Your browser doesn't support javascript.
loading
Amyloidogenic Processing of Amyloid Precursor Protein Drives Stretch-Induced Disruption of Axonal Transport in hiPSC-Derived Neurons.
Chaves, Rodrigo S; Tran, My; Holder, Andrew R; Balcer, Alexandra M; Dickey, Andrea M; Roberts, Elizabeth A; Bober, Brian G; Gutierrez, Edgar; Head, Brian P; Groisman, Alex; Goldstein, Lawrence S B; Almenar-Queralt, Angels; Shah, Sameer B.
Afiliação
  • Chaves RS; Department of Cellular and Molecular Medicine, University of California, San Diego, California 92093.
  • Tran M; Sanford Consortium for Regenerative Medicine, La Jolla, California 92037.
  • Holder AR; Department of Orthopaedic Surgery, University of California, San Diego, California 92093.
  • Balcer AM; Sanford Consortium for Regenerative Medicine, La Jolla, California 92037.
  • Dickey AM; Sanford Consortium for Regenerative Medicine, La Jolla, California 92037.
  • Roberts EA; Sanford Consortium for Regenerative Medicine, La Jolla, California 92037.
  • Bober BG; Department of Cellular and Molecular Medicine, University of California, San Diego, California 92093.
  • Gutierrez E; Sanford Consortium for Regenerative Medicine, La Jolla, California 92037.
  • Head BP; Department of Cellular and Molecular Medicine, University of California, San Diego, California 92093.
  • Groisman A; Sanford Consortium for Regenerative Medicine, La Jolla, California 92037.
  • Goldstein LSB; Department of Bioengineering, University of California, San Diego, California 92093.
  • Almenar-Queralt A; Department of Physics, University of California, San Diego, California 92093.
  • Shah SB; Research Division, VA San Diego Healthcare System, San Diego, California 92161.
J Neurosci ; 41(49): 10034-10053, 2021 12 08.
Article em En | MEDLINE | ID: mdl-34663629
ABSTRACT
Traumatic brain injury (TBI) results in disrupted brain function following impact from an external force and is a risk factor for sporadic Alzheimer's disease (AD). Although neurologic symptoms triggered by mild traumatic brain injuries (mTBI), the most common form of TBI, typically resolve rapidly, even an isolated mTBI event can increase the risk to develop AD. Aberrant accumulation of amyloid ß peptide (Aß), a cleaved fragment of amyloid precursor protein (APP), is a key pathologic outcome designating the progression of AD following mTBI and has also been linked to impaired axonal transport. However, relationships among mTBI, amyloidogenesis, and axonal transport remain unclear, in part because of the dearth of human models to study the neuronal response following mTBI. Here, we implemented a custom-microfabricated device to deform neurons derived from human-induced pluripotent stem cells, derived from a cognitively unimpaired male individual, to mimic the mild stretch experienced by neurons during mTBI. Although no cell lethality or cytoskeletal disruptions were observed, mild stretch was sufficient to stimulate rapid amyloidogenic processing of APP. This processing led to abrupt cessation of APP axonal transport and progressive formation of aberrant axonal accumulations that contained APP, its processing machinery, and amyloidogenic fragments. Consistent with this sequence of events, stretch-induced defects were abrogated by reducing amyloidogenesis either pharmacologically or genetically. In sum, we have uncovered a novel and manipulable stretch-induced amyloidogenic pathway directly responsible for APP axonal transport dysregulation. Our findings may help to understand and ultimately mitigate the risk of developing AD following mTBI.SIGNIFICANCE STATEMENT Mild traumatic brain injury is a risk factor for sporadic Alzheimer's disease (AD). Increased amyloid ß peptide generation after injury may drive this risk. Here, by using a custom-built device to impose mild stretch to human neurons, we found that stretch triggers amyloid precursor protein (APP) cleavage, and thus amyloid ß peptide generation, consequently disrupting APP axonal transport. Compellingly, protecting APP from cleavage was sufficient to spare axonal transport dysregulation and the consequent aberrant axonal accumulation of APP. Supporting such protective mechanism, the expression of the AD-protective APPA673T genetic variant conferred protection against stretch-induced APP axonal transport phenotypes. Our data reveal potential subcellular pathways contributing to the development of AD-associated phenotypes following mild traumatic brain injury, and putative strategies for intervening in these pathways.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Transporte Axonal / Precursor de Proteína beta-Amiloide / Neurônios Tipo de estudo: Etiology_studies / Prognostic_studies / Risk_factors_studies Limite: Humans / Male Idioma: En Revista: J Neurosci Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Transporte Axonal / Precursor de Proteína beta-Amiloide / Neurônios Tipo de estudo: Etiology_studies / Prognostic_studies / Risk_factors_studies Limite: Humans / Male Idioma: En Revista: J Neurosci Ano de publicação: 2021 Tipo de documento: Article