Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Int. j. morphol ; 41(6): 1610-1619, dic. 2023. ilus, tab
Article in English | LILACS | ID: biblio-1528775

ABSTRACT

SUMMARY: Keloid scar is a unique benign fibroproliferative tumor of the human skin. Previously, it was reported that early growth response 1 (EGR1), a transcription factor, promotes keloid fibrosis; however, the mechanism by which EGR1 modulates keloid formation was not elaborated. In this research, the specific function and the microRNA (miRNA) regulatory network of EGR1 in keloids was examined. Keloid fibroblasts (KFs) were transfected with EGR1-small interfering RNA (siEGR1), EGR1-overexpression plasmid (pcDNA3.1-EGR1), and microRNA (miR-183-5p)-mimics to regulate the expression of EGR1 and miR-183-5p. The study employed dual-luciferase reporter assays to explore the targeting regulation of miR-183-5p on EGR1. Additionally, Western blotting, flow cytometry, qRT-PCR, cell count kit-8 (CCK-8), transwell, and wound healing assays, and RNA sequencing were conducted. EGR1 was upregulated in KFs, and EGR1 silencing diminished proliferation, fibrosis, migration, invasion, and apoptosis of cells. In KFs, the expression of miR- 183-5p was reduced, leading to the inhibition of cell proliferation, migration, and invasion. Conversely, it enhanced apoptosis. By targeting EGR1, miR-183-5p partially counteracted the impact of EGR1 on migration, invasion, and fibrosis in KFs. The findings imply that miR-183-5p suppresses keloid formation by targeting EGR1. As a result, EGR1 holds promise as a potential therapeutic target for preventing and treating keloids.


La cicatriz queloide es un tumor fibroproliferativo benigno único de la piel humana. Anteriormente, se informó que la respuesta de crecimiento temprano 1 (EGR1), un factor de transcripción, promueve la fibrosis queloide; sin embargo, no se explicó el mecanismo por el cual EGR1 modula la formación de queloides. En esta investigación, se examinó la función específica y la red reguladora de microARN (miARN) de EGR1 en queloides. Se transfectaron fibroblastos queloides (KF) con ARN de interferencia pequeño de EGR1 (siEGR1), plásmido de sobreexpresión de EGR1 (pcDNA3.1-EGR1) y miméticos de microARN (miR-183-5p) para regular la expresión de EGR1 y miR-183. -5p. El estudio empleó ensayos de indicador de luciferasa dual para explorar la regulación dirigida de miR-183-5p en EGR1. Además, se realizaron pruebas de transferencia Western, citometría de flujo, qRT-PCR, kit de recuento celular-8 (CCK-8), transwell y curación de heridas, y secuenciación de ARN. EGR1 estaba regulado positivamente en KF, y el silenciamiento de EGR1 disminuyó la proliferación, fibrosis, migración, invasión y apoptosis de las células. En KF, la expresión de miR- 183-5p se redujo, lo que llevó a la inhibición de la proliferación, migración e invasión celular. Por el contrario, mejoró la apoptosis. Al apuntar a EGR1, miR-183-5p contrarrestó parcialmente el impacto de EGR1 en la migración, invasión y fibrosis en KF. Los hallazgos implican que miR-183-5p suprime la formación de queloides al apuntar a EGR1. Como resultado, EGR1 es prometedor como objetivo terapéutico potencial para prevenir y tratar los queloides.


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Young Adult , Early Growth Response Protein 1 , Fibroblasts , Keloid/genetics , Keloid/pathology , Wound Healing , Transfection , Down-Regulation , Cell Movement , Blotting, Western , Sequence Analysis, RNA , Apoptosis , MicroRNAs/physiology , Cell Proliferation , Real-Time Polymerase Chain Reaction
2.
Actual. osteol ; 19(1): 18-29, ago. 2023. tab
Article in English | LILACS, UNISALUD, BINACIS | ID: biblio-1511400

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNA molecules that play critical roles in post-transcriptional gene regulation. They function by binding to target messenger RNA (mRNA) molecules, leading to their degradation or inhibiting their translation into proteins. In the context of skeletal diseases, such as osteoporosis, osteoarthritis, and bone metastasis, there is growing evidence osteoblastic miRNAs, are involved in the regulation of bone formation and maintenance.Osteoblasts are bone-forming cells responsible for synthesizing and depositing the extracellular matrix, which ultimately mineralizes to form bone tissue. Osteoblastic miRNAs modulate various aspects of osteoblast function, including proliferation, differentiation, mineralization, and apoptosis. Dysregulation of these miRNAs can disrupt the balance between bone formation and resorption, leading to skeletal diseases.The therapeutic implications of targeting osteoblastic miRNAs in skeletal diseases are significant. Modulating the expression levels of specific miRNAs holds promise for developing novel therapeutic strategies to enhance bone formation, prevent bone loss, and promote bone regeneration. Potential therapeutic approaches include the use of synthetic miRNA mimics to restore miRNA expression in diseases associated with miRNA downregulation or the use of anti-miRNA oligonucleotides to inhibit miRNA function in diseases associated with miRNA upregulation.miRNA-based therapies are still in the early stages of development, and further research is needed to fully understand the complexity of miRNA networks. Additionally, the delivery of miRNAs to specific target tissues and cells remains a challenge that needs to be addressed for effective clinical translation. Nonetheless, targeting osteoblastic miRNAs represents a promising avenue for future therapeutic interventions in skeletal diseases. (AU)


