Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Toxicol In Vitro ; 75: 105192, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33984456

ABSTRACT

Targeted delivery aims to enhance cellular uptake and improve therapeutic outcome with higher disease specificity. The expression of transferrin receptor (TfR) is upregulated on tumor cells, which make the protein Tf and its receptor vastly relevant when applied to targeting strategies. Here, we proposed Tf-decorated pH-sensitive PLGA nanoparticles containing the chemosensitizer poloxamer as a carrier for doxorubicin delivery to tumor cells (Tf-DOX-PLGA-NPs), aiming at alleviating multidrug resistance (MDR). We performed a range of in vitro studies to assess whether targeted NPs have the ability to improve DOX antitumor potential on resistant NCI/ADR-RES cells. All evaluations of the Tf-decorated NPs were performed comparatively to the nontargeted counterparts, aiming to evidence the real role of NP surface functionalization, along with the benefits of pH-sensitivity and poloxamer, in the improvement of antiproliferative activity and reversal of MDR. Tf-DOX-PLGA-NPs induced higher number of apoptotic events and ROS generation, along with cell cycle arrest. Moreover, they were efficiently internalized by NCI/ADR-RES cells, increasing DOX intracellular accumulation, which supports the greater cell killing ability of these targeted NPs with respect to MDR cells. Altogether, these findings supported the effectiveness of the Tf-surface modification of DOX-PLGA-NPs for an improved antiproliferative activity. Therefore, our pH-responsive Tf-inspired NPs are a promising smart drug delivery system to overcome MDR effect at some extent, enhancing the efficacy of DOX antitumor therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Nanoparticles/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Transferrin/administration & dosage , Apoptosis/drug effects , Cell Cycle/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , HeLa Cells , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Neoplasms/drug therapy , Neoplasms/metabolism , Reactive Oxygen Species/metabolism
2.
Cytometry A ; 97(7): 662-667, 2020 07.
Article in English | MEDLINE | ID: mdl-32506725

ABSTRACT

SARS-CoV-2 pandemic and recurrent dengue epidemics in tropical countries have turned into a global health threat. While both virus-caused infections may only reveal light symptoms, they can also cause severe diseases. Here, we review the possible antibody-dependent enhancement (ADE) occurrence, known for dengue infections, when there is a second infection with a different virus strain. Consequently, preexisting antibodies do not neutralize infection, but enhance it, possibly by triggering Fcγ receptor-mediated virus uptake. No clinical data exist indicating such mechanism for SARS-CoV-2, but previous coronavirus infections or infection of SARS-CoV-2 convalescent with different SARS-CoV-2 strains could promote ADE, as experimentally shown for antibodies against the MERS-CoV or SARS-CoV spike S protein. © 2020 International Society for Advancement of Cytometry.


Subject(s)
Antibody-Dependent Enhancement/immunology , Betacoronavirus/immunology , Coinfection/immunology , Dengue Virus/immunology , Receptors, IgG/immunology , Spike Glycoprotein, Coronavirus/immunology , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Dengue/immunology , Dengue/pathology , Humans , Image Cytometry/methods , Middle East Respiratory Syndrome Coronavirus/immunology , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Severe acute respiratory syndrome-related coronavirus/immunology , SARS-CoV-2 , Virus Internalization
3.
Stem Cell Rev Rep ; 16(3): 434-440, 2020 06.
Article in English | MEDLINE | ID: mdl-32307653

ABSTRACT

The expressive number of deaths and confirmed cases of SARS-CoV-2 call for an urgent demand of effective and available drugs for COVID-19 treatment. CD147, a receptor on host cells, is a novel route for SARS-CoV-2 invasion. Thus, drugs that interfere in the spike protein/CD147 interaction or CD147 expression may inhibit viral invasion and dissemination among other cells, including in progenitor/stem cells. Studies suggest beneficial effects of azithromycin in reducing viral load of hospitalized patients, possibly interfering with ligand/CD147 receptor interactions; however, its possible effects on SARS-CoV-2 invasion has not yet been evaluated. In addition to the possible effect in invasion, azithromycin decreases the expression of some metalloproteinases (downstream to CD147), induces anti-viral responses in primary human bronchial epithelial infected with rhinovirus, decreasing viral replication and release. Moreover, resident lung progenitor/stem are extensively differentiated into myofibroblasts during pulmonary fibrosis, a complication observed in COVID-19 patients. This process, and the possible direct viral invasion of progenitor/stem cells via CD147 or ACE2, could result in the decline of these cellular stocks and failing lung repair. Clinical tests with allogeneic MSCs from healthy individuals are underway to enhance endogenous lung repair and suppress inflammation.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Azithromycin/therapeutic use , Basigin/genetics , Betacoronavirus/drug effects , Coronavirus Infections/therapy , Pandemics , Pneumonia, Viral/therapy , Spike Glycoprotein, Coronavirus/genetics , Stem Cell Transplantation , Angiotensin-Converting Enzyme 2 , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/virology , Basigin/antagonists & inhibitors , Basigin/immunology , Betacoronavirus/metabolism , Betacoronavirus/pathogenicity , COVID-19 , Clinical Trials as Topic , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Gene Expression , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Lung/immunology , Lung/virology , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/immunology , Pneumonia, Viral/epidemiology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Protein Binding/drug effects , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/immunology , Stem Cells/drug effects , Stem Cells/immunology , Stem Cells/virology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology , Viral Load/drug effects
4.
Chemosphere ; 251: 126642, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32345545

ABSTRACT

The ubiquitous presence of aluminum in the environment leads to a high likelihood of human exposure. Neurotoxicity of the trivalent cationic form of this metal (Al3+) occurs in the central nervous system via accumulation of Al in cells of neural origin, including neural progenitor cells (NPCs). NPCs play a key role in the development and regeneration of the brain throughout life; therefore, this metal may contribute to neuropathological conditions. Here, we evaluated the effects of different Al3+ concentrations (0-50 µM) on the purinergic system of NPCs isolated from embryonic telencephalons, cultured as neurospheres. Al3+ adhered to the cell surface of neurospheres reducing extracellular ATP release, as well as ATP, ADP, and AMP hydrolysis by NTPDase and 5'-nucleotidase, respectively. In addition, impaired nucleotide release by Al3+ reduced P2Y1 and adenosine A2A receptors expression in differentiated neurospheres. These receptors are crucial for NPC proliferation during brain development and self-repair against external stimuli, such as metal exposure. Thus, Al3+ represents an environmental agent linked to neurodegeneration through alterations in the ATP-signalling pathway, proving to be a potential mechanism associated with NPC proliferation and brain degeneration.


Subject(s)
Aluminum/toxicity , 5'-Nucleotidase , Adenosine Triphosphate/metabolism , Aluminum/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Central Nervous System/metabolism , GPI-Linked Proteins , Humans , Signal Transduction/drug effects , Stem Cells , Toxicity Tests
5.
Microb Pathog ; 136: 103703, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31476377

ABSTRACT

Trypanosoma evansi appears to have a significant tropism for brain tissue in its chronic and acute phases. The most common symptoms of this brain infection are motor incoordination, meningoencephalitis, demyelination, and anemia. There have only been few studies of the effects of T. evansi infection on neuronal differentiation and brain plasticity. Here, we investigated the impact of the congenital T. evansi infection on brain development in mice. We collected telencephalon-derived neural progenitor cells (NPCs) from T. evansi uninfected and infected mice, and cultivated them into neurospheres. We found that T. evansi significantly decreased the number of cells during development of neurospheres. Analysis of neurosphere differentiation revealed that T. evansi infection significantly increased neural migration. We also observed that T. evansi promoted expression of glial fibrillary acidic protein (GFAP) in infected cells. These data suggest that congenital T. evansi infection may affect embryonic brain development.


Subject(s)
Host-Pathogen Interactions , Neural Stem Cells/pathology , Neural Stem Cells/parasitology , Trypanosoma/growth & development , Animals , Cell Differentiation , Mice
6.
Cancer Microenviron ; 12(2-3): 77-94, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31420805

ABSTRACT

Tumour progression involves interactions among various cancer cell clones, including the cancer stem cell subpopulation and exogenous cellular components, termed cancer stromal cells. The latter include a plethora of tumour infiltrating immunocompetent cells, among which are also immuno-modulatory mesenchymal stem cells, which by vigorous migration to growing tumours and susequent transdifferentiation into various types of tumour-residing stromal cells, may either inhibit or support tumour progression. In the light of the scarce therapeutic options existing for the most malignant brain tumour glioblastoma, mesenchymal stem cells may represent a promising novel tool for cell therapy, e.g. drug delivery vectors. Here, we review the increasing number of reports on mutual interactions between mesenchymal stem cells and glioblastoma cells in their microenvironment. We particularly point out two novel aspects: the different responses of cancer cells to their microenvironmental cues, and to the signalling by kinin receptors that complement the immuno-modulating cytokine-signalling networks. Inflammatory glioblastoma microenvironment is characterised by increasing expression of kinin receptors during progressive glioma malignancy, thus making kinin signalling and kinins themselves rather important in this context. In general, their role in tumour microenvironment has not been explored so far. In addition, kinins also regulate blood brain barrier-related drug transfer as well as brain tumour angiogenesis. These studies support the on-going research on kinin antagonists as candidates in the development of anti-invasive agents for adjuvant glioblastoma therapy.

7.
Arch Toxicol ; 93(9): 2515-2524, 2019 09.
Article in English | MEDLINE | ID: mdl-31363819

ABSTRACT

Aluminum (Al) is a neurotoxin and is associated with the etiology of neurodegenerative diseases, such as Alzheimer's disease (AD). The Al-free ion (Al3+) is the biologically reactive and toxic form. However, the underlying mechanisms of Al toxicity in the brain remain unclear. Here, we evaluated the effects of Al3+ (in the chloride form-AlCl3) at different concentrations (0.1-100 µM) on the morphology, proliferation, apoptosis, migration and differentiation of neural progenitor cells (NPCs) isolated from embryonic telencephalons, cultured as neurospheres. Our results reveal that Al3+ at 100 µM reduced the number and diameter of neurospheres. Cell cycle analysis showed that Al3+ had a decisive function in proliferation inhibition of NPCs during neural differentiation and induced apoptosis on neurospheres. In addition, 1 µM Al3+ resulted in deleterious effects on neural phenotype determination. Flow cytometry and immunocytochemistry analysis showed that Al3+ promoted a decrease in immature neuronal marker ß3-tubulin expression and an increase in co-expression of the NPC marker nestin and glial fibrillary acidic protein. Thus, our findings indicate that Al3+ caused cellular damage and reduced proliferation and migration, resulting in global inhibition of NPC differentiation and neurogenesis.


Subject(s)
Aluminum Chloride/toxicity , Embryonic Stem Cells/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Embryonic Stem Cells/pathology , Female , Male , Mice , Neural Stem Cells/pathology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/physiopathology , Phenotype , Telencephalon/drug effects , Telencephalon/embryology
8.
Microb Pathog ; 132: 156-161, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31029718

ABSTRACT

Chagas disease (CD) affecting about 7 million people is caused by the flagellate protozoan Trypanosoma cruzi. The central nervous system (CNS) is an important site for T. cruzi persistence in the host during the chronic phase of infection, because the protozoan may pass the blood-brain barrier and may cause motor and cognitive neuronal damage. Thinking about avoiding or minimizing these negative effects, it is hypothesized that resveratrol (RSV), a component with several medicinal properties has beneficial effects on the CNS. The objective of this study was to investigate, whether T. cruzi infection interferes with neurogenesis and gliogenesis of embryos of infected mice females, and whether RSV would be able to avoid or minimize these changes caused by CD. RSV is a polyphenol found in grapes and widely studied for its neuroprotective and antioxidant properties. In addition, we investigated the role caused by the parasite during congenital infection and CNS development. Embryos and their brains were PCR-positive for T. cruzi. For this study, NPCs obtained from telencephalon of infected and uninfected embryos and were cultured in presence of resveratrol for forming neurospheres. The results demonstrated that the congenital transmission of T. cruzi influences CNS formation and neural fate, decreasing the number of neuroespheres and causing an elongation in the phases of the cell cycle. In addition, the parasite promoted an increase in neugliogenesis. Resveratrol was neuroprotective and prevented negative effects of the infection. Thus, we suggest the use of resveratrol as a therapeutic target for the treatment of neuroinflammation or as neuroprotective agent during Chagas disease, as it improves gliogenesis and restores neural migration.


Subject(s)
Cell Differentiation/drug effects , Neurons/drug effects , Resveratrol/pharmacology , Stem Cells/drug effects , Animals , Chagas Disease/drug therapy , Disease Models, Animal , Female , Mice , Neurogenesis/drug effects , Neurons/cytology , Stem Cells/metabolism , Trypanosoma cruzi
9.
Mol Neurobiol ; 56(4): 2328-2338, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30027338

ABSTRACT

The intracellular protozoan Toxoplasma gondii may cause congenital toxoplasmosis and serious brain damage in fetus. However, the underlying mechanism of neuropathogenesis in brain toxoplasmosis remains unclear. For this study, neural progenitor cells (NPCs) were obtained from embryo telencephalons (embryonic day 13) and induced to proliferation in the presence of growth factors (GFs). For gathering insights into the biological effects of resveratrol (RSV) on neurogenesis, this study aimed to investigate effects of RSV concentrations (0.1 to 100 µM) on proliferation, migration and differentiation of NPCs infected by T. gondii. T. gondii infection increased the presence of cells in Sub G1 phase, reducing the global frequency of undifferentiated cells in S and G2/M phases of cell cycle and reduced cell viability/mithochondrial activity of infected NPCs. Moreover T. gondii stimulated neural migration and gliogenesis during neutral differentation. However, the treatment with RSV stimulated cell proliferation, restored cellular viability of infected NPCs and exerted an inhibitory effect on gliogenesis of infected NPCs favorecing neuronal maturation during toxoplasmosis infection. Thus, we have successfully to demonstrated that RSV is promising as therapeutic for congenital toxoplasmosis.


Subject(s)
Neural Stem Cells/parasitology , Neurogenesis/drug effects , Neuroglia/pathology , Resveratrol/pharmacology , Toxoplasma/physiology , Animals , Brain/growth & development , Brain/parasitology , Brain/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Infectious Disease Transmission, Vertical , Mice , Neural Stem Cells/drug effects , Neuroglia/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism
10.
Purinergic Signal ; 15(1): 77-84, 2019 03.
Article in English | MEDLINE | ID: mdl-30535987

ABSTRACT

The effects of Toxoplasma gondii during embryonic development have not been explored despite the predilection of this parasite for neurons and glial cells. Here, we investigated the activation of the purinergic system and proinflammatory responses during congenital infection by T. gondii. Moreover, neuroprotective and neuromodulatory properties of resveratrol (RSV), a polyphenolic natural compound, were studied in infected neuronal progenitor cells (NPCs). For this study, NPCs were isolated from the telencephalon of infected mouse embryos and subjected to neurosphere culture in the presence of EGF and FGF2. ATP hydrolysis and adenosine deamination by adenosine deaminase activity were altered in conditions of T. gondii infection. P2X7 and adenosine A2A receptor expression rates were augmented in infected NPCs together with an increase of proinflammatory (INF-γ and TNF-α) and anti-inflammatory (IL-10) cytokine gene expression. Our results confirm that RSV counteracted T. gondii-promoted effects on enzymes hydrolyzing extracellular nucleotides and nucleosides and also upregulated P2X7 and A2A receptor expression and activity, modulating INF-γ, TNF-α, and IL-10 cytokine production, which plays an integral role in the immune response against T. gondii.


Subject(s)
Antioxidants/pharmacology , Neural Stem Cells , Receptor, Adenosine A2A/metabolism , Receptors, Purinergic P2X7/metabolism , Resveratrol/pharmacology , Toxoplasmosis/metabolism , Animals , Female , Mice , Neural Stem Cells/drug effects , Neural Stem Cells/immunology , Neural Stem Cells/microbiology , Pregnancy , Prenatal Exposure Delayed Effects/microbiology , Purines/metabolism , Receptor, Adenosine A2A/immunology , Receptors, Purinergic P2X7/immunology , Toxoplasmosis/immunology
11.
Pesqui. vet. bras ; 38(12): 2183-2189, dez. 2018. ilus
Article in English | LILACS, VETINDEX | ID: biblio-976422

ABSTRACT

Adipose tissue-derived stem cells (ADSCs) are an attractive source of mesenchymal stem cells (MSCs) for use in tissue engineering and clinical applications. This paper focuses on the characterization of ADSCs used as immunosuppressive agent in rabbits undergoing partial allograft for urine bladder restorage. For this study highlighted the characterization of the ADSCs used as immunosuppressive agents in rabbits submitted to partial allograft for restoration of the urinary vesicle, using 25 animals, six months old, New Zealand. ADSCs at the third peal were characterized by the MSC-specific CD105, CD73 and CD90 expression and by the absence of the hematopoietic marker CD45, as revealed by flow cytometry analysis. Moreover, ADSCs were efficient in preventing allograft rejection from the urinary bladder, as judged by biochemical, clinical and ultrasonography analysis. Together, these results compose characterization of protein expression profiles and immunosuppressive functionality of ADSCs in rabbits, which had undergone partial allografts of the urinary bladder, foreseeing future applications in clinical practice.(AU)


As células mesenquimais derivadas de tecido adiposo (ADSCs) são uma fonte atraente de células-tronco mesenquimais (MSCs) para uso na engenharia de tecidos e suas aplicações clínicas. Este trabalho destacou a caracterização das ADSCs utilizadas como agentes imunossupressores em coelhos submetidos a aloenxerto parcial para restauração da vesícula urinária, sendo utilizados 25 animais, de seis meses de idade, Nova Zelândia. As ADSCs, após o terceiro repique, foram caracterizadas pela expressão específica de MSC CD105, CD73 e CD90 e pela ausência do marcador hematopoiético CD45, tal como revelado por análise de citometria de fluxo. Além disso, os ADSCs foram eficientes na prevenção da rejeição de aloenxertos da vesícula urinária, conforme avaliado por análises clínica, bioquímica e ultrassonográfica. Juntos, esses resultados compõem a caracterização dos perfis de expressão proteica e a funcionalidade imunossupressora de ADSCs em coelhos, que sofreram aloenxertos parciais da bexiga, prevendo futuras aplicações na prática clínica.(AU)


Subject(s)
Animals , Rabbits , Rabbits , Urinary Bladder/transplantation , Allografts/cytology , Cell- and Tissue-Based Therapy/veterinary , Immunosuppressive Agents , Flow Cytometry/veterinary
12.
Front Pharmacol ; 9: 500, 2018.
Article in English | MEDLINE | ID: mdl-29867502

ABSTRACT

Bone marrow metastasis occurs in approximately 350,000 patients that annually die in the U.S. alone. In view of the importance of tumor cell migration into the bone marrow, we have here investigated effects of various concentrations of stromal cell-derived factor-1 (SDF-1), bradykinin- and ATP on bone marrow metastasis. We show for first time that bradykinin augmented chemotactic responsiveness of neuroblastoma cells to SDF-1 and ATP concentrations, encountered under physiological conditions. Bradykinin upregulated VEGF expression, increased metalloproteinase activity and induced adhesion of neuroblastoma cells. Bradykinin augmented SDF-1-induced intracellular Ca2+ mobilization as well as resensitization and expression of ATP-sensing P2X7 receptors. Bradykinin treatment resulted in higher gene expression levels of the truncated P2X7B receptor compared to those of the P2X7A full-length isoform. Bradykinin as pro-metastatic factor induced tumor proliferation that was significantly decreased by P2X7 receptor antagonists; however, the peptide did not enhance cell death nor P2X7A receptor-related pore activity, promoting neuroblastoma growth. Furthermore, immunodeficient nude/nude mice transplanted with bradykinin-pretreated neuroblastoma cells revealed significantly higher metastasis rates compared to animals injected with untreated cells. In contrast, animals receiving Brilliant Blue G, a P2X7 receptor antagonist, did not show any specific dissemination of neuroblastoma cells to the bone marrow and liver, and metastasis rates were drastically reduced. Our data suggests correlated actions of kinins and purines in neuroblastoma dissemination, providing novel avenues for clinic research in preventing metastasis.

13.
Front Pharmacol ; 9: 325, 2018.
Article in English | MEDLINE | ID: mdl-29692728

ABSTRACT

Since proving adenosine triphosphate (ATP) functions as a neurotransmitter in neuron/glia interactions, the purinergic system has been more intensely studied within the scope of the central nervous system. In neurological disorders with associated motor symptoms, including Parkinson's disease (PD), motor neuron diseases (MND), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Huntington's Disease (HD), restless leg syndrome (RLS), and ataxias, alterations in purinergic receptor expression and activity have been noted, indicating a potential role for this system in disease etiology and progression. In neurodegenerative conditions, neural cell death provokes extensive ATP release and alters calcium signaling through purinergic receptor modulation. Consequently, neuroinflammatory responses, excitotoxicity and apoptosis are directly or indirectly induced. This review analyzes currently available data, which suggests involvement of the purinergic system in neuro-associated motor dysfunctions and underlying mechanisms. Possible targets for pharmacological interventions are also discussed.

14.
Neurobiol Learn Mem ; 149: 135-143, 2018 03.
Article in English | MEDLINE | ID: mdl-29458098

ABSTRACT

Lipopolysaccharide (LPS) has been long known to promote neuroinflammation and learning and memory deficits. Since spermine, one of the main natural polyamines in the central nervous system, protects from LPS-induced memory deficit by a mechanism that comprises GluN2B receptors, the aim of the present study was to determine whether brain-derived neurotrophic factor (BDNF), tropomyosin-related kinase B (TrkB) receptor and cAMP response element binding (CREB) are involved in this protective effect of spermine. Adult male Swiss albino mice received, immediately after training in the novel object recognition task, saline or LPS (250 µg/kg, i.p.); 5 min later they received saline or spermine (0.3 mg/kg, i.p.) and, when specified, 5 min thereafter saline or the TrkB receptor antagonist ANA-12 (0.5 mg/kg, i.p.) in different flanks. Animals were tested 24 h after training. Spermine protected from LPS-induced memory deficit and this protective effect was reversed by ANA-12. In a subset of animals BDNF, CREB and phospho-CREB immunoreactivity was determined in the hippocampi and cerebral cortex 4 h after spermine injection. Spermine reversed the decrease of mature BDNF levels induced by LPS in both hippocampus and cerebral cortex. Spermine increased phospho-CREB content and phospho-CREB/total CREB ratio in the cerebral cortex of LPS-treated mice. The results support that the protective effect of spermine on LPS-induced memory deficits depends on TrkB receptor activation and is accompanied by restoration of mature BDNF levels in hippocampus and cerebral cortex, as well as increased CREB phosphorylation in the cerebral cortex.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/drug effects , Membrane Glycoproteins/metabolism , Memory Disorders/metabolism , Neuroprotective Agents/pharmacology , Protein-Tyrosine Kinases/metabolism , Spermine/pharmacology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Hippocampus/metabolism , Lipopolysaccharides , Male , Memory Disorders/chemically induced , Mice , Phosphorylation/drug effects , Signal Transduction/drug effects
15.
Sci Rep ; 8(1): 1299, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29358738

ABSTRACT

Glioblastoma multiforme (GBM) represents the most lethal brain tumour, and these tumours have very limited treatment options. Mesenchymal stem cells (MSC) are considered as candidates for advanced cell therapies, due to their tropism towards GBM, possibly affecting their malignancy, thus also representing a potential therapeutic vector. Therefore, we aimed to compare the effects of bone-marrow-derived versus adipose-tissue-derived MSC (BM-/AT-MSC) on heterogeneous populations of tumour cells. This cells' interplay was addressed by the in-vitro two-dimensional (monolayer) and three-dimensional (spheroid) co-culture models, using U87 and U373 GBM cell lines, expressing genotypically different mesenchymal transcriptome profiles. U87 cell low mesenchymal profile expressed high levels of kinin receptor 1 (B1R) and their invasion was greatly enhanced by the B1R agonist des-Arg9-bradykinin upon BM-MSC co-culturing in 3D co-cultures. This correlated to significantly higher cell-cell interactions in U87/BM-MSC mixed spheroids. This was not observed with the U373 cells and not in AT-MSC co-cultures. Altogether, these data support the on-going exploration of B1R as target for adjuvant approach in GBM therapy. Secondly, the results emphasize the need for further careful exploration of the selectivity regarding the origin of MSC as potential candidates for cell therapies, particular in cancer, where they may adversely affect heterogeneous tumour cell populations.


Subject(s)
Bradykinin/analogs & derivatives , Cell Communication/drug effects , Cell Movement/drug effects , Neuroglia/drug effects , Receptor, Bradykinin B1/agonists , Spheroids, Cellular/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bradykinin/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Organ Specificity , Receptor, Bradykinin B1/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Tissue Culture Techniques
16.
Prog Neuropsychopharmacol Biol Psychiatry ; 80(Pt A): 34-53, 2018 Jan 03.
Article in English | MEDLINE | ID: mdl-28476640

ABSTRACT

Neuropsychiatric disorders involve various pathological mechanisms, resulting in neurodegeneration and brain atrophy. Neurodevelopmental processes have shown to be critical for the progression of those disorders, which are based on genetic and epigenetic mechanisms as well as on extrinsic factors. We review here common mechanisms underlying the comorbidity of Bipolar Disorders and Alzheimer's Disease, such as aberrant neurogenesis and neurotoxicity, reporting current therapeutic approaches. The understanding of these mechanisms precedes stem cell-based strategies as a new therapeutic possibility for treatment and prevention of Bipolar and Alzheimer's Disease progression. Taking into account the difficulty of studying the molecular basis of disease progression directly in patients, we also discuss the importance of stem cells for effective drug screening, modeling and treating psychiatric diseases, once in vitro differentiation of patient-induced pluripotent stem cells provides relevant information about embryonic origins, intracellular pathways and molecular mechanisms.


Subject(s)
Alzheimer Disease , Bipolar Disorder , Stem Cell Transplantation/methods , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Bipolar Disorder/etiology , Bipolar Disorder/metabolism , Bipolar Disorder/therapy , Humans
17.
Platelets ; 29(8): 801-810, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29090621

ABSTRACT

Signaling mediated by purines is a widespread mechanism of cell-cell communication related to vasomotor responses and the control of platelet function in the vascular system. However, little is known about the involvement of this signaling as well as the role of reactive oxygen species (ROS) in the development of hypothyroidism. Therefore, the present study investigates changes in the purinergic system, including enzyme activities and expression in platelets, and oxidative profiles in patients with post-thyroidectomy hypothyroidism. The nucleoside triphosphate diphosphohydrolase 1 (NTPDase/CD39) expression in patients increased by 40%, and the adenosine triphosphate (ATP) or adenosine diphosphate (ADP) hydrolyzing activity increased by 82% and 70%, respectively. The activities of ecto-5´-nucleotidase and adenosine deaminase (ADA) also significantly enhanced (39% and 52%, respectively), which correlates with a 45% decrease in adenosine concentration. Furthermore, these patients demonstrated an increased production of ROS (42%), thiobarbituric acid reactive substances (TBARS) (115%), carbonyl protein (30%) and a decreased glutathione S-transferase (GST) activity (20%). This study demonstrates that hypothyroidism interferes with adenine nucleoside and nucleotide hydrolysis and this is correlated with oxidative stress, which might be responsible for the increase in ADA activity. This increase causes rapid adenosine deamination, which can generate a decrease in their concentration in the systemic circulation, which can be associated with the development of vascular complications.


Subject(s)
Apyrase/blood , Blood Platelets/enzymology , Gene Expression Regulation, Enzymologic , Hypothyroidism/blood , Reactive Oxygen Species/blood , Thyroidectomy , Adenosine Diphosphate/blood , Adenosine Triphosphate/blood , Adult , Aged , Blood Platelets/pathology , Female , Humans , Hypothyroidism/etiology , Hypothyroidism/pathology , Male , Middle Aged
18.
Microb Pathog ; 110: 450-456, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28648625

ABSTRACT

The aim of this study was to evaluate the behavior of erythrocyte and platelet, as immunological markers, as well as evaluate the involvement of these factors in hemolytic and hemorrhagic reactions in hamsters experimentally infected by Leptospira interrogans Serovar Canicola. Our experimental design was composed by two randomized groups: Infected Group (IG) (n = 12) and control group (CG) (n = 6). Ninety-six hours after the inoculation, the presence of immunoglobulins (IgG and IgM) and complement C3 levels, related to erythrocytes and platelets, was assessed. Platelet's microparticles marked by CD61, reticulocytes and reticulated platelets were also quantified. Additionally, fibrinogen, prothrombin time, partially activated thromboplastin time and sera levels of IgG and IgM were assessed. Our results showed that levels of platelet decreased in IG (P < 0.001); as well as, there was presence of IgG and C3 associated with erythrocyte surface in the infected animals (P < 0.01, P < 0.05, respectively). Levels of prothrombin time and Activated Partial Thromboplastin Time were increased, while fibrinogen level was decreased (P < 0.01) in IG. CD61 microparticles were higher (P < 0.05) in IG due to platelet activation. Thus, it was established a positive correlation (P < 0.01) between platelets count and fibrinogen (Figure 3, R = 0.84, P < 0.001). Therefore, the platelet consumption component was preponderant in relation to autoimmune causes. Finally, regarding the erythrocytes, the autoimmune component played an important role, did not causing hemolytic reaction in this acute experimental time.


Subject(s)
Autoantibodies/blood , Erythrocytes/immunology , Immunoglobulin G/blood , Leptospira interrogans serovar canicola/pathogenicity , Leptospirosis/pathology , Animals , Blood Platelets/immunology , Complement C3/analysis , Cricetinae , Disease Models, Animal , Platelet Count
19.
Neurobiol Learn Mem ; 140: 82-91, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28254465

ABSTRACT

Putrescine, spermidine and spermine are organic cations implicated in learning, memory consolidation, reconsolidation and neurogenesis. These physiological processes are closely related, and convincing evidence indicates that neurogenesis is implicated both, in the establishment and maintenance of remote contextual fear memory. Although brain-derived neurotrophic factor (BDNF) is a key mediator involved in both neurogenesis and memory consolidation, effects of spermidine on persistence of memory after reactivation (reconsolidation) and possible involvement of BDNF have not been investigated. Here, we investigated whether the intrahippocampal infusion of spermidine improves the persistence of reconsolidated contextual fear conditioning memory in rats and whether these possible changes depend on BDNF/TrkB signaling in the hippocampus. The infusion of spermidine immediately and 12h post-reactivation improved fear memory of the animals tested seven but not two days after reactivation. The facilitatory effect of spermidine on the persistence of reconsolidated memory was blocked by the TrkB inhibitor ANA-12 (73.6pmol/site) and accompanied by mature BDNF level increase in the hippocampus, indicating that it depends on the BDNF/TrkB pathway. We also investigated whether spermidine alters BDNF levels and neural progenitor cell differentiation in vitro. Spermidine increased BDNF levels in vitro, facilitating neuritogenesis and neural migration. Spermidine-induced neuritogenesis in vitro was also blocked by ANA-12 (10µM). Since spermidine increases BDNF levels and facilitates neural differentiation in vitro, similar mechanisms may be involved in spermidine-induced facilitation of the persistence of reconsolidated memory.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Fear/drug effects , Hippocampus/drug effects , Memory Consolidation/drug effects , Neurogenesis/drug effects , Spermidine/pharmacology , Animals , Azepines/pharmacology , Benzamides/pharmacology , Cell Movement/drug effects , Conditioning, Classical/drug effects , Hippocampus/metabolism , Male , Rats , Rats, Wistar , Receptor, trkB/antagonists & inhibitors
20.
J Cell Sci ; 129(18): 3437-48, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27528403

ABSTRACT

During brain development, cells proliferate, migrate and differentiate in highly accurate patterns. In this context, published results indicate that bradykinin functions in neural fate determination, favoring neurogenesis and migration. However, mechanisms underlying bradykinin function are yet to be explored. Our findings indicate a previously unidentified role for bradykinin action in inducing neuron-generating division in vitro and in vivo, given that bradykinin lengthened the G1-phase of the neural progenitor cells (NPC) cycle and increased TIS21 (also known as PC3 and BTG2) expression in hippocampus from newborn mice. This role, triggered by activation of the kinin-B2 receptor, was conditioned by ERK1/2 activation. Moreover, immunohistochemistry analysis of hippocampal dentate gyrus showed that the percentage of Ki67(+) cells markedly increased in bradykinin-treated mice, and ERK1/2 inhibition affected this neurogenic response. The progress of neurogenesis depended on sustained ERK phosphorylation and resulted in ERK1/2 translocation to the nucleus in NPCs and PC12 cells, changing expression of genes such as Hes1 and Ngn2 (also known as Neurog2). In agreement with the function of ERK in integrating signaling pathways, effects of bradykinin in stimulating neurogenesis were reversed following removal of protein kinase C (PKC)-mediated sustained phosphorylation.


Subject(s)
Bradykinin/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/enzymology , Neurons/metabolism , Animals , Calcium/metabolism , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Lineage/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Enzyme Activation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , PC12 Cells , Phenotype , Phosphorylation/drug effects , Rats , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...