Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
J Pineal Res ; 76(3): e12952, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38587234

ABSTRACT

Melatonin (5-methoxy-N-acetyltryptamine) binds with high affinity and specificity to membrane receptors. Several receptor subtypes exist in different species, of which the mammalian MT1 and MT2 receptors are the best-characterized. They are members of the G protein-coupled receptor superfamily, preferentially coupling to Gi/o proteins but also to other G proteins in a cell-context-depending manner. In this review, experts on melatonin receptors will summarize the current state of the field. We briefly report on the discovery and classification of melatonin receptors, then focus on the molecular structure of human MT1 and MT2 receptors and highlight the importance of molecular simulations to identify new ligands and to understand the structural dynamics of these receptors. We then describe the state-of-the-art of the intracellular signaling pathways activated by melatonin receptors and their complexes. Brief statements on the molecular toolbox available for melatonin receptor studies and future perspectives will round-up this review.


Subject(s)
Melatonin , Receptor, Melatonin, MT1 , Animals , Humans , Receptors, Melatonin , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/metabolism , Melatonin/metabolism , Signal Transduction , Receptors, G-Protein-Coupled , Mammals/metabolism
2.
Cell Mol Life Sci ; 79(7): 361, 2022 Jun 13.
Article in English | MEDLINE | ID: mdl-35697820

ABSTRACT

COVID-19 is a complex disease with short- and long-term respiratory, inflammatory and neurological symptoms that are triggered by the infection with SARS-CoV-2. Invasion of the brain by SARS-CoV-2 has been observed in humans and is postulated to be involved in post-COVID state. Brain infection is particularly pronounced in the K18-hACE2 mouse model of COVID-19. Prevention of brain infection in the acute phase of the disease might thus be of therapeutic relevance to prevent long-lasting symptoms of COVID-19. We previously showed that melatonin or two prescribed structural analogs, agomelatine and ramelteon delay the onset of severe clinical symptoms and improve survival of SARS-CoV-2-infected K18-hACE2 mice. Here, we show that treatment of K18-hACE2 mice with melatonin and two melatonin-derived marketed drugs, agomelatine and ramelteon, prevents SARS-CoV-2 entry in the brain, thereby reducing virus-induced damage of small cerebral vessels, immune cell infiltration and brain inflammation. Molecular modeling analyses complemented by experimental studies in cells showed that SARS-CoV-2 entry in endothelial cells is prevented by melatonin binding to an allosteric-binding site on human angiotensin-converting enzyme 2 (ACE2), thus interfering with ACE2 function as an entry receptor for SARS-CoV-2. Our findings open new perspectives for the repurposing of melatonergic drugs and its clinically used analogs in the prevention of brain infection by SARS-CoV-2 and COVID-19-related long-term neurological symptoms.


Subject(s)
COVID-19 Drug Treatment , Melatonin , Angiotensin-Converting Enzyme 2 , Animals , Brain/metabolism , Endothelial Cells/metabolism , Melatonin/pharmacology , Melatonin/therapeutic use , Mice , Mice, Transgenic , Peptidyl-Dipeptidase A , SARS-CoV-2
3.
Int J Mol Sci ; 24(18)2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37762230

ABSTRACT

Simufilam is a novel oral drug candidate in Phase 3 clinical trials for Alzheimer's disease (AD) dementia. This small molecule binds an altered form of filamin A (FLNA) that occurs in AD. This drug action disrupts FLNA's aberrant linkage to the α7 nicotinic acetylcholine receptor (α7nAChR), thereby blocking soluble amyloid beta1-42 (Aß42)'s signaling via α7nAChR that hyperphosphorylates tau. Here, we aimed to clarify simufilam's mechanism. We now show that simufilam reduced Aß42 binding to α7nAChR with a 10-picomolar IC50 using time-resolved fluorescence resonance energy transfer (TR-FRET), a robust technology to detect highly sensitive molecular interactions. We also show that FLNA links to multiple inflammatory receptors in addition to Toll-like receptor 4 (TLR4) in postmortem human AD brains and in AD transgenic mice: TLR2, C-X-C chemokine receptor type 4 (CXCR4), C-C chemokine receptor type 5 (CCR5), and T-cell co-receptor cluster of differentiation 4 (CD4). These aberrant FLNA linkages, which can be induced in a healthy control brain by Aß42 incubation, were disrupted by simufilam. Simufilam reduced inflammatory cytokine release from Aß42-stimulated human astrocytes. In the AD transgenic mice, CCR5-G protein coupling was elevated, indicating persistent activation. Oral simufilam reduced both the FLNA-CCR5 linkage and the CCR5-G protein coupling in these mice, while restoring CCR5's responsivity to C-C chemokine ligand 3 (CCL3). By disrupting aberrant FLNA-receptor interactions critical to AD pathogenic pathways, simufilam may promote brain health.


