Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cell ; 182(4): 976-991.e19, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32702314

ABSTRACT

Although complex inflammatory-like alterations are observed around the amyloid plaques of Alzheimer's disease (AD), little is known about the molecular changes and cellular interactions that characterize this response. We investigate here, in an AD mouse model, the transcriptional changes occurring in tissue domains in a 100-µm diameter around amyloid plaques using spatial transcriptomics. We demonstrate early alterations in a gene co-expression network enriched for myelin and oligodendrocyte genes (OLIGs), whereas a multicellular gene co-expression network of plaque-induced genes (PIGs) involving the complement system, oxidative stress, lysosomes, and inflammation is prominent in the later phase of the disease. We confirm the majority of the observed alterations at the cellular level using in situ sequencing on mouse and human brain sections. Genome-wide spatial transcriptomics analysis provides an unprecedented approach to untangle the dysregulated cellular network in the vicinity of pathogenic hallmarks of AD and other brain diseases.


Subject(s)
Alzheimer Disease/pathology , Sequence Analysis, DNA/methods , Transcriptome , Alzheimer Disease/genetics , Amyloid/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Complement System Proteins/genetics , Complement System Proteins/metabolism , Disease Models, Animal , Gene Expression Profiling , Humans , Lysosomes/genetics , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/metabolism , Oxidative Stress/genetics
2.
Cell ; 175(2): 400-415.e13, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30173915

ABSTRACT

Macrophages are highly heterogeneous tissue-resident immune cells that perform a variety of tissue-supportive functions. The current paradigm dictates that intestinal macrophages are continuously replaced by incoming monocytes that acquire a pro-inflammatory or tissue-protective signature. Here, we identify a self-maintaining population of macrophages that arise from both embryonic precursors and adult bone marrow-derived monocytes and persists throughout adulthood. Gene expression and imaging studies of self-maintaining macrophages revealed distinct transcriptional profiles that reflect their unique localization (i.e., closely positioned to blood vessels, submucosal and myenteric plexus, Paneth cells, and Peyer's patches). Depletion of self-maintaining macrophages resulted in morphological abnormalities in the submucosal vasculature and loss of enteric neurons, leading to vascular leakage, impaired secretion, and reduced intestinal motility. These results provide critical insights in intestinal macrophage heterogeneity and demonstrate the strategic role of self-maintaining macrophages in gut homeostasis and intestinal physiology.


Subject(s)
Intestines/immunology , Macrophages/immunology , Animals , Body Patterning/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Gastrointestinal Motility/immunology , Gastrointestinal Motility/physiology , Homeostasis , Inflammation/immunology , Intestinal Mucosa/immunology , Intestine, Small/metabolism , Mice , Monocytes/metabolism , Neurons/metabolism , Phagocytes/immunology , Transcriptome
4.
J Clin Invest ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38888964

ABSTRACT

The ß-secretase BACE1 is a central drug target for Alzheimer's disease. Clinically tested, BACE1-directed inhibitors also block the homologous protease BACE2. Yet, little is known about physiological BACE2 substrates and functions in vivo. Here, we identify BACE2 as the protease shedding the lymphangiogenic vascular endothelial growth factor receptor 3 (VEGFR3). Inactivation of BACE2, but not BACE1, inhibited shedding of VEGFR3 from primary human lymphatic endothelial cells (LECs) and reduced release of the shed, soluble VEGFR3 (sVEGFR3) ectodomain into the blood of mice, non-human primates and humans. Functionally, BACE2 inactivation increased full-length VEGFR3 and enhanced VEGFR3 signaling in LECs and also in vivo in zebrafish, where enhanced migration of LECs was observed. Thus, this study identifies BACE2 as a modulator of lymphangiogenic VEGFR3 signaling and demonstrates the utility of sVEGFR3 as a pharmacodynamic plasma marker for BACE2 activity in vivo, a prerequisite for developing BACE1-selective inhibitors for a safer prevention of Alzheimer's disease.

5.
EMBO Mol Med ; 14(4): e09824, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35352880

ABSTRACT

Single domain antibodies (VHHs) are potentially disruptive therapeutics, with important biological value for treatment of several diseases, including neurological disorders. However, VHHs have not been widely used in the central nervous system (CNS), largely because of their restricted blood-brain barrier (BBB) penetration. Here, we propose a gene transfer strategy based on BBB-crossing adeno-associated virus (AAV)-based vectors to deliver VHH directly into the CNS. As a proof-of-concept, we explored the potential of AAV-delivered VHH to inhibit BACE1, a well-characterized target in Alzheimer's disease. First, we generated a panel of VHHs targeting BACE1, one of which, VHH-B9, shows high selectivity for BACE1 and efficacy in lowering BACE1 activity in vitro. We further demonstrate that a single systemic dose of AAV-VHH-B9 produces positive long-term (12 months plus) effects on amyloid load, neuroinflammation, synaptic function, and cognitive performance, in the AppNL-G-F Alzheimer's mouse model. These results constitute a novel therapeutic approach for neurodegenerative diseases, which is applicable to a range of CNS disease targets.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Single-Domain Antibodies , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/immunology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/immunology , Aspartic Acid Endopeptidases/metabolism , Blood-Brain Barrier , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/therapeutic use , Mice , Mice, Transgenic
6.
Nat Rev Neurol ; 17(11): 703-714, 2021 11.
Article in English | MEDLINE | ID: mdl-34548654

ABSTRACT

Alzheimer disease (AD) is the most common cause of dementia in older individuals (>65 years) and has a long presymptomatic phase. Preventive therapies for AD are not yet available, and potential disease-modifying therapies targeting amyloid-ß plaques in symptomatic stages of AD have only just been approved in the United States. Small-molecule inhibitors of ß-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1; also known as ß-secretase 1) reduce the production of amyloid-ß peptide and are among the most advanced drug candidates for AD. However, to date all phase II and phase III clinical trials of BACE inhibitors were either concluded without benefit or discontinued owing to futility or the occurrence of adverse effects. Adverse effects included early, mild cognitive impairment that was associated with all but one inhibitor; preliminary results suggest that the cognitive effects are non-progressive and reversible. These discontinuations have raised questions regarding the suitability of BACE1 as a drug target for AD. In this Perspective, we discuss the status of BACE inhibitors and suggest ways in which the results of the discontinued trials can inform the development of future clinical trials of BACE inhibitors and related secretase modulators as preventative therapies. We also propose a series of experiments that should be performed to inform 'go-no-go' decisions in future trials with BACE inhibitors and consider the possibility that low levels of BACE1 inhibition could avoid adverse effects while achieving efficacy for AD prevention.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Aged , Aged, 80 and over , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/genetics , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Female , Humans , Male , Middle Aged , Plaque, Amyloid/prevention & control , Research Design
7.
Biol Psychiatry ; 89(8): 745-756, 2021 04 15.
Article in English | MEDLINE | ID: mdl-32223911

ABSTRACT

BACE1 (beta-site amyloid precursor protein cleaving enzyme 1) was initially cloned and characterized in 1999. It is required for the generation of all monomeric forms of amyloid-ß (Aß), including Aß42, which aggregates into bioactive conformational species and likely initiates toxicity in Alzheimer's disease (AD). BACE1 concentrations and rates of activity are increased in AD brains and body fluids, thereby supporting the hypothesis that BACE1 plays a critical role in AD pathophysiology. Therefore, BACE1 is a prime drug target for slowing down Aß production in early AD. Besides the amyloidogenic pathway, BACE1 has other substrates that may be important for synaptic plasticity and synaptic homeostasis. Indeed, germline and adult conditional BACE1 knockout mice display complex neurological phenotypes. Despite BACE1 inhibitor clinical trials conducted so far being discontinued for futility or safety reasons, BACE1 remains a well-validated therapeutic target for AD. A safe and efficacious compound with high substrate selectivity as well as a more accurate dose regimen, patient population, and disease stage may yet be found. Further research should focus on the role of Aß and BACE1 in physiological processes and key pathophysiological mechanisms of AD. The functions of BACE1 and the homologue BACE2, as well as the biology of Aß in neurons and glia, deserve further investigation. Cellular and molecular studies of BACE1 and BACE2 knockout mice coupled with biomarker-based human research will help elucidate the biological functions of these important enzymes and identify their substrates and downstream effects. Such studies will have critical implications for BACE1 inhibition as a therapeutic approach for AD.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Animals , Aspartic Acid Endopeptidases/genetics , Humans , Mice , Mice, Knockout
8.
Cell Rep ; 27(4): 1293-1306.e6, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31018141

ABSTRACT

Gene expression profiles of more than 10,000 individual microglial cells isolated from cortex and hippocampus of male and female AppNL-G-F mice over time demonstrate that progressive amyloid-ß accumulation accelerates two main activated microglia states that are also present during normal aging. Activated response microglia (ARMs) are composed of specialized subgroups overexpressing MHC type II and putative tissue repair genes (Dkk2, Gpnmb, and Spp1) and are strongly enriched with Alzheimer's disease (AD) risk genes. Microglia from female mice progress faster in this activation trajectory. Similar activated states are also found in a second AD model and in human brain. Apoe, the major genetic risk factor for AD, regulates the ARMs but not the interferon response microglia (IRMs). Thus, the ARMs response is the converging point for aging, sex, and genetic AD risk factors.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Biomarkers/metabolism , Brain/pathology , Disease Models, Animal , Microglia/pathology , Plaque, Amyloid/pathology , Aging/genetics , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , Biomarkers/analysis , Brain/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Presenilins/physiology , Sex Characteristics
9.
Biol Psychiatry ; 83(4): 320-327, 2018 02 15.
Article in English | MEDLINE | ID: mdl-28918941

ABSTRACT

The genetic evidence implicating amyloid-ß in the initial stage of Alzheimer's disease is unequivocal. However, the long biochemical and cellular prodromal phases of the disease suggest that dementia is the result of a series of molecular and cellular cascades whose nature and connections remain unknown. Therefore, it is unlikely that treatments directed at amyloid-ß will have major clinical effects in the later stages of the disease. We discuss the two major candidate therapeutic targets to lower amyloid-ß in a preventive mode, i.e., γ- and ß-secretase; the rationale behind these two targets; and the current state of the field.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid Precursor Protein Secretases/drug effects , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/prevention & control , Animals , Humans
10.
Life Sci Alliance ; 1(1): e201800026, 2018 Jan.
Article in English | MEDLINE | ID: mdl-30456346

ABSTRACT

ß-Site APP-cleaving enzyme 1 (BACE1) inhibition is considered one of the most promising therapeutic strategies for Alzheimer's disease, but current BACE1 inhibitors also block BACE2. As the localization and function of BACE2 in the brain remain unknown, it is difficult to predict whether relevant side effects can be caused by off-target inhibition of BACE2 and whether it is important to generate BACE1-specific inhibitors. Here, we show that BACE2 is expressed in discrete subsets of neurons and glia throughout the adult mouse brain. We uncover four new substrates processed by BACE2 in cultured glia: vascular cell adhesion molecule 1, delta and notch-like epidermal growth factor-related receptor, fibroblast growth factor receptor 1, and plexin domain containing 2. Although these substrates were not prominently cleaved by BACE2 in healthy adult mice, proinflammatory TNF induced a drastic increase in BACE2-mediated shedding of vascular cell adhesion molecule 1 in CSF. Thus, although under steady-state conditions the effect of BACE2 cross-inhibition by BACE1-directed inhibitors is rather subtle, it is important to consider that side effects might become apparent under physiopathological conditions that induce TNF expression.

11.
Oncotarget ; 7(2): 1120-43, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26701726

ABSTRACT

The protein kinase LKB1 regulates cell metabolism and growth and is implicated in intestinal and lung cancer. Bone morphogenetic protein (BMP) signaling regulates cell differentiation during development and tissue homeostasis. We demonstrate that LKB1 physically interacts with BMP type I receptors and requires Smad7 to promote downregulation of the receptor. Accordingly, LKB1 suppresses BMP-induced osteoblast differentiation and affects BMP signaling in Drosophila wing longitudinal vein morphogenesis. LKB1 protein expression and Smad1 phosphorylation analysis in a cohort of non-small cell lung cancer patients demonstrated a negative correlation predominantly in a subset enriched in adenocarcinomas. Lung cancer patient data analysis indicated strong correlation between LKB1 loss-of-function mutations and high BMP2 expression, and these two events further correlated with expression of a gene subset functionally linked to apoptosis and migration. This new mechanism of BMP receptor regulation by LKB1 has ramifications in physiological organogenesis and disease.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Smad7 Protein/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Animals, Genetically Modified , Bone Morphogenetic Protein Receptors, Type I/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Drosophila/genetics , Drosophila/growth & development , Drosophila/metabolism , Gene Expression , HEK293 Cells , Humans , Immunoblotting , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Knockout , Protein Binding , Protein Serine-Threonine Kinases/genetics , Pupa/genetics , Pupa/growth & development , Pupa/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Smad7 Protein/genetics , Wings, Animal/growth & development , Wings, Animal/metabolism
12.
Mol Neurodegener ; 11(1): 67, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27716410

ABSTRACT

BACKGROUND: The protease BACE1 (beta-site APP cleaving enzyme) is a major drug target in Alzheimer's disease. However, BACE1 therapeutic inhibition may cause unwanted adverse effects due to its additional functions in the nervous system, such as in myelination and neuronal connectivity. Additionally, recent proteomic studies investigating BACE1 inhibition in cell lines and cultured murine neurons identified a wider range of neuronal membrane proteins as potential BACE1 substrates, including seizure protein 6 (SEZ6) and its homolog SEZ6L. METHODS AND RESULTS: We generated antibodies against SEZ6 and SEZ6L and validated these proteins as BACE1 substrates in vitro and in vivo. Levels of the soluble, BACE1-cleaved ectodomain of both proteins (sSEZ6, sSEZ6L) were strongly reduced upon BACE1 inhibition in primary neurons and also in vivo in brains of BACE1-deficient mice. BACE1 inhibition increased neuronal surface levels of SEZ6 and SEZ6L as shown by cell surface biotinylation, demonstrating that BACE1 controls surface expression of both proteins. Moreover, mass spectrometric analysis revealed that the BACE1 cleavage site in SEZ6 is located in close proximity to the membrane, similar to the corresponding cleavage site in SEZ6L. Finally, an improved method was developed for the proteomic analysis of murine cerebrospinal fluid (CSF) and was applied to CSF from BACE-deficient mice. Hereby, SEZ6 and SEZ6L were validated as BACE1 substrates in vivo by strongly reduced levels in the CSF of BACE1-deficient mice. CONCLUSIONS: This study demonstrates that SEZ6 and SEZ6L are physiological BACE1 substrates in the murine brain and suggests that sSEZ6 and sSEZ6L levels in CSF are suitable markers to monitor BACE1 inhibition in mice.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Biomarkers/cerebrospinal fluid , Blotting, Western , Immunohistochemistry , Mass Spectrometry , Mice , Mice, Knockout , Substrate Specificity
13.
Autophagy ; 8(2): 252-64, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22240588

ABSTRACT

During Drosophila embryogenesis the majority of the extra-embryonic epithelium known as the amnioserosa (AS) undergoes programmed cell death (PCD) following the completion of the morphogenetic process of dorsal closure. Approximately ten percent of AS cells, however, are eliminated during dorsal closure by extrusion from the epithelium. Using biosensors that report autophagy and caspase activity in vivo, we demonstrate that AS cell extrusion occurs in the context of elevated autophagy and caspase activation. Furthermore, we evaluate AS extrusion rates, autophagy, and caspase activation in embryos in which caspase activity or autophagy are altered by genetic manipulation. This includes using the GAL4/UAS system to drive expression of p35, reaper, dINR (ACT) and Atg1 in the AS; we also analyze embryos lacking both maternal and zygotic expression of Atg1. Based on our results we suggest that autophagy can promote, but is not required for, epithelial extrusion and caspase activation in the amnioserosa.


Subject(s)
Amnion/cytology , Amnion/embryology , Autophagy , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Embryo, Nonmammalian/cytology , Epithelial Cells/cytology , Amnion/enzymology , Amnion/ultrastructure , Animals , Apoptosis , Biosensing Techniques , Caspases/metabolism , Cell Communication , Cell Polarity , Cell Shape , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/ultrastructure , Enzyme Activation , Epithelial Cells/ultrastructure , Green Fluorescent Proteins/metabolism , Mutation/genetics , Proteolysis
SELECTION OF CITATIONS
SEARCH DETAIL