• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 182
  • 53
  • 27
  • 16
  • 8
  • 7
  • 6
  • 5
  • 3
  • 3
  • 2
  • 1
  • 1
  • Tagged with
  • 369
  • 369
  • 369
  • 82
  • 73
  • 67
  • 42
  • 37
  • 34
  • 34
  • 33
  • 32
  • 31
  • 31
  • 26
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
231

Veneno e toxina da aranha Phoneutria nigriventer : ação no sistema nervoso central / Venon and toxin of Phoneutria nigriventer spider : action in the central nervous system

Carneiro, Catarina Raposo Dias 14 August 2018 (has links)
Orientador: Maria Alice da Cruz-Hofling / Tese (doutorado) - Universidade Estadual de Campinas, Instituto de Biologia / Made available in DSpace on 2018-08-14T00:39:53Z (GMT). No. of bitstreams: 1 Carneiro_CatarinaRaposoDias_D.pdf: 35688895 bytes, checksum: dc0574f3ff57450c0d5ff8840699f5e9 (MD5) Previous issue date: 2009 / Resumo: Venenos animais são fontes de substâncias neuroativas, algumas capazes de provocar paralisia e convulsão em mamíferos, com visível ação no sistema nervoso central (SNC). O veneno da aranha Phoneutria nigriventer (PNV) é composto por neurotoxinas que causam, experimentalmente, permeabilização da barreira hematoencefálica (BHE). A BHE é uma entidade tanto física, quanto molecular, composta pelos microvasos sanguíneos cerebrais, pelos pés astrocitários e pericitos adjacentes engajados no controle do tráfego de moléculas na interface sangue-cérebro. A BHE, embora imprescindível à manutenção da homeostase no SNC, pode representar um obstáculo ao acesso de drogas terapêuticas ao microambiente neural. Nossa proposta foi investigar a ação sistêmica do PNV após 15 min, 2 e 5 h da injeção i.v. em ratos Wistar adultos através de: (1) alterações na expressão das proteínas juncionais, de efluxo e transportador de glicose da BHE; (2) alterações na expressão da proteína conexina-43 (constituinte das junções comunicantes) e da proteína fosfatase pPP2A, uma vez que a fosforilação de resíduos de tirosina das proteínas juncionais tem papel no controle da integridade paracelular; (3) ativação de vias neuronais e sua modulação pelo óxido nítrico (NO). (4) Reação inflamatória e gliose reativa de astrócitos in vivo e in vitro e sua possível modulação pelo NO, (5) Purificação e identificação de toxinas do PNV com ação na BHE. A expressão das proteínas juncionais encontrava-se diminuída aos 15 min e 2 h, porém às 5 h pós-PNV a expressão das proteínas investigadas estava total ou parcialmente recuperada, sugerindo ser esse um dos mecanismos de abertura da BHE. Igualmente, a expressão da proteína de efluxo aumentou indicando mecanismo de clearance do agente tóxico. A expressão da conexina-43, e da pPP2A estavam aumentadas aos 15 min e diminuída às 5 h da injeção do PNV, mostrando não só que as comunicações célula-célula e o mecanismo de adesão célulacélula foram afetados, mas também que as alterações podem ser transitórias. Ademais, vias neuronais foram ativadas em áreas motoras e em núcleos do hipotálamo o que explicaria o comprometimento motor (convulsão, paralisia) e os sinais neurovegetativos (sialorréia, hipertensão, estresse respiratório, edema pulmonar, anúria) vistos em animais envenenados. Muitas dessas vias apontam modulação nitrérgica dos sinais tóxicos do envenenamento, uma vez que a inibição da síntese de NO pelo 7- nitroindazol (7-NI) diminuiu a ativação neuronal em algumas áreas e exacerbou em outras. Os astrócitos incubados com PNV, corroborando com estudos in vivo, expressaram citocinas pró-inflamatórias e apresentaram gliose reativa, porém a inibição da síntese do NO atenuou esses efeitos, confirmando que o NO tem um importante papel nos efeitos do PNV. Toxinas F8a-1 e