Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain Res Bull ; 207: 110864, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38157992

RESUMEN

Ischemic stroke induced inflammatory responses contribute significantly to neuronal damage and stroke outcomes. CD200 ligand and its receptor, CD200R, constitute an endogenous inhibitory signaling that is being increasingly recognized in studies of neuroinflammation in various central nervous system disorders. CD200 is a type 1 membrane glycoprotein that is broadly expressed by endothelia and neurons in the brain. In the present study, we have examined the role of endothelial CD200 signaling in acute ischemic stroke. Endothelial CD200 conditional knock out (CKO) mice were generated by breeding CD200 gene floxed mice with Cdh5Cre mice. The mice were subjected to a 60-min transient middle cerebral artery occlusion (MCAO). Flow cytometry, Immunohistochemical staining, and Western blotting were performed to assess the post-stroke inflammation; stroke outcomes (infarct volume and neurobehavioral deficits) were evaluated at 72 h after MCAO. We found CD200R was near-null expressed on microglia at 24 h after stoke. Endothelial CKO of CD200 had no impact on peripheral immune cell development. Immunohistochemical staining confirmed CD200 was expressed on CD200 floxed but not on CD200 CKO endothelia. CD200 CKO mice exhibited larger infarct size, worse neurological deficit scores (NDS), and more deficits in the adhesive removal when compared with control mice, 72 h after MCAO. Western blot results showed that endothelial CKO of CD200 did not change BBB protein expression. Together it suggests that endothelial CD200 signaling protects brains against ischemic injury through a mechanism not directly related to microglial activation.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Animales , Ratones , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Infarto/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Ratones Noqueados , Microglía/metabolismo , Accidente Cerebrovascular/metabolismo
2.
Transl Stroke Res ; 2023 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-37432594

RESUMEN

Microglia and monocytes play a critical role in immune responses to cerebral ischemia. Previous studies have demonstrated that interferon regulatory factor 4 (IRF4) and IRF5 direct microglial polarization after stroke and impact outcomes. However, IRF4/5 are expressed by both microglia and monocytes, and it is not clear if it is the microglial (central) or monocytic (peripheral) IRF4-IRF5 regulatory axis that functions in stroke. In this work, young (8-12 weeks) male pep boy (PB), IRF4 or IRF5 flox, and IRF4 or IRF5 conditional knockout (CKO) mice were used to generate 8 types of bone marrow chimeras, to differentiate the role of central (PB-to-IRF CKO) vs. peripheral (IRF CKO-to-PB) phagocytic IRF4-IRF5 axis in stroke. Chimeras generated from PB and flox mice were used as controls. All chimeras were subjected to 60-min middle cerebral artery occlusion (MCAO) model. Three days after the stroke, outcomes and inflammatory responses were analyzed. We found that PB-to-IRF4 CKO chimeras had more robust microglial pro-inflammatory responses than IRF4 CKO-to-PB chimeras, while ameliorated microglial response was seen in PB-to-IRF5 CKO vs. IRF5 CKO-to-PB chimeras. PB-to-IRF4 or IRF5 CKO chimeras had worse or better stroke outcomes respectively than their controls, whereas IRF4 or 5 CKO-to-PB chimeras had similar outcomes compared to controls. We conclude that the central IRF4/5 signaling is responsible for microglial activation and mediates stroke outcomes.

3.
Transl Stroke Res ; 14(5): 776-789, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-35906327

RESUMEN

Post-menopausal women become vulnerable to stroke and have poorer outcomes and higher mortality than age-matched men, and previous studies suggested that sex chromosomes play a vital role in mediating stroke sensitivity in the aged. It is unknown if this is due to effects of the X or Y chromosome. The present study used the XY* mouse model (with four genotypes: XX and XO gonadal females and XY and XXY gonadal males) to compare the effect of the X vs. Y chromosome compliment in stroke. Aged (18-20 months) and gonadectomized young (8-12 weeks) mice were subjected to a 60-min middle cerebral artery occlusion. Infarct volume and behavioral deficits were quantified 3 days after stroke. Microglial activation and infiltration of peripheral leukocytes in the aged ischemic brain were assessed by flow cytometry. Plasma inflammatory cytokine levels by ELISA, and brain expression of two X chromosome-linked genes, KDM6A and KDM5C by immunochemistry, were also examined. Both aged and young XX and XXY mice had worse stroke outcomes compared to XO and XY mice, respectively; however, the difference between XX vs. XXY and XO vs. XY aged mice was minimal. Mice with two copies of the X chromosome showed more robust microglial activation, higher brain-infiltrating leukocytes, elevated plasma cytokine levels, and enhanced co-localization of KDM6A and KDM5C with Iba1+ cells after stroke than mice with one X chromosome. The number of X chromosomes mediates stroke sensitivity in aged mice, which might be processed through the X chromosome-linked genes and the inflammatory responses.


Asunto(s)
Accidente Cerebrovascular , Cromosoma X , Masculino , Ratones , Femenino , Animales , Cromosoma X/genética , Cromosoma Y/genética , Accidente Cerebrovascular/genética , Genotipo , Citocinas/genética
4.
Aging (Albany NY) ; 14(15): 6047-6065, 2022 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-35963621

RESUMEN

Numerous neurochemical changes occur with aging and stroke mainly affects the elderly. Our previous study has found interferon regulatory factor 5 (IRF5) and 4 (IRF4) regulate neuroinflammation in young stroke mice. However, whether the IRF5-IRF4 regulatory axis has the same effect in aged brains is not known. In this study, aged (18-20-month-old), microglial IRF5 or IRF4 conditional knockout (CKO) mice were subjected to a 60-min middle cerebral artery occlusion (MCAO). Stroke outcomes were quantified at 3d after MCAO. Flow cytometry and ELISA were performed to evaluate microglial activation and immune responses. We found aged microglia express higher levels of IRF5 and lower levels of IRF4 than young microglia after stroke. IRF5 CKO aged mice had improved stroke outcomes; whereas worse outcomes were seen in IRF4 CKO vs. their flox controls. IRF5 CKO aged microglia had significantly lower levels of IL-1ß and CD68 than controls; whereas significantly higher levels of IL-1ß and TNF-α were seen in IRF4 CKO vs. control microglia. Plasma levels of TNF-α and MIP-1α were decreased in IRF5 CKO vs. flox aged mice, and IL-1ß/IL-6 levels were increased in IRF4 CKO vs. controls. The anti-inflammatory cytokines (IL-4/IL-10) levels were higher in IRF5 CKO, and lower in IRF4 CKO aged mice vs. their flox controls. IRF5 and IRF4 signaling drives microglial pro- and anti-inflammatory response respectively; microglial IRF5 is detrimental and IRF4 beneficial for aged mice in stroke. IRF5-IRF4 axis is a promising target for developing new, effective therapeutic strategies for the cerebral ischemia.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular , Animales , Infarto de la Arteria Cerebral Media , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Ratones , Microglía/metabolismo , Accidente Cerebrovascular/genética , Factor de Necrosis Tumoral alfa/farmacología
5.
Biol Sex Differ ; 12(1): 66, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930441

RESUMEN

BACKGROUND: Sex differences in COVID-19 are increasingly recognized globally. Although infection rates are similar between the sexes, men have more severe illness. The mechanism underlying these sex differences is unknown, but a differential immune response to COVID-19 has been implicated in several recent studies. However, how sex differences shape the immune response to COVID-19 remains understudied. METHODS: We collected demographics and blood samples from over 600 hospitalized patients diagnosed with COVID-19 from May 24th 2020 to April 28th, 2021. These patients were divided into two cohorts: Cohort 1 was further classified into three groups based on the severity of the disease (mild, moderate and severe); Cohort 2 patients were longitudinally followed at three time points from hospital admission (1 day, 7 days, and 14 days). MultiPlex and conventional ELISA were used to examine inflammatory mediator levels in the plasma in both cohorts. Flow cytometry was conducted to examine leukocyte responses in Cohort 2. RESULTS: There were more COVID+ males in the total cohort, and the mortality rate was higher in males vs. females. More male patients were seen in most age groups (in 10-year increments), and in most ethnic groups. Males with severe disease had significantly higher levels of pro-inflammatory cytokines (IL-6, IL-8, MCP-1) than females; levels of IL-8, GRO, sCD40L, MIP-1ß, MCP-1 were also significantly higher in severe vs. mild or control patients in males but not in females. Females had significantly higher anti-inflammatory cytokine IL-10 levels at 14 days compared to males, and the level of IL-10 significantly increased in moderate vs. the control group in females but not in males. At 7 days and 14 days, males had significantly more circulating neutrophils and monocytes than females; however, B cell numbers were significantly higher in females vs. males. CONCLUSION: Sex differences exist in hospitalized patients with acute COVID-19 respiratory tract infection. Exacerbated inflammatory responses were seen in male vs. female patients, even when matched for disease severity. Males appear to have a more robust innate immune response, and females mount a stronger adaptive immune response to COVID-19 respiratory tract infection.


Asunto(s)
COVID-19 , Inmunidad , COVID-19/inmunología , Femenino , Humanos , Masculino , Factores Sexuales
6.
J Neurosci Methods ; 364: 109359, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34537225

RESUMEN

BACKGROUND: Microglia play a central role in neuroinflammation in various CNS diseases.Neonatal microglial culture has been extensively used to in vitro study microglial activation; however, as many neuroinflammatory diseases occur in the elderly, the neonatal microglial culture may not fully replicate the aged microglial activity seen in these diseases. NEW METHOD: Primary microglia from both 18-24-month-old and P0-P4 C57BL/6 mice were cultured simultaneously. Morphology and activation profiles of the two age groups of microglia were examined following ischemic stimulation, by ELISA, RT-PCR, live microscopy, immunocytochemistry, and Western blotting. RESULTS: We showed that aged microglia had larger cell bodies, more cytoplasmic inclusions, and enhanced phagocytosis than neonatal microglia. Cytokine production in these cells exhibited heterogeneity either after or before ischemic stimulation. The baseline expression of microglial marker CD11b was significantly higher in aged vs. neonatal cells; ischemic stimulation increased the expression in neonatal vs. aged microglia only in males but not in females. COMPARISON WITH EXISTING METHODS: Previous primary microglia cultures have been limited to using neonatal/adult cells. This method is complementary to exiting methods and works for aged microglia, and does not suffer from potential limitations due to filtering artifacts. The protocol renders microglial culture no need for meningeal/hippocampal removal prior to brain tissue dissociation, and compares microglia between males vs. females, and between the aged vs. neonates. CONCLUSIONS: We concluded that neonatal microglial culture is not appropriate for those in vitro studies that mimic the neuroinflammatory central nervous system disorders occurring in the elderly, in which case the aged microglial culture should be applied, and sex differences should be considered.


Asunto(s)
Factores de Edad , Microglía , Caracteres Sexuales , Animales , Técnicas de Cultivo de Célula , Femenino , Hipocampo , Masculino , Ratones , Ratones Endogámicos C57BL , Fagocitosis
7.
Stroke ; 52(10): 3362-3373, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34353112

RESUMEN

Background and Purpose: CD200 (cluster of differentiation 200), a highly glycosylated protein primarily expressed on neurons in the central nervous system, binds with its receptor CD200R to form an endogenous inhibitory signal against immune responses. However, little is known about the effect of neuronal CD200 signaling in cerebral ischemia. The aim of this study was to investigate how neuronal CD200 signaling impacts poststroke inflammation and the ischemic injury. Methods: CD200 tma1lf/fl:Thy1CreER mice were treated with tamoxifen to induce conditional gene knockout (ICKO) of neuronal CD200. The mice were subjected to a 60-minute transient middle cerebral artery occlusion. Stroke outcomes, apoptotic cell death, immune cell infiltration, microglia activation, and other inflammatory profiles were evaluated at 3 and 7 days after stroke. Results: Infarct volumes were significantly larger, and behavioral deficits more severe in ICKO versus control mice at 3 days after middle cerebral artery occlusion. Terminal deoxynucleotidyl transferase dUTP nick end labeling assay also revealed a significant increase in apoptotic neuronal death in CD200 ICKO mice. An enhancement in lymphocytic infiltration and microglial proinflammatory responses were revealed by flow cytometry at 3 and 7 days after stroke in ICKO mice, accompanied by an increased microglial phagocytosis activity. Plasma proinflammatory cytokine (TNFα [tumor necrosis factor alpha] and IL [interleukin]-1ß) levels significantly increased at 3 days, and IL-1ß/IL-6 levels increased at 7 days in ICKO versus control animals. ICKO led to significantly lower baseline level of CD200 both in brain and plasma. Conclusions: Neuronal CD200 inhibits proinflammatory responses and is protective against stroke injury.


Asunto(s)
Antígenos CD/análisis , Accidente Cerebrovascular Isquémico/prevención & control , Neuronas/fisiología , Neuroprotección , Accidente Cerebrovascular/prevención & control , Animales , Antígenos CD/genética , Apoptosis , Citocinas/metabolismo , Inmunidad Celular , Infarto de la Arteria Cerebral Media/complicaciones , Inflamación/etiología , Inflamación/prevención & control , Accidente Cerebrovascular Isquémico/psicología , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Infiltración Neutrófila , Transducción de Señal , Resultado del Tratamiento
8.
Aging (Albany NY) ; 13(12): 15917-15941, 2021 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-34139671

RESUMEN

The G-quadruplex (G4-DNA or G4) is a secondary DNA structure formed by DNA sequences containing multiple runs of guanines. While it is now firmly established that stabilized G4s lead to enhanced genomic instability in cancer cells, whether and how G4s contribute to genomic instability in brain cells is still not clear. We previously showed that, in cultured primary neurons, small-molecule G4 stabilizers promote formation of DNA double-strand breaks (DSBs) and downregulate the Brca1 gene. Here, we determined if G4-dependent Brca1 downregulation is unique to neurons or if the effects in neurons also occur in astrocytes and microglia. We show that primary neurons, astrocytes and microglia basally exhibit different G4 landscapes. Stabilizing G4-DNA with the G4 ligand pyridostatin (PDS) differentially modifies chromatin structure in these cell types. Intriguingly, PDS promotes DNA DSBs in neurons, astrocytes and microglial cells, but fails to downregulate Brca1 in astrocytes and microglia, indicating differences in DNA damage and repair pathways between brain cell types. Taken together, our findings suggest that stabilized G4-DNA contribute to genomic instability in the brain and may represent a novel senescence pathway in brain aging.


Asunto(s)
Astrocitos/metabolismo , G-Cuádruplex , Microglía/metabolismo , Neuronas/metabolismo , Aminoquinolinas/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/ultraestructura , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Secuencia de Bases , Línea Celular , Cromatina/efectos de los fármacos , Cromatina/metabolismo , Cromatina/ultraestructura , Daño del ADN , Ratones , Microglía/efectos de los fármacos , Microglía/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Ácidos Picolínicos/farmacología , Regiones Promotoras Genéticas/genética , Ratas
9.
J Neuroinflammation ; 18(1): 70, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712031

RESUMEN

BACKGROUND: Stroke is a sexually dimorphic disease. Previous studies have found that young females are protected against ischemia compared to males, partially due to the protective effect of ovarian hormones, particularly estrogen (E2). However, there are also genetic and epigenetic effects of X chromosome dosage that contribute to stroke sensitivity and neuroinflammation after injury, especially in the aged. Genes that escape from X chromosome inactivation (XCI) contribute to sex-specific phenotypes in many disorders. Kdm5c and kdm6a are X escapee genes that demethylate H3K4me3 and H3K27me3, respectively. We hypothesized that the two demethylases play critical roles in mediating the stroke sensitivity. METHODS: To identify the X escapee genes involved in stroke, we performed RNA-seq in flow-sorted microglia from aged male and female wild type (WT) mice subjected to middle cerebral artery occlusion (MCAO). The expression of these genes (kdm5c/kdm6a) were confirmed in four core genotypes (FCG) mice and in post-mortem human stroke brains by immunohistochemistry (IHC), Western blot, and RT-PCR. Chromatin immunoprecipitation (ChIP) assays were conducted to detect DNA levels of inflammatory interferon regulatory factor (IRF) 4/5 precipitated by histone H3K4 and H3K27 antibodies. Manipulation of kdm5c/kdm6a expression with siRNA or lentivirus was performed in microglial culture, to determine downstream pathways and examine the regulatory roles in inflammatory cytokine production. RESULTS: Kdm5c and kdm6a mRNA levels were significantly higher in aged WT female vs. male microglia, and the sex difference also existed in ischemic brains from FCG mice and human stroke patients. The ChIP assay showed the IRF 4/5 had higher binding levels to demethylated H3K4 or H3K27, respectively, in female vs. male ischemic microglia. Knockdown or over expression of kdm5c/kdm6a with siRNA or lentivirus altered the methylation of H3K4 or H3K27 at the IRF4/5 genes, which in turn, impacted the production of inflammatory cytokines. CONCLUSIONS: The KDM-Histone-IRF pathways are suggested to mediate sex differences in cerebral ischemia. Epigenetic modification of stroke-related genes constitutes an important mechanism underlying the ischemic sexual dimorphism.


Asunto(s)
Epigénesis Genética/genética , Inflamación/genética , Accidente Cerebrovascular Isquémico/genética , Caracteres Sexuales , Cromosoma X/genética , Anciano , Anciano de 80 o más Años , Animales , Inmunoprecipitación de Cromatina , Citocinas/biosíntesis , Femenino , Genotipo , Histona Demetilasas/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , ARN Interferente Pequeño/genética , Transducción de Señal/genética
10.
Cells ; 10(2)2021 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-33573200

RESUMEN

BACKGROUND: Interferon Regulatory Factor (IRF) 5 and 4 play a determinant role in regulating microglial pro- and anti-inflammatory responses to cerebral ischemia. How microglial IRF5 and IRF4 signaling are activated has been elusive. We hypothesized that interleukin-1 receptor associated kinase 4 (IRAK4) phosphorylates and activates IRF5 and IRF4 in ischemic microglia. We aimed to explore the upstream signals of the two IRFs, and to determine how the IRAK4-IRF signaling regulates the expression of inflammatory mediators, and impacts neuropathology. METHODS: Spontaneously Immortalized Murine (SIM)-A9 microglial cell line, primary microglia and neurons from C57BL/6 WT mice were cultured and exposed to oxygen-glucose deprivation (OGD), followed by stimulation with LPS or IL-4. An IRAK4 inhibitor (ND2158) was used to examine IRAK4's effects on the phosphorylation of IRF5/IRF4 and the impacts on neuronal morphology by co-immunoprecipitation (Co-IP)/Western blot, ELISA, and immunofluorescence assays. RESULTS: We confirmed that IRAK4 formed a Myddosome with MyD88/IRF5/IRF4, and phosphorylated both IRFs, which subsequently translocated into the nucleus. Inhibition of IRAK4 phosphorylation quenched microglial pro-inflammatory response primarily, and increased neuronal viability and neurite lengths after ischemia. CONCLUSIONS: IRAK4 signaling is critical for microglial inflammatory responses and a potential therapeutic target for neuroinflammatory diseases including cerebral ischemia.


Asunto(s)
Isquemia Encefálica/genética , Mediadores de Inflamación/metabolismo , Factores Reguladores del Interferón/metabolismo , Microglía/metabolismo , Animales , Isquemia Encefálica/patología , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Transgénicos , Fosforilación , Transducción de Señal
11.
Proc Natl Acad Sci U S A ; 117(3): 1742-1752, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31892541

RESUMEN

Microglial activation plays a central role in poststroke inflammation and causes secondary neuronal damage; however, it also contributes in debris clearance and chronic recovery. Microglial pro- and antiinflammatory responses (or so-called M1-M2 phenotypes) coexist and antagonize each other throughout the disease progress. As a result of this balance, poststroke immune responses alter stroke outcomes. Our previous study found microglial expression of interferon regulatory factor 5 (IRF5) and IRF4 was related to pro- and antiinflammatory responses, respectively. In the present study, we genetically modified the IRF5 and IRF4 signaling to explore their roles in stroke. Both in vitro and in vivo assays were utilized; IRF5 or IRF4 small interfering RNA (siRNA), lentivirus, and conditional knockout (CKO) techniques were employed to modulate IRF5 or IRF4 expression in microglia. We used a transient middle cerebral artery occlusion model to induce stroke and examined both acute and chronic stroke outcomes. Poststroke inflammation was evaluated with flow cytometry, RT-PCR, MultiPlex, and immunofluorescence staining. An oscillating pattern of the IRF5-IRF4 regulatory axis function was revealed. Down-regulation of IRF5 signaling by siRNA or CKO resulted in increased IRF4 expression, enhanced M2 activation, quenched proinflammatory responses, and improved stroke outcomes, whereas down-regulation of IRF4 led to increased IRF5 expression, enhanced M1 activation, exacerbated proinflammatory responses, and worse functional recovery. Up-regulation of IRF4 or IRF5 by lentivirus induced similar results. We conclude that the IRF5-IRF4 regulatory axis is a key determinant in microglial activation. The IRF5-IRF4 regulatory axis is a potential therapeutic target for neuroinflammation and ischemic stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Inflamación/metabolismo , Factores Reguladores del Interferón/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/metabolismo , Factores Reguladores del Interferón/genética , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño , Transducción de Señal , Transcriptoma
12.
Artículo en Inglés | MEDLINE | ID: mdl-31993106

RESUMEN

CD200 and its receptor, CD200R, constitutes an endogenous inhibitory signaling, and is being increasingly recognized in studies of various central nervous system (CNS) disorders. Emerging data have demonstrated that neuronal CD200 binds to CD200R to modulate immune responses to pathogenic stimuli. However, on which component of the immune response that CD200-CD200R signaling acts is not well understood. In this review, we focused on cellular expression of the signaling, the effects on immune cell activation, and the function in pathological procedures of neurodegenerative diseases, in both clinical and experimental disease models. Essential functions of CD200-CD200R interaction and the treatment relevance have been elaborated. Immune responses to diseases under the control of CD200-CD200R axis were also discussed in the review.

13.
Contrast Media Mol Imaging ; 2018: 1693513, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30538612

RESUMEN

Fluorine-19 (19F) magnetic resonance imaging (MRI) has the potential for a wide range of in vivo applications but is limited by lack of flexibility in exogenous probe formulation. Most 19F MRI probes are composed of perfluorocarbons (PFCs) or perfluoropolyethers (PFPEs) with intrinsic properties which limit formulation options. Hydrophilic organofluorine molecules can provide more flexibility in formulation options. We report herein a hyperfluorinated hydrophilic organoflourine, ET1084, with ∼24 wt. % 19F content. It dissolves in water and aqueous buffers to give solutions with ≥8 M 19F. 19F MRI phantom studies at 9.4T employing a 10-minute multislice multiecho (MSME) scan sequence show a linear increase in signal-to-noise ratio (SNR) with increasing concentrations of the molecule and a detection limit of 5 mM. Preliminary cytotoxicity and genotoxicity assessments suggest it is safe at concentrations of up to 20 mM.


Asunto(s)
Medios de Contraste/química , Flúor , Fluorocarburos/química , Imagen por Resonancia Magnética/métodos , Medios de Contraste/toxicidad , Fluorocarburos/toxicidad , Interacciones Hidrofóbicas e Hidrofílicas , Límite de Detección , Fantasmas de Imagen , Relación Señal-Ruido , Solubilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...