Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Biomaterials ; 299: 122161, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37236138

RESUMEN

We previously constructed a three-dimensional gelatin sponge (3D-GS) scaffold as a delivery vehicle for therapeutic cells and trophic factors in the treatment of spinal cord injury (SCI), and this study aimed to assess the biosafety and efficacy of the scaffold in a non-human primate SCI model. However, because it has only been tested in rodent and canine models, the biosafety and efficacy of the scaffold should ideally be assessed in a non-human primate SCI model before its use in the clinic. No adverse reactions were observed over 8 weeks following 3D-GS scaffold implantation into in a Macaca fascicularis with hemisected SCI. Scaffold implantation also did not add to neuroinflammatory or astroglial responses already present at the injured site, suggesting good biocompatibility. Notably, there was a significant reduction in α-smooth muscle actin (αSMA)-positive cells at the injury/implantation interface, leading to alleviation of fibrotic compression of the residual spinal cord tissue. The regenerating tissue in the scaffold showed numerous cells migrating into the implant secreting abundant extracellular matrix, resulting in a pro-regenerative microenvironment. Consequently, nerve fiber regeneration, myelination, vascularization, neurogenesis, and electrophysiological improvements were achieved. These results indicated that the 3D-GS scaffold had good histocompatibility and effectiveness in the structural repair of injured spinal cord tissue in a non-human primate and is suitable for use in the treatment of patients with SCI.


Asunto(s)
Gelatina , Traumatismos de la Médula Espinal , Animales , Perros , Gelatina/química , Andamios del Tejido/química , Traumatismos de la Médula Espinal/terapia , Regeneración Nerviosa/fisiología , Médula Espinal , Primates
2.
ACS Nano ; 16(1): 823-836, 2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35025206

RESUMEN

The microorganism has become a promising therapeutic tool for many diseases because it is a kind of cell factory that can efficiently synthesize a variety of bioactive substances. However, the metabolic destiny of microorganisms is difficult to predict in vivo. Here, a timing bionic dormant body with programmable destiny is reported, which can predict the metabolic time and location of microorganisms in vivo and can prevent it from being damaged by the complex biological environment in vivo. Taking the complex digestive system as an example, the bionic dormant body exists in the upper digestive tract as a nonmetabolic dormant body after oral administration and will be awakened to synthesize bioactive substances about 2 h after reaching the intestine. Compared with oral microorganisms alone, the bioavailability of the biomimetic dormant body in the intestine is almost 3.5 times higher. The utilization rate of the oral bionic dormant body to synthesize drugs is 2.28 times higher than oral drugs. We demonstrated the significant efficacies of treatment using Parkinson's disease (PD) mice by dormant body capable of timed neurotransmitter production after oral delivery. The timed bionic dormant body with programmable destiny may provide an effective technology to generate advanced microbial therapies for the treatment of various diseases.


Asunto(s)
Biónica , Enfermedad de Parkinson , Ratones , Animales , Preparaciones Farmacéuticas
3.
Mol Med Rep ; 23(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33495805

RESUMEN

Subsequently to the publication of the above paper, an interested reader drew to the authors' attention that several pairings of panels in Fig. 5, as shown on p. 5599, were strikingly similar. After having examined their original data, the authors realized that they uploaded some images incorrectly during the process of compiling this figure, and that there were duplicated data panels in this figure. However, the authors were able to consult their original data, and had access to the correct images. The revised version of Fig. 5, showing the correct data for the Akt/Control, p­Akt/Control, mTOR/0.05 µM Ouabain, HIF­1α/0.05 µM Ouabain and Akt/0.5 µM Ouabain experiments, is shown opposite. Note that the replacement of the erroneous data does not affect either the results or the conclusions reported in this paper, and all the authors agree to this Corrigendum. The authors are grateful to the Editor of Molecular Medicine Reports for granting them this opportunity to publish a Corrigendum, and apologize to the readership for any inconvenience caused. [the original article was published in Molecular Medicine Reports 17: 5595­5600, 2018; DOI: 10.3892/mmr.2018.8587].

4.
Front Neurosci ; 14: 403, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32581664

RESUMEN

Traumatic brain injury (TBI) contributes to hypocoagulopathy associated with prolonged bleeding and hemorrhagic progression. Bloodletting puncture therapy at hand twelve Jing-well points (BL-HTWP) has been applied as a first aid measure in various emergent neurological diseases, but the detailed mechanisms of the modulation between the central nervous system and systemic circulation after acute TBI in rodents remain unclear. To investigate whether BL-HTWP stimulation modulates hypocoagulable state and exerts neuroprotective effect, experimental TBI model of mice was produced by the controlled cortical impactor (CCI), and treatment with BL-HTWP was immediately made after CCI. Then, the effects of BL-HTWP on the neurological function, cerebral perfusion state, coagulable state, and cerebrovascular histopathology post-acute TBI were determined, respectively. Results showed that BL-HTWP treatment attenuated cerebral hypoperfusion and improve neurological recovery post-acute TBI. Furthermore, BL-HTWP stimulation reversed acute TBI-induced hypocoagulable state, reduced vasogenic edema and cytotoxic edema by regulating multiple hallmarks of coagulopathy in TBI. Therefore, we conclude for the first time that hypocoagulopathic state occurs after acute experimental TBI, and the neuroprotective effect of BL-HTWP relies on, at least in part, the modulation of hypocoagulable state. BL-HTWP therapy may be a promising strategy for acute severe TBI in the future.

5.
Zhongguo Zhen Jiu ; 39(10): 1075-80, 2019 Oct 12.
Artículo en Chino | MEDLINE | ID: mdl-31621260

RESUMEN

OBJECTIVE: To observe the effect of bloodletting acupuncture at twelve jing-well points of hand on microcirculatory disturbance in mice with traumatic brain injury (TBI), and to explore the protective effect of bloodletting therapy on TBI. METHODS: Sixty clean adult male C57BL/6J mice were randomly divided into a sham-operation group, a model group and a treatment group, 20 mice in each group. The TBI model was established by using electronic controlled cerebral cortex impact instrument in the model group and the treatment group. The mice in the treatment group were treated with bloodletting acupuncture at bilateral "Shaoshang" (LU 11), "Shangyang" (LI 1), "Zhongchong" (PC 9), "Guanchong" (TE 1), "Shaochong" (HT 9) and "Shaoze" (SI 1) immediately after trauma. The mice in the sham-operation group only opened the bone window but did not receive the strike. The regional cerebral blood flow (rCBF) was monitored by laser speckle contrast analysis (LASCA) using a PeriCam PSI System before trauma, immediately after trauma and 1, 2, 12, 24, 48, 72 h after trauma. The brain water content was measured by wet-dry weight method 24 h after trauma. The severity of functional impairment at 2, 12, 24, 48 and 72 h after trauma was evaluated by modified neurological scale scores (mNSS). RESULTS: ① 2 h after trauma, the mNSS in the model group and treatment group were >7 points, suggesting the successful establishment of model; compared with the sham-operation group, the mNSS was increased significantly from 12 to 72 h after trauma in the model group ( all P<0.01), but the mNSS in the treatment group was significantly lower than that in the model group from 2 to 24 h after trauma (P<0.01, P<0.05). ② Compared with the sham-operation group, rCBF in the model group was decreased significantly immediately after trauma (P<0.01), and the rCBF in the model group was lower than that in the sham-operation group from 1 to 72 h after trauma ( all P<0.01); rCBF in the treatment group began to rise and was significantly higher than that in the model group 1-2 h after trauma (P<0.01); 12-48 h after trauma, the increasing of rCBF in the two groups tended to be gentle until 72 h after injury, and rCBF in the model group was decreased while that in the treatment group continued to rise and was higher than that in the model group (P<0.01). ③ 24 h after trauma, the brain water content in the model group was significantly higher than that in the sham-operation group (P<0.01), and brain water content in the treatment group was significantly lower than that in the model group (P<0.01). CONCLUSION: The bloodletting acupuncture at twelve jing-well points of hand could improve microcirculation disturbance, increase microcirculation perfusion, alleviate secondary brain edema and promote the recovery of nerve function in mice with TBI.


Asunto(s)
Terapia por Acupuntura , Venodisección , Lesiones Traumáticas del Encéfalo , Puntos de Acupuntura , Animales , Lesiones Traumáticas del Encéfalo/terapia , Masculino , Ratones , Ratones Endogámicos C57BL , Microcirculación , Distribución Aleatoria
6.
Biomed Res Int ; 2019: 5653212, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31355268

RESUMEN

OBJECTIVE: Casein kinase 2 interacting protein-1 (CKIP-1) has exhibited multiple functions in regulating cell proliferation, apoptosis, differentiation, and cytoskeleton. CKIP-1 also plays an important role as a critical regulator in tumorigenesis. The aim of this study is to further examine the function of CKIP-1 in glioma cells. METHODS: The expression level of CKIP-1 protein was determined in gliomas tissues and cell lines by immunohistochemistry stain and western blotting while the association of CKIP-1 expression with prognosis was analyzed by Kaplan-Meier method and compared by log-rank test. CKIP-1 was overexpressed or silenced in gliomas cell lines. CCK-8, colony formation assay, and BrdU incorporation assay were used to determine cell proliferation and DNA synthesis. Cell cycle and apoptosis rate were determined with fluorescence-activated cell sorting (FACS) method. Then, expression of key members in AKT/GSK3ß/ß-catenin pathway was detected by western blot analysis. RESULTS: In the present study, we reported new evidence that CKIP-1 was reversely associated with the proliferation of glioma cells and survival in glioma patients. Additionally, the overexpressed CKIP-1 significantly inhibited glioma cell proliferation. Further experiments revealed that CKIP-1 functioned through its antiproliferative and proapoptotic activity in glioma cells. Importantly, mechanistic investigations suggested that CKIP-1 sharply suppressed the activity of AKT by inhibiting the phosphorylation, markedly downregulated the phosphorylated GSK3ß at Ser9, and promoted ß-catenin degradation. CONCLUSIONS: Overall, our results provided new insights into the clinical significance and molecular mechanism of CKIP-1 in glioma, which indicated CKIP1 might function as a therapeutic target for clinical treatment of glioma.


Asunto(s)
Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glioma , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Adulto , Línea Celular Tumoral , Femenino , Glioma/metabolismo , Glioma/mortalidad , Glioma/patología , Humanos , Masculino , Persona de Mediana Edad
7.
Neural Regen Res ; 14(6): 991-996, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30762010

RESUMEN

An accurate and effective neurological evaluation is indispensable in the treatment and rehabilitation of traumatic brain injury. However, most of the existing evaluation methods in basic research and clinical practice are not objective or intuitive for assessing the neurological function of big animals, and are also difficult to use to qualify the extent of damage and recovery. In the present study, we established a big animal model of traumatic brain injury by impacting the cortical motor region of beagles. At 2 weeks after successful modeling, we detected neurological deficiencies in the animal model using a series of techniques, including three-dimensional motion capture, electromyogram and ground reaction force. These novel technologies may play an increasingly important role in the field of traumatic brain injury diagnosis and rehabilitation in the future. The experimental protocol was approved by the Animal Care and Use Committee of Logistics University of People's Armed Police Force (approval No. 2017-0006.2).

8.
Mol Med Rep ; 17(4): 5595-5600, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29436645

RESUMEN

Glioma is one of the most malignant forms of brain tumor, and has been of persistent concern due to its high recurrence and mortality rates, and limited therapeutic options. As a cardiac glycoside, ouabain has widespread applications in congestive heart diseases due to its positive cardiac inotropic effect by inhibiting Na+/K+­ATPase. Previous studies have demonstrated that ouabain has antitumor activity in several types of human tumor, including glioma. However, the exact underlying mechanism remains to be elucidated. The purpose of present study was to elucidate the effect of ouabain on human glioma cell apoptosis and investigate the exact mechanism. U­87MG cells were treated with various concentrations of ouabain for 24 h, following which cell viability and survival rate were assessed using a 3­(4,5-dimethylthiazol-2­yl)­2,5­diphenyltetrazolium bromide assay. The dynamic changes and cell motility were observed using digital holographic microscopy. Additionally, western blot analysis and high­content screening assays were used to detect the protein expression levels of phosphorylated (p­)Akt, mammalian target of rapamycin (mTOR), p­mTOR and hypoxia­inducible factor (HIF)­1α, respectively. Compared with the control group, ouabain suppressed U­87MG cell survival, and attenuated cell motility in a dose­dependent manner (P<0.01). The downregulation of p­Akt, mTOR, p­mTOR and HIF­1α were observed following treatment with 2.5 and 25 µmol/l of ouabain. These results suggested that ouabain exerted suppressive effects on tumor cell growth and motility, leading to cell death via regulating the intracellular Akt/mTOR signaling pathway and inhibiting the expression of HIF­1α in glioma cells. The present study examined the mechanism underlying the antitumor property of ouabain, providing a novel potential therapeutic agent for glioma treatment.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ouabaína/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glioma/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
9.
Neurotherapeutics ; 15(1): 216-232, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29247448

RESUMEN

This study aimed to investigate the effects of targeted temperature management (TTM) modulation on traumatic brain injury (TBI) and the involved mechanisms using quantitative proteomics technology. SH-SY5Y and HT-22 cells were subjected to moderate stretch injury using the cell injury controller (CIC), followed by incubation at TTM (mild hypothermia, 32°C), or normothermia (37°C). The real-time morphological changes, cell cycle phase distribution, death, and cell viability were evaluated. Moderate TBI was produced by the controlled cortical impactor (CCI), and the effects of TTM on the neurological damage, neurodegeneration, cerebrovascular histopathology, and behavioral outcome were determined in vivo. Results showed that TTM treatment prevented TBI-induced neuronal necrosis in the brain, achieved a substantial reduction in neuronal death both in vitro and in vivo, reduced cortical lesion volume and neuronal loss, attenuated cerebrovascular histopathological damage, brain edema, and improved behavioral outcome. Using an iTRAQ proteomics approach, proteins that were significantly associated with TTM in experimental TBI were identified. Importantly, changes in four candidate molecules (plasminogen [PLG], antithrombin III [AT III], fibrinogen gamma chain [FGG], transthyretin [TTR]) were verified using TBI rat brain tissues and TBI human cerebrospinal fluid (CSF) samples. This study is one of the first to investigate the neuroprotective effects of TTM on the proteome of human and experimental models of TBI, providing an overall landscape of the TBI brain proteome and a scientific foundation for further assessment of candidate molecules associated with TTM for the promotion of reparative strategies post-TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/terapia , Hipotermia Inducida/métodos , Proteómica , Animales , Lesiones Traumáticas del Encéfalo/patología , Ciclo Celular , Línea Celular Tumoral , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/patología , Frío , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Aprendizaje por Laberinto , Ratones , Necrosis , Neuronas/metabolismo , Neuronas/patología , Estimulación Física , Ratas , Ratas Sprague-Dawley
10.
Sci Rep ; 7(1): 5818, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28724891

RESUMEN

Necroptosis, a novel type of programmed cell death, is involved in stroke-induced ischemic brain injury. Although studies have sought to explore the mechanisms of necroptosis, its signaling pathway has not yet to be completely elucidated. Thus, we used oxygen-glucose deprivation (OGD) and middle cerebral artery occlusion (MCAO) models mimicking ischemic stroke (IS) conditions to investigate mechanisms of necroptosis. We found that OGD and MCAO induced cell death, local brain ischemia and neurological deficit, while zVAD-fmk (zVAD, an apoptotic inhibitor), GSK'872 (a receptor interacting protein kinase-3 (RIP3) inhibitor), and combined treatment alleviated cell death and ischemic brain injury. Moreover, OGD and MCAO upregulated protein expression of the triggers of necroptosis: receptor interacting protein kinase-1 (RIP1), RIP3 and mixed lineage kinase domain-like protein (MLKL). The upregulation of these proteins was inhibited by GSK'872, combination treatments and RIP3 siRNA but not zVAD treatment. Intriguingly, hypoxia-inducible factor-1 alpha (HIF-1α), an important transcriptional factor under hypoxic conditions, was upregulated by OGD and MCAO. Similar to their inhibitory effects on aforementioned proteins upregulation, GSK'872, combination treatments and RIP3 siRNA decreased HIF-1α protein level. These findings indicate that necroptosis contributes to ischemic brain injury induced by OGD and MCAO and implicate HIF-1α, RIP1, RIP3, and MLKL in necroptosis.


Asunto(s)
Apoptosis , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Proteínas Activadoras de GTPasa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Clorometilcetonas de Aminoácidos , Animales , Lesiones Encefálicas/complicaciones , Isquemia Encefálica/complicaciones , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Glucosa/deficiencia , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones Endogámicos C57BL , Necrosis , Oxígeno , ARN Interferente Pequeño/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
11.
Brain Inj ; 31(3): 406-415, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28140659

RESUMEN

AIM: To investigate the protective effect of hypothermia (HT) on brain injury in moderate traumatic brain injury (TBI) rat models and the potential mechanisms, especially the involvement of RIPK1 in apoptosis and necroptosis. METHODS: Adult Sprague-Dawley rats were randomized to four groups: sham+normothermia (sham+NT), sham+hypothermia (sham+HT), moderate TBI+normothermia (TBI+NT) and moderate TBI+hypothermia (TBI+HT). The sham+HT and TBI+HT groups were submitted to 32°C for 6 hours. The regional cerebral blood flow (rCBF) was assessed 24 hours after TBI; 24 and 48 hours after TBI, the modified neurological severity score (mNSS) was assessed. Immediately after behavioural tests, rats were sacrificed to harvest the brain tissues. RESULTS: mNSS scores were lower in the TBI+HT group compared with the TBI+NT group (p < 0.01) and cerebral blood flow was better (p < 0.01). H&E staining of the cortex and ipsilateral hippocampus showed pyknotic and irregularly shaped neurons in TBI+NT rats, which were less frequent in TBI+HT rats. The TBI+NT and TBI+HT groups showed higher TNF-α, TRAIL, FasL, FADD, caspase-3, caspase-8, PARP-1, RIPK-1 and RIPK-3 levels than the sham+NT group (all p < 0.05), but the levels of these proteins were all lower in the TBI+HT group compared with the TBI+NT group (all p < 0.01). CONCLUSION: HT treatment significantly reduced RIPK-1 upregulation, which may inhibit necroptosis and apoptosis pathways after moderate TBI.


Asunto(s)
Apoptosis/fisiología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/terapia , Regulación de la Expresión Génica/fisiología , Hipotermia Inducida , Animales , Caspasas/genética , Caspasas/metabolismo , Circulación Cerebrovascular/fisiología , Modelos Animales de Enfermedad , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Masculino , Necrosis/etiología , Necrosis/metabolismo , Examen Neurológico , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
12.
Neural Regen Res ; 11(6): 931-6, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27482221

RESUMEN

Bloodletting at Jing points has been used to treat coma in traditional Chinese medicine. Mild induced hypothermia has also been shown to have neuroprotective effects. However, the therapeutic effects of bloodletting at Jing points and mild induced hypothermia alone are limited. Therefore, we investigated whether combined treatment might have clinical effectiveness for the treatment of acute severe traumatic brain injury. Using a rat model of traumatic brain injury, combined treatment substantially alleviated cerebral edema and blood-brain barrier dysfunction. Furthermore, neurological function was ameliorated, and cellular necrosis and the inflammatory response were lessened. These findings suggest that the combined effects of bloodletting at Jing points (20 µL, twice a day, for 2 days) and mild induced hypothermia (6 hours) are better than their individual effects alone. Their combined application may have marked neuroprotective effects in the clinical treatment of acute severe traumatic brain injury.

13.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 31(3): 249-53, 2015 May.
Artículo en Chino | MEDLINE | ID: mdl-26387189

RESUMEN

OBJECTIVE: To investigate the influence of therapeutic bloodletting at Jing-well points and hypothermia on acute cerebral edema after traumatic brain injury (TBI) in rats. METHODS: Seventy-five SD rats were randomly divided into sham-operation group (Sham), TBI group (TBI), bloodletting group (BL), mild-induced hypothermia group (MIH), and bloodletting plus MIH group (BL + MIH) (n = 15). The model of TBI was established by electric controlled cortical impactor (eCCI). The rats of BL group were bloodletting at Jing-well points immediately after injury, twice daily. While the MIH group was settled on a hypothermia blanket promptly after TBI for 6 hours, so that the temperature dropped to 32 degrees. Each of measurement was performed after 48 hours. Magnetic resonance imaging (MRI) was used to evaluate the dynamic impairment of cerebral edema after TBI (n = 3). In addition, mNSS score, measurements of wet and dry brain weight, and Evans Blue assay were performed to investigate the neurologic deficit, cerebral water content (n = 8), and blood-brain barrier permeability (BBB), (n = 4), respectively. RESULTS: MRI analysis showed that the cerebral edema, hematoma and midline shifting of rats in TBI group was more serious than other treatment group. Meanwhile compared with TBI group, the mNSS scores of every treatment group were meaningfully lower (all P < 0.05). Furthermore, treatment with BL+ MIH group was superior to the separated BL and MIH group (all P < 0.01). In addition, brain water content of each intervention group reduced to varying degrees (all P < 0.05), especially that of MIH group and BL + MIH group (P <0.01). BBB permeability of each treatment group was also significantly improved (all P < 0.01), and the improvement in MIH group and BL + MIH group was much better than the BL alone group (P < 0.05, P < 0.01). CONCLUSION: Our major finding is that bloodletting at Jing-well points and MIH can reduce cerebral edema and BBB dysfunction and exert neuroprotective effects after TBI. The results suggest that the combination of BL and MIH is more effective than other treatment being used alone.


Asunto(s)
Venodisección , Edema Encefálico/prevención & control , Lesiones Encefálicas/terapia , Hipotermia Inducida , Animales , Barrera Hematoencefálica , Encéfalo/patología , Ratas , Ratas Sprague-Dawley
14.
Zhongguo Zhen Jiu ; 33(9): 779-83, 2013 Sep.
Artículo en Chino | MEDLINE | ID: mdl-24298764

RESUMEN

OBJECTIVE: To verify the effect of bloodletting therapy at Jing-well points and semen coicis on patients with traumatic cerebral infarction. METHODS: Ninety patients were randomized into a bloodletting therapy at Jing-well points group (bloodletting group), a semen coicis group and a comprehensive therapy group, 30 cases in each one. The conventional basic medication was applied in all of the three groups. In the bloodletting group, the bloodletting therapy was done at twelve Jing-well points with three-edged needle, 3 drops of blood required at each one, three times a day. In the semen coicis group, the semen coicis preparation was applied via nasal feeding or oral administration, 90 g each day, three times a day. In the comprehensive therapy group, the bloodletting therapy at twelve Jing-well points and semen coicis preparation were used in combination and the methods were same as the above two groups. After 4 weeks of treatment, the efficacy was assessed with nerve function defectscale (NDS). Fugl-Meyer scale of the upper and lower limb function was used to evaluate the motor function of the affected limbs of the patients before and after treatment. RESULTS: The scores of Fugl-Meyer scale of the upper and lower limb function were increased apparently after treatment in the patients of every group (P < 0.01, P < 0.05). The score increase was much more obvious in the bloodletting group and the comprehensive therapy group as compared with the semen coicis group (all P < 0.01). The result in the comprehensive therapy group was superior to the bloodletting group (all P < 0.05). The total effective rates of NDS in the comprehensive therapy group, bloodletting group and semen coicis group were 96.7% (29/30), 83.3% (25/30) and 76.7% (23/30) separately. The result in the comprehensive therapy group was higher apparently than those in the bloodletting group and semen coicis group separately (both P < 0.05). The result in the bloodletting group was better than that in the semen coicis group (P < 0.05). CONCLUSION: The bloodletting therapy at Jing-well points and semen coicis alleviate apparently nerve function defect, improve the motor function of the affected limbs and achieve the better efficacy.


Asunto(s)
Venodisección , Encéfalo/efectos de los fármacos , Infarto Cerebral/terapia , Coix/química , Medicamentos Herbarios Chinos/administración & dosificación , Puntos de Acupuntura , Adulto , Anciano , Infarto Cerebral/tratamiento farmacológico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Adulto Joven
15.
Ther Hypothermia Temp Manag ; 3(3): 132-139, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24066267

RESUMEN

Previous studies have reported that mild induced hypothermia (MIH) treatment has positive effects on traumatic brain injury (TBI) outcomes, which have recently been linked to ß-amyloid (Aß)-induced secondary brain injury (SBI) extent in hippocampal tissues. We therefore investigate the relationship between MIH treatment and expression of Aß and related proteins following TBI. Adult Sprague-Dawley rats were randomly divided into three equal groups (S: sham-operated, N: normothermia, and H: mild hypothermia). After TBI induced by fluid percussion, group N remained at normal temperature, and group H underwent MIH (32°C) for 6 hours. Behavioral scale scores were then assessed. All rats were sacrificed 24 hours and hippocampal tissues were harvested, stained with hematoxylin and eosin. mRNA and protein expressions of Aß, ß-amyloid protein precursor (APP), and ß-secretase (BACE) were analyzed. Our results revealed significantly improved behavioral scale scores and the surviving neuron numbers were observed in group H compared to group N (p<0.05). Additionally, group N increased APP, Aß, and BACE levels compared to group S (all p<0.05). Reduced expression of APP-, Aß-, and BACE were apparent in group H compared to group N (all p<0.05). However, no statistically significant difference was observed between groups H and S in behavioral scale scores and the expression of APP-, Aß-, and BACE (p>0.05). In conclusion, MIH treatment significantly improves the survival of neuron and reduced Aß, BACE, and APP upregulation after TBI, which may provide a better understanding of the mechanisms by which hypothermia reduces SBI in TBI patients.

16.
PLoS One ; 8(8): e72376, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23991102

RESUMEN

Forkhead box M1 (FoxM1) is a member of the forkhead transcription factor family and is overexpression in malignant gliomas. However, the molecular mechanisms by which FoxM1lead to glioma carcinogenesis and progression are still not well known. In the present study, we show that Anxa1 was overexpression in gliomas and predicted the poor outcome. Furthermore, Anxa1 closely related to the FoxM1 expression and was a direct transcriptional target of FoxM1. Overexpression of FoxM1 up-regulated Anxa1 expression, whereas suppression of FoxM1 expression down-regulated Anxa1 expression in glioma cells. Finally, FoxM1 enhanced the proliferation, migration, and angiogenesis in Anxa1-dependent manner both in vitro and in vivo. Our findings provide both clinical and mechanistic evidences that FoxM1 contributes to glioma development by directly up-regulating Anxa1 expression.


Asunto(s)
Anexina A1/metabolismo , Factores de Transcripción Forkhead/fisiología , Glioblastoma/patología , Regulación hacia Arriba , Anexina A1/genética , Secuencia de Bases , Western Blotting , Inmunoprecipitación de Cromatina , Cartilla de ADN , Progresión de la Enfermedad , Proteína Forkhead Box M1 , Glioblastoma/metabolismo , Humanos , Pronóstico , Regiones Promotoras Genéticas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
17.
Mol Neurobiol ; 48(3): 490-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23475611

RESUMEN

Accumulating evidence shows that Sirt1 regulates a variety of neurological functions through the deacetylation of many proteins besides histone; however, the literature on the relationship between Sirt1 and axonal outgrowth is limited. Here, we first demonstrated that Sirt1 was located in the axon, especially in the growth cone. Then, we found that genetic inhibition of Sirt1 retarded axonal development in embryonic hippocampal neurons, whereas genetic and pharmacologic upregulation of Sirt1 promoted not only the formation but also the elongation of axons. Sirt1 can deacetylate and thus activate Akt, and inhibition of Akt significantly reversed the axonogenesis induced by Sirt1 overexpression. We also found that Sirt1 inhibited the activity of glycogen synthase kinase 3 (GSK3), whereas activation of GSK3 could abolish the effect of Sirt1. These results suggest that Sirt1 promotes axonogenesis by deacetylating Akt and thereby activates the Akt/GSK3 pathway, which could be a promising therapeutic target for axonopathy.


Asunto(s)
Axones/enzimología , Axones/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Neurogénesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirtuina 1/metabolismo , Acetilación , Animales , Células Cultivadas , Regulación hacia Abajo , Activación Enzimática , Conos de Crecimiento/metabolismo , Neuritas/metabolismo , Ratas , Regulación hacia Arriba
18.
PLoS One ; 8(1): e55693, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23383267

RESUMEN

BACKGROUND: Forkhead box Q1 (FoxQ1) is a member of the forkhead transcription factor family, and it has recently been found to participate in cancer development. However, whether FoxQ1 expression contributes to glioma development and progression is not known. We investigate FoxQ1 expression in gliomas and the role of FoxQ1 during tumorgenesis. METHODS: Reverse transcription quantitative real-time PCR (RT-qPCR) and Western blot were used to determine the FoxQ1 and Neurexins 3 (NRXN3) expression in gliomas. Chromatin immunoprecipitation (ChIP) and luciferase assays were used to determine the regulation between FoxQ1 and NRXN3. We established depleted FoxQ1 stable clones in U-87MG cells and overexpressed FoxQ1 stable clones in SW1088 cells. MTT and transwell were used to evaluate the ability of proliferation and migration, respectively. RESULTS: FoxQ1 mRNA and protein were up-regulated in gliomas and negatively related to the NRXN3 expression (r = -0.373, P = 0.042). FoxQ1 directly binds to NRXN3 promoter region and suppresses the promoter activity. Furthermore, the ability of proliferation and migration is reduced in depleted FoxQ1 cells. CONCLUSION: FoxQ1 promotes glioma cell proliferation and migration by down-regulation of NRXN3 expression.


Asunto(s)
Movimiento Celular , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/metabolismo , Proteínas del Tejido Nervioso/genética , Animales , Secuencia de Bases , Sitios de Unión , Encéfalo/metabolismo , Línea Celular Tumoral , Proliferación Celular , Factores de Transcripción Forkhead/genética , Humanos , Ratones , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica , Trasplante Heterólogo
19.
Neurosci Lett ; 525(2): 95-9, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-22897877

RESUMEN

Ferulic acid (4-hydroxy-3-methoxycinnamic acid, FA) is a widely distributed natural phenolic compound that is abundant in many plant tissues and foods. This study investigated possible mechanisms underlying the sedative-hypnotic effect of FA through behavioral pharmacology methods. FA showed dose-dependent sedative effects on locomotion activity in normal mice. FA also significantly potentiated pentobarbital-induced (45 mg/kg, i.p.) sleep by prolonging sleeping time and shortening sleep latency in a dose-dependent manner. These effects were augmented by the administration of 5-hydroxytryptophan (5-HTP), a precursor of 5-hydroxytryptamine (5-HT). With a sub-hypnotic dose of pentobarbital (25 mg/kg, i.p.), FA significantly increased the rate of sleep onset and exhibited a synergistic effect with 5-HTP (2.5 mg/kg, i.p.). Pretreatment with p-chlorophenylalanine (PCPA, an inhibitor of tryptophan hydroxylase) significantly decreased the duration of pentobarbital-induced sleep, whereas FA significantly reversed this effect. These results suggest that FA has sedative-hypnotic activity, possibly mediated by the serotonergic system.


Asunto(s)
Ácidos Cumáricos/farmacología , Hipnóticos y Sedantes/farmacología , Fenobarbital/farmacología , Serotonina/metabolismo , Sueño/efectos de los fármacos , 5-Hidroxitriptófano/farmacología , Animales , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Fenclonina/farmacología , Masculino , Ratones , Ratones Endogámicos ICR , Actividad Motora/efectos de los fármacos , Triptófano Hidroxilasa/antagonistas & inhibidores
20.
J Neurotrauma ; 29(14): 2393-403, 2012 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-22655683

RESUMEN

Stem cell transplantation holds great potential for the treatment of traumatic brain injury (TBI). However, the micro-environment of reduced oxygen and accumulated toxins leads to low survival rates of grafted cells, which dramatically limits their clinical application. Mild hypothermia has been demonstrated to improve the micro-environment after severe TBI. Thus, we speculate that combinational therapy of mild hypothermia may promote survival of grafted cells, especially temperature-sensitive stem cells, which show the most activity in mild temperatures. In this study, we first isolated mesenchymal stem cells from umbilical cord (UCSMCs) and generated the temperature-sensitive UCSMCs (tsUCSMCs) by infection with a retrovirus carrying the temperature-sensitive tsA58 SV40 LT antigen gene. We demonstrated that tsUCSMCs grew and proliferated with more activity at 33°C than at 37°C by counting cell numbers with a hematocytometer, measuring the cell cycle with flow cytometry, and detecting proliferating cell nuclear antigen (PCNA) with immunofluorescence staining. Thereafter, we established the rat severe TBI model by fluid percussion, and injected PBS, UCSMCs, or tsUCSMCs into the injured region, and subject the animals to normothermia or mild hypothermia (33°C). We found that, compared with UCSMC or tsUCSMC treatment alone, their combination with hypothermia could significantly improve motor and cognitive function with more survival of the grafted cells. Furthermore, we observed that combined therapy with hypothermia and tsUCSMCs exerted the most protective effect on the recovery of neurological function of all the tested treatments, with the highest survival and proliferation rates, and the lowest apoptosis rate. Thus this may represent a new therapeutic strategy for the treatment of severe TBI.


Asunto(s)
Lesiones Encefálicas/cirugía , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Hipotermia Inducida/métodos , Puntaje de Gravedad del Traumatismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Animales , Temperatura Corporal/fisiología , Lesiones Encefálicas/fisiopatología , Terapia Combinada/métodos , Humanos , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...