clarify its role in the maintenance of the intestinal epithelial barrier.


Asunto(s)
Apoptosis/fisiología , Colitis/patología , Células Epiteliales/metabolismo , Mucosa Intestinal/patología , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Animales , Colitis/metabolismo , Células Epiteliales/patología , Mucosa Intestinal/metabolismo , Ratones , Transducción de Señal/fisiología
19.
J Vis Exp ; (172)2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34180884

RESUMEN

Epithelial cells lining the intestinal mucosa create a physical barrier that separates the luminal content from the interstitium. Epithelial barrier impairment has been associated with the development of various pathologies such as inflammatory bowel diseases (IBD). In the inflamed mucosa, superficial erosions or micro-erosions that corrupt epithelial monolayers correspond to sites of high permeability. Several mechanisms have been implicated in the formation of micro-erosions including cell shedding and apoptosis. These micro-erosions often represent microscopic epithelial gaps randomly distributed in the colon. Visualization and quantification of those epithelial gaps has emerged as an important tool to investigate intestinal epithelial barrier function. Here, we describe a new method to visualize the specific location of where transcellular and paracellular permeability is enhanced in the inflamed colonic mucosa. In this assay, we apply a 10 kDa fluorescent dye conjugated to a lysine fixable dextran to visualize high permeability regions (HPR) in the colonic mucosa. Additional use of cell death markers revealed that HPR encompass apoptotic foci where epithelial extrusion/shedding occurs. The protocol described here provides a simple but effective approach to visualize and quantify micro-erosions in the intestine, which is a very useful tool in disease models, in which the intestinal epithelial barrier is compromised.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Mucosa Intestinal , Colon , Células Epiteliales , Técnica del Anticuerpo Fluorescente , Humanos , Permeabilidad , Coloración y Etiquetado
20.
Mol Pharmacol ; 100(2): 155-169, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34031189

RESUMEN

The 14-3-3 proteins constitute a family of adaptor proteins with many binding partners and biological functions, and they are considered promising drug targets in cancer and neuropsychiatry. By screening 1280 small-molecule drugs using differential scanning fluorimetry (DSF), we found 15 compounds that decreased the thermal stability of 14-3-3ζ Among these compounds, ebselen was identified as a covalent, destabilizing ligand of 14-3-3 isoforms ζ, ε, γ, and η Ebselen bonding decreased 14-3-3ζ binding to its partner Ser19-phosphorylated tyrosine hydroxylase. Characterization of site-directed mutants at cysteine residues in 14-3-3ζ (C25, C94, and C189) by DSF and mass spectroscopy revealed covalent modification by ebselen of all cysteines through a selenylsulfide bond. C25 appeared to be the preferential site of ebselen interaction in vitro, whereas modification of C94 was the main determinant for protein destabilization. At therapeutically relevant concentrations, ebselen and ebselen oxide caused decreased 14-3-3 levels in SH-SY5Y cells, accompanied with an increased degradation, most probably by the ubiquitin-dependent proteasome pathway. Moreover, ebselen-treated zebrafish displayed decreased brain 14-3-3 content, a freezing phenotype, and reduced mobility, resembling the effects of lithium, consistent with its proposed action as a safer lithium-mimetic drug. Ebselen has recently emerged as a promising drug candidate in several medical areas, such as cancer, neuropsychiatric disorders, and infectious diseases, including coronavirus disease 2019. Its pleiotropic actions are attributed to antioxidant effects and formation of selenosulfides with critical cysteine residues in proteins. Our work indicates that a destabilization of 14-3-3 may affect the protein interaction networks of this protein family, contributing to the therapeutic potential of ebselen. SIGNIFICANCE STATEMENT: There is currently great interest in the repurposing of established drugs for new indications and therapeutic targets. This study shows that ebselen, which is a promising drug candidate against cancer, bipolar disorder, and the viral infection coronavirus disease 2019, covalently bonds to cysteine residues in 14-3-3 adaptor proteins, triggering destabilization and increased degradation in cells and intact brain tissue when used in therapeutic concentrations, potentially explaining the behavioral, anti-inflammatory, and antineoplastic effects of this drug.


Asunto(s)
Proteínas 14-3-3/química , Proteínas 14-3-3/metabolismo , Cisteína/genética , Isoindoles/farmacología , Compuestos de Organoselenio/farmacología , Proteínas 14-3-3/genética , Animales , Sitios de Unión/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Dicroismo Circular , Regulación hacia Abajo , Femenino , Humanos , Masculino , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica/efectos de los fármacos , Conformación Proteica , Estabilidad Proteica/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo , Pez Cebra , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...