Elsevier

Experimental Neurology

Volume 190, Issue 1, November 2004, Pages 192-203
Experimental Neurology

Alzheimer's pathology in human temporal cortex surgically excised after severe brain injury

https://doi.org/10.1016/j.expneurol.2004.06.011Get rights and content

Abstract

Traumatic brain injury (TBI) is a risk factor for the development of Alzheimer's disease (AD). This immunohistochemical study determined the extent of AD-related changes in temporal cortex resected from individuals treated surgically for severe TBI. Antisera generated against species (total , Aβ1–42, and Aβ1–40), the C-terminal of the Aβ precursor protein (APP), apolipoprotein E (apoE), and markers of neuron structure and degeneration (tau, ubiquitin, α-, β-, and γ-synuclein) were used to examine the extent of Aβ plaque deposition and neurodegenerative changes in 18 TBI subjects (ages 18–64 years). Diffuse cortical Aβ deposits were observed in one third of subjects (aged 35–62 years) as early as 2 h after injury, with only one (35-year old) individual exhibiting “mature”, dense-cored plaques. Plaque-like deposits, neurons, glia, and axonal changes were also immunostained with APP and apoE antibodies. In plaque-positive cases, the only statistically significant change in cellular immunostaining was increased neuronal APP (P = 0.013). There was no significant correlation between the distribution of Aβ plaques and markers of neuronal degeneration. Diffuse tau immunostaining was localized to neuronal cell soma, axons or glial cells in a larger subset of individuals. Tau-positive, neurofibrillary tangle (NFT)-like changes were detected in only two subjects, both of more advanced age and who were without Aβ deposits. Other neurodegenerative changes, evidenced by ubiquitin- and synuclein-immunoreactive neurons, were abundant in the majority of cases. Our results demonstrate a differential distribution and course of intra- and extra-cellular AD-like changes during the acute phase following severe TBI in humans. Aβ plaques and early evidence of neuronal degenerative changes can develop rapidly after TBI, while fully developed NFTs most likely result from more chronic disease- or injury-related processes. These observations lend further support to the hypothesis that head trauma significantly increases the risk of developing pathological and clinical symptoms of AD, and provide insight into the molecular mechanisms that initiate these pathological cascades very early during severe brain injury.

Introduction

Traumatic brain injury (TBI) is an environmental risk factor for the development of chronic neurodegenerative disorders, including Alzheimer's disease (AD). TBI correlates with accelerated cognitive decline in head-injured, aged subjects (Luukinen et al., 1999), and there is an increased risk for AD dementia in patients who, in adulthood, sustained a severe head injury Guo et al., 2000, McKenzie et al., 1994, Nemetz et al., 1999, Plassman et al., 2000, Roberts et al., 1991, Roberts et al., 1994, Roberts et al., 1993, Schofield et al., 1997. One of the detrimental consequences of TBI involves altered processing of amyloid precursor protein (APP), which potentially contributes to AD-like pathological cascades DeKosky et al., 1998, McIntosh et al., 1998. In AD, amyloid β peptide (Aβ) overproduction and deposition induce inflammatory cytokine infiltration with microglial activation and oxidative stress Altstiel and Sperber, 1991, Butterfield et al., 2002, Buxbaum et al., 1992, Forloni et al., 1992, Griffin et al., 1989, Griffin et al., 1995, the sequelae of which are also involved in trauma-related neurological dysfunction (Rothwell et al., 1997). TBI studies in both animal models Ciallella et al., 2002, Masumura et al., 2000, Murakami et al., 1998, Pierce et al., 1996, Smith et al., 1999, Uryu et al., 2002 and post-mortem human tissue Gentleman et al., 1993, Graham et al., 1995, McKenzie et al., 1994, Roberts et al., 1991, Roberts et al., 1994 demonstrated that increased APP production and/or accumulation can develop after injury. However, evidence of AD-like extracellular Aβ deposition has been described only in post-mortem human brain tissue so current knowledge of amyloidogenic APP metabolism and Aβ accumulation after human TBI derives almost exclusively from studies of victims of severe, lethal TBI (Roberts et al., 1994). Notably, AD-like neurodegenerative changes as well as intracellular accumulation of neurofibrillary tangle (NFT)-like pathologies were not found in these patients, with exception of subjects with repetitive head injury Geddes et al., 1999, Schmidt et al., 2001. In the present study, we collected freshly resected human brain tissue from subjects who underwent surgical treatment within hours of TBI and assessed the extent of AD-like extracellular and intracellular changes during this acute post-injury period. We further examined possible associations between these changes and variables such as subjects' age, primary lesion characteristics, hypothermia treatment, and functional outcome.

Section snippets

Subjects

The study examined 18 patients (age range 18–64 years), admitted to the University of Pittsburgh Medical Center (UPMC) between June 1999 and May 2001 for surgical treatment of severe closed head injuries (Glasgow Coma Scale, GCS score < 9). Patients' demographic and clinical variables are listed in Table 1. Data on the initial status (GCS; Teasdale and Jennett, 1974) and functional outcome at 3, 6, and 12 months after injury (Glasgow Outcome Scale, GOS; Jennett et al., 1981) were obtained for

AD plaque markers after TBI

In eight of the 18 subjects suffering severe TBI, temporal cortex contained AD-like plaque deposits (plaque-positive cases) of Aβ and/or related proteins (e.g., APP, apoE). There were no such plaques detectable in the remaining 10 (plaque-negative) TBI subjects, although some of these cases showed immunoreactive pyramidal neurons, axons, and glia (Table 3, Figs. 1D–F). Plaque-positive and plaque-negative groups were similar when compared by demographic variables and APOE genotype (percent of

Discussion

This analysis of surgically resected temporal cortex from survivors of severe TBI reveals that the formation of immunohistochemically detectable extracellular Aβ deposits can occur as early as 2 h after injury, demonstrating the extraordinary rapidity with which AD-like pathology can develop after acute neuronal insult. Extracellular deposits of Aβ, APP, and apoE were documented in 30% of subjects examined, corroborating and extending several previous post-mortem analyses Horsburgh et al., 2000

Acknowledgements

We thank Yetta I. Wilbur, Barbara A. Isanski, and Daniel Martinez for expert technical assistance. We are grateful to the University of Pittsburgh Alzheimer's Disease Research Center (AG05133), the Brain Trauma Research Center (NS30318), and Ava Puccio for brain tissue processing. We also thank Dr. Peter Davies for providing PHF-1 antibodies. Supported by NINDS NS30318, NIA AG05133, NIA AG11542, MH18273.

References (71)

  • G.W. Roberts et al.

    βA4 amyloid protein deposition in brain after head trauma

    Lancet

    (1991)
  • D.H. Smith et al.

    Brain trauma induces massive hippocampal neuron death linked to a surge in beta-amyloid levels in mice overexpressing mutant amyloid precursor protein

    Am. J. Pathol

    (1998)
  • J.R. Stone et al.

    Antibodies to the C-terminus of the beta-amyloid precursor protein (APP): a site specific marker for the detection of traumatic axonal injury

    Brain Res

    (2000)
  • G. Teasdale et al.

    Assessment of coma and impaired consciousness. A practical scale

    Lancet

    (1974)
  • J.Q. Trojanowski et al.

    Parkinson's disease and related synucleinopathies are a new class of nervous system amyloidoses

    Neurotoxicology

    (2002)
  • Anonymous

    National Institute on Aging and Reagan Institute working group on diagnosis criteria for the neuropathological assessment of Alzheimer's disease. Consensus recommendations for the postmortem diagnosis of AD

    Neurobiol. Aging

    (1997)
  • T. Arai et al.

    A high incidence of apolipoprotein E epsilon4 allele in middle-aged non-demented subjects with cerebral amyloid beta protein deposits

    Acta Neuropathol

    (1999)
  • D.A. Butterfield et al.

    Amyloid beta-peptide and amyloid pathology are central to the oxidative stress and inflammatory cascades under which Alzheimer's disease brain exists

    J. Alzheimer's Dis

    (2002)
  • J.D. Buxbaum et al.

    Cholinergic agonists and interleukin-1 regulate processing and secretion of the Alzheimer ß/A4 amyloid protein precursor

    Proc. Natl. Acad. Sci. U. S. A

    (1992)
  • J.R. Ciallella et al.

    Changes in expression of amyloid precursor protein and interleukin-1beta after experimental traumatic brain injury in rats

    J. Neurotrauma

    (2002)
  • R.S.B. Clark et al.

    Increases in Bcl-2 and cleavage of caspase-1 and caspase-3 in human brain after head injury

    FASEB

    (1999)
  • G.L. Clifton et al.

    A phase II study of moderate hypothermia in severe brain injury

    J. Neurotrauma

    (1993)
  • J. Clinton et al.

    Post-traumatic Alzheimer's disease: preponderance of a single plaque type

    Neuropathol. Appl. Neurobiol

    (1991)
  • H. Crystal et al.

    Clinico-pathologic studies in dementia: nondemented subjects with pathologically confirmed Alzheimer's disease

    Neurology

    (1988)
  • A.D. Dayan

    Quantitative histological studies on the aged human brain. I. Senile plaques and neurofibrillary tangles in “normal” patients

    Acta Neuropathol

    (1970)
  • S.T. DeKosky et al.

    Secondary injury after head trauma: subacute and long-term mechanisms

    Semin. Clin. Neuropsychiatry

    (1998)
  • D.W. Dickson et al.

    Identification of normal and pathological aging in prospectively studied nondemented elderly humans

    Neurobiol. Aging

    (1991)
  • J.F. Geddes et al.

    Neuronal cytoskeletal changes are an early consequence of repetitive head injury

    Acta Neuropathol

    (1999)
  • S.M. Gentleman et al.

    A beta 42 is the predominant form of amyloid beta-protein in the brains of short-term survivors of head injury

    NeuroReport

    (1997)
  • T. Gomez-Isla et al.

    Clinical and pathological correlates of apolipoprotein E epsilon 4 in Alzheimer's disease

    Ann. Neurol

    (1996)
  • D.I. Graham et al.

    Distribution of B-amyloid protein in the brain following severe head injury

    Neuropathol. Appl. Neurobiol

    (1995)
  • W.S.T. Griffin et al.

    Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer's disease

    Proc. Natl. Acad. Sci. U. S. A

    (1989)
  • W.S. Griffin et al.

    Interleukin-1 expression in different plaque types in Alzheimer's disease: significance in plaque evolution

    J. Neuropathol. Exp. Neurol

    (1995)
  • Z. Guo et al.

    Head injury and the risk of AD in the MIRAGE study

    Neurology

    (2000)
  • R.E. Hartman et al.

    Apolipoprotein E4 influences amyloid deposition but not cell loss after traumatic brain injury in a mouse model of Alzheimer's disease

    J. Neurosci

    (2002)
  • Cited by (362)

    View all citing articles on Scopus
    View full text