Subject(s)
Alzheimer Disease , Mice , Humans , Animals , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Filamins/metabolism , Mice, Transgenic , Peptide Fragments/metabolism
4.
FASEB J ; 35(1): e21161, 2021 01.
Article in English | MEDLINE | ID: mdl-33156577

ABSTRACT

Association of G protein-coupled receptors into heterodimeric complexes has been reported for over 50 receptor pairs in vitro but functional in vivo validation remains a challenge. Our recent in vitro studies defined the functional fingerprint of heteromers composed of Gi -coupled melatonin MT2 receptors and Gq -coupled serotonin 5-HT2C receptors, in which melatonin transactivates phospholipase C (PLC) through 5-HT2C . Here, we identified this functional fingerprint in the mouse brain. Gq protein activation was probed by [35 S]GTPγS incorporation followed by Gq immunoprecipitation, and PLC activation by determining the inositol phosphate levels in brain lysates of animals previously treated with melatonin. Melatonin concentration-dependently activated Gq proteins and PLC in the hypothalamus and cerebellum but not in cortex. These effects were inhibited by the 5-HT2C receptor-specific inverse agonist SB-243213, and were absent in MT2 and 5-HT2C knockout mice, fully recapitulating previous in vitro data and indicating the involvement of MT2 /5-HT2C heteromers. The antidepressant agomelatine had a similar effect than melatonin when applied alone but blocked the melatonin-promoted Gq activation due to its 5-HT2C antagonistic component. Collectively, we provide strong functional evidence for the existence of MT2 /5-HT2C heteromeric complexes in mouse brain. These heteromers might participate in the in vivo effects of agomelatine.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Enzymologic , Protein Multimerization , Receptor, Melatonin, MT2/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Transcriptional Activation , Type C Phospholipases/biosynthesis , Acetamides/pharmacology , Animals , Indoles/pharmacology , Male , Mice , Mice, Knockout , Pyridines/pharmacology , Receptor, Melatonin, MT2/genetics , Receptor, Serotonin, 5-HT2C/genetics , Type C Phospholipases/genetics
5.
J Pineal Res ; 72(1): e12772, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34586649

ABSTRACT

As the COVID-19 pandemic grows, several therapeutic candidates are being tested or undergoing clinical trials. Although prophylactic vaccination against SARS-CoV-2 infection has been shown to be effective, no definitive treatment exists to date in the event of infection. The rapid spread of infection by SARS-CoV-2 and its variants fully warrants the continued evaluation of drug treatments for COVID-19, especially in the context of repurposing of already available and safe drugs. Here, we explored the therapeutic potential of melatonin and melatonergic compounds in attenuating COVID-19 pathogenesis in mice expressing human ACE2 receptor (K18-hACE2), strongly susceptible to SARS-CoV-2 infection. Daily administration of melatonin, agomelatine, or ramelteon delays the occurrence of severe clinical outcome with improvement of survival, especially with high melatonin dose. Although no changes in most lung inflammatory cytokines are observed, treatment with melatonergic compounds limits the exacerbated local lung production of type I and type III interferons, which is likely associated with the observed improved symptoms in treated mice. The promising results from this preclinical study should encourage studies examining the benefits of repurposing melatonergic drugs to treat COVID-19 and related diseases in humans.


Subject(s)
Acetamides/pharmacology , COVID-19 Drug Treatment , COVID-19 , Indenes/pharmacology , Melatonin/pharmacology , SARS-CoV-2/drug effects , Animals , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Viral Load/drug effects
6.
Neuroendocrinology ; 111(4): 370-387, 2021.
Article in English | MEDLINE | ID: mdl-32335558

ABSTRACT

INTRODUCTION: Metabolic dysfunction is now recognized as a pivotal component of Alzheimer's disease (AD), the most common dementia worldwide. However, the precise molecular mechanisms linking metabolic dysfunction to AD remain elusive. OBJECTIVE: Here, we investigated the direct impact of soluble oligomeric amyloid beta (Aß) peptides, the main molecular hallmark of AD, on the leptin system, a major component of central energy metabolism regulation. METHODS: We developed a new time-resolved fluorescence resonance energy transfer-based Aß binding assay for the leptin receptor (LepR) and studied the effect of Aß on LepR function in several in vitro assays. The in vivo effect of Aß on LepR function was studied in an Aß-specific AD mouse model and in pro-opiomelanocortin (POMC) neurons of the hypothalamic arcuate nucleus. RESULTS: We revealed specific and high-affinity (Ki = 0.1 nM) binding of Aß to LepR. Pharmacological characterization of this interaction showed that Aß binds allosterically to the extracellular domain of LepR and negatively affects receptor function. Negative allosteric modulation of LepR by Aß was detected at the level of signaling pathways (STAT-3, AKT, and ERK) in vitroand in vivo. Importantly, the leptin-induced response of POMC neurons, key players in the regulation of metabolic function, was completely abolished in the presence of Aß. CONCLUSION: Our data indicate that Aß is a negative allosteric modulator of LepR, resulting in impaired leptin action, and qualify LepR as a new and direct target of Aß oligomers. Preventing the interaction of Aß with LepR might improve both the metabolic and cognitive dysfunctions in AD condition.


Subject(s)
Allosteric Regulation/physiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Leptin/metabolism , Pro-Opiomelanocortin/metabolism , Receptors, Leptin/metabolism , Animals , Cell Line , Disease Models, Animal , HEK293 Cells , Humans , Male , Mice , Signal Transduction/physiology
7.
J Pineal Res ; 70(2): e12712, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33332653

ABSTRACT

A multitude of effects has been attributed to melatonin at pmol/L to mmol/L concentrations. More than fifteen targets have been proposed for melatonin but only few of them are well characterized. The current guidelines intend to provide a framework to improve and rationalize the characterization of melatonin targets and effects. They should be considered as mandatory guidelines and minimum requirements for manuscripts submitted to the Journal of Pineal Research.


Subject(s)
Melatonin/metabolism , Quinone Reductases/metabolism , Receptors, Melatonin/metabolism , Animals
8.
J Pineal Res ; 68(4): e12641, 2020 May.
Article in English | MEDLINE | ID: mdl-32080899

ABSTRACT

G protein-coupled receptors (GPCRs) transmit extracellular signals into cells by activating G protein- and ß-arrestin-dependent pathways. Extracellular signal-regulated kinases (ERKs) play a central role in integrating these different linear inputs coming from a variety of GPCRs to regulate cellular functions. Here, we investigated human melatonin MT1 and MT2 receptors signaling through the ERK1/2 cascade by employing different biochemical techniques together with pharmacological inhibitors and siRNA molecules. We show that ERK1/2 activation by both receptors is exclusively G protein-dependent, without any participation of ß-arrestin1/2 in HEK293 cells. ERK1/2 activation by MT1 is only mediated though Gi/o proteins, while MT2 is dependent on the cooperative activation of Gi/o and Gq/11 proteins. In the absence of Gq/11 proteins, however, MT2 -induced ERK1/2 activation switches to a ß-arrestin1/2-dependent mode. The signaling cascade downstream of G proteins is the same for both receptors and involves activation of the PI3K/PKCζ/c-Raf/MEK/ERK cascade. The differential G protein dependency of MT1 - and MT2 -mediated ERK activation was confirmed at the level of EGR1 and FOS gene expression, two ERK1/2 target genes. Gi/o /Gq/11 cooperativity was also observed in Neuroscreen-1 cells expressing endogenous MT2 , whereas in the mouse retina, where MT2 is engaged into MT1 /MT2 heterodimers, ERK1/2 signaling is exclusively Gi/o -dependent. Collectively, our data reveal differential signaling modes of MT1 and MT2 in terms of ERK1/2 activation, with an unexpected Gi/o /Gq/11 cooperativity exclusively for MT2 . The plasticity of ERK activation by MT2 is highlighted by the switch to a ß-arrestin1/2-dependent mode in the absence of Gq/11 proteins and by the switch to a Gi/o mode when engaged into MT1 /MT2 heterodimers, revealing a new mechanism underlying tissue-specific responses to melatonin.


Subject(s)
GTP-Binding Proteins/metabolism , MAP Kinase Signaling System/physiology , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/metabolism
9.
Proc Natl Acad Sci U S A ; 114(38): E7997-E8006, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28874589

ABSTRACT

G protein-coupled receptors (GPCRs) are classically characterized as cell-surface receptors transmitting extracellular signals into cells. Here we show that central components of a GPCR signaling system comprised of the melatonin type 1 receptor (MT1), its associated G protein, and ß-arrestins are on and within neuronal mitochondria. We discovered that the ligand melatonin is exclusively synthesized in the mitochondrial matrix and released by the organelle activating the mitochondrial MT1 signal-transduction pathway inhibiting stress-mediated cytochrome c release and caspase activation. These findings coupled with our observation that mitochondrial MT1 overexpression reduces ischemic brain injury in mice delineate a mitochondrial GPCR mechanism contributing to the neuroprotective action of melatonin. We propose a new term, "automitocrine," analogous to "autocrine" when a similar phenomenon occurs at the cellular level, to describe this unexpected intracellular organelle ligand-receptor pathway that opens a new research avenue investigating mitochondrial GPCR biology.


Subject(s)
Brain Injuries/metabolism , Brain Ischemia/metabolism , Melatonin/biosynthesis , Mitochondria/metabolism , Receptor, Melatonin, MT1/metabolism , Signal Transduction , Animals , Brain Injuries/genetics , Brain Ischemia/genetics , Cytochromes c/genetics , Cytochromes c/metabolism , Male , Melatonin/genetics , Mice , Mitochondria/genetics , Receptor, Melatonin, MT1/genetics
10.
J Pineal Res ; 67(4): e12606, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31442321

ABSTRACT

The tryptophan derivative melatonin is an evolutionary old molecule that is involved in a pleiotropy of physiological functions. In humans, age-related decline of circulating melatonin levels and/or dysregulation of its circadian synthesis pattern have been associated with several disorders and disease states. Several molecular targets have been proposed for melatonin since its discovery, in 1959. Among them, melatonin MT1 and MT2 receptors are the best characterized melatonin targets, mediating melatonin effects in a variety of tissues. They belong to the superfamily of G protein-coupled receptors. Two back-to-back articles published in the "Nature" Journal earlier this year present the first crystal structures of the human MT1 and MT2 in its inactive states. Here, we will briefly outline the discovery path of melatonin receptors until their structural elucidation and discuss how these new findings will guide future research toward a better understanding of their function and rational drug design.


Subject(s)
Melatonin/chemistry , Melatonin/metabolism , Receptor, Melatonin, MT1 , Receptor, Melatonin, MT2 , Signal Transduction , Animals , History, 20th Century , History, 21st Century , Humans , Melatonin/history , Protein Structure, Tertiary , Receptor, Melatonin, MT1/chemistry , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/chemistry , Receptor, Melatonin, MT2/metabolism
11.
J Pineal Res ; 66(2): e12540, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30475390

ABSTRACT

Melatonin receptors play important roles in the regulation of circadian and seasonal rhythms, sleep, retinal functions, the immune system, depression, and type 2 diabetes development. Melatonin receptors are approved drug targets for insomnia, non-24-hour sleep-wake disorders, and major depressive disorders. In mammals, two melatonin receptors (MTRs) exist, MT1 and MT2 , belonging to the G protein-coupled receptor (GPCR) superfamily. Similar to most other GPCRs, reliable antibodies recognizing melatonin receptors proved to be difficult to obtain. Here, we describe the development of the first monoclonal antibodies (mABs) for mouse MT1 and MT2 . Purified antibodies were extensively characterized for specific reactivity with mouse, rat, and human MT1 and MT2 by Western blot, immunoprecipitation, immunofluorescence, and proximity ligation assay. Several mABs were specific for either mouse MT1 or MT2 . None of the mABs cross-reacted with rat MTRs, and some were able to react with human MTRs. The specificity of the selected mABs was validated by immunofluorescence microscopy in three established locations (retina, suprachiasmatic nuclei, pituitary gland) for MTR expression in mice using MTR-KO mice as control. MT2 expression was not detected in mouse insulinoma MIN6 cells or pancreatic beta-cells. Collectively, we report the first monoclonal antibodies recognizing recombinant and native mouse melatonin receptors that will be valuable tools for future studies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Receptor, Melatonin, MT1/analysis , Receptor, Melatonin, MT2/analysis , Animals , Mice , Protein Domains , Receptor, Melatonin, MT1/immunology , Receptor, Melatonin, MT2/immunology
12.
J Pineal Res ; 64(3)2018 Apr.
Article in English | MEDLINE | ID: mdl-29247541

ABSTRACT

Recent genetic studies have highlighted the potential involvement of melatonin receptor 1 (MT1 ) and melatonin receptor 2 (MT2 ) in the pathogenesis of type 2 diabetes. Here, we report that mice lacking MT1 (MT1 KO) tend to accumulate more fat mass than WT mice and exhibit marked systemic insulin resistance. Additional experiments revealed that the main insulin signaling pathway affected by the loss of MT1 was the activation of phosphatidylinositol-3-kinase (PI3K). Transcripts of both catalytic and regulatory subunits of PI3K were strongly downregulated within MT1 KO mice. Moreover, the suppression of nocturnal melatonin levels within WT mice, by exposing mice to constant light, resulted in impaired PI3K activity and insulin resistance during the day, similar to what was observed in MT1 KO mice. Inversely, administration of melatonin to WT mice exposed to constant light was sufficient and necessary to restore insulin-mediated PI3K activity and insulin sensitivity. Hence, our data demonstrate that the activation of MT1 signaling at night modulates insulin sensitivity during the day via the regulation of the PI3K transcription and activity. Lastly, we provide evidence that decreased expression of MTNR1A (MT1 ) in the liver of diabetic individuals is associated with poorly controlled diabetes.


Subject(s)
Circadian Rhythm/physiology , Insulin Resistance/physiology , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Melatonin, MT1/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Male , Mice , Mice, Knockout
13.
Int J Mol Sci ; 19(8)2018 Jul 26.
Article in English | MEDLINE | ID: mdl-30049944

ABSTRACT

Melatonin (5-methoxy-N-acetylserotonin), the pineal hormone, is also synthesized by immune-competent cells. The pineal hormone signals darkness, while melatonin synthesized on demand by activated macrophages at any hour of the day acts locally, favoring regulatory/tolerant phenotypes. Activation of ß-adrenoceptors in pinealocytes is the main route for triggering melatonin synthesis. However, despite the well-known role of ß-adrenoceptors in the resolution macrophage phenotype (M2), and the relevance of macrophage synthesized melatonin in facilitating phagocytic activity, there is no information regarding whether activation of ß-adrenoceptors would induce melatonin synthesis by monocytes. Here we show that catecholamines stimulate melatonin synthesis in bone marrow-derived dendritic cells and RAW 264.7 macrophages. Activation of ß-adrenoceptors promotes the synthesis of melatonin by stimulating cyclic AMP/protein kinase A (PKA) pathway and by activating the nuclear translocation of NF-κB. Considering the great number of macrophages around sympathetic nerve terminals, and the relevance of this system for maintaining macrophages in stages compatible to low-grade inflammation, our data open the possibility that extra-pineal melatonin acts as an autocrine/paracrine signal in macrophages under resolution or tolerant phenotypes.


Subject(s)
Macrophages/metabolism , Melatonin/metabolism , Phagocytes/metabolism , Receptors, Adrenergic, beta/metabolism , Animals , Biosynthetic Pathways , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Male , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , Signal Transduction
14.
Neuroendocrinology ; 104(2): 126-134, 2017.
Article in English | MEDLINE | ID: mdl-26954684

ABSTRACT

BACKGROUND/AIM: The nocturnal production of melatonin by the pineal gland is triggered by sympathetic activation of adrenoceptors and may be modulated by immunological signals. The effect of glucocorticoids on nocturnal melatonin synthesis is controversial; both stimulatory and inhibitory effects have been reported. During pathophysiological processes, an increased sympathetic tonus could result in different patterns of adrenoceptor activation in the pineal gland. Therefore, in this investigation, we evaluated whether the pattern of adrenergic stimulation of the pineal gland drives the direction of the glucocorticoid effect on melatonin production. METHODS: The corticosterone effect on the pineal hormonal production induced by ß-adrenoceptor or ß+α1-adrenoceptor activation was evaluated in cultured glands. We also investigated whether the in vivo lipopolysaccharide (LPS)-induced inhibition of melatonin is dependent on the interaction of glucocorticoids and the α1-adrenoceptor in adrenalectomized animals and on the in vivo blockade of glucocorticoid receptors (GRs) or the α1-adrenoceptor. RESULTS: Corticosterone potentiated ß-adrenoceptor-induced pineal melatonin synthesis, whilst corticosterone-dependent inhibition was observed when melatonin production was induced by ß+α1-adrenoceptors agonists. The inhibitory effect of corticosterone is mediated by GR, as it was abolished in the presence of a GR antagonist. Moreover, LPS-induced reduction in melatonin nocturnal plasma content was reversed by adrenalectomy and by antagonizing GR or α1-adrenoceptors. CONCLUSIONS: The dual effect of corticosterone on pineal melatonin synthesis is determined by the activation pattern of adrenoceptors (ß or ß+α1) in the gland during GR activation, suggesting that increased activation of the sympathetic system and the hypothalamic-pituitary-adrenal axis are necessary for the control of melatonin production during defense responses.


Subject(s)
Catecholamines/metabolism , Corticosterone/administration & dosage , Melatonin/biosynthesis , Pineal Gland/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Agonists/administration & dosage , Animals , Inflammation/metabolism , Isoproterenol/administration & dosage , Lipopolysaccharides , Male , Pineal Gland/drug effects , Rats , Rats, Wistar
15.
J Cell Physiol ; 231(9): 1953-63, 2016 09.
Article in English | MEDLINE | ID: mdl-26728119

ABSTRACT

The phenotype of primary cells in culture varies according to the donor environmental condition. We recently showed that the time of the day imposes a molecular program linked to the inflammatory response that is heritable in culture. Here we investigated whether microRNAs (miRNAs) would show differential expression according to the time when cells were obtained, namely daytime or nighttime. Cells obtained from explants of cremaster muscle and cultivated until confluence (∼20 days) presented high CD133 expression. Global miRNA expression analysis was performed through deep sequencing in order to compare both cultured cells. A total of 504 mature miRNAs were identified, with a specific miRNA signature being associated to the light versus dark phase of a circadian cycle. miR-1249 and miR-129-2-3p were highly expressed in daytime cells, while miR-182, miR-96-5p, miR-146a-3p, miR-146a-5p, and miR-223-3p were highly expressed in nighttime cells. Nighttime cells are regulated for programs involved in cell processes and development, as well as in the inflammation, cell differentiation and maturation; while daytime cells express miRNAs that control stemness and cytoskeleton remodeling. In summary, the time of the day imposes a differential profile regarding to miRNA signature on CD133(+) cells in culture. Understanding this daily profile in the phenotype of cultured cells is highly relevant for clinical outputs, including cellular therapy approaches. J. Cell. Physiol. 231: 1953-1963, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Differentiation/genetics , Inflammation/genetics , MicroRNAs/genetics , Photoperiod , AC133 Antigen/immunology , Animals , Cells, Cultured , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Male , Rats, Wistar
16.
FASEB J ; 29(6): 2566-82, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25757565

ABSTRACT

Melatonin is the hormone produced by the pineal gland known to regulate physiologic rhythms and to display immunomodulatory and neuroprotective properties. It has been reported that Alzheimer disease patients show impaired melatonin production and altered expression of the 2 G protein-coupled melatonin receptors (MTRs), MT1 and MT2, but the underlying mechanisms are not known. Here we evaluated whether this dysfunction of the melatonergic system is directly caused by amyloid ß peptides (Aß(1-40) and Aß(1-42)). Aß treatment of rat pineal glands elicited an inflammatory response within the gland, evidenced by the up-regulation of 52 inflammatory genes, and decreased the production of melatonin up to 75% compared to vehicle-treated glands. Blocking NF-κB activity prevented this effect. Exposure of HEK293 cells stably expressing recombinant MT1 or MT2 receptors to Aß lead to a 40% reduction in [(125)I]iodomelatonin binding to MT1. ERK1/2 activation triggered by MTRs, but not by the ß2-adrenergic receptor, was markedly impaired by Aß in HEK293 transfected cells, as well as in primary rat endothelial cells expressing endogenous MTRs. Our data reveal the melatonergic system as a new target of Aß, opening new perspectives to Alzheimer disease diagnosis and therapeutic intervention.


Subject(s)
Amyloid beta-Peptides/pharmacology , MAP Kinase Signaling System/drug effects , Melatonin/biosynthesis , Peptide Fragments/pharmacology , Pineal Gland/drug effects , Receptors, Melatonin/metabolism , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/drug effects , HEK293 Cells , Humans , Immunoblotting , Male , Pineal Gland/metabolism , Protein Multimerization/drug effects , Rats, Wistar , Receptors, Melatonin/chemistry , Receptors, Melatonin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tissue Culture Techniques
17.
Int J Mol Sci ; 14(6): 10979-97, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23708099

ABSTRACT

Pineal gland melatonin is the darkness hormone, while extra-pineal melatonin produced by the gonads, gut, retina, and immune competent cells acts as a paracrine or autocrine mediator. The well-known immunomodulatory effect of melatonin is observed either as an endocrine, a paracrine or an autocrine response. In mammals, nuclear translocation of nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) blocks noradrenaline-induced melatonin synthesis in pinealocytes, which induces melatonin synthesis in macrophages. In addition, melatonin reduces NF-κB activation in pinealocytes and immune competent cells. Therefore, pathogen- or danger-associated molecular patterns transiently switch the synthesis of melatonin from pinealocytes to immune competent cells, and as the response progresses melatonin inhibition of NF-κB activity leads these cells to a more quiescent state. The opposite effect of NF-κB in pinealocytes and immune competent cells is due to different NF-κB dimers recruited in each phase of the defense response. This coordinated shift of the source of melatonin driven by NF-κB is called the immune-pineal axis. Finally, we discuss how this concept might be relevant to a better understanding of pathological conditions with impaired melatonin rhythms and hope it opens new horizons for the research of side effects of melatonin-based therapies.


Subject(s)
Immune System/cytology , Immune System/metabolism , Melatonin/metabolism , NF-kappa B/metabolism , Pineal Gland/cytology , Pineal Gland/metabolism , Animals , Humans , Melatonin/biosynthesis , Signal Transduction
18.
Eur J Med Chem ; 249: 115152, 2023 Mar 05.
Article in English | MEDLINE | ID: mdl-36724633

ABSTRACT

COVID-19 is a complex disease with short-term and long-term respiratory, inflammatory and neurological symptoms that are triggered by the infection with SARS-CoV-2. As many drugs targeting single targets showed only limited effectiveness against COVID-19, here, we aimed to explore a multi-target strategy. We synthesized a focused compound library based on C2-substituted indolealkylamines (tryptamines and 5-hydroxytryptamines) with activity for three potential COVID-19-related proteins, namely melatonin receptors, calmodulin and human angiotensin converting enzyme 2 (hACE2). Two molecules from the library, 5e and h, exhibit affinities in the high nanomolar range for melatonin receptors, inhibit the calmodulin-dependent calmodulin kinase II activity and the interaction of the SARS-CoV-2 Spike protein with hACE2 at micromolar concentrations. Both compounds inhibit SARS-CoV-2 entry into host cells and 5h decreases SARS-CoV-2 replication and MPro enzyme activity in addition. In conclusion, we provide a proof-of-concept for the successful design of multi-target compounds based on the tryptamine scaffold. Optimization of these preliminary hit compounds could potentially provide drug candidates to treat COVID-19 and other coronavirus diseases.


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , COVID-19 Drug Treatment , Calmodulin , Receptors, Melatonin
19.
Elife ; 122023 03 14.
Article in English | MEDLINE | ID: mdl-36917493

ABSTRACT

Aggregates of the tau protein are a well-known hallmark of several neurodegenerative diseases, collectively referred to as tauopathies, including frontal temporal dementia and Alzheimer's disease (AD). Monitoring the transformation process of tau from physiological monomers into pathological oligomers or aggregates in a high-throughput, quantitative manner and in a cellular context is still a major challenge in the field. Identifying molecules able to interfere with those processes is of high therapeutic interest. Here, we developed a series of inter- and intramolecular tau biosensors based on the highly sensitive Nanoluciferase (Nluc) binary technology (NanoBiT) able to monitor the pathological conformational change and self-interaction of tau in living cells. Our repertoire of tau biosensors reliably reports i. molecular proximity of physiological full-length tau at microtubules; ii. changes in tau conformation and self-interaction associated with tau phosphorylation, as well as iii. tau interaction induced by seeds of recombinant tau or from mouse brain lysates of a mouse model of tau pathology. By comparing biosensors comprising different tau forms (i.e. full-length or short fragments, wild-type, or the disease-associated tau(P301L) variant) further insights into the tau transformation process are obtained. Proof-of-concept data for the high-throughput suitability and identification of molecules interfering with the pathological tau transformation processes are presented. This novel repertoire of tau biosensors is aimed to boost the disclosure of molecular mechanisms underlying pathological tau transformation in living cells and to discover new drug candidates for tau-related neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Tauopathies , Mice , Animals , tau Proteins/genetics , tau Proteins/metabolism , Alzheimer Disease/metabolism , Tauopathies/pathology , Microtubules/metabolism , Neurons/physiology , Brain/metabolism
20.
PLoS One ; 18(4): e0283941, 2023.
Article in English | MEDLINE | ID: mdl-37014877

ABSTRACT

Intracellular accumulation of tau protein is a hallmark of Alzheimer's Disease and Progressive Supranuclear Palsy, as well as other neurodegenerative disorders collectively known as tauopathies. Despite our increasing understanding of the mechanisms leading to the initiation and progression of tau pathology, the field still lacks appropriate disease models to facilitate drug discovery. Here, we established a novel and modulatable seeding-based neuronal model of full-length 4R tau accumulation using humanized mouse cortical neurons and seeds from P301S human tau transgenic animals. The model shows specific and consistent formation of intraneuronal insoluble full-length 4R tau inclusions, which are positive for known markers of tau pathology (AT8, PHF-1, MC-1), and creates seeding competent tau. The formation of new inclusions can be prevented by treatment with tau siRNA, providing a robust internal control for use in qualifying the assessment of potential therapeutic candidates aimed at reducing the intracellular pool of tau. In addition, the experimental set up and data analysis techniques used provide consistent results in larger-scale designs that required multiple rounds of independent experiments, making this is a versatile and valuable cellular model for fundamental and early pre-clinical research of tau-targeted therapies.


Subject(s)
Alzheimer Disease , Tauopathies , Mice , Animals , Humans , tau Proteins/genetics , tau Proteins/metabolism , Mice, Transgenic , Brain/metabolism , Tauopathies/metabolism , Alzheimer Disease/pathology , Neurons/metabolism , Drug Discovery
SELECTION OF CITATIONS
SEARCH DETAIL