Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 7497, 2023 11 18.
Article in English | MEDLINE | ID: mdl-37980356

ABSTRACT

The degenerative process in Parkinson's disease (PD) causes a progressive loss of dopaminergic neurons (DaNs) in the nigrostriatal system. Resolving the differences in neuronal susceptibility warrants an amenable PD model that, in comparison to post-mortem human specimens, controls for environmental and genetic differences in PD pathogenesis. Here we generated high-quality profiles for 250,173 cells from the substantia nigra (SN) and putamen (PT) of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkinsonian macaques and matched controls. Our primate model of parkinsonism recapitulates important pathologic features in nature PD and provides an unbiased view of the axis of neuronal vulnerability and resistance. We identified seven molecularly defined subtypes of nigral DaNs which manifested a gradient of vulnerability and were confirmed by fluorescence-activated nuclei sorting. Neuronal resilience was associated with a FOXP2-centered regulatory pathway shared between PD-resistant DaNs and glutamatergic excitatory neurons, as well as between humans and nonhuman primates. We also discovered activation of immune response common to glial cells of SN and PT, indicating concurrently activated pathways in the nigrostriatal system. Our study provides a unique resource to understand the mechanistic connections between neuronal susceptibility and PD pathophysiology, and to facilitate future biomarker discovery and targeted cell therapy.


Subject(s)
Parkinson Disease , Parkinsonian Disorders , Animals , Humans , Mice , Parkinson Disease/metabolism , Parkinsonian Disorders/metabolism , Substantia Nigra/metabolism , Dopaminergic Neurons/metabolism , Macaca , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Disease Models, Animal , Mice, Inbred C57BL
3.
Nat Protoc ; 18(6): 1930-1957, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37045994

ABSTRACT

The low number of neural progenitor cells (NPCs) present in the adult and aged primate brains represents a challenge for generating high-yield and viable in vitro cultures of primary brain cells. Here we report a step-by-step approach for the fast and reproducible isolation of high-yield and viable primary brain cells, including mature neurons, immature cells and NPCs, from adult and aged macaques. We describe the anesthesia, transcardial perfusion and brain tissue preparation; the subsequent microdissection of the regions of interest and their enzymatic dissociation, leading to the separation of single cells. The cell isolation steps of our protocol can also be used for routine cell culturing, in particular for NPC expansion and differentiation, suitable for studies of hippocampal neurogenesis in the adult macaque brain. The purified primary brain cells are largely free from myelin debris and erythrocytes, paving the way for multiple downstream applications in vitro and in vivo. When combined with single-cell profiling techniques, this approach allows an unbiased and comprehensive mapping of cell states in the adult and aged macaque brain, which is needed to advance our understanding of human cognitive and neurological diseases. The neural cell isolation protocol requires 4 h and a team of four to six users with expertize in primary brain cell isolation to avoid tissue hypoxia during the time-sensitive steps of the procedure.


Subject(s)
Cell Culture Techniques , Neural Stem Cells , Animals , Adult , Humans , Aged , Cell Culture Techniques/methods , Neurons , Cells, Cultured , Cell Differentiation/physiology , Cell Separation
4.
Zool Res ; 44(1): 153-168, 2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36484227

ABSTRACT

Strabismus and amblyopia are common ophthalmologic developmental diseases caused by abnormal visual experiences. However, the underlying pathogenesis and visual defects are still not fully understood. Most studies have used experimental interference to establish disease-associated animal models, while ignoring the natural pathophysiological mechanisms. This study was designed to investigate whether natural strabismus and amblyopia are associated with abnormal neurological defects. We screened one natural strabismic monkey ( Macaca fascicularis) and one natural amblyopic monkey from hundreds of monkeys, and retrospectively analyzed one human strabismus case. Neuroimaging, behavioral, neurophysiological, neurostructural, and genovariation features were systematically evaluated using magnetic resonance imaging (MRI), behavioral tasks, flash visual evoked potentials (FVEP), electroretinogram (ERG), optical coherence tomography (OCT), and whole-genome sequencing (WGS), respectively. Results showed that the strabismic patient and natural strabismic and amblyopic monkeys exhibited similar abnormal asymmetries in brain structure, i.e., ipsilateral impaired right hemisphere. Visual behavior, visual function, retinal structure, and fundus of the monkeys were impaired. Aberrant asymmetry in binocular visual function and structure between the strabismic and amblyopic monkeys was closely related, with greater impairment of the left visual pathway. Several similar known mutant genes for strabismus and amblyopia were also identified. In conclusion, natural strabismus and amblyopia are accompanied by abnormal asymmetries of the visual system, especially visual neurophysiological and neurostructural defects. Our results suggest that future therapeutic and mechanistic studies should consider defects and asymmetries throughout the entire visual system.


Subject(s)
Evoked Potentials, Visual , Visual Pathways , Animals , Humans , Retrospective Studies , Haplorhini
5.
Exp Eye Res ; 227: 109365, 2023 02.
Article in English | MEDLINE | ID: mdl-36577484

ABSTRACT

Retinal pigment epithelial (RPE) cellular senescence is regarded as an initiator for age-related macular degeneration (AMD). We previously demonstrated that by the coculture way, embryonic stem cells (ESCs) can reverse the senescence of RPE cells, but xenograft cells can cause a plethora of adverse effects. Extracellular vesicles (EVs) derived from ESCs can act as messengers to mediate nearby cell activities and have the same potential as ESCs to reverse RPE senescence. Furthermore, ESC-EVs have achieved preliminary efficacy while treating many age-related diseases. The present study aimed to test the effect of ESC-EVs on the replicative senescence model of RPE cells as well as its mechanism. The results showed that ESC-EVs enhanced the proliferative ability and cell cycle transition of senescent RPE cells, whereas reduced the senescence-associated galactosidase (SA-ß-gal) staining rate, as well as the levels of mitochondrial membrane potential (MMP) and reactive oxygen species (ROS). Moreover, classical markers of cellular senescence p21WAF1/CIP1 (p21) and p16INK4a (p16) were downregulated. The bioinformatic analysis and further study showed that the inhibition of the p38MAPK pathway by ESC-EVs played a pivotal role in RPE cellular senescence-reversing effect, which was ameliorated or even abolished when dehydrocorydaline were administrated simultaneously, demonstrating that ESC-EVs can effectively reverse RPE cellular senesence by inhibiting the p38MAPK pathway, thus highlights the potential of ESC-derived EVs as biomaterials for preventative and protective therapy in AMD.


Subject(s)
Extracellular Vesicles , Retinal Pigment Epithelium , Humans , Retinal Pigment Epithelium/metabolism , Embryonic Stem Cells , Epithelial Cells , Retinal Pigments/metabolism , Cellular Senescence
6.
Cell Rep ; 40(11): 111322, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36103830

ABSTRACT

Retinal ganglion cells (RGCs) are the brain's gateway to the visual world. They can be classified into different types on the basis of their electrophysiological, transcriptomic, or morphological characteristics. Here, we characterize the transcriptomic, morphological, and functional features of 472 high-quality RGCs using Patch sequencing (Patch-seq), providing functional and morphological annotation of many transcriptomic-defined cell types of a previously established RGC atlas. We show a convergence of different modalities in defining the RGC identity and reveal the degree of correspondence for well-characterized cell types across multimodal data. Moreover, we complement some RGC types with detailed morphological and functional properties. We also identify differentially expressed genes among ON, OFF, and ON-OFF RGCs such as Vat1l, Slitrk6, and Lmo7, providing candidate marker genes for functional studies. Our research suggests that the molecularly distinct clusters may also differ in their roles of encoding visual information.


Subject(s)
Retinal Ganglion Cells , Transcriptome , Animals , Mammals , Phenotype , Retinal Ganglion Cells/metabolism , Transcriptome/genetics
7.
ACS Omega ; 6(36): 23203-23210, 2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34549121

ABSTRACT

Synthesizing novel photocatalysts that can effectively harvest photon energy over a wide range of the solar spectrum for practical applications is vital. Porphyrin-derived nanostructures with properties similar to those of chlorophyll have emerged as promising candidates to meet this requirement. In this study, tetrakis(4-carboxyphenyl) porphyrin (TCPP) nanofibers were formed on the surface of ZnO nanoparticles using a simple self-assembly approach. The obtained ZnO/TCPP nanofiber composites were characterized via scanning electron microscopy, X-ray diffraction analysis, and ultraviolet-visible absorbance and reflectance measurements. The results demonstrated that the ZnO nanoparticles with an average size of approximately 37 nm were well integrated in the TCPP nanofiber matrix. The resultant composite showed photocatalytic activity of ZnO and TCPP nanofibers concomitantly, with band gap energies of 3.12 and 2.43 eV, respectively. The ZnO/TCPP photocatalyst exhibited remarkable photocatalytic performance for RhB degradation with a removal percentage of 97% after 180 min of irradiation under simulated sunlight because of the synergetic activity of ZnO and TCPP nanofibers. The dominant active species participating in the photocatalytic reaction were •O2 - and OH•, resulting in enhanced charge separation by exciton-coupled charge-transfer processes between the hybrid materials.

8.
Front Oncol ; 11: 584884, 2021.
Article in English | MEDLINE | ID: mdl-33898304

ABSTRACT

Acute myeloid leukemia (AML) is malignant hematologic tumors with frequent recurrence and cause high mortality. Its fate is determined by abnormal intracellular competitive endogenous RNA (ceRNA) network and extracellular tumor microenvironment (TME). This study aims to build a ceRNA network related to AML TME to explore new prognostic and therapeutic targets. The RNA expression data of AML were obtained from The Cancer Genome Atlas (TCGA) database. First, we used the ESTIMATE algorithm to calculate the immune cells and stromal cells infiltration scores in the TME and found that all scores were highly correlated with AML's prognostic characteristics. Subsequently, differentially expressed mRNAs and lncRNAs between high and low score groups were identified to construct a TME-related ceRNA network. Further, the Cox-lasso survival model was employed to screen out the hub prognostic ceRNA network composed of two mRNAs (EPB41L3, COL2A1), three miRNAs (hsa-mir-26a-5p, hsa-mir-148b-3p, hsa-mir-148a-3p), and two lncRNAs (CYP1B1-AS1, C9orf106), and construct nomograms. Finally, we used CIBERSORT algorithm and Kaplan-Meier survival analysis to identify the prognostic TME immune cells and found that naive B cells, M2-type macrophages, and helper follicular T cells were related to prognosis, and the hub ceRNAs were highly correlated with immune cell infiltration. This study provided a new perspective to elucidate how TME regulates AML process and put forward the new therapy strategies combining targeting tumor cells with disintegrating TME.

9.
J Cell Physiol ; 236(7): 4783-4796, 2021 07.
Article in English | MEDLINE | ID: mdl-33368322

ABSTRACT

Senescent cells can secrete a plethora of cytokines which induce senescent phenotype of neighboring cells and was called senescence-associated secretory phenotype. Previously, it was believed that cancer was caused by the infinite division and uncontrolled proliferation of cells. Based on this, anticancer treatments were all aimed at killing cancer cells. Cancer is now considered an age-related disease. Cancer cells are not exogenous, but one of the worst results of injuries which initially induce cell senescence. Therefore, reversing cell senescence can fundamentally prevent and treat cancer. Though current anticancer treatments induce the cancer cells apoptosis, they induce senescence of normal cells at the same time, thus promoting the occurrence and development of cancer and forming a vicious circle. Extracellular vesicles (EVs) are nano-sized vesicles which partially mirror their parent cells. In the tumor microenvironment, EVs of senescent cells can change the expression profile of cancer cells, contributing to their resistance to chemotherapy. There is growing evidence indicates that stem cell EVs exert effective antiaging and anticancer actions by transferring functional microRNAs and proteins. This review will summarize the therapeutic role of stem cell EVs in reversing aging and cancer, which suggests the broad clinical application perspective.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Extracellular Vesicles/metabolism , Neoplasms/pathology , Neoplasms/therapy , Neoplastic Stem Cells/metabolism , Apoptosis , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/genetics , Tumor Microenvironment/physiology
10.
Transl Cancer Res ; 10(1): 487-498, 2021 Jan.
Article in English | MEDLINE | ID: mdl-35116278

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is the most common malignant tumor of the central nervous system, accounting for 48.6% of malignant tumors. The current standard treatment plan includes the widest range of safe surgical resection, supplemented by local brain radiotherapy and temozolomide concurrent chemotherapy; this can cause serious side effects. Even so, the median survival time of GBM patients is only 8 months, and the 5-year survival rate is only 5.5%. It is imminent to find new treatments. Early studies have shown that chicken and zebrafish embryos can reprogram cancer cells into a non-tumorigenic phenotype through the embryonic microenvironment. However, the effect of embryonic stem cell microenvironment on GBM and its possible mechanism are not clear. METHODS: In this study, the glioblastoma cell line, U118, in the brain was investigated. There were four experimental groups: GB, GE, GA and GT. U118 cells were harvested after culturing for 72 hours. Cell proliferation, apoptosis, reactive oxygen species (ROS) were examined using vasculogenic mimicry assays, quantitative real-time polymerase chain reaction (QRT-PCR), western blotting (WB) and flow cytometry. The differences in the biological function of U118 cells and the PI3K/protein kinase B (AKT) signaling pathway were compared between the groups. RESULTS: Compared with the GB control group, the GE co-culture group and GT chemotherapy group showed reduced cell proliferation, increased apoptosis, increased ROS, as well as decreased or inhibited vasculogenic mimicry. Expressions of cyclin B1 and cyclin D1 were also notably reduced, while that of Bax, Bcl-2, p53, Caspase-3, GSK-3ß, p21, and p27 were significantly increased. Moreover, the expression of PI3K, AKT, and mTOR were markedly decreased, whereas expression of PTEN increased considerably. Also, the expression of positive regulatory factors significantly increased, however negative regulatory factors decreased in the GA group compared to the GE group. CONCLUSIONS: The ESC microenvironment reverses glioma malignancy, partially via inhibition of the PI3K signaling pathway. Our study may have a significant impact and important clinical implications for cell therapy in the treatment of glioma.

11.
Front Oncol ; 10: 577072, 2020.
Article in English | MEDLINE | ID: mdl-33194692

ABSTRACT

Cutaneous melanoma is the most life-threatening skin malignant tumor due to its increasing metastasis and mortality rate. The abnormal competitive endogenous RNA network promotes the development of tumors and becomes biomarkers for the prognosis of various tumors. At the same time, the tumor immune microenvironment (TIME) is of great significance for tumor outcome and prognosis. From the perspective of TIME and ceRNA network, this study aims to explain the prognostic factors of cutaneous melanoma systematically and find novel and powerful biomarkers for target therapies. We obtained the transcriptome data of cutaneous melanoma from The Cancer Genome Atlas (TCGA) database, 3 survival-related mRNAs co-expression modules and 2 survival-related lncRNAs co-expression modules were identified through weighted gene co-expression network analysis (WCGNA), and 144 prognostic miRNAs were screened out by univariate Cox proportional hazard regression. Cox regression model and Kaplan-Meier survival analysis were employed to identify 4 hub prognostic mRNAs, and the prognostic ceRNA network consisting of 7 lncRNAs, 1 miRNA and 4 mRNAs was established. After analyzing the composition and proportion of total immune cells in cutaneous melanoma microenvironment through CIBERSORT algorithm, it is found through correlation analysis that lncRNA-TUG1 in the ceRNA network was closely related to the TIME. In this study, we first established cutaneous melanoma's TIME-related ceRNA network by WGCNA. Cutaneous melanoma prognostic markers have been identified from multiple levels, which has important guiding significance for clinical diagnosis, treatment, and further scientific research on cutaneous melanoma.

12.
Genes (Basel) ; 11(8)2020 07 31.
Article in English | MEDLINE | ID: mdl-32751923

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) is one of the most common malignant and aggressive hematologic tumors, and its pathogenesis is associated with abnormal post-transcriptional regulation. Unbalanced competitive endogenous RNA (ceRNA) promotes tumorigenesis and progression, and greatly contributes to tumor risk classification and prognosis. However, the comprehensive analysis of the circular RNA (circRNA)-long non-coding RNA (lncRNA)-miRNA-mRNA ceRNA network in the prognosis of AML is still rarely reported. METHOD: We obtained transcriptome data of AML and normal samples from The Cancer Genome Atlas (TCGA), Genotype-tissue Expression (GTEx), and Gene Expression Omnibus (GEO) databases, and identified differentially expressed (DE) mRNAs, lncRNAs, and circRNAs. Then, the targeting relationships among lncRNA-miRNA, circRNA-miRNA, and miRNA-mRNA were predicted, and the survival related hub mRNAs were further screened by univariate and multivariate Cox proportional hazard regression. Finally, the AML prognostic circRNA-lncRNA-miRNA-mRNA ceRNA regulatory network was established. RESULTS: We identified prognostic 6 hub mRNAs (TM6SF1, ZMAT1, MANSC1, PYCARD, SLC38A1, and LRRC4) through Cox regression model, and divided the AML samples into high and low risk groups according to the risk score obtained by multivariate Cox regression. Survival analysis verified that the survival rate of the high-risk group was significantly reduced (p < 0.0001). The prognostic ceRNA network of 6 circRNAs, 32 lncRNAs, 8 miRNAs, and 6 mRNAs was established according to the targeting relationship between 6 hub mRNAs and other RNAs. CONCLUSION: In this study, ceRNA network jointly participated by circRNAs and lncRNAs was established for the first time. It comprehensively elucidated the post-transcriptional regulatory mechanism of AML, and identified novel AML prognostic biomarkers, which has important guiding significance for the clinical diagnosis, treatment, and further scientific research of AML.


Subject(s)
Biomarkers, Tumor/genetics , Gene Regulatory Networks , Leukemia, Myeloid, Acute/genetics , Transcriptome , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism , Biomarkers, Tumor/metabolism , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Cell Prolif ; 53(8): e12865, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32588948

ABSTRACT

The tumour microenvironment (TME) plays a pivotal role in tumour fate determination. The TME acts together with the genetic material of tumour cells to determine their initiation, metastasis and drug resistance. Stromal cells in the TME promote the growth and metastasis of tumour cells by secreting soluble molecules or exosomes. The abnormal microenvironment reduces immune surveillance and tumour killing. The TME causes low anti-tumour drug penetration and reactivity and high drug resistance. Tumour angiogenesis and microenvironmental hypoxia limit the drug concentration within the TME and enhance the stemness of tumour cells. Therefore, modifying the TME to effectively attack tumour cells could represent a comprehensive and effective anti-tumour strategy. Normal cells, such as stem cells and immune cells, can penetrate and disrupt the abnormal TME. Reconstruction of the TME with healthy cells is an exciting new direction for tumour treatment. We will elaborate on the mechanism of the TME to support tumours and the current cell therapies for targeting tumours and the TME-such as immune cell therapies, haematopoietic stem cell (HSC) transplantation therapies, mesenchymal stem cell (MSC) transfer and embryonic stem cell-based microenvironment therapies-to provide novel ideas for producing breakthroughs in tumour therapy strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Mesenchymal Stem Cells/drug effects , Neovascularization, Pathologic/drug therapy , Tumor Microenvironment/drug effects , Exosomes/drug effects , Exosomes/pathology , Humans , Neoplasms/drug therapy , Neoplasms/pathology
14.
Genes (Basel) ; 11(2)2020 02 21.
Article in English | MEDLINE | ID: mdl-32098099

ABSTRACT

Growing evidence has revealed that abnormal alternative splicing (AS) events are closely related to carcinogenic processes. However, the comprehensive study on the prognostic value of splicing events involved in uveal melanoma (UM) is still lacking. Therefore, splicing data of 80 UM patients were obtained from the Cancer Genome Atlas (TCGA) SpliceSeq and RNA sequence data of UM and patient clinical features were downloaded from the Cancer Genome Atlas (TCGA) database to identify survival related splicing events in UM. As a result, a total of 37996 AS events of 17911 genes in UM were detected, among which 5299 AS events of 3529 genes were significantly associated with UM patients' survival. Functional enrichment analysis revealed that this survival related splicing genes are corelated with mRNA catabolic process and ribosome pathway. Based on survival related splicing events, seven types of prognostic markers and the final overall prognostic signature could independently predict the overall survival of UM patients. Finally, an 11 spliced gene was identified in the final signature. On the basis of these 11 genes, we constructed a Support Vector Machine (SVM) classifier and evaluated it with leave-one-out cross-validation. The results showed that the 11 genes could determine short- and long-term survival with a predicted accuracy of 97.5%. Besides, the splicing factors and alternative splicing events correlation network was constructed to serve as therapeutic targets for UM treatment. Thus, our study depicts a comprehensive landscape of alternative splicing events in the prognosis of UM. The correlation network and associated pathways would provide additional potential targets for therapy and prognosis.


Subject(s)
Alternative Splicing/genetics , Melanoma/genetics , Melanoma/mortality , Uveal Neoplasms/genetics , Uveal Neoplasms/mortality , Alternative Splicing/physiology , Biomarkers, Tumor/genetics , Databases, Genetic , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Humans , Male , Melanoma/metabolism , Prognosis , RNA Splicing Factors/genetics , RNA, Messenger/genetics , Uveal Neoplasms/metabolism
15.
Theranostics ; 9(16): 4764-4778, 2019.
Article in English | MEDLINE | ID: mdl-31367256

ABSTRACT

The currently used anti-cancer therapies work by killing cancer cells but result in adverse effects and resistance to treatment, which accelerates aging and causes damage to normal somatic cells. On one hand, chicken and zebrafish embryos can reprogram cancer cells towards a non-tumorigenic phenotype; however, they cannot be used in the clinical practice. On the other hand, embryonic stem cells (ESCs) mimic the early embryonic microenvironment and are easily available. We investigated the therapeutic efficacy of the ESC microenvironment (ESCMe) in human uveal melanoma in vitro and in vivo. Methods: Human uveal melanoma C918 cells co-cultured with ESCs were used to measure the levels of mRNA and protein of the phosphoinositide 3-kinase (PI3K) pathway. Cell proliferation, invasiveness, and tumorigenicity of C918 cells were also analyzed. To mimic the tumor microenvironment in vivo, we co-cultured C918 cells and normal somatic cells with ESCs in a co-culture system and evaluated the therapeutic potential of ESCMe in both cell types. For an in vivo study, a mouse tumor model was used to test the safety and efficacy of the transplanted ESC. Elimination of the transplanted ESCs in mice was carried out by using the ESC-transfected with a thymidine kinase suicidal gene followed by administration of ganciclovir to prevent the formation of teratomas by ESCs. Results: In vitro studies confirmed that ESCMe inhibits the proliferation, invasiveness, and tumorigenicity of C918 cells, and the PI3K agonist abolished these effects. ESCMe suppressed the various malignant behaviors of uveal melanoma cells but enhanced the proliferation of normal somatic cells both in vitro and in vivo. Further, we demonstrated that ESCMe suppressed the PI3K pathway in tumor cells but activated in somatic cells. Conclusions: The ESCMe can effectively suppress the malignant phenotype of uveal melanoma cells and modulate the tumor-promoting aging environment by preventing the senescence of normal cells through the bidirectional regulation of the PI3K signaling. Our results suggest that ESC transplantation can serve as an effective and safe approach for treating cancer without killing cells.


Subject(s)
Embryonic Stem Cells/cytology , Melanoma/physiopathology , Tumor Microenvironment , Uveal Neoplasms/physiopathology , Animals , Cell Line, Tumor , Cell Proliferation , Embryonic Stem Cells/metabolism , Humans , Male , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Uveal Neoplasms/genetics , Uveal Neoplasms/metabolism
16.
Oxid Med Cell Longev ; 2019: 3632169, 2019.
Article in English | MEDLINE | ID: mdl-31249643

ABSTRACT

Age-related macular degeneration (AMD) is a blinding disease caused by multiple factors and is the primary cause of vision loss in the elderly. The morbidity of AMD increases every year. Currently, there is no effective treatment option for AMD. Intravitreal injection of antivascular endothelial growth factor (anti-VEGF) is currently the most widely used therapy, but it only aims at neovascularization, which is an intermediate pathological phenomenon of wet AMD, not at the etiological treatment. Anti-VEGF therapy can only temporarily delay the degeneration process of wet AMD, and AMD is easy to relapse after drug withdrawal. Therefore, it is urgent to deepen our understanding of the pathophysiological processes underlying AMD and to identify integrated or new strategies for AMD prevention and treatment. Recent studies have found that autophagy dysfunction in retinal pigment epithelial (RPE) cells, cellular senescence, and abnormal immune-inflammatory responses play key roles in the pathogenesis of AMD. For many age-related diseases, the main focus is currently the clearing of senescent cells (SNCs) as an antiaging treatment, thereby delaying diseases. However, in AMD, there is no relevant antiaging application. This review will discuss the pathogenesis of AMD and how interactions among RPE autophagy dysfunction, cellular senescence, and abnormal immune-inflammatory responses are involved in AMD, and it will summarize the three antiaging strategies that have been developed, with the aim of providing important information for the integrated prevention and treatment of AMD and laying the ground work for the application of antiaging strategies in AMD treatment.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Autophagy , Cellular Senescence , Inflammation/drug therapy , Macular Degeneration/pathology , Macular Degeneration/prevention & control , Neuroprotective Agents/therapeutic use , Humans , Macular Degeneration/immunology , Macular Degeneration/metabolism , Prognosis
17.
Cancer Med ; 8(9): 4265-4277, 2019 08.
Article in English | MEDLINE | ID: mdl-31173492

ABSTRACT

Malignant cancer cells engage in a dynamic reciprocity with the tumor microenvironment (TME) that promotes tumor growth, development, and resistance to therapy. Early embryonic blastocyst microenvironments can reverse the tumorigenic phenotype of malignant cancer cells via ameliorating of TME. It is potential to apply embryonic stem cell (ESC) microenvironment to suppress the malignant behaviors of cancer cells. This study aimed to investigate a better method and the mechanism of ESC microenvironment supplied by ESCs on suppressing the malignancy of cutaneous melanoma cells. Cutaneous melanoma cell line A2058 were cultured and divided into four groups: (a) A2058-only (Control); (b) A2058 and ESCs continuously co-cultured (Group One); (c) A2058 co-cultured with daily refreshed ESCs (Group two); (d) Group one with VO-Ohpic, inhibitor of PTEN (VO-Ohpic Group). The results showed that, compared to control group, A2058 cells in group one exhibited decreased cellular proliferation, migration, invasiveness and vasculogenic mimicry concomitant with an increase in cell apoptosis, accompanied by down-regulation of PI3K/AKT pathway. Besides, the above mentioned anti-tumor effects on A2058 cells were significantly enhanced in group two but statistically weakened after administration of VO-Ohpic compared to group one. We demonstrate that ESC microenvironment reduces the malignancy of A2058 by down-regulating PI3K/AKT pathway. Notably, such anti-tumor effects can be enhanced by appropriately increasing the quality and quantity of ESCs in co-culture system. Our results suggest that ESC microenvironment could be an effective and safe approach to treating cancer.


Subject(s)
Embryonic Stem Cells/cytology , Gene Regulatory Networks , Melanoma/genetics , Signal Transduction , Skin Neoplasms/genetics , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Coculture Techniques , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks/drug effects , Humans , Melanoma/metabolism , Melanoma/therapy , Organometallic Compounds/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/therapy , Stem Cell Niche , Tumor Microenvironment , Melanoma, Cutaneous Malignant
18.
Int J Ophthalmol ; 11(9): 1496-1502, 2018.
Article in English | MEDLINE | ID: mdl-30225224

ABSTRACT

AIM: To explore a new diagnostic index for differentiating the evaporative dry eye (EDE) subtypes by analysis of their respective clinical characteristics. METHODS: A cross-sectional study of 139 patients (139 eyes) with EDE who were enrolled and classified as obstructive meibomian gland dysfunction (MGD) (n=81) and non-obstructive MGD (n=58) EDE. All patients completed a Standard Patient Evaluation of Eye Dryness (SPEED) questionnaire and were evaluated for average lipid layer thickness (LLT), tear meniscus height measurements (TMH), tear break-up time (TBUT), ocular surface staining score, Schirmer I test (SIT), lid margin abnormalities, and meibomian gland function and morphology. RESULTS: Age, average LLT, TMH, scores of lid margin abnormalities, meibum quality, meibomian gland loss (MGL) (all P≤0.001), and TBUT (P=0.03) were all significantly different between obstructive MGD EDE patients and non-obstructive MGD EDE patients. Average LLT in obstructive MGD EDE was correlated with meibomian expressibility (r=-0.541, P≤0.001), lid margin abnormalities were marginally not significant (r=0.197, P=0.077), and TMH was correlated with MGL (total MGL: r=0.552, P≤0.001; upper MGL: r=0.438, P≤0.001; lower MGL: r=0.407, P≤0.001). Average LLT in non-obstructive MGD EDE, was correlated with meibomian expressibility and Oxford staining (r=-0.396, P=0.002; r=-0.461, P≤0.001). The efficiency of combining average LLT and TMH was optimal, with a sensitivity of 80.2% and a specificity of 74.1%. Obstructive MGD EDE patients had an average LLT≥69 nm and TMH≥0.25 mm, while non-obstructive MGD EDE patients had an average LLT<69 nm and TMH<0.25 mm. CONCLUSION: Obstructive MGD EDE and non-obstructive MGD EDE have significantly different clinical characteristics. Combining average LLT and TMH measurements enhanced their reliability for differentiating these two subtypes and provided guidance for offering more precise treatments for EDE subtypes.

19.
Int J Ophthalmol ; 11(3): 389-394, 2018.
Article in English | MEDLINE | ID: mdl-29600171

ABSTRACT

AIM: To investigate the effects of different concentrations of artificial tears on lipid layer thickness (LLT) and blink rate (BR) in dry eye patients. METHODS: This study included 106 eyes of 58 patients with dry eye. The lipid deficiency type was defined as the LLT baseline <75 nm. The LLT and BR were measured at baseline and 1, 5 and 15min after the instillation of 0.1% or 0.3% sodium hyaluronate (SH) eye drops by using the LipiView ocular surface interferometer. RESULTS: In the lipid deficiency group, the LLT increased from baseline at 1min post instillation. The LLT after the instillation of 0.1% SH was significantly higher than that after the instillation of 0.3% SH (P<0.001). The LLT returned to baseline at 15min post instillation of 0.1% SH and at 5min post instillation of 0.3% SH. In the non-lipid deficiency group, the LLT decreased from baseline at 1min and returned to baseline at 5min for both treatments. The BRs were not significantly different at different time points for both treatments. CONCLUSION: SH eye drops induce a short-term increase in LLT of patients with lipid deficiency. A low concentration of artificial tears have a stronger effect than a high concentration of artificial tears on the increase in LLT. In comparison, SH eye drops induce a transient and slight decrease in LLT of patients without lipid deficiency. A low concentration of artificial tears might be better for patients with lipid deficiency.

SELECTION OF CITATIONS
SEARCH DETAIL
...