Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mar Drugs ; 22(5)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38786617

RESUMEN

Utilizing plant-based resources, particularly their by-products, aligns with sustainability principles and circular bioeconomy, contributing to environmental preservation. The therapeutic potential of plant extracts is garnering increasing interest, and this study aimed to demonstrate promising outcomes from an extract obtained from an underutilized plant waste. Chaetomorpha linum, an invasive macroalga found in the Orbetello Lagoon, thrives in eutrophic conditions, forming persistent mats covering approximately 400 hectares since 2005. The biomass of C. linum undergoes mechanical harvesting and is treated as waste, requiring significant human efforts and economic resources-A critical concern for municipalities. Despite posing challenges to local ecosystems, the study identified C. linum as a natural source of bioactive metabolites. Phytochemical characterization revealed lipids, amino acids, and other compounds with potential anti-inflammatory activity in C. linum extract. In vitro assays with LPS-stimulated RAW 264.7 and TNF-α/IFN-γ-stimulated HaCaT cells showed the extract inhibited reactive oxygen species (ROS), nitric oxide (NO), and prostaglandin E2 (PGE2) productions, and reduced inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expressions via NF-κB nuclear translocation, in RAW 264.7 cells. It also reduced chemokines (TARC/CCL17, RANTES/CCL5, MCP-1/CCL2, and IL-8) and the cytokine IL-1ß production in HaCaT cells, suggesting potential as a therapeutic candidate for chronic diseases like atopic dermatitis. Finally, in silico studies indicated palmitic acid as a significant contributor to the observed effect. This research not only uncovered the untapped potential of C. linum but also laid the foundation for its integration into the circular bioeconomy, promoting sustainable practices, and innovative applications across various industries.


Asunto(s)
Antiinflamatorios , Fitoquímicos , Extractos Vegetales , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/química , Ratones , Células RAW 264.7 , Humanos , Fitoquímicos/farmacología , Fitoquímicos/química , Extractos Vegetales/farmacología , Extractos Vegetales/química , Células HaCaT , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ciclooxigenasa 2/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , FN-kappa B/metabolismo , Dinoprostona/metabolismo , Chlorophyta , Algas Marinas
2.
Int J Mol Sci ; 25(5)2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38473765

RESUMEN

Currently, many environmental and energy-related problems are threatening the future of our planet. In October 2022, the Worldmeter recorded the world population as 7.9 billion people, estimating that there will be an increase of 2 billion by 2057. The rapid growth of the population and the continuous increase in needs are causing worrying conditions, such as pollution, climate change, global warming, waste disposal, and natural resource reduction. Looking for novel and innovative methods to overcome these global troubles is a must for our common welfare. The circular bioeconomy represents a promising strategy to alleviate the current conditions using biomass-like natural wastes to replace commercial products that have a negative effect on our ecological footprint. Applying the circular bioeconomy concept, we propose an integrated in silico and in vitro approach to identify antioxidant bioactive compounds extracted from chestnut burrs (an agroforest waste) and their potential biological targets. Our study provides a novel and robust strategy developed within the circular bioeconomy concept aimed at target and drug discovery for a wide range of diseases. Our study could open new frontiers in the circular bioeconomy related to target and drug discovery, offering new ideas for sustainable scientific research aimed at identifying novel therapeutical strategies.


Asunto(s)
Antioxidantes , Cambio Climático , Humanos , Biomasa , Descubrimiento de Drogas , Contaminación Ambiental
3.
Brief Bioinform ; 22(5)2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-33538294

RESUMEN

Alkaptonuria (AKU, OMIM: 203500) is an autosomal recessive disorder caused by mutations in the Homogentisate 1,2-dioxygenase (HGD) gene. A lack of standardized data, information and methodologies to assess disease severity and progression represents a common complication in ultra-rare disorders like AKU. This is the reason why we developed a comprehensive tool, called ApreciseKUre, able to collect AKU patients deriving data, to analyse the complex network among genotypic and phenotypic information and to get new insight in such multi-systemic disease. By taking advantage of the dataset, containing the highest number of AKU patient ever considered, it is possible to apply more sophisticated computational methods (such as machine learning) to achieve a first AKU patient stratification based on phenotypic and genotypic data in a typical precision medicine perspective. Thanks to our sufficiently populated and organized dataset, it is possible, for the first time, to extensively explore the phenotype-genotype relationships unknown so far. This proof of principle study for rare diseases confirms the importance of a dedicated database, allowing data management and analysis and can be used to tailor treatments for every patient in a more effective way.


