Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Exp Med ; 23(7): 3651-3662, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36943594

ABSTRACT

Sepsis is a life-threatening organ dysfunction caused by a dysregulated inflammatory response to infection. To date, there is no specific treatment established for sepsis. In the extracellular compartment, purines such as adenosine triphosphate (ATP) and adenosine play essential roles in the immune/inflammatory responses during sepsis and septic shock. The balance of extracellular levels among ATP and adenosine is intimately involved in the signals related to immune stimulation/immunosuppression balance. Specialized enzymes, including CD39, CD73, and adenosine deaminase (ADA), are responsible to metabolize ATP to adenosine which will further sensitize the P2 and P1 purinoceptors, respectively. Disruption of the purinergic pathway had been described in the sepsis pathophysiology. Although purinergic signaling has been suggested as a potential target for sepsis treatment, the majority of data available were obtained using pre-clinical approaches. We hypothesized that, as a reflection of deregulation on purinergic signaling, septic patients exhibit differential measurements of serum, neutrophils and monocytes purinergic pathway markers when compared to two types of controls (healthy and ward). It was observed that ATP and ADP serum levels were increased in septic patients, as well as the A2a mRNA expression in neutrophils and monocytes. Both ATPase/ADPase activities were increased during sepsis. Serum ATP and ADP levels, and both ATPase and ADPase activities were associated with the diagnosis of sepsis, representing potential biomarkers candidates. In conclusion, our results advance the translation of purinergic signaling from pre-clinical models into the clinical setting opening opportunities for so much needed new strategies for sepsis and septic shock diagnostics and treatment.


Subject(s)
Sepsis , Shock, Septic , Humans , Apyrase/metabolism , Adenosine , Adenosine Triphosphate/metabolism , Biomarkers , Sepsis/diagnosis , Adenosine Diphosphate , Adenosine Triphosphatases
2.
Cancer Chemother Pharmacol ; 85(6): 1177-1182, 2020 06.
Article in English | MEDLINE | ID: mdl-32417936

ABSTRACT

Glioblastoma is the most devastating primary brain tumor and effective therapies are not available. Treatment is based on surgery followed by radio and chemotherapy with temozolomide (TMZ), but TMZ increases patient survival only by 2 months. CD73, an enzyme responsible for adenosine production, emerges as a target for glioblastoma treatment. Indeed, adenosine causes tumor-promoting actions and CD73 inhibition increases sensitivity to TMZ in vitro. Here, a cationic nanoemulsion to nasal delivery of siRNA CD73 (NE-siRNA CD73) aiming glioblastoma treatment was employed alone or in combination with TMZ. In vitro, two glioblastoma cell lines (C6 and U138MG) with a chemo-resistant profile were used. Treatment alone with NE-siRNA CD73 reduced C6 and U138MG glioma cell viability by 70% and 25%, respectively. On the other hand, when NE-siRNA + TMZ combined treatment was employed, a reduction of 85% and 33% of cell viability was observed. Notably, treatment with NE-siRNA CD73 of glioma-bearing Wistar rats reduced tumor size by 80%, 60% more than the standard chemotherapy with TMZ, but no synergistic or additive effect was observed in vivo. Additionally, NE-siRNA CD73, TMZ or combined therapy decreased adenosine levels in liquor confirming the importance of this nucleoside on in vivo GB growth. Finally, no hemolytic potential was observed. These results suggest that nasal administration of NE-siRNA CD73 exhibits higher antiglioma effect when compared to TMZ. However, no synergistic or additive in vivo was promoted by the therapeutic regimen employed in this study.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , RNA, Small Interfering/genetics , Temozolomide/pharmacology , 5'-Nucleotidase/genetics , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation , Drug Evaluation, Preclinical , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , RNA, Small Interfering/administration & dosage , Rats , Rats, Wistar , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Mol Neurobiol ; 57(2): 635-649, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31407144

ABSTRACT

Glioblastoma is the most devastating primary brain tumor. Effective therapies are not available, mainly due to high tumor heterogeneity, chemoresistance, and the difficulties imposed by blood-brain barrier. CD73, an enzyme responsible for adenosine (ADO) production, is overexpressed in cancer cells and emerges as a target for glioblastoma treatment. Indeed, ADO causes a variety of tumor-promoting actions, particularly by inducing tumor immune escape, whereas CD73 inhibition impairs tumor progression. Here, a cationic nanoemulsion to deliver CD73siRNA (NE-siRNA CD73R) via nasal route aiming glioblastoma treatment was developed. NE-siRNA CD73R was uptaken by glioma cells in culture, resulting in a parallel 60-80% decrease in AMPase activity and 30-50% in cell viability. Upon nasal delivery, NE-siRNA CD73R was detected in rat brain and serum. Notably, treatment with CD73siRNA complexes of glioma-bearing Wistar rats reduced tumor growth by 60%. Additionally, NE-siRNA CD73R treatment decreased 95% ADO levels in liquor and tumor CD73 expression, confirming in vivo CD73 silencing. Finally, no toxicity was observed in either primary astrocytes or rats with this cationic nanoemulsion. These results suggest that nasal administration of cationic NE as CD73 siRNA delivery system represents a novel potential treatment for glioblastoma. Graphical Abstract Glioblastoma is the most common and devastating form of primary brain tumor. CD73, a protein involved in cell-cell adhesion and migration processes and also responsible for extracellular adenosine (ADO) production, is overexpressed by glioma cells and emerges as an important target for glioma treatment. Indeed, ADO participates in tumor immune escape, cell proliferation, and angiogenesis, and CD73 inhibition impairs those processes. Here, a cationic nanoemulsion to deliver CD73 siRNA (NE-siRNA CD73R) via nasal route aiming glioblastoma treatment was developed. NE-siRNA CD73R knockdown in vitro and in vivo CD73. Upon nasal delivery of NE-siRNA CD73R, the treatment markedly reduced tumor volume by 60% in a rat preclinical glioblastoma model. The treatment was well tolerated, and did not induce kidney, liver, lung, olfactory, bone marrow, or behavior alterations. These results indicate that the nasal administration of NE as a CD73 siRNA delivery system offered an efficient means of gene knockdown and may represent a potential alternative for glioblastoma treatment.


Subject(s)
5'-Nucleotidase/metabolism , Emulsions/administration & dosage , Gene Transfer Techniques , Glioblastoma/therapy , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , Administration, Intranasal , Animals , Astrocytes/pathology , Brain Neoplasms/therapy , Cations , Cell Line, Tumor , Cell Proliferation , Cell Survival , GPI-Linked Proteins/metabolism , Glioblastoma/pathology , Humans , Male , Rats, Wistar
4.
Mol Neurobiol ; 56(5): 3260-3279, 2019 May.
Article in English | MEDLINE | ID: mdl-30117104

ABSTRACT

Glioblastoma is the worst and most common primary brain tumor. Here, we demonstrated the role of CD73, an enzyme responsible for adenosine (ADO) production, in glioblastoma progression. ADO increased glioma cell viability via A1 receptor sensitization. CD73 downregulation decreased glioma cell migration and invasion by reducing metalloproteinase-2 and vimentin expression and reduced cell proliferation by 40%, which was related to necrosis and sub-G1 phase blockage of cell cycle. Those effects also involved the stimulation of Akt/NF-kB pathways. Additionally, CD73 knockdown or enzyme inhibition potentiated temozolomide cytotoxic effect on glioma cells by decreasing the IC50 value and sensitizing cells to a non-cytotoxic drug concentration. CD73 inhibition also decreased in vivo rat glioblastoma progression. Delivery of siRNA-CD73 or APCP reduced tumor size by 45 and 40%, respectively, when compared with control. This effect was followed by a parallel 95% reduction of ADO levels in cerebrospinal fluid, indicating the role of extracellular ADO in in vivo glioma growth. Treatment did not induce systemic damage or mortality. Altogether, we conclude that CD73 is an interesting target for glioblastoma treatment and its inhibition may provide new opportunities to improve the treatment of brain tumors. Graphical Abstract ᅟ.


Subject(s)
5'-Nucleotidase/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Down-Regulation/genetics , Glioblastoma/genetics , Glioblastoma/pathology , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Animals , Biomarkers, Tumor/blood , Brain Neoplasms/blood , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/genetics , Cell Survival , Disease Progression , Gene Knockdown Techniques , Glioblastoma/blood , Glioblastoma/drug therapy , Humans , Matrix Metalloproteinase 2/metabolism , NF-kappa B/metabolism , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptors, Purinergic P1/metabolism , Signal Transduction , Temozolomide/pharmacology , Temozolomide/therapeutic use , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...