Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 337
Filtrar
1.
Am J Cardiol ; 125(12): 1906-1912, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32331711

RESUMEN

To determine the impact of radiation therapy (XRT) in addition to trastuzumab (TZB) adjuvant chemotherapy for HER2+ breast cancer on left ventricular systolic function, we assessed demographics, oncologic treatment history including XRT exposure, and serial measurements of left ventricular ejection fraction (LVEF) in 135 consecutively identified women receiving TZB for treatment of adjuvant breast cancer. Longitudinal mixed effects models were fit to identify baseline to treatment changes in LVEF among those receiving TZB with or without concomitant anthracycline or XRT. Women averaged 53 ± 3 years in age, 77% were white, 62% patients had 1 or more cardiovascular risk factors at baseline, and mean duration of TZB was 11 ± 5 months. Seventy-seven women were treated with XRT and received between 4000 and 5500 cGy of radiation. The LVEF declined by an average of 3.4% after 1 year for those in the study. Relative to baseline upon completion of adjuvant TZB, LVEF remained reduced for those receiving anthracycline with or without XRT (p=0.002 for both), or XRT alone (p=0.002), but not in those without these therapies. Amongst patients treated only with XRT and TZB, LVEF declined 3.1% on average in those with left-sided disease and 6.9% on average in those with right-sided disease (p= 0.06, p= 0.008 respectively). Among women receiving TZB for adjuvant treatment of HER-2 positive breast cancer, the administration of XRT, anthracycline, or the combination of the 2 is associated with a persistent post-treatment as opposed to a temporary treatment related decline in LVEF.


Asunto(s)
Antraciclinas/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Enfermedades Cardiovasculares/etiología , Volumen Sistólico , Trastuzumab/uso terapéutico , Quimioterapia Adyuvante , Femenino , Humanos , Persona de Mediana Edad , Receptor ErbB-2 , Factores de Riesgo , Volumen Sistólico/efectos de los fármacos , Volumen Sistólico/efectos de la radiación
2.
J Pharmacol Exp Ther ; 371(1): 121-129, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31320494

RESUMEN

Cyclodextrins (CDs) have a variety of uses from acting as excipients to aiding the ability of lipid soluble drugs to cross the blood-brain barrier (BBB). They are being investigated as an active pharmaceutical ingredient, most recently for the treatment of Niemann-Pick disease, a lysosomal storage disease. Cyclodextrins are helpful in animal models and human subjects/patients afflicted with Neimann-Pick disease, including improving the neurologic component of the disease. The improvement in brain disease by intravenous administration implies that CDs can cross the BBB; however, there are only a few studies that have directly addressed this. In the current studies, multiple-time regression analysis indicated that 2-hydroxypropyl-ß-cyclodextrin [Kleptose (Klep)] radioactively labeled with 14C (C-Klep) crossed the BBB at a slow rate by a nonsaturable mechanism consistent with transcellular diffusion. However, the rate of transport varied greatly by the brain region with no detectable uptake by the spinal cord; additionally, many regions rapidly reached equilibrium between the brain and blood. The presence of a brain-to-blood efflux system was also detected and much of the C-Klep did not completely cross the BBB, but loosely adhered to the luminal surface of brain endothelial cells. Peripheral tissues also took up C-Klep, with the kidney taking up the most, which is consistent with renal clearance. In conclusion, we demonstrated minimal uptake of the ß-cyclodextrin Kleptose by the brain with accumulation being affected by efflux and reversible luminal binding. SIGNIFICANCE STATEMENT: This cyclodextrin, which produces therapeutic effects on the central nervous system after peripheral administration, penetrates the BBB poorly. Uptake by the brain to a therapeutic level will likely be difficult to achieve without giving high peripheral doses, bypassing the BBB, or otherwise altering penetration into the brain.


Asunto(s)
2-Hidroxipropil-beta-Ciclodextrina/farmacocinética , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Animales , Masculino , Ratones
3.
Int J Obes (Lond) ; 42(3): 391-397, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28990588

RESUMEN

OBJECTIVE: Resistance at the brain receptors for leptin and insulin has been associated with increased feeding, obesity and cognitive impairments. The causal agent for central resistance is unknown but could be derived from the blood. Here we postulate whether hypertriglyceridemia, the major dyslipidemia of the metabolic syndrome, could underlie central leptin and insulin resistance. DESIGN: We used radioactively labeled triglycerides to measure blood-brain barrier (BBB) penetration, western blots to measure receptor activation, and feeding and cognitive tests to assess behavioral endpoints. RESULTS: Human CSF was determined to contain triglycerides, a finding previously unclear. The radioactive triglyceride triolein readily crossed the BBB and centrally administered triolein and peripherally administered lipids induced in vivo leptin and/or insulin resistance at hypothalamic receptors. Central triolein blocked the satiety effect of centrally administered leptin. Decreasing serum triglycerides with gemfibrozil improved both learning and memory inversely proportionate to triglyceride levels. CONCLUSIONS: Triglycerides cross the blood-brain barrier rapidly, are found in human cerebrospinal fluid, and induce central leptin and insulin receptor resistance, decreasing satiety and cognition.


Asunto(s)
Antígenos CD/metabolismo , Barrera Hematoencefálica/metabolismo , Resistencia a la Insulina/fisiología , Leptina/metabolismo , Receptor de Insulina/metabolismo , Triglicéridos/metabolismo , Anciano , Animales , Cognición/efectos de los fármacos , Femenino , Gemfibrozilo/farmacología , Humanos , Leptina/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Respuesta de Saciedad/efectos de los fármacos , Triglicéridos/sangre , Triglicéridos/líquido cefalorraquídeo , Trioleína/metabolismo , Trioleína/farmacología
4.
Prostate Cancer Prostatic Dis ; 20(1): 20-27, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27645128

RESUMEN

BACKGROUND: Epidemiologic and in vitro studies suggest that SLCO-encoded organic anion transporting polypeptide (OATP) transporters influence the response of prostate cancer (PCa) to androgen deprivation by altering intratumor androgens. We have previously shown that castration-resistant metastases express multiple SLCO transporters at significantly higher levels than primary PCa, suggesting that OATP-mediated steroid transport is biologically relevant in advanced disease. However, whether OATP-mediated steroid transport can actually modify prostate tumor androgen levels in vivo has never been demonstrated. METHODS: We sought to determine whether OATP-mediated steroid transport can measurably alter PCa androgen levels in vivo. We evaluated the uptake of dehydroepiandrosterone (DHEAS), E1S and testosterone in LNCaP cells engineered to express OATP1B1, 1B3, 2B1 or 4A1. We measured the uptake via administration of tritiated steroids to castrate mice bearing vector control or OATP1B1-, 2B1- or 4A1-expressing xenografts. We treated tumor-bearing mice with DHEAS and testosterone at physiologically relevant levels and measured intratumor accumulation of administered steroids by mass spectrometry. RESULTS: OATP1B1- and 2B-expressing xenografts each showed a threefold increase in tritiated-DHEAS uptake vs vector controls (P=0.002 and P=0.036, respectively). At circulating DHEAS levels similar to those in abiraterone-treated men (~15 µg dl-1), OATP1B1- and 2B1-expressing xenografts showed a 3.9-fold (P=0.057) and 1.9-fold (P=0.048) increase in tumor accumulation of DHEAS and a 1.6-fold (P=0.057) and 2.7-fold (P=0.095) increase in DHEA, respectively. At the substantial circulating testosterone levels found in eugonadal men, a consistent effect of OATP1B1, 2B1 or 4A1 on testosterone uptake in vivo was not detected. CONCLUSIONS: OATP transporters measurably alter DHEAS uptake and intratumor androgen levels in prostate tumors in vivo, even at circulating androgen levels achieved in abiraterone-treated patients. These novel data emphasize the continued need to inhibit ligand-mediated androgen receptor signaling in PCa tumors, and support prospective evaluation of studies designed to test inhibition of OATP-mediated DHEAS uptake and utilization.


Asunto(s)
Transportadores de Anión Orgánico/metabolismo , Neoplasias de la Próstata/metabolismo , Esteroides/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Masculino , Ratones , Transportadores de Anión Orgánico/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Neuroscience ; 319: 206-20, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-26777891

RESUMEN

Exposure to blast overpressure (BOP) is associated with behavioral, cognitive, and neuroimaging abnormalities. We investigated the dynamic responses of cortical vasculature and its relation to microglia/macrophage activation in mice using intravital two-photon microscopy following mild blast exposure. We found that blast caused vascular dysfunction evidenced by microdomains of aberrant vascular permeability. Microglial/macrophage activation was specifically associated with these restricted microdomains, as evidenced by rapid microglial process retraction, increased ameboid morphology, and escape of blood-borne Q-dot tracers that were internalized in microglial/macrophage cell bodies and phagosome-like compartments. Microdomains of cortical vascular disruption and microglial/macrophage activation were also associated with aberrant tight junction morphology that was more prominent after repetitive (3×) blast exposure. Repetitive, but not single, BOPs also caused TNFα elevation two weeks post-blast. In addition, following a single BOP we found that aberrantly phosphorylated tau rapidly accumulated in perivascular domains, but cleared within four hours, suggesting it was removed from the perivascular area, degraded, and/or dephosphorylated. Taken together these findings argue that mild blast exposure causes an evolving CNS insult that is initiated by discrete disturbances of vascular function, thereby setting the stage for more protracted and more widespread neuroinflammatory responses.


Asunto(s)
Traumatismos por Explosión/patología , Lesiones Encefálicas/patología , Macrófagos/patología , Microglía/patología , Animales , Barrera Hematoencefálica/patología , Western Blotting , Encéfalo/irrigación sanguínea , Encéfalo/patología , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Microscopía Intravital , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/patología
6.
Brain Behav Immun ; 44: 1-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25172555

RESUMEN

Neuroimmunology is concerned with the relations between the central nervous and immune systems and with the mechanisms that drive those relations. The blood-brain barrier (BBB) employs mechanisms that both separate and connect these two systems. In fact, the relative immune privilege of the central nervous system (CNS) is largely attributable to the BBB's ability to prevent the unregulated exchange of immune cells and their secretions between the CNS and blood. Having separated the two systems, the BBB then participates in mechanisms that allow them to influence, communicate, and interact with one another. Likewise, the BBB itself is influenced by immune events that are occurring in the periphery and in the CNS so that these three components (the BBB, the immune system, and the CNS) form neuroimmune axes that adapt to physiological and pathological conditions. To date, four major themes have emerged by which the BBB participates in these neuroimmune axes. The first of these four, the formation of the barrier, acts to separate the immune and central nervous systems. The other three themes provide mechanisms for re-establishing communication: response of the BBB to immunomodulatory molecules (e.g., prostaglandins, cytokines, chemokines, nitric oxide) secreted by immune and CNS cells; the controlled, regulated exchange of chemokines, cytokines, and immune cells between the CNS and the blood (i.e., transport across the BBB); the secretion of immunomodulatory molecules by the BBB, often in a polarized fashion. Taken together, these mechanisms reveal the BBB to be a dynamic, interactive, and adaptable interface between the immune system and the CNS, separating them on the one hand and fostering their interactions on the other hand, adjusting to physiological changes, while being a target for disease processes. This review examines specific examples by which the BBB plays an interactive, defining role in neuroimmunology.


Asunto(s)
Barrera Hematoencefálica/inmunología , Neuroinmunomodulación , Animales , Citocinas/inmunología , Humanos , Óxido Nítrico/inmunología , Prostaglandinas/inmunología
7.
Peptides ; 62: 197-202, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25278492

RESUMEN

Alpha-synuclein (α-Syn), a small protein with multiple physiological and pathological functions, is one of the dominant proteins found in Lewy Bodies, a pathological hallmark of Lewy body disorders, including Parkinson's disease (PD). More recently, α-Syn has been found in body fluids, including blood and cerebrospinal fluid, and is likely produced by both peripheral tissues and the central nervous system. Exchange of α-Syn between the brain and peripheral tissues could have important pathophysiologic and therapeutic implications. However, little is known about the ability of α-Syn to cross the blood-brain barrier (BBB). Here, we found that radioactively labeled α-Syn crossed the BBB in both the brain-to-blood and the blood-to-brain directions at rates consistent with saturable mechanisms. Low-density lipoprotein receptor-related protein-1 (LRP-1), but not p-glycoprotein, may be involved in α-Syn efflux and lipopolysaccharide (LPS)-induced inflammation could increase α-Syn uptake by the brain by disrupting the BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Inflamación/mortalidad , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Transporte Biológico , Barrera Hematoencefálica/fisiología , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Cuerpos de Lewy/metabolismo , Cuerpos de Lewy/patología , Lipopolisacáridos/toxicidad , Lipoproteínas LDL , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Enfermedad de Parkinson/fisiopatología , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo
8.
Cardiorenal Med ; 3(4): 265-82, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24474955

RESUMEN

The prevalence of the cardiorenal metabolic syndrome (CRS) is increasing in parallel with obesity, type 2 diabetes mellitus, Alzheimer's disease, and other forms of dementia. Along with metabolic, inflammatory, and immunological abnormalities, there is maladaptive structural remodeling of the heart, kidney, and brain. The term 'diabetic cognopathy' (DC) may be used when discussing functional and structural changes in the brain of the diabetic patient. DC likely represents an advanced form of these changes in the brain that evolve with increasing duration of the CRS and subsequent clinical diabetes. We posit that DC develops due to a convergence of aging, genetic and lifestyle abnormalities (overnutrition and lack of exercise), which result in multiple injurious metabolic and immunologic toxicities such as dysfunctional immune responses, oxidative stress, inflammation, insulin resistance, and dysglycemia (systemically and in the brain). These converging abnormalities may lead to endothelial blood-brain barrier tight junction/adherens junction (TJ/AJ) complex remodeling and microglia activation, which may result in neurodegeneration, impaired cognition, and dementia. Herein, we describe the brain ultrastructural changes evolving from a normal state to maladaptive remodeling in rodent models of CRS including microglia activation/polarization and attenuation and/or loss of the TJ/AJ complexes, pericytes and astrocytes of the neurovascular unit. Further, we discuss the potential relationship between these structural changes and the development of DC, potential therapeutic strategies, and future directions.

9.
Neuroscience ; 226: 89-100, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22986172

RESUMEN

The blood-brain barrier is a restrictive interface between the brain parenchyma and the intravascular compartment. Tight junctions contribute to the integrity of the blood-brain barrier. Hypoxic-ischemic damage to the blood-brain barrier could be an important component of fetal brain injury. We hypothesized that increases in blood-brain barrier permeability after ischemia depend upon the duration of reperfusion and that decreases in tight junction proteins are associated with the ischemia-related impairment in blood-brain barrier function in the fetus. Blood-brain barrier function was quantified with the blood-to-brain transfer constant (K(i)) and tight junction proteins by Western immunoblot in fetal sheep at 127 days of gestation without ischemia, and 4, 24, or 48 h after ischemia. The largest increase in K(i) (P<0.05) was 4 h after ischemia. Occludin and claudin-5 expressions decreased at 4 h, but returned toward control levels 24 and 48 h after ischemia. Zonula occludens-1 and -2 decreased after ischemia. Inverse correlations between K(i) and tight junction proteins suggest that the decreases in tight junction proteins contribute to impaired blood-brain barrier function after ischemia. We conclude that impaired blood-brain barrier function is an important component of hypoxic-ischemic brain injury in the fetus, and that increases in quantitatively measured barrier permeability (K(i)) change as a function of the duration of reperfusion after ischemia. The largest increase in permeability occurs 4 h after ischemia and blood-brain barrier function improves early after injury because the blood-brain barrier is less permeable 24 and 48 than 4 h after ischemia. Changes in the tight junction molecular composition are associated with increases in blood-brain barrier permeability after ischemia.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Feto/fisiología , Daño por Reperfusión/fisiopatología , Proteínas de Uniones Estrechas/biosíntesis , Uniones Estrechas/metabolismo , Algoritmos , Ácidos Aminoisobutíricos , Animales , Volumen Sanguíneo/fisiología , Western Blotting , Encéfalo/embriología , Isquemia Encefálica/fisiopatología , Arterias Carótidas/fisiología , Claudina-1/biosíntesis , Claudina-5/biosíntesis , Densitometría , Electroencefalografía , Eritrocitos/fisiología , Femenino , Ocludina/biosíntesis , Ovinos , Tecnecio
10.
Physiol Behav ; 105(1): 145-9, 2011 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21669216

RESUMEN

The blood-brain barrier (BBB) presents a tremendous challenge for the delivery of drugs to the central nervous system (CNS). This includes drugs that target brain receptors for the treatment of obesity and anorexia. Strategic drug delivery to brain (SDDB) is an approach that considers in depth the relations among the BBB, the candidate therapeutic, the CNS target, and the disease state to be treated. Here, we illustrate principles of SDDB with two different approaches to developing drugs based on leptin. In normal body weight humans and in non-obese rodents, leptin is readily transported across the BBB and into the CNS where it inhibits feeding and enhances thermogenesis. However, in obesity, the transport of leptin across the BBB is impaired, resulting in a resistance to leptin. As a result, it is difficult to treat obesity with leptin or its analogs that depend on the leptin transporter for access to the CNS. To treat obesity, we developed a leptin agonist modified by the addition of pluronic block copolymers (P85-leptin). P85-leptin retains biological activity and is capable of crossing the BBB by a mechanism that is not dependent on the leptin transporter. As such, P85-leptin is able to cross the BBB of obese mice at a rate similar to that of native leptin in lean mice. To treat anorexia, we developed a leptin antagonist modified by pegylation (PEG-MLA) that acts primarily by blocking the BBB transporter for endogenous, circulating leptin. This prevents blood-borne, endogenous leptin from entering the CNS, essentially mimicking the leptin resistance seen in obesity, and resulting in a significant increase in adiposity. These examples illustrate two strategies in which an understanding of the interactions among the BBB, CNS targets, and candidate therapeutics under physiologic and diseased conditions can be used to develop drugs effective for the treatment of brain disease.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Leptina/análogos & derivados , Leptina/administración & dosificación , Animales , Encéfalo/metabolismo , Diseño de Fármacos , Ratones
11.
Int J Androl ; 34(1): 55-68, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20403060

RESUMEN

Testosterone regulates energy metabolism and skeletal muscle mass in males, but the molecular mechanisms are not fully understood. This study investigated the response of skeletal muscle to castration and testosterone replacement in 8-week-old male mice. Using microarray analyses of mRNA levels in gastrocnemius muscle, 91 genes were found to be negatively regulated by testosterone and 68 genes were positively regulated. The mRNA levels of the insulin signalling suppressor molecule Grb10 and the glycogen synthesis inhibitors, protein phosphatase inhibitor-1 and phosphorylase kinase-γ, were negatively regulated by testosterone. The insulin-sensitive glucose and amino acid transporters, Glut3 and SAT2, the lipodystrophy gene, Lpin1 and protein targeting to glycogen were positively regulated. These changes would be expected to increase nutrient availability and sensing within skeletal muscle, increase metabolic rate and carbohydrate utilization and promote glycogen accumulation. The observed positive regulation of atrogin-1 (Fbxo32) by testosterone could be explained by the phosphorylation of Akt and Foxo3a, as determined by Western blotting. Testosterone prevented the castration-induced increase in interleukin-1α, the decrease in interferon-γ and the atrophy of the levator ani muscle, which were all correlated with testosterone-regulated gene expression. These findings identify specific mechanisms by which testosterone may regulate skeletal muscle glucose and protein metabolism.


Asunto(s)
Regulación de la Expresión Génica , Glucosa/metabolismo , Músculo Esquelético/metabolismo , Proteínas/metabolismo , Testosterona/administración & dosificación , Acetiltransferasas/genética , Animales , Proteína Adaptadora GRB10/genética , Expresión Génica , Perfilación de la Expresión Génica , Transportador de Glucosa de Tipo 3/genética , Interferón gamma/genética , Interleucina-1alfa/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Proteínas Musculares/genética , Proteínas Nucleares/genética , Orquiectomía , Fosfatidato Fosfatasa , Fosforilasa Quinasa/genética , ARN Mensajero/análisis , Distribución Aleatoria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Ligasas SKP Cullina F-box/genética , Transducción de Señal , Testosterona/sangre
12.
Exp Neurol ; 218(1): 162-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19422824

RESUMEN

The cellular form of the prion protein (PrP(c)) is necessary for the development of prion diseases and is a highly conserved protein that may play a role in neuroprotection. PrP(c) is found in both blood and cerebrospinal fluid and is likely produced by both peripheral tissues and the central nervous system (CNS). Exchange of PrP(c) between the brain and peripheral tissues could have important pathophysiologic and therapeutic implications, but it is unknown whether PrP(c) can cross the blood-brain barrier (BBB). Here, we found that radioactively labeled PrP(c) crossed the BBB in both the brain-to-blood and blood-to-brain directions. PrP(c) was enzymatically stable in blood and in brain, was cleared by liver and kidney, and was sequestered by spleen and the cervical lymph nodes. Circulating PrP(c) entered all regions of the CNS, but uptake by the lumbar and cervical spinal cord, hypothalamus, thalamus, and striatum was particularly high. These results show that PrP(c) has bidirectional, saturable transport across the BBB and selectively targets some CNS regions. Such transport may play a role in PrP(c) function and prion replication.


Asunto(s)
Barrera Hematoencefálica/fisiología , Priones/metabolismo , Animales , Permeabilidad Capilar , Isótopos de Yodo/farmacocinética , Masculino , Ratones , Priones/farmacocinética , Transporte de Proteínas/fisiología , Factores de Tiempo , Distribución Tisular
13.
Neurobiol Aging ; 28(8): 1170-8, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16846666

RESUMEN

Accumulation of toxic amyloid-beta (Abeta)-peptide is suggested to cause oxidative stress in Alzheimer's disease (AD) brain, and decrease the content of polyunsaturated fatty acids (PUFA) in neuronal membrane lipids. The senescence accelerated prone mice (SAMP8) have age-related increases in the level of hippocampal Abeta-peptide, learning and memory deficits, and a shorter lifespan than their controls. The effects of age-related oxidative damage on PUFA content in membrane phospholipids (PL), and alpha-tocopherol concentration were investigated in hippocampus and amygdala of 2-, 4-, 12-, and 18-month-old SAMP8 mice. In comparison to the younger SAMP8 mice, the hippocampus of the 12-month-old mice contained lower proportions of docosahexaenoic acid (DHA) in phosphatidylserine (PS) and phosphatidylinositol (PI), and higher proportions of arachidonic acid (AA) in PS. Their amygdala contained a lower proportion of AA in phosphatidylcholine (PC). In the hippocampus of the oldest age group, the proportions of DHA in PS, and AA in PC and PI were higher than in the younger age groups. At 2 months of age, the amygdala contained a higher concentration of alpha-tocopherol than the hippocampus, but this difference between the two brain regions was lost with aging. The oldest age group contained the highest concentration of alpha-tocopherol, indicating a protection against oxidative damage of PUFA in brain membrane phospholipids.


Asunto(s)
Envejecimiento/genética , Envejecimiento/metabolismo , Encéfalo/metabolismo , Peroxidación de Lípido , Factores de Edad , Análisis de Varianza , Animales , Encéfalo/efectos de los fármacos , Química Encefálica , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Mutantes Neurológicos , Oligodesoxirribonucleótidos Antisentido/farmacología , alfa-Tocoferol/metabolismo
14.
Aging Male ; 8(3-4): 135-40, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16390735

RESUMEN

Frailty occurs in aging males for a variety of reasons. It is less common in males than females. Diseases which are particularly associated with frailty are diabetes mellitus, atherosclerosis, anemia and chronic obstructive pulmonary disease. Insulin resistance syndrome plays a pathogenetic role in the "fat-frail" syndrome. Sarcopenia occurs predominantly because of hormone deficiency and cytokine excess. Pain and anorexia are also associated with frailty. Stem cell research represents a potential promise for the treatment of frailty.


Asunto(s)
Envejecimiento/fisiología , Anciano Frágil , Factores de Edad , Anciano , Envejecimiento/patología , Citocinas , Progresión de la Enfermedad , Femenino , Evaluación Geriátrica , Humanos , Resistencia a la Insulina , Masculino , Estudios Seroepidemiológicos
15.
Neuroscience ; 128(1): 143-53, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15450361

RESUMEN

Human immunodeficiency virus (HIV)-1 within the CNS induces neuro-acquired immunodeficiency syndrome and acts as a reservoir for reinfection of peripheral tissues. HIV-1 crosses the blood-brain barrier (BBB) within infected immune cells and as cell-free virus by a CD4-independent mechanism. Which proteins control free virus transport across the BBB are unknown, but work with wheatgerm agglutinin (WGA) and heparin suggests that heparan sulfate proteoglycans, sialic acid, and N-acetyl-beta-D-glucosaminyl acid bind HIV-1. Here, we found that an HIV-1 T-tropic virus was taken up by mouse brain endothelial cells in vitro and crossed the BBB in vivo and could be effluxed as intact virus. Uptake was stimulated by WGA and protamine sulfate (PS) and inhibited by heparin. BBB uptake of virus involved four distinguishable binding sites: i) reversible cell surface binding involving gp120 and sensitive to PS/heparin but insensitive to WGA; internalization with a ii) WGA-sensitive site binding gp120 and iii) a PS/heparin-sensitive site not involving gp120; iv) membrane incorporation not affected by WGA, heparin, or PS. In conclusion, binding, internalization, and membrane incorporation are separately regulated steps likely determining whether HIV-1 is incorporated into brain endothelial cells, transported across them, or returned to the circulation.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Endocitosis/fisiología , Endotelio Vascular/metabolismo , VIH-1/metabolismo , Animales , Barrera Hematoencefálica/virología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/virología , Heparina/metabolismo , Masculino , Ratones , Protaminas/metabolismo , Aglutininas del Germen de Trigo/metabolismo
16.
Neuroscience ; 126(4): 915-26, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15207326

RESUMEN

The senescence-accelerated mouse (SAM) is a murine model of accelerated senescence that was established using phenotypic selection. The SAMP series includes nine substrains, each of which exhibits characteristic disorders. SAMP8 is known to exhibit age-dependent learning and memory deficits. In our previous study, we reported that brains from 12-month-old SAMP8 have greater protein oxidation, as well as lipid peroxidation, compared with brains from 4-month-old SAMP8 mice. In order to investigate the relation between age-associated oxidative stress on specific protein oxidation and age-related learning and memory deficits in SAMP8, we used proteomics to identify proteins that are expressed differently and/or modified oxidatively in aged SAMP8 brains. We report here that in 12 month SAMP8 mice brains the expressions of neurofilament triplet L protein, lactate dehydrogenase 2 (LDH-2), heat shock protein 86, and alpha-spectrin are significantly decreased, while the expression of triosephosphate isomerase (TPI) is increased compared with 4-month-old SAMP8 brains. We also report that the specific protein carbonyl levels of LDH-2, dihydropyrimidinase-like protein 2, alpha-spectrin and creatine kinase, are significantly increased in the brain of 12-month-old SAMP8 mice when compared with the 4-month-old SAMP8 brain. These findings are discussed in reference to the effect of specific protein oxidation and changes of expression on potential mechanisms of abnormal alterations in metabolism and neurochemicals, as well as to the learning and memory deficits in aged SAMP8 mice.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Regulación de la Expresión Génica/fisiología , Estrés Oxidativo/fisiología , Proteínas/metabolismo , Envejecimiento/genética , Animales , Western Blotting/métodos , Electroforesis en Gel Bidimensional/métodos , Masculino , Espectrometría de Masas/métodos , Ratones , Ratones Endogámicos , Ratones Mutantes Neurológicos , Proteómica/métodos
17.
Neuroscience ; 121(2): 487-92, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14522007

RESUMEN

Brain to blood transport is believed to be a major determinant of the amount of amyloid beta protein (AbetaP) found in brain. Impaired efflux has been suggested as a mechanism by which AbetaP can accumulate in the CNS and so lead to Alzheimer's disease (AD). To date, however, no study of the efflux of the form of AbetaP most relevant to AD, AbetaP1-42, has been conducted, even though a single amino acid substitution in AbetaP can greatly alter efflux. Here, we examined the efflux of AbetaP mouse1-42, mouse1-40, human1-42, and human1-40 in young CD-1, young senesence accelerated mouse (SAM) P8, and aged SAMP8 mice. The SAMP8 mouse with aging spontaneously overproduces AbetaP and develops cognitive impairments reversed by AbetaP-directed antibody or phosphorothioate antisense oligonucleotide. CD-1 mice transported all forms of AbetaP, although mouse1-42 and human1-40 were transported faster than the other forms. There was a decrease in the saturable transport of mouse1-42 in SAMP8 mice regardless of age. Efflux of mouse1-40 and human1-42 was only by a non-saturable mechanism in young SAMP8 mice and their efflux was totally absent in aged SAMP8 mice. These differences in the efflux of the various forms of AbetaP among the three groups of mice supports the hypothesis that impaired efflux is an important factor in the accumulation of AbetaP in the CNS.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Fragmentos de Péptidos/metabolismo , Transporte de Proteínas/fisiología , Envejecimiento , Animales , Transporte Biológico , Sangre/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Mutantes Neurológicos , Ensayo de Unión Radioligante , Homología de Secuencia de Aminoácido , Especificidad de la Especie
18.
J Pharmacol Exp Ther ; 301(3): 878-83, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12023514

RESUMEN

Human obesity may be caused by a resistance to circulating leptin. Evidence from rodents and humans suggests that a major component of this resistance is an impairment in the ability of the blood-brain barrier (BBB) to transport leptin from the blood to the brain. One potential way to bypass the BBB is by administering leptin into the intrathecal (i.t.) space. To be effective, i.t. leptin would have to move caudally from the site of injection, enter the cranium, and reach the hypothalamic arcuate nucleus at the base of the pituitary fossa. However, many substances, especially small, lipid-soluble molecules, do not diffuse far from the site of i.t. injection but are resorbed back into blood. To determine whether i.t. leptin can move caudally, we injected leptin conjugated to diethylenetriaminepentaacetic acid (DTPA) and labeled with (68)Ga (G-Ob) into the lumbar space of three baboons. We also studied unconjugated DTPA labeled with (68)Ga, which did not move up the spinal cord but rapidly appeared in blood after i.t. injection. In contrast, G-Ob steadily moved toward the cranium and had reached the hypothalamus 91 and 139 min after i.t. injection in two baboons. We estimated the concentration of leptin in the hypothalamic region to be at least 8 ng/ml, which is about 40 times higher than cerebrospinal fluid levels in normal weight humans and about 4 times higher than the highest level ever recorded after the peripheral administration of leptin. In a third baboon, the leptin neither moved caudally nor appeared in the blood. We conclude that leptin administered i.t. can reach the hypothalamus in therapeutic concentrations, although there is considerable individual variation.


Asunto(s)
Hipotálamo/diagnóstico por imagen , Leptina/administración & dosificación , Tomografía Computarizada de Emisión , Animales , Mapeo Encefálico/métodos , Femenino , Hipotálamo/metabolismo , Inyecciones Espinales/métodos , Inyecciones Espinales/estadística & datos numéricos , Leptina/farmacocinética , Masculino , Papio , Ácido Pentético/administración & dosificación , Ácido Pentético/farmacocinética , Tomografía Computarizada de Emisión/métodos , Tomografía Computarizada de Emisión/estadística & datos numéricos
19.
Life Sci ; 69(23): 2765-73, 2001 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-11720081

RESUMEN

Leptin regulates body adiposity by decreasing feeding and increasing thermogenesis. Obese humans and some obese rodents are resistant to peripherally administered leptin, suggesting a defect in the transport of leptin across the blood-brain barrier (BBB). Defective transport of exogenous leptin occurs in some models of obesity, but in other models transport is normal. This shows that factors other than obesity are associated with impairment of leptin transport across the BBB. In order to further investigate these factors, we determined leptin transport in rats made obese by lesioning of the ventromedial hypothalamus (VMH), paraventricular nucleus (PVN), or posterodorsal amygdala (PDA). These regions all contain leptin receptors and lesions there induce obesity and hyperleptinemia and alter the levels of many feeding hormones which might participate in leptin transporter regulation. We measured the uptake of radioactively labeled leptin by the BBB by multiple-time regression analysis which divides uptake into a reversible phase (Vi, e.g., receptor/transporter binding to the brain endothelial cell) and an irreversible phase (Ki, complete transport across the BBB). Leptin uptake was not affected in rats with VMH lesions. No significant change occurred in the entry rate (Ki) for any group, although Ki declined by over 35% in rats with PVN lesions. Decreased uptake was observed in rats with PVN lesions and with PDA lesions. This was primarily due to a reduced Vi (about 21% for the PDA). This decreased uptake is most likely explained by decreased binding of leptin to the brain endothelial cell, which could be because of decreased binding by either receptors or transporters. This suggests that some of the feeding hormones controlled by the PVN and PDA may participate in regulating leptin uptake by the BBB.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Barrera Hematoencefálica/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Amígdala del Cerebelo/lesiones , Animales , Femenino , Hipotálamo/lesiones , Obesidad/etiología , Núcleo Hipotalámico Paraventricular/lesiones , Ratas , Ratas Long-Evans , Aumento de Peso
20.
Life Sci ; 69(14): 1683-9, 2001 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-11589508

RESUMEN

LY303870 (LY) is a non-peptide neurokinin-1 receptor antagonist that has effects on the brain after peripheral administration. We determined whether LY given by intravenous (iv) injection can cross the blood-brain barrier (BBB). Multiple-time regression analysis showed the unidirectional influx rate (Ki) from blood to brain for LY labeled with tritium to be 6.41+/-0.85 microl/g-min and influx was inhibited by unlabeled LY. HPLC and mass spectrometry showed LY was stable in blood and brain. LY reached a brain/serum ratio of 190+/-12 microl/g with about 0.07% of the injected dose entering each gram of brain. These results show that LY is transported across the BBB from serum into brain by a saturable system.


Asunto(s)
Barrera Hematoencefálica , Indoles/farmacocinética , Antagonistas del Receptor de Neuroquinina-1 , Piperidinas/farmacocinética , Animales , Encéfalo/metabolismo , Cromatografía Líquida de Alta Presión , Indoles/administración & dosificación , Indoles/sangre , Indoles/metabolismo , Inyecciones Intravenosas , Masculino , Espectrometría de Masas , Piperidinas/administración & dosificación , Piperidinas/sangre , Piperidinas/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...