Asunto(s)
Alcaptonuria/genética , Bases de Datos Genéticas , Genotipo , Aprendizaje Automático , Selección de Paciente , Medicina de Precisión , Alcaptonuria/enzimología , Femenino , Homogentisato 1,2-Dioxigenasa/genética , Humanos , Masculino , Mutación , Enfermedades Raras
4.
Int J Mol Sci ; 24(6)2023 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-36982893

RESUMEN

Whenever a protein fails to fold into its native structure, a profound detrimental effect is likely to occur, and a disease is often developed. Protein conformational disorders arise when proteins adopt abnormal conformations due to a pathological gene variant that turns into gain/loss of function or improper localization/degradation. Pharmacological chaperones are small molecules restoring the correct folding of a protein suitable for treating conformational diseases. Small molecules like these bind poorly folded proteins similarly to physiological chaperones, bridging non-covalent interactions (hydrogen bonds, electrostatic interactions, and van der Waals contacts) loosened or lost due to mutations. Pharmacological chaperone development involves, among other things, structural biology investigation of the target protein and its misfolding and refolding. Such research can take advantage of computational methods at many stages. Here, we present an up-to-date review of the computational structural biology tools and approaches regarding protein stability evaluation, binding pocket discovery and druggability, drug repurposing, and virtual ligand screening. The tools are presented as organized in an ideal workflow oriented at pharmacological chaperones' rational design, also with the treatment of rare diseases in mind.


Asunto(s)
Chaperonas Moleculares , Pliegue de Proteína , Chaperonas Moleculares/metabolismo , Conformación Proteica , Biología , Biología Computacional
5.
Arch Biochem Biophys ; 717: 109137, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35090868

RESUMEN

Alkaptonuria (AKU) is an ultra-rare genetic disease caused by a deficient activity of the enzyme homogentisate 1,2-dioxygenase (HGD) leading to the accumulation of homogentisic acid (HGA) on connective tissues. Even though AKU is a multi-systemic disease, osteoarticular cartilage is the most affected system and the most damaged tissue by the disease. In chondrocytes, HGA causes oxidative stress dysfunctions, which induce a series of not fully characterized cellular responses. In this study, we used a human chondrocytic cell line as an AKU model to evaluate, for the first time, the effect of HGA on autophagy, the main homeostasis system in articular cartilage. Cells responded timely to HGA treatment with an increase in autophagy as a mechanism of protection. In a chronic state, HGA-induced oxidative stress decreased autophagy, and chondrocytes, unable to restore balance, activated the chondroptosis pathway. This decrease in autophagy also correlated with the accumulation of ochronotic pigment, a hallmark of AKU. Our data suggest new perspectives for understanding AKU and a mechanistic model that rationalizes the damaging role of HGA.


Asunto(s)
Alcaptonuria/prevención & control , Autofagia/efectos de los fármacos , Biomarcadores/metabolismo , Homogentisato 1,2-Dioxigenasa/metabolismo , Ácido Homogentísico/metabolismo , Alcaptonuria/metabolismo , Apoptosis/efectos de los fármacos , Cartílago Articular/efectos de los fármacos , Línea Celular , Condrocitos/citología , Ácido Homogentísico/farmacología , Humanos , Ocronosis/metabolismo , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal
6.
Int J Mol Sci ; 23(24)2022 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-36555443

RESUMEN

Alkaptonuria (AKU) is an ultra-rare metabolic disease caused by the accumulation of homogentisic acid (HGA), an intermediate product of phenylalanine and tyrosine degradation. AKU patients carry variants within the gene coding for homogentisate-1,2-dioxygenase (HGD), which are responsible for reducing the enzyme catalytic activity and the consequent accumulation of HGA and formation of a dark pigment called the ochronotic pigment. In individuals with alkaptonuria, ochronotic pigmentation of connective tissues occurs, leading to inflammation, degeneration, and eventually osteoarthritis. The molecular mechanisms underlying the multisystemic development of the disease severity are still not fully understood and are mostly limited to the metabolic pathway segment involving HGA. In this view, untargeted metabolomics of biofluids in metabolic diseases allows the direct investigation of molecular species involved in pathways alterations and their interplay. Here, we present the untargeted metabolomics study of AKU through the nuclear magnetic resonance of urine from a cohort of Italian patients; the study aims to unravel molecular species and mechanisms underlying the AKU metabolic disorder. Dysregulation of metabolic pathways other than the HGD route and new potential biomarkers beyond homogentisate are suggested, contributing to a more comprehensive molecular signature definition for AKU and the development of future adjuvant treatment.


Asunto(s)
Alcaptonuria , Dioxigenasas , Humanos , Alcaptonuria/genética , Metabolómica , Ácido Homogentísico/metabolismo , Biomarcadores , Espectroscopía de Resonancia Magnética
7.
J Cell Physiol ; 236(8): 6011-6024, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33469937

RESUMEN

Alkaptonuria (AKU) is an ultra-rare disease caused by the deficient activity of homogentisate 1,2-dioxygenase enzyme, leading the accumulation of homogentisic acid (HGA) in connective tissues implicating the formation of a black pigmentation called "ochronosis." Although AKU is a multisystemic disease, the most affected tissue is the articular cartilage, which during the pathology appears to be highly damaged. In this study, a model of alkaptonuric chondrocytes and cartilage was realized to investigate the role of HGA in the alteration of the extracellular matrix (ECM). The AKU tissues lost its architecture composed of collagen, proteoglycans, and all the proteins that characterize the ECM. The cause of this alteration in AKU cartilage is attributed to a degeneration of the cytoskeletal network in chondrocytes caused by the accumulation of HGA. The three cytoskeletal proteins, actin, vimentin, and tubulin, were analyzed and a modification in their amount and disposition in AKU chondrocytes model was identified. Cytoskeleton is involved in many fundamental cellular processes; therefore, the aberration in this complex network is involved in the manifestation of AKU disease.


Asunto(s)
Cartílago Articular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Ácido Homogentísico/farmacología , Actinas/efectos de los fármacos , Actinas/metabolismo , Alcaptonuria/metabolismo , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Humanos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Ocronosis/tratamiento farmacológico , Vimentina/efectos de los fármacos , Vimentina/metabolismo
8.
Expert Rev Proteomics ; 18(4): 315-327, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33861161

RESUMEN

Introduction: The term 'orphan diseases' includes conditions meeting prevalence-based or commercial viability criteria: they affect a small number of individuals and are considered an unviable market for drug development. Proteomics is an important technology to study them, providing information on mechanisms and evolution, biomarkers, and effects of therapeutic interventions.Areas covered: Herein, we review how proteomics and bioinformatic tools could be applied to the study of rare diseases and discuss pitfalls and potential.Expert opinion: Research in the field of rare diseases has to face many challenges, and implementation plans should foresee highly specialized collaborative consortia to create multidisciplinary frameworks for data sharing, advancing research, supporting clinical studies, and accelerating drug development. The integration of different technologies will allow better knowledge of disease pathophysiology, and the inclusion of proteomics and other omics technologies in this context will be pivotal to this aim.Several aspects of rare diseases, often perceived as limiting factors, might actually be advantages for a precision medicine approach: the limited number of patients, the collaboration with patient societies, and the availability of curated clinical registries could allow the development of homogeneous clinical databases and ultimately a better control over the data to be analyzed.


