Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Technol Cancer Res Treat ; 22: 15330338231164359, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36938678

RESUMEN

Background: Emerging evidence suggests that long noncoding RNAs (lncRNAs) play an important role in the progression of multiple human cancers including breast cancer. In this study, we aimed to research novel functions of long intergenic noncoding RNA 460 (LINC00460) on cell proliferation and ferroptosis in breast cancer. Method: UALCAN, TANRIC, and GSE16446 data were used to analyze the expression of LINC00460 in breast cancer tissues. Furthermore, real-time quantitative PCR, western blot, cell proliferation assay, iron assay, and malondialdehyde (MDA) assay were applied to detect the function and mechanism of particular molecules. Results: The LINC00460 expression was significantly increased in breast cancer tissues compared with normal tissues. Importantly, patients with high LINC00460 expression showed a longer overall survival rate. LINC00460 knockdown markedly suppressed the proliferation of breast cancer cells and promoted ferroptosis. Mechanistic analysis revealed that LINC00460 promoted myelin and lymphocyte protein 2 (MAL2) expression by sponging miR-320a. Moreover, both miR-320a knockdown and MAL2 overexpression could reverse the effects of LINC00460 silencing on cell proliferation and ferroptosis. Conclusions: In summary, our results reveal an alternative mechanism by which breast cancer cells can acquire resistance to ferroptosis via the LINC00460/miR-320a/MAL2 axis.


Asunto(s)
Neoplasias de la Mama , Ferroptosis , MicroARNs , ARN Largo no Codificante , Femenino , Humanos , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/genética , Ferroptosis/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/genética , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba
2.
Acta Pharmacol Sin ; 44(8): 1612-1624, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36747104

RESUMEN

Suprachiasmatic nucleus (SCN) in mammals functions as the master circadian pacemaker that coordinates temporal organization of physiological processes with the environmental light/dark cycles. But the causative links between SCN and cardiovascular diseases, specifically the reparative responses after myocardial infarction (MI), remain largely unknown. In this study we disrupted mouse SCN function to investigate the role of SCN in cardiac dysfunction post-MI. Bilateral ablation of the SCN (SCNx) was generated in mice by electrical lesion; myocardial infarction was induced via ligation of the mid-left anterior descending artery (LAD); cardiac function was assessed using echocardiography. We showed that SCN ablation significantly alleviated MI-induced cardiac dysfunction and cardiac fibrosis, and promoted angiogenesis. RNA sequencing revealed differentially expressed genes in the heart of SCNx mice from D0 to D3 post-MI, which were functionally associated with the inflammatory response and cytokine-cytokine receptor interaction. Notably, the expression levels of insulin-like growth factor 2 (Igf2) in the heart and serum IGF2 concentration were significantly elevated in SCNx mice on D3 post-MI. Stimulation of murine peritoneal macrophages in vitro with serum isolated from SCNx mice on D3 post-MI accelerated the transition of anti-inflammatory macrophages, while antibody-mediated neutralization of IGF2 receptor blocked the macrophage transition toward the anti-inflammatory phenotype in vitro as well as the corresponding cardioprotective effects observed in SCNx mice post-MI. In addition, disruption of mouse SCN function by exposure to a desynchronizing condition (constant light) caused similar protective effects accompanied by elevated IGF2 expression on D3 post-MI. Finally, mice deficient in the circadian core clock genes (Ckm-cre; Bmal1f/f mice or Per1/2 double knockout) did not lead to increased serum IGF2 concentration and showed no protective roles in post-MI, suggesting that the cardioprotective effect observed in this study was mediated particularly by the SCN itself, but not by self-sustained molecular clock. Together, we demonstrate that inhibition of SCN function promotes Igf2 expression, which leads to macrophage transition and improves cardiac repair post-MI.


Asunto(s)
Ritmo Circadiano , Infarto del Miocardio , Animales , Ratones , Ritmo Circadiano/genética , Macrófagos , Mamíferos , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Núcleo Supraquiasmático/metabolismo
3.
Curr Probl Cardiol ; 48(5): 101570, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36584729

RESUMEN

Resident macrophages (R-mac) are a subset of macrophages with self-renewal functions, which play a pivotal role in the homeostasis, inflammation, injury, and repair of the heart. In this paper, we summarize the knowledge related to cardiac R-mac and describe their dominating functions in myocardial infarction, such as inhibiting fibrosis and adverse remodeling, promoting revascularization and improving arrhythmia, etc. In the last, we sketch out the extended application of R-mac in tissue engineering, providing a novel direction of research and application for the therapy in the future.


Asunto(s)
Infarto del Miocardio , Humanos , Infarto del Miocardio/terapia , Corazón , Macrófagos , Miocardio
4.
J Headache Pain ; 23(1): 6, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35032999

RESUMEN

BACKGROUND: Migraine is a common and disabling primary headache, which is associated with a wide range of psychiatric comorbidities. However, the mechanisms of emotion processing in migraine are not fully understood yet. The present study aimed to investigate the neural network during neutral, positive, and negative emotional stimuli in the migraine patients. METHODS: A total of 24 migraine patients and 24 age- and sex-matching healthy controls were enrolled in this study. Neuromagnetic brain activity was recorded using a whole-head magnetoencephalography (MEG) system upon exposure to human facial expression stimuli. MEG data were analyzed in multi-frequency ranges from 1 to 100 Hz. RESULTS: The migraine patients exhibited a significant enhancement in the effective connectivity from the prefrontal lobe to the temporal cortex during the negative emotional stimuli in the gamma frequency (30-90 Hz). Graph theory analysis revealed that the migraine patients had an increased degree and clustering coefficient of connectivity in the delta frequency range (1-4 Hz) upon exposure to positive emotional stimuli and an increased degree of connectivity in the delta frequency range (1-4 Hz) upon exposure to negative emotional stimuli. Clinical correlation analysis showed that the history, attack frequency, duration, and neuropsychological scales of the migraine patients had a negative correlation with the network parameters in certain frequency ranges. CONCLUSIONS: The results suggested that the individuals with migraine showed deviant effective connectivity in viewing the human facial expressions in multi-frequencies. The prefrontal-temporal pathway might be related to the altered negative emotional modulation in migraine. These findings suggested that migraine might be characterized by more universal altered cerebral processing of negative stimuli. Since the significant result in this study was frequency-specific, more independent replicative studies are needed to confirm these results, and to elucidate the neurocircuitry underlying the association between migraine and emotional conditions.


Asunto(s)
Magnetoencefalografía , Trastornos Migrañosos , Mapeo Encefálico , Emociones , Cefalea , Humanos , Imagen por Resonancia Magnética , Corteza Prefrontal
5.
J Cell Mol Med ; 25(17): 8103-8114, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34378345

RESUMEN

Transplantation of stem cells is a promising, emerging treatment for cardiovascular diseases in the modern era. Mesenchymal stem cells (MSCs) derived from the umbilical cord are one of the most promising cell sources because of their capacity for differentiation into cardiomyocytes, endothelial cells and vascular smooth muscle cells in vitro/in vivo. In addition, umbilical cord-derived MSCs (UC-MSCs) secrete many effective molecules regulating apoptosis, fibrosis and neovascularization. Another important and specific characteristic of UC-MSCs is their low immunogenicity and immunomodulatory properties. However, the application of UC-MSCs still faces some challenges, such as low survivability and tissue retention in a harmful disease environment. Gene engineering and pharmacological studies have been implemented to overcome these difficulties. In this review, we summarize the differentiation ability, secretion function, immunoregulatory properties and preclinical/clinical studies of UC-MSCs, highlighting the advantages of UC-MSCs for the treatment of cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas , Cordón Umbilical , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Miocitos Cardíacos/citología , Cordón Umbilical/citología , Cordón Umbilical/metabolismo
6.
Bioact Mater ; 6(2): 520-528, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32995677

RESUMEN

Myocardial infarction (MI) is one of cardiovascular diseases that pose a serious threat to human health. The pathophysiology of MI is complex and contains several sequential phases including blockage of a coronary artery, necrosis of myocardial cells, inflammation, and myocardial fibrosis. Aiming at the treatment of different stages of MI, in this work, an injectable alginate based composite hydrogel is developed to load vascular endothelial active factor (VEGF) and silk fibroin (SF) microspheres containing bone morphogenetic protein 9 (BMP9) for releasing VEGF and BMP9 to realize their respective functions. The results of in vitro experiments indicate a rapid initial release of VEGF during the first few days and a relatively slow and sustained release of BMP9 for days, facilitating the formation of blood vessels in the early stage and inhibiting myocardial fibrosis in the long-term stage, respectively. Intramyocardial injection of such composite hydrogel into the infarct border zone of mice MI model via multiple points promotes angiogenesis and reduces the infarction size. Taken together, these results indicate that the dual-release of VEGF and BMP9 from the composite hydrogel results in a collaborative effect on the treatment of MI and improvement of heart function, showing a promising potential for cardiac clinical application.

7.
J Cell Mol Med ; 24(23): 13775-13788, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33084169

RESUMEN

Myocardial infarction (MI) results in cardiomyocyte death and ultimately leads to heart failure. Pyroptosis is a type of the inflammatory programmed cell death that has been found in various diseased tissues. However, the role of pyroptosis in MI heart remains unknown. Here, we showed that CXADR-like membrane protein (CLMP) was involved in pyroptosis in the mouse MI heart. Our data showed that CLMP was strongly expressed in fibroblasts of the infarcted mouse hearts. The Clmp+/- mice showed more serious myocardial fibrosis and ventricular dysfunction post-MI than wild-type (Clmp+/+ ) mice, indicating a protective effect of the fibroblast-expressed CLMP against MI-induced heart damage. Transcriptome analyses by RNA sequencing indicated that Il-1ß mRNA was significantly increased in the MI heart of Clmp+/- mouse, which indicated a more serious inflammatory response. Meanwhile, cleaved caspase-1 and Gasdermin D were significantly increased in the Clmp+/- MI heart, which demonstrated enhanced pyroptosis in the Clmp knockdown heart. Further analysis revealed that the pyroptosis mainly occurred in cardiac fibroblasts (CFs). Compared to wild-type fibroblasts, Clmp+/- CFs showed more serious pyroptosis and inflammatory after LPS plus nigericin treatment. Collectively, our results indicate that CLMP participates in the pyroptotic and inflammatory response of CFs in MI heart. We have provided a novel pyroptotic insight into the ischaemic heart, which might hold substantial potential for the treatment of MI.


Asunto(s)
Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Piroptosis/genética , Animales , Biomarcadores , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Ecocardiografía , Fibroblastos/metabolismo , Expresión Génica , Genotipo , Inmunohistoquímica , Mediadores de Inflamación/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Mutación , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/etiología , Fenotipo
8.
Stem Cell Res Ther ; 11(1): 373, 2020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32859268

RESUMEN

BACKGROUND: Myocardial infarction (MI) is a severe disease that often associated with dysfunction of angiogenesis. Cell-based therapies for MI using mesenchymal stem cell (MSC)-derived exosomes have been well studied due to their strong proangiogenic effect. Genetic modification is one of the most common methods to enhance exosome therapy. This study investigated the proangiogenic and cardioprotective effect of exosomes derived from hypoxia-inducible factor 1-alpha (HIF-1α)-modified MSCs. METHODS: Lentivirus containing HIF-1α overexpressing vector was packaged and used to infect MSCs. Exosomes were isolated from MSC-conditioned medium by ultracentrifugation. Human umbilical vein endothelial cells (HUVECs) were treated under hypoxia condition for 48 h co-cultured with PBS, control exosomes, or HIF-1α-overexpressed exosomes, respectively. Then the preconditioned HUVECs were subjected to tube formation assay, Transwell assay, and EdU assay to evaluate the protective effect of exosomes. Meanwhile, mRNA and secretion levels of proangiogenic factors were measured by RT-qPCR and ELISA assays. In vivo assays were conducted using the rat myocardial infarction model. PBS, control exosomes, or HIF-1α-overexpressed exosomes were injected through tail vein after MI surgery. Heart function was assessed by echocardiography at days 3, 14, and 28. At day 7, mRNA and protein expression levels of proangiogenic factors in the peri-infarction area and circulation were evaluated, respectively. At day 28, hearts were collected and subjected to H&E staining, Masson's trichrome staining, and immunofluorescent staining. RESULTS: HIF-1α-overexpressed exosomes rescued the impaired angiogenic ability, migratory function, and proliferation of hypoxia-injured HUVECs. Simultaneously, HIF-1α-overexpressed exosomes preserved heart function by promoting neovessel formation and inhibiting fibrosis in the rat MI model. In addition, both in vitro and in vivo proangiogenic factors mRNA and protein expression levels were elevated after HIF-1α-overexpressed exosome application. CONCLUSION: HIF-1α-overexpressed exosomes could rescue the impaired angiogenic ability, migration, and proliferation of hypoxia-pretreated HUVECs in vitro and mediate cardioprotection by upregulating proangiogenic factors and enhancing neovessel formation.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Infarto del Miocardio , Animales , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Neovascularización Patológica , Neovascularización Fisiológica , Ratas
9.
Cell Death Dis ; 11(5): 349, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32393744

RESUMEN

Mesenchymal stem cells (MSCs) are derived from a wide range of sources and easily isolated and cultured. MSCs have the capacity for in vitro amplification and self-renewal, low immunogenicity and immunomodulatory properties, and under certain conditions, MSCs can be differentiated into a variety of cells. In the cardiovascular system, MSCs can protect the myocardium by reducing the level of inflammation, promoting the differentiation of myocardial cells around infarct areas and angiogenesis, increasing apoptosis resistance, and inhibiting fibrosis, which are ideal qualities for cardiovascular repair. Preclinical studies have shown that MSCs can be transplanted and improve cardiac repair, but challenges, such as their low rate of migration to the ischemic myocardium, low tissue retention, and low survival rate after transplantation, remain. This article reviews the potential and methods of MSC transplantation in the treatment of cardiovascular diseases (CVDs) and the challenges of the clinical use of MSCs.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Animales , Enfermedades Cardiovasculares/patología , Diferenciación Celular , Ensayos Clínicos como Asunto , Humanos , Inmunomodulación , Neovascularización Fisiológica
10.
Oxid Med Cell Longev ; 2020: 7242836, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32148656

RESUMEN

Bone marrow-derived mesenchymal stem cells (MSCs) have shown great promise in tissue engineering and regenerative medicine; however, the regenerative capacity of senescent MSCs is greatly reduced, thus exhibiting limited therapy potential. Previous studies uncovered that microRNA-206 (miR-206) could largely regulate cell functions, including cell proliferation, survival, and apoptosis, but whether miR-206 is involved in the senescent process of MSCs remains unknown. In this study, we mainly elucidated the effects of miR-206 on MSC senescence and the underlying mechanism. We discovered that miR-206 was upregulated in the senescent MSCs induced by H2O2, and abrogation of miR-206 could alleviate this tendency. Besides, we determined that by targeting Alpl, miR-206 could ameliorate the impaired migration and paracrine function in MSCs reduced by H2O2. In vivo study, we revealed that inhibition of miR-206 in senescent MSCs could effectively protect their potential for myocardial infarction treatment in a rat MI model. In summary, we examined that inhibition of miR-206 in MSCs can alleviate H2O2-induced senescence and dysfunction, thus protecting its therapeutic potential.


Asunto(s)
Fosfatasa Alcalina/genética , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Animales , Senescencia Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Humanos , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Transfección
11.
J Tissue Eng Regen Med ; 14(4): 588-599, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32068957

RESUMEN

Bone marrow-derived mesenchymal stromal cells (MSCs) have been wildly applied to cell-based strategies for tissue engineering and regenerative medicine; however, they have to undergo the senescence process and thus appeared to be less therapeutic effective. HMGA2, a protein belonged to high mobility group A (HMGA) family, exhibits an inverse expression level related to embryonic development and acts as a developmental regulator in stem cell self-renewal progression. Therefore, we performed senescence-associated ß-galactosidase (SA-ß-gal) staining, transwell assay, to examine the changes of MSCs in different stages and then over-expressed HMGA2 in MSCs by lentivirus transfection. We found the percentage of SA-ß-gal staining positive cells in MSCs from 24-month-old Sprague-Dawley (SD) rats (O-MSCs) was significantly higher compared with MSCs from 2-week-old SD rats (Y-MSCs), and the expression levels of P21 and P53, two senescence-related molecules, were also significantly up-regulated in O-MSCs than in Y-MSCs. In contrast, the HMGA2 expression level in O-MSCs was dramatically down-regulated in contrast to Y-MSCs. In additional, the migration ability in O-MSCs was significantly attenuated than in Y-MSCs. After successfully over-expressed HMGA2 in O-MSCs, the percentage of SA-ß-gal staining positive cells and the expression levels of P21 and P53 were reduced, and the migration ability was improved compared with O-MSCs without treatment. Further, mRNA sequencing analysis revealed that overexpression of HMGA2 changed the expression of genes related to cell proliferation and senescence, such as Lyz2, Pf4, Rgs2, and Mstn. Knockdown of Rgs2 in HMGA2 overexpression O-MSCs could antagonize the protective effect of HMGA2 in the senescence process of O-MSCs.


Asunto(s)
Células de la Médula Ósea/metabolismo , Senescencia Celular , Proteína HMGA2/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Células de la Médula Ósea/citología , Proteína HMGA2/genética , Células Madre Mesenquimatosas/citología , Ratas , Ratas Sprague-Dawley
12.
Mol Immunol ; 117: 84-93, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31743856

RESUMEN

Inflammatory bowel disease (IBD) is an autoimmune disease characterized by an abnormal immune response. Fibrinogen-like protein 2 (FGL2) is known to have immunoregulatory and anti-inflammatory activity. The level of FGL2 is elevated in patients with IBD; however, its comprehensive function in IBD is almost unknown. In our study, we explored the effect of FGL2 on dextran sulfate sodium (DSS)-induced colitis in mice and on NF-κB signaling in intestinal epithelial cells (IECs) and lamina propria dendritic cells (LPDCs). We founded that FGL2-/- mice in the colitis model showed more severe colitis manifestations than WT mice did, including weight loss, disease activity index (DAI), and colon histological scores. FGL2-/- mice treated with DSS produced more proinflammatory cytokines (IL-1ß, IL-6, TNF-α) in serum than WT mice did and demonstrated upregulated expression of TNF-α and inflammatory marker enzymes, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (Cox-2) in the colon tissue. Our data suggested that DSS-treated FGL2-/- mice showed stronger activation of NF-κB signaling, especially in IECs. Next, we demonstrated that recombinant FGL2 (rFGL2) inhibited the production of proinflammatory cytokines and the expression of inflammatory marker enzymes by downregulating the NF-κB signaling in HT-29 cells. Finally, we discovered that LPDCs from the colon of DSS-treated FGL2-/- mice showed significantly upregulated expression of surface maturation co-stimulatory molecules, including CD80, CD86, CD40, and MHC class II molecules compared with that in WT mice. In addition, LPDCs in FGL2-/- treated with DSS exhibited excessive NF-κB activity and the administration of rFGL2 to FGL2-/- mice could rescue the aggravated results of FGL2-/- mice. Taken together, our findings demonstrated that FGL2 might be a target for further therapy of IBD.


Asunto(s)
Colitis/inmunología , Células Dendríticas/inmunología , Fibrinógeno/inmunología , Mucosa Intestinal/inmunología , Transducción de Señal/inmunología , Animales , Colitis/inducido químicamente , Colitis/metabolismo , Sulfato de Dextran/toxicidad , Fibrinógeno/metabolismo , Quinasa I-kappa B/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/inmunología , FN-kappa B/inmunología
13.
J Mater Chem B ; 8(2): 308-315, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31808500

RESUMEN

Low circulating levels of insulin-like growth factor 1 (IGF-1) have been correlated with an increased risk for cardiovascular diseases in humans. In this work, an injectable alginate hydrogel containing silk fibroin (SF) microspheres with the capability to sustain the release of IGF-1 was prepared to induce myocardial repair after myocardial infarction (MI). First, IGF-1 was physically adsorbed onto SF microspheres prepared by the coaxial needle system, and these IGF-1-containing microspheres were subsequently encapsulated into sodium alginate solutions at different concentrations (1.0-2.5%). Finally, this solution was crosslinked with 0.68% calcium gluconate solution to prepare the composite injectable hydrogel. The composite hydrogel prepared using a sodium alginate solution at a concentration of 1.5% could promote proliferation of H9C2 cardiomyocytes and reduce the cellular apoptosis rate under hypoxic conditions. The enzyme-linked immunosorbent assay results indicated that SF microspheres as microcarriers could effectively enhance the sustained release of IGF-1 from the hydrogels, causing the composite hydrogel to possess a better sustained release ability than the system without the SF microspheres. Moreover, echocardiography, hematoxylin-eosin staining, and Masson trichrome staining results indicated that an intramyocardial injection of the composite hydrogel into the peripheral region of a MI rat model could reduce the infarct size and improve the cardiac function after 28 days. The applications of such a composite hydrogel may comprise a powerful platform in cardiac tissue engineering.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Preparaciones de Acción Retardada/farmacología , Portadores de Fármacos/síntesis química , Corazón/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina , Infarto del Miocardio/tratamiento farmacológico , Alginatos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Femenino , Fibroínas/uso terapéutico , Hidrogeles/uso terapéutico , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Microesferas , Ratas , Ratas Sprague-Dawley
14.
Stem Cell Res Ther ; 10(1): 295, 2019 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-31547872

RESUMEN

BACKGROUND: Interleukin 33 is known to have an important influence in the process of myocardial infarction, and the immunoregulatory function of MSCs could be influenced by cell factors. In this study, we evaluated the therapeutic efficacy of IL-33-overexpressing bone marrow mesenchymal stem cells (IL33-MSCs) on myocardial infarction (MI) and detected the inflammatory level and cardiac function in rats. METHODS AND RESULTS: First, we evaluated the proliferation of T cells and polarization of macrophages that had been co-cultured with Vector-MSCs or IL33-MSCs. Co-culture experiments indicated that IL33-MSCs reduced T cell proliferation and enhanced CD206+ macrophage polarization. Second, we determined the inflammation level and cardiac function of PBS-, Vector-MSC-, and IL33-MSC-injected rats. Echocardiography indicated that left ventricular ejection fraction (LVEF) was enhanced in IL33-MSC-injected rats compared with Vector-MSC-injected rats. Postmortem analysis of rat heart tissue showed reduced fibrosis and less inflammation in IL33-MSC-injected rats. CONCLUSION: These studies indicated that the IL33-MSC injection improved heart function and reduces inflammation in rats with MI compared with PBS or Vector-MSC injections. IL-33 overexpression enhances the immunomodulatory function and therapeutic effects of MSCs on acute MI via enhancing the polarization of macrophages toward M2, enhancing the differentiation of CD4+ T cells toward CD4+IL4+Th2 cells, and finally, reducing heart inflammation and enhancing heart function.


Asunto(s)
Interleucina-33/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Animales , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Interleucina-33/genética , Macrófagos/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley , Linfocitos T/efectos de los fármacos
15.
J Headache Pain ; 20(1): 3, 2019 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-30626318

RESUMEN

BACKGROUND: Although altered neural networks have been demonstrated in recent MEG (magnetoencephalography) research in migraine patients during resting state, it is unknown whether this alteration can be detected in task-related networks. The present study aimed to investigate the abnormalities of the frequency-specific somatosensory-related network in migraine patients by using MEG. METHODS: Twenty-two migraineurs in the interictal phase and twenty-two sex- and age-matched healthy volunteers were studied using a whole-head magnetoencephalography (MEG) system. Electrical stimuli were delivered alternately to the median nerve on the right wrists of all subjects. MEG data were analyzed in a frequency range of 1-1000 Hz in multiple bands. RESULTS: The brain network patterns revealed that the patients with migraine exhibited remarkably increased functional connectivity in the high-frequency (250-1000 Hz) band between the sensory cortex and the frontal lobe. The results of quantitative analysis of graph theory showed that the patients had (1) an increased degree of connectivity in the theta (4-8 Hz), beta (13-30 Hz) and gamma (30-80 Hz) bands; (2) an increased connectivity strength in the beta (13-30 Hz) and gamma (30-80 Hz) bands; (3) an increased path length in the beta (13-30 Hz), gamma (30-80 Hz) and ripple (80-250 Hz) bands; and (4) an increased clustering coefficient in the theta (4-8 Hz), beta (13-30 Hz) and gamma (30-80 Hz) bands. CONCLUSIONS: The results indicate that migraine is associated with aberrant connections from the somatosensory cortex to the frontal lobe. The frequency-specific increases in connectivity in terms of strength, path length and clustering coefficients support the notion that migraineurs have elevated cortical networks. This alteration in functional connectivity may be involved in somatosensory processing in migraine patients and may contribute to understanding migraine pathophysiology and to providing convincing evidence for a spatially targeted migraine therapy.


Asunto(s)
Encéfalo/fisiopatología , Trastornos Migrañosos/fisiopatología , Red Nerviosa/fisiopatología , Adulto , Mapeo Encefálico/métodos , Femenino , Humanos , Magnetoencefalografía , Masculino , Corteza Somatosensorial/fisiopatología , Adulto Joven
16.
Hum Gene Ther ; 30(3): 286-301, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30101604

RESUMEN

Follistatin-like 1 (Fstl1) protects cardiomyocytes from a broad spectrum of pathologic injuries including myocardial infarction (MI). It is worthy of note that although cardiac Fstl1 is elevated in post-MI microenvironment, its cardioprotective role is still restricted to a limited extent considering the frequency and severity of adverse cardiac remodeling following MI. We therefore propose that intrinsic Fstl1-suppressing microRNA (miRNA) may exist in the heart and its neutralization may further facilitate post-MI recovery. Here, miR-9-5p is predicted as one of the potential Fstl1-targeting miRNAs whose expression is decreased in ischemic myocardium and reversely correlated with Fstl1. Luciferase activity assay further validated Fstl1 as a direct target of miR-9-5p. In addition, forced expression of miR-9-5p in H9c2 cells is concurrent with diminished expression of Fstl1 and vice versa. Importantly, transfection of miR-9-5p mimics in hypoxic H9c2 cells exacerbates cardiac cell death, lactate dehydrogenase release, reactive oxygen species accumulation, and malonyldialdehyde concentration. More importantly, in vivo silencing of miR-9-5p by a specific antagomir in a murine acute MI model effectively preserves post-MI heart function with attenuated fibrosis and inflammatory response. Further studies demonstrated that antagomir treatment stabilizes Fstl1 expression as well as blocks cardiac cell death and reactive oxygen species generation in both ischemia-challenged hearts and hypoxia-treated cardiomyoblasts. Finally, cytoprotection against hypoxic challenge by miR-9-5p inhibitor is partially reversed by knockdown of Fstl1, indicating a novel role of miR-9-5p/Fstl1 axis in survival defense against hypoxic challenge. In summary, these findings identified miR-9-5p as a mediator of hypoxic injury in cardiomyoblasts and miR-9-5p suppression prevents cardiac remodeling after acute MI, providing a potential strategy for early treatment against MI.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Interferencia de ARN , Remodelación Ventricular/genética , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/genética , Biomarcadores , Modelos Animales de Enfermedad , Ecocardiografía , Fibrosis , Proteínas Relacionadas con la Folistatina/genética , Pruebas de Función Cardíaca , Hipoxia/genética , Hipoxia/metabolismo , Masculino , Ratones , MicroARNs/administración & dosificación , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocarditis/genética , Miocarditis/metabolismo , Miocarditis/patología , Miocarditis/fisiopatología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , ARN Mensajero/genética
17.
J Am Heart Assoc ; 7(15): e008737, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30371236

RESUMEN

Background Exosomes are membranous vesicles generated by almost all cells. Recent studies demonstrated that mesenchymal stem cell-derived exosomes possessed many effects, including antiapoptosis, anti-inflammatory effects, stimulation of angiogenesis, anticardiac remodeling, and recovery of cardiac function on cardiovascular diseases. However, targeting of exosomes to recipient cells precisely in vivo still remains a problem. Ligand fragments or homing peptides discovered by phage display and in vivo biopanning methods fused to the enriched molecules on the external part of exosomes have been exploited to improve the ability of exosomes to target specific tissues or organs carrying cognate receptors. Herein, we briefly elucidated how to improve targeting ability of exosomes to ischemic myocardium. Methods and Results We used technology of molecular cloning and lentivirus packaging to engineer exosomal enriched membrane protein (Lamp2b) fused with ischemic myocardium-targeting peptide CSTSMLKAC (IMTP). In vitro results showed that IMTP-exosomes could be internalized by hypoxia-injured H9C2 cells more efficiently than blank-exosomes. Compared with blank-exosomes, IMTP-exosomes were observed to be increasingly accumulated in ischemic heart area ( P<0.05). Meanwhile, attenuated inflammation and apoptosis, reduced fibrosis, enhanced vasculogenesis, and cardiac function were detected by mesenchymal stem cell-derived IMTP-exosome treatment in ischemic heart area. Conclusions Our research concludes that exosomes engineered by IMTP can specially target ischemic myocardium, and mesenchymal stem cell-derived IMTP-exosomes exert enhanced therapeutic effects on acute myocardial infarction.


Asunto(s)
Exosomas/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Miocitos Cardíacos/metabolismo , Péptidos/administración & dosificación , Animales , Línea Celular , Células HEK293 , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Infarto del Miocardio/metabolismo , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/metabolismo , Plásmidos , Ratas , Transfección
18.
Stem Cells Int ; 2018: 3290372, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30271437

RESUMEN

BACKGROUND: To cure ischemic diseases, angiogenesis needs to be improved by various strategies in ischemic area. Considering that microRNA-132 (miR-132) regulates endothelial cell behavior during angiogenesis and the safe and efficacious delivery of microRNAs in vivo is rarely achieved, an ideal vehicle for miR-132 delivery could bring the promise for ischemic diseases. As a natural carrier of biological molecules, exosomes are more and more developed as an ideal vehicle for miRNA transfer. Meanwhile, mesenchymal stem cells could release large amounts of exosomes. Thus, this study aimed to investigate whether MSC-derived exosomes can be used for miR-132 delivery in the treatment of myocardial ischemia. METHODS: MSC-derived exosomes were electroporated with miR-132 mimics and inhibitors. After electroporation, miR-132 exosomes were labelled with DiI and added to HUVECs. Internalization of DiI-labelled exosomes was examined by fluorescent microscopy. Expression levels of miR-132 in exosomes and HUVECs were quantified by real-time PCR. The mRNA levels of miR-132 target gene RASA1 in HUVECs were quantified by real-time PCR. Luciferase reporter assay was performed to examine the targeting relationship between miR-132 and RASA1. The effects of miR-132 exosomes on the angiogenic ability of endothelial cells were evaluated by tube formation assay. Matrigel plug assay and myocardial infarction model were used to determine whether miR-132 exosomes can promote angiogenesis in vivo. RESULTS: miR-132 mimics were effectively electroporated and highly detected in MSC-derived exosomes. The expression level of miR-132 was high in HUVECs preincubated with miR-132 mimic-electroporated exosomes and low in HUVECs preincubated with miR-132 inhibitor-electroporated exosomes. The expression level of RASA1, miR-132 target gene, was reversely correlated with miR-132 expression in HUVECs pretreated with exosomes. Luciferase reporter assay further confirmed that RASA1 was a direct target of miR-132. Exosomes loaded with miR-132, as a vehicle for miRNA transfer, significantly increased tube formation of endothelial cells. Moreover, subcutaneous injection of HUVECs pretreated with miR-132 exosomes in nude mice significantly increased their angiogenesis capacity in vivo. In addition, transplantation of miR-132 exosomes in the ischemic hearts of mice markedly enhanced the neovascularization in the peri-infarct zone and preserved heart functions. CONCLUSIONS: The findings suggest that the export of miR-132 via MSC-derived exosomes represents a novel strategy to enhance angiogenesis in ischemic diseases.

19.
Biosci Rep ; 38(6)2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30305382

RESUMEN

Background: Preclinical models have suggested a role for sex hormones in the development of glioblastoma multiforme (GBM). However, the impact of gender on the survival time of patients with GBM has not been fully understood. The objective of the present study was to clarify the association between gender and survival of patients with GBM by analyzing population-based data.Methods: We searched the Surveillance, Epidemiology, and End-Results database who were diagnosed with GBM between 2000 and 2008 and were treated with surgery. Five-year cancer specific survival data were obtained. Kaplan-Meier methods and multivariable Cox regression models were used to analyze long-term survival outcomes and risk factors.Results: A total of 6586 patients were identified; 61.5% were men and 38.5% were women. The 5-year cancer-specific survival (CSS) rates in the male and female groups were 6.8% and 8.3%, respectively (P=0.002 by univariate and P<0.001 by multivariate analysis). A stratified analysis showed that male patients always had the lowest CSS rate across localized cancer stage and different age subgroups.Conclusions: Gender has prognostic value for determining GBM risk. The role of sex hormones in the development of GBM warrants further investigation.


Asunto(s)
Neoplasias Encefálicas/epidemiología , Glioblastoma/epidemiología , Pronóstico , Adulto , Anciano , Neoplasias Encefálicas/patología , Supervivencia sin Enfermedad , Femenino , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Factores de Riesgo , Programa de VERF , Caracteres Sexuales , Tasa de Supervivencia
20.
Front Behav Neurosci ; 12: 169, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30174594

RESUMEN

Here, we aimed to investigate brain activity in migraineurs in response to emotional stimulation. Magnetoencephalography (MEG) was used to examine 20 patients with episodic migraine (EM group), 15 patients with chronic migraine (CM group), and 35 healthy participants (control group). Neuromagnetic brain activity was elicited by emotional stimulation using photographs of facial expressions. We analyzed the latency and amplitude of M100 and M170 components and used Morlet wavelet and beamformers to analyze the spectral and spatial signatures of MEG signals in gamma band (30-100 Hz). We found that the timing and frequency of MEG activity differed across the three groups in response negative emotional stimuli. First, peak M170 amplitude was significantly lower in the CM group than in the control group. Second, compared with the control group, the average spectral power was significantly lower in the EM group and CM group at M100 and M170. Third, the average spectral powers of the M100 and M170 in the CM group were negatively correlated with either HAM-D scores or migraine attack frequency. No significant differences across groups was found for positive or neutral emotional stimuli. Furthermore, after negative emotional stimuli, the MEG source analysis demonstrated that the CM group showed a significantly higher percentage of amygdala activation than the control group for M100 and M170. Thus, during headache free phases, migraineurs have abnormal brain activity in the gamma band in response to negative emotional stimuli. Trial Registration: ChiCTR-RNC-17012599. Registered 7 September, 2017.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...