Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Int J Mol Sci ; 24(23)2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38068900

ABSTRACT

S100B, a homodimeric Ca2+-binding protein, is produced and secreted by astrocytes, and its extracellular levels have been used as a glial marker in brain damage and neurodegenerative and psychiatric diseases; however, its mechanism of secretion is elusive. We used primary astrocyte cultures and calcium measurements from real-time fluorescence microscopy to investigate the role of intracellular calcium in S100B secretion. In addition, the dimethyl sulfoxide (DMSO) effect on S100B was investigated in vitro and in vivo using Wistar rats. We found that DMSO, a widely used vehicle in biological assays, is a powerful S100B secretagogue, which caused a biphasic response of Ca2+ mobilization. Our data show that astroglial S100B secretion is triggered by the increase in intracellular Ca2+ and indicate that this increase is due to Ca2+ mobilization from the endoplasmic reticulum. Also, blocking plasma membrane Ca2+ channels involved in the Ca2+ replenishment of internal stores decreased S100B secretion. The DMSO-induced S100B secretion was confirmed in vivo and in ex vivo hippocampal slices. Our data support a nonclassic vesicular export of S100B modulated by Ca2+, and the results might contribute to understanding the mechanism underlying the astroglial release of S100B.


Subject(s)
Astrocytes , Dimethyl Sulfoxide , Rats , Animals , Rats, Wistar , Dimethyl Sulfoxide/pharmacology , Dimethyl Sulfoxide/metabolism , Astrocytes/metabolism , Colforsin/pharmacology , Secretagogues/pharmacology , Calcium/metabolism , Nerve Growth Factors/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Endoplasmic Reticulum/metabolism , Cells, Cultured
2.
Biomolecules ; 11(6)2021 06 12.
Article in English | MEDLINE | ID: mdl-34204735

ABSTRACT

The receptor for advanced glycation-end products (RAGE) is a multiligand receptor with a role in inflammatory and pulmonary pathologies. Hyperactivation of RAGE by its ligands has been reported to sustain inflammation and oxidative stress in common comorbidities of severe COVID-19. RAGE is essential to the deleterious effects of the renin-angiotensin system (RAS), which participates in infection and multiorgan injury in COVID-19 patients. Thus, RAGE might be a major player in severe COVID-19, and appears to be a useful therapeutic molecular target in infections by SARS-CoV-2. The role of RAGE gene polymorphisms in predisposing patients to severe COVID-19 is discussed. .


Subject(s)
COVID-19/metabolism , Inflammation/metabolism , Oxidative Stress , Receptor for Advanced Glycation End Products/metabolism , Renin-Angiotensin System , Animals , COVID-19/genetics , COVID-19/pathology , Humans , Inflammation/genetics , Inflammation/pathology , Polymorphism, Genetic , Receptor for Advanced Glycation End Products/genetics , Risk Factors , SARS-CoV-2/physiology , Severity of Illness Index
3.
Life Sci ; 272: 119251, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33636175

ABSTRACT

A novel infectious disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was detected in December 2019 and declared as a global pandemic by the World Health. Approximately 15% of patients with COVID-19 progress to severe pneumonia and eventually develop acute respiratory distress syndrome (ARDS), septic shock and/or multiple organ failure with high morbidity and mortality. Evidence points towards a determinant pathogenic role of members of the renin-angiotensin system (RAS) in mediating the susceptibility, infection, inflammatory response and parenchymal injury in lungs and other organs of COVID-19 patients. The receptor for advanced glycation end-products (RAGE), a member of the immunoglobulin superfamily, has important roles in pulmonary pathological states, including fibrosis, pneumonia and ARDS. RAGE overexpression/hyperactivation is essential to the deleterious effects of RAS in several pathological processes, including hypertension, chronic kidney and cardiovascular diseases, and diabetes, all of which are major comorbidities of SARS-CoV-2 infection. We propose RAGE as an additional molecular target in COVID-19 patients for ameliorating the multi-organ pathology induced by the virus and improving survival, also in the perspective of future infections by other coronaviruses.


Subject(s)
COVID-19/complications , Drug Discovery , Multiple Organ Failure/etiology , Multiple Organ Failure/prevention & control , Receptor for Advanced Glycation End Products/antagonists & inhibitors , SARS-CoV-2/physiology , Animals , COVID-19/metabolism , COVID-19/pathology , Humans , Molecular Targeted Therapy , Multiple Organ Failure/metabolism , Multiple Organ Failure/pathology , Receptor for Advanced Glycation End Products/metabolism , Renin-Angiotensin System/drug effects , SARS-CoV-2/drug effects , Signal Transduction/drug effects , COVID-19 Drug Treatment
4.
J Cachexia Sarcopenia Muscle ; 11(4): 929-946, 2020 08.
Article in English | MEDLINE | ID: mdl-32159297

ABSTRACT

BACKGROUND: Cachexia, a multifactorial syndrome affecting more than 50% of patients with advanced cancer and responsible for ~20% of cancer-associated deaths, is still a poorly understood process without a standard cure available. Skeletal muscle atrophy caused by systemic inflammation is a major clinical feature of cachexia, leading to weight loss, dampening patients' quality of life, and reducing patients' response to anticancer therapy. RAGE (receptor for advanced glycation end-products) is a multiligand receptor of the immunoglobulin superfamily and a mediator of muscle regeneration, inflammation, and cancer. METHODS: By using murine models consisting in the injection of colon 26 murine adenocarcinoma (C26-ADK) or Lewis lung carcinoma (LLC) cells in BALB/c and C57BL/6 or Ager-/- (RAGE-null) mice, respectively, we investigated the involvement of RAGE signalling in the main features of cancer cachexia, including the inflammatory state. In vitro experiments were performed using myotubes derived from C2C12 myoblasts or primary myoblasts isolated from C57BL/6 wild type and Ager-/- mice treated with the RAGE ligand, S100B (S100 calcium-binding protein B), TNF (tumor necrosis factor)α±IFN (interferon) γ, and tumour cell- or masses-conditioned media to analyse hallmarks of muscle atrophy. Finally, muscles of wild type and Ager-/- mice were injected with TNFα/IFNγ or S100B in a tumour-free environment. RESULTS: We demonstrate that RAGE is determinant to activate signalling pathways leading to muscle protein degradation in the presence of proinflammatory cytokines and/or tumour-derived cachexia-inducing factors. We identify the RAGE ligand, S100B, as a novel factor able to induce muscle atrophy per se via a p38 MAPK (p38 mitogen-activated protein kinase)/myogenin axis and STAT3 (signal transducer and activator of transcription 3)-dependent MyoD (myoblast determination protein 1) degradation. Lastly, we found that in cancer conditions, an increase in serum levels of tumour-derived S100B and HMGB1 (high mobility group box 1) occurs leading to chronic activation/overexpression of RAGE, which induces hallmarks of cancer cachexia (i.e. muscle wasting, systemic inflammation, and release of tumour-derived pro-cachectic factors). Absence of RAGE in mice translates into reduced serum levels of cachexia-inducing factors, delayed loss of muscle mass and strength, reduced tumour progression, and increased survival. CONCLUSIONS: RAGE is a molecular determinant in inducing the hallmarks of cancer cachexia, and molecular targeting of RAGE might represent a therapeutic strategy to prevent or counteract the cachectic syndrome.


Subject(s)
Cachexia/prevention & control , Neoplasms/complications , Receptor for Advanced Glycation End Products/physiology , Animals , Humans , Mice , Neoplasms/physiopathology
5.
Cell Mol Life Sci ; 77(18): 3547-3565, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32072237

ABSTRACT

Reductive stress is defined as a condition of sustained increase in cellular glutathione/glutathione disulfide and NADH/NAD+ ratios. Reductive stress is emerging as an important pathophysiological event in several diseased states, being as detrimental as is oxidative stress. Occurrence of reductive stress has been documented in several cardiomyopathies and is an important pathophysiological factor particularly in coronary artery disease and myocardial infarction. Excess activation of the transcription factor, Nrf2-the master regulator of the antioxidant response-, consequent in most cases to defective autophagy, can lead to reductive stress. In addition, hyperglycemia-induced activation of the polyol pathway can lead to increased NADH/NAD+ ratio, which might translate into increased levels of hydrogen sulfide-via enhanced activity of cystathionine ß-synthase-that would fuel reductive stress through inhibition of mitochondrial complex I. Reductive stress may be either a potential weapon against cancer priming tumor cells to apoptosis or a cancer's ally promoting tumor cell proliferation and making tumor cells resistant to reactive oxygen species-inducing drugs. In non-cancer pathological states reductive stress is definitely harmful paradoxically leading to reactive oxygen species overproduction via excess NADPH oxidase 4 activity. In face of the documented occurrence of reductive stress in several heart diseases, there is much less information about the occurrence and effects of reductive stress in skeletal muscle tissue. In the present review we describe relevant results emerged from studies of reductive stress in the heart and review skeletal muscle conditions in which reductive stress has been experimentally documented and those in which reductive stress might have an as yet unrecognized pathophysiological role. Establishing whether reductive stress has a (patho)physiological role in skeletal muscle will hopefully contribute to answer the question whether antioxidant supplementation to the general population, athletes, and a large cohort of patients (e.g. heart, sarcopenic, dystrophic, myopathic, cancer, and bronco-pulmonary patients) is harmless or detrimental.


Subject(s)
Muscle Cells/metabolism , Oxidative Stress , Antioxidants/pharmacology , Autophagy , Glutathione/metabolism , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism
6.
Cell Mol Life Sci ; 77(1): 129-147, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31363816

ABSTRACT

Obesity is an endemic pathophysiological condition and a comorbidity associated with hypercholesterolemia, hypertension, cardiovascular disease, type 2 diabetes mellitus, and cancer. The adipose tissue of obese subjects shows hypertrophic adipocytes, adipocyte hyperplasia, and chronic low-grade inflammation. S100 proteins are Ca2+-binding proteins exclusively expressed in vertebrates in a cell-specific manner. They have been implicated in the regulation of a variety of functions acting as intracellular Ca2+ sensors transducing the Ca2+ signal and extracellular factors affecting cellular activity via ligation of a battery of membrane receptors. Certain S100 proteins, namely S100A4, the S100A8/S100A9 heterodimer and S100B, have been implicated in the pathophysiology of obesity-promoting macrophage-based inflammation via toll-like receptor 4 and/or receptor for advanced glycation end-products ligation. Also, serum levels of S100A4, S100A8/S100A9, S100A12, and S100B correlate with insulin resistance/type 2 diabetes, metabolic risk score, and fat cell size. Yet, secreted S100B appears to exert neurotrophic effects on sympathetic fibers in brown adipose tissue contributing to the larger sympathetic innervation of this latter relative to white adipose tissue. In the present review we first briefly introduce S100 proteins and then critically examine their role(s) in adipose tissue and obesity.


Subject(s)
Adipose Tissue/metabolism , Obesity/metabolism , S100 Proteins/metabolism , Adipose Tissue/physiopathology , Animals , Cytokines/analysis , Cytokines/metabolism , Humans , Inflammation/complications , Inflammation/metabolism , Inflammation/physiopathology , Macrophages/metabolism , Macrophages/pathology , Obesity/complications , Obesity/physiopathology , S100 Proteins/analysis
7.
Int J Dev Neurosci ; 77: 26-38, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31026497

ABSTRACT

The presence of immune cells in the central nervous system has long been the subject of research to find out their role. For a long time it was believed that the CNS was a privileged area from an immunological point of view, due to the presence of the blood-brain barrier (BBB), as circulating immune cells were unable to penetrate the brain parenchyma, at least until the integrity of the BBB was preserved. For this reason the study of the CNS immune system has focused on the functions of microglia, the immunocompetent resident element of the brain parenchyma that retain the ability to divide and self-renew during lifespan without any significant contribution from circulating blood cells. More recently, the presence of lymphatic vessels in the dural sinuses has been demonstrated with accompanying lymphocytes, monocytes and other immune cells. Moreover, meningeal macrophages, that is macrophages along the blood vessels and in the choroid plexus (CP), are also present. These non-parenchymal immune cells, together with microglia, can affect multiple CNS functions. Here, we discuss the functional role of parenchymal and non-parenchymal immune cells and their contribution to the regulation of neurogenesis.


Subject(s)
Brain/physiology , Macrophages/physiology , Mast Cells/physiology , Microglia/physiology , Neurogenesis/physiology , Animals , Brain/cytology , Brain/immunology , Humans
8.
Cell Prolif ; 52(3): e12599, 2019 May.
Article in English | MEDLINE | ID: mdl-30912260

ABSTRACT

Sertoli cells (SeC) are responsible for the immunoprivileged status of the testis thanks to which allogeneic or xenogeneic engraftments can survive without pharmacological immune suppression if co-injected with SeC. This peculiar ability of SeC is dependent on secretion of a plethora of factors including maturation factors, hormones, growth factors, cytokines and immunomodulatory factors. The anti-inflammatory and trophic properties of SeC have been largely exploited in several experimental models of diseases, diabetes being the most studied. Duchenne muscular dystrophy (DMD) is a lethal X-linked recessive pathology in which lack of functional dystrophin leads to progressive muscle degeneration culminating in loss of locomotion and premature death. Despite a huge effort to find a cure, DMD patients are currently treated with anti-inflammatory steroids. Recently, encapsulated porcine SeC (MC-SeC) have been injected ip in the absence of immunosuppression in an animal model of DMD resulting in reduction of muscle inflammation and amelioration of muscle morphology and functionality, thus opening an additional avenue in the treatment of DMD. The novel protocol is endowed with the advantage of being potentially applicable to all the cohort of DMD patients regardless of the mutation. This mini-review addresses several issues linked to the possible use of MC-SeC injected ip in dystrophic people.


Subject(s)
Cell Transplantation/methods , Muscular Dystrophy, Duchenne/therapy , Sertoli Cells/transplantation , Animals , Disease Models, Animal , Heterografts , Humans , Immune Privilege , Injections, Intraperitoneal , Male , Mice , Mice, Inbred mdx , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Sertoli Cells/immunology , Swine , Transplantation Immunology
9.
J Cachexia Sarcopenia Muscle ; 9(7): 1255-1268, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30499235

ABSTRACT

Primary sarcopenia is a condition of reduced skeletal muscle mass and strength, reduced agility, and increased fatigability and risk of bone fractures characteristic of aged, otherwise healthy people. The pathogenesis of primary sarcopenia is not completely understood. Herein, we review the essentials of the cellular and molecular mechanisms of skeletal mass maintenance; the alterations of myofiber metabolism and deranged properties of muscle satellite cells (the adult stem cells of skeletal muscles) that underpin the pathophysiology of primary sarcopenia; the role of the Ca2+ -sensor protein, S100B, as an intracellular factor and an extracellular signal regulating cell functions; and the functional role of S100B in muscle tissue. Lastly, building on recent results pointing to S100B as to a molecular determinant of myoblast-brown adipocyte transition, we propose S100B as a transducer of the deleterious effects of accumulation of reactive oxygen species in myoblasts and, potentially, myofibers concurring to the pathophysiology of sarcopenia.


Subject(s)
Muscle, Skeletal/metabolism , Sarcopenia/etiology , Sarcopenia/metabolism , Biomarkers , Humans , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/pathology , S100 Calcium Binding Protein beta Subunit/genetics , S100 Calcium Binding Protein beta Subunit/metabolism , Sarcopenia/pathology , Satellite Cells, Skeletal Muscle/metabolism
10.
J Cachexia Sarcopenia Muscle ; 9(7): 1213-1234, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30334619

ABSTRACT

Emerging evidence suggests that the signalling of the Receptor for Advanced Glycation End products (RAGE) is critical for skeletal muscle physiology controlling both the activity of muscle precursors during skeletal muscle development and the correct time of muscle regeneration after acute injury. On the other hand, the aberrant re-expression/activity of RAGE in adult skeletal muscle is a hallmark of muscle wasting that occurs in response to ageing, genetic disorders, inflammatory conditions, cancer, and metabolic alterations. In this review, we discuss the mechanisms of action and the ligands of RAGE involved in myoblast differentiation, muscle regeneration, and muscle pathological conditions. We highlight potential therapeutic strategies for targeting RAGE to improve skeletal muscle function.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Diseases/etiology , Muscular Diseases/metabolism , Receptor for Advanced Glycation End Products/metabolism , Animals , Gene Expression Regulation , Humans , Ligands , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Diseases/pathology , Protein Isoforms , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Receptor for Advanced Glycation End Products/chemistry , Receptor for Advanced Glycation End Products/genetics , Rhabdomyosarcoma/etiology , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Signal Transduction
11.
Nanomaterials (Basel) ; 8(11)2018 Oct 23.
Article in English | MEDLINE | ID: mdl-30360511

ABSTRACT

The synthesis of ultrasmall UiO-66 nanoparticles (NPs) with an average size of 25 nm, determined by X-ray powder diffraction and electron microscopies analysis, is reported. The NPs were stabilized in water by dialyzing the NP from the DMF used for the synthesis. DLS measurements confirmed the presence of particles of 100 nm, which are spherical aggregates of smaller particles of 20⁻30 nm size. The NP have a BET surface area of 700 m²/g with an external surface area of 300 m²/g. UiO-66_N (UiO-66 nanoparticles) were loaded with acridine orange as fluorescent probe. UV-vis spectroscopy analysis revealed no acridine loss after 48 h of agitation in simulated body fluid. The biocompatibility of UiO-66_N was evaluated in human glioblastoma (GBM) cell line U251, the most malignant (IV grade of WHO classification) among brain tumors. In U251 cells, UiO-66_N are inert since they do not alter the cell cycle, the viability, migration properties, and the expression of kinases involved in cancer cell growth. The internalization process was evident after a few hours of incubation. After 24 h, UiO-66_N@Acr (UiO-66_N loaded with acridine orange) were detectable around the nuclei of the cells. These data suggest that small UiO-66 are biocompatible NP and could represent a potential carrier for drug delivery in glioblastoma therapies.

12.
Hum Mol Genet ; 27(21): 3734-3746, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30085099

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal X-linked disease affecting striated muscles, which undergo progressive degeneration and chronic inflammation. Receptor for advanced glycation end-products (RAGE), a multiligand receptor involved in myogenesis and inflammation, is absent in healthy adult muscles but is re-expressed in myoblasts, regenerating myofibers and activated immune cells upon acute muscle injury, and in certain myopathies. We show here that RAGE is expressed and chronically stimulated in muscles of mdx mice, an experimental model of DMD, which also release high amounts of the RAGE ligands, HMGB1 and S100B. We generated a double mutant, mdx/Ager-/- mouse lacking dystrophin and RAGE. Compared to mdx mice, muscles of mdx/Ager-/- mice show restrained inflammation, unaffected fibrosis and higher muscle strength. Mdx/Ager-/- macrophages are less responsive to proinflammatory stimuli and express lower levels of Ccr2, Ccl2 and Ccl7, which are involved in monocyte/macrophage chemotaxis and migration. In vivo treatment of dystrophic muscles with a RAGE blocking antibody results in reduced necrosis and inflammatory infiltrate. Our results suggest that RAGE sustains muscle inflammation and necrosis in DMD muscles and that reducing RAGE activity might represent a potential therapeutic tool to counteract muscle inflammation and rescue muscle morphology in DMD conditions.


Subject(s)
Inflammation , Muscle Strength , Muscular Dystrophy, Duchenne/metabolism , Receptor for Advanced Glycation End Products/physiology , Animals , Disease Models, Animal , Dystrophin/genetics , Fibrosis , Male , Mice , Mice, Inbred mdx , Mice, Knockout , Muscular Dystrophy, Duchenne/physiopathology , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism
13.
Front Cell Neurosci ; 12: 99, 2018.
Article in English | MEDLINE | ID: mdl-29692710

ABSTRACT

Glioblastoma multiforme (GBM) is the most malignant brain tumor and is associated with poor prognosis due to its thorny localization, lack of efficacious therapies and complex biology. Among the numerous pathways driving GBM biology studied so far, PTEN/phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT/mechanistic target of rapamycin (mTOR) signaling plays a pivotal role, as it controls cell survival, proliferation and metabolism and is involved in stem cell maintenance. In front of recent and numerous evidences highlighting mTOR upregulation in GBM, all the strategies developed to inhibit this pathway have been substantially unsuccessful. Our study focused on mTOR complex 2 (mTORC2) to understand its involvement in GBM cell growth, proliferation, migration and invasiveness. We utilized an in vitro model, characterized by various genetic alterations (i.e., GL15, U257, U87MG and U118MG cell lines) in order to achieve the clonal heterogeneity observed in vivo. Additionally, being the U87MG cell line endowed with glioblastoma stem cells (GSCs), we also investigated the role of the PTEN/PI3K/AKT/mTOR pathway in this specific cell population, which is responsible for GBM relapse. We provide further insights that explain the reasons for the failure of numerous clinical trials conducted to date targeting PI3K or mTOR complex 1 (mTORC1) with rapamycin and its analogs. Additionally, we show that mTORC2 might represent a potential clinically valuable target for GBM treatment, as proliferation, migration and GSC maintenance appear to be mTORC2-dependent. In this context, we demonstrate that the novel ATP-competitive mTOR inhibitor PP242 effectively targets both mTORC1 and mTORC2 activation and counteracts cell proliferation via the induction of high autophagy levels, besides reducing cell migration, invasiveness and stemness properties.

14.
Biochim Biophys Acta Mol Cell Res ; 1865(5): 721-733, 2018 May.
Article in English | MEDLINE | ID: mdl-29499228

ABSTRACT

Nrf2 and its endogenous inhibitor, Keap1, function as a ubiquitous, evolutionarily conserved intracellular defense mechanism to counteract oxidative stress. Sequestered by cytoplasmic Keap1 and targeted to proteasomal degradation in basal conditions, in case of oxidative stress Nrf2 detaches from Keap1 and translocates to the nucleus, where it heterodimerizes with one of the small Maf proteins. The heterodimers recognize the AREs, that are enhancer sequences present in the regulatory regions of Nrf2 target genes, essential for the recruitment of key factors for transcription. In the present review we briefly introduce the Nrf2-Keap1 system and describe Nrf2 functions, illustrate the Nrf2-NF-κB cross-talk, and highlight the effects of the Nrf2-Keap1 system in the physiology and pathophysiology of striated muscle tissue taking into account its role(s) in oxidative stress and reductive stress.


Subject(s)
Kelch-Like ECH-Associated Protein 1/genetics , NF-E2-Related Factor 2/genetics , Oxidative Stress/genetics , Cell Nucleus/genetics , Humans , Maf Transcription Factors/genetics , NF-kappa B/genetics , Oxidation-Reduction , Signal Transduction
15.
Int J Mol Sci ; 19(1)2018 Jan 22.
Article in English | MEDLINE | ID: mdl-29361745

ABSTRACT

Depending on the species, microglial cells represent 5-20% of glial cells in the adult brain. As the innate immune effector of the brain, microglia are involved in several functions: regulation of inflammation, synaptic connectivity, programmed cell death, wiring and circuitry formation, phagocytosis of cell debris, and synaptic pruning and sculpting of postnatal neural circuits. Moreover, microglia contribute to some neurodevelopmental disorders such as Nasu-Hakola disease (NHD), and to aged-associated neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and others. There is evidence that human and rodent microglia may become senescent. This event determines alterations in the microglia activation status, associated with a chronic inflammation phenotype and with the loss of neuroprotective functions that lead to a greater susceptibility to the neurodegenerative diseases of aging. In the central nervous system (CNS), Triggering Receptor Expressed on Myeloid Cells 2-DNAX activation protein 12 (TREM2-DAP12) is a signaling complex expressed exclusively in microglia. As a microglial surface receptor, TREM2 interacts with DAP12 to initiate signal transduction pathways that promote microglial cell activation, phagocytosis, and microglial cell survival. Defective TREM2-DAP12 functions play a central role in the pathogenesis of several diseases. The CX3CL1 (fractalkine)-CX3CR1 signaling represents the most important communication channel between neurons and microglia. The expression of CX3CL1 in neurons and of its receptor CX3CR1 in microglia determines a specific interaction, playing fundamental roles in the regulation of the maturation and function of these cells. Here, we review the role of the TREM2-DAP12 and CX3CL1-CX3CR1 axes in aged microglia and the involvement of these pathways in physiological CNS aging and in age-associated neurodegenerative diseases.


Subject(s)
Aging/physiology , Microglia/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain/metabolism , Brain/pathology , CX3C Chemokine Receptor 1/metabolism , Central Nervous System/metabolism , Central Nervous System/pathology , Chemokine CX3CL1/metabolism , Gene Expression Regulation , Humans , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Receptors, Immunologic/metabolism , Signal Transduction
16.
Dis Markers ; 2018: 9230479, 2018.
Article in English | MEDLINE | ID: mdl-30662577

ABSTRACT

The mechanistic target of rapamycin (mTOR) drives several physiologic and pathologic cellular processes and is frequently deregulated in different types of tumors, including glioblastoma (GBM). Despite recent advancements in understanding the molecular mechanisms involved in GBM biology, the survival rates of this tumor are still disappointing, primarily due to the lack of efficacious treatments. The phosphatase and tensin homolog (PTEN)/phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mTOR pathway has emerged as a crucial player in GBM development and progression. However, to date, all the attempts to target this pathway with PI3K, AKT, or mTORC1 inhibitors failed to improve the outcome of patients with GBM. Despite these discouraging results, recent evidence pointed out that the blockade of mTORC2 might provide a useful therapeutic strategy for GBM, with the potential to overcome the limitations that mTORC1 inhibitors have shown so far. In this review, we analyzed the rationale of targeting mTOR in GBM and the available preclinical and clinical evidence supporting the choice of this therapeutic approach, highlighting the different roles of mTORC1 and mTORC2 in GBM biology.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Evidence-Based Medicine , Glioblastoma/drug therapy , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Molecular Targeted Therapy , Signal Transduction/drug effects
17.
Sci Rep ; 7(1): 12537, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28970581

ABSTRACT

Regeneration of injured skeletal muscles relies on a tightly controlled chain of cellular and molecular events. We show that appropriate levels of S100B protein are required for timely muscle regeneration after acute injury. S100B released from damaged myofibers and infiltrating macrophages expands the myoblast population, attracts macrophages and promotes their polarization into M2 (pro-regenerative) phenotype, and modulates collagen deposition, by interacting with RAGE (receptor for advanced glycation end-products) or FGFR1 (fibroblast growth factor receptor 1) depending on the muscle repair phase and local conditions. However, persistence of high S100B levels compromises the regeneration process prolonging myoblast proliferation and macrophage infiltration, delaying M1/M2 macrophage transition, and promoting deposition of fibrotic tissue via RAGE engagement. Interestingly, S100B is released in high abundance from degenerating muscles of mdx mice, an animal model of Duchenne muscular dystrophy (DMD), and blocking S100B ameliorates histopathology. Thus, levels of S100B differentially affect skeletal muscle repair upon acute injury and in the context of muscular dystrophy, and S100B might be regarded as a potential molecular target in DMD.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophy, Duchenne/genetics , Regeneration/genetics , S100 Calcium Binding Protein beta Subunit/genetics , Animals , Disease Models, Animal , Humans , Macrophage Activation/genetics , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred mdx , Muscle Strength/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/physiopathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Receptor for Advanced Glycation End Products/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics
18.
Cell Death Differ ; 24(12): 2077-2088, 2017 12.
Article in English | MEDLINE | ID: mdl-28885620

ABSTRACT

Muscles of sarcopenic people show hypotrophic myofibers and infiltration with adipose and, at later stages, fibrotic tissue. The origin of infiltrating adipocytes resides in fibro-adipogenic precursors and nonmyogenic mesenchymal progenitor cells, and in satellite cells, the adult stem cells of skeletal muscles. Myoblasts and brown adipocytes share a common Myf5+ progenitor cell: the cell fate depends on levels of bone morphogenetic protein 7 (BMP-7), a TGF-ß family member. S100B, a Ca2+-binding protein of the EF-hand type, is expressed at relatively high levels in myoblasts from sarcopenic humans and exerts anti-myogenic effects via NF-κB-dependent inhibition of MyoD, a myogenic transcription factor acting upstream of the essential myogenic factor, myogenin. Adipogenesis requires high levels of ROS, and myoblasts of sarcopenic subjects show elevated ROS levels. Here we show that: (1) ROS overproduction in myoblasts results in upregulation of S100B levels via NF-κB activation; and (2) ROS/NF-κB-induced accumulation of S100B causes myoblast transition into brown adipocytes. S100B activates an NF-κB/Ying Yang 1 axis that negatively regulates the promyogenic and anti-adipogenic miR-133 with resultant accumulation of the brown adipogenic transcription regulator, PRDM-16. S100B also upregulates BMP-7 via NF-κB/Ying Yang 1 with resultant BMP-7 autocrine activity. Interestingly, myoblasts from sarcopenic humans show features of brown adipocytes. We also show that S100B levels and NF-κB activity are elevated in brown adipocytes obtained by culturing myoblasts in adipocyte differentiation medium and that S100B knockdown or NF-κB inhibition in myoblast-derived brown adipocytes reconverts them into fusion-competent myoblasts. At last, interstitial cells and, unexpectedly, a subpopulation of myofibers in muscles of geriatric but not young mice co-express S100B and the brown adipocyte marker, uncoupling protein-1. These results suggest that S100B is an important intracellular molecular signal regulating Myf5+ progenitor cell differentiation into fusion-competent myoblasts or brown adipocytes depending on its levels.


Subject(s)
Adipocytes, Brown/metabolism , MicroRNAs/metabolism , Myoblasts/metabolism , NF-kappa B/metabolism , Oxidative Stress/physiology , S100 Calcium Binding Protein beta Subunit/metabolism , Adipocytes, Brown/cytology , Animals , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Humans , Male , Mice , MicroRNAs/genetics , Myoblasts/cytology , Reactive Oxygen Species/metabolism , S100 Calcium Binding Protein beta Subunit/genetics , Transfection , YY1 Transcription Factor/metabolism
19.
Front Mol Neurosci ; 10: 191, 2017.
Article in English | MEDLINE | ID: mdl-28674485

ABSTRACT

In vertebrates, during an early wave of hematopoiesis in the yolk sac between embryonic day E7.0 and E9.0, cells of mesodermal leaflet addressed to macrophage lineage enter in developing central nervous system (CNS) and originate the developing native microglial cells. Depending on the species, microglial cells represent 5-20% of glial cells resident in adult brain. Here, we briefly discuss some canonical functions of the microglia, i.e., cytokine secretion and functional transition from M1 to M2 phenotype. In addition, we review studies on the non-canonical functions of microglia such as regulation of phagocytosis, synaptic pruning, and sculpting postnatal neural circuits. In this latter context the contribution of microglia to some neurodevelopmental disorders is now well established. Nasu-Hakola (NHD) disease is considered a primary microgliopathy with alterations of the DNAX activation protein 12 (DAP12)-Triggering receptor expressed on myeloid cells 2 (TREM-2) signaling and removal of macromolecules and apoptotic cells followed by secondary microglia activation. In Rett syndrome Mecp2-/- microglia shows a substantial impairment of phagocytic ability, although the role of microglia is not yet clear. In a mouse model of Tourette syndrome (TS), microglia abnormalities have also been described, and deficient microglia-mediated neuroprotection is obvious. Here we review the role of microglial cells in neurodevelopmental disorders without inflammation and on the complex role of microglia in developing CNS.

20.
Cell Mol Life Sci ; 74(15): 2749-2760, 2017 08.
Article in English | MEDLINE | ID: mdl-28417162

ABSTRACT

S100A6 protein belongs to the A group of the S100 protein family of Ca2+-binding proteins. It is expressed in a limited number of cell types in adult normal tissues and in several tumor cell types. As an intracellular protein, S100A6 has been implicated in the regulation of several cellular functions, such as proliferation, apoptosis, the cytoskeleton dynamics, and the cellular response to different stress factors. S100A6 can be secreted/released by certain cell types which points to extracellular effects of the protein. RAGE (receptor for advanced glycation endproducts) and integrin ß1 transduce some extracellular S100A6's effects. Dosage of serum S100A6 might aid in diagnosis in oncology.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Proliferation , Neoplasms/metabolism , S100 Proteins/metabolism , Animals , Apoptosis , Cell Cycle Proteins/blood , Cell Cycle Proteins/genetics , Cell Movement , Cytoskeleton/genetics , Cytoskeleton/metabolism , Gene Expression Regulation , Humans , Integrin beta1/metabolism , Neoplasms/blood , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Receptor for Advanced Glycation End Products/metabolism , S100 Calcium Binding Protein A6 , S100 Proteins/blood , S100 Proteins/genetics , Signal Transduction , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...