Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 231
Filter
1.
Hum Brain Mapp ; 44(10): 3998-4010, 2023 07.
Article in English | MEDLINE | ID: mdl-37162380

ABSTRACT

There has been growing attention on the effect of COVID-19 on white-matter microstructure, especially among those that self-isolated after being infected. There is also immense scientific interest and potential clinical utility to evaluate the sensitivity of single-shell diffusion magnetic resonance imaging (MRI) methods for detecting such effects. In this work, the performances of three single-shell-compatible diffusion MRI modeling methods are compared for detecting the effect of COVID-19, including diffusion-tensor imaging, diffusion-tensor decomposition of orthogonal moments and correlated diffusion imaging. Imaging was performed on self-isolated patients at the study initiation and 3-month follow-up, along with age- and sex-matched controls. We demonstrate through simulations and experimental data that correlated diffusion imaging is associated with far greater sensitivity, being the only one of the three single-shell methods to demonstrate COVID-19-related brain effects. Results suggest less restricted diffusion in the frontal lobe in COVID-19 patients, but also more restricted diffusion in the cerebellar white matter, in agreement with several existing studies highlighting the vulnerability of the cerebellum to COVID-19 infection. These results, taken together with the simulation results, suggest that a significant proportion of COVID-19 related white-matter microstructural pathology manifests as a change in tissue diffusivity. Interestingly, different b-values also confer different sensitivities to the effects. No significant difference was observed in patients at the 3-month follow-up, likely due to the limited size of the follow-up cohort. To summarize, correlated diffusion imaging is shown to be a viable single-shell diffusion analysis approach that allows us to uncover opposing patterns of diffusion changes in the frontal and cerebellar regions of COVID-19 patients, suggesting the two regions react differently to viral infection.


Subject(s)
COVID-19 , White Matter , COVID-19/diagnostic imaging , COVID-19/pathology , Diffusion Tensor Imaging , Feasibility Studies , White Matter/diagnostic imaging , White Matter/ultrastructure , Frontal Lobe/diagnostic imaging , Frontal Lobe/ultrastructure , Humans , Male , Female , Young Adult , Adult , Middle Aged , Aged
2.
Nature ; 601(7891): 139-143, 2022 01.
Article in English | MEDLINE | ID: mdl-34880495

ABSTRACT

The abnormal aggregation of TAR DNA-binding protein 43 kDa (TDP-43) in neurons and glia is the defining pathological hallmark of the neurodegenerative disease amyotrophic lateral sclerosis (ALS) and multiple forms of frontotemporal lobar degeneration (FTLD)1,2. It is also common in other diseases, including Alzheimer's and Parkinson's. No disease-modifying therapies exist for these conditions and early diagnosis is not possible. The structures of pathological TDP-43 aggregates are unknown. Here we used cryo-electron microscopy to determine the structures of aggregated TDP-43 in the frontal and motor cortices of an individual who had ALS with FTLD and from the frontal cortex of a second individual with the same diagnosis. An identical amyloid-like filament structure comprising a single protofilament was found in both brain regions and individuals. The ordered filament core spans residues 282-360 in the TDP-43 low-complexity domain and adopts a previously undescribed double-spiral-shaped fold, which shows no similarity to those of TDP-43 filaments formed in vitro3,4. An abundance of glycine and neutral polar residues facilitates numerous turns and restricts ß-strand length, which results in an absence of ß-sheet stacking that is associated with cross-ß amyloid structure. An uneven distribution of residues gives rise to structurally and chemically distinct surfaces that face external densities and suggest possible ligand-binding sites. This work enhances our understanding of the molecular pathogenesis of ALS and FTLD and informs the development of diagnostic and therapeutic agents that target aggregated TDP-43.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Cryoelectron Microscopy , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/ultrastructure , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Amino Acid Sequence , Amyloid beta-Peptides/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Frontal Lobe/ultrastructure , Humans , Male , Middle Aged , Motor Cortex/metabolism , Motor Cortex/pathology , Motor Cortex/ultrastructure , Mutation
3.
J Comp Neurol ; 529(16): 3676-3708, 2021 11.
Article in English | MEDLINE | ID: mdl-34259349

ABSTRACT

In the current study, we examined the number, distribution, and aspects of the neurochemical identities of infracortical white matter neurons, also termed white matter interstitial cells (WMICs), in the brains of a southern lesser galago (Galago moholi), a black-capped squirrel monkey (Saimiri boliviensis boliviensis), and a crested macaque (Macaca nigra). Staining for neuronal nuclear marker (NeuN) revealed WMICs throughout the infracortical white matter, these cells being most dense close to inner cortical border, decreasing in density with depth in the white matter. Stereological analysis of NeuN-immunopositive cells revealed estimates of approximately 1.1, 10.8, and 37.7 million WMICs within the infracortical white matter of the galago, squirrel monkey, and crested macaque, respectively. The total numbers of WMICs form a distinct negative allometric relationship with brain mass and white matter volume when examined in a larger sample of primates where similar measures have been obtained. In all three primates studied, the highest densities of WMICs were in the white matter of the frontal lobe, with the occipital lobe having the lowest. Immunostaining revealed significant subpopulations of WMICs containing neuronal nitric oxide synthase (nNOS) and calretinin, with very few WMICs containing parvalbumin, and none containing calbindin. The nNOS and calretinin immunopositive WMICs represent approximately 21% of the total WMIC population; however, variances in the proportions of these neurochemical phenotypes were noted. Our results indicate that both the squirrel monkey and crested macaque might be informative animal models for the study of WMICs in neurodegenerative and psychiatric disorders in humans.


Subject(s)
Brain Chemistry/physiology , Brain/cytology , Galagidae/physiology , Macaca/physiology , Neurons/ultrastructure , Saimiri/physiology , White Matter/cytology , Animals , Calbindin 2/metabolism , Calbindins/metabolism , Cell Count , Frontal Lobe/cytology , Frontal Lobe/ultrastructure , Immunohistochemistry , Male , Neurons/chemistry , Nitric Oxide Synthase Type I/metabolism , Occipital Lobe/cytology , Occipital Lobe/ultrastructure , Parvalbumins/metabolism , Species Specificity , White Matter/chemistry
4.
J Stroke Cerebrovasc Dis ; 30(6): 105766, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33866227

ABSTRACT

BACKGROUND AND PURPOSE: In subarachnoid hemorrhage (SAH), impairments in motor and cognitive functions may occur and continue in later periods. MicroRNAs (miRNAs) are small non-coding RNAs that can directly or indirectly affect synaptic reconstruction. mir-132, mir-134, and mir-138 are the leading miRNAs that can be effective on some neurological functions through its effects on synaptic plasticity in the relevant brain areas. In our study, it was aimed to determine the levels of miRNAs in the hippocampus and frontal lobe of rats exposed to different environmental conditions after the experimental SAH. METHODS: SAH was created using the cisterna magna double blood-injection method. Brain tissues were collected at different times after the last blood injection. Rats were grouped according to the different environmental conditions in which they were kept. Expression levels of miRNAs were performed by qPCR and ultrastructural changes in samples were determined by transmission electron microscopy (TEM). RESULTS: After SAH, miR-132, miR-134, and miR-138 expressions in the frontal lobes of rats increased in impoverished environment on the 7th day and in the enriched environment on the 14th day. It was observed that the myelin and microtubule structures in the axons that were disrupted after SAH were more organized and stable in the enriched environment. CONCLUSIONS: After SAH, different environmental conditions may affect the miRNA levels associated with synaptic plasticity and microtubule organization in the frontal lobe, and this might have some effects especially on cognitive and motor functions related to this brain area.


Subject(s)
Frontal Lobe/metabolism , Hippocampus/metabolism , MicroRNAs/metabolism , Microtubules/metabolism , Neuronal Plasticity , Neurons/metabolism , Subarachnoid Hemorrhage/metabolism , Animals , Disease Models, Animal , Female , Frontal Lobe/ultrastructure , Hippocampus/pathology , MicroRNAs/genetics , Microtubules/genetics , Microtubules/ultrastructure , Neurons/ultrastructure , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/genetics , Subarachnoid Hemorrhage/pathology
5.
J Gerontol A Biol Sci Med Sci ; 75(2): 236-243, 2020 01 20.
Article in English | MEDLINE | ID: mdl-30285083

ABSTRACT

Physical activity has positive effects on brain health and cognitive function throughout the life span. Thus far, few studies have examined the effects of physical activity on white matter microstructure and psychomotor speed within the same, population-based sample (critical if conclusions are to extend to the wider population). Here, using diffusion tensor imaging and a simple reaction time task within a relatively large population-derived sample (N = 399; 18-87 years) from the Cambridge Centre for Ageing and Neuroscience (Cam-CAN), we demonstrate that physical activity mediates the effect of age on white matter integrity, measured with fractional anisotropy. Higher self-reported daily physical activity was associated with greater preservation of white matter in several frontal tracts, including the genu of corpus callosum, uncinate fasciculus, external capsule, and anterior limb of the internal capsule. We also show that the age-related slowing is mediated by white matter integrity in the genu. Our findings contribute to a growing body of work, suggesting that a physically active lifestyle may protect against age-related structural disconnection and slowing.


Subject(s)
Aging/physiology , Cognition/physiology , Diffusion Magnetic Resonance Imaging , Exercise/physiology , Frontal Lobe/ultrastructure , Psychomotor Performance/physiology , White Matter/ultrastructure , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Anisotropy , England , Female , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Reaction Time/physiology , Surveys and Questionnaires
6.
Psicothema (Oviedo) ; 30(2): 159-164, mayo 2018.
Article in English | IBECS | ID: ibc-173330

ABSTRACT

BACKGROUND: Since the mid 19th century, cognitive and behavioral neurosciences have attempted to find the neurological bases of intellectual abilities. During the early 20th century the psychometric concept of "intelligence" was coined; and toward the end of the 20th century the neuropsychological concept of "executive functions" was introduced. Controversies, however, remain about the unity or heterogeneity of so-called executive functions. METHOD: It is proposed that two major executive functions could be separated: metacognitive - or intellectual - and emotional/motivational. A similar distinction has been suggested by several authors. Standard definitions of intelligence implicitly assume that executive functions represent the fundamental components of intelligence. RESULTS: Research has demonstrated that, if considered as a whole, executive functions only partially correspond to the psychometric concept of intelligence; whereas some specific executive functions clearly correspond to intelligence, some others do not involve intelligence. CONCLUSIONS: If using a major distinction between metacognitive -or simply "intellectual"-executive functions, and emotional/ motivational - or simply non-intellectual-executive functions, it becomes evident that general intelligence can be equated with metacognitive executive functions but not with emotional/ motivational executive functions


ANTECEDENTES: desde mediados del siglo XIX las neurociencias cognitivas y comportamentales han intentado hallar las bases neurológicas de las habilidades intelectuales. A comienzos del siglo XX se acuña el concepto psicométrico de "inteligencia"; y hacia finales del siglo XX se introduce el concepto neuropsicológico de "funciones ejecutivas". Sin embargo, continúan existiendo controversias acerca de la unidad o heterogeneidad de las llamadas funciones ejecutivas. MÉTODO: se propone que es posible distinguir dos funciones ejecutivas básicas: metacognitivas - o intelectuales - y emocionales/motivacionales. Diversos autores han propuesto una distinción similar. Las definiciones estándar de inteligencia implícitamente asumen que las funciones ejecutivas representan los componentes fundamentales de la inteligencia. RESULTADOS: la investigación ha demostrado que, consideradas en conjunto, las funciones ejecutivas corresponden solo en forma parcial al concepto psicométrico de inteligencia; en tanto que algunas funciones ejecutivas claramente corresponden a inteligencia, otras no se asocian con la inteligencia. CONCLUSIONES: utilizando la distinción entre funciones ejecutivas metacognitivas "o simplemente "intelectuales" -y "funciones ejecutivas emocionales/motivacionales" o simplemente "no intelectuales"-, se hace evidente que la inteligencia general se puede equiparar con las funciones ejecutivas metacognitivas, pero no con las funciones ejecutivas emocionales/motivacionales


Subject(s)
Humans , Adolescent , Young Adult , Adult , Executive Function/physiology , Intelligence/physiology , Thinking/physiology , Neurodegenerative Diseases/physiopathology , Metacognition/physiology , Models, Psychological , Brain Injuries/physiopathology , Brain Injuries/psychology , Emotions/physiology , Frontal Lobe/physiology , Frontal Lobe/ultrastructure , Motivation/physiology , Neurodegenerative Diseases/psychology , Prefrontal Cortex/physiology , Prefrontal Cortex/ultrastructure , Verbal Behavior/physiology
7.
Psicothema ; 30(2): 159-164, 2018 05.
Article in English | MEDLINE | ID: mdl-29694315

ABSTRACT

BACKGROUND: Since the mid 19th century, cognitive and behavioral neurosciences have attempted to find the neurological bases of intellectual abilities. During the early 20th century the psychometric concept of "intelligence" was coined; and toward the end of the 20th century the neuropsychological concept of "executive functions" was introduced. Controversies, however, remain about the unity or heterogeneity of so-called executive functions. METHODS: It is proposed that two major executive functions could be separated: metacognitive -or intelectual- and emotional/motivational. A similar distinction has been suggested by several authors. Standard definitions of intelligence implicitly assume that executive functions represent the fundamental components of intelligence. RESULTS: Research has demonstrated that, if considered as a whole, executive functions only partially correspond to the psychometric concept of intelligence; whereas some specific executive functions clearly correspond to intelligence, some others do not involve intelligence. CONCLUSIONS: If using a major distinction between metacognitive -or simply "intellectual"-executive functions, and emotional/ motivational -or simply non-intellectual-executive functions, it becomes evident that general intelligence can be equated with metacognitive executive functions but not with emotional/ motivational executive functions.


Subject(s)
Executive Function , Intelligence , Metacognition , Models, Psychological , Adolescent , Adult , Brain Injuries/physiopathology , Brain Injuries/psychology , Child , Emotions/physiology , Executive Function/physiology , Frontal Lobe/physiology , Frontal Lobe/ultrastructure , Humans , Intelligence/physiology , Memory, Short-Term/physiology , Metacognition/physiology , Motivation/physiology , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/psychology , Prefrontal Cortex/physiology , Prefrontal Cortex/ultrastructure , Psychological Tests , Thinking/physiology , Verbal Behavior/physiology
8.
Acta Neuropathol Commun ; 6(1): 26, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29615128

ABSTRACT

The amyloid cascade hypothesis posits that the initiating event in Alzheimer's disease (AD) is the aggregation and deposition of the ß-amyloid (Aß) peptide, which is a proteolytic cleavage product of the amyloid precursor protein (APP). Mounting evidence suggests that the formation and spread of prion-like Aß aggregates during AD may contribute to disease progression. Inoculation of transgenic mice that overexpress APP with pre-formed Aß aggregates results in the prion-like induction of cerebral Aß deposition. To determine whether Aß deposition can also be induced when physiological APP levels are present in the brain, we inoculated AppNL-F mice, a knock-in model of AD that avoids potential artifacts associated with APP overexpression, with Aß aggregates derived from the brains of AD patients or transgenic mice. In all cases, induced Aß deposition was apparent in the corpus callosum, olfactory bulb, and meningeal blood vessels of inoculated mice at 130-150 days post-inoculation, whereas uninoculated and buffer-inoculated animals exhibited minimal or no Aß deposits at these ages. Interestingly, despite being predominantly composed of protease-resistant Aß42 aggregates, the induced parenchymal Aß deposits were largely diffuse and were unreactive to an amyloid-binding dye. These results demonstrate that APP overexpression is not a prerequisite for the prion-like induction of cerebral Aß deposition. Accordingly, spreading of Aß deposition may contribute to disease progression in AD patients.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Frontal Lobe/metabolism , Plaque, Amyloid/pathology , Prions/metabolism , Protein Aggregation, Pathological/etiology , Aged , Alzheimer Disease/genetics , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Female , Frontal Lobe/ultrastructure , Gene Expression Regulation/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Middle Aged , Mutation/genetics , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Protein Aggregates/physiology , Silver Staining
9.
Brain Struct Funct ; 223(1): 255-265, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28779306

ABSTRACT

Atomic force microscopy (AFM) is emerging as an innovative tool to phenotype the brain. This study demonstrates the utility of AFM to determine nanomechanical and nanostructural features of the murine dorsolateral frontal cortex from weaning to adulthood. We found an increase in tissue stiffness of the primary somatosensory cortex with age, along with an increased cortical mechanical heterogeneity. To characterize the features potentially responsible for this heterogeneity, we applied AFM scan mode to directly image the topography of thin sections of the primary somatosensory cortical layers II/III, IV and V/VI. Topographical mapping of the cortical layers at successive ages showed progressive smoothing of the surface. Topographical images were also compared with histochemically derived morphological information, which demonstrated the deposition of perineuronal nets, important extracellular components and markers of maturity. Our work demonstrates that high-resolution AFM images can be used to determine the nanostructural properties of cortical maturation, well beyond embryonic and postnatal development. Furthermore, it may offer a new method for brain phenotyping and screening to uncover topographical changes in early stages of neurodegenerative diseases.


Subject(s)
Brain Mapping , Frontal Lobe/growth & development , Frontal Lobe/ultrastructure , Microscopy, Atomic Force , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Biomechanical Phenomena , Biotin , Male , Mice , Mice, Inbred C57BL , Plant Lectins/metabolism , Receptors, N-Acetylglucosamine/metabolism
10.
Article in English | MEDLINE | ID: mdl-29079139

ABSTRACT

Clozapine, a representative atypical antipsychotic, has superior efficacy compared to other antipsychotic agents and is used for the treatment of severe psychotic disorders. Therefore, studies on its mechanisms of action are important for understanding the mechanisms of therapeutic approaches to psychosis. Adenosine monophosphate-activated protein kinase (AMPK) is a serine-threonine kinase that plays a major role in maintaining metabolic homeostasis. Unc-51-like kinase 1 (ULK1) and Beclin1 are downstream substrates of AMPK and activate the autophagic process. In this study, we examined the effects of clozapine on the AMPK-ULK1-Beclin1 signaling pathway and autophagy in the frontal cortex of the rat. Clozapine (10mg/kg) administration increased the immunoreactivity of p-Thr172-AMPKα in the rat frontal cortex at 1, 2, and 4h after injection, as we previously reported. The immunoreactivity of p-Ser317-ULK1 and p-Ser93-Beclin1 was also increased at 2 and 4h after clozapine injection. At the same time, the immunoreactivity of LC3-II and the Atg5-Atg12 conjugate, which indicate activation of autophagy, was increased. Transmission electron microscopy clearly showed an increase in autophagosome number in the rat frontal cortex at 2h after clozapine injection. To investigate the role of AMPK in clozapine-induced autophagy, the effects of intracerebroventricular injection of compound C, an AMPK inhibitor, were examined. Administration of compound C attenuated the clozapine-induced increase in ULK1 and Beclin1 phosphorylation, as well the protein levels of LC3-II and the Atg5-Atg12 conjugate in the frontal cortex. In summary, the results showed that clozapine activates autophagy through the AMPK-ULK1-Beclin1 signaling pathway in the frontal cortex of the rat.


Subject(s)
Antipsychotic Agents/pharmacology , Autophagy/drug effects , Clozapine/pharmacology , Frontal Lobe/drug effects , Adenylate Kinase/metabolism , Animals , Autophagy/physiology , Autophagy-Related Protein-1 Homolog/metabolism , Beclin-1/metabolism , Frontal Lobe/metabolism , Frontal Lobe/ultrastructure , Male , Microscopy, Electron, Transmission , Phosphorylation/drug effects , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Signal Transduction/drug effects
11.
Mol Cell Neurosci ; 86: 65-71, 2018 01.
Article in English | MEDLINE | ID: mdl-29180229

ABSTRACT

Doxorubicin, a commonly used anti-neoplastic agent, causes severe neurotoxicity. Doxorubicin promotes thinning of the brain cortex and accelerates brain aging, leading to cognitive impairment. Oxidative stress induced by doxorubicin contributes to cellular damage. In addition to mitochondria, peroxisomes also generate reactive oxygen species (ROS) and promote cell senescence. Here, we investigated if doxorubicin affects peroxisomal homeostasis in neurons. We demonstrate that the number of peroxisomes is increased in doxorubicin-treated neurons and in the brains of mice which underwent doxorubicin-based chemotherapy. Pexophagy, the specific autophagy of peroxisomes, is downregulated in neurons, and peroxisomes produce more ROS. 2-hydroxypropyl-ß-cyclodextrin (HPßCD), an activator of the transcription factor TFEB, which regulates expression of genes involved in autophagy and lysosome function, mitigates damage of pexophagy and decreases ROS production induced by doxorubicin. We conclude that peroxisome-associated oxidative stress induced by doxorubicin may contribute to neurotoxicity, cognitive dysfunction, and accelerated brain aging in cancer patients and survivors. Peroxisomes might be a valuable new target for mitigating neuronal damage caused by chemotherapy drugs and for slowing down brain aging in general.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Neurons/drug effects , Oxidative Stress/drug effects , Peroxisomes/drug effects , Animals , Cells, Cultured , Female , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Frontal Lobe/ultrastructure , Mice , Neurons/metabolism , Neurons/ultrastructure , Oxidative Stress/physiology , Peroxisomes/metabolism , Peroxisomes/ultrastructure , Rats , Reactive Oxygen Species/metabolism
12.
Int J Immunopathol Pharmacol ; 30(4): 371-382, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29171783

ABSTRACT

It has been reported that hyperhomocysteinemia (HHcy) is associated with neurodegenerative and cardiovascular diseases. However, little is known about brain histomorphology, neuronal organelles, and hairy enhancer of split ( hes) expression under HHcy. In this study, non-HHcy and HHcy induced by high-methionine diet in apolipoprotein E-deficient (Apo E-/-) mice were comparatively investigated. The histomorphology, ultrastructure, autophagosomes, apoptosis, and expression of proteins, HES1, HES5 and P62, were designed to assess the effects of HHcy on brain. The results showed that compared to the non-HHcy mice, the HHcy group had an increase in autophagosomes, vacuolization in mitochondria, and neuron apoptosis; treatment with folate and vitamin B12 reduced the extent of these lesions. However, the elementary histomorphology, the numbers of cortical neurons, and Nissl bodies had no significant difference between the HHcy and the non-HHcy groups or the group treated with folate and vitamin B12. Immunohistochemistry and immunofluorescence demonstrated a decrease in HES1- or HES5-positive neurons in the HHcy group when compared to the non-HHcy groups, wild-type, and Apo E-/- controls, or the HHcy mice with folate and vitamin B12 supplement. Western blots showed that HHcy induced a decreased expression of HES1 and HES5, or P62, in which the expression of HES1 and P62 was elevated by treating with folate and vitamin B12 supplement. These results suggest that HHcy-enhanced brain damage is associated with increased autophagy and neuronal apoptosis in Apo E-/- mice, in which downregulation of hes1 and hes5 is involved.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hyperhomocysteinemia/metabolism , Neurons/metabolism , Repressor Proteins/metabolism , Transcription Factor HES-1/metabolism , Animals , Apoptosis , Autophagy , Down-Regulation , Frontal Lobe/metabolism , Frontal Lobe/ultrastructure , Homocysteine/blood , Hyperhomocysteinemia/blood , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Microscopy, Electron, Transmission , Neurons/ultrastructure
13.
Bull Exp Biol Med ; 164(2): 285-292, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29177876

ABSTRACT

Permeability of the blood-brain barrier for protein fractions 50-100 kDa (PF50-100) of Cellex Daily preparation labeled with fluorescent tracer FITC and non-conjugated FITC were compared after intranasal administration of the preparations to healthy rats. Fluorimetrical analysis of the serum and cerebrospinal fluid samples showed that Cellex Daily PF50-100-FITC administered intranasally penetrated into the blood and cerebrospinal fluid with maximum accumulation in 2 h after administration and persists in the circulation for 24 h probably due to binding with plasma proteins. The differences in the kinetic profile of PF50-100-FITC and free FITC indirectly suggest that the major part of the preparation is not degraded within 24 h and FITC is probably not cleaved from the protein components of the preparation. In vivo fluorescence analysis showed significant fluorescent signal in the olfactory bulbs in 6 h after intranasal administration; hence, the preparation administered via this route can bypass the blood-brain barrier. Scanning laser confocal microscopy of rat brain sections confirmed penetration of the high-molecular weight protein fraction PF50-100-FITC into CNS structures. The most pronounced accumulation of the labeled drug was observed in the olfactory bulb in 6 and 12 h after administration. In contrast to free FITC administered in the control group, significant accumulation of PF50-100-FITC in the olfactory cortex and frontal cortex neurons with functionally active nuclei was observed in 6, 12 and 24 h after intranasal administration.


Subject(s)
Blood-Brain Barrier/metabolism , Frontal Lobe/metabolism , Nerve Tissue Proteins/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Olfactory Bulb/metabolism , Peptides/pharmacokinetics , Administration, Intranasal , Animals , Biological Availability , Biological Transport , Blood-Brain Barrier/ultrastructure , Fetus , Fluorescein-5-isothiocyanate/chemistry , Fluorometry , Frontal Lobe/ultrastructure , Luminescent Measurements , Male , Nerve Tissue Proteins/blood , Nerve Tissue Proteins/chemistry , Neuroprotective Agents/blood , Neuroprotective Agents/chemistry , Olfactory Bulb/ultrastructure , Peptides/blood , Peptides/chemistry , Rats , Rats, Wistar , Staining and Labeling/methods , Swine
14.
Alzheimers Res Ther ; 9(1): 54, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28750690

ABSTRACT

BACKGROUND: The apolipoprotein E ε4 (APOE4) genotype is a prominent late-onset Alzheimer's disease (AD) risk factor. ApoE4 disrupts memory function in rodents and may contribute to both plaque and tangle formation. METHODS: Coimmunoprecipitation and Western blot detection were used to determine: 1) the effects of select fragments from the apoE low-density lipoprotein (LDL) binding domain and recombinant apoE subtypes on amyloid beta (Aß)42-α7 nicotinic acetylcholine receptor (α7nAChR) interaction and tau phosphorylation in rodent brain synaptosomes; and 2) the level of Aß42-α7nAChR complexes in matched controls and patients with mild cognitive impairment (MCI) and dementia due to AD with known APOE genotypes. RESULTS: In an ex vivo study using rodent synaptosomes, apoE141-148 of the apoE promotes Aß42-α7nAChR association and Aß42-induced α7nAChR-dependent tau phosphorylation. In a single-blind study, we examined lymphocytes isolated from control subjects, patients with MCI and dementia due to AD with known APOE genotypes, sampled at two time points (1 year apart). APOE ε4 genotype was closely correlated with heightened Aß42-α7nAChR complex levels and with blunted exogenous Aß42 effects in lymphocytes derived from AD and MCI due to AD cases. Similarly, plasma from APOE ε4 carriers enhanced the Aß42-induced Aß42-α7nAChR association in rat cortical synaptosomes. The progression of cognitive decline in APOE ε4 carriers correlated with higher levels of Aß42-α7nAChR complexes in lymphocytes and greater enhancement by their plasma of Aß42-induced Aß42-α7nAChR association in rat cortical synaptosomes. CONCLUSIONS: Our data suggest that increased lymphocyte Aß42-α7nAChR-like complexes may indicate the presence of AD pathology especially in APOE ε4 carriers. We show that apoE, especially apoE4, promotes Aß42-α7nAChR interaction and Aß42-induced α7nAChR-dependent tau phosphorylation via its apoE141-148 domain. These apoE-mediated effects may contribute to the APOE ε4-driven neurodysfunction and AD pathologies.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Lymphocytes/metabolism , Peptide Fragments/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/pharmacology , Animals , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Dose-Response Relationship, Drug , Female , Frontal Lobe/ultrastructure , Humans , Lymphocytes/drug effects , Male , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptors, LDL/metabolism , Statistics as Topic , Synaptosomes/metabolism , Synaptosomes/ultrastructure , tau Proteins/metabolism
15.
Sci Transl Med ; 9(385)2017 04 12.
Article in English | MEDLINE | ID: mdl-28404863

ABSTRACT

Heterozygous mutations in the GRN gene lead to progranulin (PGRN) haploinsufficiency and cause frontotemporal dementia (FTD), a neurodegenerative syndrome of older adults. Homozygous GRN mutations, on the other hand, lead to complete PGRN loss and cause neuronal ceroid lipofuscinosis (NCL), a lysosomal storage disease usually seen in children. Given that the predominant clinical and pathological features of FTD and NCL are distinct, it is controversial whether the disease mechanisms associated with complete and partial PGRN loss are similar or distinct. We show that PGRN haploinsufficiency leads to NCL-like features in humans, some occurring before dementia onset. Noninvasive retinal imaging revealed preclinical retinal lipofuscinosis in heterozygous GRN mutation carriers. Increased lipofuscinosis and intracellular NCL-like storage material also occurred in postmortem cortex of heterozygous GRN mutation carriers. Lymphoblasts from heterozygous GRN mutation carriers accumulated prominent NCL-like storage material, which could be rescued by normalizing PGRN expression. Fibroblasts from heterozygous GRN mutation carriers showed impaired lysosomal protease activity. Our findings indicate that progranulin haploinsufficiency caused accumulation of NCL-like storage material and early retinal abnormalities in humans and implicate lysosomal dysfunction as a central disease process in GRN-associated FTD and GRN-associated NCL.


Subject(s)
Haploinsufficiency/physiology , Intercellular Signaling Peptides and Proteins/deficiency , Neuronal Ceroid-Lipofuscinoses/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Animals , Cells, Cultured , Frontal Lobe/metabolism , Frontal Lobe/ultrastructure , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Haploinsufficiency/genetics , Heterozygote , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lysosomes , Mice , Microscopy, Electron , Mutation/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Progranulins , Retina/metabolism , Retina/ultrastructure
16.
J Comp Neurol ; 525(9): 2175-2191, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28256708

ABSTRACT

The excitatory glutamatergic synapse is the principal site of communication between cortical pyramidal neurons and their targets, a key locus of action of many drugs, and highly vulnerable to dysfunction and loss in neurodegenerative disease. A detailed knowledge of the structure of these synapses in distinct cortical areas and across species is a prerequisite for understanding the anatomical underpinnings of cortical specialization and, potentially, selective vulnerability in neurological disorders. We used serial electron microscopy to assess the ultrastructural features of excitatory (asymmetric) synapses in the layers 2-3 (L2-3) neuropil of visual (V1) and frontal (FC) cortices of the adult mouse and compared findings to those in the rhesus monkey (V1 and lateral prefrontal cortex [LPFC]). Analyses of multiple ultrastructural variables revealed four organizational features. First, the density of asymmetric synapses does not differ between frontal and visual cortices in either species, but is significantly higher in mouse than in monkey. Second, the structural properties of asymmetric synapses in mouse V1 and FC are nearly identical, by stark contrast to the significant differences seen between monkey V1 and LPFC. Third, while the structural features of postsynaptic entities in mouse and monkey V1 do not differ, the size of presynaptic boutons are significantly larger in monkey V1. Fourth, both presynaptic and postsynaptic entities are significantly smaller in the mouse FC than in the monkey LPFC. The diversity of synaptic ultrastructural features demonstrated here have broad implications for the nature and efficacy of glutamatergic signaling in distinct cortical areas within and across species.


Subject(s)
Frontal Lobe/ultrastructure , Macaca mulatta/anatomy & histology , Mice/anatomy & histology , Synapses/ultrastructure , Visual Cortex/ultrastructure , Analysis of Variance , Animals , Female , Frontal Lobe/metabolism , Imaging, Three-Dimensional , Male , Microscopy, Immunoelectron , Neurons/metabolism , Neurons/ultrastructure , Neuropil/metabolism , Neuropil/ultrastructure , Presynaptic Terminals/ultrastructure , Species Specificity , Synapses/classification , Synapses/metabolism , Visual Cortex/metabolism
17.
Neuroreport ; 27(12): 916-22, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27341212

ABSTRACT

Mitochondrial dysfunction induced by mitochondria-related ß-amyloid (Aß) accumulation is increasingly being considered a novel risk factor for sporadic Alzheimer's disease pathophysiology. The close relationship between chronic sleep restriction (CSR) and cortical Aß elevation was confirmed recently. By assessing frontal cortical mitochondrial function (electron microscopy manifestation, cytochrome C oxidase concentration, ATP level, and mitochondrial membrane potential) and the levels of mitochondria-related Aß in 9-month-old adult male C57BL/6J mice subjected to CSR and as an environmental control (CO) group, we aimed to evaluate the association of CSR with mitochondrial dysfunction and mitochondria-related Aß accumulation. In this study, frontal cortical mitochondrial dysfunction was significantly more severe in CSR mice compared with CO animals. Furthermore, CSR mice showed higher mitochondria-associated Aß, total Aß, and mitochondria-related ß-amyloid protein precursor (AßPP) levels compared with CO mice. In the CSR model, mouse frontal cortical mitochondrial dysfunction was correlated with mitochondria-associated Aß and mitochondria-related AßPP levels. However, frontal cortical mitochondria-associated Aß levels showed no significant association with cortical total Aß and mitochondrial AßPP concentrations. These findings indicated that CSR-induced frontal cortical mitochondrial dysfunction and mitochondria-related Aß accumulation, which was closely related to mitochondrial dysfunction under CSR.


Subject(s)
Amyloid beta-Peptides/metabolism , Frontal Lobe/metabolism , Mitochondria/metabolism , Sleep Deprivation/metabolism , Animals , Frontal Lobe/ultrastructure , Male , Mice, Inbred C57BL , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism
18.
Cereb Cortex ; 26(6): 2689-2704, 2016 06.
Article in English | MEDLINE | ID: mdl-26045568

ABSTRACT

Most glutamatergic inputs in the neocortex originate from the thalamus or neocortical pyramidal cells. To test whether thalamocortical afferents selectively innervate specific cortical cell subtypes and surface domains, we investigated the distribution patterns of thalamocortical and corticocortical excitatory synaptic inputs in identified postsynaptic cortical cell subtypes using intracellular and immunohistochemical staining combined with confocal laser scanning and electron microscopic observations in 2 thalamorecipient sublayers, lower layer 2/3 (L2/3b) and lower layer 5 (L5b) of rat frontal cortex. The dendrites of GABAergic parvalbumin (PV) cells preferentially received corticocortical inputs in both sublayers. The somata of L2/3b PV cells received thalamic inputs in similar proportions to the basal dendritic spines of L2/3b pyramidal cells, whereas L5b PV somata were mostly innervated by cortical inputs. The basal dendrites of L2/3b pyramidal and L5b corticopontine pyramidal cells received cortical and thalamic glutamatergic inputs in proportion to their local abundance, whereas crossed-corticostriatal pyramidal cells in L5b exhibited a preference for thalamic inputs, particularly in their distal dendrites. Our data demonstrate an exquisite selectivity among thalamocortical afferents in which synaptic connectivity is dependent on the postsynaptic neuron subtype, cortical sublayer, and cell surface domain.


Subject(s)
Frontal Lobe/physiology , Neurons/physiology , Synapses/physiology , Thalamus/physiology , Animals , Frontal Lobe/ultrastructure , Glutamic Acid/metabolism , Immunohistochemistry , Male , Microscopy, Confocal , Microscopy, Electron , Neuroanatomical Tract-Tracing Techniques , Neurons/ultrastructure , Rats, Wistar , Thalamus/ultrastructure
19.
Sleep ; 39(4): 861-74, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26715225

ABSTRACT

STUDY OBJECTIVE: The adolescent brain may be uniquely affected by acute sleep deprivation (ASD) and chronic sleep restriction (CSR), but direct evidence is lacking. We used electron microscopy to examine how ASD and CSR affect pyramidal neurons in the frontal cortex of adolescent mice, focusing on mitochondria, endosomes, and lysosomes that together perform most basic cellular functions, from nutrient intake to prevention of cellular stress. METHODS: Adolescent (1-mo-old) mice slept (S) or were sleep deprived (ASD, with novel objects and running wheels) during the first 6-8 h of the light period, chronically sleep restricted (CSR) for > 4 days (using novel objects, running wheels, social interaction, forced locomotion, caffeinated water), or allowed to recover sleep (RS) for ∼32 h after CSR. Ultrastructural analysis of 350 pyramidal neurons was performed (S = 82; ASD = 86; CSR = 103; RS = 79; 4 to 5 mice/group). RESULTS: Several ultrastructural parameters differed in S versus ASD, S versus CSR, CSR versus RS, and S versus RS, although the different methods used to enforce wake may have contributed to some of the differences between short and long sleep loss. Differences included larger cytoplasmic area occupied by mitochondria in CSR versus S, and higher number of secondary lysosomes in CSR versus S and RS. We also found that sleep loss may unmask interindividual differences not obvious during baseline sleep. Moreover, using a combination of 11 ultrastructural parameters, we could predict in up to 80% of cases whether sleep or wake occurred at the single cell level. CONCLUSIONS: Ultrastructural analysis may be a powerful tool to identify which cellular organelles, and thus which cellular functions, are most affected by sleep and sleep loss.


Subject(s)
Aging , Frontal Lobe/pathology , Pyramidal Cells/pathology , Pyramidal Cells/ultrastructure , Sleep Deprivation/pathology , Sleep Deprivation/physiopathology , Animals , Female , Frontal Lobe/ultrastructure , Lysosomes/pathology , Lysosomes/ultrastructure , Male , Mice , Mitochondria/pathology , Mitochondria/ultrastructure , Sexual Maturation , Sleep/physiology , Time Factors
20.
Neurobiol Dis ; 86: 154-69, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26611103

ABSTRACT

HIV-associated neurocognitive disorders (HAND) still occur in approximately 50% of HIV patients, and therapies to combat HAND progression are urgently needed. HIV proteins are released from infected cells and cause neuronal damage, possibly through mitochondrial abnormalities. Altered mitochondrial fission and fusion is implicated in several neurodegenerative disorders. Here, we hypothesized that mitochondrial fission/fusion may be dysregulated in neurons during HAND. We have identified decreased mitochondrial fission protein (dynamin 1-like; DNM1L) in frontal cortex tissues of HAND donors, along with enlarged and elongated mitochondria localized to the soma of damaged neurons. Similar pathology was observed in the brains of GFAP-gp120 tg mice. In vitro, recombinant gp120 decreased total and active DNM1L levels, reduced the level of Mitotracker staining, and increased extracellular acidification rate (ECAR) in primary neurons. DNM1L knockdown enhanced the effects of gp120 as measured by reduced Mitotracker signal in the treated cells. Interestingly, overexpression of DNM1L increased the level of Mitotracker staining in primary rat neurons and reduced neuroinflammation and neurodegeneration in the GFAP-gp120-tg mice. These data suggest that mitochondrial biogenesis dynamics are shifted towards mitochondrial fusion in brains of HAND patients and this may be due to gp120-induced reduction in DNM1L activity. Promoting mitochondrial fission during HIV infection of the CNS may restore mitochondrial biogenesis and prevent neurodegeneration.


Subject(s)
Brain/metabolism , Cognition Disorders/metabolism , Cognition Disorders/virology , HIV Infections/complications , Mitochondria/metabolism , Mitochondrial Dynamics , Neurons/metabolism , Adult , Animals , Brain/ultrastructure , Brain/virology , Dynamins , Encephalitis/metabolism , Encephalitis/virology , Female , Frontal Lobe/metabolism , Frontal Lobe/ultrastructure , Frontal Lobe/virology , GTP Phosphohydrolases/metabolism , HIV Envelope Protein gp120/metabolism , Humans , Male , Mice , Microtubule-Associated Proteins/metabolism , Middle Aged , Mitochondria/ultrastructure , Mitochondria/virology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Neurons/ultrastructure , Neurons/virology , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL