Your browser doesn't support javascript.
loading
Soluble TREM2 in CSF and its association with other biomarkers and cognition in autosomal-dominant Alzheimer's disease: a longitudinal observational study.
Morenas-Rodríguez, Estrella; Li, Yan; Nuscher, Brigitte; Franzmeier, Nicolai; Xiong, Chengjie; Suárez-Calvet, Marc; Fagan, Anne M; Schultz, Stephanie; Gordon, Brian A; Benzinger, Tammie L S; Hassenstab, Jason; McDade, Eric; Feederle, Regina; Karch, Celeste M; Schlepckow, Kai; Morris, John C; Kleinberger, Gernot; Nellgard, Bengt; Vöglein, Jonathan; Blennow, Kaj; Zetterberg, Henrik; Ewers, Michael; Jucker, Mathias; Levin, Johannes; Bateman, Randall J; Haass, Christian.
  • Morenas-Rodríguez E; German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany. Electronic address: estrella.morenas.rodriguez@gmail.com.
  • Li Y; Division of Biostatistics, Washington University School of Medicine, St Louis, MO, USA.
  • Nuscher B; German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany.
  • Franzmeier N; Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany.
  • Xiong C; Division of Biostatistics, Washington University School of Medicine, St Louis, MO, USA.
  • Suárez-Calvet M; Barcelonaßeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Servei de Neurologia, Hospital del Mar Medical Research Institute, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Madrid, Spain.
  • Fagan AM; Department of Neurology, Washington University School of Medicine, St Louis, MO, USA.
  • Schultz S; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA.
  • Gordon BA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA.
  • Benzinger TLS; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA.
  • Hassenstab J; Department of Neurology, Washington University School of Medicine, St Louis, MO, USA.
  • McDade E; Department of Neurology, Washington University School of Medicine, St Louis, MO, USA.
  • Feederle R; German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute for Diabetes and Obesity, Monoclonal Antibody Core Facility, Helmholtz Center, Munich, Germany; German Research Center for Environmental Health, Neuherberg, Germa
  • Karch CM; Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA.
  • Schlepckow K; German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany.
  • Morris JC; Department of Neurology, Washington University School of Medicine, St Louis, MO, USA.
  • Kleinberger G; German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany.
  • Nellgard B; Department of Anesthesiology and Intensive Care, Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
  • Vöglein J; German Center for Neurodegenerative Diseases, Munich, Germany; Department of Neurology, University Hospital of Munich, Ludwig-Maximilians University, Munich, Germany.
  • Blennow K; Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
  • Zetterberg H; Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Queens Square Institute of Neurology, University College
  • Ewers M; German Center for Neurodegenerative Diseases, Munich, Germany; Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany.
  • Jucker M; German Center for Neurodegenerative Diseases, Tübingen, Germany; Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, Tübingen, Germany.
  • Levin J; German Center for Neurodegenerative Diseases, Munich, Germany; Department of Neurology, University Hospital of Munich, Ludwig-Maximilians University, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
  • Bateman RJ; Department of Neurology, Washington University School of Medicine, St Louis, MO, USA.
  • Haass C; German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
Lancet Neurol ; 21(4): 329-341, 2022 04.
Article en En | MEDLINE | ID: mdl-35305339
ABSTRACT

BACKGROUND:

Therapeutic modulation of TREM2-dependent microglial function might provide an additional strategy to slow the progression of Alzheimer's disease. Although studies in animal models suggest that TREM2 is protective against Alzheimer's pathology, its effect on tau pathology and its potential beneficial role in people with Alzheimer's disease is still unclear. Our aim was to study associations between the dynamics of soluble TREM2, as a biomarker of TREM2 signalling, and amyloid ß (Aß) deposition, tau-related pathology, neuroimaging markers, and cognitive decline, during the progression of autosomal dominant Alzheimer's disease.

METHODS:

We did a longitudinal analysis of data from the Dominantly Inherited Alzheimer Network (DIAN) observational study, which includes families with a history of autosomal dominant Alzheimer's disease. Participants aged over 18 years who were enrolled in DIAN between Jan 1, 2009, and July 31, 2019, were categorised as either carriers of pathogenic variants in PSEN1, PSEN2, and APP genes (n=155) or non-carriers (n=93). We measured amounts of cleaved soluble TREM2 using a novel immunoassay in CSF samples obtained every 2 years from participants who were asymptomatic (Clinical Dementia Rating [CDR]=0) and annually for those who were symptomatic (CDR>0). CSF concentrations of Aß40, Aß42, total tau (t-tau), and tau phosphorylated on threonine 181 (p-tau) were measured by validated immunoassays. Predefined neuroimaging measurements were total cortical uptake of Pittsburgh compound B PET (PiB-PET), cortical thickness in the precuneus ascertained by MRI, and hippocampal volume determined by MRI. Cognition was measured using a validated cognitive composite (including DIAN word list test, logical memory delayed recall, digit symbol coding test [total score], and minimental status examination). We based our statistical analysis on univariate and bivariate linear mixed effects models.

FINDINGS:

In carriers of pathogenic variants, a high amyloid burden at baseline, represented by low CSF Aß42 (ß=-4·28 × 10-2 [SE 0·013], p=0·0012), but not high cortical uptake in PiB-PET (ß=-5·51 × 10-3 [0·011], p=0·63), was the only predictor of an augmented annual rate of subsequent increase in soluble TREM2. Augmented annual rates of increase in soluble TREM2 were associated with a diminished rate of decrease in amyloid deposition, as measured by Aß42 in CSF (r=0·56 [0·22], p=0·011), in presymptomatic carriers of pathogenic variants, and with diminished annual rate of increase in PiB-PET (r=-0·67 [0·25], p=0·0060) in symptomatic carriers of pathogenic variants. Presymptomatic carriers of pathogenic variants with annual rates of increase in soluble TREM2 lower than the median showed a correlation between enhanced annual rates of increase in p-tau in CSF and augmented annual rates of increase in PiB-PET signal (r=0·45 [0·21], p=0·035), that was not observed in those with rates of increase in soluble TREM2 higher than the median. Furthermore, presymptomatic carriers of pathogenic variants with rates of increase in soluble TREM2 above or below the median had opposite associations between Aß42 in CSF and PiB-PET uptake when assessed longitudinally. Augmented annual rates of increase in soluble TREM2 in presymptomatic carriers of pathogenic variants correlated with decreased cortical shrinkage in the precuneus (r=0·46 [0·22]), p=0·040) and diminished cognitive decline (r=0·67 [0·22], p=0·0020).

INTERPRETATION:

Our findings in autosomal dominant Alzheimer's disease position the TREM2 response within the amyloid cascade immediately after the first pathological changes in Aß aggregation and further support the role of TREM2 on Aß plaque deposition and compaction. Furthermore, these findings underpin a beneficial effect of TREM2 on Aß deposition, Aß-dependent tau pathology, cortical shrinkage, and cognitive decline. Soluble TREM2 could, therefore, be a key marker for clinical trial design and interpretation. Efforts to develop TREM2-boosting therapies are ongoing.

FUNDING:

German Research Foundation, US National Institutes of Health.
Asunto(s)

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Glicoproteínas de Membrana / Enfermedad de Alzheimer / Disfunción Cognitiva Tipo de estudio: Observational_studies / Prognostic_studies / Risk_factors_studies Límite: Adult / Humans / Middle aged País como asunto: America do norte Idioma: En Año: 2022 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Glicoproteínas de Membrana / Enfermedad de Alzheimer / Disfunción Cognitiva Tipo de estudio: Observational_studies / Prognostic_studies / Risk_factors_studies Límite: Adult / Humans / Middle aged País como asunto: America do norte Idioma: En Año: 2022 Tipo del documento: Article