Los micro-ARNs (miARNss) son pequeños ARN no codificantes que desempeñan un papel fundamental en la regulación génica postranscripcional. Ejercen su función al unir-se a moléculas de ARN mensajero (ARNm), promoviendo su degradación e inhibiendo su traducción en proteínas. En el contexto de las enfermedades esqueléticas, como la osteoporosis, la osteoartritis y la metástasis ósea existe evidencia de que los miARNs osteoblásticos están involucrados en la regulación de la formación y del mantenimiento óseo. Los osteoblastos son células formadoras de hueso responsables de sintetizar y depositar la matriz extracelular, que finalmente se mineraliza para formar el hueso. Los miARNs derivados de osteoblastos modulan varios aspectos de la función de estas células, incluida la proliferación, diferenciación, mineralización y la apoptosis. La desregulación de estos miARNs puede alterar el equilibrio entre la formación y la resorción ósea, lo que lleva a enfermedades óseas. Las implicaciones terapéuticas de los miARNs osteoblásticos en enfermedades esqueléticas son significativas. La modulación de los niveles de expresión de miARNs específicos es prometedora para desarrollar nuevas estrate-gias terapéuticas a fin de mejorar la formación, prevenir la pérdida y promover la regeneración ósea. Los enfoques terapéuticos potenciales incluyen el uso de miméticos de miARNs para restaurar la expresión de miARNs o el uso de oligonucleótidos anti-miARNs para inhibir su función. Las terapias basadas en miARNs aún se encuentran en las primeras etapas de desarrollo. La administración de miARNs a las células y los tejidos específicos sigue siendo un desafío para lograr una aplicación clínica eficaz. (AU)


Subject(s)
Humans , Osteoblasts/cytology , Osteogenesis/genetics , MicroRNAs/genetics , Osteoclasts/cytology , Bone Diseases/prevention & control , Signal Transduction , Gene Expression Regulation , MicroRNAs/biosynthesis , MicroRNAs/physiology , MicroRNAs/therapeutic use
3.
Int. j. morphol ; 38(6): 1700-1706, Dec. 2020. graf
Article in English | LILACS | ID: biblio-1134501

ABSTRACT

SUMMARY: The treatment of chronic wounds has become a public health issue in recent years mainly due to comorbidities associated with an older population and bacterial resistance. Honey has emerged as an alternative treatment for chronic wounds but lack of knowledge of its mechanism of actionin the treated tissue and low quality of evidence in clinical triads has distanced the medical community from honey as a possible treatment. One of the main processes that is altered in chronic wounds is re-epithelialization mediated by keratinocytes, where proliferation and migration processes are altered. Markers of proliferation, migration and activation of keratinocytes, such as adhesion molecules, growth factors, membrane receptors, signal translating proteins, transcription factors, microRNAs, among others are deregulated in this process. In general, honeys from different floral origins have a positive effect on markers of proliferation and migration in keratinocytes. In conclusion there are still few studies that focus on the molecular action of honey in keratinocytes and fail to report details on the honey used not allowing to achieve the same results.


RESUMEN: El tratamiento de heridas crónicas (HC) se ha vuelto un tema de salud pública en los últimos años, principalmente debido a comorbilidades asociadas a una población de mayor edad y a la resistencia bacteriana. La miel ha surgido como un tratamiento alternativo para HC pero la falta de conocimiento de su mecanismo de acción en el tejido tratado y de la baja calidad de la evidencia en triadas clínicas, ha distanciado a la comunidad médica de la miel como posible tratamiento. Uno de los principales procesos que se ve alterado en las HC es la re-epitelización mediada por queratinocitos, donde se ven alterados los procesos de proliferación y migración. Marcadores de proliferación, migración y activación de queratinocitos, como moléculas de adhesión, factores de crecimiento, receptores de membrana, proteínas traductores de señales, factores de transcripción, microARNs, entre otras, se ven desreguladas en éste proceso. De manera general las mieles de diferentes orígenes florales tienen un efecto positivo en marcadores de proliferación y migración en queratinocitos. En conclusión aún existen pocos estudios que se enfoquen en la acción molecular de la miel en queratinocitos y los pocos que existen fallan en la entrega de información en relación a la miel utilizada que pueda hacer reproducibles los resultados.


Subject(s)
Wound Healing/physiology , Keratinocytes/physiology , Re-Epithelialization/physiology , Honey , Wound Healing/genetics , MicroRNAs/physiology , MicroRNAs/genetics , Re-Epithelialization/genetics
4.
Actual. osteol ; 16(3): 188-210, 2020. ilus, tab
Article in Spanish | LILACS | ID: biblio-1253839

ABSTRACT

Los micro-ARNs (miARNs) son pequeñas moléculas de ARN no codificante (de aproximadamente 15-25 nucleótidos), que regulan la expresión de genes involucrados en numerosas funciones biológicas, a través de la inhibición o degradación de un ARN mensajero diana. La homeostasis ósea se mantiene por el balance entre la formación osteoblástica y la resorción osteoclástica. La sobreexpresión o inhibición de miARNs específicos afecta la proliferación, diferenciación y actividad de osteoblastos, osteocitos y osteoclastos. Estas acciones son llevadas a cabo modulando la expresión de distintos factores transcripcionales y moléculas de señalización de las vías esenciales para la osteoblastogénesis u osteoclastogénesis. Estos efectos modifican el balance entre la formación y la resorción, determinando cambios en la homeostasis ósea. Esta revisión enumera una serie de miARNs que participan en la homeostasis ósea. Profundizando en el conocimiento de los mecanismos por medio de los cuales los miARNs actúan sobre el hueso, podrían revelarse nuevos usos potenciales futuros, entre los que se encuentran su utilidad como nuevos biomarcadores óseos o como agentes terapéuticos para el tratamiento de trastornos metabólicos óseos, pérdida de masa ósea o enfermedades óseas. (AU)


MicroRNAs (miRNAs) are endogenous small noncoding RNA molecules (of approximately 15­25 nucleotides), which regulate the expression of genes controlling numerous biological functions, through the inhibition or degradation of the target messenger RNA. Bone homeostasis is maintained by a balance between osteoblastic bone formation and osteoclastic bone resorption. The overexpression or inhibition of specific miRNAs affects cell proliferation, differentiation and activity of osteoblast, osteocytes and osteoclast. This action is done by modulating the expression of different transcription factors and signaling molecules of the most relevant pathways of osteoblastogenesis or osteoclastogenesis. This effect is able to modify the balance between bone formation and resorption, determining changes in bone homeostasis. The present review is an overview of a series of miRNAs involved in bone homeostasis. An in depth knowledge of the mechanisms by which miRNAs act on bone may reveal potential uses in the future as new bone biomarkers or therapeutic agents for treating metabolic bone disorders, bone loss and bone diseases. (AU)


Subject(s)
Humans , Bone Remodeling , MicroRNAs/therapeutic use , Osteoblasts , Osteoclasts , Osteocytes , Skeleton/metabolism , Bone Diseases/therapy , Bone Resorption/therapy , Biomarkers , MicroRNAs/physiology , Fractures, Bone/prevention & control
5.
Biol. Res ; 53: 27, 2020. graf
Article in English | LILACS | ID: biblio-1124212

ABSTRACT

BACKGROUND: Circular RNA (circRNA) is highly expressed in the brain tissue, but its molecular mechanism in cerebral ischemia-reperfusion remains unclear. Here, we explored the role and underlying mechanisms of circRNA antisense non-coding RNA in the INK4 locus (circ_ANRIL) in oxygen-glucose deprivation and reoxygenation (OGD/R)-induced cell injury. RESULTS: The expression of circ_ANRIL in OGD/R-induced human brain microvascular endothelial cells (HBMECs) was significantly up-regulated, while that of miR-622 was significantly down-regulated. Overexpression of circ_ANRIL significantly inhibited the proliferation of OGD/R-induced HBMECs and aggravated OGD/R-induced cell apoptosis. Moreover, circ_ANRIL overexpression further increased the secretion of interleukin (IL)-1ß, IL-6, tumor necrosis factor-a, and monocyte chemoattractant protein-1 in OGD/R-treated HBMECs. The results of bioinformatics analysis and luciferase reporter assay indicated that circ_ANRIL served as an miR-622 sponge to negatively regulate the expression of miR-622 in OGD/R-treated HBMECs. Additionally, circ_ANRIL silencing exerted anti-apoptotic and anti-inflammatory effects by positively regulating the expression of miR-622. Furthermore, inhibition of OGD/R-induced activation of the nuclear factor (NF)-kB pathway by circ_ANRIL silencing was significantly reversed by treatment with miR-622 inhibitor. CONCLUSIONS: Knockdown of circ_ANRIL improved OGD/R-induced cell damage, apoptosis, and inflammatory responses by inhibiting the NF-κB pathway through sponging miR-622.


Subject(s)
Humans , Reperfusion Injury/metabolism , Hypoxia, Brain/metabolism , MicroRNAs/physiology , MicroRNAs/genetics , RNA, Circular , Oxygen , Brain , Apoptosis , Cyclin-Dependent Kinase Inhibitor p16 , Endothelial Cells , RNA, Long Noncoding , Glucose/metabolism , Inflammation
6.
Biol. Res ; 53: 43, 2020. tab, graf
Article in English | LILACS | ID: biblio-1131887

ABSTRACT

BACKGROUND: Breast cancer, the most common cancer in women worldwide, causes the vast majority of cancer-related deaths. Undoubtedly, tumor metastasis and recurrence are responsible for more than 90 percent of these deaths. MicroRNAs are endogenous noncoding RNAs that have been integrated into almost all the physiological and pathological processes, including metastasis. In the present study, the role of miR-128 in breast cancer was investigated. RESULTS: Compared to the corresponding adjacent normal tissue, the expression of miR-128 was significantly suppressed in human breast cancer specimens. More importantly, its expression level was reversely correlated to histological grade of the cancer. Ectopic expression of miR-128 in the aggressive breast cancer cell line MDA-MB-231 could inhibit cell motility and invasive capacity remarkably. Afterwards, Metadherin (MTDH), also known as AEG-1 (Astrocyte Elevated Gene 1) and Lyric that implicated in various aspects of cancer progression and metastasis, was further identified as a direct target gene of miR-128 and its expression level was up-regulated in clinical samples as expected. Moreover, knockdown of MTDH in MDA-MB-231 cells obviously impaired the migration and invasion capabilities, whereas re-expression of MTDH abrogated the suppressive effect caused by miR-128. CONCLUSIONS: Overall, these findings demonstrate that miR-128 could serve as a novel biomarker for breast cancer metastasis and a potent target for treatment in the future.


Subject(s)
Humans , Female , Breast Neoplasms/genetics , MicroRNAs/physiology , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Gene Expression Regulation, Neoplastic , RNA-Binding Proteins , Cell Line, Tumor , Membrane Proteins , Neoplasm Recurrence, Local
7.
Arq. bras. cardiol ; 112(2): 154-162, Feb. 2019. tab, graf
Article in English | LILACS | ID: biblio-983823

ABSTRACT

Abstract Background: Diabetes mellitus (DM) is one of the major risk factors for cardiovascular disease, leading to endothelial dysfunction and angiogenesis impairment . MiR-126 and miR-210 support angiogenic response in endothelial cells. Objective: The present study sought to explore the effect of garlic and voluntary exercise, alone or together, on miR-126 and miR-210 expressions and cardiac angiogenesis in rats with type 1 diabetes. Methods: Male Wistar rats were divided into five groups (n = 7): Control, Diabetes, Diabetes+Garlic, Diabetes+Exercise, and Diabetes+Garlic+Exercise. Diabetes was induced in the animals by streptozotocin (ip, 50 mg/kg). The rats were then fed raw fresh garlic homogenate (250 mg/kg) or were subjected to voluntary exercise, or to combined garlic and voluntary exercise for 6 weeks. MiR-126 and miR-210 expressions in the myocardium were determined by real time PCR, and the serum lipid profile was measured by enzymatic kits. Angiogenesis was evaluated by immunostaining for PECAM-1/ CD31 in the myocardium. Results: Diabetes reduced both cardiac miR-126 expression and angiogenesis (p < 0.05). On the other hand, there was a miR-210 expression increase in the myocardium of diabetic animals (p < 0.001). However, those effects reversed either with garlic or voluntary exercise (p < 0.01). Moreover, treating diabetic rats with garlic and voluntary exercise combined had an additional effect on the expressions of miR-126 and miR-210 (p < 0.001). Furthermore, both voluntary exercise and garlic significantly improved serum lipid profiles (p < 0.001). Conclusion: The induction of diabetes decreased angiogenesis in the myocardium, whereas our treatment using long-term voluntary exercise and garlic improved myocardial angiogenesis. These changes were possibly owing to the enhancement of myocardial miR-126 and miR-210 expressions.


Resumo Fundamento: O diabetes mellitus (DM) é um dos principais fatores de risco para doenças cardiovasculares, levando à disfunção endotelial e inibição da angiogênese. O miRNA-126 e o miRNA-210 promovem a resposta angiogênica em células endoteliais. Objetivo: O presente estudo buscou explorar o efeito do alho e de exercícios físicos voluntários, isoladamente ou em conjunto, nas expressões do miRNA-126 e do miR-210 e na angiogênese cardíaca em ratos com diabetes tipo 1. Métodos: Ratos Wistar machos foram divididos em cinco grupos (n = 7): Controle, Diabetes, Diabetes+Alho, Diabetes+Exercícios e Diabetes+Alho+Exercícios. Introduziu-se diabetes nos animais por estreptozotocina (ip, 50 mg/kg). Os ratos foram então alimentados com homogenato de alho fresco cru (250 mg/kg), ou foram submetidos a exercícios voluntários, ou a uma combinação de alho e exercícios voluntários, durante 6 semanas. As expressões do miRNA-126 e do miRNA-210 no miocárdio foram determinadas por PCR em tempo real, e o perfil lipídico sérico foi medido por kits enzimáticos. A angiogênese foi avaliada por imunocoloração por PECAM-1/CD31 no miocárdio Resultados: O diabetes reduziu a expressão do miRNA-126 cardíaco e da angiogênese (p < 0,05). Por outro lado, houve um aumento da expressão do miRNA-210 no miocárdio dos animais diabéticos (p < 0,001). No entanto, tais efeitos foram revertidos com alho ou exercícios voluntários (p < 0,01). Além disso, o tratamento de ratos diabéticos conjuntamente com alho e exercícios voluntários teve um efeito adicional sobre as expressões do miRNA-126 e do miRNA-210 (p < 0,001). Além disso, tanto os exercícios voluntários quanto o alho melhoraram significativamente os perfis lipídicos séricos (p < 0,001). Conclusões: A indução de diabetes diminuiu a angiogênese no miocárdio, enquanto nosso tratamento com exercícios voluntários de longa duração e alho melhorou a angiogênese miocárdica. Estas alterações devem-se, possivelmente, ao aumento das expressões do miRNA-126 e do miRNA no miocárdio.


Subject(s)
Animals , Male , Physical Conditioning, Animal/physiology , Neovascularization, Physiologic/physiology , Coronary Vessels/physiopathology , MicroRNAs/analysis , Diabetes Mellitus, Type 1/physiopathology , Garlic/chemistry , Triglycerides/blood , Immunohistochemistry , Random Allocation , Cholesterol/blood , Reproducibility of Results , Treatment Outcome , Rats, Wistar , Platelet Endothelial Cell Adhesion Molecule-1/analysis , MicroRNAs/physiology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/therapy , Real-Time Polymerase Chain Reaction , Heart/physiopathology
8.
Arq. bras. cardiol ; 111(5): 738-746, Nov. 2018. graf
Article in English | LILACS | ID: biblio-973801

ABSTRACT

Abstract MiRNA (or microRNA) is a subclass of non-coding RNAs that is responsible for post-transcriptional gene regulation. It has approximately 22 nucleotides and regulates gene expression in plants and animals at the post-transcriptional level, by the cleavage of a target mRNA or by suppression of its translation. Although many of the processes and mechanisms have not yet been fully elucidated, there is a strong association between miRNA expression and several diseases. It is known that miRNAs are expressed in the cardiovascular system, but their role in cardiovascular diseases (CVDs) has not been clearly established. In this non-systematic review of the literature, we first present the definition of miRNAs and their action at the cellular level. Afterward, we discuss the role of miRNAs as circulating biomarkers of CVDs, and then their role in cardiac remodeling and atherosclerosis. Despite the complexity and challenges, it is crucial to identify deregulated miRNAs in CVDs, as it allows a better understanding of underlying cellular and molecular mechanisms and helps in the development of more accurate diagnostic and prognostic circulating biomarkers, and new therapeutic strategies for different stages of CVDs.


Resumo O miRNA (ou microRNA) constitui uma subclasse de RNAs não codificantes responsáveis pela regulação gênica pós-transcricional. Ele possui aproximadamente 22 nucleotídeos e regula a expressão gênica em plantas e animais ao nível pós-transcricional, pela clivagem de um mRNA alvo ou da repressão de sua tradução. Embora muitos processos e mecanismos ainda não estejam completamente elucidados, existe uma forte associação entre a expressão de miRNAs e diversas doenças que acometem o organismo. Os miRNAs são expressos no sistema cardiovascular, contudo o seu papel no desenvolvimento das doenças cardiovasculares (DCVs) ainda não está totalmente elucidado. Diante disso, realizou-se uma revisão não sistemática da literatura a fim de se discutir a relação entre os miRNAs e as DCVs. Nesta revisão, primeiramente é discutido o que são os miRNAs e a sua ação a nível celular. Após, é discutido o papel dos miRNAs como biomarcadores circulantes de DCVs e então o seu papel no remodelamento cardíaco e na aterosclerose. Apesar da complexidade e dos desafios, a identificação dos miRNAs desregulados nas DCVs é crucial, uma vez que possibilita uma melhor compressão dos mecanismos celulares e moleculares envolvidos, assim como auxilia o desenvolvimento de marcadores circulantes de diagnóstico e prognóstico mais acurados e de novas estratégias terapêuticas para os diferentes estágios da DCV.


Subject(s)
Humans , Cardiovascular Diseases/physiopathology , MicroRNAs/physiology , Biomarkers , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Gene Expression Regulation/genetics , Ventricular Remodeling/genetics , MicroRNAs/genetics , Atherosclerosis/physiopathology , Atherosclerosis/genetics , Atherosclerosis/metabolism
9.
Biol. Res ; 51: 50, 2018. graf
Article in English | LILACS | ID: biblio-1011394

ABSTRACT

BACKGROUND: Accumulating studies have demonstrated that high-mobility group A2 (HMGA2), an oncofetal protein, plays a role in tumor development and progression. However, the molecular role of HMGA2 in ovarian carcinoma is yet to be established. MicroRNAs (miRNAs), a group of small noncoding RNAs, negatively regulate gene expression and their dysregulation has been implicated in tumorigenesis. The aim of this study was to investigate the potential involvement of a specific miRNA, miR-219-5p, in HMGA2-induced ovarian cancer. METHODS: The ovarian cancer cell line, SKOV3, was employed, and miR-219-5p and HMGA2 overexpression vectors constructed. The CCK-8 kit was used to determine cell proliferation and the Transwell® assay used to measure cell invasion and migration. RT-PCR and western blot analyses were applied to analyze the expression of miR-219-5p and HMGA2, and the luciferase reporter assay used to examine the interactions between miR-219-5p and HMGA2. Nude mice were employed to characterize in vivo tumor growth regulation. RESULTS: Expression of miR-219-5p led to suppression of proliferation, invasion and migration of the ovarian cancer cell line, SKOV3, by targeting HMGA2. The inhibitory effects of miR-219-5p were reversed upon overexpression of HMGA2. Data from the luciferase reporter assay showed that miR-219-5p downregulates HMGA2 via direct integration with its 3'-UTR. Consistent with in vitro findings, expression of miR-219-5p led to significant inhibition of tumor growth in vivo. CONCLUSION: Our results collectively suggest that miR-219-5p inhibits tumor growth and metastasis by targeting HMGA2.


Subject(s)
Humans , Animals , Female , Mice , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , HMGA2 Protein/metabolism , MicroRNAs/physiology , Ovarian Neoplasms/genetics , Cell Movement/genetics , HMGA2 Protein/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Neoplasm Invasiveness , Neoplasm Metastasis
10.
Biol. Res ; 51: 51, 2018. tab, graf
Article in English | LILACS | ID: biblio-1011395

ABSTRACT

BACKGROUND: Emerging evidence showed that microRNAs (miRs) play critical roles in human cancers by functioning as either tumor suppressor or oncogene. MIR-382 was found to function as tumor suppressor in certain cancers. However, the role of MIR-382 in colorectal cancer (CRC) is largely unknown. Specificity protein 1 (SP1) is highly expressed in several cancers including CRC and is correlated with poor prognosis, but it is unclear whether or not MIR-382 can regulate the expression of SP1. METHODS: MIR-382 expression level was measured by reverse transcription-quantitative polymerase chain reaction. The connection between MIR-382 and SP1 was validated by luciferase activity reporter assay and western blot assay. Cell counting kit-8 assay and wound-healing assay were conducted to investigate the biological functions of MIR-382 in CRC. RESULTS: In this study, we found MIR-382 expression was downregulated in CRC tissues and cell lines, and the transfection of MIR-382 mimic decreased cell growth and migration. Furthermore, we identified SP1 was a direct target of MIR-382. Overexpression of MIR-382 decreased the expression of SP1, whereas MIR-382 knockdown promoted SP1 expression. We also observed an inversely correlation between MIR-382 and SP1 in CRC tissues. Additionally, we showed that knockdown of SP1 inhibited cell growth and migration and attenuated the effect of MIR-382 inhibitor on cell behaviors. CONCLUSIONS: In conclusion, the present study describes a potential mechanism underlying a MIR-382/SP1 link contributing to CRC development. Thus, MIR-382 may be able to be developed as a novel treatment target for CRC.


Subject(s)
Humans , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/genetics , Sp1 Transcription Factor/metabolism , MicroRNAs/physiology , Transfection , Colorectal Neoplasms/pathology , Down-Regulation , Cell Movement , Sp1 Transcription Factor/genetics , MicroRNAs/genetics , Cell Line, Tumor , Cell Proliferation , Neoplasm Invasiveness/genetics
11.
Biol. Res ; 51: 56, 2018. graf
Article in English | LILACS | ID: biblio-1011400

ABSTRACT

BACKGROUND: Glioma is the most prevalent malignant tumor in human central nervous systems. Recently, the development of resistance to radiotherapy in glioma patients markedly vitiates the therapy outcome. MiR-153-3p has been reported to be closely correlated with tumor progression, but its effect and molecular mechanism underlying radioresistance remains unclear in glioma. METHODS: The expression of miR-153-3p was determined in radioresistant glioma clinical specimens as well as glioma cell lines exposed to irradiation (IR) using quantitative real-time PCR. Cell viability, proliferation and apoptosis were then evaluated by MTT assay, colony formation assay, Flow cytometry analysis and caspase-3 activity assay in glioma cells (U87 and U251). Tumor forming was evaluated by nude mice model in vivo. TUNEL staining was used to detect cell apoptosis in nude mice model. The target genes of miR-153-3p were predicted and validated using integrated bioinformatics analysis and a luciferase reporter assay. RESULTS: Here, we found that miR-153-3p was down-regulated in radioresistant glioma clinical specimens as well as glioma cell lines (U87 and U251) exposed to IR. Enhanced expression of miR-153-3p promoted the radiosensitivity, promoted apoptosis and elevated caspase-3 activity in glioma cells in vitro, as well as the radiosensitivity in U251 cell mouse xenografs in vivo. Mechanically, B cell lymphoma-2 gene (BCL2) was identified as the direct and functional target of miR-153-3p. Moreover, restoration of BCL2 expression reversed miR-153-3p-induced increase of radiosensitivity, apoptosis and caspase-3 activity in U251 cells in vitro. In addition, clinical data indicated that the expression of miR-153-3p was significantly negatively associated with BCL2 in radioresistance of glioma samples. CONCLUSIONS: Our findings suggest that miR-153-3p is a potential target to enhance the effect of radiosensitivity on glioma cells, thus representing a new potential therapeutic target for glioma.


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Radiation Tolerance/genetics , Genes, bcl-2/physiology , MicroRNAs/radiation effects , MicroRNAs/physiology , Glioma/genetics , Time Factors , Down-Regulation , Gene Expression Regulation, Neoplastic , Cell Survival/radiation effects , Blotting, Western , Analysis of Variance , Gene Targeting/methods , Genes, bcl-2/radiation effects , In Situ Nick-End Labeling , MicroRNAs/analysis , Cell Line, Tumor , Cell Proliferation/radiation effects , Caspase 3/analysis , Real-Time Polymerase Chain Reaction , Flow Cytometry , Glioma/radiotherapy
12.
Braz. j. med. biol. res ; 51(8): e7566, 2018. tab, graf
Article in English | LILACS | ID: biblio-951742

ABSTRACT

Protein coding sequences represent only 2% of the human genome. Recent advances have demonstrated that a significant portion of the genome is actively transcribed as non-coding RNA molecules. These non-coding RNAs are emerging as key players in the regulation of biological processes, and act as "fine-tuners" of gene expression. Neurological disorders are caused by a wide range of genetic mutations, epigenetic and environmental factors, and the exact pathophysiology of many of these conditions is still unknown. It is currently recognized that dysregulations in the expression of non-coding RNAs are present in many neurological disorders and may be relevant in the mechanisms leading to disease. In addition, circulating non-coding RNAs are emerging as potential biomarkers with great potential impact in clinical practice. In this review, we discuss mainly the role of microRNAs and long non-coding RNAs in several neurological disorders, such as epilepsy, Huntington disease, fragile X-associated ataxia, spinocerebellar ataxias, amyotrophic lateral sclerosis (ALS), and pain. In addition, we give information about the conditions where microRNAs have demonstrated to be potential biomarkers such as in epilepsy, pain, and ALS.


Subject(s)
Humans , MicroRNAs/physiology , RNA, Long Noncoding/physiology , Nervous System Diseases/genetics , Genetic Markers/physiology , Gene Expression Regulation , Neurodegenerative Diseases/genetics , Circulating MicroRNA , Neuromuscular Diseases/genetics
13.
Braz. j. med. biol. res ; 51(12): e7574, 2018. graf
Article in English | LILACS | ID: biblio-974257

ABSTRACT

Bone fracture is a common medical condition, which may occur due to traumatic injury or disease-related conditions. Evidence suggests that microRNAs (miRNAs) can regulate osteoblast differentiation and function. In this study, we explored the effects and mechanism of miR-221 on the growth and migration of osteoblasts using MC3T3-E1 cells. The expression levels of miR-221 in the different groups were measured by qRT-PCR. Then, miR-221 mimic and inhibitor were transfected into MC3T3-E1 cells, and cell viability and migration were measured using the CCK-8 assay and the Transwell migration assay. Additionally, the expression levels of differentiation-related factors (Runx2 and Ocn) and ZFPM2 were measured by qRT-PCR. Western blot was used to measure the expression of cell cycle-related proteins, epithelial-mesenchymal transition (EMT)-related proteins, ZFPM2, and Wnt/Notch, and Smad signaling pathway proteins. miR-221 was significantly up-regulated in the patients with lumbar compression fracture (LCM) and trochanteric fracture (TF). miR-221 promoted ALP, Runx2, and OPN expressions in MC3T3-E1 cells. miR-221 overexpression significantly increased cell proliferation, migration, differentiation, and matrix mineralization, whereas suppression of miR-221 reversed these effects. Additionally, the results displayed that ZFPM2 was a direct target gene of miR-221, and overexpression of ZFPM2 reversed the promoting effects of miR-221 overexpression on osteoblasts. Mechanistic study revealed that overexpression of miR-221 inactivated the Wnt/Notch and Smad signaling pathways by regulating ZFPM2 expression. We drew the conclusions that miR-221 overexpression promoted osteoblast proliferation, migration, and differentiation by regulation of ZFPM2 expression and deactivating the Wnt/Notch and Smad signaling pathways.


Subject(s)
Humans , Animals , Rabbits , Cell Differentiation/physiology , Cell Movement/physiology , MicroRNAs/physiology , Cell Proliferation/physiology , DNA-Binding Proteins/physiology , Fractures, Bone/blood , Osteoblasts/physiology , Reference Values , Transcription Factors/blood , Cell Survival/physiology , Blotting, Western , Analysis of Variance , 3T3 Cells , MicroRNAs/blood , DNA-Binding Proteins/blood
14.
Braz. j. med. biol. res ; 51(11): e7655, 2018. tab, graf
Article in English | LILACS | ID: biblio-974247

ABSTRACT

Previous studies have indicated that propofol has immunomodulatory and antioxidative properties. However, the renoprotection effect and the precise mechanisms of propofol in sepsis-induced renal injury remain unclear. The purpose of the present study was to investigate the role of miR-290-5p/CCL-2 signaling in septic mice treatment with propofol. Mice were treated with propofol (50 mg/kg) twice within 24 h. Survival outcome was monitored within 48 h. The mRNA and protein levels were assayed by qRT-PCR and western blotting, respectively. Mouse podocytes (MPC5) were treated with lipopolysaccharide (LPS) to establish the cell model in vitro. The proliferation of MPC5 was monitored using the MTS assay. Cell apoptosis was analyzed by flow cytometry. Propofol improved survival outcome and alleviated acute kidney injury in cecal ligation and puncture-operated mice. Propofol increased miR-290-5p expression and decreased CCL-2 and inflammatory cytokines levels in the kidney for septic mice. We found that miR-290-5p was a direct regulator of CCL-2 in MPC5. Propofol could abrogate LPS-induced growth inhibition and apoptosis in MPC5. Meanwhile, propofol inhibited CCL-2 expression in LPS-treated MPC5, however, knockdown of miR-290-5p abrogated the inhibitory effect propofol on the mRNA and protein expressions of CCL-2. Propofol could serve as an effective therapeutic medication to suppress sepsis-induced renal injury in vivo and in vitro by regulating the miR-290-5p/CCL-2 signaling pathway.


Subject(s)
Animals , Male , Rabbits , Signal Transduction/drug effects , Propofol/pharmacology , Sepsis/complications , Chemokine CCL2/drug effects , MicroRNAs/drug effects , Acute Kidney Injury/prevention & control , Blotting, Western , Sepsis/metabolism , Chemokine CCL2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , MicroRNAs/physiology , Acute Kidney Injury/etiology , Flow Cytometry
15.
Int. braz. j. urol ; 43(6): 1060-1067, Nov.-Dec. 2017. graf
Article in English | LILACS | ID: biblio-892928

ABSTRACT

ABSTRACT Objective: miR-483-5p has been identified as a miRNA oncogene in certain cancers. However, its role in prostate cancer has not been sufficiently investigated. In this study, we investigated the role of miR-483-5p in prostate cancer and examined RBM5 regulation by miR-483-5p. Material and methods: Expression levels of miR-483-5p were determined by quantitative real-time PCR. The effect of miR-483-5p on proliferation was evaluated by MTT assay, cell invasion was evaluated by trans-well invasion assays, and target protein expression was determined by western blotting in LNCaP, DU-145, and PC-3 cells. Luciferase reporter plasmids were constructed to confirm the action of miR-483-5p on downstream target gene RBM5 in HEK-293T cells. Results: we observed that miR-483-5p was upregulated in prostate cancer cell lines and tissues. A miR-483-5p inhibitor inhibited prostate cancer cell growth and invasion in DU-145 and PC-3 cells. miR-483-5p directly bound to the 3' untranslated region (3'UTR) of RBM5 in HEK-293T cells. RBM5 overexpression inhibited prostate cancer cell growth and invasion in LNCaP cells. Enforced RBM5 expression alleviated miR-483-5p promotion of prostate cancer cell growth and invasion in LNCaP cells. Conclusion: The present study describes a potential mechanism underlying a miR-483-5p/RBM5 link that contributes to prostate cancer development.


Subject(s)
Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Cell Cycle Proteins/metabolism , Untranslated Regions/genetics , Tumor Suppressor Proteins/metabolism , MicroRNAs/physiology , Cell Proliferation/genetics , DNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Prostatic Neoplasms/mortality , Down-Regulation , Up-Regulation , RNA-Binding Proteins/metabolism , MicroRNAs/antagonists & inhibitors , Cell Line, Tumor , Neoplasm Invasiveness
16.
Rev. bras. cir. cardiovasc ; 32(3): 215-224, May-June 2017. tab, graf
Article in English | LILACS | ID: biblio-897914

ABSTRACT

Abstract Objective: Abdominal aortic aneurysm is an important cause of morbidity and mortality in the elderly. Currently, the only way to prevent rupture and death related to abdominal aortic aneurysms is through surgical intervention. Endovascular treatment is associated with less morbidity than conventional treatment. The formation of an aneurysm is a complex multifactorial process, involving destructive remodeling of the connective tissue around the affected segment of the aorta wall. MicroRNAs are small sequences of non-coding RNAs that control diverse cellular functions by promoting degradation or inhibition of translation of specific mRNAs. A profile aberrant expression of miRNAs has been linked to human diseases, including cardiovascular dysfunction.


Subject(s)
Humans , Aorta, Abdominal/physiopathology , Aortic Aneurysm, Abdominal/etiology , Aortic Aneurysm, Abdominal/physiopathology , MicroRNAs/physiology , Sex Factors , Risk Factors , Aortic Aneurysm, Abdominal/therapy , Medical Illustration
17.
Braz. j. med. biol. res ; 50(2): e5760, 2017. graf
Article in English | LILACS | ID: biblio-839255

ABSTRACT

Cardiomyocyte apoptosis plays key roles in the pathogenesis of heart diseases such as myocardial infarction. MicroRNAs are important regulators of gene expression, which are also involved in the regulation of cardiomyocyte apoptosis. However, cardiomyocyte apoptosis regulated by microRNA (miR)-122 is largely unexplored. The aim of this study focused on the role of miR-122 in cardiomyocyte apoptosis. Cardiomyocytes were isolated from neonatal mice and primarily cultured. MiR-122 mimic and inhibitor were transfected to cardiomyocytes and verified by qRT-PCR. Cell viability and apoptosis post-transfection were assessed by MTT assay and flow cytometry, respectively. Changes in expression of caspase-8 were quantified by qRT-PCR and western blot. Results showed that miR-122 mimic and inhibitor successfully induced changes in miR-122 levels in cultured cardiomyocytes (P<0.01). MiR-122 overexpression suppressed viability and promoted apoptosis of cardiomyocytes (P<0.05), and miR-122 knockdown promoted cell viability and inhibited apoptosis (P<0.05). The mRNA and protein levels of caspase-8 were elevated by miR-122 overexpression (P<0.01) and reduced by miR-122 knockdown (P<0.001). These results suggest an inductive role of miR-122 in cardiomyocyte apoptosis, which may be related to its regulation on caspase-8.


Subject(s)
Animals , Mice , Apoptosis/genetics , Caspase 8/genetics , Gene Expression/genetics , MicroRNAs/genetics , MicroRNAs/physiology , Myocytes, Cardiac/pathology , Animals, Newborn , Gene Expression/physiology , Mice, Inbred BALB C
18.
Braz. j. med. biol. res ; 50(5): e6359, 2017. graf
Article in English | LILACS | ID: biblio-839294

ABSTRACT

MicroRNAs (miRNAs) play an important role in drug resistance and modulate the efficiency of chemotherapy. A recent study indicated that miR-340 functions as a tumor suppressor in various types of cancer. However, the role of miR-340 in chemotherapy has not been reported yet. In this study, we found that miR-340 enhanced cisplatin (CDDP)-induced cell death. Induction of miR-340-5p expression decreased the IC50 of CDDP and increased the apoptosis of CDDP-resistant MG-63 and Saos-2 cells. Moreover, miR-340-5p decreased the accumulation of MRP1 and MDR1. We further explored the mechanism underlying the promoting effects of miR-340-5p on CDDP-induced cell death. We identified a potential target of miR-340 in the 3′ untranslated region of lysophosphatidic acid acyltransferase (LPAATβ) using the online program Targetscan (http://www.microrna.org). Luciferase reporter assays showed that miR-340 binds to the 3′UTR of LPAATβ. Enforced expression of miR-340-5p decreased the accumulation of LPAATβ in both MG-63 and Saos-2 cells. Silencing LPAATβ decreased the IC50 of CDDP and increased the apoptosis of CDDP-resistant MG-63 and Saos-2 cells, which is consistent with the effect of miR-340-5p on CDDP-induced cell death. Moreover, induced expression of LPAATβ compromised the effects of miR-340-5p on CDDP-induced cell death and accumulation of MRP1 and MDR1. Taken together, our data indicated that miR-340-5p enhanced the sensitivity to CDDP by targeting LPAATβ.


Subject(s)
Humans , Acyltransferases/physiology , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Cisplatin/pharmacology , Drug Resistance, Neoplasm/physiology , MicroRNAs/physiology , Osteosarcoma/drug therapy , Acyltransferases/analysis , Acyltransferases/drug effects , Apoptosis/drug effects , Blotting, Western , Bone Neoplasms/physiopathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Luciferases , MicroRNAs/analysis , MicroRNAs/drug effects , Osteosarcoma/physiopathology , Real-Time Polymerase Chain Reaction
19.
Yonsei Medical Journal ; : 234-240, 2017.
Article in English | WPRIM | ID: wpr-126252

ABSTRACT

PURPOSE: MicroRNAs are small non-coding RNAs that play important roles in vascular smooth muscle cell (VSMC) function. This study investigated the role of miR-379 on proliferation, invasion, and migration of VSMCs and explored underlying mechanisms thereof. MATERIALS AND METHODS: MicroRNA, mRNA, and protein levels were determined by quantitative real-time PCR and western blot. The proliferative, invasive, and migratory abilities of VSMCs were measured by CCK-8, invasion, and wound healing assay, respectively. Luciferase reporter assay was used to confirm the target of miR-379. RESULTS: Platelet-derived growth factor-bb was found to promote cell proliferation and suppress miR-379 expression in VSMCs. Functional assays demonstrated that miR-379 inhibited cell proliferation, cell invasion, and migration. Flow cytometry results further showed that miR-379 induced apoptosis in VSMCs. TargetScan analysis and luciferase report assay confirmed that insulin-like growth factor-1 (IGF-1) 3'UTR is a direct target of miR-379, and mRNA and protein levels of miR-379 and IGF-1 were inversely correlated. Rescue experiments showed that enforced expression of IGF-1 sufficiently overcomes the inhibitory effect of miR-379 on cell proliferation, invasion, and migration in VSMCs. CONCLUSION: Our results suggest that miR-379 plays an important role in regulating VSMCs proliferation, invasion, and migration by targeting IGF-1.


Subject(s)
Humans , Apoptosis , Cell Movement/physiology , Cell Proliferation/physiology , Insulin , Insulin-Like Growth Factor I/physiology , MicroRNAs/physiology , Muscle, Smooth, Vascular/cytology , Proto-Oncogene Proteins c-sis/physiology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Sincalide/physiology , Wound Healing/physiology
20.
Actual. osteol ; 12(2): 126-135, 2016. ilus
Article in English | LILACS, UNISALUD, BINACIS | ID: biblio-1372418

ABSTRACT

Coronary heart disease, a leading cause of death in western societies, is caused by the presence of atherosclerotic plaques in the coronary arteries. Calcification is a frequent complication of atherosclerotic plaques, and often a contributing factor to their instability and rupture. Endothelial cells, smooth muscle cells and plaque macrophages, all contribute to the calcification process, which is reminiscent of that underlying bone formation. In particular, the role of macrophages in calcification has long been recognized, but whether or not distinct macrophage subsets ­v.g., M1 or inflammatory, and M2 or antinflammatory have specific functions in osteogenic signaling within the context of plaque calcification remains poorly understood. Over the past few years, accumulated evidence has revealed novel roles of non-coding micro-RNAs (miRs) in atherorelevant functions of macrophages and in mechanisms linked to macrophage divergence into different subtypes. In this article we discuss some salient findings on potential roles of miRs in vascular calcification, with focus on those miRs that have also been associated to macrophage differentiation, and speculate on their potential relation to M1 and M2 macrophages in the context of calcification of atherosclerotic plaques. (AU)


La enfermedad cardíaca coronaria, principal causa de muerte en occidente, es causada por la presencia de placas ateroscleróticas en las arterias coronarias. La presencia de depósitos de calcificación es una complicación frecuente de la placa, y puede contribuir a la inestabilidad y ruptura de la misma. El proceso de calcificación de la placa es similar al que ocurre en hueso, y contribuyen al mismo, mecanismos dependientes de células endoteliales, células musculares lisas y macrófagos, células que están presentes en todas las etapas de desarrollo de la placa aterosclerótica. El rol de los macrófagos en la calcificación de la placa se conoce desde hace tiempo, pero la contribución de los distintos tipos de macrófagos ­por ejemplo, M1 o tipo inflamatorio, y M2 o tipo antiinflamatorio a mecanismos de señalización osteogénica en dicho contexto aún no se conoce. Recientemente varios trabajos experimentales han revelado la existencia de nuevos roles de micro-ARNs no codificantes (miRs) en varias funciones de los macrófagos que son de relevancia en el proceso aterogénico, como así también en mecanismos relacionados a la diferenciación de macrófagos en subtipos específicos. En este artículo discutimos algunos de los hallazgos más importantes sobre posibles nuevos roles de miRs en calcificación vascular, poniendo énfasis en aquellos miRs que han sido también asociados a la diferenciación de macrófagos, y especulamos acerca de su posible relación con macrófagos M1 y M2 en el contexto de la calcificación de la placa aterosclerótica. (AU)


Subject(s)
Humans , MicroRNAs/physiology , Plaque, Atherosclerotic/classification , Plaque, Atherosclerotic/physiopathology , Vascular Calcification/physiopathology , Macrophages/physiology , Osteogenesis/physiology , Atherosclerosis/complications , Vascular Calcification/prevention & control , Macrophages/classification
SELECTION OF CITATIONS
SEARCH DETAIL