Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 15: 1386224, 2024.
Article in English | MEDLINE | ID: mdl-38595916

ABSTRACT

Introduction: Alzheimer's disease (AD) is the most common neurodegenerative disorder affecting the elderly population worldwide. Due to the multifactorial nature of the disease, involving impairment of cholinergic neurotransmission and immune system, previous attempts to find effective treatments have faced challenges. Methods: In such scenario, we attempted to investigate the effects of alpha-glyceryl-phosphoryl-choline (α-GPC), a cholinomimetic molecule, on neuroinflammation and memory outcome in the triple transgenic mouse model of AD (3xTg-AD). Mice were enrolled at 4 months of age, treated orally with α-GPC dissolved in drinking water at a concentration resulting in an average daily dose of 100 mg/kg for 8 months and sacrificed at 12 months of age. Thereafter, inflammatory markers, as well as cognitive parameters, were measured. Results: Chronic α-GPC treatment reduced accumulation of amyloid deposits and led to a substantial re-balance of the inflammatory response of resident innate immune cells, astrocytes and microglia. Specifically, fluorescent immunohistochemistry and Western blot analysis showed that α-GPC contributed to reduction of cortical and hippocampal reactive astrocytes and pro-inflammatory microglia, concurrently increasing the expression of anti-inflammatory molecules. Whereas α-GPC beneficially affect the synaptic marker synaptophysin in the hippocampus. Furthermore, we observed that α-GPC was effective in restoring cognitive dysfunction, as measured by the Novel Object Recognition test, wherein 3xTg-AD mice treated with α-GPC significantly spent more time exploring the novel object compared to 3xTg-AD untreated mice. Discussion: In conclusion, chronic treatment with α-GPC exhibited a significant anti-inflammatory activity and sustained the key function of hippocampal synapses, crucial for the maintenance of a regular cognitive status. In light of our results, we suggest that α-GPC could be exploited as a promising therapeutic approach in early phases of AD.

2.
Cells ; 13(4)2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38391922

ABSTRACT

Alzheimer's disease (AD), marked by cognitive impairment, predominantly affects the brain regions regulated by cholinergic innervation, such as the cerebral cortex and hippocampus. Cholinergic dysfunction, a key contributor to age-related cognitive decline, has spurred investigations into potential therapeutic interventions. We have previously shown that choline alphoscerate (α-GPC), a cholinergic neurotransmission-enhancing agent, protects from Aß-mediated neurotoxicity. Herein, we investigated the effects of α-GPC on the microglial phenotype in response to Aß via modulation of the nicotinic alpha-7 acetylcholine receptor (α7 nAChR). BV2 microglial cells were pre-treated for 1 h with α-GPC and were treated for 24, 48, and 72 h with Aß1-42 and/or α-BTX, a selective α7nAchR antagonist. Fluorescent immunocytochemistry and Western blot analysis showed that α-GPC was able to antagonize Aß-induced inflammatory effects. Of note, α-GPC exerted its anti-inflammatory effect by directly activating the α7nAChR receptor, as suggested by the induction of an increase in [Ca2+]i and Ach-like currents. Considering that cholinergic transmission appears crucial in regulating the inflammatory profiles of glial cells, its modulation emerges as a potential pharmaco-therapeutic target to improve outcomes in inflammatory neurodegenerative disorders, such as AD.


Subject(s)
Alzheimer Disease , Receptors, Nicotinic , Humans , Alzheimer Disease/drug therapy , Microglia/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Glycerylphosphorylcholine/pharmacology , Amyloid beta-Peptides/metabolism , Receptors, Nicotinic/metabolism , Synaptic Transmission , Cholinergic Agents
3.
Int J Mol Sci ; 23(19)2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36232931

ABSTRACT

TRAIL, a member of TNF superfamily, is a potent inducer of neuronal death. Neurotoxic effects of TRAIL appear mediated by its death receptor TRAIL-R2/DR5. To assess the role of TRAIL/TRAIL-R2 pathway in AD-related neurodegeneration, we studied the impact of the treatment with amyloid-ß (Aß) upon cell viability and inflammation in TRAIL-R-deficient mice (TRAIL-R-/-). Here, we demonstrate that the lack of TRAIL-R2 protects from death cultured TRAIL-R-/- mouse embryonic hippocampal cells after treatment with either Aß1-42 or TRAIL. Consistently, stereotaxic injection of Aß1-42 resulted in blunted caspase activation, as well as in reduction of JNK phosphorylation and increased AKT phosphorylation in TRAIL-R-/- mice. Moreover, the lack of TRAIL-R2 was associated with blunted constitutive p53 expression in mice that have undergone Aß1-42 treatment, as well as in decrease of phosphorylated forms of tau and GSK3ß proteins. Likewise, TRAIL-R2 appears essential to both TRAIL and Aß-mediated neurotoxicity and inflammation. Indeed, hippocampi of TRAIL-R-/- mice challenged with Aß1-42, showed a slight expression of microglial (Iba-1) and astrocytic (GFAP) markers along with attenuated levels of IL-1ß, TNF-α, NOS2 and COX2. In conclusion, the bulk of these results demonstrate that the constitutive lack of TRAIL-R2 is associated with a substantial reduction of noxious effects of Aß1-42, providing further evidence on the prominent role played by TRAIL in course of Aß-related neurodegeneration and confirming that the TRAIL system represents a potential target for innovative AD therapy.


Subject(s)
Neurotoxicity Syndromes , Receptors, TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha , Amyloid beta-Peptides/metabolism , Animals , Caspases/metabolism , Cyclooxygenase 2/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Inflammation , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53
4.
Cells ; 11(17)2022 09 01.
Article in English | MEDLINE | ID: mdl-36078138

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia worldwide, with a complex, poorly understood pathogenesis. Cerebral atrophy, amyloid-ß (Aß) plaques, and neurofibrillary tangles represent the main pathological hallmarks of the AD brain. Recently, neuroinflammation has been recognized as a prominent feature of the AD brain and substantial evidence suggests that the inflammatory response modulates disease progression. Additionally, dysregulation of calcium (Ca2+) homeostasis represents another early factor involved in the AD pathogenesis, as intracellular Ca2+ concentration is essential to ensure proper cellular and neuronal functions. Although growing evidence supports the involvement of Ca2+ in the mechanisms of neurodegeneration-related inflammatory processes, scant data are available on its contribution in microglia and astrocytes functioning, both in health and throughout the AD continuum. Nevertheless, AD-related aberrant Ca2+ signalling in astrocytes and microglia is crucially involved in the mechanisms underpinning neuroinflammatory processes that, in turn, impact neuronal Ca2+ homeostasis and brain function. In this light, we attempted to provide an overview of the current understanding of the interactions between the glia cells-mediated inflammatory responses and the molecular mechanisms involved in Ca2+ homeostasis dysregulation in AD.


Subject(s)
Alzheimer Disease , Microglia , Alzheimer Disease/pathology , Astrocytes/pathology , Homeostasis , Humans , Microglia/pathology , Neuroinflammatory Diseases , Plaque, Amyloid
5.
Infect Dis Rep ; 14(3): 360-371, 2022 May 11.
Article in English | MEDLINE | ID: mdl-35645219

ABSTRACT

Two years have passed since WHO declared a pandemic state for SARS-CoV-2 infection. COVID-19 pathogenesis consists of a first viral phase responsible for early symptoms followed by an inflammatory phase, cytokine-mediated, responsible for late-onset manifestations up to ARDS. The dysregulated immune response has an outstanding role in the progression of pulmonary damage in COVID-19. IL-6, through the induction of pro-inflammatory chemokines and cytokines, plays a key role in the development and maintenance of inflammation, acting as a pioneer of the hyperinflammatory condition and cytokine storm in severe COVID-19. Therefore, drugs targeting both IL-6 and IL-6 receptors have been evaluated in order to blunt the abnormal SARS-CoV-2-induced cytokine release. Sarilumab, a high-affinity anti-IL-6 receptor antibody, may represent a promising weapon to treat the fearsome hyperinflammatory phase by improving the outcome of patients with moderate-to-severe COVID-19 pneumonia. Further prospective and well-designed clinical studies with larger sample sizes and long-term follow-up are needed to assess the efficacy and the safety of this therapeutic approach to achieve improved outcomes in COVID-19.

6.
IDCases ; 28: e01462, 2022.
Article in English | MEDLINE | ID: mdl-35265458

ABSTRACT

Enterococcus faecalis infective endocarditis (EFIE) continues to be a very serious disease, showing considerable morbidity and mortality rates which are influenced by the spread of multi-drug resistant strains occurred in the last decades. Although aminoglycosides were considered the treatment of choice of EIFE, in recent years several studies have investigated alternative therapeutic approaches, including combinations of beta-lactams, mainly because of the aminoglycoside-renowned nephrotoxicity and the widespread development of high-level aminoglycosides resistance (HLAR). In this scenario, we reported a case involving a prosthetic valve infective endocarditis caused by an aminoglycoside-resistant E. faecalis strain which was successfully treated with ampicillin plus ceftriaxone despite the presence of artificial heart valve and the patient's severe clinical conditions.

7.
Antibiotics (Basel) ; 12(1)2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36671250

ABSTRACT

Multidrug resistant Gram-negative bacteremia represents a therapeutic challenge clinicians have to deal with. This concern becomes more difficult when causing germs are represented by carbapenem resistant Acinetobacter baumannii or difficult-to-treat Pseudomonas aeruginosa. Few antibiotics are available against these cumbersome bacteria, although literature data are not conclusive, especially for Acinetobacter. Cefiderocol could represent a valid antibiotic choice, being a molecule with an innovative mechanism of action capable of overcoming common resistance pathways, whereas intravenous fosfomycin may be an appropriate partner either enhancing cefiderocol activity or avoiding resistance development. Here we report two patients with MDR Gram negative bacteremia who were successfully treated with a cefiderocol/fosfomycin combination.

8.
Pharmaceuticals (Basel) ; 16(1)2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36678564

ABSTRACT

Psilocybin has been suggested as a promising transdiagnostic treatment strategy for a wide range of psychiatric disorders. Recent findings showed that psychedelic-assisted/"psycholitic" psychotherapy should provide significant and sustained alleviation of depressive symptoms. However, to date, there have been several study limitations (e.g., small sample sizes, blinding, limited follow-up, highly screened treatment populations) and some health/political issues, including practitioners' experience, lack of standardized protocols, psychedelics' legal status, ethical concerns, and potential psychological/psychopathological/medical untoward effects. The focus here is on a range of clinical and methodological issues, also aiming at outlining some possible suggestions. We are confident that newer evidence, more precise protocols, and eventual reclassification policies may allow a better understanding of the real potential of psilocybin as a transdiagnostic therapeutic molecule.

9.
J Clin Med ; 10(19)2021 Oct 06.
Article in English | MEDLINE | ID: mdl-34640612

ABSTRACT

Enterococcus faecalis infective endocarditis (EFIE) continues to represent a potentially fatal infectious disease characterized by elevated morbidity and mortality. Despite advances in antimicrobial therapy, changing demographics and the reduced availability of useful antibiotics combined with the dissemination of multi-drug resistant strains, the mortality rate remained unchanged in the last decades. Nowadays, optimizing the antibiotic regimen is still of paramount importance. Historically, aminoglycosides were considered as a cornerstone for treatment even though their use is associated with a high risk of kidney failure. It is against this background that, in recent years, several studies have been carried in order to assess the validity of alternative therapeutic approaches, including combinations of beta-lactams, that, acting synergistically, have yielded useful results in different clinical settings. In this scenario, we searched and critically report clinical studies assessing the efficacy and safety of double beta-lactam therapy in treating EFIE.

10.
Cell Death Dis ; 12(10): 905, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34611142

ABSTRACT

Age-related disorders, such as Alzheimer's disease (AD) and age-related macular degeneration (AMD) share common features such as amyloid-ß (Aß) protein accumulation. Retinal deposition of Aß aggregates in AMD patients has suggested a potential link between AMD and AD. In the present study, we analyzed the expression pattern of a focused set of miRNAs, previously found to be involved in both AD and AMD, in the retina of a triple transgenic mouse model of AD (3xTg-AD) at different time-points. Several miRNAs were differentially expressed in the retina of 3xTg-AD mice, compared to the retina of age-matched wild-type (WT) mice. In particular, bioinformatic analysis revealed that miR-155 had a central role in miRNA-gene network stability, regulating several pathways, including apoptotic and inflammatory signaling pathways modulated by TNF-related apoptosis-inducing ligand (TNFSF10). We showed that chronic treatment of 3xTg-AD mice with an anti-TNFSF10 monoclonal antibody was able to inhibit the retinal expression of miR-155, which inversely correlated with the expression of its molecular target SOCS-1. Moreover, the fine-tuned mechanism related to TNFSF10 immunoneutralization was tightly linked to modulation of TNFSF10 itself and its death receptor TNFRSF10B, along with cytokine production by microglia, reactive gliosis, and specific AD-related neuropathological hallmarks (i.e., Aß deposition and Tau phosphorylation) in the retina of 3xTg-AD mice. In conclusion, immunoneutralization of TNFSF10 significantly preserved the retinal tissue in 3xTg-AD mice, suggesting its potential therapeutic application in retinal degenerative disorders.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Inflammation/pathology , MicroRNAs/metabolism , Retina/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Base Sequence , Calcium-Binding Proteins/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Glial Fibrillary Acidic Protein/metabolism , Gliosis/complications , Gliosis/pathology , Inflammation/complications , Inflammation/genetics , Interleukin-10/metabolism , Mice, Transgenic , MicroRNAs/genetics , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Phosphorylation/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Signal Transduction/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism , tau Proteins/metabolism
11.
Cells ; 10(8)2021 08 05.
Article in English | MEDLINE | ID: mdl-34440761

ABSTRACT

Neurodegenerative diseases are characterized by the progressive loss of structure and/or function of both neurons and glial cells, leading to different degrees of pathology and loss of cognition. The hypothesis of circuit reconstruction in the damaged brain via direct cell replacement has been pursued extensively so far. In this context, stem cells represent a useful option since they provide tissue restoration through the substitution of damaged neuronal cells with exogenous stem cells and create a neuro-protective environment through the release of bioactive molecules for healthy neurons, as well. These peculiar properties of stem cells are opening to potential therapeutic strategies for the treatment of severe neurodegenerative disorders, for which the absence of effective treatment options leads to an increasingly socio-economic burden. Currently, the introduction of new technologies in the field of stem cells and the implementation of alternative cell tissues sources are pointing to exciting frontiers in this area of research. Here, we provide an update of the current knowledge about source and administration routes of stem cells, and review light and shadows of cells replacement therapy for the treatment of the three main neurodegenerative disorders (Amyotrophic lateral sclerosis, Parkinson's, and Alzheimer's disease).


Subject(s)
Central Nervous System/physiopathology , Nerve Degeneration , Nerve Regeneration , Neural Stem Cells/transplantation , Neurodegenerative Diseases/surgery , Stem Cell Transplantation , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Alzheimer Disease/surgery , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Amyotrophic Lateral Sclerosis/surgery , Animals , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/pathology , Humans , Neural Stem Cells/immunology , Neural Stem Cells/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Neuroimmunomodulation , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Parkinson Disease/surgery , Phenotype , Recovery of Function , Stem Cell Transplantation/adverse effects
12.
Curr Alzheimer Res ; 18(4): 298-309, 2021.
Article in English | MEDLINE | ID: mdl-34102970

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the most common form of neurodegenerative disorder characterized by cognitive impairment, which represents an urgent public health concern. Given the worldwide impact of AD, there is a compelling need for effective therapies to slow down or halt this disorder. OBJECTIVE: Choline alphoscerate (α-GPC) represents a potentially effective cholinergic neurotransmission enhancing agent with an interesting clinical profile in cognitive dysfunctions improvement, although only scanty data are available about the mechanisms underlying such beneficial effects. METHODS: The SH-SY5Y neuronal cell line, differentiated for 1 week with 10 µm of all-trans-retinoic acid (RA), to achieve a switch towards a cholinergic phenotype, was used as an in vitro model of AD. SH-SY5Y cells were pre-treated for 1h with α-GPC (100nM) and treated for 72 h with Aß25-35 (10µM). RESULTS: α-GPC was able to antagonize Aß25-35 mediated neurotoxicity and attenuate the Aß-induced phosphorylation of the Tau protein. Moreover, α-GPC exerted its beneficial effects by employing the NGF/TrkA system, knocked down in AD and, consequently, by sustaining the expression level of synaptic vesicle proteins, such as synaptophysin. CONCLUSION: Taken together, our data suggest that α-GPC can have a role in neuroprotection in the course of toxic challenges with Aß. Thus, a deeper understanding of the mechanism underlying its beneficial effect, could provide new insights into potential future pharmacological applications of its functional cholinergic enhancement, with the aim to mitigate AD and could represent the basis for innovative therapy.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/toxicity , Cell Differentiation/drug effects , Glycerylphosphorylcholine/pharmacology , Neurons/drug effects , Peptide Fragments/toxicity , tau Proteins/metabolism , Animals , Humans , In Vitro Techniques , Mice , Phosphorylation
13.
Rev Endocr Metab Disord ; 22(4): 1013-1025, 2021 12.
Article in English | MEDLINE | ID: mdl-33945051

ABSTRACT

Obesity is a major public health problem worldwide. Only relatively few treatment options are, at present, available for the management of obese patients. Furthermore, treatment of obesity is affected by the widespread misuse of drugs and food supplements. Ephedra sinica is an old medicinal herb, commonly used in the treatment of respiratory tract diseases. Ephedra species contain several alkaloids, including pseudoephedrine, notably endowed with indirect sympathomimetic pharmacodynamic properties. The anorexigenic effect of pseudoephedrine is attributable primarily to the inhibition of neurons located in the hypothalamic paraventricular nucleus (PVN), mediating satiety stimuli. Pseudoephedrine influences lipolysis and thermogenesis through interaction with ß3 adrenergic receptors and reduces fat accumulation through down-regulation of transcription factors related to lipogenesis. However, its use is associated with adverse events that involve to a large extent the cardiovascular and the central nervous system. Adverse events of pseudoephedrine also affect the eye, the intestine, and the skin, and, of relevance, sudden cardiovascular death related to dietary supplements containing Ephedra alkaloids has also been reported. In light of the limited availability of clinical data on pseudoephedrine in obesity, along with its significantly unbalanced risk/benefit profile, as well as of the psychophysical susceptibility of obese patients, it appears reasonable to preclude the prescription of pseudoephedrine in obese patients of any order and degree.


Subject(s)
Alkaloids , Ephedra sinica , Ephedrine/adverse effects , Humans , Obesity/chemically induced , Obesity/drug therapy , Pseudoephedrine/therapeutic use
14.
Cancers (Basel) ; 13(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33921021

ABSTRACT

Since the start of the global spread of coronavirus disease (COVID-19) pandemic, cancer patients were identified as a specifically susceptible subgroup of the patient population. Several reports have shown that cancer patients have an increased risk of both contracting the infection and of experiencing a more severe disease course, with a rapidly evolving picture associated with higher mortality. The assumption of cancer patients as "COVID-19 vulnerable" has led, irretrievably, to profound changes in the decision making of oncological treatments. Potential justifications for such concerns encompass the cancer-dependent suppression of the immune response, as well as the influence of administration of systemic anticancer treatments, including chemotherapy and immunotherapy. Nevertheless, to date, it is not clear whether the use of immune checkpoint inhibitors (ICIs) in cancer patients is safe, given their modulating effects on the immune system, or that they may rather conceal detrimental consequences. Theoretically, on the one hand, ICIs may enhance the immunological control of viral infections through their immunostimulating mechanisms; on the other hand, they could contribute to the hyper-inflammatory phase of COVID-19, worsening its clinical outcomes. In this study, we report the foremost clinical observations on the safety of ICI administration in cancer patients affected by COVID-19.

15.
Front Neurosci ; 14: 614643, 2020.
Article in English | MEDLINE | ID: mdl-33343293

ABSTRACT

Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder characterized by cognitive decline and by the presence of amyloid ß plaques and neurofibrillary tangles in the brain. Despite recent advances in understanding its pathophysiological mechanisms, to date, there are no disease-modifying therapeutic options, to slow or halt the evolution of neurodegenerative processes in AD. Current pharmacological treatments only transiently mitigate the severity of symptoms, with modest or null overall improvement. Emerging evidence supports the concept that AD is affected by the impaired ability of the immune system to restrain the brain's pathology. Deep understanding of the relationship between the nervous and the immune system may provide a novel arena to develop effective and safe drugs for AD treatment. Considering the crucial role of inflammatory/immune pathways in AD, here we discuss the current status of the immuno-oncological, immunomodulatory and anti-TNF-α drugs which are being used in preclinical studies or in ongoing clinical trials by means of the drug-repositioning approach.

16.
J Neuroinflammation ; 17(1): 298, 2020 Oct 13.
Article in English | MEDLINE | ID: mdl-33050925

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive degeneration and loss of neurons in specific regions of the central nervous system. Chronic activation of the immune cells resident in the brain, peripheral immune cell trafficking across the blood-brain barrier, and release of inflammatory and neurotoxic factors, appear critical contributors of the neuroinflammatory response that drives the progression of neurodegenerative processes in AD. As the neuro-immune network is impaired in course of AD, this review is aimed to point out the essential supportive role of innate and adaptive immune response either in normal brain as well as in brain recovery from injury. Since a fine-tuning of the immune response appears crucial to ensure proper nervous system functioning, we focused on the role of the TNF superfamily member, TNF-related apoptosis-inducing ligand (TRAIL), which modulates both the innate and adaptive immune response in the pathogenesis of several immunological disorders and, in particular, in AD-related neuroinflammation. We here summarized mounting evidence of potential involvement of TRAIL signaling in AD pathogenesis, with the aim to provide clearer insights about potential novel therapeutic approaches in AD.


Subject(s)
Alzheimer Disease/immunology , Brain/immunology , Immunity, Cellular/physiology , TNF-Related Apoptosis-Inducing Ligand/immunology , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Humans , TNF-Related Apoptosis-Inducing Ligand/metabolism
18.
Pharmacol Res ; 130: 374-384, 2018 04.
Article in English | MEDLINE | ID: mdl-29438781

ABSTRACT

In the last several years a large number of studies have demonstrated the neurobiological and clinical continuum between depression and Alzheimer's disease (AD). Depression is a risk factor for the development of AD, and the presence of depressive symptoms significantly increases the conversion of Mild Cognitive Impairment (MCI) into AD. Common pathophysiological events have been identified in depression and AD, including neuroinflammation with an aberrant Tumor Necrosis Factor-α (TNF-α) signaling, and an impairment of Brain-Derived Neurotrophic Factor (BDNF) and Transforming-Growth-Factor-ß1 (TGF-ß1) signaling. TGF-ß1 is an anti-inflammatory cytokine that exerts neuroprotective effects against amyloid-ß (Aß)-induced neurodegeneration, and it has a key role in memory formation and synaptic plasticity. TGF-ß1 plasma levels are reduced in major depressed patients (MDD), correlate with depression severity, and significantly contribute to treatment resistance in MDD. The deficit of Smad-dependent TGF-ß1 signaling is also an early event in AD pathogenesis, which contributes to inflammaging and cognitive decline in AD. A long-term treatment with antidepressants such as selective-serotonin-reuptake inhibitors (SSRIs) is known to reduce the risk of AD in patients with depression and, SSRIs, such as fluoxetine, increase the release of TGF-ß1 from astrocytes and exert relevant neuroprotective effects in experimental models of AD. We propose the TGF-ß1 signaling pathway as a common pharmacological target in depression and AD, and discuss the potential rescue of TGF-ß1 signaling by antidepressants as a way to prevent the transition from depression to AD.


Subject(s)
Alzheimer Disease/drug therapy , Depression/drug therapy , Transforming Growth Factor beta1/metabolism , Alzheimer Disease/epidemiology , Alzheimer Disease/metabolism , Animals , Cognitive Dysfunction/metabolism , Depression/epidemiology , Depression/metabolism , Humans , Signal Transduction
19.
Front Pharmacol ; 7: 389, 2016.
Article in English | MEDLINE | ID: mdl-27826242

ABSTRACT

Selective reuptake inhibitors (SSRIs), such as fluoxetine and sertraline, increase circulating Transforming-Growth-Factor-ß1 (TGF-ß1) levels in depressed patients, and are currently studied for their neuroprotective properties in Alzheimer's disease. TGF-ß1 is an anti-inflammatory cytokine that exerts neuroprotective effects against ß-amyloid (Aß)-induced neurodegeneration. In the present work, the SSRI, fluoxetine, was tested for the ability to protect cortical neurons against 1 µM oligomeric Aß1-42-induced toxicity. At therapeutic concentrations (100 nM-1 µM), fluoxetine significantly prevented Aß-induced toxicity in mixed glia-neuronal cultures, but not in pure neuronal cultures. Though to a lesser extent, also sertraline was neuroprotective in mixed cultures, whereas serotonin (10 nM-10 µM) did not mimick fluoxetine effects. Glia-conditioned medium collected from astrocytes challenged with fluoxetine protected pure cortical neurons against Aß toxicity. The effect was lost in the presence of a neutralizing antibody against TGF-ß1 in the conditioned medium, or when the specific inhibitor of type-1 TGF-ß1 receptor, SB431542, was added to pure neuronal cultures. Accordingly, a 24 h treatment of cortical astrocytes with fluoxetine promoted the release of active TGF-ß1 in the culture media through the conversion of latent TGF-ß1 to mature TGF-ß1. Unlike fluoxetine, both serotonin and sertraline did not stimulate the astrocyte release of active TGF-ß1. We conclude that fluoxetine is neuroprotective against Aß toxicity via a paracrine signaling mediated by TGF-ß1, which does not result from a simplistic SERT blockade.

SELECTION OF CITATIONS
SEARCH DETAIL
...