Asunto(s)
Proteómica , Enfermedades Raras , Biomarcadores , Biología Computacional , Humanos , Medicina de Precisión
9.
Anal Biochem ; 618: 114125, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33524411

RESUMEN

Protein Thiolation Index (PTI) has been recently proposed as a new biomarker of oxidative stress. It is calculated by measuring both free thiols and S-thiolated proteins in plasma with the assumption that this redox ratio is altered by a pro-oxidant stimulus. Here the original protocol was modified and adapted to the use of microvolumes of blood collected by finger prick and down to 3 µl blood was shown to be the lowest volume suitable for this kind of analysis. The new procedure was used to evaluate both the circadian rhythm and the annual fluctuations of PTI in healthy humans.


Asunto(s)
Proteínas Sanguíneas/química , Plasma/química , Compuestos de Sulfhidrilo/química , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad
10.
Mar Drugs ; 19(9)2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34564160

RESUMEN

Increasing frequency of native jellyfish proliferations and massive appearance of non-indigenous jellyfish species recently concur to impact Mediterranean coastal ecosystems and human activities at sea. Nonetheless, jellyfish biomass may represent an exploitable novel resource to coastal communities, with reference to its potential use in the pharmaceutical, nutritional, and nutraceutical Blue Growth sectors. The zooxanthellate jellyfish Cassiopea andromeda, Forsskål, 1775 (Cnidaria, Rhizostomeae) entered the Levant Sea through the Suez Canal and spread towards the Western Mediterranean to reach Malta, Tunisia, and recently also the Italian coasts. Here we report on the biochemical characterization and antioxidant activity of C. andromeda specimens with a discussion on their relative biological activities. The biochemical characterization of the aqueous (PBS) and hydroalcoholic (80% ethanol) soluble components of C. andromeda were performed for whole jellyfish, as well as separately for umbrella and oral arms. The insoluble components were hydrolyzed by sequential enzymatic digestion with pepsin and collagenase. The composition and antioxidant activity of the insoluble and enzymatically digestible fractions were not affected by the pre-extraction types, resulting into collagen- and non-collagen-derived peptides with antioxidant activity. Both soluble compounds and hydrolyzed fractions were characterized for the content of proteins, phenolic compounds, and lipids. The presence of compounds coming from the endosymbiont zooxanthellae was also detected. The notable yield and the considerable antioxidant activity detected make this species worthy of further study for its potential biotechnological sustainable exploitation.


Asunto(s)
Suplementos Dietéticos , Escifozoos , Animales , Antioxidantes , Organismos Acuáticos , Ecosistema , Mar Mediterráneo
11.
Int J Mol Sci ; 22(3)2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530326

RESUMEN

ApreciseKUre is a multi-purpose digital platform facilitating data collection, integration and analysis for patients affected by Alkaptonuria (AKU), an ultra-rare autosomal recessive genetic disease. It includes genetic, biochemical, histopathological, clinical, therapeutic resources and quality of life scores that can be shared among registered researchers and clinicians in order to create a Precision Medicine Ecosystem (PME). The combination of machine learning application to analyse and re-interpret data available in the ApreciseKUre shows the potential direct benefits to achieve patient stratification and the consequent tailoring of care and treatments to a specific subgroup of patients. In this study, we have developed a tool able to investigate the most suitable treatment for AKU patients in accordance with their Quality of Life scores, which indicates changes in health status before/after the assumption of a specific class of drugs. This fact highlights the necessity of development of patient databases for rare diseases, like ApreciseKUre. We believe this is not limited to the study of AKU, but it represents a proof of principle study that could be applied to other rare diseases, allowing data management, analysis, and interpretation.


Asunto(s)
Alcaptonuria/terapia , Aprendizaje Automático , Medicina de Precisión/métodos , Algoritmos , Alcaptonuria/diagnóstico , Alcaptonuria/etiología , Bases de Datos Factuales , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Humanos , Modelos Teóricos , Calidad de Vida
12.
Int J Mol Sci ; 22(22)2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34830297

RESUMEN

During angiogenesis, cell adhesion molecules expressed on the endothelial cell surface promote the growth and survival of newly forming vessels. Hence, elucidation of the signaling pathways activated by cell-to-matrix adhesion may assist in the discovery of new targets to be used in antiangiogenic therapy. In proliferating endothelial cells, the single-pass transmembrane glycoprotein CD93 has recently emerged as an important endothelial cell adhesion molecule regulating vascular maturation. In this study, we unveil a signaling pathway triggered by CD93 that regulates actin cytoskeletal dynamics responsible of endothelial cell adhesion. We show that the Src-dependent phosphorylation of CD93 and the adaptor protein Cbl leads to the recruitment of Crk, which works as a downstream integrator in the CD93-mediated signaling. Moreover, confocal microscopy analysis of FRET-based biosensors shows that CD93 drives the coordinated activation of Rac1 and RhoA at the cell edge of spreading cells, thus promoting the establishment of cell polarity and adhesion required for cell motility.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Complemento/metabolismo , Transducción de Señal/genética , Proteína de Unión al GTP rhoA/metabolismo , Adhesión Celular/genética , Moléculas de Adhesión Celular/genética , Polaridad Celular/genética , Células Cultivadas , Humanos , Glicoproteínas de Membrana/genética , Fosforilación/genética , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Proto-Oncogénicas c-crk/metabolismo , Interferencia de ARN , Receptores de Complemento/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Familia-src Quinasas/metabolismo
13.
J Cell Physiol ; 235(10): 6808-6816, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31989660

RESUMEN

Alkaptonuria (AKU) is a rare disease correlated with deficiency of the enzyme homogentisate 1,2 dioxygenase, which causes homogentisic acid (HGA) accumulation. HGA is subjected to oxidation/polymerization reactions, leading to the production of a peculiar melanin-like pigmentation (ochronosis) after chronic inflammation, which is considered as a triggering event for the generation of oxidative stress. Clinical manifestations of AKU are urine darkening, sclera pigmentation, early severe osteoarthropathy, and cardiovascular and renal complication. Despite major clinical manifestations of AKU being observed in the bones and skeleton, the molecular and functional parameters are so far unknown in AKU. In the present study, we used human osteoblasts supplemented with HGA as a AKU cellular model. We observed marked oxidative stress, and for the first time, we were able to correlate HGA deposition with an impairment in the Wnt/ß-catenin signaling pathway, opening a range of possible therapeutic strategies for a disease still lacking a known cure.


Asunto(s)
Ácido Homogentísico/farmacología , Osteoblastos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo , Alcaptonuria/metabolismo , Huesos/efectos de los fármacos , Huesos/metabolismo , Células Cultivadas , Humanos , Inflamación/metabolismo , Melaninas/metabolismo , Ocronosis/metabolismo , Osteoblastos/metabolismo , Oxidación-Reducción/efectos de los fármacos , Pigmentación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
14.
Arch Biochem Biophys ; 690: 108416, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32502471

RESUMEN

Alkaptonuria (AKU) is a rare metabolic disease correlated with the deficiency of homogentisate 1,2-dioxygenase and leading to an accumulation of the metabolite homogentisic acid (HGA) which can be subjected to oxidation and polymerization reactions. These events are considered a trigger for the induction of oxidative stress in AKU but, despite the large description of an altered redox status, the underlying pathogenetic processes are still unstudied. In the present study, we investigated the molecular mechanisms responsible for the oxidative damage present in an osteoblast-based cellular model of AKU. Bone, in fact, is largely affected in AKU patients: severe osteoclastic resorption, osteoporosis, even for pediatric cases, and an altered rate of remodeling biomarkers have been reported. In our AKU osteoblast cell model, we found a clear altered redox homeostasis, determined by elevated hydrogen peroxide (H2O2) levels and 4HNE protein adducts formation. These findings were correlated with increased NADPH oxidase (NOX) activity and altered mitochondrial respiration. In addition, we observed a decreased activity of superoxide dismutase (SOD) and reduced levels of thioredoxin (TRX) that parallel the decreased Nrf2-DNA binding. Overall, our results reveal that HGA is able to alter the cellular redox homeostasis by modulating the endogenous ROS production via NOX activation and mitochondrial dysfunctions and impair the cellular response mechanism. These findings can be useful for understanding the pathophysiology of AKU, not yet well studied in bones, but which is an important source of comorbidities that affect the life quality of the patients.


Asunto(s)
Alcaptonuria/metabolismo , Homeostasis/fisiología , Línea Celular , Proteínas de Unión al ADN/metabolismo , Ácido Homogentísico/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , NADPH Oxidasas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Osteoblastos/citología , Oxidación-Reducción , Estrés Oxidativo/fisiología , Transducción de Señal , Superóxido Dismutasa/metabolismo , Tiorredoxinas/metabolismo
15.
FASEB J ; 33(11): 12696-12703, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31462106

RESUMEN

Alkaptonuria (AKU) is an ultrarare autosomal recessive disorder (MIM 203500) that is caused byby a complex set of mutations in homogentisate 1,2-dioxygenasegene and consequent accumulation of homogentisic acid (HGA), causing a significant protein oxidation. A secondary form of amyloidosis was identified in AKU and related to high circulating serum amyloid A (SAA) levels, which are linked with inflammation and oxidative stress and might contribute to disease progression and patients' poor quality of life. Recently, we reported that inflammatory markers (SAA and chitotriosidase) and oxidative stress markers (protein thiolation index) might be disease activity markers in AKU. Thanks to an international network, we collected genotypic, phenotypic, and clinical data from more than 200 patients with AKU. These data are currently stored in our AKU database, named ApreciseKUre. In this work, we developed an algorithm able to make predictions about the oxidative status trend of each patient with AKU based on 55 predictors, namely circulating HGA, body mass index, total cholesterol, SAA, and chitotriosidase. Our general aim is to integrate the data of apparently heterogeneous patients with AKUAKU by using specific bioinformatics tools, in order to identify pivotal mechanisms involved in AKU for a preventive, predictive, and personalized medicine approach to AKU.-Cicaloni, V., Spiga, O., Dimitri, G. M., Maiocchi, R., Millucci, L., Giustarini, D., Bernardini, G., Bernini, A., Marzocchi, B., Braconi, D., Santucci, A. Interactive alkaptonuria database: investigating clinical data to improve patient care in a rare disease.


Asunto(s)
Alcaptonuria , Biología Computacional , Bases de Datos Genéticas , Medicina de Precisión , Enfermedades Raras , Alcaptonuria/metabolismo , Alcaptonuria/patología , Alcaptonuria/terapia , Femenino , Humanos , Masculino , Enfermedades Raras/metabolismo , Enfermedades Raras/patología , Enfermedades Raras/terapia
16.
J Inherit Metab Dis ; 43(4): 737-747, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31609457

RESUMEN

The clinical effects of alkaptonuria (AKU) are delayed and ageing influences disease progression. Morbidity of AKU is secondary to high circulating homogentisic acid (HGA) and ochronosis. It is not known whether HGA is produced by or processed in the kidney in AKU. Data from AKU patients from four studies were merged to form a single AKU group. A control group of non-AKU subjects was generated by merging data from two non-AKU studies. Data were used to derive renal clearance and fractional excretion (FE) ratios for creatinine, HGA, phenylalanine (PHE) and tyrosine (TYR) using standard calculations, for comparison between the AKU and the control groups. There were 225 AKU patients in the AKU group and 52 in the non-AKU control group. Circulating HGA increased with age (P < 0.001), and was significantly associated with decreased HGA clearance (CLHGA ) (P < 0.001) and FEHGA (P < 0.001). CLHGA and FEHGA were increased beyond the theoretical maximum renal plasma flow, confirming renal production and emphasising the greater contribution of net tubular secretion than glomerular filtration to renal elimination of HGA. The kidneys are crucial to elimination of HGA. Elimination of HGA is impaired with age resulting in worsening disease over time. The kidney is an important site for production of HGA. Tubular secretion of HGA contributes more to elimination of HGA in AKU than glomerular filtration.


Asunto(s)
Alcaptonuria/metabolismo , Tasa de Filtración Glomerular , Ácido Homogentísico/metabolismo , Riñón/metabolismo , Ocronosis/etiología , Adulto , Alcaptonuria/fisiopatología , Estudios de Casos y Controles , Creatinina/metabolismo , Femenino , Humanos , Modelos Lineales , Masculino , Persona de Mediana Edad , Ocronosis/fisiopatología , Fenilalanina/metabolismo , Factores Sexuales , Tirosina/metabolismo
17.
Int J Mol Sci ; 21(15)2020 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-32717819

RESUMEN

Osteosarcomas (OSs) are bone tumors most commonly found in pediatric and adolescent patients characterized by high risk of metastatic progression and recurrence after therapy. Effective therapeutic management of this disease still remains elusive as evidenced by poor patient survival rates. To achieve a more effective therapeutic management regimen, and hence patient survival, there is a need to identify more focused targeted therapies for OSs treatment in the clinical setting. The role of the OS tumor stroma microenvironment plays a significant part in the development and dissemination of this disease. Important components, and hence potential targets for treatment, are the tumor-infiltrating macrophages that are known to orchestrate many aspects of OS stromal signaling and disease progression. In particular, increased infiltration of M2-like tumor-associated macrophages (TAMs) has been associated with OS metastasis and poor patient prognosis despite currently used aggressive therapies regimens. This review aims to provide a summary update of current macrophage-centered knowledge and to discuss the possible roles that macrophages play in the process of OS metastasis development focusing on the potential influence of stromal cross-talk signaling between TAMs, cancer-stem cells and additional OSs tumoral microenvironment factors.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores , Neoplasias Óseas/inmunología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/terapia , Humanos , Metástasis de la Neoplasia , Osteosarcoma/inmunología , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/terapia , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/patología
18.
J Cell Physiol ; 234(5): 6696-6708, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30341892

RESUMEN

Alkaptonuria (AKU) is a disease caused by a deficient homogentisate 1,2-dioxygenase activity leading to systemic accumulation of homogentisic acid (HGA), that forms a melanin-like polymer that progressively deposits onto connective tissues causing a pigmentation called "ochronosis" and tissue degeneration. The effects of AKU and ochronotic pigment on the biomechanical properties of articular cartilage need further investigation. To this aim, AKU cartilage was studied using thermal (thermogravimetry and differential scanning calorimetry) and rheological analysis. We found that AKU cartilage had a doubled mesopore radius compared to healthy cartilage. Since the mesoporous structure is the main responsible for maintaining a correct hydrostatic pressure and tissue homoeostasis, drastic changes of thermal and rheological parameters were found in AKU. In particular, AKU tissue lost its capability to enhance chondrocytes metabolism (decreased heat capacity) and hence the production of proteoglycans. A drastic increase in stiffness and decrease in dissipative and lubricant role ensued in AKU cartilage. Multiphoton and scanning electron microscopies revealed destruction of cell-matrix microstructure and disruption of the superficial layer. Such observations on AKU specimens were confirmed in HGA-treated healthy cartilage, indicating that HGA is the toxic responsible of morphological and mechanical alterations of cartilage in AKU.


Asunto(s)
Alcaptonuria/tratamiento farmacológico , Condrocitos/efectos de los fármacos , Ácido Homogentísico/farmacología , Ocronosis/tratamiento farmacológico , Alcaptonuria/metabolismo , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Humanos , Oxidación-Reducción/efectos de los fármacos , Pigmentación/efectos de los fármacos
19.
Chem Res Toxicol ; 32(6): 1096-1102, 2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-30945548

RESUMEN

Glutathione (GSH) is one of the most well-studied biomarkers of oxidative stress. Under oxidizing conditions, GSH is transformed into its disulfide forms, glutathione disulfide (GSSG) and S-glutathionylated proteins (PSSG), which are considered to be reliable biomarkers of oxidative stress. In red blood cells (RBCs), the main targets of S-glutathionylation are hemoglobin and membrane-associated skeletal proteins, but S-glutathionylated hemoglobin (HbSSG) has been more thoroughly studied as a biomarker of oxidative stress than S-glutahionylated RBC membrane skeletal proteins. Here, we have investigated whether and how all these biomarkers are altered in human RBCs treated with a slow and cyclically intermittent flux of the oxidant tert-butyl hydroperoxide. To this aim, a new device for sample treatment and collection was developed. During and at the end of the treatment, GSH, GSSG, and PSSG (discriminating between HbSSG and membrane PSSG) were measured by the use of spectrophotometer (for GSSG) and HPLC (for GSH, HbSSG, and membrane PSSG). The main results of our study are as follows: (i) GSH decreased and GSSG increased, but only in the presence of the oxidant, and recovered their initial values at the end of the infusion; (ii) the increase in total PSSG concentration was lower than that of GSSG, but it kept on throughout the experiments; (iii) membrane skeletal proteins did not recover their initial values, whereas HbSSG levels recovered their initial values similarly to GSH and GSSG; (d) membrane skeletal PSSG were more stable and also more abundant than HbSSG. Western blot analysis indicated spectrin, ankyrin, and bands 3, 4.1, and 4.2 as the proteins most susceptible to S-glutathionylation in RBC membrane. These results suggest that S-glutathionylated membrane skeletal proteins can be considered as a suitable biomarker of oxidative stress. Mostly when the oxidant insult is slight and intermittent, PSSG in RBC membranes are worth measuring in addition to GSSG by virtue of their greater stability.


Asunto(s)
Eritrocitos/metabolismo , Glutatión/metabolismo , Proteínas de la Membrana/metabolismo , Estrés Oxidativo , Disulfuros/análisis , Eritrocitos/química , Glutatión/análisis , Humanos , Proteínas de la Membrana/análisis
20.
Cell Commun Signal ; 17(1): 55, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31138217

RESUMEN

BACKGROUND: In the endothelium, the single-pass membrane protein CD93, through its interaction with the extracellular matrix protein Multimerin-2, activates signaling pathways that are critical for vascular development and angiogenesis. Trafficking of adhesion molecules through endosomal compartments modulates their signaling output. However, the mechanistic basis coordinating CD93 recycling and its implications for endothelial cell (EC) function remain elusive. METHODS: Human umbilical vein ECs (HUVECs) and human dermal blood ECs (HDBEC) were used in this study. Fluorescence confocal microscopy was employed to follow CD93 retrieval, recycling, and protein colocalization in spreading cells. To better define CD93 trafficking, drug treatments and transfected chimeric wild type and mutant CD93 proteins were used. The scratch assay was used to evaluate cell migration. Gene silencing strategies, flow citometry, and quantification of migratory capability were used to determine the role of Rab5c during CD93 recycling to the cell surface. RESULTS: Here, we identify the recycling pathway of CD93 following EC adhesion and migration. We show that the cytoplasmic domain of CD93, by its interaction with Moesin and F-actin, is instrumental for CD93 retrieval in adhering and migrating cells and that aberrant endosomal trafficking of CD93 prevents its localization at the leading edge of migration. Moreover, the small GTPase Rab5c turns out to be a key component of the molecular machinery that is able to drive CD93 recycling to the EC surface. Finally, in the Rab5c endosomal compartment CD93 forms a complex with Multimerin-2 and active ß1 integrin, which is recycled back to the basolaterally-polarized cell surface by clathrin-independent endocytosis. CONCLUSIONS: Our findings, focusing on the pro-angiogenic receptor CD93, unveil the mechanisms of its polarized trafficking during EC adhesion and migration, opening novel therapeutic opportunities for angiogenic diseases.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Adhesión Celular , Movimiento Celular , Integrina beta1/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Complemento/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA