Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Brain Behav Immun ; 36: 9-14, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24145049

ABSTRACT

Immunological privilege appears to be a product of unique lymphatic drainage systems for the brain and receptor-mediated entry of inflammatory cells through the blood-brain barrier. Most organs of the body have well-defined lymphatic vessels that carry extracellular fluid, antigen presenting cells, lymphocytes, neoplastic cells and even bacteria to regional lymph nodes. The brain has no such conventional lymphatics, but has perivascular pathways that drain interstitial fluid (ISF) from brain parenchyma and cerebrospinal fluid (CSF) from the subarachnoid space to cervical lymph nodes. ISF and solutes drain along narrow, ∼100 nm-thick basement membranes within the walls of cerebral capillaries and arteries to cervical lymph nodes; this pathway does not allow traffic of lymphocytes or antigen presenting cells from brain to lymph nodes. Although CSF drains into blood through arachnoid villi, CSF also drains from the subarachnoid space through channels in the cribriform plate of the ethmoid bone into nasal lymphatics and thence to cervical lymph nodes. This pathway does allow the traffic of lymphocytes and antigen presenting cells from CSF to cervical lymph nodes. Efferent pathways by which lymphocytes enter the brain are regulated by selected integrins on lymphocytes and selective receptors on vascular endothelial cells. Here we review: (1) the structure and function of afferent lymphatic drainage of ISF and CSF, (2) mechanisms involved in the efferent pathways by which lymphocytes enter the brain and (3) the failure of lymphatic drainage of the brain parenchyma with age and the role of such failure in the pathogenesis of Alzheimer's disease.


Subject(s)
Alzheimer Disease/immunology , Brain/immunology , Lymphatic System/immunology , Lymphocytes/immunology , Animals , Cerebrospinal Fluid/physiology , Extracellular Fluid/physiology , Humans
2.
Neuropathol Appl Neurobiol ; 39(6): 593-611, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23489283

ABSTRACT

Failure of elimination of proteins from the brain is a major feature in many neurodegenerative diseases. Insoluble proteins accumulate in brain parenchyma and in walls of cerebral capillaries and arteries. Cerebral amyloid angiopathy (CAA) is a descriptive term for amyloid in vessel walls. Here, we adopt the term protein elimination failure angiopathy (PEFA) to focus on mechanisms involved in the pathogenesis of a spectrum of disorders that exhibit both unique and common features of protein accumulation in blood vessel walls. We review (a) normal pathways and mechanisms by which proteins and other soluble metabolites are eliminated from the brain along 100- to 150-nm-thick basement membranes in walls of cerebral capillaries and arteries that serve as routes for lymphatic drainage of the brain; (b) a spectrum of proteins involved in PEFA; and (c) changes that occur in artery walls and contribute to failure of protein elimination. We use accumulation of amyloid beta (Aß), prion protein and granular osmiophilic material (GOM) in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) as examples of different factors involved in the aetiology and pathogenesis of PEFA. Finally, we discuss how knowledge of factors involved in PEFA may help to focus on new therapies for neurodegenerative diseases. When Aß (following immunotherapy) and prion protein are released from brain parenchyma they deposit in walls of cerebral capillaries and arteries; GOM in CADASIL accumulates primarily in artery walls. Therefore, the focus of therapy for protein clearance in neurodegenerative disease should perhaps be on facilitating perivascular elimination of proteins and reducing PEFA.


Subject(s)
CADASIL/etiology , Cerebral Amyloid Angiopathy/etiology , Cerebral Arterial Diseases/etiology , Neurodegenerative Diseases/therapy , Prion Diseases/etiology , Amyloidogenic Proteins/metabolism , Brain/blood supply , Brain/pathology , CADASIL/metabolism , Cerebral Amyloid Angiopathy/metabolism , Cerebral Arterial Diseases/metabolism , Cerebrovascular Circulation , Humans , Prion Diseases/metabolism
3.
Pathophysiology ; 17(4): 295-306, 2010 Sep.
Article in English | MEDLINE | ID: mdl-19954936

ABSTRACT

In most organs of the body, immunological reactions involve the drainage of antigens and antigen presenting cells (APCs) along defined lymphatic channels to regional lymph nodes. The CNS is considered to be an immunologically privileged organ with no conventional lymphatics. However, immunological reactions do occur in the CNS in response to infections and in immune-mediated disorders such as multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). Here, we review evidence that cervical lymph nodes play a role in B and T cell mediated immune reactions in the CNS. Then we define the separate pathways by which interstitial fluid (ISF) and CSF drain to cervical lymph nodes. ISF and solutes drain from the brain along the 100-150nm-wide basement membranes in the walls of capillaries and arteries. In humans, this perivascular pathway is outlined by the deposition of insoluble amyloid (Abeta) in capillary and artery walls in cerebral amyloid angiopathy in Alzheimer's disease. The failure of APCs to migrate to lymph nodes along perivascular lymphatic drainage pathways may be a major factor in immunological privilege of the brain. Lymphatic drainage of CSF is predominantly through the cribriform plate into nasal lymphatics. Lymphatic drainage of ISF and CSF and the specialised cervical lymph nodes to which they drain play significant roles in the induction of immunological tolerance and of adaptive immunological responses in the CNS. Understanding the afferent and efferent arms of the CNS lymphatic system will be valuable for the development of therapeutic strategies for diseases such as MS.

4.
Clin Anat ; 23(1): 43-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19918869

ABSTRACT

The brain has no conventional lymphatics, but solutes injected into it drain along artery walls and reach lymph nodes in the neck. This study seeks to identify cervical lymph nodes related to the human internal carotid artery (ICA) that could act as the first regional lymph nodes for the brain. Bilateral dissections were carried out on four embalmed human heads, from the level of the carotid bifurcation in the neck, to the base of the skull. Lymph nodes from every specimen were processed for histological examination. A total of 51 deep cervical lymph nodes were identified: 12 lymph nodes (confirmed by histological examination) were observed to be in direct relationship with the ICA. These lymph nodes were found within the carotid sheath and had average diameters of 13.5 x 4.8 mm. Solutes and interstitial fluid from the brain may drain along the walls of cerebral arteries and reach these lymph nodes. They may be sites of stimulation of immune responses against antigens from the brain.


Subject(s)
Carotid Artery, Internal/anatomy & histology , Lymph Nodes/anatomy & histology , Skull Base/anatomy & histology , Aged , Aged, 80 and over , Female , Humans , Male
5.
Brain ; 131(Pt 12): 3299-310, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18953056

ABSTRACT

A major feature of Alzheimer's disease is the accumulation of amyloid-beta peptide (Abeta) in the brain both in the form of plaques in the cerebral cortex and in blood vessel as cerebral amyloid angiopathy (CAA). Experimental models and human clinical trials have shown that accumulation of Abeta plaques can be reversed by immunotherapy. In this study, we hypothesized that Abeta in plaques is solubilized by antibodies generated by immunization and drains via the perivascular pathway, detectable as an increase in cerebrovascular Abeta. We have performed a follow up study of Alzheimer's disease patients immunized against Abeta42. Neuropathological examination was performed on nine patients who died between four months and five years after their first immunization. Immunostaining for Abeta40 and Abeta42 was quantified and compared with that in unimmunized Alzheimer's disease controls (n = 11). Overall, compared with these controls, the group of immunized patients had approximately 14 times as many blood vessels containing Abeta42 in the cerebral cortex (P<0.001) and seven times more in the leptomeninges (P = 0.013); among the affected blood vessels in the immunized cases, most of them had full thickness and full circumference involvement of the vessel wall in the cortex (P = 0.001), and in the leptomeninges (P = 0.015). There was also a significantly higher level of cerebrovascular Abeta40 in the immunized cases than in the unimmunized cases (cortex: P = 0.009 and leptomeninges: P = 0.002). In addition, the immunized patients showed a higher density of cortical microhaemorrhages and microvascular lesions than the unimmunized controls, though none had major CAA-related intracerebral haemorrhages. The changes in cerebral vascular Abeta load did not appear to substantially influence the structural proteins of the blood vessels. Unlike most of the immunized patients, two of the longest survivors, four to five years after first immunization, had virtually complete absence of both plaques and CAA, raising the possibility that, given time, Abeta is eventually cleared from the cerebral vasculature. The findings are consistent with the hypothesis that Abeta immunization results in solubilization of plaque Abeta42 which, at least in part, exits the brain via the perivascular pathway, causing a transient increase in the severity of CAA. The extent to which these vascular alterations following Abeta immunization in Alzheimer's disease are reflected in changes in cognitive function remains to be determined.


Subject(s)
Alzheimer Disease/therapy , Alzheimer Vaccines/therapeutic use , Amyloid beta-Peptides/immunology , Cerebral Amyloid Angiopathy/therapy , Peptide Fragments/immunology , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Blood Vessels/metabolism , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Cerebral Hemorrhage/etiology , Female , Follow-Up Studies , Humans , Immunotherapy, Active/methods , Male , Meninges/blood supply , Meninges/metabolism , Middle Aged , Peptide Fragments/metabolism , Solubility
6.
Neuropathol Appl Neurobiol ; 34(2): 131-44, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18208483

ABSTRACT

UNLABELLED: Elimination of interstitial fluid and solutes plays a role in homeostasis in the brain, but the pathways are unclear. Previous work suggests that interstitial fluid drains along the walls of arteries. AIMS: to define the pathways within the walls of capillaries and arteries for drainage of fluid and solutes out of the brain. METHODS: Fluorescent soluble tracers, dextran (3 kDa) and ovalbumin (40 kDa), and particulate fluospheres (0.02 microm and 1.0 microm in diameter) were injected into the corpus striatum of mice. Brains were examined from 5 min to 7 days by immunocytochemistry and confocal microscopy. RESULTS: soluble tracers initially spread diffusely through brain parenchyma and then drain out of the brain along basement membranes of capillaries and arteries. Some tracer is takenf up by vascular smooth muscle cells and by perivascular macrophages. No perivascular drainage was observed when dextran was injected into mouse brains following cardiac arrest. Fluospheres expand perivascular spaces between vessel walls and surrounding brain, are ingested by perivascular macrophages but do not appear to leave the brain even following an inflammatory challenge with lipopolysaccharide or kainate. CONCLUSIONS: capillary and artery basement membranes act as 'lymphatics of the brain' for drainage of fluid and solutes; such drainage appears to require continued cardiac output as it ceases following cardiac arrest. This drainage pathway does not permit migration of cells from brain parenchyma to the periphery. Amyloid-beta is deposited in basement membrane drainage pathways in cerebral amyloid angiopathy, and may impede elimination of amyloid-beta and interstitial fluid from the brain in Alzheimer's disease. Soluble antigens, but not cells, drain from the brain by perivascular pathways. This atypical pattern of drainage may contribute to partial immune privilege of the brain and play a role in neuroimmunological diseases such as multiple sclerosis.


Subject(s)
Basement Membrane/metabolism , Brain/physiology , Cerebral Amyloid Angiopathy/physiopathology , Extracellular Fluid/metabolism , Extracellular Space/metabolism , Animals , Arteries/metabolism , Brain/blood supply , Capillaries/metabolism , Dextrans/metabolism , Immunohistochemistry , Mice , Microscopy, Confocal , Ovalbumin/metabolism
8.
J Theor Biol ; 238(4): 962-74, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16112683

ABSTRACT

Experimental studies and observations in the human brain indicate that interstitial fluid and solutes, such as amyloid-beta (Abeta), are eliminated from grey matter of the brain along pericapillary and periarterial pathways. It is unclear, however, what constitutes the motive force for such transport within blood vessel walls, which is in the opposite direction to blood flow. In this paper the potential for global pressure differences to achieve such transport are considered. A mathematical model is constructed in order to test the hypothesis that perivascular drainage of interstitial fluid and solutes out of brain tissue is driven by pulsations of the blood vessel walls. Here it is assumed that drainage occurs through a thin layer between astrocytes and endothelial cells or between smooth muscle cells. The model suggests that, during each pulse cycle, there are periods when fluid and solutes are driven along perivascular spaces in the reverse direction to the flow of blood. It is shown that successful drainage may depend upon some attachment of solutes to the lining of the perivascular space, in order to produce a valve-like effect, although an alternative without this requirement is also postulated. Reduction in pulse amplitude, as in ageing cerebral vessels, would prolong the attachment time, encourage precipitation of Abeta peptides in vessel walls, and impair elimination of Abeta from the brain. These factors may play a role in the pathogenesis of cerebral amyloid angiopathy and in the accumulation of Abeta in the brain in Alzheimer's disease.


Subject(s)
Brain/metabolism , Cerebrovascular Circulation/physiology , Models, Neurological , Amyloid beta-Peptides/blood , Biological Transport, Active , Cerebral Arteries/physiology , Elasticity , Extracellular Fluid/metabolism , Humans , Pulsatile Flow
9.
Morphologie ; 89(284): 22-34, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15943078

ABSTRACT

The meninges comprise the dura mater and the leptomeninges (arachnoid and pia mater). Dura forms an outer endosteal layer related to the bones of the skull and spine and an inner layer closely applied to the arachnoid mater. Leptomeninges have multiple functions and anatomical relationships. The outer parietal layer of arachnoid is impermeable to CSF due to tight intercellular junctions; elsewhere leptomeningeal cells form demosomes and gap junctions. Trabeculae of leptomeninges compartmentalize the subarachnoid space and join the pia to arachnoid mater. In bacterial meningitis leptomeningeal cells secrete cytokines. Pia mater is reflected from the surface of the brain and spinal cord onto arteries and veins, thus separating the subarachnoid space from the brain and cord. A sheath of leptomeninges accompanies arteries into the brain and is related to the pathways for the drainage of interstitial fluid that play a role in inflammatory responses in the brain and appear to be blocked by amyloid-beta in Alzheimer's disease. Specialised leptomeningeal cells in the stroma of the choroid plexus form collagen whorls that become calcified with age. Leptomeningeal cells also form channels in the core and apical cap of arachnoid granulations for the drainage of CSF into venous sinuses. In the spine, leptomeninges form highly perforated intermediate sheets of arachnoid and delicate ligaments that compartmentalize the subarachnoid space; dentate ligaments anchor subpial collagen to the dura mater and stabilize the spinal cord. Despite the multiple anatomical arrangements and physiological functions, leptomeningeal cells retain many histological features that are similar from site to site.


Subject(s)
Meninges/anatomy & histology , Humans , Meninges/ultrastructure , Microscopy, Electron
10.
Neuropathol Appl Neurobiol ; 30(4): 374-84, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15305983

ABSTRACT

UNLABELLED: Multiple sclerosis and experimental autoimmune encephalomyelitis (EAE) are autoimmune inflammatory diseases in which cytokines are intimately involved. Here we test the hypothesis that injection of pro-inflammatory cytokines, tumour necrosis factor-alpha (TNFalpha) and interferon gamma (IFNgamma) into the brain of animals in the prodromal phase of EAE significantly enhances inflammation in the central nervous system (CNS). We were particularly interested to learn whether a local increase in cytokines influenced the pathology locally, or more extensively, within the CNS. EAE was induced in female adult Lewis rats. Eight days post-inoculation, TNFalpha or INFgamma was injected into one cerebral hemisphere. Days 11 and 13 post-inoculation (3 and 5 days after the injection of cytokine) inflammation was quantified by the number of perivascular cuffs and the degree of major histocompatibility complex (MHC) class II expression by microglia. Normal animals injected with cytokines, and EAE animals with saline injection served as controls. RESULTS: microglial activation was increased three- to fourfold in the brain and eightfold in the spinal cord (P

Subject(s)
Autoimmune Diseases/pathology , Cytokines/metabolism , Encephalitis/pathology , Multiple Sclerosis/pathology , Myelitis/pathology , Animals , Autoimmune Diseases/metabolism , Axons/pathology , Brain/metabolism , Brain/pathology , Brain Chemistry , Encephalitis/metabolism , Female , Interferon-gamma/metabolism , Lymphocytes/pathology , Lymphocytes/physiology , Microglia/pathology , Microglia/physiology , Multiple Sclerosis/metabolism , Myelitis/metabolism , Nerve Degeneration/pathology , Rats , Rats, Inbred Lew , Spinal Cord/metabolism , Spinal Cord/pathology , Stereotaxic Techniques , Tumor Necrosis Factor-alpha/pharmacology
11.
Synapse ; 53(1): 36-43, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15150739

ABSTRACT

Vigabatrin, the gamma-aminobutyric acid transaminase (GABA-T)-inhibiting anticonvulsant drug, was given orally at a dose of 275 mg/kg/day to rats (n = 6) in their feed for a period of 12 weeks, during which T2-weighted magnetic resonance images (MRIs) and diffusion-weighted MRIs (DWIs) were collected at weeks 1, 3, 6, 9, and 12. Half the rats (n = 3; and half their age-matched littermate controls; n = 3) were then killed for histopathological confirmation of the observed VGB-induced cerebellar and cortical white-matter lesions. VGB was removed from the diet and additional MRIs of the remaining rats taken at weeks 14, 17, 20, and 24, at which time they (n = 3), along with remaining controls (n = 3), were also killed for histopathology. The T2-weighted MRIs acquired were used to compute T2 relaxation time maps. Statistically significant VGB-induced T2 increases were observed in the frontal and occipital cortices and in the cerebellar white matter (CWM). The cerebellar lesions were more clearly discerned by eye in the DWIs than by T2-contrast alone. During the recovery period the VGB-treatment group CWM-T2 and CWM-DWI hyperintensity greatly decreased as the reversible lesion disappeared. As expected, histological and immunocytochemical examinations demonstrated the presence of intra-myelinic edema, microvacuolation, and reactive astrocytosis in the CWM and cortex after 12 weeks VGB-treatment. In the remaining animals microvacuolation of the white matter had not completely resolved during the 12-week recovery phase. The data show that quantitative MRI T2-relaxometry can be used to detect VGB-induced CNS pathology, and also suggest that DWI is particularly sensitive to the cerebellar lesion. The reversible neurotoxicity of global GABA-elevation in experimental animals is discussed.


Subject(s)
Anticonvulsants/toxicity , Brain/drug effects , Brain/pathology , Vigabatrin/toxicity , Animals , Immunohistochemistry , Magnetic Resonance Imaging , Male , Rats , Rats, Sprague-Dawley , Time Factors
12.
Panminerva Med ; 46(4): 239-51, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15876980

ABSTRACT

Dementia is a disease of the elderly, and although there are many causes of dementia, Alzheimer's disease (AD) and vascular dementia (VaD) account for the majority of cases world- wide. Many patients with dementia have radiological and neuropathological features of AD and VaD, with the classical neurofibrillary tangles and senile amyloid-beta (Abeta) plaques of AD together with the cerebral infarcts of VaD. In this review we examine the close relationship between AD and VaD and suggest that the age changes in cerebral blood vessels that are the basis of cerebrovascular disease and VaD may also be responsible for the failure of elimination of Abeta from the brain in AD. Abeta appears to be eliminated along the perivascular pathways by which interstitial fluid (ISF) drains from the brain (effectively the lymphatics of the brain). In aged individuals, insoluble Abeta amyloid fibrils are deposited in the ISF drainage pathways resulting in cerebral amyloid angiopathy (CAA). We review the evidence that age changes in cerebral arteries and cerebrovascular disease inhibit the perivascular drainage of ISF and Abeta along the walls of cerebral arteries resulting in the accumulation of insoluble and soluble Abeta in the brain in AD. Therapies for AD are reviewed, especially those involving immunotherapy for the removal of insoluble Abeta from the cerebral cortex and the facilitation of drainage of ISF and soluble Abeta from the brain.


Subject(s)
Alzheimer Disease/physiopathology , Cerebrovascular Disorders/physiopathology , Aged , Aging/pathology , Aging/physiology , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Brain/physiopathology , Cerebrovascular Disorders/pathology , Dementia, Vascular/pathology , Dementia, Vascular/physiopathology , Dementia, Vascular/therapy , Extracellular Fluid/metabolism , Humans , Models, Neurological , Neurofibrillary Tangles/pathology , Risk Factors
13.
Lancet ; 362(9380): 295-6, 2003 Jul 26.
Article in English | MEDLINE | ID: mdl-12892962

ABSTRACT

In animals, high doses of intramuscular artemether and artemotil have been shown to cause an unusual pattern of selective damage to certain brainstem nuclei, especially those implicated in hearing and balance. We aimed to investigate whether a similar pattern arises in human adults. We examined the brainstems of adults who died after treatment with high dose artemether or quinine for severe falciparum malaria for evidence of a pattern of selective neuronal damage. Neuropathological findings were similar in recipients of quinine (n=15) and artemether (n=6; total artemether doses received 4-44 mg/kg). No evidence was recorded for artemether-induced neurotoxic effects.


Subject(s)
Antimalarials/adverse effects , Artemisinins/adverse effects , Brain Diseases/chemically induced , Brain Diseases/pathology , Malaria, Falciparum/drug therapy , Sesquiterpenes/adverse effects , Adult , Antimalarials/therapeutic use , Artemether , Artemisinins/therapeutic use , Brain Stem/drug effects , Brain Stem/pathology , Female , Humans , Malaria, Falciparum/pathology , Male , Quinine/adverse effects , Quinine/therapeutic use , Sesquiterpenes/therapeutic use
14.
Neuropathol Appl Neurobiol ; 29(2): 106-17, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12662319

ABSTRACT

Accumulation of amyloid beta (Abeta) in the extracellular spaces of the cerebral cortex and in blood vessel walls as cerebral amyloid angiopathy is a characteristic of Alzheimer's disease (AD) and the ageing human brain. Studies in animals suggest that Abeta is eliminated from the brain either directly into the blood or along perivascular interstitial fluid drainage channels. The aim of the present study is to define the perivascular route for the drainage of Abeta from the human brain. Smears and paraffin sections of post-mortem cortical tissue from 17 cases of AD and from two controls were stained with thioflavin and for Abeta by immunohistochemistry. Histology and confocal microscopy showed that deposits of Abeta in the cortical parenchyma were continuous with Abeta in capillary walls but Abeta in artery walls was not in continuity with Abeta in brain parenchyma. Quantitative studies supported these observations. The results of this study suggest that when Abeta is eliminated from the extracellular spaces of the human brain by the perivascular route, it enters pericapillary spaces and from there drains along the walls of cortical arteries to leptomeningeal arteries. Factors such as overproduction of Abeta, entrapment of Abeta in drainage pathways and poor drainage of Abeta due to functional changes in ageing arteries might result in the failure of elimination of Abeta from the ageing brain and play a major role in the pathogenesis of AD. Such factors might affect therapies for AD that entail administration of anti-Abeta antibodies to eliminate Abeta from the human brain.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Arteries/pathology , Capillaries/pathology , Cerebral Amyloid Angiopathy/metabolism , Cerebral Cortex/metabolism , Female , Humans , Immunohistochemistry , Male , Microscopy, Confocal , Middle Aged
17.
J Pathol ; 194(1): 1-3, 2001 May.
Article in English | MEDLINE | ID: mdl-11329133

ABSTRACT

Analysis of lumbar cerebrospinal fluid (CSF) plays a major role in the investigation of central nervous system disease, but how well do the changes in the CSF reflect pathology within the brain and spinal cord parenchyma? Both Creutzfeldt-Jakob (CJD) and Alzheimer's disease (AD) are characterized by the deposition of insoluble beta-pleated sheet peptides [prion protein (PrP) and beta-amyloid (Abeta), respectively] in the extracellular spaces of grey matter in the brain, but there is discordance in both diseases between the peptide levels in the brain and in the CSF. Experimental studies using tracers have shown that interstitial fluid (ISF) drains through very narrow intercellular spaces within grey matter into bulk flow perivascular channels that surround penetrating arteries. ISF then flows to the surface of the brain and joins CSF to drain to cervical lymph nodes. Such drainage of ISF and CSF to regional lymph nodes in the rat plays a significant role in B-cell and T-cell immune reactions within the brain. In man, the pia mater separates the periarterial ISF drainage pathways from the CSF in the subarachnoid space. The almost complete lack of insoluble protease-resistant PrP entering the CSF from the brain in patients with CJD, reported by Wong et al. in this issue of the Journal of Pathology, illustrates the limitations of ISF drainage pathways for the elimination of insoluble peptides from brain tissue. Insoluble Abeta accumulates in the extracellular spaces as plaques in AD and in periarterial ISF drainage pathways as cerebral amyloid angiopathy. Soluble Abeta appears to become entrapped by the insoluble Abeta in the ISF drainage pathways; thus, as the level of soluble Abeta in the brain rises in AD, the level in the CSF falls. Thus, the changes in the CSF do not accurately reflect the accumulation of the abnormal peptides in the brain parenchyma in either CJD or AD. In both diseases, facilitation of ISF drainage and elimination of PrP and Abeta peptides from the extracellular spaces of the brain may lead to practical therapeutic strategies for these devastating disorders.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Creutzfeldt-Jakob Syndrome/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Humans , Prions/cerebrospinal fluid
19.
Mol Med ; 7(9): 609-18, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11778650

ABSTRACT

BACKGROUND: High levels of A beta in the cerebral cortex distinguish demented Alzheimer's disease (AD) from nondemented elderly individuals, suggesting that decreased amyloid-beta (A beta) peptide clearance from the brain is a key precipitating factor in AD. MATERIALS AND METHODS: The levels of A beta in brain and plasma as well as apolipoprotein E (ApoE) in brain were investigated by enzyme-linked immunosorbent assay (ELISA) and Western blotting at various times during the life span of the APP23 transgenic (Tg) and control mice. Histochemistry and immunocytochemistry were used to assess the morphologic characteristics of the brain parenchymal and cerebrovascular amyloid deposits and the intracellular amyloid precursor protein (APP) deposits in the APP23 Tg mice. RESULTS: No significant differences were found in the plasma levels of A beta between the APP23 Tg and control mice from 2-20 months of age. In contrast, soluble A beta levels in the brain were continually elevated, increasing 4-fold at 2 months and 33-fold in the APP23 Tg mice at 20 months of age when compared to the control mice. Soluble A beta42 was about 60% higher than A beta40. In the APP23 Tg mice, insoluble A beta40 remained at basal levels in the brain until 9 months and then rose to 680 microg/g cortex by 20 months. Insoluble A beta40 was negligible in non-Tg mice at all ages. Insoluble A beta42 in APP23 Tg mice rose to 60 microg/g cortex at 20 months, representing 24 times the control A beta42 levels. Elevated levels of ApoE in the brain were observed in the APP23 Tg mice at 2 months of age, becoming substantially higher by 20 months. ApoE colocalized with A beta in the plaques. Beta-amyloid precursor protein (betaAPP) deposits were detected within the neuronal cytoplasm from 4 months of age onward. Amyloid angiopathy in the APP23 Tg mice increased markedly with age, being by far more severe than in the Tg2576 mice. CONCLUSIONS: We suggest that the APP23 Tg mouse may develop an earlier blockage in A beta clearance than the Tg2576 mice, resulting in a more severe accumulation of A beta in the perivascular drainage pathways and in the brain. Both Tg mice reflect decreased A beta elimination and as models for the amyloid cascade they are useful to study AD pathophysiology and therapy.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Peptide Fragments/metabolism , Aging/physiology , Alzheimer Disease/etiology , Amyloid beta-Peptides/blood , Animals , Apolipoproteins E/metabolism , Brain/blood supply , Brain/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Peptide Fragments/blood , Time Factors
20.
Mol Med ; 6(5): 430-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10952022

ABSTRACT

BACKGROUND: Amyloid-beta (A beta) accumulates in plaques and as cerebral amyloid angiopathy (CAA) in the brains of both Alzheimer's disease (AD) patients and transgenic A betaPPswe/tg2576 (tg2576) mice. Increasingly, evidence in humans and mice shows this process to be modulated by apolipoprotein E (apoE). MATERIALS AND METHODS: To explore this relationship, we measured apoE and A beta levels in brains of tg2576 mice and controls at intervals between 2 and 20 months. In addition, A beta concentrations in plasma and muscle of these animals were also quantified. RESULTS: Quite strikingly, we found that the amount of tg2576 mice brain apoE was elevated by an average of 45%, relative to the control mice from 2 months on. The level of brain apoE soared after 14 months to almost 60% greater than the level found in control mice. A beta concentrations in brains before 9 months were less than 2 ng/mg of protein, but by 14 months concentrations rose to 8.7 ng/mg, and by 20 months to 47 ng/mg. In plasma, we noted that the levels of A beta in tg2576 mice declined from above 30 ng/ml prior to 12 months to 14 ng/ml by 14 months. Histology showed that A beta plaques and CAA began to be discernible in the tg2576 mice at about 9 and 20 months of age, respectively. CONCLUSIONS: ApoE was immunocytochemically detected in neuritic plaques that were positive for thioflavine-S. We suggest that the elevation of brain apoE in tg2576 mice participates in an age-related dysregulation of A beta clearance and signals the start of A beta sequestration during the time of cognitive dysfunction.


Subject(s)
Aging , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Apolipoproteins E/genetics , Brain/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid/analysis , Amyloid/metabolism , Animals , Apolipoproteins E/metabolism , Brain/growth & development , Brain/pathology , Humans , Learning Disabilities/etiology , Memory Disorders/etiology , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...