F10a-1, purificadas do PNV, foram identificadas como responsáveis pela permeabilização da BHE, embora não esteja excluída a contribuição de outros componentes. O entendimento da ação do PNV e de suas toxinas no tecido neural e na BHE pode contribuir para o desenvolvimento de ferramentas úteis para uso clínico e em pesquisa. / Abstract: Animal venoms are source of neuroactive substances, some of them able to provoke paralysis and convulsion in mammals, indicating action on the central nervous system (CNS). The Phoneutria nigriventer spider venom (PNV) is composed of neurotoxins that cause, experimentally, blood-brain barrier (BBB) permeabilization. The BBB is both a physical and molecular entity, constituted by the cerebral microvessels and surrounding astrocytic end-feet and pericytes, all involved in the control of the traffic of molecules at the blood-brain interface. Even so the BBB presence is essential for the maintenance of CNS homeostasis; it also represents an obstacle for the therapeutical drugs access into the neural microenvironment. Our proposal was to investigate acute changes (15 min, 2 and 5 h) after PNV i.v. injection in adult Wistar rats through evaluation of the: (1) alterations in the expression of the BBB-junctional proteins, -efflux proteins, and -glucose transporter; (2) alterations in the expression of connexin-43 protein (gap junctions constituent) and protein phosphatase 2A (pPP2A, since phosphorylation of tyrosine residues from junctional proteins plays a role in controlling junctional integrity); (3) activation of neuronal pathways and its modulation by the nitric oxide (NO). (4) In vivo and in vitro astrocytes inflammatory reaction and reactive gliosis after incubation with PNV and its possible modulation by NO. (5) Purification and determination of BBB-acting from toxins PNV. The expression of the junctional proteins was diminished at 15 min and 2 h post-PNV exposure; however at 5 h the expression of most of the proteins investigated was total or partially recovered, suggesting that this might be one of the BBB opening mechanisms. Similarly, the venom increased the efflux protein expression, indicating ongoing clearance of toxic agents from the neural tissue. The expression of both connexin-43 and pPP2A increased after 15 min and diminished after 5 h of PNV injection, showing not only that cell-cell communication and cell-cell adhesion mechanism were affected, but that these alterations were transitory. Activated neuronal pathways have been observed in brain motor areas and hypothalamic nucleus, explaining the motor impairment (convulsion, paralysis) and neurovegetative signs (salivation, hypertension, respiratory stress, pulmonary edema, anuria) observed in the envenomed animals. Many of these neuronal pathways point to a nitrergic modulation of the envenomed toxic signs, since that NO synthesis inhibition by 7-nitroindazol (7-NI) decreased the neuronal activation in some areas and enhanced in others. The astrocytes incubated with PNV, in agreement with in vivo studies, expressed pro-inflammatory cytokines and presented reactive gliosis, however the pretreatment with 7- NI attenuated these effects, confirming that NO have an important role in the PNV effects. The F8a-1 and F10a-1 toxins, fractionated from the crude PNV, were identified as responsible by BBB permeabilization; despite, other venom components contribution can not be discarded. The understanding of the action of the venom of Phoneutria nigriventer and its toxins in the neural tissue and BBB can contribute for the development of useful tools for clinical and research purposes. / Doutorado / Biologia Celular / Doutor em Biologia Celular e Estrutural
232

