SwePub
Tyck till om SwePub Sök här!
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Ruiz Rocío) "

Sökning: WFRF:(Ruiz Rocío)

  • Resultat 1-10 av 12
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Cruz, Raquel, et al. (författare)
  • Novel genes and sex differences in COVID-19 severity
  • 2022
  • Ingår i: Human Molecular Genetics. - : Oxford University Press. - 0964-6906 .- 1460-2083. ; 31:22, s. 3789-3806
  • Tidskriftsartikel (refereegranskat)abstract
    • Here, we describe the results of a genome-wide study conducted in 11 939 coronavirus disease 2019 (COVID-19) positive cases with an extensive clinical information that were recruited from 34 hospitals across Spain (SCOURGE consortium). In sex-disaggregated genome-wide association studies for COVID-19 hospitalization, genome-wide significance (P < 5 × 10−8) was crossed for variants in 3p21.31 and 21q22.11 loci only among males (P = 1.3 × 10−22 and P = 8.1 × 10−12, respectively), and for variants in 9q21.32 near TLE1 only among females (P = 4.4 × 10−8). In a second phase, results were combined with an independent Spanish cohort (1598 COVID-19 cases and 1068 population controls), revealing in the overall analysis two novel risk loci in 9p13.3 and 19q13.12, with fine-mapping prioritized variants functionally associated with AQP3 (P = 2.7 × 10−8) and ARHGAP33 (P = 1.3 × 10−8), respectively. The meta-analysis of both phases with four European studies stratified by sex from the Host Genetics Initiative (HGI) confirmed the association of the 3p21.31 and 21q22.11 loci predominantly in males and replicated a recently reported variant in 11p13 (ELF5, P = 4.1 × 10−8). Six of the COVID-19 HGI discovered loci were replicated and an HGI-based genetic risk score predicted the severity strata in SCOURGE. We also found more SNP-heritability and larger heritability differences by age (<60 or ≥60 years) among males than among females. Parallel genome-wide screening of inbreeding depression in SCOURGE also showed an effect of homozygosity in COVID-19 hospitalization and severity and this effect was stronger among older males. In summary, new candidate genes for COVID-19 severity and evidence supporting genetic disparities among sexes are provided.
  •  
2.
  • Aleix-Mata, Gael, et al. (författare)
  • Development and characterization of 15 novel polymorphic microsatellite loci for two important bot flies (Diptera, Oestridae) by next-generation sequencing
  • 2020
  • Ingår i: Parasitology Research. - : Springer Nature. - 0932-0113 .- 1432-1955. ; 119:9, s. 2829-2835
  • Tidskriftsartikel (refereegranskat)abstract
    • Cephenemyia stimulatorandOestrus ovisare two important parasitic bot flies (Oestridae) species causing myiasis, with a potential negative impact on the welfare of the host. Using next-generation sequencing approach and bioinformatics tools, a large panel of possible microsatellites loci was obtained in both species. Primer pairs were designed for 15 selected microsatellite loci inC. stimulatorand other 15 loci inO. ovisfor PCR amplification. Loci amplification and analysis were performed in four populations of each species. The results demonstrated that all selected loci were polymorphic, with the number of alleles ranging from 2 to 6 per locus inC. stimulatorand 3 to 13 per locus inO. ovis. This is the first time to describe these microsatellite loci forC. stimulatorandO. ovis. These two sets of microsatellite markers could be further used for biogeographic and population genetics studies.
  •  
3.
  • Alonso-Bellido, Isabel M., et al. (författare)
  • The Other Side of SARS-CoV-2 Infection : Neurological Sequelae in Patients
  • 2021
  • Ingår i: Frontiers in Aging Neuroscience. - : Frontiers Media SA. - 1663-4365. ; 13
  • Forskningsöversikt (refereegranskat)abstract
    • The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread around the globe causing coronavirus disease 2019 (COVID-19). Because it affects the respiratory system, common symptoms are cough and breathing difficulties with fever and fatigue. Also, some cases progress to acute respiratory distress syndrome (ARDS). The acute phase of COVID-19 has been also related to nervous system symptoms, including loss of taste and smell as well as encephalitis and cerebrovascular disorders. However, it remains unclear if neurological complications are due to the direct viral infection of the nervous system, or they appear as a consequence of the immune reaction against the virus in patients who presented pre-existing deficits or had a certain detrimental immune response. Importantly, the medium and long-term consequences of the infection by SARS-CoV-2 in the nervous system remain at present unknown. This review article aims to give an overview of the current neurological symptoms associated with COVID-19, as well as attempting to provide an insight beyond the acute affectation.
  •  
4.
  • Espinosa-Oliva, Ana M., et al. (författare)
  • Inflammatory bowel disease induces pathological α-synuclein aggregation in the human gut and brain
  • 2024
  • Ingår i: Neuropathology and Applied Neurobiology. - 0305-1846. ; 50:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Aims: According to Braak's hypothesis, it is plausible that Parkinson's disease (PD) originates in the enteric nervous system (ENS) and spreads to the brain through the vagus nerve. In this work, we studied whether inflammatory bowel diseases (IBDs) in humans can progress with the emergence of pathogenic α-synuclein (α-syn) in the gastrointestinal tract and midbrain dopaminergic neurons. Methods: We have analysed the gut and the ventral midbrain from subjects previously diagnosed with IBD and form a DSS-based rat model of gut inflammation in terms of α-syn pathology. Results: Our data support the existence of pathogenic α-syn in both the gut and the brain, thus reinforcing the potential role of the ENS as a contributing factor in PD aetiology. Additionally, we have analysed the effect of a DSS-based rat model of gut inflammation to demonstrate (i) the appearance of P-α-syn inclusions in both Auerbach's and Meissner's plexuses (gut), (ii) an increase in α-syn expression in the ventral mesencephalon (brain) and (iii) the degeneration of nigral dopaminergic neurons, which all are considered classical hallmarks in PD. Conclusion: These results strongly support the plausibility of Braak's hypothesis and emphasise the significance of peripheral inflammation and the gut-brain axis in initiating α-syn aggregation and transport to the substantia nigra, resulting in neurodegeneration.
  •  
5.
  • García-Revilla, Juan, et al. (författare)
  • Galectin-3, a rising star in modulating microglia activation under conditions of neurodegeneration
  • 2022
  • Ingår i: Cell Death and Disease. - : Springer Science and Business Media LLC. - 2041-4889. ; 13:7
  • Forskningsöversikt (refereegranskat)abstract
    • The advent of high-throughput single-cell transcriptomic analysis of microglia has revealed different phenotypes that are inherently associated with disease conditions. A common feature of some of these activated phenotypes is the upregulation of galectin-3. Representative examples of these phenotypes include disease-associated microglia (DAM) and white-associated microglia (WAM), whose role(s) in neuroprotection/neurotoxicity is a matter of high interest in the microglia community. In this review, we summarise the main findings that demonstrate the ability of galectin-3 to interact with key pattern recognition receptors, including, among others, TLR4 and TREM2 and the importance of galectin-3 in the regulation of microglia activation. Finally, we discuss increasing evidence supporting the involvement of this lectin in the main neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, multiple sclerosis, traumatic brain injury, and stroke.
  •  
6.
  • Arroyo-García, Luis Enrique, et al. (författare)
  • Targeting galectin-3 to counteract spike-phase uncoupling of fast-spiking interneurons to gamma oscillations in Alzheimer’s disease
  • 2023
  • Ingår i: Translational Neurodegeneration. - : Springer Science and Business Media LLC. - 2047-9158. ; 12:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Background: Alzheimer’s disease (AD) is a progressive multifaceted neurodegenerative disorder for which no disease-modifying treatment exists. Neuroinflammation is central to the pathology progression, with evidence suggesting that microglia-released galectin-3 (gal3) plays a pivotal role by amplifying neuroinflammation in AD. However, the possible involvement of gal3 in the disruption of neuronal network oscillations typical of AD remains unknown. Methods: Here, we investigated the functional implications of gal3 signaling on experimentally induced gamma oscillations ex vivo (20–80 Hz) by performing electrophysiological recordings in the hippocampal CA3 area of wild-type (WT) mice and of the 5×FAD mouse model of AD. In addition, the recorded slices from WT mice under acute gal3 application were analyzed with RT-qPCR to detect expression of some neuroinflammation-related genes, and amyloid-β (Aβ) plaque load was quantified by immunostaining in the CA3 area of 6-month-old 5×FAD mice with or without Gal3 knockout (KO). Results: Gal3 application decreased gamma oscillation power and rhythmicity in an activity-dependent manner, which was accompanied by impairment of cellular dynamics in fast-spiking interneurons (FSNs) and pyramidal cells. We found that the gal3-induced disruption was mediated by the gal3 carbohydrate-recognition domain and prevented by the gal3 inhibitor TD139, which also prevented Aβ42-induced degradation of gamma oscillations. Furthermore, the 5×FAD mice lacking gal3 (5×FAD-Gal3KO) exhibited WT-like gamma network dynamics and decreased Aβ plaque load. Conclusions: We report for the first time that gal3 impairs neuronal network dynamics by spike-phase uncoupling of FSNs, inducing a network performance collapse. Moreover, our findings suggest gal3 inhibition as a potential therapeutic strategy to counteract the neuronal network instability typical of AD and other neurological disorders encompassing neuroinflammation and cognitive decline.
  •  
7.
  • Bachiller, Sara, et al. (författare)
  • The Ubiquitin Proteasome System in Neuromuscular Disorders : Moving Beyond Movement
  • 2020
  • Ingår i: International Journal of Molecular Sciences. - : MDPI AG. - 1422-0067. ; 21:17
  • Forskningsöversikt (refereegranskat)abstract
    • Neuromuscular disorders (NMDs) affect 1 in 3000 people worldwide. There are more than 150 different types of NMDs, where the common feature is the loss of muscle strength. These disorders are classified according to their neuroanatomical location, as motor neuron diseases, peripheral nerve diseases, neuromuscular junction diseases, and muscle diseases. Over the years, numerous studies have pointed to protein homeostasis as a crucial factor in the development of these fatal diseases. The ubiquitin-proteasome system (UPS) plays a fundamental role in maintaining protein homeostasis, being involved in protein degradation, among other cellular functions. Through a cascade of enzymatic reactions, proteins are ubiquitinated, tagged, and translocated to the proteasome to be degraded. Within the ubiquitin system, we can find three main groups of enzymes: E1 (ubiquitin-activating enzymes), E2 (ubiquitin-conjugating enzymes), and E3 (ubiquitin-protein ligases). Only the ubiquitinated proteins with specific chain linkages (such as K48) will be degraded by the UPS. In this review, we describe the relevance of this system in NMDs, summarizing the UPS proteins that have been involved in pathological conditions and neuromuscular disorders, such as Spinal Muscular Atrophy (SMA), Charcot-Marie-Tooth disease (CMT), or Duchenne Muscular Dystrophy (DMD), among others. A better knowledge of the processes involved in the maintenance of proteostasis may pave the way for future progress in neuromuscular disorder studies and treatments.
  •  
8.
  • Boza-Serrano, Antonio, et al. (författare)
  • Galectin-3, a novel endogenous TREM2 ligand, detrimentally regulates inflammatory response in Alzheimer’s disease
  • 2019
  • Ingår i: Acta Neuropathologica. - : Springer Science and Business Media LLC. - 0001-6322 .- 1432-0533. ; 138:2, s. 251-273
  • Tidskriftsartikel (refereegranskat)abstract
    • Alzheimer’s disease (AD) is a progressive neurodegenerative disease in which the formation of extracellular aggregates of amyloid beta (Aβ) peptide, fibrillary tangles of intraneuronal tau and microglial activation are major pathological hallmarks. One of the key molecules involved in microglial activation is galectin-3 (gal3), and we demonstrate here for the first time a key role of gal3 in AD pathology. Gal3 was highly upregulated in the brains of AD patients and 5xFAD (familial Alzheimer’s disease) mice and found specifically expressed in microglia associated with Aβ plaques. Single-nucleotide polymorphisms in the LGALS3 gene, which encodes gal3, were associated with an increased risk of AD. Gal3 deletion in 5xFAD mice attenuated microglia-associated immune responses, particularly those associated with TLR and TREM2/DAP12 signaling. In vitro data revealed that gal3 was required to fully activate microglia in response to fibrillar Aβ. Gal3 deletion decreased the Aβ burden in 5xFAD mice and improved cognitive behavior. Interestingly, a single intrahippocampal injection of gal3 along with Aβ monomers in WT mice was sufficient to induce the formation of long-lasting (2 months) insoluble Aβ aggregates, which were absent when gal3 was lacking. High-resolution microscopy (stochastic optical reconstruction microscopy) demonstrated close colocalization of gal3 and TREM2 in microglial processes, and a direct interaction was shown by a fluorescence anisotropy assay involving the gal3 carbohydrate recognition domain. Furthermore, gal3 was shown to stimulate TREM2–DAP12 signaling in a reporter cell line. Overall, our data support the view that gal3 inhibition may be a potential pharmacological approach to counteract AD.
  •  
9.
  • García-Revilla, Juan, et al. (författare)
  • Galectin-3 shapes toxic alpha-synuclein strains in Parkinson’s disease
  • 2023
  • Ingår i: Acta Neuropathologica. - 0001-6322. ; 146:1, s. 51-75
  • Tidskriftsartikel (refereegranskat)abstract
    • Parkinson’s Disease (PD) is a neurodegenerative and progressive disorder characterised by intracytoplasmic inclusions called Lewy bodies (LB) and degeneration of dopaminergic neurons in the substantia nigra (SN). Aggregated α-synuclein (αSYN) is known to be the main component of the LB. It has also been reported to interact with several proteins and organelles. Galectin-3 (GAL3) is known to have a detrimental function in neurodegenerative diseases. It is a galactose-binding protein without known catalytic activity and is expressed mainly by activated microglial cells in the central nervous system (CNS). GAL3 has been previously found in the outer layer of the LB in post-mortem brains. However, the role of GAL3 in PD is yet to be elucidated. In post-mortem samples, we identified an association between GAL3 and LB in all the PD subjects studied. GAL3 was linked to less αSYN in the LB outer layer and other αSYN deposits, including pale bodies. GAL3 was also associated with disrupted lysosomes. In vitro studies demonstrate that exogenous recombinant Gal3 is internalised by neuronal cell lines and primary neurons where it interacts with endogenous αSyn fibrils. In addition, aggregation experiments show that Gal3 affects spatial propagation and the stability of pre-formed αSyn fibrils resulting in short, amorphous toxic strains. To further investigate these observations in vivo, we take advantage of WT and Gal3KO mice subjected to intranigral injection of adenovirus overexpressing human αSyn as a PD model. In line with our in vitro studies, under these conditions, genetic deletion of GAL3 leads to increased intracellular αSyn accumulation within dopaminergic neurons and remarkably preserved dopaminergic integrity and motor function. Overall, our data suggest a prominent role for GAL3 in the aggregation process of αSYN and LB formation, leading to the production of short species to the detriment of larger strains which triggers neuronal degeneration in a mouse model of PD.
  •  
10.
  • Mobley, Kenyon B., et al. (författare)
  • No evidence that stickleback spines directly increase risk of predation by an invertebrate predator
  • 2013
  • Ingår i: Evolutionary Ecology Research. - 1522-0613 .- 1937-3791. ; 15:2, s. 189-198
  • Tidskriftsartikel (refereegranskat)abstract
    • Background: Some populations of stickleback have a reduced number and/or relative size of spines. Hypothesis: Macroinvertebrate predators such as dragonfly larvae cause selective pressure against spines by capturing more stickleback with pelvic spines than stickleback that are spineless. Organisms: Ninespine stickleback (Pungitius pungitius) and dragonfly larvae (Aeshna grandis). Methods: We used 10 stickleback, five with pelvic spines and five with their pelvic spines removed. We put them in containers with two dragonfly larvae. Every day for 4 days we monitored how many stickleback were captured by the larvae. We repeated this experiment ten times at two different densities of fish and predators. We also developed a model to determine whether selection for spinelessness can be distinguished from drift. Results: Dragonfly larvae caught as many stickleback with spines as without. The absence of spines was not associated with a decrease in predation risk. We substituted Bayesian estimates of the selection coefficient into quantitative genetic models of allele frequency change, and the results of the models suggest that the selective advantage of spine loss is so small that its effects cannot be distinguished from drift.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-10 av 12
Typ av publikation
tidskriftsartikel (9)
forskningsöversikt (3)
Typ av innehåll
refereegranskat (12)
Författare/redaktör
Ruiz, Rocío (8)
Deierborg, Tomas (7)
Venero, José L. (4)
Bachiller, Sara (3)
Alonso-Bellido, Isab ... (2)
Real, Luis M. (2)
visa fler...
Leffler, Hakon (1)
Boada, Mercè (1)
Alarcón-Riquelme, Ma ... (1)
Karlsen, Tom H (1)
Linse, Sara (1)
Gago Dominguez, Manu ... (1)
Ayuso, Carmen (1)
Cao, Yang, Associate ... (1)
Castelao, Jose E. (1)
Gonzalez-Neira, Anna (1)
Pita, Guillermo (1)
Englund, Elisabet (1)
Frithiof, Robert (1)
Englund, Martin (1)
Turkiewicz, Aleksand ... (1)
Ganna, Andrea (1)
Önnerfjord, Patrik (1)
Guillén-Navarro, Enc ... (1)
Aleix-Mata, Gael (1)
Exposito, Miriam (1)
Ruiz-Ruano, Francisc ... (1)
Maria Lopez-Beceiro, ... (1)
Fidalgo, Luis E. (1)
Martinez-Carrasco, C ... (1)
Ruiz de Ybanez, Mari ... (1)
Boos, Mathieu (1)
Perez, Jesus M. (1)
Sanchez, Antonio (1)
Carracedo, Angel (1)
Ali, Neserin (1)
Vázquez, Guillermo (1)
Cruz-Hernández, Luis (1)
Martínez, Emilio (1)
Ruiz-Mateos, Ezequie ... (1)
Stegmayr, John (1)
Herrero, Maria José (1)
Schulte, Eva C (1)
Fernandez-Cadenas, I ... (1)
Johansson, Frank (1)
Andrade-Talavera, Yu ... (1)
Arroyo-García, Luis ... (1)
Fisahn, André (1)
Blanco, Francisco J. (1)
Prati, Daniele (1)
visa färre...
Lärosäte
Lunds universitet (8)
Uppsala universitet (3)
Karolinska Institutet (3)
Umeå universitet (2)
Örebro universitet (1)
Språk
Engelska (12)
Forskningsämne (UKÄ/SCB)
Medicin och hälsovetenskap (10)
Naturvetenskap (3)

År

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy