Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 176: 116806, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38796971

ABSTRACT

Central nervous system (CNS) damage is usually irreversible owing to the limited regenerative capability of neurons. Following CNS injury, astrocytes are reactively activated and are the key cells involved in post-injury repair mechanisms. Consequently, research on the reprogramming of reactive astrocytes into neurons could provide new directions for the restoration of neural function after CNS injury and in the promotion of recovery in various neurodegenerative diseases. This review aims to provide an overview of the means through which reactive astrocytes around lesions can be reprogrammed into neurons, to elucidate the intrinsic connection between the two cell types from a neurogenesis perspective, and to summarize what is known about the neurotranscription factors, small-molecule compounds and MicroRNA that play major roles in astrocyte reprogramming. As the malignant proliferation of astrocytes promotes the development of glioblastoma multiforme (GBM), this review also examines the research advances on and the theoretical basis for the reprogramming of GBM cells into neurons and discusses the advantages of such approaches over traditional treatment modalities. This comprehensive review provides new insights into the field of GBM therapy and theoretical insights into the mechanisms of neurological recovery following neurological injury and in GBM treatment.


Subject(s)
Astrocytes , Brain Neoplasms , Cellular Reprogramming , Glioblastoma , Neurons , Humans , Astrocytes/metabolism , Astrocytes/pathology , Glioblastoma/pathology , Neurons/metabolism , Neurons/pathology , Animals , Brain Neoplasms/pathology , Neurogenesis , Central Nervous System/pathology
2.
Ageing Res Rev ; 98: 102324, 2024 07.
Article in English | MEDLINE | ID: mdl-38762100

ABSTRACT

Progressive neuronal dysfunction and death are key features of neurodegenerative diseases; therefore, promoting neurogenesis in neurodegenerative diseases is crucial. With advancements in proteomics and high-throughput sequencing technology, it has been demonstrated that histone post-transcriptional modifications (PTMs) are often altered during neurogenesis when the brain is affected by disease or external stimuli and that the degree of histone modification is closely associated with the development of neurodegenerative diseases. This review aimed to show the regulatory role of histone modifications in neurogenesis and neurodegenerative diseases by discussing the changing patterns and functional significance of histone modifications, including histone methylation, acetylation, ubiquitination, phosphorylation, and lactylation. Finally, we explored the control of neurogenesis and the development of neurodegenerative diseases by artificially modulating histone modifications.


Subject(s)
Histones , Neurodegenerative Diseases , Neurogenesis , Protein Processing, Post-Translational , Neurogenesis/physiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/genetics , Humans , Histones/metabolism , Animals , Histone Code
3.
Redox Biol ; 69: 103030, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38181705

ABSTRACT

Ferroptosis is a type of programmed cell death resulting from iron overload-dependent lipid peroxidation, and could be promoted by activating transcription factor 3 (ATF3). SIRT1 is an enzyme accounting for removing acetylated lysine residues from target proteins by consuming NAD+, but its role remains elusive in ferroptosis and activating ATF3. In this study, we found SIRT1 was activated during the process of RSL3-induced glioma cell ferroptosis. Moreover, the glioma cell death was aggravated by SIRT1 activator SRT2183, but suppressed by SIRT inhibitor EX527 or when SIRT1 was silenced with siRNA. These indicated SIRT1 sensitized glioma cells to ferroptosis. Furthermore, we found SIRT1 promoted RSL3-induced expressional upregulation and nuclear translocation of ATF3. Silence of ATF3 with siRNA attenuated RSL3-induced increases of ferrous iron and lipid peroxidation, downregulation of SLC7A11 and GPX4 and depletion of cysteine and GSH. Thus, SIRT1 promoted glioma cell ferroptosis by inducting ATF3 activation. Mechanistically, ATF3 activation was reinforced when RSL3-induced decline of NAD+ was aggravated by FK866 that could inhibit NAD + synthesis via salvage pathway, but suppressed when intracellular NAD+ was maintained at higher level by supplement of exogenous NAD+. Notably, the NAD + decline caused by RSL3 was enhanced when SIRT1 was further activated by SRT2183, but attenuated when SIRT1 activation was inhibited by EX527. These indicated SIRT1 promoted ATF3 activation via consumption of NAD+. Finally, we found RSL3 activated SIRT1 by inducing reactive oxygen species-dependent upregulation of AROS. Together, our study revealed SIRT1 activated by AROS sensitizes glioma cells to ferroptosis via activation of ATF3-dependent inhibition of SLC7A11 and GPX4.