Vectorisation de molécules thérapeutiques aux tissus cérébraux / Drug delivery to the central nervous system

Nieto Montesinos, Rita Milagros 19 February 2014 (has links)
La présence de la glycoprotéine P (P-gp) dans la barrière hémato-encéphalique (BHE) conduit à l’échec de nombreuses thérapies ciblant le système nerveux central (SNC). Cependant la P-gp protège aussi le cerveau contre des composés nocifs, essentiellement lipophiles, endogènes et exogènes susceptibles de passer la BHE par diffusion simple. Par conséquent, toute inhibition de la P-gp qui vise à améliorer la distribution des agents pharmacologiques dans le cerveau doit prendre en compte la neurotoxicité potentielle de cette inhibition. Les premiers travaux ont montré que l’elacridar et le tariquidar, deux modulateurs de la P-gp de troisième génération, augmentaient la distribution dans le cerveau de plusieurs de ses substrats. Malheureusement, d’autres études plus récentes, suggèrent l’utilisation de doses élevées de l’elacridar et du tariquidar pour moduler efficacement l’activité de la P-gp dans la BHE. Néanmoins, ces doses élevées en co-administration avec des substrats de la P-gp peuvent être associées à des interactions pharmacocinétiques et à des profils toxiques, limitant ainsi l'utilisation de ces inhibiteurs.Dans ce contexte, l’objectif principal de cette thèse est d’obtenir une modulation transitoire mais efficace de la P-gp dans la BHE par administration intraveineuse de doses faibles mais thérapeutiques de l’elacridar et du tariquidar sous leur forme libre ou co-encapsulé dans les liposomes. Le lopéramide, substrat de la P-gp, a été également administré sous sa forme libre comme une preuve in vivo d’une inhibition efficace de la P-gp dans la BHE.L'administration simultanée de ces deux modulateurs de la P-gp n’a pas modifié leurs concentrations plasmatiques ou celles du lopéramide, mais a entraîné une importante distribution du lopéramide dans le cerveau en raison de leur activité inhibitrice non- compétitive. De plus, la co-encapsulation de l’elacridar et du tariquidar dans des immunoliposomes stabilisées stériquement a amélioré la demi-vie et la distribution dans le cerveau des ceux deux composés. Par conséquent, la distribution dans le cerveau du lopéramide a été considérablement augmentée, sans aucune modification de sa pharmacocinétique ou distribution tissulaire. Par ailleurs, la diminution partielle de l'activité inhibitrice du tariquidar par des liposomes vides suggère l’utilisation de ce nanovecteur comme une approche de bio-détoxification pour le traitement des surdoses de tariquidar. En résumé, cette thèse propose différentes approches pour exploiter pleinement l’elacridar et le tariquidar. Les résultats décrits dans ce manuscrit devraient ouvrir des pistes intéressantes pour atteindre une inhibition efficace de la P-gp dans la BHE et pour réussir des thérapies ciblant le système nerveux central / Although the P-glycoprotein (P-gp) represents an obstacle in several central nervous system (CNS) pharmacotherapies, the P-gp also protects the brain from intoxication by endogenous and exogenous harmful lipophilic compounds that otherwise could penetrate the blood-brain barrier (BBB) by simple diffusion. Therefore, any modulation of the efflux transporter has to consider the potential neurotoxicity of such modulation. Early studies showed that elacridar and tariquidar, two third-generation P-gp modulators, increase the distribution of several P-gp substrates in the brain. Unfortunately, recent studies suggest the use of high doses of elacridar and tariquidar to efficiently modulate the P-gp at the BBB. Nevertheless, when co-administered with P-gp substrates, these high doses may be associated with pharmacokinetic interactions and toxic profiles, thus limiting the use of these compounds.Hence, this thesis aimed to attain a transient but efficient modulation of the P-gp at the BBB using elacridar and tariquidar but avoiding the use of large doses of these compounds. For this purpose we took advantage of the possible in vivo intravenous co-administration of low but therapeutic doses of elacridar and tariquidar, under their free form or co-encapsulated in liposomes. The brain distribution of free loperamide was determined as an in vivo probe of full inhibition of the P-gp activity at the BBB.The concurrent administration of both free P-gp modulators does not modify their plasma concentrations or those of the P-gp substrate but significantly increased the brain uptake of loperamide as a result of their non-competitive modulatory activity. Moreover, the co-encapsulation of elacridar and tariquidar in targeted sterically stabilized immunoliposomes improved the half-lives and brain distribution of both compounds. Consequently, the brain uptake of free loperamide was significantly enhanced without any modification of its pharmacokinetics or tissue distribution. Moreover, the partial impairment of the modulatory activity of tariquidar by empty liposomes, supports the use of this nanocarrier as a bio-detoxifying approach for the treatment of tariquidar overdoses.In summary, this thesis proposes different approaches for full exploitation of elacridar and tariquidar. The findings described in this manuscript should open interesting avenues to achieve an efficient overcoming of the P-gp at the BBB and succeed CNS pharmacotherapies.
233

