Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
ACS Omega ; 8(43): 40934-40943, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37929090

RESUMO

Sepsis-associated encephalopathy (SAE) is the most common complication of sepsis, with increased morbidity and mortality. To date, there has still been no established pharmacological therapy. Memantine, as an NMDA (N-methyl-d-aspartate) receptor antagonist, exhibited neuroprotective effects against cognitive and emotional dysfunction in many disorders. We performed cecal ligation and puncture (CLP) inducing sepsis as the ideal animal model of SAE. CLP-induced septic mice were given a memantine treatment through intragastric administration. The novel object recognition test indicated that memantine significantly improved cognitive dysfunction in septic mice. The open field test revealed that the anxiety-like behaviors and locomotion ability of septic mice were relieved by memantine. The pole test further confirmed the protective effects of memantine against immobility. Memantine significantly inhibited the excessive glutamate production and improved impaired neurogenesis on first and seventh day after sepsis, accompanying with reducing proinflammatory cytokines production (tumor necrosis factor alpha (TNF-α), interleukin (IL)-1beta (IL-1ß), and IL-10) and microglia activation in the brain of SAE. In addition, memantine treatment also reducing sepsis-induced brain blood barrier disruption via inhibiting the expression of metalloproteinase-9 (MMP-9). In conclusion, memantine exerted neuro-protective effects against cognitive and emotional defects, which might be considered as a promising therapy for SAE.

2.
J Colloid Interface Sci ; 650(Pt A): 67-80, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37393769

RESUMO

Due to the complexity of tumors, multimodal therapy for them has always been of concern to researchers. How to design a multifunctional drug nanoplatform with cascade effect and capable of responding to specific stimuli in the tumor microenvironment is the key to achieve efficient multimodal synergistic therapy of cancer. Here, we prepare a kind of GNRs@SiO2@PDA-CuO2-l-Arg (GSPRs-CL) nanomotors for systematic treatment of tumor. First, under near-infrared (NIR) irradiation, GSPRs-CL can generate heat and exhibit excellent photothermal therapy effect. Then under acidic conditions, CuO2 can be decomposed to release Cu2+ and generate H2O2, which not only complemented the limited endogenous H2O2 in cells, but also further triggered Fenton-like reaction, converting H2O2 into •OH to kill cancer cells, thereby achieving chemodynamic therapy. Furthermore, both endogenous and exogenous H2O2 can release nitric oxide (NO) in response to the occurrence of l-Arg of nanomotors to enhance gas therapy. In addition, as a dual-mode drive, NIR laser and NO can promote the penetration ability of nanomotors at tumor sites. The experimental results in vivo show that the drug nanoplatform had good biosafety and significant tumor killing effect triggered by NIR light and acidic conditions of tumor. It provide a promising strategy for the development of advanced drug nanoplatform for cancer therapy.


Assuntos
Nanopartículas , Neoplasias , Humanos , Fototerapia/métodos , Dióxido de Silício/uso terapêutico , Peróxido de Hidrogênio/farmacologia , Linhagem Celular Tumoral , Raios Infravermelhos , Neoplasias/tratamento farmacológico , Microambiente Tumoral
3.
Adv Healthc Mater ; 12(22): e2300167, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37223944

RESUMO

Due to their complexity and variability, tumors need to be treated with multimodal combined therapy, which requires the development of therapeutic agents that can provide multimodal therapeutic effects. Herein, CuMoO4 nanodots smaller than 10 nm that can be prepared by simple hydrothermal method are reported. These nanodots can be well dispersed in water and have good biosafety and biodegradability. Further studies show that these nanodots also present multienzyme activities, such as catalase, peroxidase and glutathione peroxidase. In addition, CuMoO4 nanodots exhibit high photothermal conversion efficiency (41%) under 1064 nm near-infrared laser irradiation. In vitro and in vivo experimental results indicate that CuMoO4 nanodots can effectively inhibit the instinctive regulation of tumor cells to oxidative stress, provide sustained treatment to achieve photothermal synergistic ferroptosis, and trigger immune responses to immunogenic cell death. It is worth mentioning that the CuMoO4 nanodots also cause cuproptosis of tumor cells. This study provides a promising nanoplatform for multimodal combined therapy of cancer.


Assuntos
Nanopartículas , Neoplasias , Humanos , Linhagem Celular Tumoral , Fototerapia , Neoplasias/tratamento farmacológico , Terapia Combinada , Raios Infravermelhos , Nanopartículas/uso terapêutico
4.
Artigo em Inglês | MEDLINE | ID: mdl-36883991

RESUMO

A kind of drug-loaded nanomotors with deep penetration was developed to improve the therapeutic effect of ferroptosis on tumor. The nanomotors were constructed by co-loading hemin and ferrocene (Fc) on the surface of bowl-shaped polydopamine (PDA) nanoparticles. The near-infrared response of PDA makes the nanomotor have high tumor penetration capability. In vitro experiments show that the nanomotors can exhibit good biocompatibility, high light to heat conversion efficiency, and deep tumor permeability. It is worth noting that under the catalysis of H2O2 overexpressed in the tumor microenvironment, the Fenton-like reagents hemin and Fc loaded on the nanomotors can increase the concentration of toxic •OH. Furthermore, hemin can consume glutathione in tumor cells and trigger the up-regulation of heme oxygenase-1, which can efficiently decompose hemin to Fe2+, thus producing the Fenton reaction and causing a ferroptosis effect. Notably, thanks to the photothermal effect of PDA, it can enhance the generation of reactive oxygen species and thus intervene in the Fenton reaction process, thereby enhancing the ferroptosis effect photothermally. In vivo antitumor results show that the drug-loaded nanomotors with high penetrability showed an effective antitumor therapeutic effect.

5.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 35(3): 274-277, 2023 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-36916340

RESUMO

OBJECTIVE: To explore whether barium chloride (BaCl2) preconditioning has the protective effect on lipopolysaccharide (LPS)-induced acute respiratory distress syndrome (ARDS) model in mice and the possible mechanism. METHODS: Sixty 8-12 week old healthy C57BL/6 male mice were randomly divided into control group, ARDS model group and BaCl2 pretreatment group, with 20 mice in each group. The BaCl2 pretreatment group was continuously injected with BaCl2 (4 mg/kg through the tail vein) for 3 days before ARDS model establishment. ARDS model was established by intratracheally injecting (3 mg/kg) LPS. The control group was intratracheally given the same volume of 0.9% normal saline. On 24th hour after ARDS model establishment, some mice were sacrificed for obtaining fresh lung tissue. And the right lower lobe of the lung was separated for observing the pathological changes of lung tissue while the left lung tissue was used to measure the wet/dry weight ratio (W/D) of the lung. Some mice were sacrificed for observing pulmonary microvascular permeability at 2nd hours after injecting Evans blue (EB) through tail vein. The left mice were killed for alveolar lavage to measure the levels of tumor necrosis factor-α (TNF-α) via enzyme linked immunosorbent assay (ELISA). RESULTS: Comparing with the control group, ARDS model group showed typical ARDS pathological changes, which included the increased W/D ratio (4.951±0.161 vs. 3.449±0.299, P < 0.01) and the content of EB in the lung tissue (µg/g: 0.130±0.027 vs. 0.085±0.011, P < 0.01), the damaged alveolar wall structure, lung congestion and exudates in the alveoli, as well as amounts of inflammatory cells. The pathological score of lung injury (10.33±1.15 vs. 1.67±0.58) and the level of TNF-α in BALF (ng/L: 900.85±247.80 vs. 68.21±5.79) were significantly increased in the ARDS model group (both P < 0.01). Comparing with the ARDS model group, the lung W/D ratio (4.620±0.125 vs. 4.951±0.161) and the EB content in the lung tissue (µg/g: 0.108±0.011 vs. 0.130±0.027) of BaCl2 pretreatment group were significantly reduced (both P < 0.01). And the damaged pulmonary structural BaCl2 pretreatment group were significantly alleviated. In addition, the pulmonary pathological score (5.00±1.00 vs. 10.33±1.15) and the level of TNF-α in BALF (ng/L: 169.16±73.33 vs. 900.85±247.80) were significantly decreased (both P < 0.01). CONCLUSIONS: Barium chloride pretreatment can improve the lung histopathological changes of ARDS model mice induced by LPS by reducing the permeability of pulmonary capillaries and local inflammatory reaction.Barium chloride has the protective effect against LPS attack in mice model of ARDS.


Assuntos
Lesão Pulmonar Aguda , Síndrome do Desconforto Respiratório , Camundongos , Masculino , Animais , Lipopolissacarídeos , Fator de Necrose Tumoral alfa , Camundongos Endogâmicos C57BL , Pulmão
6.
Curr Med Sci ; 43(1): 86-92, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36752938

RESUMO

OBJECTIVE: Despite the recent advances in diagnosis and treatment, sepsis continues to lead to high morbidity and mortality. Early diagnosis and prompt treatment are essential to save lives. However, most biomarkers can only help to diagnose sepsis, but cannot predict the development of septic shock in high-risk patients. The present study determined whether the combined measurement of procalcitonin (PCT), thromboelastography (TEG) and platelet (PLT) count can predict the development of septic shock. METHODS: A retrospective study was conducted on 175 septic patients who were admitted to the intensive care unit between January 2017 and February 2021. These patients were divided into two groups: 73 patients who developed septic shock were assigned to the septic shock group, while the remaining 102 patients were assigned to the sepsis group. Then, the demographic, clinical and laboratory data were recorded, and the predictive values of PCT, TEG and PLT count for the development of septic shock were analyzed. RESULTS: Compared to the sepsis group, the septic shock group had statistically lower PLT count and TEG measurements in the R value, K value, α angle, maximum amplitude, and coagulation index, but had longer prothrombin time (DT), longer activated partial thromboplastin time (APTT), and higher PCT levels. Furthermore, the Sequential Organ Failure Assessment (SOFA) score was higher in the septic shock group. The multivariate logistic regression analysis revealed that PCT, TEG and PLT count were associated with the development of septic shock. The area under the curve analysis revealed that the combined measurement of PCT, TEG and PLT count can be used to predict the development of septic shock with higher accuracy, when compared to individual measurements. CONCLUSION: The combined measurement of PCT, TEG and PLT count is a novel approach to predict the development of septic shock in high-risk patients.


Assuntos
Sepse , Choque Séptico , Humanos , Choque Séptico/diagnóstico , Pró-Calcitonina , Tromboelastografia , Contagem de Plaquetas , Estudos Retrospectivos , Unidades de Terapia Intensiva
7.
J Colloid Interface Sci ; 633: 679-690, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36473358

RESUMO

The limited penetration depth of nanodrugs in the tumor and the severe hypoxia inside the tumor significantly reduce the efficacy of photothermal-photodynamic synergistic therapy (PTT-PDT). Here, we synthesized a methoxypolyethylene glycol amine (mPEG-NH2)-modified walnut-shaped polydopamine nanomotor (PDA-PEG) driven by near-infrared light (NIR). At the same time, it also loaded the photosensitizer indocyanine green (ICG) via electrostatic/hydrophobicinteractions and chelated with ferric ion (Fe3+). Under the irradiation of NIR, the asymmetry of PDA-PEG morphology led to the asymmetry of local photothermal effects and the formation of thermal gradient, which can make the nanomotor move autonomously. This ability of autonomous movement was proved to be used to improve the permeability of the nanomotor in three-dimensional (3D) tumor sphere. Fe3+ can catalyze endogenous hydrogen peroxide to produce oxygen, so as to overcome the hypoxia of tumor microenvironment and thereby generate more singlet oxygen to kill tumor cells. Animal experiments in vivo confirmed that the nanomotors had a good PTT-PDT synergistic treatment effect. The introduction of nanomotor technology has brought new ideas for cancer optical therapy.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Animais , Verde de Indocianina/farmacologia , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Neoplasias/tratamento farmacológico , Hipóxia/tratamento farmacológico , Linhagem Celular Tumoral , Microambiente Tumoral
8.
Bioconjug Chem ; 33(4): 726-735, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35312294

RESUMO

The combination of photothermal therapy and chemical drug therapy shows good prospects in cancer treatment, but there are also some limitations such as low permeability of therapeutic agents and uneven photothermal therapy. Here, we synthesized a walnut-shaped polydopamine (PDA) nanomotor driven by near infrared (NIR) light. The nanomotor was modified by methoxy polyethylene glycol amine (mPEG-NH2) for improving water solubility. PDA-PEG loaded adriamycin through π-π accumulation and hydrogen bonding. The experimental results showed that the PDA nanomotors had good biocompatibility and photothermal effect. Further, the NIR light irradiation and tumor cell microenvironment are conducive to drug release. In addition, under the irradiation of an NIR laser, the asymmetry of walnut-shaped nanoparticles makes the particles obtain the ability of autonomous movement, which can improve the permeability of particles in 3D tumor balls, which can provide support for drug penetration and heat dispersion. This strategy offers potential innovative materials for photothermal/chemotherapy synergistic therapy of tumors.


Assuntos
Juglans , Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Liberação Controlada de Fármacos , Humanos , Indóis , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fototerapia/métodos , Polímeros/uso terapêutico , Microambiente Tumoral
9.
Neurosci Lett ; 738: 135359, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32949939

RESUMO

hNSCs (human neural stem cells) derived from embryonic tissue and aborted fetal brains are considered to be the most promising candidates for neurodegenerative and other CNS(central nervous system) diseases. However, the most common problem, which limited successful use of these allogeneic hNSC therapy, is immune rejection. Mesenchymal stem cells (MSCs) from human umbilical cord (hUC-MSCs) are receiving increasing attention for their immune-modulatory properties. In the current studies, we firstly investigated the immunogenecity of hNSCs as well as their lineages in cultures with the presence or absence of interferon gamma (IFNγ), a pro-inflammatory factors. Our data revealed that the majority of hNSCs and astrocytes expressed MHCI (major histocompatibility complex class I) while neurons hardly expressed MHCI (<5%) in the absence of IFNγ. In addition, neither hNSCs nor neurons expressed MHCII while a subpopulation (about 18 %) of astrocytes expressed MHCII without IFNγ stimulation. However, the addition of IFNγ in cultures significantly increased the expressions of MHCII on hNSCs and astrocytes. However, IFNγ did not affect the expression of MHCI on hNSCs and astrocytes. We then investigated whether hUC-MSCs had the capacity of regulating the immunogenecity of hNSCs as well as their lineages in a co-culture system. We found that hUC-MSCs did not affect the expression of MHCI on hNSCs and their lineages, however, these cells were able to significantly inhibit the IFNγ-induced up-regulation of MHCII on hNSCs and astrocytes (p < 0.001). Thus, our results suggest that hUC-MSCs may serve as potentially useful modulators to reduce the immunogenicity of allogeneic hNSCs in clinical application.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Neurais/citologia , Cordão Umbilical/citologia , Astrócitos/metabolismo , Antígenos de Histocompatibilidade/farmacologia , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Neurais/efeitos dos fármacos , Neurônios/metabolismo
10.
Eur J Histochem ; 64(s2)2020 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-32705859

RESUMO

Alzheimer's disease (AD) is a progressive, neurodegenerative disease. Accumulating evidence suggests that protein isoaspartate methyltransferase 1 (PCMT1) is highly expressed in brain tissue (substantia nigra, blue plaque, paraventricular nucleus). In this study, we investigated the effect of neural stem cell conditioned medium alleviates Aß25-35 damage to SH-SY5Y cells by PCMT1/MST1 pathway. Results demonstrated that Aß25-35 significantly decreased the cell viability in time and dose dependent manner. However, Neural stem cell-complete medium (NSC-CPM) or NSC-CDM had inhibitory effect on toxicity when fibrillation of Aß25-35 occurred in their presence and NSC-CDM had a better inhibitor result. An increase of the PCMT1 expression levels was found in Aß25-35 + NSC-CDM group. sh-PCMT1 significantly reduced the PCMT1, the cell viability and inhibited the protective effect; induced apoptosis and increased the expression of p-MST1. Overexpression of PCMT1 group reversed the effect of Aß25-35 inhibited the cell viability and Aß25-35 induced the apoptosis. In conclusion, NSC-CDM corrects the damage of Aß25-35 to cells by increasing PCMT1, reducing MST phosphorylation.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Meios de Cultivo Condicionados/farmacologia , Células-Tronco Neurais/metabolismo , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/toxicidade , Transdução de Sinais/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Estresse Oxidativo/efeitos dos fármacos , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/metabolismo , Proteínas Proto-Oncogênicas/metabolismo
11.
Neurosci Lett ; 698: 76-80, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30605704

RESUMO

Even though opioid tolerance is both a common and a major challenge in medicine, treatment with opioids is currently the primary method used to treat acute and chronic pain. The cAMP accumulation induced by chronic morphine is regarded as one of the molecular mechanisms leading to its tolerance and dependence characteristics. In the present study, we differentiated SH-SY5Y cells into neuron-like cells by retinoic acid (RA), pretreated these cells with morphine, and tested their cAMP levels under different conditions, including co-culture with bone marrow-derived human mesenchymal stem cells from bone marrow (hMSCs-BM) at various hMSCs-BM/SH-SY5Y ratios (1:5, 1:25, and 1:125), by direct cell-to-cell contact or without cell-to-cell contact, and by conditioned medium (CM) from hMSCs-BM. We found that chronic treatment with 10 µM morphine led to cAMP upregulation in those RA-differentiated SH-SY5Y cells while the morphine induced-cAMP accumulation was significantly attenuated by co-culturing with hMSCs-BM by direct cell-to-cell contact at a lower cell ratio (1:25) and a higher cell ratio (1:5). However, at neither the low or higher cell ratios could hMSCs-BM inhibit morphine-induced cAMP accumulation in RA-differentiated SH-SY5Y cells without cell-to-cell contact. In summary, hMSCs-BM can successfully inhibit morphine-induced cAMP up-regulation in RA-differentiated SH-SY5Y cells by cell-to-cell contact at a higher ratio, suggesting that hMSCs-BM may serve as valuable therapeutics to minimize the risk of drug abuse and addiction in the treatment of morphine tolerance and dependence.


Assuntos
Analgésicos Opioides/farmacologia , AMP Cíclico/metabolismo , Células-Tronco Mesenquimais/fisiologia , Morfina/farmacologia , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados , Tolerância a Medicamentos , Humanos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Tretinoína/farmacologia
12.
Stem Cells Int ; 2018: 4659159, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29765412

RESUMO

INTRODUCTION: Our previous study suggested that NSC-CM (neural stem cell-conditioned medium) inhibited cell apoptosis in vitro. In addition, many studies have shown that neurotrophic factors and microparticles secreted into a conditioned medium by NSCs had neuroprotective effects. Thus, we hypothesized that NSC-CM had the capacity of protecting against cerebral I/R injury. METHODS: Adult male Sprague-Dawley rats receiving middle cerebral artery occlusion surgery as an animal model of cerebral I/R injury were randomly assigned to two groups: the control group and NSC-CM-treated group. 1.5 ml NSC-CM or PBS (phosphate buffer saline) was administrated slowly by tail vein at 3 h, 24 h, and 48 h after ischemia onset. RESULTS: NSC-CM significantly ameliorated neurological defects and reduced cerebral infarct volume, accompanied by preserved mitochondrial ultrastructure. In addition, we also found that NSC-CM significantly inhibited cell apoptosis in the ischemic hemisphere via improving the expression of Bcl-2 (B-cell lymphoma-2). CONCLUSION: NSC-CM might be an alternative and effective therapeutic intervention for ischemic stroke.

13.
J Stroke Cerebrovasc Dis ; 27(6): 1697-1704, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29525080

RESUMO

BACKGROUND: It is well accepted that both rosuvastatin and resveratrol exert neuroprotective effects on cerebral ischemia/reperfusion injury through some common pathways. Resveratrol has also been demonstrated to protect against cerebral ischemia/reperfusion injury through enhancing autophagy. Thus, we hypothesized that combined rosuvastatin and resveratrol pretreatment had synergistic effects on cerebral ischemia/reperfusion injury. MATERIALS AND METHODS: Adult male Sprague Dawley rats receiving middle cerebral artery occlusion surgery as animal model of cerebral ischemia/reperfusion injury were randomly assigned to 4 groups: control, resveratrol alone pretreatment, rosuvastatin alone pretreatment, and combined rosuvastatin and resveratrol pretreatment. Rosuvastatin (10 mg/kg) or resveratrol (50 mg/kg) was administrated once a day for 7 days before cerebral ischemia onset. RESULTS: We found that combined rosuvastatin and resveratrol pretreatment not only significantly decreased the neurologic defective score, cerebral infarct volume, the levels of caspase-3, and Interleukin-1ß (IL-1ß) but also significantly increased the ratios of Bcl-2/Bax and LC3II/LC3I, as well as the level of Becline-1, compared with resveratrol alone or rosuvastatin alone pretreatment group. Rosuvastatin alone pretreatment significantly increased the ratio of LC3II/LC3I and the level of Beclin-1. However, there were no significant differences in the neurologic defective score, cerebral infarct volume, the levels of caspase-3, IL-1ß, and Beclin-1, and the ratios of Bcl-2/Bax and LC3II/LC3I between resveratrol pretreatment group and rosuvastatin pretreatment group. CONCLUSIONS: Synergistically enhanced antiapoptosis, anti-inflammation, and autophagy activation might be responsible for the synergistic neuroprotective effects of combining rosuvastatin with resveratrol on cerebral ischemia/reperfusion injury.


Assuntos
Encéfalo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Rosuvastatina Cálcica/farmacologia , Estilbenos/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/efeitos dos fármacos , Proteínas Relacionadas à Autofagia/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Citoproteção , Modelos Animais de Doenças , Sinergismo Farmacológico , Quimioterapia Combinada , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Mediadores da Inflamação/metabolismo , Masculino , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Resveratrol , Transdução de Sinais/efeitos dos fármacos
14.
Front Neurol ; 7: 181, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27822198

RESUMO

Rhinocerebral mucomycosis (RCM) as an emerging opportunistic, angioinvasive, and devastating fungi infection with high mortality is difficult to be diagnosed early because of the lack of specific clinical features or manifestations. Garcin syndrome is more often caused by skull base and rhinopharyngeal tumors or metastases, and basal meningitis. We reported that an aged diabetic man, involved nearly all cranial nerves (Garcin syndrome), who was at first suspected to be suffered from tuberculous meningitis, ultimately developed typically progressing RCM. Diagnosis was made to find the presence of mucormycosis in the infected tissue by biopsy.

15.
Oncotarget ; 7(45): 72537-72545, 2016 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-27705922

RESUMO

The immunoregulatory function of T regulatory cells (Tregs) is impaired in multiple sclerosis (MS). Recent studies have shown that umbilical cord-derived mesenchymal stem cells (UC-MSCs) exert regulatory effect on the functions of immune cells. Thus, we investigated whether UC-MSCs could improve the impaired function of Tregs from MS patients. Co-cultures of UC-MSCs with PBMCs of MS patients were performed for 3 days. Flow cytometry was used to determine the frequency of Tregs. A cell proliferation assay was used to evaluate the suppressive capacity of Tregs. ELISA was conducted for cytokine analysis in the co-cultures. Our results showed that UC-MSCs significantly increased the frequency of CD4+CD25+CD127low/- Tregs in resting CD4+ T cells (p<0.01) from MS, accompanied by the significantly augmented production of cytokine prostaglandin E2, transforming growth factor (-ß1, and interleukin-10, along with a reduced interferon-γ production in these co-cultures (p<0.05 - 0.01). More importantly, UC-MSC-primed Tregs of MS patients significantly inhibited the proliferation of PHA-stimulated autologous and allogeneic CD4+CD25- T effector cells (Teffs) from MS patients and healthy individuals compared to non-UC-MSC-primed (naïve) Tregs from the same MS patients (p<0.01). Furthermore, no remarkable differences in suppressing the proliferation of PHA-stimulated CD4+CD25- Teffs was observed in UC-MSC-primed Tregs from MS patients and naïve Tregs from healthy subjects. The impaired suppressive function of Tregs from MS can be completely reversed in a co-culture by UC-MSC modulation. This report is the first to demonstrate that functional defects of Tregs in MS can be repaired in vitro using a simple UC-MSC priming approach.


Assuntos
Técnicas de Cocultura/métodos , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Esclerose Múltipla/genética , Linfócitos T Reguladores/imunologia , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Imunomodulação , Masculino , Esclerose Múltipla/metabolismo
16.
Sci Rep ; 6: 32096, 2016 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-27554341

RESUMO

More than 240 million opioid prescriptions are dispensed annually to treat pain in the US. The use of opioids is commonly associated with opioid tolerance (OT) and opioid-induced hyperalgesia (OIH), which limit efficacy and compromise safety. The dearth of effective way to prevent or treat OT and OIH is a major medical challenge. We hypothesized that mesenchymal stem cells (MSCs) attenuate OT and OIH in rats and mice based on the understanding that MSCs possess remarkable anti-inflammatory properties and that both OT and chronic pain are associated with neuroinflammation in the spinal cord. We found that the development of OT and OIH was effectively prevented by either intravenous or intrathecal MSC transplantation (MSC-TP), which was performed before morphine treatment. Remarkably, established OT and OIH were significantly reversed by either intravenous or intrathecal MSCs when cells were transplanted after repeated morphine injections. The animals did not show any abnormality in vital organs or functions. Immunohistochemistry revealed that the treatments significantly reduced activation level of microglia and astrocytes in the spinal cord. We have thus demonstrated that MSC-TP promises to be a potentially safe and effective way to prevent and reverse two of the major problems of opioid therapy.


Assuntos
Tolerância a Medicamentos/fisiologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Morfina/farmacologia , Animais , Astrócitos/citologia , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Microglia/citologia , Microglia/efeitos dos fármacos , Ratos Sprague-Dawley , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos
17.
Front Neural Circuits ; 10: 29, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27147976

RESUMO

Transplantation of dopaminergic (DA) neurons is considered to be the most promising therapeutic strategy for replacing degenerated dopamine cells in the midbrain of Parkinson's disease (PD), thereby restoring normal neural circuit function and slow clinical progression of the disease. Human neural stem cells (hNSCs) derived from fetal forebrain are thought to be the important cell sources for producing DA neurons because of their multipotency for differentiation and long-term expansion property in cultures. However, low DA differentiation of the forebrain-derived hNSCs limited their therapeutic potential in PD. In the current study, we explored a combined application of Pramipexole (PRX), bone morphogenetic proteins 7 (BMP-7), and growth factors, including acidic fibroblast factor (aFGF), forskolin, and phorbol-12-myristae-13-acetate (TPA), to induce differentiation of forebrain-derived hNSCs toward DA neurons in cultures. We found that DA neuron-associated genes, including Nurr1, Neurogenin2 (Ngn2), and tyrosine hydroxylase (TH) were significantly increased after 24 h of differentiation by RT-PCR analysis (p < 0.01). Fluorescent examination showed that about 25% of cells became TH-positive neurons at 24 h, about 5% of cells became VMAT2 (vascular monoamine transporter 2)-positive neurons, and less than 5% of cells became DAT (dopamine transporter)-positive neurons at 72 h following differentiation in cultures. Importantly, these TH-, VMAT2-, and DAT-expressing neurons were able to release dopamine into cultures under both of the basal and evoked conditions. Dopamine levels released by DA neurons produced using our protocol were significantly higher compared to the control groups (P < 0.01), as examined by ELISA. Our results demonstrated that the combination of PRX, BMP-7, and growth factors was able to greatly promote differentiation of the forebrain-derived hNSCs into DA-releasing neurons.


Assuntos
Benzotiazóis/farmacologia , Proteína Morfogenética Óssea 7/farmacologia , Diferenciação Celular/efeitos dos fármacos , Agonistas de Dopamina/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Análise de Variância , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Combinação de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Pramipexol , Prosencéfalo/citologia , RNA Mensageiro/metabolismo , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas Vesiculares de Transporte de Monoamina/genética , Proteínas Vesiculares de Transporte de Monoamina/metabolismo
18.
J Stroke Cerebrovasc Dis ; 25(8): 1914-21, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27184615

RESUMO

BACKGROUND: It is well accepted that repetitive resveratrol (RV) pretreatment (PRC) exerts neuroprotective effect on ischemic stroke. RV was shown to be able to enhance the production of T regulatory cells (Tregs) in autoimmune diseases whereas Tregs are considered to be the cerebroprotective immunomodulator in ischemic stroke. Thus, we hypothesized whether Tregs contributed to PRC-induced neuroprotection against cerebral ischemia/reperfusion (I/R) injury. METHODS: Cerebral I/R injury was induced by middle cerebral artery occlusion for 90 minutes in rats. Adult male Sprague-Dawley rats were randomly assigned to 2 groups: I/R and RV I/R. RV (50 mg/kg) was administrated intraperitoneally once a day for 7 days prior to ischemia onset. RESULTS: PRC significantly ameliorated neurological defects and reduced cerebral infarct volume, accompanied by the significantly increased frequencies of Tregs in the spleens and ischemic hemisphere, the significantly increased levels of interleukin-10 (IL-10) in the plasma and ischemic hemisphere, and the significantly decreased levels of tumor necrosis factor-α and IL-6 in the plasma and ischemic hemisphere at 24 hours after ischemia onset. In addition, we also found that PRC significantly improved the frequency and suppressive function of Tregs in the spleens prior to ischemia onset. CONCLUSIONS: Thus, PRC-induced neuroprotection was in part mediated by more Treg accumulation and activation in vivo prior to ischemia onset except for less inflammation response at 24 hours after ischemia onset.


Assuntos
Antioxidantes/administração & dosagem , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle , Estilbenos/administração & dosagem , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Encéfalo/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Esquema de Medicação , Citometria de Fluxo , Infarto da Artéria Cerebral Média/complicações , Masculino , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/etiologia , Resveratrol , Baço/patologia , Linfócitos T Reguladores/fisiologia
19.
Cell Immunol ; 302: 26-31, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26774852

RESUMO

Bone marrow-derived mesenchymal stem cells (MSCs) are promising candidate cells for therapeutic application in autoimmune diseases due to their immunomodulatory properties. Unused human umbilical cords (UC) offer an abundant and noninvasive source of MSCs without ethical issues and are emerging as a valuable alternative to bone marrow tissue for producing MSCs. We thus investigated the immunomodulation effect of umbilical cord-derived MSCs (UC-MSCs) on human peripheral blood mononuclear cells (PBMCs), T cells in particular, in a co-culture system. We found that UC-MSCs efficiently suppressed the proliferation of phytohaemagglutinin (PHA)-stimulated PBMCs (p<0.01). Kinetic analysis revealed that UC-MSCs primarily inhibited the division of generation 3 (G3) and 4 (G4) of PBMCs. In addition, UC-MSCs augmented the expression of CD127(+) and CD45RA(+) but reduced the expression of CD25(+) in PBMCs stimulated by PHA (p<0.05). Furthermore, UC-MSCs inhibited PHA-resulted increase in the frequency of CD4(+)CD25(+)CD127(low/-) Tregs significantly (p<0.01) but augmented PHA-resulted increase in the frequency of CD4(+)CD25(high)CD45RA(+) Tregs to about three times in PBMCs. The levels of anti-inflammatory cytokines, PEG2, TGF-ß, and IL-10 were greatly up-regulated, accompanied by a significant down-regulation of pro-inflammatory IFN-γ in the co-culture (p<0.01). Our results showed that UC-MSCs are able to suppress mitogen-induced PBMC activation and proliferation in vitro by altering T lymphocyte phenotypes, increasing the frequency of CD4(+)CD25(high)CD45RA(+) Tregs, and modulating the associated cytokine production. Further studies are warranted to investigate the therapeutic potential of UC-MSCs in immunologically-diseased conditions.


Assuntos
Antígenos CD/metabolismo , Citocinas/metabolismo , Linfócitos/citologia , Células-Tronco Mesenquimais/fisiologia , Linfócitos T/citologia , Cordão Umbilical/citologia , Antígenos CD4 , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Humanos , Subunidade alfa de Receptor de Interleucina-2 , Antígenos Comuns de Leucócito , Ativação Linfocitária , Linfócitos/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Fito-Hemaglutininas/farmacologia , Linfócitos T/imunologia
20.
Neurosci Lett ; 610: 43-7, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26497914

RESUMO

It is well known that neurons differentiated from SH-SY5Y cells can serve as cell models for neuroscience research; i.e., neurotoxicity and tolerance to morphine in vitro. To differentiate SH-SY5Y cells into neurons, RA (retinoic acid) is commonly used to produce the inductive effect. However, the percentage of neuronal cells produced from SH-SY5Y cells is low, either from the use of RA treatment alone or from the combined application of RA and other chemicals. In the current study, we used CM-hNSCs (conditioned medium of human neural stem cells) as the combinational inducer with RA to prompt neuronal differentiation of SH-SY5Y cells. We found that neuronal differentiation was improved and that neurons were greatly increased in the differentiated SH-SY5Y cells using a combined treatment of CM-hNSCs and RA compared to RA treatment alone. The neuronal percentage was higher than 80% (about 88%) on the 3rd day and about 91% on the 7th day examined after a combined treatment with CM-hNSCs and RA. Cell maturation and neurite growth of these neuronal cells were also improved. In addition, the use of CM-hNSCs inhibited the apoptosis of RA-treated SH-SY5Y cells in culture. We are the first to report the use of CM-hNSCs in combination with RA to induce neuronal differentiation of RA-treated SH-SY5Y cells. Our method can rapidly and effectively promote the neuronal production of SH-SY5Y cells in culture conditions.


Assuntos
Células-Tronco Neurais/citologia , Neurônios/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Meios de Cultivo Condicionados , Humanos , Células-Tronco Neurais/efeitos dos fármacos , Neuroblastoma , Neurônios/efeitos dos fármacos , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA