Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Neuroinflammation ; 19(1): 65, 2022 Mar 11.
Article in English | MEDLINE | ID: mdl-35277173

ABSTRACT

BACKGROUND: Membrane-type matrix metalloproteinase 5 (MT5-MMP) deficiency in the 5xFAD mouse model of Alzheimer's disease (AD) reduces brain neuroinflammation and amyloidosis, and prevents deficits in synaptic activity and cognition in prodromal stages of the disease. In addition, MT5-MMP deficiency prevents interleukin-1 beta (IL-1ß)-mediated inflammation in the peripheral nervous system. In this context, we hypothesized that the MT5-MMP/IL-1ß tandem could regulate nascent AD pathogenic events in developing neural cells shortly after the onset of transgene activation. METHODS: To test this hypothesis, we used 11-14 day in vitro primary cortical cultures from wild type, MT5-MMP-/-, 5xFAD and 5xFAD/MT5-MMP-/- mice, and evaluated the impact of MT5-MMP deficiency and IL-1ß treatment for 24 h, by performing whole cell patch-clamp recordings, RT-qPCR, western blot, gel zymography, ELISA, immunocytochemistry and adeno-associated virus (AAV)-mediated transduction. RESULTS: 5xFAD cells showed higher levels of MT5-MMP than wild type, concomitant with higher basal levels of inflammatory mediators. Moreover, MT5-MMP-deficient cultures had strong decrease of the inflammatory response to IL-1ß, as well as decreased stability of recombinant IL-1ß. The levels of amyloid beta peptide (Aß) were similar in 5xFAD and wild-type cultures, and IL-1ß treatment did not affect Aß levels. Instead, the absence of MT5-MMP significantly reduced Aß by more than 40% while sparing APP metabolism, suggesting altogether no functional crosstalk between IL-1ß and APP/Aß, as well as independent control of their levels by MT5-MMP. The lack of MT5-MMP strongly downregulated the AAV-induced neuronal accumulation of the C-terminal APP fragment, C99, and subsequently that of Aß. Finally, MT5-MMP deficiency prevented basal hyperexcitability observed in 5xFAD neurons, but not hyperexcitability induced by IL-1ß treatment. CONCLUSIONS: Neuroinflammation and hyperexcitability precede Aß accumulation in developing neural cells with nascent expression of AD transgenes. MT5-MMP deletion is able to tune down basal neuronal inflammation and hyperexcitability, as well as APP/Aß metabolism. In addition, MT5-MMP deficiency prevents IL-1ß-mediated effects in brain cells, except hyperexcitability. Overall, this work reinforces the idea that MT5-MMP is at the crossroads of pathogenic AD pathways that are already incipiently activated in developing neural cells, and that targeting MT5-MMP opens interesting therapeutic prospects.


Subject(s)
Alzheimer Disease , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/metabolism , Disease Models, Animal , Matrix Metalloproteinases/metabolism , Mice , Mice, Transgenic , Neuroinflammatory Diseases , Neurons/metabolism
2.
FASEB J ; 35(7): e21727, 2021 07.
Article in English | MEDLINE | ID: mdl-34117802

ABSTRACT

We previously discovered the implication of membrane-type 5-matrix metalloproteinase (MT5-MMP) in Alzheimer's disease (AD) pathogenesis. Here, we shed new light on pathogenic mechanisms by which MT5-MMP controls the processing of amyloid precursor protein (APP) and the fate of amyloid beta peptide (Aß) as well as its precursor C99, and C83. We found in human embryonic kidney cells (HEK) carrying the APP Swedish familial mutation (HEKswe) that deleting the C-terminal non-catalytic domains of MT5-MMP hampered its ability to process APP and release the soluble 95 kDa form (sAPP95). Catalytically inactive MT5-MMP variants increased the levels of Aß and promoted APP/C99 sorting in the endolysosomal system, likely through interactions of the proteinase C-terminal portion with C99. Most interestingly, the deletion of the C-terminal domain of MT5-MMP caused a strong degradation of C99 by the proteasome and prevented Aß accumulation. These discoveries reveal new control of MT5-MMP over APP by proteolytic and non-proteolytic mechanisms driven by the C-terminal domains of the proteinase. The targeting of these non-catalytic domains of MT5-MMP could, therefore, provide new insights into the therapeutic regulation of APP-related pathology in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Matrix Metalloproteinases, Membrane-Associated/metabolism , Peptide Fragments/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cell Line , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proteolysis
3.
Mar Drugs ; 19(10)2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34677445

ABSTRACT

Sugar-based molecules such as heparins or natural heparan sulfate polysaccharides have been developed and widely studied for controlling heparanase (HPSE) enzymatic activity, a key player in extracellular matrix remodelling during cancer pathogenesis. However, non-enzymatic functions of HPSE have also been described in tumour mechanisms. Given their versatile properties, we hypothesized that sugar-based inhibitors may interfere with enzymatic but also non-enzymatic HPSE activities. In this work, we assessed the effects of an original marine λ-carrageenan derived oligosaccharide (λ-CO) we previously described, along with those of its native counterpart and heparins, on cell viability, proliferation, migration, and invasion of MDA-MB-231 breast cancer cells but also of sh-MDA-MB-231 cells, in which the expression of HPSE was selectively downregulated. We observed no cytotoxic and no anti-proliferative effects of our compounds but surprisingly λ-CO was the most efficient to reduce cell migration and invasion compared with heparins, and in a HPSE-dependent manner. We provided evidence that λ-CO tightly controlled a HPSE/MMP-14/MMP-2 axis, leading to reduced MMP-2 activity. Altogether, this study highlights λ-CO as a potent HPSE "modulator" capable of reducing not only the enzymatic activity of HPSE but also the functions controlled by the HPSE levels.


Subject(s)
Antineoplastic Agents/pharmacology , Carrageenan/pharmacology , Cell Line, Tumor/drug effects , Glucuronidase/metabolism , Rhodophyta , Animals , Antineoplastic Agents/chemistry , Aquatic Organisms , Breast Neoplasms , Carrageenan/chemistry , Cell Movement/drug effects , Female , Humans , Inhibitory Concentration 50 , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism
4.
FASEB J ; 33(2): 2910-2927, 2019 02.
Article in English | MEDLINE | ID: mdl-30332299

ABSTRACT

We previously demonstrated that membrane type 1 (MT1) matrix metalloproteinase (MMP) was up-regulated in the hippocampus of the model of transgenic mice bearing 5 familial mutations on human amyloid precursor protein (APP) and presenilin 1 of Alzheimer disease (AD), and that the proteinase increased the levels of amyloid ß peptide (Aß) and its APP C-terminal fragment of 99 aa in a heterologous cell system. Here we provide further evidence that MT1-MMP interacts with APP and promotes amyloidogenesis in a proteolytic-dependent manner in Swedish APP-expressing human embryonic kidney 293 (HEKswe) cells. MT1-MMP-mediated processing of APP releases a soluble APP fragment, sAPP95. This process partly requires the activation of endogenous MMP-2 but is independent of ß-site APP cleaving enzyme 1 (BACE-1) or α-secretase activities. In contrast, MT1-MMP-mediated increase of Aß levels involved BACE-1 activity and was inhibited by tissue inhibitor of MMP-2, a natural inhibitor of both MT1-MMP and MMP-2. Interestingly, near abolishment of basal Aß production upon BACE-1 inhibition was rescued by MT1-MMP, indicating that the latter could mimic ß-secretase-like activity. Moreover, MT1-MMP promoted APP/Aß localization in endosomes, where Aß production mainly occurs. These data unveil new mechanistic insights to support the proamyloidogenic role of MT1-MMP based on APP processing and trafficking, and reinforce the idea that this proteinase may become a new potential therapeutic target in AD.-Paumier, J.-M., Py, N. A., González, L. G., Bernard, A., Stephan, D., Louis, L., Checler, F., Khrestchatisky, M., Baranger, K., Rivera, S. Proamyloidogenic effects of membrane type 1 matrix metalloproteinase involve MMP-2 and BACE-1 activities, and the modulation of APP trafficking.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid/chemistry , Aspartic Acid Endopeptidases/metabolism , Gene Expression Regulation/drug effects , Matrix Metalloproteinase 14/pharmacology , Matrix Metalloproteinase 2/metabolism , Alzheimer Disease , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/genetics , HEK293 Cells , Humans , Matrix Metalloproteinase 2/genetics , Mice , Mice, Transgenic , Protein Transport
5.
Cell Mol Life Sci ; 76(16): 3167-3191, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31197405

ABSTRACT

As life expectancy increases worldwide, age-related neurodegenerative diseases will increase in parallel. The lack of effective treatment strategies may soon lead to an unprecedented health, social and economic crisis. Any attempt to halt the progression of these diseases requires a thorough knowledge of the pathophysiological mechanisms involved to facilitate the identification of new targets and the application of innovative therapeutic strategies. The metzincin superfamily of metalloproteinases includes matrix metalloproteinases (MMP), a disintegrin and metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS). These multigenic and multifunctional proteinase families regulate the functions of an increasing number of signalling and scaffolding molecules involved in neuroinflammation, blood-brain barrier disruption, protein misfolding, synaptic dysfunction or neuronal death. Metalloproteinases and their physiological inhibitors, the tissue inhibitors of metalloproteinases (TIMPs), are therefore, at the crossroads of molecular and cellular mechanisms that support neurodegenerative processes, and emerge as potential new therapeutic targets. We provide an overview of current knowledge on the role and regulation of metalloproteinases and TIMPs in four major neurodegenerative diseases: Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and Huntington's disease.


Subject(s)
Alzheimer Disease/pathology , Matrix Metalloproteinases/metabolism , Neurodegenerative Diseases/pathology , Tissue Inhibitor of Metalloproteinases/metabolism , ADAM Proteins/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Humans , Huntington Disease/metabolism , Huntington Disease/pathology , Neurodegenerative Diseases/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology
6.
Cell Mol Life Sci ; 73(1): 217-36, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26202697

ABSTRACT

Membrane-type 5-matrix metalloproteinase (MT5-MMP) is a proteinase mainly expressed in the nervous system with emerging roles in brain pathophysiology. The implication of MT5-MMP in Alzheimer's disease (AD), notably its interplay with the amyloidogenic process, remains elusive. Accordingly, we crossed the genetically engineered 5xFAD mouse model of AD with MT5-MMP-deficient mice and examined the impact of MT5-MMP deficiency in bigenic 5xFAD/MT5-MMP(-/-) mice. At early stages (4 months) of the pathology, the levels of amyloid beta peptide (Aß) and its amyloid precursor protein (APP) C-terminal fragment C99 were largely reduced in the cortex and hippocampus of 5xFAD/MT5-MMP(-/-), compared to 5xFAD mice. Reduced amyloidosis in bigenic mice was concomitant with decreased glial reactivity and interleukin-1ß (IL-1ß) levels, and the preservation of long-term potentiation (LTP) and spatial learning, without changes in the activity of α-, ß- and γ-secretases. The positive impact of MT5-MMP deficiency was still noticeable at 16 months of age, as illustrated by reduced amyloid burden and gliosis, and a better preservation of the cortical neuronal network and synaptophysin levels in bigenic mice. MT5-MMP expressed in HEKswe cells colocalized and co-immunoprecipitated with APP and significantly increased the levels of Aß and C99. MT5-MMP also promoted the release of a soluble APP fragment of 95 kDa (sAPP95) in HEKswe cells. sAPP95 levels were significantly reduced in brain homogenates of 5xFAD/MT5-MMP(-/-) mice, supporting altogether the idea that MT5-MMP influences APP processing. MT5-MMP emerges as a new pro-amyloidogenic regulator of APP metabolism, whose deficiency alleviates amyloid pathology, neuroinflammation and cognitive decline.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/physiopathology , Hippocampus/enzymology , Hippocampus/physiopathology , Matrix Metalloproteinases, Membrane-Associated/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/analysis , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/analysis , Amyloid beta-Protein Precursor/metabolism , Animals , Cognition , Female , Gene Deletion , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Long-Term Potentiation , Male , Matrix Metalloproteinases, Membrane-Associated/analysis , Matrix Metalloproteinases, Membrane-Associated/genetics , Mice, Inbred C57BL , Mice, Transgenic , Spatial Learning
7.
Neurobiol Dis ; 88: 107-17, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26774030

ABSTRACT

Clinical and experimental evidence point to a possible role of cerebrovascular dysfunction in Alzheimer's disease (AD). The 5xFAD mouse model of AD expresses human amyloid precursor protein and presenilin genes with mutations found in AD patients. It remains unknown whether amyloid deposition driven by these mutations is associated with cerebrovascular changes. 5xFAD and wild type mice (2 to 12months old; M2 to M12) were used. Thinned skull in vivo 2-photon microscopy was used to determine Aß accumulation on leptomeningeal or superficial cortical vessels over time. Parenchymal microvascular damage was assessed using FITC-microangiography. Collagen-IV and CD31 were used to stain basal lamina and endothelial cells. Methoxy-XO4, Thioflavin-S or 6E10 were used to visualize Aß accumulation in living mice or in fixed brain tissues. Positioning of reactive IBA1 microglia and GFAP astrocytes at the vasculature was rendered using confocal microscopy. Platelet-derived growth factor receptor beta (PDGFRß) staining was used to visualize perivascular pericytes. In vivo 2-photon microscopy revealed Methoxy-XO4(+) amyloid perivascular deposits on leptomeningeal and penetrating cortical vessels in 5xFAD mice, typical of cerebral amyloid angiopathy (CAA). Amyloid deposits were visible in vivo at M3 and aggravated over time. Progressive microvascular damage was concomitant to parenchymal Aß plaque accumulation in 5xFAD mice. Microvascular inflammation in 5xFAD mice presented with sporadic FITC-albumin leakages at M4 becoming more prevalent at M9 and M12. 3D colocalization showed inflammatory IBA1(+) microglia proximal to microvascular FITC-albumin leaks. The number of perivascular PDGFRß(+) pericytes was significantly decreased at M4 in the fronto-parietal cortices, with a trend decrease observed in the other structures. At M9-M12, PDGFRß(+) pericytes displayed hypertrophic perivascular ramifications contiguous to reactive microglia. Cerebral amyloid angiopathy and microvascular inflammation occur in 5xFAD mice concomitantly to parenchymal plaque deposition. The prospect of cerebrovascular pharmacology in AD is discussed.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Blood Vessels/pathology , Cerebrovascular Circulation/genetics , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Calcium-Binding Proteins/metabolism , Collagen Type IV/metabolism , Disease Models, Animal , Disease Progression , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Mutation/genetics , Pericytes/metabolism , Pericytes/pathology , Plaque, Amyloid/metabolism , Platelet Endothelial Cell Adhesion Molecule-1 , Presenilin-1/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism
8.
J Neuroinflammation ; 13(1): 167, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27349644

ABSTRACT

We have recently identified in a transgenic mouse model of Alzheimer's disease (AD) membrane-type 5-MMP (MT5-MMP) as a new player in Alzheimer's pathogenesis, which displays pro-amyloidogenic features and proteolytic processing of amyloid precursor protein (APP). Another group has reported that MT5-MMP processing of APP may release a novel neurotoxic APP fragment. Although MT5-MMP-mediated APP processing appears to be a key pathogenic step, we hypothesize that MT5-MMP may also contribute to AD pathogenesis through complementary mechanisms that involve the activation of pro-inflammatory pathways and/or APP trafficking.


Subject(s)
Alzheimer Disease/chemically induced , Alzheimer Disease/genetics , Matrix Metalloproteinases, Membrane-Associated/metabolism , Matrix Metalloproteinases, Membrane-Associated/toxicity , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Humans , Mice , Mice, Transgenic
9.
Hippocampus ; 24(7): 762-72, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24596271

ABSTRACT

The 5XFAD mice are an early-onset transgenic model of Alzheimer's disease (AD) in which amyloid plaques are first observed between two and four months of age in the cortical layer five and in the subiculum of the hippocampal formation. Although cognitive alterations have been described in these mice, there are no studies that focused on the onset of hippocampus-dependent memory deficits, which are a hallmark of the prodromal stage of AD. To identify when the first learning and memory impairments appear, 5XFAD mice of two, four, and six months of age were compared with their respective wild-type littermates using the olfactory tubing maze, which is a very sensitive hippocampal-dependent task. Deficits in learning and memory started at four months with a substantial increase at six months of age while no olfactory impairments were observed. The volumetric study using magnetic resonance imaging of the whole brain and specific areas (olfactory bulb, striatum, and hippocampus) did not reveal neuro-anatomical difference. Slight memory deficits appeared at 4 months of age in correlation with an increased astrogliosis and amyloid plaque formation. This early impairment in learning and memory related to the hippocampal dysfunction is particularly suited to assess preclinical therapeutic strategies aiming to delay or suppress the onset of AD.


Subject(s)
Alzheimer Disease/psychology , Hippocampus/physiopathology , Learning Disabilities/etiology , Memory Disorders/etiology , Age of Onset , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Animals , Astrocytes/pathology , Corpus Striatum/pathology , Gliosis/etiology , Gliosis/pathology , Hippocampus/pathology , Learning Disabilities/pathology , Learning Disabilities/physiopathology , Magnetic Resonance Imaging , Male , Maze Learning , Memory Disorders/pathology , Memory Disorders/physiopathology , Mice , Mice, Transgenic , Olfactory Bulb/pathology , Organ Size , Plaque, Amyloid/etiology , Plaque, Amyloid/pathology , Point Mutation , Presenilin-1/genetics , Smell/physiology
10.
J Neuroinflammation ; 9: 10, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-22248015

ABSTRACT

BACKGROUND: Alzheimer's disease has become a growing socio-economical concern in developing countries where increased life expectancy is leading to large aged populations. While curing Alzheimer's disease or stopping its progression does not appear within reach in a foreseeable future, new therapies capable of delaying the pathogenesis would represent major breakthroughs. PRESENTATION OF THE HYPOTHESIS: The growing number of medical benefits of cannabinoids, such as their ability to regulate age-related processes like neuroinflammation, neurogenesis and memory, raise the question of their potential role as a preventive treatment of AD. TESTING THE HYPOTHESIS: To test this hypothesis, epidemiological studies on long term, chronic cannabinoid users could enlighten us on the potential benefits of these compounds in normal and pathological ageing processes. Systematic pharmacological (and thus more mechanistic) investigations using animal models of Alzheimer's disease that have been developed would also allow a thorough investigation of the benefits of cannabinoid pharmacotherapy in the pathogenesis of Alzheimer's disease. IMPLICATIONS OF THE HYPOTHESIS: The chronic administration of non-selective cannabinoids may delay the onset of cognitive deficits in AD patients; this will dramatically reduce the socio-economic burden of AD and improve the quality of life of the patients and their families.


Subject(s)
Aging , Alzheimer Disease/metabolism , Encephalitis/prevention & control , Memory/physiology , Neurogenesis/physiology , Receptors, Cannabinoid/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Humans , Memory/drug effects , Models, Biological , Neurogenesis/drug effects
11.
J Alzheimers Dis ; 90(1): 251-262, 2022.
Article in English | MEDLINE | ID: mdl-36093693

ABSTRACT

BACKGROUND: The 5XFAD model of Alzheimer's disease (AD) bearing five familial mutations of Alzheimer's disease on human APP and PSEN1 transgenes shows deposits of amyloid-ß peptide (Aß) as early as 2 months, while deficits in long-term memory can be detected at 4 months using the highly sensitive olfactory-dependent tests that we previously reported. OBJECTIVE: Given that detecting early dysfunctions in AD prior to overt pathology is of major interest in the field, we sought to detect memory deficits at earlier stages of the disease in 3-month-old male 5XFAD mice. METHODS: To this end, we used the Helico Maze, a behavioral task that was recently developed and patented. This device allows deeper analysis of learning and subcategories of hippocampal-dependent long-term memory using olfactory cues. RESULTS: Eight male 5XFAD and 6 male wild-type (WT: C57Bl6 background) mice of 3 months of age were tested in the Helico Maze. The results demonstrated, for the first time, a starting deficit of pure reference long-term memory. Interestingly, memory impairment was clearly correlated with Aß deposits in the hippocampus. While we also found significant differences in astrogliosis between 5XFAD and WT mice, this was not correlated with memory abilities. CONCLUSION: Our results underline the efficiency of this new olfactory-dependent behavioral task, which is easy to use, with a small cohort of mice. Using the Helico Maze may open new avenues to validate the efficacy of treatments that target early events related to the amyloid-dependent pathway of the disease and AD progression.


Subject(s)
Alzheimer Disease , Humans , Animals , Mice , Male , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Mice, Transgenic , Amyloid beta-Peptides/metabolism , Disease Models, Animal , Memory Disorders/genetics , Memory Disorders/pathology , Mice, Inbred C57BL , Maze Learning
12.
Cell Rep ; 40(7): 111200, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977506

ABSTRACT

Apolipoprotein E4 (APOEε4) is the major allelic risk factor for late-onset sporadic Alzheimer's disease (sAD). Inflammation is increasingly considered as critical in sAD initiation and progression. Identifying brain molecular mechanisms that could bridge these two risk factors remain unelucidated. Leveraging induced pluripotent stem cell (iPSC)-based strategies, we demonstrate that APOE controls inflammation in human astrocytes by regulating Transgelin 3 (TAGLN3) expression and, ultimately, nuclear factor κB (NF-κB) activation. We uncover that APOE4 specifically downregulates TAGLN3, involving histone deacetylases activity, which results in low-grade chronic inflammation and hyperactivated inflammatory responses. We show that APOE4 exerts a dominant negative effect to prime astrocytes toward a pro-inflammatory state that is pharmacologically reversible by TAGLN3 supplementation. We further confirm that TAGLN3 is downregulated in the brain of patients with sAD. Our findings highlight the APOE-TAGLN3-NF-κB axis regulating neuroinflammation in human astrocytes and reveal TAGLN3 as a molecular target to modulate neuroinflammation, as well as a potential biomarker for AD.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Apolipoproteins E/metabolism , Nerve Tissue Proteins/metabolism , Alzheimer Disease/metabolism , Apolipoprotein E3/metabolism , Apolipoprotein E4/metabolism , Apolipoproteins E/genetics , Astrocytes/metabolism , Humans , Inflammation/metabolism , NF-kappa B/metabolism
13.
Biochem Soc Trans ; 39(5): 1441-6, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21936830

ABSTRACT

It is now clear that NSPs (neutrophil serine proteases), including elastase, Pr3 (proteinase 3) and CatG (cathepsin G) are major pathogenic determinants in chronic inflammatory disorders of the lungs. Two unglycosylated natural protease inhibitors, SLPI (secretory leucocyte protease inhibitor) and elafin, and its precursor trappin-2 that are found in the lungs, have therapeutic potential for reducing the protease-induced inflammatory response. This review examines the multifaceted roles of SLPI and elafin/trappin-2 in the context of their possible use as inhaled drugs for treating chronic lung diseases such as CF (cystic fibrosis) and COPD (chronic obstructive pulmonary disease).


Subject(s)
Elafin/metabolism , Inflammation/enzymology , Lung Diseases/enzymology , Secretory Leukocyte Peptidase Inhibitor/metabolism , Serine Proteases/metabolism , Serine Proteinase Inhibitors/metabolism , Aerosols , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/therapeutic use , Antifungal Agents/metabolism , Antifungal Agents/therapeutic use , Elafin/therapeutic use , Humans , Inflammation/drug therapy , Lung Diseases/drug therapy , Proteinase Inhibitory Proteins, Secretory/metabolism , Proteinase Inhibitory Proteins, Secretory/therapeutic use , Secretory Leukocyte Peptidase Inhibitor/therapeutic use , Serine Proteinase Inhibitors/therapeutic use , Transglutaminases/metabolism
14.
J Med Chem ; 63(19): 10705-10725, 2020 10 08.
Article in English | MEDLINE | ID: mdl-32459966

ABSTRACT

Although matrix metalloproteinases (MMPs) are implicated in the regulation of numerous physiological processes, evidence of their pathological roles have also been obtained in the last decades, making MMPs attractive therapeutic targets for several diseases. Recent discoveries of their involvement in central nervous system (CNS) disorders, and in particular in Alzheimer's disease (AD), have paved the way to consider MMP modulators as promising therapeutic strategies. Over the past few decades, diverse approaches have been undertaken in the design of therapeutic agents targeting MMPs for various purposes, leading, more recently, to encouraging developments. In this article, we will present recent examples of inhibitors ranging from small molecules and peptidomimetics to biologics. We will also discuss the scientific knowledge that has led to the development of emerging tools and techniques to overcome the challenges of selective MMP inhibition.


Subject(s)
Alzheimer Disease/drug therapy , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinases/drug effects , Alzheimer Disease/enzymology , Central Nervous System/physiology , Drug Delivery Systems , Humans , Matrix Metalloproteinases/metabolism , Peptidomimetics
15.
Br J Pharmacol ; 177(9): 1988-2005, 2020 05.
Article in English | MEDLINE | ID: mdl-31881553

ABSTRACT

BACKGROUND AND PURPOSE: We recently identified donecopride as a pleiotropic compound able to inhibit AChE and to activate 5-HT4 receptors. Here, we have assessed the potential therapeutic effects of donecopride in treating Alzheimer's disease (AD). EXPERIMENTAL APPROACH: We used two in vivo animal models of AD, transgenic 5XFAD mice and mice exposed to soluble amyloid-ß peptides and, in vitro, primary cultures of rat hippocampal neurons. Pro-cognitive and anti-amnesic effects were evaluated with novel object recognition, Y-maze, and Morris water maze tests. Amyloid load in mouse brain was measured ex vivo and effects of soluble amyloid-ß peptides on neuronal survival and neurite formation determined in vitro. KEY RESULTS: In vivo, chronic (3 months) administration of donecopride displayed potent anti-amnesic properties in the two mouse models of AD, preserving learning capacities, including working and long-term spatial memories. These behavioural effects were accompanied by decreased amyloid aggregation in the brain of 5XFAD mice and, in cultures of rat hippocampal neurons, reduced tau hyperphosphorylation. In vitro, donecopride increased survival in neuronal cultures exposed to soluble amyloid-ß peptides, improved the neurite network and provided neurotrophic benefits, expressed as the formation of new synapses. CONCLUSIONS AND IMPLICATIONS: Donecopride acts like a Swiss army knife, exhibiting a range of sustainable symptomatic therapeutic effects and potential disease-modifying effects in models of AD. Clinical trials with this promising drug candidate will soon be undertaken to confirm its therapeutic potential in humans.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aniline Compounds , Animals , Brain/metabolism , Disease Models, Animal , Maze Learning , Mice , Mice, Transgenic , Piperidines , Rats
16.
Front Aging Neurosci ; 11: 244, 2019.
Article in English | MEDLINE | ID: mdl-31607898

ABSTRACT

Processing of amyloid beta precursor protein (APP) into amyloid-beta peptide (Aß) by ß-secretase and γ-secretase complex is at the heart of the pathogenesis of Alzheimer's disease (AD). Targeting this proteolytic pathway effectively reduces/prevents pathology and cognitive decline in preclinical experimental models of the disease, but therapeutic strategies based on secretase activity modifying drugs have so far failed in clinical trials. Although this may raise some doubts on the relevance of ß- and γ-secretases as targets, new APP-cleaving enzymes, including meprin-ß, legumain (δ-secretase), rhomboid-like protein-4 (RHBDL4), caspases and membrane-type matrix metalloproteinases (MT-MMPs/η-secretases) have confirmed that APP processing remains a solid mechanism in AD pathophysiology. This review will discuss recent findings on the roles of all these proteinases in the nervous system, and in particular on the roles of MT-MMPs, which are at the crossroads of pathological events involving not only amyloidogenesis, but also inflammation and synaptic dysfunctions. Assessing the potential of these emerging proteinases in the Alzheimer's field opens up new research prospects to improve our knowledge of fundamental mechanisms of the disease and help us establish new therapeutic strategies.

17.
Neurotherapeutics ; 16(4): 1237-1254, 2019 10.
Article in English | MEDLINE | ID: mdl-31267473

ABSTRACT

The low-molecular weight thiol pantethine, known as a hypolipidemic and hypocholesterolemic agent, is the major precursor of co-enzyme A. We have previously shown that pantethine treatment reduces amyloid-ß (Aß)-induced IL-1ß release and alleviates pathological metabolic changes in primary astrocyte cultures. These properties of pantethine prompted us to investigate its potential benefits in vivo in the 5XFAD (Tg) mouse model of Alzheimer's disease (AD).1.5-month-old Tg and wild-type (WT) male mice were submitted to intraperitoneal administration of pantethine or saline control solution for 5.5 months. The effects of such treatments were investigated by performing behavioral tests and evaluating astrogliosis, microgliosis, Αß deposition, and whole genome expression arrays, using RNAs extracted from the mice hippocampi. We observed that long-term pantethine treatment significantly reduced glial reactivity and Αß deposition, and abrogated behavioral alteration in Tg mice. Moreover, the transcriptomic profiles revealed that after pantethine treatment, the expression of genes differentially expressed in Tg mice, and in particular those known to be related to AD, were significantly alleviated. Most of the genes overexpressed in Tg compared to WT were involved in inflammation, complement activation, and phagocytosis and were found repressed upon pantethine treatment. In contrast, pantethine restored the expression of a significant number of genes involved in the regulation of Αß processing and synaptic activities, which were downregulated in Tg mice. Altogether, our data support a beneficial role for long-term pantethine treatment in preserving CNS crucial functions altered by Aß pathogenesis in Tg mice and highlight the potential efficiency of pantethine to alleviate AD pathology.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Disease Models, Animal , Pantetheine/analogs & derivatives , Aggression/drug effects , Aggression/physiology , Alzheimer Disease/pathology , Animals , Drug Administration Schedule , Hippocampus/drug effects , Hippocampus/pathology , Humans , Male , Mice , Mice, Transgenic , Pantetheine/administration & dosage , Phagocytosis/drug effects , Phagocytosis/physiology , Time Factors
18.
FEBS J ; 275(9): 2008-20, 2008 May.
Article in English | MEDLINE | ID: mdl-18341586

ABSTRACT

Trappin-2 (also known as pre-elafin) is an endogenous inhibitor of neutrophil serine proteases and is involved in the control of excess proteolysis, especially in inflammatory events, along with the structurally related secretory leucocyte proteinase inhibitor. Secretory leucocyte proteinase inhibitor has been shown to have antibacterial and antifungal properties, whereas recent data indicate that trappin-2 has antimicrobial activity against Pseudomonas aeruginosa and Staphylococcus aureus. In the present study, we tested the antibacterial properties of trappin-2 towards other respiratory pathogens. We found that trappin-2, at concentrations of 5-20 microm, has significant activity against Klebsiella pneumoniae, Haemophilus influenzae, Streptococcus pneumoniae, Branhamella catarrhalis and the pathogenic fungi Aspergillus fumigatus and Candida albicans, in addition to P. aeruginosa and S. aureus. A similar antimicrobial activity was observed with trappin-2 A62D/M63L, a trappin-2 variant that has lost its antiprotease properties, indicating that trappin-2 exerts its antibacterial effects through mechanisms independent from its intrinsic antiprotease capacity. Furthermore, the antibacterial and antifungal activities of trappin-2 were sensitive to NaCl and heparin, demonstrating that its mechanism of action is most probably dependent on its cationic nature. This enables trappin-2 to interact with the membranes of target organisms and disrupt them, as shown by our scanning electron microscopy analyses. Thus, trappin-2 not only provides an antiprotease shield, but also may play an important role in the innate defense of the human lungs and mucosae against pathogenic microorganisms.


Subject(s)
Anti-Bacterial Agents/metabolism , Antifungal Agents/metabolism , Elafin/metabolism , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/metabolism , Disulfides/chemistry , Dose-Response Relationship, Drug , Elafin/pharmacology , Fibronectins/metabolism , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/ultrastructure , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/ultrastructure , Heparin/pharmacology , Kinetics , Models, Chemical , Mutation , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/isolation & purification , Sodium Chloride/pharmacology
19.
Biochimie ; 90(2): 284-95, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17964057

ABSTRACT

Elafin and SLPI are low-molecular weight proteins that were first identified as protease inhibitors in mucous fluids including lung secretions, where they help control excessive proteolysis due to neutrophil serine proteases (elastase, proteinase 3 and cathepsin G). Elafin and SLPI are structurally related in that both have a fold with a four-disulfide core or whey acidic protein (WAP) domain responsible for inhibiting proteases. Elafin is derived from a precursor, trappin-2 or pre-elafin, by proteolysis. Trappin-2, which is itself a protease inhibitor, has a unique N-terminal domain that enables it to become cross-linked to extracellular matrix proteins by transglutaminase(s). SLPI and elafin/trappin-2 are attractive candidates as therapeutic molecules for inhibiting neutrophil serine proteases in inflammatory lung diseases. Hence, they have become the WAP proteins most studied over the last decade. This review focuses on recent findings revealing that SLPI and elafin/trappin-2 have many biological functions as diverse as anti-bacterial, anti-fungal, anti-viral, anti-inflammatory and immuno-modulatory functions, in addition to their well-recognized role as protease inhibitors.


Subject(s)
Elafin/physiology , Secretory Leukocyte Peptidase Inhibitor/physiology , Amino Acid Sequence , Anti-HIV Agents/pharmacology , Anti-Infective Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Elafin/chemistry , Elafin/pharmacology , Humans , Lung Diseases/drug therapy , Molecular Sequence Data , Secretory Leukocyte Peptidase Inhibitor/chemistry , Secretory Leukocyte Peptidase Inhibitor/pharmacology , Transglutaminases/metabolism
20.
Mol Neurobiol ; 55(8): 6463-6479, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29318446

ABSTRACT

The impairment of hippocampal neurogenesis at the early stages of Alzheimer's disease (AD) is believed to support early cognitive decline. Converging studies sustain the idea that vitamin D might be linked to the pathophysiology of AD and to hippocampal neurogenesis. Nothing being known about the effects of vitamin D on hippocampal neurogenesis in AD, we assessed them in a mouse model of AD. In a previous study, we observed that dietary vitamin D supplementation in female AD-like mice reduced cognitive decline only when delivered during the symptomatic phase. With these data in hand, we wondered whether the consequences of vitamin D administration on hippocampal neurogenesis are stage-dependent. Male wild-type and transgenic AD-like mice (5XFAD model) were fed with a diet containing either no vitamin D (0VD) or a normal dose of vitamin D (NVD) or a high dose of vitamin D (HVD), from month 1 to month 6 (preventive arm) or from month 4 to month 9 (curative arm). Working memory was assessed using the Y-maze, while amyloid burden, astrocytosis, and neurogenesis were quantified using immunohistochemistry. In parallel, the effects of vitamin D on proliferation and differentiation were assayed on primary cultures of murine neural progenitor cells. Improved working memory and neurogenesis were observed when high vitamin D supplementation was administered during the early phases of the disease, while a normal dose of vitamin D increased neurogenesis during the late phases. Conversely, an early hypovitaminosis D increased the number of amyloid plaques in AD mice while a late hypovitaminosis D impaired neurogenesis in AD and WT mice. The observed in vivo vitamin D-associated increased neurogenesis was partially substantiated by an augmented in vitro proliferation but not an increased differentiation of neural progenitors into neurons. Finally, a sexual dimorphism was observed. Vitamin D supplementation improved the working memory of males and females, when delivered during the pre-symptomatic and symptomatic phases, respectively. Our study establishes that (i) neurogenesis is improved by vitamin D in a male mouse model of AD, in a time-dependent manner, and (ii) cognition is enhanced in a gender-associated way. Additional pre-clinical studies are required to further understand the gender- and time-specific mechanisms of action of vitamin D in AD. This may lead to an adaptation of vitamin D supplementation in relation to patient's gender and age as well as to the stage of the disease.


Subject(s)
Alzheimer Disease/physiopathology , Cognition/drug effects , Neurogenesis/drug effects , Vitamin D/pharmacology , Alzheimer Disease/pathology , Amyloid/metabolism , Animals , Calcitriol/pharmacology , Cell Differentiation , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Female , Hippocampus/pathology , Male , Memory, Short-Term/drug effects , Mice, Transgenic , Neural Stem Cells/metabolism , Neurons/metabolism , Sex Characteristics , Time Factors , Vitamin D/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL