Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
PLoS Pathog ; 18(11): e1010929, 2022 11.
Article in English | MEDLINE | ID: mdl-36395147

ABSTRACT

The amyloid cascade hypothesis, focusing on pathological proteins aggregation, has so far failed to uncover the root cause of Alzheimer's disease (AD), or to provide an effective therapy. This traditional paradigm essentially explains a mechanism involved in the development of sporadic AD rather than its cause. The failure of an overwhelming majority of clinical studies (99.6%) demonstrates that a breakthrough in therapy would be difficult if not impossible without understanding the etiology of AD. It becomes more and more apparent that the AD pathology might originate from brain infection. In this review, we discuss a potential role of bacteria, viruses, fungi, and eukaryotic parasites as triggers of AD pathology. We show evidence from the current literature that amyloid beta, traditionally viewed as pathological, actually acts as an antimicrobial peptide, protecting the brain against pathogens. However, in case of a prolonged or excessive activation of a senescent immune system, amyloid beta accumulation and aggregation becomes damaging and supports runaway neurodegenerative processes in AD. This is paralleled by the recent study by Alam and colleagues (2022) who showed that alpha-synuclein, the protein accumulating in synucleinopathies, also plays a critical physiological role in immune reactions and inflammation, showing an unforeseen link between the 2 unrelated classes of neurodegenerative disorders. The multiplication of the amyloid precursor protein gene, recently described by Lee and collegues (2018), and possible reactivation of human endogenous retroviruses by pathogens fits well into the same picture. We discuss these new findings from the viewpoint of the infection hypothesis of AD and offer suggestions for future research.


Subject(s)
Alzheimer Disease , Anti-Infective Agents , Humans , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/pathology , Anti-Infective Agents/metabolism , Anti-Bacterial Agents/therapeutic use
2.
Biomolecules ; 10(9)2020 08 21.
Article in English | MEDLINE | ID: mdl-32825572

ABSTRACT

Progressive mitochondrial dysfunction due to the accumulation of amyloid beta (Aß) peptide within the mitochondrial matrix represents one of the key characteristics of Alzheimer's disease (AD) and appears already in its early stages. Inside the mitochondria, Aß interacts with a number of biomolecules, including cyclophilin D (cypD) and 17ß-hydroxysteroid dehydrogenase type 10 (17ß-HSD10), and affects their physiological functions. However, despite intensive ongoing research, the exact mechanisms through which Aß impairs mitochondrial functions remain to be explained. In this work, we studied the interactions of Aß with cypD and 17ß-HSD10 in vitro using the surface plasmon resonance (SPR) method and determined the kinetic parameters (association and dissociation rates) of these interactions. This is the first work which determines all these parameters under the same conditions, thus, enabling direct comparison of relative affinities of Aß to its mitochondrial binding partners. Moreover, we used the determined characteristics of the individual interactions to simulate the concurrent interactions of Aß with cypD and 17ß-HSD10 in different model situations associated with the progression of AD. This study not only advances the understanding of Aß-induced processes in mitochondria during AD, but it also provides a new perspective on research into complex multi-interaction biomolecular processes in general.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Mitochondrial Proteins/metabolism , Peptidyl-Prolyl Isomerase F/metabolism , 17-Hydroxysteroid Dehydrogenases/chemistry , Amyloid beta-Peptides/chemistry , Biosensing Techniques , Peptidyl-Prolyl Isomerase F/chemistry , Humans , Mitochondrial Proteins/chemistry , Surface Plasmon Resonance
3.
Neurochem Res ; 45(4): 915-927, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31997103

ABSTRACT

The nucleus-encoded 17ß-hydroxysteroid dehydrogenase type 10 (17ß-HSD10) regulates cyclophilin D (cypD) in the mitochondrial matrix. CypD regulates opening of mitochondrial permeability transition pores. Both mechanisms may be affected by amyloid ß peptides accumulated in mitochondria in Alzheimer's disease (AD). In order to clarify changes occurring in brain mitochondria, we evaluated interactions of both mitochondrial proteins in vitro (by surface plasmon resonance biosensor) and detected levels of various complexes of 17ß-HSD10 formed in vivo (by sandwich ELISA) in brain mitochondria isolated from the transgenic animal model of AD (homozygous McGill-R-Thy1-APP rats) and in cerebrospinal fluid samples of AD patients. By surface plasmon resonance biosensor, we observed the interaction of 17ß-HSD10 and cypD in a direct real-time manner and determined, for the first time, the kinetic parameters of the interaction (ka 2.0 × 105 M1s-1, kd 5.8 × 104 s-1, and KD 3.5 × 10-10 M). In McGill-R-Thy1-APP rats compared to controls, levels of 17ß-HSD10-cypD complexes were decreased and those of total amyloid ß increased. Moreover, the levels of 17ß-HSD10-cypD complexes were decreased in cerebrospinal fluid of individuals with AD (in mild cognitive impairment as well as dementia stages) or with Frontotemporal lobar degeneration (FTLD) compared to cognitively normal controls (the sensitivity of the complexes to AD dementia was 92.9%, that to FTLD 73.8%, the specificity to AD dementia equaled 91.7% in a comparison with the controls but only 26.2% with FTLD). Our results demonstrate the weakened ability of 17ß-HSD10 to regulate cypD in the mitochondrial matrix probably via direct effects of amyloid ß. Levels of 17ß-HSD10-cypD complexes in cerebrospinal fluid seem to be the very sensitive indicator of mitochondrial dysfunction observed in neurodegeneration but unfortunately not specific to AD pathology. We do not recommend it as the new biomarker of AD.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Alzheimer Disease/metabolism , Peptidyl-Prolyl Isomerase F/metabolism , 17-Hydroxysteroid Dehydrogenases/cerebrospinal fluid , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Humans , Kinetics , Male , Mitochondria/metabolism , Rats, Transgenic , Rats, Wistar , Surface Plasmon Resonance
4.
Int J Mol Sci ; 21(24)2020 Dec 20.
Article in English | MEDLINE | ID: mdl-33419257

ABSTRACT

In early stages of Alzheimer's disease (AD), amyloid beta (Aß) accumulates in the mitochondrial matrix and interacts with mitochondrial proteins, such as cyclophilin D (cypD) and 17ß-hydroxysteroid dehydrogenase 10 (17ß-HSD10). Multiple processes associated with AD such as increased production or oligomerization of Aß affect these interactions and disbalance the equilibrium between the biomolecules, which contributes to mitochondrial dysfunction. Here, we investigate the effect of the ionic environment on the interactions of Aß (Aß1-40, Aß1-42) with cypD and 17ß-HSD10 using a surface plasmon resonance (SPR) biosensor. We show that changes in concentrations of K+ and Mg2+ significantly affect the interactions and may increase the binding efficiency between the biomolecules by up to 35% and 65% for the interactions with Aß1-40 and Aß1-42, respectively, in comparison with the physiological state. We also demonstrate that while the binding of Aß1-40 to cypD and 17ß-HSD10 takes place preferentially around the physiological concentrations of ions, decreased concentrations of K+ and increased concentrations of Mg2+ promote the interaction of both mitochondrial proteins with Aß1-42. These results suggest that the ionic environment represents an important factor that should be considered in the investigation of biomolecular interactions taking place in the mitochondrial matrix under physiological as well as AD-associated conditions.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/chemistry , Biosensing Techniques/methods , Surface Plasmon Resonance/methods , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/genetics , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Peptidyl-Prolyl Isomerase F/chemistry , Peptidyl-Prolyl Isomerase F/genetics , Humans , Ions/chemistry , Mitochondria/chemistry , Mitochondrial Proteins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/genetics
5.
Neurotherapeutics ; 17(1): 329-339, 2020 01.
Article in English | MEDLINE | ID: mdl-31820275

ABSTRACT

Tuberous sclerosis complex (TSC) is a genetic disorder characterized by frequent noncancerous neoplasia in the brain, which can induce a range of severe neuropsychiatric symptoms in humans, resulting from out of control tissue growth. The causative spontaneous loss-of-function mutations have been also identified in rats. Herein, we studied histopathological and molecular changes in brain lesions of the Eker rat model carrying germline mutation of the tsc2 gene, predisposed to multiple neoplasias. Predominant subcortical tumors were analyzed, along with a rare form occurring within the pyriform lobe. The uniform composition of lesions supports the histochemical parity of malformations, with immunofluorescence data supporting their neuro-glial origin. Massive depletion of mature neurons and axonal loss were evident within lesions, with occasional necrotic foci implying advanced stage of pathology. Enrichment of mesenchymal-derived cell markers with hallmarks of neurogenesis and active microglia imply enhanced cell proliferation, with local immune response. The depletion of capillaries within the core was complemented by the formation of dense mesh of nascent vessels at the interface of neoplasia with healthy tissue, implying large-scale vascular remodeling. Taken as a whole, these findings present several novel features of brain tumors in Eker rat model, rendering it suitable for studies of the pathobiology and progression of primary brain tumors, with therapeutic interventions.


Subject(s)
Brain Neoplasms/pathology , Microglia/pathology , Neurons/pathology , Tuberous Sclerosis/pathology , Vascular Remodeling , Animals , Astrocytes/pathology , Axons/pathology , Brain/blood supply , Brain/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/etiology , Female , Male , Rats, Long-Evans , Tuberous Sclerosis/complications , Tuberous Sclerosis Complex 2 Protein/genetics
6.
Sci Rep ; 9(1): 16700, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31723183

ABSTRACT

In early stages of Alzheimer's disease (AD), amyloid-ß (Aß) accumulates in neuronal mitochondria where it interacts with a number of biomolecules including 17beta-hydroxysteroide dehydrogenase 10 (17ß-HSD10) and cyclophilin D (cypD). It has been hypothesized that 17ß-HSD10 interacts with cypD preventing it from opening mitochondrial permeability transition pores and that its regulation during AD may be affected by the accumulation of Aß. In this work, we demonstrate for the first time that 17ß-HSD10 and cypD form a stable complex in vitro. Furthermore, we show that factors, such as pH, ionic environment and the presence of Aß, affect the ability of 17ß-HSD10 to bind cypD. We demonstrate that K+ and Mg2+ ions present at low levels may facilitate this binding. We also show that different fragments of Aß (Aß1-40 and Aß1-42) affect the interaction between 17ß-HSD10 and cypD differently and that Aß1-42 (in contrast to Aß1-40) is capable of simultaneously binding both 17ß-HSD10 and cypD in a tri-complex.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Calcium/metabolism , Mitochondria/metabolism , Peptidyl-Prolyl Isomerase F/metabolism , Alzheimer Disease/metabolism , Humans , In Vitro Techniques , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore
7.
Int J Mol Sci ; 20(13)2019 Jul 03.
Article in English | MEDLINE | ID: mdl-31277281

ABSTRACT

Aging and chronic sleep deprivation (SD) are well-recognized risk factors for Alzheimer's disease (AD), with N-methyl-D-aspartate receptor (NMDA) and downstream nitric oxide (NO) signalling implicated in the process. Herein, we investigate the impact of the age- and acute or chronic SD-dependent changes on the expression of NMDA receptor subunits (NR1, NR2A, and NR2B) and on the activities of NO synthase (NOS) isoforms in the cortex of Wistar rats, with reference to cerebral lateralization. In young adult controls, somewhat lateralized seasonal variations in neuronal and endothelial NOS have been observed. In aged rats, overall decreases in NR1, NR2A, and NR2B expression and reduction in neuronal and endothelial NOS activities were found. The age-dependent changes in NR1 and NR2B significantly correlated with neuronal NOS in both hemispheres. Changes evoked by chronic SD (dysfunction of endothelial NOS and the increasing role of NR2A) differed from those evoked by acute SD (increase in inducible NOS in the right side). Collectively, these results demonstrate age-dependent regulation of the level of NMDA receptor subunits and downstream NOS isoforms throughout the rat brain, which could be partly mimicked by SD. As described herein, age and SD alterations in the prevalence of NMDA receptors and NOS could contribute towards cognitive decline in the elderly, as well as in the pathobiology of AD and the neurodegenerative process.


Subject(s)
Aging/metabolism , Cerebral Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction , Sleep Deprivation/metabolism , Age Factors , Alzheimer Disease/epidemiology , Alzheimer Disease/etiology , Animals , Gene Expression Regulation , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Risk Factors , Sleep Deprivation/physiopathology
8.
Biomark Med ; 13(4): 267-277, 2019 03.
Article in English | MEDLINE | ID: mdl-30888831

ABSTRACT

AIM: Surface tension of biological fluids can be influenced by changes in oligomerization or aggregation of surfactant peptides or proteins. Amphiphilic peptides of amyloid-ß or other amyloidogenic peptides/proteins display properties of surfactants, oligomerization and aggregation increase also their fluorescence intensity compared with native structures. Results/methodology: We estimated surface tension and native/ThioflavinT-based/intrinsic amyloid fluorescence intensity in serum and cerebrospinal fluid samples for their evalution as diagnostic biomarkers for Alzheimer´s disease (AD). DISCUSSION/CONCLUSION: Our results indicate that values of surface tension are not a suitable biomarker for AD. However, the ratio of ThioflavinT-based fluorescence to intrinsic amyloid fluorescence in cerebrospinal fluid appears to be an acceptable supportive diagnostic biomarker for AD (its sensitivity was 61.1%, and the specificity 70.8% when compared with aged controls).


Subject(s)
Alzheimer Disease/diagnosis , Amyloid/blood , Amyloid/cerebrospinal fluid , Biomarkers/analysis , Fluorescence , Adult , Aged , Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Case-Control Studies , Female , Follow-Up Studies , Humans , Male , Prognosis , Surface Tension
9.
Biomark Med ; 12(12): 1331-1340, 2018 12.
Article in English | MEDLINE | ID: mdl-30520659

ABSTRACT

AIM: We aimed to characterize the role of mitochondrial 17ß-hydroxysteroid dehydrogenase type 10 (17ß-HSD10) overexpression in multiple sclerosis (MS) and to evaluate its use as a biomarker. Materials & methods: We estimated levels of 17ß-HSD10, amyloid ß 1-42, cyclophilin D, 17ß-HSD10-cyclophilin D complexes or 17ß-HSD10-parkin complexes in cerebrospinal fluid (CSF) samples. RESULTS: The increase in 17ß-HSD10 levels or in 17ß-HSD10-parkin complexes and links to leukocytes were found only in relapsing-remitting MS. The sensitivity of the biomarker was 64%, the specificity equaled 60-63% compared with controls. CONCLUSION: Increased CSF levels of 17ß-HSD10 in later stages of MS could be interpreted via its upregulation in demyelinated neuronal axons. CSF levels of 17ß-HSD10 are not the valuable biomarker for the early diagnosis or for the progression of MS.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/cerebrospinal fluid , Multiple Sclerosis/cerebrospinal fluid , Adult , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Female , Humans , Male , Peptide Fragments/cerebrospinal fluid
10.
Front Aging Neurosci ; 10: 250, 2018.
Article in English | MEDLINE | ID: mdl-30210330

ABSTRACT

The McGill-R-Thy1-APP transgenic rat is an animal model of the familial form of Alzheimer's disease (AD). This model mirrors several neuropathological hallmarks of the disease, including the accumulation of beta-amyloid and the formation of amyloid plaques (in homozygous animals only), neuroinflammation and the gradual deterioration of cognitive functions even prior to plaque formation, although it lacks the tauopathy observed in human victims of AD. The goal of the present study was a thorough characterization of the homozygous model with emphasis on its face validity in several domains of behavior known to be affected in AD patients, including cognitive functions, motor coordination, emotionality, sociability, and circadian activity patterns. On the behavioral level, we found normal locomotor activity in spontaneous exploration, but problems with balance and gait coordination, increased anxiety and severely impaired spatial cognition in 4-7 month old homozygous animals. The profile of social behavior and ultrasonic communication was altered in the McGill rats, without a general social withdrawal. McGill rats also exhibited changes in circadian profile, with a shorter free-running period and increased total activity during the subjective night, without signs of sleep disturbances during the inactive phase. Expression of circadian clock gene Bmal1 was found to be increased in the parietal cortex and cerebellum, while Nr1d1 expression was not changed. The clock-controlled gene Prok2 expression was found to be elevated in the parietal cortex and hippocampus, which might have contributed to the observed changes in circadian phenotype. We conclude that the phenotype in the McGill rat model is not restricted to the cognitive domain, but also includes gait problems, changes in emotionality, social behavior, and circadian profiles. Our findings show that the model should be useful for the development of new therapeutic approaches targeting not only memory decline but also other symptoms decreasing the quality of life of AD patients.

11.
Dement Geriatr Cogn Disord ; 43(1-2): 45-58, 2017.
Article in English | MEDLINE | ID: mdl-27988521

ABSTRACT

BACKGROUND: Reversible acetylcholinesterase inhibitors are used in Alzheimer disease therapy. However, tacrine and its derivatives have severe side effects. Derivatives of the tacrine analogue 7-methoxytacrine (MEOTA) are less toxic. METHODS: We evaluated new derivatives of 7-MEOTA (2 homodimers linked by 2 C4-C5 chains and 5 N-alkylated C4-C8 side chain derivatives) in vitro, using the rat hippocampal choline transporter CHT1. RESULTS: Some derivatives were effective inhibitors of rat acetylcholinesterase and comparable with 7-MEOTA. All derivatives were able to inhibit CHT1, probably via quaternary ammonium, and this interaction could be involved in the enhancement of their detrimental side effects and/or in the attenuation of their promising effects. Under conditions of disrupted lipid rafts, the unfavorable effects of some derivatives were weakened. Only tacrine was probably able to stereospecifically interact with the naturally occurring amyloid-ß isoform and to simultaneously stimulate CHT1. Some derivatives, when coincubated with amyloid ß, did not influence CHT1. All derivatives also increased the fluidity of the cortical membranes. CONCLUSION: The N-alkylated derivative of 7-MEOTA bearing from C4 side chains appears to be the most promising compound and should be evaluated in future in vivo research.


Subject(s)
Cation Transport Proteins/drug effects , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Tacrine/analogs & derivatives , Tacrine/pharmacology , Alkylation , Amyloid beta-Peptides/drug effects , Animals , Cerebral Cortex/drug effects , Cholesterol/metabolism , Choline/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Membrane Fluidity/drug effects , Membrane Microdomains/drug effects , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/pharmacology , Rats , Rats, Wistar , Stereoisomerism , Synaptosomes/drug effects , Synaptosomes/metabolism , Tacrine/chemical synthesis
12.
Neurochem Res ; 41(8): 1911-23, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27038442

ABSTRACT

Methamphetamine (MA) is the most commonly used psychostimulant drug, the chronic abuse of which leads to neurodegenerative changes in the brain. The global use of MA is increasing, including in pregnant women. Since MA can cross both placental and haematoencephalic barriers and is also present in maternal milk, children of chronically abused mothers are exposed prenatally as well as postnatally. Women seem to be more vulnerable to some aspects of MA abuse than men. MA is thought to exert its effects among others via direct interactions with dopamine transporters (DATs) in the brain tissue. Sexual dimorphism of the DAT system could be a base of sex-dependent actions of MA observed in behavioural and neurochemical studies. Possible sex differences in the DATs of preadolescent offspring exposed to MA prenatally and/or postnatally have not yet been evaluated. We examined the striatal synaptosomal DATs (the activity and density of surface expressed DATs and total DAT expression) in preadolescent male and female Wistar rats (31-35-day old animals) exposed prenatally and/or postnatally to MA (daily 5 mg/kg, s.c. to mothers during pregnancy and lactation). To distinguish between specific and nonspecific effects of MA on DATs, we also evaluated the in vitro effects of lipophilic MA on the fluidity of striatal membranes isolated from preadolescent and young adult rats of both sexes. We observed similar changes in the DATs of preadolescent rats exposed prenatally or postnatally (MA-mediated drop in the reserve pool but no alterations in surface-expressed DATs). However, prenatal exposure evoked significant changes in males and postnatal exposure in females. A significant decrease in the activity of surface-expressed DATs was found only in postnatally exposed females sensitized to MA via prenatal exposure. MA applied in vitro increased the fluidity of striatal membranes of preadolescent female but not male rats. In summary, DATs of preadolescent males are more sensitive to prenatal MA exposure via changes in the reserve pool and those of preadolescent females to postnatal MA exposure via the same mechanism. The combination of prenatal and postnatal MA exposure increases the risk of dopaminergic deficits via alterations in the activity of surface-expressed DATs especially in preadolescent females. MA-mediated changes in DATs of preadolescent females could be still enhanced via nonspecific disordering actions of MA on striatal membranes.


Subject(s)
Corpus Striatum/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine/metabolism , Methamphetamine/toxicity , Prenatal Exposure Delayed Effects/metabolism , Sex Characteristics , Animals , Animals, Newborn , Corpus Striatum/drug effects , Female , Male , Methamphetamine/administration & dosage , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...