Subject(s)
Ferroptosis , Glioma , Humans , NAD , Activating Transcription Factor 3/genetics , Cell Line, Tumor , Sirtuin 1/genetics , Glioma/genetics , Glioma/metabolism , RNA, Small Interfering
4.
Mol Cell Biochem ; 2023 Dec 22.
Article in English | MEDLINE | ID: mdl-38129625

ABSTRACT

Polypyrimidine tract-binding protein 1 (PTBP1) is a member of the heterogeneous nuclear ribonucleoprotein (hnRNP) family, which plays a key role in alternative splicing of precursor mRNA and RNA metabolism. PTBP1 is universally expressed in various tissues and binds to multiple downstream transcripts to interfere with physiological and pathological processes such as the tumor growth, body metabolism, cardiovascular homeostasis, and central nervous system damage, showing great prospects in many fields. The function of PTBP1 involves the regulation and interaction of various upstream molecules, including circular RNAs (circRNAs), microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). These regulatory systems are inseparable from the development and treatment of diseases. Here, we review the latest knowledge regarding the structure and molecular functions of PTBP1 and summarize its functions and mechanisms of PTBP1 in various diseases, including controversial studies. Furthermore, we recommend future studies on PTBP1 and discuss the prospects of targeting PTBP1 in new clinical therapeutic approaches.

7.
Aging (Albany NY) ; 15(18): 9544-9560, 2023 Sep 18.
Article in English | MEDLINE | ID: mdl-37724890

ABSTRACT

Inflammation of chondrocytes plays a critical role in the occurrence and development of osteoarthritis (OA). Recent evidence indicated exosomes derived from mesenchymal stem cells (MSCs-Exos) exhibit excellent anti-inflammatory ability in many troublesome inflammatory diseases including OA. In the present study, we aimed to explore the role of human umbilical cord-derived MSCs-Exos (hUC-MSCs-Exos) in treating the inflammation of chondrocytes and its related mechanisms. Ultracentrifugation was applied to isolate hUC-MSCs-Exos from the culture supernatant of hUC-MSCs. Two OA-like in vitro inflammation models of human articular chondrocytes induced with interleukin 1ß (IL-1ß) and co-incubation with macrophage utilizing transwell cell culture inserts were both used to evaluate the anti-inflammatory effects of hUC-MSCs-Exos. The mRNA sequencing of chondrocytes after treatment and microRNA (miRNA) sequencing of hUC-MSCs-Exos were detected and analyzed for possible mechanism analysis. The results of the study confirmed that hUC-MSCs-Exos had a reversed effect of IL-1ß on chondrocytes in the expression of collagen type II alpha 1 (COL2A1) and matrix metalloproteinase 13 (MMP13). The addition of hUC-MSCs-Exos to M1 macrophages in the upper chamber showed down-regulation of IL-1ß and tumor necrosis factor α (TNF-α), up-regulation of IL-10 and arginase1 (ARG1), and reversed the gene and protein expression of COL2A1 and MMP13 of the chondrocytes seeded in the lower chamber. The results of this study confirmed the anti-inflammatory effects of hUC-MSCs-Exos in the human articular chondrocytes inflammation model. hUC-MSCs-Exos may be used as a potential cell-free treatment strategy for chondrocyte inflammation in OA.

8.
Acta Pharmacol Sin ; 44(10): 2125-2138, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37277492

ABSTRACT

Parthanatos is a type of programmed cell death dependent on hyper-activation of poly (ADP-ribose) polymerase 1 (PARP-1). SIRT1 is a highly conserved nuclear deacetylase and often acts as an inhibitor of parthanatos by deacetylation of PARP1. Our previous study showed that deoxypodophyllotoxin (DPT), a natural compound isolated from the traditional herb Anthriscus sylvestris, triggered glioma cell death via parthanatos. In this study, we investigated the role of SIRT1 in DPT-induced human glioma cell parthanatos. We showed that DPT (450 nmol/L) activated both PARP1 and SIRT1, and induced parthanatos in U87 and U251 glioma cells. Activation of SIRT1 with SRT2183 (10 µmol/L) enhanced, while inhibition of SIRT1 with EX527 (200 µmol/L) or knockdown of SIRT1 attenuated DPT-induced PARP1 activation and glioma cell death. We demonstrated that DPT (450 nmol/L) significantly decreased intracellular NAD+ levels in U87 and U251 cells. Further decrease of NAD+ levels with FK866 (100 µmol/L) aggravated, but supplement of NAD+ (0.5, 2 mmol/L) attenuated DPT-induced PARP1 activation. We found that NAD+ depletion enhanced PARP1 activation via two ways: one was aggravating ROS-dependent DNA DSBs by upregulation of NADPH oxidase 2 (NOX2); the other was reinforcing PARP1 acetylation via increase of N-acetyltransferase 10 (NAT10) expression. We found that SIRT1 activity was improved when being phosphorylated by JNK at Ser27, the activated SIRT1 in reverse aggravated JNK activation via upregulating ROS-related ASK1 signaling, thus forming a positive feedback between JNK and SIRT1. Taken together, SIRT1 activated by JNK contributed to DPT-induced human glioma cell parthanatos via initiation of NAD+ depletion-dependent upregulation of NOX2 and NAT10.


Subject(s)
Glioma , Parthanatos , Sirtuin 1 , Humans , Glioma/drug therapy , N-Terminal Acetyltransferases/genetics , N-Terminal Acetyltransferases/metabolism , NAD/metabolism , NADPH Oxidase 2/metabolism , Parthanatos/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Up-Regulation
9.
Acta Pharmacol Sin ; 44(9): 1906-1919, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37186123

ABSTRACT

Parthanatos is a type of programmed cell death initiated by over-activated poly (ADP-ribose) polymerase 1 (PARP1). Nuclear translocation of apoptosis inducing factor (AIF) is a prominent feature of parthanatos. But it remains unclear how activated nuclear PARP1 induces mitochondrial AIF translocation into nuclei. Evidence has shown that deoxypodophyllotoxin (DPT) induces parthanatos in glioma cells via induction of excessive ROS. In this study we explored the downstream signal of activated PARP1 to induce nuclear translocation of AIF in DPT-triggered glioma cell parthanatos. We showed that treatment with DPT (450 nM) induced PARP1 over-activation and Tax1 binding protein 1 (TAX1BP1) distribution to mitochondria in human U87, U251 and U118 glioma cells. PARP1 activation promoted TAX1BP1 distribution to mitochondria by depleting nicotinamide adenine dinucleotide (NAD+). Knockdown of TAX1BP1 with siRNA not only inhibited TAX1BP1 accumulation in mitochondria, but also alleviated nuclear translocation of AIF and glioma cell death. We demonstrated that TAX1BP1 enhanced the activity of respiratory chain complex I not only by upregulating the expression of ND1, ND2, NDUFS2 and NDUFS4, but also promoting their assemblies into complex I. The activated respiratory complex I generated more superoxide to cause mitochondrial depolarization and nuclear translocation of AIF, while the increased mitochondrial superoxide reversely reinforced PARP1 activation by inducing ROS-dependent DNA double strand breaks. In mice bearing human U87 tumor xenograft, administration of DPT (10 mg· kg-1 ·d-1, i.p., for 8 days) markedly inhibited the tumor growth accompanied by NAD+ depletion, TAX1BP1 distribution to mitochondria, AIF distribution to nuclei as well as DNA DSBs and PARP1 activation in tumor tissues. Taken together, these data suggest that TAX1BP1 acts as a downstream signal of activated PARP1 to trigger nuclear translocation of AIF by activation of mitochondrial respiratory chain complex I.


Subject(s)
Glioma , Parthanatos , Humans , Mice , Animals , Apoptosis Inducing Factor/genetics , Superoxides/metabolism , Reactive Oxygen Species/metabolism , NAD/metabolism , Electron Transport , Electron Transport Complex I , Glioma/metabolism , Neoplasm Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
10.
J Alzheimers Dis ; 91(1): 33-42, 2023.
Article in English | MEDLINE | ID: mdl-36373321

ABSTRACT

Millions of patients worldwide are affected by Alzheimer's disease (AD), and the number of patients with AD is increasing. However, current treatment can only improve symptoms but cannot cure the disease. Astrocytes, glial cells in the central nervous system, play important roles in support, nutrition, protection, and information transmission in the nervous system. Pathological changes in astrocytes are closely associated with the development and progression of AD. As carriers for material and information exchange between astrocytes and other neural cells, astrocyte-derived exosomes (ADEs) have been widely studied in recent years, and ADE secretion has been shown to be increased in patients with AD and animal models of AD. ADEs contain a variety of substances, including nucleic acids, proteins, and lipids. The contents of ADEs can effectively control oxidative stress and detoxification during the early development of AD, thereby playing positive and negative roles in the occurrence and development of AD. In this review, we elaborate on the functions of ADEs and their components in AD and discuss their applications in AD research and clinical practice.


Subject(s)
Alzheimer Disease , Exosomes , Animals , Alzheimer Disease/pathology , Astrocytes/metabolism , Amyloid beta-Peptides/metabolism , Exosomes/metabolism , Central Nervous System/metabolism
11.
Cell Mol Neurobiol ; 43(5): 2035-2052, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36112332

ABSTRACT

Substance-P (SP) is an 11 amino acid neuropeptide that is known to stimulate the peripheral mobilization of bone marrow mesenchymal stem cells and M2 polarization in monocytes/macrophages in a variety of acute and chronic tissue injuries. To examine the role of SP in protection and recovery from acute ischemic brain injury, experimental ischemic stroke was induced by transient middle cerebral artery occlusion (tMCAo) in rats for 1 h with subsequent reperfusion. Two injections of SP, immediately and one day post-tMCAo, resulted in approximately threefold lower mortality and 40% less infarct volume than those of saline-treated rats at seven days post-tMCAo. At 4.5 h, SP markedly increased CD11b/c+CD163+/CD 206+ cells in the blood, which were concomitantly decreased in the bone marrow, suggesting that SP preferentially mobilized M2-polarized monocytes. After two days, SP increased the expression of neuroprotective and anti-inflammatory genes in the ischemic brain and induced neuronal survival in the brain penumbra. Additionally, SP markedly increased CD68+CD163+ and CD68+CD206+ M2 microglia/macrophages in the ischemic brain during seven days post-tMCAo. Furthermore, SP preserved the blood‒brain barrier in the ischemic brain, which was confirmed by the abundant levels of SMI71+ brain endothelial cells that colocalized with α-SMA+ pericytes. The beneficial effects of SP on functional recovery and tissue preservation were maintained for six weeks. Collectively, SP treatment in the early phase of ischemic stroke markedly suppressed the destructive inflammatory response and improved the microenvironment for tissue protection and repair.


Subject(s)
Brain Injuries , Ischemic Stroke , Rats , Animals , Microglia , Neuroprotection , Substance P/pharmacology , Endothelial Cells , Macrophages , Brain , Infarction, Middle Cerebral Artery/complications
12.
J Neuroinflammation ; 19(1): 314, 2022 Dec 26.
Article in English | MEDLINE | ID: mdl-36572898

ABSTRACT

BACKGROUND: Brain lactate concentrations are enhanced in response to cerebral ischemia and promote the formation of reactive astrocytes, which are major components of the neuroinflammatory response and functional recovery, following cerebral ischemia. NDRG2 is upregulated during reactive astrocyte formation. However, its regulation and function are unclear. We studied the relationship between lactate and NDRG2 in astrocytes under conditions of ischemia or oxygen-glucose deprivation (OGD). METHODS: We examined astrocytic NDRG2 expression after middle cerebral artery occlusion (MCAO) using western blot and immunofluorescence staining. Under hypoxia conditions, we added exogenous L-lactate sodium (lactate) to cultured primary astrocytes to explore the effects of lactate on the ubiquitination modification of NDRG2. We profiled the transcriptomic features of NDRG2 silencing in astrocytes after 8 h of OGD conditions as well as exogenous lactate treatment by performing RNA-seq. Finally, we evaluated the molecular mechanisms of NDRG2 in regulating TNFα under OGD conditions using western blot and immunohistochemistry. RESULTS: Reactive astrocytes strongly expressed NDRG2 in a rat model of MCAO. We also showed that lactate stabilizes astrocytic NDRG2 by inhibiting its ubiquitination. NDRG2 inhibition in astrocytes increased inflammation and upregulated immune-associated genes and signaling pathways. NDRG2 knockdown induced TNFα expression and secretion via c-Jun phosphorylation. CONCLUSIONS: We revealed that under OGD conditions, lactate plays an important anti-inflammatory role and inhibits TNFα expression by stabilizing NDRG2, which is beneficial for neurological functional recovery. NDRG2 may be a new therapeutic target for cerebral ischemia.


Subject(s)
Astrocytes , Brain Ischemia , Animals , Rats , Astrocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Lactic Acid , Glucose/metabolism , Brain Ischemia/metabolism , Oxygen/metabolism , Infarction, Middle Cerebral Artery/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Ubiquitination , Nerve Tissue Proteins/metabolism
13.
Ann Transl Med ; 10(18): 976, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36267713

ABSTRACT

Background: Osteoarthritis (OA) is one of the most common joint diseases and a major global public health concern. Mesenchymal stem cells (MSCs) have been widely used for the treatment of OA owing to their paracrine secretion of trophic factors, a phenomenon in which exosomes may play a major role. Here, we investigate the potential of exosomes from human umbilical cord-derived MSCs (hUC-MSCs-Exos) in alleviating OA. Methods: The hUC-MSCs-Exos were harvested from hUC-MSC-conditioned medium using ultracentrifugation. Rats with surgically-induced OA were intra-articularly injected with hUC-MSCs-Exos. The effect of hUC-MSCs-Exos in repairing osteoarticular cartilage was assessed using hematoxylin and eosin (HE) staining, safranin-O and fast green staining and immunohistochemistry. The in vitro experiments were further carried out to verify the therapeutic effect. The effects of hUC-MSCs-Exos on the proliferation and migration of human chondrocytes were evaluated using the cell counting kit-8, EdU-555 cell proliferation kit, and transwell assays. Annexin V-FITC/PI staining were used to evaluate the effect of exosomes on chondrocyte apoptosis. An in vitro model of human articular chondrocytes treated with interleukin 1 beta (IL-1ß) was used to evaluate the effects of exosomes, analyses involved using quantitative real-time polymerase chain reaction (qRT-PCR), immunofluorescence, and western blotting. The role of hUC-MSCs-Exos in macrophage polarization was examined in the monocyte cell line, Tohoku Hospital Pediatrics-1 (THP-1) by qRT-PCR and immunofluorescence. Results: The results showed that hUC-MSCs-Exos prevented severe damage to the knee articular cartilage in the rat OA model. We confirmed the high efficacy of hUC-MSCs-Exos in promoting chondrocyte proliferation and migration and inhibiting chondrocyte apoptosis. Additionally, hUC-MSCs-Exos could reverse IL-1ß-induced injury of chondrocytes and regulate the polarization of macrophages in vitro. Conclusions: There is potential for hUC-MSCs-Exos to be used as a treatment strategy for OA.

14.
Cell Mol Neurobiol ; 42(7): 2031-2053, 2022 Oct.
Article in English | MEDLINE | ID: mdl-33886036

ABSTRACT

Central nervous system injuries and diseases, such as ischemic stroke, spinal cord injury, neurodegenerative diseases, glioblastoma, multiple sclerosis, and the resulting neuroinflammation often lead to death or long-term disability. MicroRNAs are small, non-coding, single-stranded RNAs that regulate posttranscriptional gene expression in both physiological and pathological cellular processes, including central nervous system injuries and disorders. Studies on miR-124, one of the most abundant microRNAs in the central nervous system, have shown that its dysregulation is related to the occurrence and development of pathology within the central nervous system. Herein, we review the molecular regulatory functions, underlying mechanisms, and effective delivery methods of miR-124 in the central nervous system, where it is involved in pathological conditions. The review also provides novel insights into the therapeutic target potential of miR-124 in the treatment of human central nervous system injuries or diseases.


Subject(s)
Central Nervous System Diseases , MicroRNAs , Neurodegenerative Diseases , Spinal Cord Injuries , Trauma, Nervous System , Central Nervous System , Humans
15.
Acta Pharmacol Sin ; 42(10): 1690-1702, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34112960

ABSTRACT

Ferroptotic cell death is characterized by iron-dependent lipid peroxidation that is initiated by ferrous iron and H2O2 via Fenton reaction, in which the role of activating transcription factor 3 (ATF3) remains elusive. Brucine is a weak alkaline indole alkaloid extracted from the seeds of Strychnos nux-vomica, which has shown potent antitumor activity against various tumors, including glioma. In this study, we showed that brucine inhibited glioma cell growth in vitro and in vivo, which was paralleled by nuclear translocation of ATF3, lipid peroxidation, and increases of iron and H2O2. Furthermore, brucine-induced lipid peroxidation was inhibited or exacerbated when intracellular iron was chelated by deferoxamine (500 µM) or improved by ferric ammonium citrate (500 µM). Suppression of lipid peroxidation with lipophilic antioxidants ferrostatin-1 (50 µM) or liproxstatin-1 (30 µM) rescued brucine-induced glioma cell death. Moreover, knockdown of ATF3 prevented brucine-induced accumulation of iron and H2O2 and glioma cell death. We revealed that brucine induced ATF3 upregulation and translocation into nuclei via activation of ER stress. ATF3 promoted brucine-induced H2O2 accumulation via upregulating NOX4 and SOD1 to generate H2O2 on one hand, and downregulating catalase and xCT to prevent H2O2 degradation on the other hand. H2O2 then contributed to brucine-triggered iron increase and transferrin receptor upregulation, as well as lipid peroxidation. This was further verified by treating glioma cells with exogenous H2O2 alone. Moreover, H2O2 reversely exacerbated brucine-induced ER stress. Taken together, ATF3 contributes to brucine-induced glioma cell ferroptosis via increasing H2O2 and iron.


Subject(s)
Activating Transcription Factor 3/metabolism , Antineoplastic Agents/therapeutic use , Ferroptosis/drug effects , Hydrogen Peroxide/metabolism , Iron/metabolism , Strychnine/analogs & derivatives , Amino Acid Transport System y+/metabolism , Animals , Antineoplastic Agents/pharmacology , Catalase/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , NADPH Oxidase 4/metabolism , Neoplasms/drug therapy , Strychnine/pharmacology , Strychnine/therapeutic use , Superoxide Dismutase-1/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
16.
Aging (Albany NY) ; 13(8): 11433-11454, 2021 04 04.
Article in English | MEDLINE | ID: mdl-33839696

ABSTRACT

Autologous chondrocyte implantation (ACI) is an effective method for treating chronic articular cartilage injury and degeneration; however, it requires large numbers of hyaline chondrocytes, and human hyaline chondrocytes often undergo dedifferentiation in vitro. Moreover, although long non-coding RNAs (lncRNAs) regulate gene expression in many pathological and physiological processes, their role in human hyaline chondrocyte dedifferentiation remains unclear. Here, we examined lncRNA and mRNA expression profiles in human hyaline chondrocyte dedifferentiation using microarray analysis. Among the many lncRNAs and mRNAs that showed differential expression, lncRNA AP001505.9 (ENST00000569966) was significantly downregulated in chondrocytes after dedifferentiation. We next performed gene ontology, pathway, and CNC (coding-non-coding gene co-expression) analyses to investigate potential regulatory mechanisms for AP001505.9. Pellet cultures were then used to redifferentiate dedifferentiated chondrocytes, and AP001505.9 expression was upregulated after redifferentiation. Finally, both in vitro and in vivo experiments demonstrated that AP001505.9 overexpression inhibited dedifferentiation of chondrocytes. This study characterizes lncRNA expression profiles in human hyaline chondrocyte dedifferentiation, thereby identifying new potential mechanisms of chondrocyte dedifferentiation worthy of further investigation.


Subject(s)
Cartilage, Articular/growth & development , Cell Dedifferentiation/genetics , Chondrocytes/physiology , Chondrogenesis/genetics , RNA, Long Noncoding/metabolism , Adult , Aged , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cells, Cultured , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Primary Cell Culture
17.
Acta Pharmacol Sin ; 42(8): 1324-1337, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33879840

ABSTRACT

FOXO3a (forkhead box transcription factor 3a) is involved in regulating multiple biological processes in cancer cells. BNIP3 (Bcl-2/adenovirus E1B 19-kDa-interacting protein 3) is a receptor accounting for priming damaged mitochondria for autophagic removal. In this study we investigated the role of FOXO3a in regulating the sensitivity of glioma cells to temozolomide (TMZ) and its relationship with BNIP3-mediated mitophagy. We showed that TMZ dosage-dependently inhibited the viability of human U87, U251, T98G, LN18 and rat C6 glioma cells with IC50 values of 135.75, 128.26, 142.65, 155.73 and 111.60 µM, respectively. In U87 and U251 cells, TMZ (200 µM) induced DNA double strand breaks (DSBs) and nuclear translocation of apoptosis inducing factor (AIF), which was accompanied by BNIP3-mediated mitophagy and FOXO3a accumulation in nucleus. TMZ treatment induced intracellular ROS accumulation in U87 and U251 cells via enhancing mitochondrial superoxide, which not only contributed to DNA DSBs and exacerbated mitochondrial dysfunction, but also upregulated FOXO3a expression. Knockdown of FOXO3a aggravated TMZ-induced DNA DSBs and mitochondrial damage, as well as glioma cell death. TMZ treatment not only upregulated BNIP3 and activated autophagy, but also triggered mitophagy by prompting BNIP3 translocation to mitochondria and reinforcing BNIP3 interaction with LC3BII. Inhibition of mitophagy by knocking down BNIP3 with SiRNA or blocking autophagy with 3MA or bafilomycin A1 exacerbated mitochondrial superoxide and intracellular ROS accumulation. Moreover, FOXO3a knockdown inhibited TMZ-induced BNIP3 upregulation and autophagy activation. In addition, we showed that treatment with TMZ (100 mg·kg-1·d-1, ip) for 12 days in C6 cell xenograft mice markedly inhibited tumor growth accompanied by inducing FOXO3a upregulation, oxidative stress and BNIP3-mediated mitophagy in tumor tissues. These results demonstrate that FOXO3a attenuates temozolomide-induced DNA double strand breaks in human glioma cells via promoting BNIP3-mediated mitophagy.


Subject(s)
Antineoplastic Agents/therapeutic use , DNA Breaks, Double-Stranded/drug effects , Forkhead Box Protein O3/metabolism , Glioma/metabolism , Mitophagy/drug effects , Temozolomide/therapeutic use , Animals , Autophagy/drug effects , Cell Line, Tumor , Glioma/drug therapy , Humans , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins/metabolism , Rats , Up-Regulation/drug effects
18.
Mol Med Rep ; 23(5)2021 05.
Article in English | MEDLINE | ID: mdl-33760132

ABSTRACT

Hair loss, including alopecia, is a common and distressing problem for men and women, and as a result, there is considerable interest in developing treatments that can prevent or reverse hair loss. Dermal papillae closely interact with epidermal cells and play a key role during hair follicle induction and hair morphogenesis. As dermal papilla cells (DPCs) lose their hair­inducing ability in monolayer cultures in vitro, it is difficult to obtain de novo hair follicle structures following DPC transplantation in vivo. The present study aimed to explore culture conditions to maintain DPC characteristics using conditioned media (CM) from the supernatant of cultured HaCaT keratinocyte cells supplemented with other components. Initially, it was observed that during passaging of in vitro monolayer DPC cultures, the Wnt/ß­catenin pathway was repressed, while the TGF­ß/Smad pathway was activated, and that HaCaT cells cultivated in 1% fetal bovine serum had higher levels of expression of Wnt3a and Wnt10b compared with normal keratinocytes. Culturing of high­passage (P7) DPCs in CM from HaCaT cells (HaCaT­CM) actively stimulated cell proliferation and maintained Sox2 and Versican expression levels. Supplementation of HaCaT­CM with SB431542 (SB, a TGF­ß receptor inhibitor), CHIR99021, (CHIR, a GSK3α/ß inhibitor and activator of Wnt signaling) and platelet­derived growth factor (PDGF)­AA further increased the expression levels of Sox2, Versican and alkaline phosphatase (ALP) in P7 DPCs. Three­dimensional culture of P7 DPCs using hanging drop cultures in HaCaT­CM supplemented with SB, CHIR and PDGF­AA resulted in larger cell aggregates and a further significant upregulation of Sox2, ALP and Versican expression levels. Taken together, these findings demonstrated that HaCaT­CM supplemented with SB, CHIR and PDGF­AA may preserve the hair­inducing ability of high­passage DPCs and may therefore be useful in reconstructing new hair follicles in vivo.


Subject(s)
Alopecia/genetics , Cell Dedifferentiation/drug effects , Dermis/growth & development , Platelet-Derived Growth Factor/genetics , Alopecia/drug therapy , Alopecia/pathology , Benzamides/pharmacology , Cell Culture Techniques , Culture Media, Conditioned , Dermis/cytology , Dioxoles/pharmacology , Hair/growth & development , Hair/metabolism , Hair Follicle/cytology , Hair Follicle/growth & development , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , Wnt Signaling Pathway/drug effects
19.
Mol Neurobiol ; 58(5): 2447-2464, 2021 May.
Article in English | MEDLINE | ID: mdl-33725319

ABSTRACT

Irreversible neuron loss caused by central nervous system injuries usually leads to persistent neurological dysfunction. Reactive astrocytes, because of their high proliferative capacity, proximity to neuronal lineage, and significant involvement in glial scarring, are ideal starting cells for neuronal regeneration. Having previously identified several small molecules as important regulators of astrocyte-to-neuron reprogramming, we established herein that miR-124, ruxolitinib, SB203580, and forskolin could co-regulate rat cortical reactive astrocyte-to-neuron conversion. The induced cells had reduced astroglial properties, displayed typical neuronal morphologies, and expressed neuronal markers, reflecting 25.9% of cholinergic neurons and 22.3% of glutamatergic neurons. Gene analysis revealed that induced neuron gene expression patterns were more similar to that of primary neurons than of initial reactive astrocytes. On the molecular level, miR-124-driven neuronal differentiation of reactive astrocytes was via targeting of the SOX9-NFIA-HES1 axis to inhibit HES1 expression. In conclusion, we present a novel approach to inducing endogenous rat cortical reactive astrocytes into neurons through co-regulation involving miR-124 and three small molecules. Thus, our research has potential implications for inhibiting glial scar formation and promoting neuronal regeneration after central nervous system injury or disease.


Subject(s)
Astrocytes/cytology , Cerebral Cortex/cytology , MicroRNAs/metabolism , Neurogenesis/physiology , Neurons/cytology , Animals , Astrocytes/metabolism , Cerebral Cortex/metabolism , Glial Fibrillary Acidic Protein/metabolism , MicroRNAs/genetics , Neurons/metabolism , Rats , Rats, Wistar
20.
Mol Cell Biochem ; 476(5): 2111-2124, 2021 May.
Article in English | MEDLINE | ID: mdl-33528706

ABSTRACT

MicroRNAs (miRNA), endogenous non-coding RNAs approximately 22 nucleotides long, regulate gene expression by mediating translational inhibition or mRNA degradation. Exosomes are a tool for intercellular transmission of information in which miRNA exchange plays an important role. Under pathophysiological conditions in the central nervous system (CNS), cellular transmission of exosomal miRNAs can regulate signaling pathways. Exosomal miRNAs are involved in the occurrence and development of diverse CNS diseases, such as traumatic brain injury, spinal cord injury, stroke, neurodegenerative diseases, epilepsy, and glioma. The use of exosomes as transport vehicles for certain miRNAs provides a novel therapeutic strategy for CNS diseases. Furthermore, the exosomes in body fluids change with the occurrence of diseases, indicating that subtle changes in physiological and pathological processes in vivo could be recognized by analyzing exosomes. Exosomal analysis is expected to act as a novel tool for diagnosis and prediction of neurological diseases. In this review, we present the current understanding of the implications of miRNAs in CNS diseases and summarize the role and mechanism of action of exosomal miRNA in nervous system disease models.


Subject(s)
Cell Communication , Central Nervous System Diseases/metabolism , Circulating MicroRNA/metabolism , Exosomes/metabolism , Signal Transduction , Animals , Central Nervous System Diseases/pathology , Exosomes/pathology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...