Models for predicting efflux transport over the blood-brain barrier

Janani, Marjaneh January 2020 (has links)
Aim: The general aim of this research is development and evaluation of novel methods for predicting active transport over the human blood-brain-barrier (BBB), while this project specifically aims to i) review the literature and select suitable methods and substrates, ii) develop models for determining in vitro kinetic properties of selected compounds, analyze the in vitro data using the developed models and to use Maximum Transport Activity (MTA) approach (Karlgren et al., 2012), iii) perform Physiology Based Pharmacokinetic (PBPK) modelling and compare to in vivo literature data. Background: Drug permeation to the brain through blood circulation is primarily limited by blood-brain barrier (BBB), due to existence of tight junctions in endothelial cells of blood vessels as well as active efflux and influx transporters in the barrier. Toxicity and CNS related side effects can be caused by peripheral targeted drugs crossing BBB. Hence, prediction of BBB permeability and estimation of drug concentration in the brain tissue are challenging in drug discovery. To resolve this, estimating the human BBB permeability using improved in vitro and in silico predictive models can be a facilitator. Methods: In vitro data provided by the Drug Delivery research group was used to develop in vitro predictive models for BBB penetration of Verapamil, Risperidone, and Prazosin using R-studio 1.2.5. The MTA approach was adjusted for extrapolation of BBB in vitro transporter activity to in vivo condition. For PBPK modelling, we took advantage of PK-Sim® to simulate drug disposition and time profile of Risperidone in human and animal species. Results: It was shown that MDR1 is the major transporter for efflux transport of Prazosin and Risperidone in brain while both BCRP and MDR1 have similar impact on transport of Verapamil. Furthermore, it was presented in PBPK models that the predicted brain concentration of Risperidone increases in rat and nonhuman primate (NHP) when MDR1 And BCRP are knocked out while the brain concentration of Risperidone in dog is not affected by expression level of the efflux transporters. Conclusion: Both MDR1 and BCRP are contributing in efflux transport of Verapamil, Risperidone, and Prazosin across the BBB. Additionally, expression of the efflux transporters shown to have an impact on brain exposure of Risperidone in animal PBPK models.
234

Evaluation of TiO2 exposure impact on adult and vulnerable brains / Evaluation des Effets de l'Exposition au TiO2 sur le Cerveau Adulte et Vulnérable

Disdier, Clémence 11 April 2016 (has links)
La présence croissante de nanoparticules (NPs) dans les produits de la vie quotidienne (alimentation, médicaments, cosmétiques, textiles…) soulève de sérieuses inquiétudes quant à leurs potentiels effets nocifs pour la santé humaine. Les NPs de dioxyde de titane (TiO2) sont produites à l’échelle industrielle et peuvent déjà être trouvées dans plusieurs produits commerciaux tels que les peintures, les cosmétiques ou dans les systèmes de décontamination de l’eau ou de l’air. Dans le passé, les NPs de TiO2 étaient considérées comme inertes, mais, très récemment, l'Agence Internationale pour la Recherche sur le Cancer les a classées comme possiblement cancérogènes (groupe 2B) pour l’homme. De nombreuses études in vitro et in vivo ont démontré la potentielle neuro-toxicité des NPs de TiO2, mais très peu d'études se sont concentrées plus spécifiquement sur la barrière hémato-encéphalique (BHE), protégeant le cerveau. Aujourd'hui, en dépit des avancées constatées, la bio-cinétique et la bio-accumulation des NPs de TiO2 ainsi que les conséquences sur la physiologie de la barrière hémato-encéphalique (BHE) in vivo restent très peu documentées. De plus, dans l’évaluation du risque lié à l’exposition aux NPs, des facteurs de risque tel que l’âge ont jusqu’ici été quasiment ignorés. Dans ce contexte, l’objectif de ce projet est donc d’évaluer chez le rat adulte et âgé, l’impact d’une exposition aux NPs de TiO2 sur les fonctions de la BHE et sur le métabolisme cérébral. Nos résultats ont montré que les NPs de TiO2 s’accumulent dans certains organes et tissus (principalement dans les poumons, la rate et le foie) et ne sont pas distribuées au système nerveux central (SNC) que ce soit après injection intra-veineuse (IV) ou après une inhalation subaiguë à un nano-aérosol de TiO2. Après administration IV, une interaction directe entre NPs et les cellules endothéliales microvasculaires conduit à des altérations fonctionnelles au niveau de la BHE. Malgré l'absence de translocation vers le SNC, la bio-persistance du titane dans les organes périphériques semble être la cause de modulations de perméabilité de la BHE et d’une inflammation cérébrale. L'implication de médiateurs circulants faisant le lien entre la bio-persitance de titane dans les organes périphériques et les modulations observées au niveau cérébral a été démontré en utilisant un modèle in vitro de BHE. Une réponse exacerbée en termes de neuro-inflammation et de modulation de perméabilité de la BHE établit la vulnérabilité du cerveau âgé à la toxicité des NPs inhalées. Ces résultats ont démontré que malgré l'absence de translocation cérébrale, l'exposition aux NPs de TiO2 induit des altérations fonctionnelles de la BHE et une neuro-inflammation qui pourraient conduire à des troubles neurologiques. L’identification des médiateurs et la description des effets neurotoxiques restent encore à préciser. / The overwhelming presence of nanoparticles (NPs) in products including foods, medications, cosmetics, or textiles raises serious concerns about their potential harmful effects on human health. In the wide diversity of NPs, titanium dioxide (TiO2) NPs are among those produced on a large industrial scale and can already be found in several commercial products such as paints, cosmetics or in environmental decontamination systems. In the past, TiO2 NPs was considered inert, but, very recently, the International Agency for Research in Cancer (IARC) has classified TiO2 as possibly carcinogenic (group 2B) to human beings. Numerous in vitro and in vivo studies have shown the potential neuro-toxicity of TiO2 NPs, but very few studies focus on the central nervous system (CNS), Nowadays, notwithstanding the reported advances, the biokinetic and bioaccumulation ofTiO2 NPs and the consequences on the physiology of the blood-brain barrier (BBB) in vivo are unknown. In addition, NPs effect on susceptible population such as the elderly have been mostly ignored. In this context, the target of the present studies is to evaluate the in vivo impact of exposure to NPs on the BBB physiology and brain inflammation which could promote neurotoxicity in young adults and aging. Our results have shown that TiO2 NPs bioaccumulate in organs and tissues (lungs, spleen and liver especially) and don’t translocate to the brain either after IV or subacute inhalation exposure. In IV administration case, the direct interaction between NPs and brain endothelial cells induces BBB functional alterations. Despite the lack of CNS translocation, the biopersistence of titanium in peripheral organs may be indirectly the cause of BBB permeability alteration and brain inflammation. The involvement of circulating mediators linking titanium biopersitence in peripheral organs and brain impact has been demonstrated using an in vitro BBB model. An exacerbated response in term of neuro-inflammation and BBB permeability modulation has established the vulnerability of the aging brain to inhaled NPs toxicity. Taken together, our findings demonstrated that despite lack of brain translocation, exposure to TiO2 NPs induce BBB physiology alteration and neuro-inflammation that may lead to CNS disorders. Thereafter, identification of mediators and description of the neurotoxic effects may complete the assessment of the impact of TiO2 NPs exposure on the brain.
235

Therapeutic approaches for two distinct CNS pathologies

Stumpf, Sina Kristin 25 June 2018 (has links)
No description available.
236

Sex Differences In the Enduring Neuroinflammatory and Behavioural Sequelae of Systemic Immune Challenge During Puberty

Kolmogorova, Daria 19 May 2021 (has links)
Puberty is a critical period for sexual maturation during which the sex-specific reorganization and remodelling of the pubertal brain facilitate sex biases in stress sensitivity. Pubertal (i.e., six-week-old) CD-1 mice treated with the bacterial endotoxin lipopolysaccharide (LPS; 1.5 mg/kg body weight, ip) show several sex-specific changes to the neuroendocrine and behavioural systems of several reproductive and non-reproductive functions. One promising explanation for the elusive mechanisms driving the sex-specific outcomes of pubertal immune challenge may lie in the cascade of neuroimmune events induced by this systemic immune stressor. This doctoral thesis tested the hypothesis that sex-specific responses of the pubertal neuroimmune network contribute to sex differences in the enduring outcomes of pubertal immune challenge on hippocampus-dependent cognitive processes. Male and female CD-1 mice are equally vulnerable to enduring impairments in spatial memory following pubertal LPS exposure. Across brain regions for cognition and stress regulation, pubertal LPS treatment alters baseline sex differences in microglial expression and morphology in a sex-dependent manner. The temporary female-specific increase in whole-brain blood-brain barrier permeability during LPS-induced sickness may have facilitated the apparent female bias in LPS-induced changes to pubertal microglia. In the context of sex- and region-specific residual effects of pubertal LPS-induced sickness on microglial expression and morphology, pubertal LPS treatment may accelerate certain neurodevelopmental processes in males but not females. The innate sex differences in the pubertal neuroimmune network highlighted by these studies underscore how a systemic immune challenge precipitates sex biases in immune-mediated disorders of brain and behaviour during adulthood.
237

Développements précliniques de nouveaux outils utilisant les ultrasons transcraniens guidés par IRM haut champ pour la délivrance de médicaments dans le cerveau et la stimulation non invasive de circuits neuronaux / Development of preclinical tool using transcranial ultrasound under high field MR-guidance for drug delivery to the brain and non-invasive neurostimulation

Magnin, Rémi 07 January 2016 (has links)
La Barrière Hémato-Encéphalique (BHE) représente aujourd’hui un obstacle majeur pour le développement de nouveaux traitements des pathologies cérébrales puisqu’elle empêche le passage de la majorité des agents thérapeutiques vers le cerveau. Afin de contourner cet obstacle, une technique proposée dans les années 2000 a montré son potentiel pour perméabiliser la BHE de façon non-invasive, locale et transitoire grâce à l’utilisation conjointe de microbulles circulantes et d’ultrasons focalisés, permettant une augmentation significative de la quantité de molécules délivrée aux tissus cérébraux. Ce protocole peut néanmoins présenter des risques (œdèmes, micro-hémorragies) qu’il est possible de maîtriser grâce à un bon contrôle du faisceau acoustique. A ce titre, l’imagerie par résonance magnétique (IRM) représente un outil de choix permettant à la fois de planifier la procédure, puis de la suivre et enfin d’étudier ses effets grâce à l’utilisation d’agents de contraste et de séquences d’imagerie quantitative (relaxométrie T1 / T2). Durant cette thèse, nous avons développé de nouveaux outils permettant l’étude de la perméabilisation de la BHE chez le rongeur. Dans un premier temps, nous avons développé et validé un système motorisé compatible IRM, permettant de déplacer le transducteur ultrasonore à l’intérieur d’un scanner préclinique à 7T, avec un rétro-contrôle en temps réel du faisceau ultrasonore sur la base des images IRM de la force de radiation acoustique (MR-ARFI). Nous avons montré que ce dispositif permettait de réaliser l’ensemble du protocole de perméabilisation de BHE guidé par IRM en choisissant la structure anatomique à traiter de façon reproductible. Nous avons également montré qu’il pouvait être utilisé pour délivrer des molécules sur des régions étendues du cerveau selon des trajectoires arbitrairement programmées. Dans une seconde partie de cette thèse, nous avons réalisé plusieurs études en vue d’étudier l’innocuité de la technique. Nous avons mis en évidence l’influence de certains paramètres acoustiques sur l’efficacité du protocole (pression acoustique, temps de cycle), puis nous avons réalisé une étude histologique des dommages engendrés par une perméabilisation chez des animaux sains pour plusieurs pressions acoustiques, entre 0 et 14 jours post-ultrasons. Enfin, dans une troisième partie, nous avons étudié la diffusion de différents agents de contraste paramagnétiques dans les tissus cérébraux suivant une perméabilisation focale de la BHE. Nous avons montré que cette technique permettait d’obtenir des mesures précises de la tortuosité des tissus cérébraux de façon non-invasive et que cette tortuosité n’était pas modifiée par les ultrasons, contrairement à ce qui est observé suite à une injection intracérébrale. / By preventing most of the molecules from penetrating the brain in sufficient quantitiy, the Blood Brain Barrier represents a major obstacle for the development of new therapeutic drugs for brain diseases. A new technique introduced in the early 2000’s combining focused ultrasound and circulating microbubbles has however shown promising results, allowing to induce a local and transient permeabilization of the BBB in a non-invasive manner, thus significantly improving the amount of drugs delivered to the Central Nervous System (CNS). However, this protocol may present some risks (oedema, small hemorrages) which can be avoided by a good control of the acoustic beam properties. To do so, Magnetic Resonance Imaging (MRI) represents a very useful tool since it allows planning, monitoring and following the permeabilization effects by using MRI contrast agents and quantitative imaging sequences (T1/T2 relaxometry). During this PhD, we worked on developing new tools for the study of ultrasound induced BBB permeabilization in rodents. The first part of this work consisted in developing a MR compatible motorized device, allowing the displacment of the ultrasound transducer within a 7T preclinic MRI scanner, with a realtime feedback on the acoustic beam position thanks to MR Acoustic Radiation Force Imaging (MR-ARFI). We have shown that this system allowed performing a full BBB permeabilization protocol under MR-guidance, with an accurate and reproducible choice of the targeted anatomical structure. This system was also used to deliver drugs along arbitrary trajectories over extended regions of the brain. Another part of the work was dedicated to study and improve the safety of the procedure. The influence of different acoustic parameters (acoustic pressure, duty cycle) on the permeabilization efficacy was studied, as well as histologic investigations of short and mid-term effects of BBB permeabilization for different acoustic pressures on healthy rats. Finally, we investigated the diffusion process of contrast agents within the brain tissues following BBB permeabilization. We have shown that this technique allowed accurate measurements of brain tissues tortuosity in a non-invasive way, and found that the tortuosity was not modified by the ultrasound application.
238

Treating Metastatic Brain Cancers With Stem Cells

Sadanandan, Nadia, Shear, Alex, Brooks, Beverly, Saft, Madeline, Cabantan, Dorothy Anne Galang, Kingsbury, Chase, Zhang, Henry, Anthony, Stefan, Wang, Zhen Jie, Salazar, Felipe Esparza, Lezama Toledo, Alma R., Rivera Monroy, Germán, Vega Gonzales-Portillo, Joaquin, Moscatello, Alexa, Lee, Jea Young, Borlongan, Cesario V. 24 November 2021 (has links)
Stem cell therapy may present an effective treatment for metastatic brain cancer and glioblastoma. Here we posit the critical role of a leaky blood-brain barrier (BBB) as a key element for the development of brain metastases, specifically melanoma. By reviewing the immunological and inflammatory responses associated with BBB damage secondary to tumoral activity, we identify the involvement of this pathological process in the growth and formation of metastatic brain cancers. Likewise, we evaluate the hypothesis of regenerating impaired endothelial cells of the BBB and alleviating the damaged neurovascular unit to attenuate brain metastasis, using the endothelial progenitor cell (EPC) phenotype of bone marrow-derived mesenchymal stem cells. Specifically, there is a need to evaluate the efficacy for stem cell therapy to repair disruptions in the BBB and reduce inflammation in the brain, thereby causing attenuation of metastatic brain cancers. To establish the viability of stem cell therapy for the prevention and treatment of metastatic brain tumors, it is crucial to demonstrate BBB repair through augmentation of vasculogenesis and angiogenesis. BBB disruption is strongly linked to metastatic melanoma, worsens neuroinflammation during metastasis, and negatively influences the prognosis of metastatic brain cancer. Using stem cell therapy to interrupt inflammation secondary to this leaky BBB represents a paradigm-shifting approach for brain cancer treatment. In this review article, we critically assess the advantages and disadvantages of using stem cell therapy for brain metastases and glioblastoma. / National Institutes of Health / Revisión por pares
239

Optimization of Focused Ultrasound Mediated Blood-Brain Barrier Opening

Ji, Robin January 2022 (has links)
Treatment of brain diseases remains extremely challenging partly due to the fact that critical drug delivery is hindered by the blood-brain barrier (BBB), a specialized and highly selective barrier lining the brain vasculature. Focused ultrasound (FUS), combined with systematically administered microbubbles (MBs), has been established as a technique to noninvasively, locally, and transiently open the BBB. The primary mechanism for temporarily opening the BBB using FUS is microbubble cavitation, a phenomenon that occurs when the circulating microbubbles interact with the FUS beam in the brain vasculature. Over the past two decades, many preclinical and clinical applications of FUS-induced BBB opening have been developed, but certain challenges, such as drug delivery route, cavitation control, inflammation onset, and overall accessibility of the technology, have affected its efficient translation to the clinic. This dissertation focuses on optimizing three aspects of FUS-induced BBB opening for therapeutic applications. The first specific aim investigated FUS-induced BBB opening for drug delivery through the intranasal route. Optimal sonication parameters were determined and applied to FUS-enhanced intranasal delivery of neurotrophic factors in a Parkinson’s Disease mouse model. In the second specific aim, cavitation levels affecting the inflammatory response due to BBB opening with FUS were optimized. The relationship between cavitation during FUS-induced BBB opening and the local inflammation was examined, and a cavitation-based controller system was developed to modulate the inflammatory response. In the third specific aim, the devices used for FUS-induced BBB opening were streamlined. A conventional system for FUS-induced BBB opening includes two transducers: one for therapy and another for cavitation monitoring (single element) or imaging (multi-element). In this aim, a single linear array transducer capable of synchronous BBB opening and cavitation imaging was developed, creating a cost-effective and highly accessible “theranostic ultrasound” device. The feasibility of theranostic ultrasound (TUS) was demonstrated in vivo in both mice and non-human primates. In summary, the findings and methodologies in this dissertation optimized FUS-enhanced intranasal delivery across the BBB, developed a cavitation-controlled system to modulate inflammation in the brain, which has been advantageous in reducing pathology and designed a new system for theranostic ultrasound for drug delivery to the brain. Taken altogether, this thesis contributes to the efficient advancement and optimization of FUS-induced BBB opening technology, thus enhancing its clinical adoption in the fight to treat many challenging brain diseases.
240

Amelioration Of Amyloid Burden In Advanced Human And Mouse Alzheimer's Disease Brains By Oral Delivery Of Myelin Basic Protein Bioencapsulated In Plant Cells

Kohli, Neha 01 January 2012 (has links)
One of the pathological hallmarks of Alzheimer's disease (AD) is the amyloid plaque deposition in aging brains by aggregation of amyloid-β (Aβ) peptides. In this study, the effect of chloroplast derived myelin basic protein (MBP) fused with cholera toxin subunit B (CTB) was investigated in advanced diseased stage of human and mouse AD brains. The CTB-fusion protein in chloroplasts facilitates transmucosal delivery in the gut by the natural binding ability of CTB pentameric form with GM1 receptors on the intestinal epithelium. Further, bioencapsulation of the MBP within plant cells confers protection from enzymes and acids in the digestive system. Here, 12-14 months old triple transgenic AD mice were fed with CTB-MBP bioencapsulated in the plant cells for 3 months. A reduction of 67.3% and 33.3% amyloid levels in hippocampal and cortical regions, respectively were observed by immunostaining of brain sections with anti- Aβ antibody. Similarly, 70% decrease in plaque number and 40% reduction of plaque intensity was observed through thioflavin S (ThS) staining that specifically stains amyloid in the AD brain. Furthermore, ex vivo 3xTg AD mice brain sections showed up to 45% reduction of ThS stained amyloid levels when incubated with enriched CTB-MBP in a concentration dependent manner. Similarly, incubation of enriched CTB-MBP with ex vivo postmortem human brain tissue sections with advanced stage of AD resulted up to 47% decrease of ThS stained amyloid plaque intensity. Lastly, lyophilization of plant material facilitates dehydration and long term storage of capsules at room temperature, in addition to increasing CTB-MBP concentration by 17 fold. These observations offer a low cost solution for treatment of even advanced stages of the AD by facilitating delivery of therapeutic proteins to central nervous system to address other neurodegenerative disease.

Page generated in 0.0373 seconds