Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Neurochem Res ; 42(1): 306-319, 2017 Jan.
Article in English | MEDLINE | ID: mdl-26980008

ABSTRACT

Cytosolic and mitochondrial human branched chain aminotransferase (hBCATc and hBCATm, respectively) play an integral role in brain glutamate metabolism. Regional increased levels of hBCATc in the CA1 and CA4 region of Alzheimer's disease (AD) brain together with increased levels of hBCATm in frontal and temporal cortex of AD brains, suggest a role for these proteins in glutamate excitotoxicity. Glutamate toxicity is a key pathogenic feature of several neurological disorders including epilepsy associated dementia, AD, vascular dementia (VaD) and dementia with Lewy bodies (DLB). To further understand if these increases are specific to AD, the expression profiles of hBCATc and hBCATm were examined in other forms of dementia including DLB and VaD. Similar to AD, levels of hBCATm were significantly increased in the frontal and temporal cortex of VaD cases and in frontal cortex of DLB cases compared to controls, however there were no observed differences in hBCATc between groups in these areas. Moreover, multiple forms of hBCATm were observed that were particular to the disease state relative to matched controls. Real-time PCR revealed similar expression of hBCATm mRNA in frontal and temporal cortex for all cohort comparisons, whereas hBCATc mRNA expression was significantly increased in VaD cases compared to controls. Collectively our results suggest that hBCATm protein expression is significantly increased in the brains of DLB and VaD cases, similar to those reported in AD brain. These findings indicate a more global response to altered glutamate metabolism and suggest common metabolic responses that might reflect shared neurodegenerative mechanisms across several forms of dementia.


Subject(s)
Dementia, Vascular/enzymology , Gene Expression Regulation, Enzymologic , Lewy Body Disease/enzymology , Transaminases/biosynthesis , Aged , Aged, 80 and over , Brain/enzymology , Brain/pathology , Cohort Studies , Dementia, Vascular/genetics , Dementia, Vascular/pathology , Female , Humans , Lewy Body Disease/genetics , Lewy Body Disease/pathology , Male , Minor Histocompatibility Antigens/biosynthesis , Pregnancy Proteins/biosynthesis , Transaminases/genetics
2.
J Alzheimers Dis ; 50(4): 1191-203, 2016.
Article in English | MEDLINE | ID: mdl-26836178

ABSTRACT

Epidemiological data associate hypertension with a predisposition to Alzheimer's disease (AD), and a number of postmortem and in vivo studies also demonstrate that hypertension increases amyloid-ß (Aß) pathology. In contrast, anti-hypertensive medications reportedly improve cognition and decrease the risk of AD, while certain classes of anti-hypertensive drugs are associated with decreased AD-related pathology. We investigated the effects of hypertension and anti-hypertensive treatment on Aß plaque load in postmortem frontal cortex in AD. Aß load was significantly increased in hypertensive (n = 20) relative to normotensive cases (n = 62) and was also significantly higher in treated (n = 9) than untreated hypertensives (n = 11). We then looked into mechanisms by which hypertension and treatment might increase Aß load, focusing on Aß-synthesizing enzymes, ß- and γ-secretase, and Aß-degrading enzymes, angiotensin-converting enzyme (ACE), insulin-degrading enzyme (IDE) and neprilysin. ACE and IDE protein levels were significantly lower in hypertensive (n = 21) than normotensive cases (n = 64), perhaps translating to decreased Aß catabolism in hypertensives. ACE level was significantly higher in treated (n = 9) than untreated hypertensives (n = 12), possibly reflecting feedback upregulation of the renin-angiotensin system. Prospective studies in larger cohorts stratified according to anti-hypertensive drug class are needed to confirm these initial findings and to elucidate the interactions between hypertension, anti-hypertensive treatments, and Aß metabolism.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Antihypertensive Agents/therapeutic use , Frontal Lobe/pathology , Hypertension/pathology , Plaque, Amyloid/pathology , Aged , Aged, 80 and over , Female , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Humans , Hypertension/drug therapy , Hypertension/metabolism , Immunohistochemistry , Insulysin/metabolism , Male , Neprilysin/metabolism , Peptidyl-Dipeptidase A/metabolism , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism , Retrospective Studies
3.
Neuropathol Appl Neurobiol ; 41(4): 428-44, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25556395

ABSTRACT

AIMS: Deposition of amyloid beta (Aß) in the brain is one of the defining abnormalities of Alzheimer's disease (AD). Phosphorylation of Aß at serine 8 (pAß) has been implicated in its aggregation in vitro and pAß level has been shown to be significantly elevated in AD. We aimed to assess the specificity of pAß for AD and have investigated associations of pAß with parenchymal and cerebrovascular accumulation of Aß, disease progression, angiotensin-converting enzyme activity and APOE genotype. METHODS: The distribution of pAß was studied by immunohistochemistry in sporadic and familial AD, pure dementia with Lewy bodies (DLB), pure vascular dementia (VaD) and age-matched controls. Soluble and insoluble (guanidine-extractable) pAß level was measured by enzyme-linked immunosorbent assay (ELISA) in the midfrontal and parahippocampal cortex in sporadic AD (n = 20, 10 with Braak tangle stages of III-IV and 10 of stages V-VI), DLB (n = 10), VaD (n = 10) and age-matched controls (n = 20). RESULTS: We found pAß to be associated with only a subset of Aß plaques and vascular deposits in sporadic and familial AD, with absent or minimal immunohistochemically detectable pAß in control, DLB and VaD brains. In both brain regions, insoluble pAß level was significantly elevated only in advanced AD (Braak tangle stage of V or VI) and in the parahippocampus soluble and insoluble pAß level increased with the number of APOE ε4 alleles. CONCLUSIONS: These results indicate that pAß accumulation in the parenchyma and vasculature is largely restricted to late-stage AD (Braak tangle stage V-VI).


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Dementia, Vascular/metabolism , Dementia/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Apolipoproteins E/genetics , Dementia/pathology , Dementia, Vascular/pathology , Humans , Lewy Bodies , Neocortex/metabolism , Neocortex/pathology , Phosphorylation , Serine/metabolism , Severity of Illness Index
4.
J Alzheimers Dis ; 44(1): 153-62, 2015.
Article in English | MEDLINE | ID: mdl-25201786

ABSTRACT

BACKGROUND: Higher angiotensin-converting enzyme (ACE) activity might increase the risk of Alzheimer's disease by increasing blood pressure, and subsequent development of cerebral small vessel disease (CSVD). Yet, it may also decrease this risk, as it functions to degrade amyloid-ß, thereby reducing brain atrophy. OBJECTIVE: To examine the cross-sectional associations of serum and cerebrospinal fluid (CSF) ACE protein levels and activity with brain atrophy and CSVD in a memory clinic cohort. METHODS: In 118 subjects from the memory clinic based Amsterdam Dementia Cohort (mean age 66 ± 8 years), ACE protein levels (ng/ml) and activity in CSF and serum were investigated. Poisson regression analyses were used to associate ACE measurements with rated global cortical atrophy, medial temporal lobe atrophy, lacunar infarcts, white matter hyperintensities, and microbleeds on brain MRI. RESULTS: Higher CSF ACE activity was associated with a reduced risk of global brain atrophy. The relative risk (95% CI) of having global cortical atrophy ≥2 per SD increase in CSF ACE activity was 0.67 (0.49; 0.93). ACE levels were not significantly related to measures of CSVD. CONCLUSIONS: These results show that high ACE might have protective effects on the brain. This could suggest that ACE inhibitors, which may lower CSF ACE levels, are not preferred as antihypertensive treatment in patients at risk for Alzheimer's disease.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Brain Diseases/cerebrospinal fluid , Brain Diseases/pathology , Brain/pathology , Peptidyl-Dipeptidase A/cerebrospinal fluid , Aged , Aged, 80 and over , Alzheimer Disease/blood , Apolipoproteins E/genetics , Atrophy/cerebrospinal fluid , Atrophy/pathology , Brain Diseases/blood , Cerebral Small Vessel Diseases/enzymology , Female , Humans , Magnetic Resonance Imaging , Male , Memory Disorders/cerebrospinal fluid , Memory Disorders/etiology , Middle Aged , Peptidyl-Dipeptidase A/blood
5.
Front Aging Neurosci ; 6: 238, 2014.
Article in English | MEDLINE | ID: mdl-25309424

ABSTRACT

There is increasing evidence that deficient clearance of ß-amyloid (Aß) contributes to its accumulation in late-onset Alzheimer disease (AD). Several Aß-degrading enzymes, including neprilysin (NEP), endothelin-converting enzyme (ECE), and angiotensin-converting enzyme (ACE) reduce Aß levels and protect against cognitive impairment in mouse models of AD. In post-mortem human brain tissue we have found that the activity of these Aß-degrading enzymes rise with age and increases still further in AD, perhaps as a physiological response that helps to minimize the build-up of Aß. ECE-1/-2 and ACE are also rate-limiting enzymes in the production of endothelin-1 (ET-1) and angiotensin II (Ang II), two potent vasoconstrictors, increases in the levels of which are likely to contribute to reduced blood flow in AD. This review considers the possible interdependence between Aß-degrading enzymes, ischemia and Aß in AD: ischemia has been shown to increase Aß production both in vitro and in vivo, whereas increased Aß probably enhances ischemia by vasoconstriction, mediated at least in part by increased ECE and ACE activity. In contrast, NEP activity may help to maintain cerebral perfusion, by reducing the accumulation of Aß in cerebral blood vessels and lessening its toxicity to vascular smooth muscle cells. In assessing the role of Aß-degrading proteases in the pathogenesis of AD and, particularly, their potential as therapeutic agents, it is important to bear in mind the multifunctional nature of these enzymes and to consider their effects on other substrates and pathways.

6.
Alzheimers Res Ther ; 6(3): 27, 2014.
Article in English | MEDLINE | ID: mdl-24987467

ABSTRACT

INTRODUCTION: Lower angiotensin-converting enzyme (ACE) activity could increase the risk of Alzheimer's disease (AD) as ACE functions to degrade amyloid-ß (Aß). Therefore, we investigated whether ACE protein and activity levels in cerebrospinal fluid (CSF) and serum were associated with CSF Aß, total tau (tau) and tau phosphorylated at threonine 181 (ptau). METHODS: We included 118 subjects from our memory clinic-based Amsterdam Dementia Cohort (mean age 66 ± 8 years) with subjective memory complaints (n = 40) or AD (n = 78), who did not use antihypertensive drugs. We measured ACE protein levels (ng/ml) and activity (RFU) in CSF and serum, and amyloid ß1-42, tau and ptau (pg/ml) in CSF. RESULTS: Cross-sectional regression analyses showed that ACE protein level and activity in CSF and serum were lower in patients with AD compared to controls. Lower CSF ACE protein level, and to a lesser extent serum ACE protein level and CSF ACE activity, were associated with lower CSF Aß, indicating more brain Aß pathology; adjusted regression coefficients (B) (95% CI) per SD increase were 0.09 (0.04; 0.15), 0.06 (0.00; 0.12) and 0.05 (0.00; 0.11), respectively. Further, lower CSF ACE protein level was associated with lower CSF tau and ptau levels; adjusted B's (95% CI) per SD increase were 0.15 (0.06; 0.25) and 0.17 (0.10; 0.25), respectively. CONCLUSIONS: These results strengthen the hypothesis that ACE degrades Aß. This could suggest that lowering ACE levels by for example ACE-inhibitors might have adverse consequences for patients with, or at risk for AD.

7.
Brain ; 137(Pt 5): 1524-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24618270

ABSTRACT

Little is known about the contributors and physiological responses to white matter hypoperfusion in the human brain. We previously showed the ratio of myelin-associated glycoprotein to proteolipid protein 1 in post-mortem human brain tissue correlates with the degree of ante-mortem ischaemia. In age-matched post-mortem cohorts of Alzheimer's disease (n = 49), vascular dementia (n = 17) and control brains (n = 33) from the South West Dementia Brain Bank (Bristol), we have now examined the relationship between the ratio of myelin-associated glycoprotein to proteolipid protein 1 and several other proteins involved in regulating white matter vascularity and blood flow. Across the three cohorts, white matter perfusion, indicated by the ratio of myelin-associated glycoprotein to proteolipid protein 1, correlated positively with the concentration of the vasoconstrictor, endothelin 1 (P = 0.0005), and negatively with the concentration of the pro-angiogenic protein, vascular endothelial growth factor (P = 0.0015). The activity of angiotensin-converting enzyme, which catalyses production of the vasoconstrictor angiotensin II was not altered. In samples of frontal white matter from an independent (Oxford, UK) cohort of post-mortem brains (n = 74), we confirmed the significant correlations between the ratio of myelin-associated glycoprotein to proteolipid protein 1 and both endothelin 1 and vascular endothelial growth factor. We also assessed microvessel density in the Bristol (UK) samples, by measurement of factor VIII-related antigen, which we showed to correlate with immunohistochemical measurements of vessel density, and found factor VIII-related antigen levels to correlate with the level of vascular endothelial growth factor (P = 0.0487), suggesting that upregulation of vascular endothelial growth factor tends to increase vessel density in the white matter. We propose that downregulation of endothelin 1 and upregulation of vascular endothelial growth factor in the context of reduced ratio of myelin-associated glycoprotein to proteolipid protein 1 are likely to be protective physiological responses to reduced white matter perfusion. Further analysis of the Bristol cohort showed that endothelin 1 was reduced in the white matter in Alzheimer's disease (P < 0.05) compared with control subjects, but not in vascular dementia, in which endothelin 1 tended to be elevated, perhaps reflecting abnormal regulation of white matter perfusion in vascular dementia. Our findings demonstrate the potential of post-mortem measurement of myelin proteins and mediators of vascular function, to assess physiological and pathological processes involved in the regulation of cerebral perfusion in Alzheimer's disease and vascular dementia.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Dementia, Vascular/pathology , Nerve Fibers, Myelinated/physiology , Aged , Aged, 80 and over , Cohort Studies , Endothelin-1/metabolism , Factor VIII/metabolism , Female , Humans , Male , Middle Aged , Myelin-Associated Glycoprotein/metabolism , Nerve Fibers, Myelinated/pathology , Peptidyl-Dipeptidase A/metabolism , Vascular Endothelial Growth Factor A/metabolism
8.
Expert Opin Investig Drugs ; 22(10): 1229-42, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23800077

ABSTRACT

INTRODUCTION: Hypertension is a modifiable risk factor for Alzheimer's disease (AD) and other dementias. Yet, despite this well-documented association, few of the current strategies to treat AD are directed at this possible target. The renin-aldosterone angiotensin system (RAAS) is a centrally active modifiable pathway that is involved in cerebral blood flow regulation. Currently, three classes of RAAS-targeting drugs are licensed for treatment of peripheral hypertension--angiotensin-converting enzyme inhibitors (ACE-Is), angiotensin II receptor blockers (ARBs) and direct renin inhibitors (DRIs). All of these are generally well tolerated and have been shown to offer varying degrees of protection on aspects of cognition and dementia, thus making them an attractive therapeutic option for AD. AREAS COVERED: This review summarises existing evidence regarding the plausibility of using RAAS-targeting drugs as a strategy to treat AD and highlights unresolved aspects to such approaches, namely the potential impact of altering angiotensin II-mediated processes in the central nervous system. EXPERT OPINION: Continued biochemical research of the RAAS pathway in combination with formal investigation of current RAAS-modifying drugs in randomised clinical trials is now necessary to determine their therapeutic value in AD.


Subject(s)
Alzheimer Disease/drug therapy , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Antihypertensive Agents/therapeutic use , Renin-Angiotensin System/drug effects , Alzheimer Disease/etiology , Alzheimer Disease/physiopathology , Angiotensin Receptor Antagonists/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Antihypertensive Agents/administration & dosage , Blood Pressure/drug effects , Cerebrovascular Circulation/drug effects , Clinical Trials as Topic , Cognition/drug effects , Humans , Hypertension/complications , Hypertension/drug therapy , Hypertension/physiopathology , Renin/antagonists & inhibitors , Renin-Angiotensin System/physiology
9.
Neurobiol Aging ; 33(7): 1345-55, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21074291

ABSTRACT

Decreased cerebral blood flow and blood-brain barrier disruption are features of Alzheimer's disease (AD). The plasma kallikrein-kinin system modulates cerebrovascular tone through release of vasoactive bradykinin (BK). Cerebroventricular infusion of Aß1-40 enhances BK release, suggesting that the activity of this system may be elevated in AD. We investigated the profile of the activating protease of this system, plasma kallikrein (PK), in frontal and temporal brain tissue from postmortem confirmed cases of AD, vascular dementia (VaD), and controls. Measurements of neuron specific enolase messenger ribonucleic acid (mRNA) and protein were used to adjust for neuronal loss. Adjusted PK mRNA was significantly increased in the frontal cortex in AD, and the frontal and temporal cortex in VaD. Similar trends were seen for PK protein level in AD and VaD. PK activity was significantly increased in the frontal and temporal cortex in AD. Increased PK activity in AD is likely to contribute to increased BK release and may thereby influence cerebral blood flow and vascular permeability.


Subject(s)
Alzheimer Disease/metabolism , Dementia, Vascular/metabolism , Frontal Lobe/metabolism , Kallikrein-Kinin System/physiology , Temporal Lobe/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/blood , Alzheimer Disease/enzymology , Bradykinin/blood , Bradykinin/metabolism , Cohort Studies , Dementia, Vascular/blood , Dementia, Vascular/enzymology , Female , Frontal Lobe/enzymology , Humans , Kallikreins/blood , Kallikreins/metabolism , Male , Middle Aged , Temporal Lobe/enzymology
10.
Brain Res ; 1363: 1-10, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-20846516

ABSTRACT

Human kallikrein-related peptidase 6 (KLK6) is highly expressed in the central nervous system. Although the physiological roles of this serine protease are unknown, in vitro substrates include amyloid precursor protein and components of the extracellular matrix, which are altered in neurological disease, particularly Alzheimer's disease (AD). We have compared KLK6 expression in post-mortem brain tissue in AD, vascular dementia (VaD) and controls. We studied the distribution of KLK6 in the temporal cortex and white matter by immunohistochemistry, and measured KLK6 mRNA and protein levels in the frontal and temporal cortex from 15 AD, 15 VaD and 15 control brains. Immunohistochemistry showed KLK6 to be restricted to endothelial cells. After adjustment for variations in vessel density by measurement of factor VIII-related antigen, we found KLK6 protein and mRNA levels to be significantly decreased in the frontal but not the temporal cortex in AD. In VaD, KLK6 protein level was significantly increased in the frontal cortex. Our findings suggest that an altered KLK6 expression may contribute to vascular abnormalities in AD and VaD.


Subject(s)
Alzheimer Disease/physiopathology , Cerebral Cortex/physiology , Dementia, Vascular/physiopathology , Kallikreins/genetics , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Cerebral Cortex/cytology , Dementia, Vascular/metabolism , Endothelial Cells/physiology , Factor VIII/metabolism , Female , Fluorescent Antibody Technique , Frontal Lobe/cytology , Frontal Lobe/physiology , Humans , Kallikreins/metabolism , Male , Middle Aged , Neocortex/cytology , Neocortex/physiology , Neurons/physiology , RNA, Messenger/metabolism , Temporal Lobe/cytology , Temporal Lobe/physiology
11.
Am J Transl Res ; 1(2): 163-77, 2009 Jan 18.
Article in English | MEDLINE | ID: mdl-19956428

ABSTRACT

Angiotensin-converting enzyme (ACE) has been implicated in Alzheimer's disease (AD): ACE1 variations influence plasma ACE and risk of AD, and ACE is increased in AD brain. We measured frontal ACE level and activity in 89 AD and 51 control brains, and post-mortem CSF from 101 cases and 19 controls. Neuron-specific enolase (NSE) level and Braak stage were used to indicate neuronal preservation and disease progression. We genotyped the common ACE insertion/deletion polymorphism, rs4343, rs1800764 and rs4921. ACE activity was elevated in AD and correlated with Braak stage. Crude ACE levels were unchanged but adjustment for NSE suggested increased neuronal ACE production with Braak stage. Exposing SH-SY-5Y neurons to oligomeric Abeta1-42 increased ACE level and activity, suggesting Abeta may upregulate ACE in AD. In CSF, ACE level but not activity was reduced in AD. ACE1 genotype did not predict ACE level or activity in brain or CSF. ACE activity and neuronal production increase in AD brain, possibly in response to Abeta. Peripheral measurements do not reflect ACE activity in the brain.

SELECTION OF CITATIONS
SEARCH DETAIL
...