Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Biol Rep ; 51(1): 654, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735002

ABSTRACT

BACKGROUND: Cervical cancer is a common gynecologic malignant tumor, but the critical factors affecting cervical cancer progression are still not well demonstrated. Mesencephalic astrocyte-derived neurotrophic factor (MANF) has been widely recognized as an anti-inflammatory factor to regulate macrophage polarization. In this study, the effect and mechanism of MANF on cervical cancer were preliminarily explored. METHODS AND RESULTS: Kaplan-Meier curve was used to show the overall survival time of the involved cervical cancer patients with high and low MANF expression in cervical cancer tissues. MANF was highly expressed in peritumoral tissues of cervical carcinoma by using immunohistochemistry and western blot. MANF mRNA level was detected by using qRT-PCR. Dual-labeled immunofluorescence showed MANF was mainly expressed in macrophages of cervical peritumoral tissues. Moreover, MANF-silenced macrophages promoted HeLa and SiHa cells survival, migration, invasion and EMT via NF-κB signaling activation. The results of tumor formation in nude mice indicated MANF-silenced macrophages promoted cervical tumor formation in vivo. CONCLUSION: Our study reveals an inhibitory role of MANF in cervical cancer progression, indicating MANF as a new and valuable therapeutic target for cervical cancer treatment.


Subject(s)
Macrophages , Nerve Growth Factors , Uterine Cervical Neoplasms , Animals , Female , Humans , Mice , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , HeLa Cells , Macrophages/metabolism , Mice, Nude , Nerve Growth Factors/metabolism , Nerve Growth Factors/genetics , NF-kappa B/metabolism , Phenotype , Signal Transduction , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism
2.
BMC Psychiatry ; 24(1): 47, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38216957

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is a debilitating health condition that has significant morbidity and mortality rates. Depression can be caused due to social, biological, environmental, psychological, and genetic factors. A few biological processes have been proposed as the pathophysiological pathways of depression. Neurotrophic factors and inflammatory cytokines have been linked to depression. Thus, we aimed to investigate the serum interleukin-33 (IL-33) and mesencephalic astrocyte-derived neurotrophic factor (MANF) in MDD patients and corresponding healthy controls (HCs). METHOD: This study involved the inclusion of 129 MDD patients and 125 HCs matched by sex and age. A psychiatrist evaluated the study participants following DSM-5 criteria. The severity of the illness was assessed utilizing the Hamilton Depression Rating Scale (Ham-D). The serum concentrations of IL-33 and MANF were measured using enzyme-linked immunosorbent assay (ELISA) kits. RESULTS: The mean serum levels of IL-33 were decreased (159.12 ± 6.07 pg/ml vs. 180.60 ± 8.64 pg/ml, p = 0.042), and the MANF levels were increased (5.40 ± 0.19 ng/ml vs. 4.46 ± 0.21 ng/ml, p = 0.001) in MDD patients when compared to HCs. CONCLUSIONS: The current study proposes that lower IL-33 and higher MANF serum levels are associated with MDD progression and depression severity. These biomarkers could be used as risk assessment tools for MDD. We recommend more investigation, including a significant population, to determine the precise function of IL-33 and MANF in depression.


Subject(s)
Depressive Disorder, Major , Humans , Astrocytes/metabolism , Cross-Sectional Studies , Interleukin-33 , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism
3.
Cell Mol Neurobiol ; 43(8): 3897-3913, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37751132

ABSTRACT

This study employs bibliometric analysis through CiteSpace to comprehensively evaluate the status and trends of MANF (mesencephalic astrocyte-derived neurotrophic factor) research spanning 25 years (1997-2022). It aims to fill the gap in objective and comprehensive reviews of MANF research. MANF-related studies were extracted from the Web of Science database. MANF publications were quantitatively and qualitatively analyzed for various factors by CiteSpace, including publication volume, journals, countries/regions, institutions, and authors. Keywords and references were visually analyzed to unveil research evolution and hotspot. Analysis of 353 MANF-related articles revealed escalating annual publications, indicating growing recognition of MANF's importance. High-impact journals such as the International Journal of Molecular Sciences and Journal of Biological Chemistry underscored MANF's interdisciplinary significance. Collaborative networks highlighted China and the USA's pivotal roles, while influential figures and partnerships drove understanding of MANF's mechanisms. Co-word analysis of MANF-related keywords exposed key evolutionary hotspots, encompassing neurotrophic effects, cytoprotective roles, MANF-related diseases, and the CDNF/MANF family. This progression from basic understanding to clinical potential showcased MANF's versatility from cellular protection to therapy. Bibliometric analysis reveals MANF's diverse research trends and pathways, from basics to clinical applications, driving medical progress. This comprehensive assessment enriches understanding and empowers researchers for dynamic evolution, advancing innovation, and benefiting patients. Bibliometric analysis of MANF research. The graphical abstract depicts the bibliometric analysis of MANF research, highlighting its aims, methods, and key results.


Subject(s)
Nerve Growth Factors , Humans , Nerve Growth Factors/physiology , Bibliometrics , Biomedical Research/trends
4.
FASEB J ; 36(6): e22349, 2022 06.
Article in English | MEDLINE | ID: mdl-35567505

ABSTRACT

Excessive lipid accumulation, inflammation, and fibrosis in the liver are the major characteristics of non-alcoholic steatohepatitis (NASH). Mesencephalic astrocyte-derived neurotrophic factor (MANF) plays an important role in metabolic homeostasis, raising the possibility that it is involved in NASH. Here, we reduced and increased MANF levels in mice in order to explore its influence on hepatic triglyceride homeostasis, inflammation, and fibrosis during NASH progression. The MANF expression was decreased in Western diet-induced NASH mice. In vivo, liver-specific MANF knockout exacerbated hepatic lipid accumulation, inflammation, and fibrosis of mice induced by Western diet, while liver-specific MANF overexpression mitigated these NASH pathogenic features. In vitro, knocking down MANF in primary hepatocyte cultures aggravated hepatic steatosis and inflammation, which MANF overexpression markedly attenuated. Studies in vitro and in vivo suggested that MANF regulated hepatic lipid synthesis by modulating SREBP1 expression. Inhibiting SREBP1 in primary hepatocytes blocked lipid accumulation after MANF knockdown. MANF overexpression reversed LXRs agonist GW3965 induced SREBP1 and LIPIN1 expression. MANF decreased the expression of pro-inflammatory cytokines by inhibiting NF-κB phosphorylation. These results suggest that MANF can protect against NASH by regulating SREBP1 expression and NF-κB signaling.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Astrocytes/metabolism , Diet, Western , Fibrosis , Inflammation/metabolism , Lipids , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism
5.
BMC Neurol ; 23(1): 213, 2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37268902

ABSTRACT

OBJECTIVE: Mesencephalic astrocyte-derived neurotrophic factor (MANF) expressions are dramatically up-regulated in injured brain tissues, thereby conferring neurological protective effects. We intended to determine significance of serum MANF as a prognostic biomarker of intracerebral hemorrhage (ICH). METHODS: In this prospective, observational study done from February 2018 to July 2021, 124 patients with new-onset primary supratentorial ICH were consecutively enrolled. Also, a group of 124 healthy individuals constituted controls. Their serum MANF levels were detected using the Enzyme-Linked Immunosorbent Assay. National Institutes of Health Stroke Scale (NIHSS) and hematoma volume were designated as the two severity indicators. Early neurologic deterioration (END) was referred to as an increase of 4 or greater points in NIHSS scores or death at post-stroke 24 h. Post-stroke 90-day modified Rankin scale (mRS) scores of 3-6 was considered as a poor prognosis. Serum MANF levels were analyzed using multivariate analysis with respect to its association with stroke severity and prognosis. RESULTS: Patients, in comparison to controls, displayed markedly elevated serum MANF levels (median, 24.7 versus 2.7 ng/ml; P < 0.001), and serum MANF levels were independently correlated with NIHSS scores (beta, 3.912; 95% confidence interval (CI), 1.623-6.200; VIF = 2.394; t = 3.385; P = 0.002), hematoma volumes (beta, 1.688; 95% CI, 0.764-2.612; VIF = 2.661; t = 3.617; P = 0.001) and mRS scores (beta, 0.018; 95% CI, 0.013-0.023; VIF = 1.984; t = 2.047; P = 0.043). Serum MANF levels significantly predicted END and poor 90-day prognosis with areas under receiver operating characteristic curve at 0.752 and 0.787 respectively. END and prognostic predictive abilities were similar between serum MANF levels and NIHSS scores plus hematoma volumes (all P > 0.05). Combination of serum MANF levels with NIHSS scores and hematoma volumes had significantly higher prognostic capability than each of them (both P < 0.05). Serum MANF levels above 52.5 ng/ml and 62.0 ng/ml distinguished development of END and poor prognosis respectively with median-high sensitivity and specificity values. Using multivariate analysis, serum MANF levels > 52.5 ng/ml predicted END with odds ratio (OR) value of 2.713 (95% CI, 1.004-7.330; P = 0.042) and > 62.0 ng/ml predicted a poor prognosis with OR value of 3.848 (95% CI, 1.193-12.417; P = 0.024). Using restricted cubic spline, there was a linear correlation between serum MANF levels and poor prognosis or END risk (both P > 0.05). Nomograms were well established to predict END and a poor 90-day prognosis. Under calibration curve, such combination models were comparatively stable (using Hosmer & Lemeshow test, both P > 0.05). CONCLUSION: Increased serum MANF levels after ICH, in independent correlation with disease severity, independently distinguished risks of END and 90-day poor prognosis. Therefore, serum MANF may be a potential prognostic biomarker of ICH.


Subject(s)
Astrocytes , Cerebral Hemorrhage , Nerve Growth Factors , Stroke , Humans , Biomarkers , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/diagnosis , Hematoma , Prognosis , Prospective Studies , Nerve Growth Factors/blood
6.
Acta Pharmacol Sin ; 44(1): 157-168, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35655095

ABSTRACT

Hepatic steatosis plays a detrimental role in the onset and progression of alcohol-associated liver disease (ALD). Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an evolutionarily conserved protein related to the unfolded protein response. Recent studies have demonstrated that MANF plays an important role in liver diseases. In this study, we investigated the role of MANF in ethanol-induced steatosis and the underlying mechanisms. We showed that the hepatic MANF expression was markedly upregulated in mouse model of ALD by chronic-plus-single-binge ethanol feeding. Moreover, after chronic-plus-binge ethanol feeding, hepatocyte-specific MANF knockout (HKO) mice displayed more severe hepatic steatosis and liver injury than wild-type (WT) control mice. Immunoprecipitation-coupled MS proteomic analysis revealed that arginosuccinate synthase 1 (ASS1), a rate-limiting enzyme in the urea cycle, resided in the same immunoprecipitated complex with MANF. Hepatocyte-specific MANF knockout led to decreased ASS1 activity, whereas overexpression of MANF contributed to enhanced ASS1 activity in vitro. In addition, HKO mice displayed unique urea cycle metabolite patterns in the liver with elevated ammonia accumulation after ethanol feeding. ASS1 is known to activate AMPK by generating an intracellular pool of AMP from the urea cycle. We also found that MANF supplementation significantly ameliorated ethanol-induced steatosis in vivo and in vitro by activating the AMPK signaling pathway, which was partly ASS1 dependent. This study demonstrates a new mechanism in which MANF acts as a key molecule in maintaining hepatic lipid homeostasis by enhancing ASS1 activity and uncovers an interesting link between lipid metabolism and the hepatic urea cycle under excessive alcohol exposure.


Subject(s)
Fatty Liver , Liver Diseases, Alcoholic , Animals , Mice , AMP-Activated Protein Kinases/metabolism , Astrocytes/metabolism , Ethanol/toxicity , Fatty Liver/chemically induced , Hepatocytes/metabolism , Liver/metabolism , Mice, Knockout , Nerve Growth Factors/metabolism , Proteomics , Urea/metabolism
7.
J Biol Chem ; 296: 100295, 2021.
Article in English | MEDLINE | ID: mdl-33460650

ABSTRACT

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER)-stress-regulated protein exhibiting cytoprotective properties through a poorly understood mechanism in various in vitro and in vivo models of neuronal and non-neuronal damage. Although initially characterized as a secreted neurotrophic factor for midbrain dopamine neurons, MANF has recently gained more interest for its intracellular role in regulating the ER homeostasis, including serving as a cofactor of the chaperone glucose-regulated protein 78 (GRP78). We aimed for a better understanding of the neuroprotective mechanisms of MANF. Here we show for the first time that MANF promotes the survival of ER-stressed neurons in vitro as a general unfolded protein response (UPR) regulator, affecting several UPR pathways simultaneously. Interestingly, MANF does not affect naïve neurons. We hypothesize that MANF regulates UPR signaling toward a mode more compatible with neuronal survival. Screening of MANF interacting proteins from two mammalian cell lines revealed a conserved interactome of 15 proteins including several ER chaperones such as GRP78, GRP170, protein disulfide isomerase family A member 1, and protein disulfide isomerase family A member 6. Further characterization confirmed previously published finding that MANF is a cofactor of GRP78 interacting with its nucleotide binding domain. Using microscale thermophoresis and nuclear magnetic resonance spectroscopy, we discovered that MANF is an ATP binding protein and that ATP blocks the MANF-GRP78 interaction. Interestingly, functional analysis of the antiapoptotic properties of MANF mutants in cultured neurons revealed divergent roles of MANF as a GRP78 cofactor and as an antiapoptotic regulator of UPR. We conclude that the co-factor type interaction with GRP78 is dispensable for the survival-promoting activity of MANF in neurons.


Subject(s)
Dopaminergic Neurons/metabolism , Endoplasmic Reticulum/genetics , Heat-Shock Proteins/genetics , Nerve Growth Factors/genetics , Unfolded Protein Response , Animals , Apoptosis/genetics , Cell Survival , Dopaminergic Neurons/cytology , Embryo, Mammalian , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Mesencephalon/cytology , Mesencephalon/metabolism , Mice , Nerve Growth Factors/metabolism , Primary Cell Culture , Protein Binding , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Interaction Mapping , Signal Transduction
8.
Mol Ther ; 29(9): 2821-2840, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33940158

ABSTRACT

A molecular hallmark in Parkinson's disease (PD) pathogenesis are α-synuclein aggregates. Cerebral dopamine neurotrophic factor (CDNF) is an atypical growth factor that is mostly resident in the endoplasmic reticulum but exerts its effects both intracellularly and extracellularly. One of the beneficial effects of CDNF can be protecting neurons from the toxic effects of α-synuclein. Here, we investigated the effects of CDNF on α-synuclein aggregation in vitro and in vivo. We found that CDNF directly interacts with α-synuclein with a KD = 23 ± 6 nM and reduces its auto-association. Using nuclear magnetic resonance (NMR) spectroscopy, we identified interaction sites on the CDNF protein. Remarkably, CDNF reduces the neuronal internalization of α-synuclein fibrils and induces the formation of insoluble phosphorylated α-synuclein inclusions. Intra-striatal CDNF administration alleviates motor deficits in rodents challenged with α-synuclein fibrils, though it did not reduce the number of phosphorylated α-synuclein inclusions in the substantia nigra. CDNF's beneficial effects on rodent behavior appear not to be related to the number of inclusions formed in the current context, and further study of its effects on the aggregation mechanism in vivo are needed. Nonetheless, the interaction of CDNF with α-synuclein, modifying its aggregation, spreading, and associated behavioral alterations, provides novel insights into the potential of CDNF as a therapeutic strategy in PD and other synucleinopathies.


Subject(s)
Nerve Growth Factors/chemistry , Nerve Growth Factors/metabolism , Parkinson Disease/physiopathology , Substantia Nigra/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Animals , Binding Sites , Cell Line , Disease Models, Animal , Dopamine/metabolism , Humans , Magnetic Resonance Spectroscopy , Male , Mice , Models, Molecular , Parkinson Disease/metabolism , Phosphorylation , Primary Cell Culture , Protein Aggregates , Protein Binding , Protein Conformation , Rats
9.
J Stroke Cerebrovasc Dis ; 31(11): 106752, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36087374

ABSTRACT

OBJECTIVES: Although Mesencephalic astrocyte-derived neurotrophic factor (MANF) shows protection in multiple cells, the role of circulating MANF in patients with acute ischemic stroke (AIS) and transient ischemic attack (TIA) remains unclear. Here, we aimed to explore the value of circulating MANF levels in cerebral ischemic events. MATERIALS AND METHODS: Using a rat cerebral ischemic model, MANF expression in ischemic brains and serum was detected. 50 AIS patients, 56 TIA patients and 48 controls were enrolled, and MANF mRNA, inflammatory cytokines and MANF concentrations in serum and different blood cell types were detected. The National Institutes of Health Stroke Scale (NIHSS) score and Alberta Stroke Program Early CT Score (ASPECTS) were used to evaluate stroke severity. Cerebrovascular recurrence within 90 d was documented during TIA follow-up. RESULTS: MANF expression increased at 2h, peaking at 24h and decreased to baseline at 7d in rat ischemic brains and serum. Serum MANF concentrations increased at 24h and 7d in AIS patients compared to controls and were correlated with NIHSS score, ASPECTS and inflammatory cytokines. MANF protein was present in blood cells, while MANF mRNA levels did not differ between AIS patients and controls. MANF levels revealed a good value to diagnose TIA with area under the curve (AUC) of 0.949 (95% CI: 0.9093-0.9892). MANF levels were lower in TIA patients with recurrence compared to non-recurrence patients. The AUC for MANF to predict a re-event was 0.80 (95% CI: 0.6746-0.9282). CONCLUSIONS: Serum MANF levels correlate with neuroprotection, stroke severity, inflammation, and TIA recurrence.


Subject(s)
Ischemic Attack, Transient , Ischemic Stroke , Stroke , Animals , Rats , Ischemic Stroke/diagnosis , Ischemic Attack, Transient/diagnosis , Ischemic Attack, Transient/metabolism , Astrocytes/metabolism , Nerve Growth Factors/genetics , Stroke/genetics , Cytokines/metabolism , RNA, Messenger/metabolism
10.
J Biol Chem ; 295(22): 7566-7583, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32327487

ABSTRACT

We have previously demonstrated that ischemia/reperfusion (I/R) impairs endoplasmic reticulum (ER)-based protein folding in the heart and thereby activates an unfolded protein response sensor and effector, activated transcription factor 6α (ATF6). ATF6 then induces mesencephalic astrocyte-derived neurotrophic factor (MANF), an ER-resident protein with no known structural homologs and unclear ER function. To determine MANF's function in the heart in vivo, here we developed a cardiomyocyte-specific MANF-knockdown mouse model. MANF knockdown increased cardiac damage after I/R, which was reversed by AAV9-mediated ectopic MANF expression. Mechanistically, MANF knockdown in cultured neonatal rat ventricular myocytes (NRVMs) impaired protein folding in the ER and cardiomyocyte viability during simulated I/R. However, this was not due to MANF-mediated protection from reactive oxygen species generated during reperfusion. Because I/R impairs oxygen-dependent ER protein disulfide formation and such impairment can be caused by reductive stress in the ER, we examined the effects of the reductive ER stressor DTT. MANF knockdown in NRVMs increased cell death from DTT-mediated reductive ER stress, but not from nonreductive ER stresses caused by thapsigargin-mediated ER Ca2+ depletion or tunicamycin-mediated inhibition of ER protein glycosylation. In vitro, recombinant MANF exhibited chaperone activity that depended on its conserved cysteine residues. Moreover, in cells, MANF bound to a model ER protein exhibiting improper disulfide bond formation during reductive ER stress but did not bind to this protein during nonreductive ER stress. We conclude that MANF is an ER chaperone that enhances protein folding and myocyte viability during reductive ER stress.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Molecular Chaperones/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Nerve Growth Factors/metabolism , Animals , Cell Survival , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Glycosylation , HeLa Cells , Humans , Mice , Mice, Knockout , Molecular Chaperones/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Nerve Growth Factors/genetics , Reactive Oxygen Species
11.
Biochem Biophys Res Commun ; 550: 197-203, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33713857

ABSTRACT

Alcoholic fatty liver disease (AFLD) is induced by alcohol consumption and may progress to more severe liver diseases such as alcoholic steatohepatitis, fibrosis and cirrhosis, and even hepatocellular carcinoma. Mesencephalic astrocyte-derived neurotrophic factor (MANF) participates in maintaining lipid homeostasis. However, the role of MANF in the pathogenesis of AFLD remains unclear. We established an AFLD mouse model following the US National Institute on Alcohol Abuse and Alcoholism procedure. Both mRNA and protein levels of MANF were significantly increased in the chronic binge alcohol feeding model. Liver-specific knockout of MANF aggravated hepatic lipid accumulation. Similarly, liver-specific overexpression of MANF alleviated AFLD in mouse livers. MANF affected hepatic lipid metabolism by modulating autophagy. The levels of LC3-II and Atg5-Atg12 were decreased in mouse livers with MANF liver-specific knockout and increased with MANF liver-specific overexpression. Furthermore, MANF changed the phosphorylation of Stat3 and its nuclear localization. MANF may have a protective role in the development of AFLD.


Subject(s)
Autophagy , Fatty Liver, Alcoholic/metabolism , Nerve Growth Factors/metabolism , STAT3 Transcription Factor/metabolism , Animals , Autophagy/drug effects , Binge Drinking , Ethanol/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factors/deficiency , Phosphorylation
12.
Bipolar Disord ; 23(1): 76-83, 2021 02.
Article in English | MEDLINE | ID: mdl-33037686

ABSTRACT

BACKGROUND: Bipolar Disorder (BD) is associated with a decrease in cellular resilience. Despite the half a century old discovery of lithium's efficacy for the treatment of BD, its exact mechanisms remain elusive. Accumulating data suggest that lithium's cytoprotective properties involve the modulation of several UPR proteins, such as GRP78. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum resident protein that regulates proteostasis through directly interacting with GRP78. The purpose of this study was to determine whether lithium increases MANF expression using cellular and rodent models and, if so, to elucidate the cellular mechanisms of action. PROCEDURE: Mouse striatal neuroblasts were treated with PBS, lithium, or lithium + Activator Protein-1 (AP-1) inhibitor for 24-72 hours. Once cells were harvested, mRNA was extracted. In vivo experiments included, intraperitoneal injections of lithium or saline to male Sprague Dawley rats twice daily for 14 consecutive days. Following drug treatment, brain tissue was isolated, and mRNA was extracted from various regions. MANF gene expression was measured using RT-qPCR. RESULTS: In vitro studies showed lithium-treated cells displayed a significant increase in MANF mRNA expression compared to controls. In contrast, cells treated with lithium and AP-1 inhibitor showed no increase in expression. Similarly, in vivo studies revealed that lithium-treated rats compared to controls had a significant increase in MANF expression in the PFC and striatum. CONCLUSION: Taken together, these data suggest that lithium's therapeutic mechanism involves the maintenance of ER homeostasis via increased MANF gene expression mediated by the AP-1 transcription factor.


Subject(s)
Bipolar Disorder , Lithium , Animals , Bipolar Disorder/drug therapy , Endoplasmic Reticulum , Endoplasmic Reticulum Chaperone BiP , Lithium/pharmacology , Male , Mice , Nerve Growth Factors , Rats , Rats, Sprague-Dawley , Transcription Factors
13.
Liver Int ; 41(3): 623-639, 2021 03.
Article in English | MEDLINE | ID: mdl-33064897

ABSTRACT

BACKGROUND: Endoplasmic reticulum (ER) perturbations are novel subcellular effectors involved in the ischaemia-reperfusion injury. As an ER stress-inducible protein, mesencephalic astrocyte-derived neurotrophic factor (MANF) has been proven to be increased during ischaemic brain injury. However, the role of MANF in liver ischaemia reperfusion (I/R) injury has not yet been studied. METHODS: To investigate the role of MANF in the process of liver ischaemia-reperfusion, Hepatocyte-specific MANF knockout (MANFhep-/- ) mice and their wild-type (WT) littermates were used in our research. Mice partial (70%) warm hepatic I/R model was established by vascular occlusion. We detected the serum levels of MANF in both liver transplant patients and WT mice before and after liver I/R injury. Recombinant human MANF (rhMANF) was injected into the tail vein before 1 hour occlusion. AST, ALT and Suzuki score were used to evaluate the extent of I/R injury. OGD/R test was performed on primary hepatocytes to simulate IRI in vitro. RNA sequence and RT-PCR were used to detect the cellular signal pathway activation while MANF knockout. RESULTS: We found that MANF expression and secretion are dramatically up-regulated during hepatic I/R. Hepatocyte-specific MANF knockout aggravates the I/R injury through the over-activated ER stress. The systemic administration of rhMANF before ischaemia has the potential to ameliorate I/R-triggered UPR and liver injury. Further study showed that MANF deficiency activated ATF4/CHOP and JNK/c-JUN/CHOP pathways, and rhMANF inhibited the activation of the two proapoptotic pathways caused by MANF deletion. CONCLUSION: Collectively, our study unravels a previously unknown relationship among MANF, UPR and hepatic I/R injury.


Subject(s)
Endoplasmic Reticulum Stress , Nerve Growth Factors , Reperfusion Injury , Animals , Apoptosis , Astrocytes , Hepatocytes , Humans , Liver , Mice
14.
Cell Biochem Funct ; 39(1): 98-106, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32613670

ABSTRACT

Glaucoma is optic neuropathy that is characterized by progressive neurodegeneration of the retinal ganglion cells (RGCs) and axons. This condition will lead to visual impairment and bring glaucoma to become the second cause of blindness globally. Neuroprotection in glaucoma is needed to prevent the progression of optic neuropathy. In this study, we examined the effects of the superior colliculus (SC), and mesencephalic astrocyte-derived neurotrophic factor (MANF) secreted from the SC, on RGC survival after oxidative stress. SC slices and RGCs from rats (3-day old) were co-cultured using a 3D-transwell system. In addition, primary RGCs from 4 to 5-day-old rats were cultured and treated with 100 µM hydrogen peroxide (H2 O2 ), together with stimulation by MANF. Immunoblot and immunofluorescence analyses indicated down-regulated expression levels of several survival markers of RGCs. Extension of neurites was decreased in RGCs treated with 100 µM H2 O2 . Following co-culture with SC slices, or the addition of MANF, we found that both the down-regulated expression of neural markers and extension of neurites caused by oxidative stress in RGCs were blocked. Furthermore, we found a decrease in the expression of neural markers and extension of neurites after co-culture with MANF siRNA-treated SC slices compared with slices treated with mock siRNA, but, RGCs co-cultured with SC slices treated with MANF siRNA displayed no-changed about to apoptosis. These results suggest that MANF secreted from the SC may play an important role in maintenance of function and survival of RGCs. It is also possible that MANF is an important factor in neuroprotection of RGCs. SIGNIFICANCE OF THE STUDY: Glaucoma is a progressive neurodegenerative disease of the retinal ganglion cell (RGC) and their axons. Neuroprotection is aimed at protecting those neurons that are damaged glaucomatous optic neuropathy. We have now examined the effects of superior colliculus, or msencephalic astrocyte-derived neutrophic factor (MANF), secreted from superior colliculus, on RGC survival using co-culture system. Our results suggested that MANF may important key factor in neuroprotection of RGC.


Subject(s)
Glaucoma/metabolism , Nerve Growth Factors/metabolism , Neurites/metabolism , Oxidative Stress , Retinal Ganglion Cells/metabolism , Animals , Cell Survival , Glaucoma/pathology , Hydrogen Peroxide/pharmacology , Rats , Rats, Wistar , Retinal Ganglion Cells/pathology
15.
Int J Mol Sci ; 22(18)2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34575854

ABSTRACT

Mesencephalic astrocyte derived neurotrophic factor (MANF) and cerebral dopamine neurotrophic factor (CDNF) are novel evolutionary conserved trophic factors, which exhibit cytoprotective activity via negative regulation of unfolded protein response (UPR) and inflammation. Despite multiple reports demonstrating detrimental effect of MANF/CDNF downregulation, little is known about the control of their expression. miRNAs-small non-coding RNAs-are important regulators of gene expression. Their dysregulation was demonstrated in multiple pathological processes and their ability to modulate levels of other neurotrophic factors, glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF), was previously reported. Here, for the first time we demonstrated direct regulation of MANF and CDNF by miRNAs. Using bioinformatic tools, reporter assay and analysis of endogenous MANF and CDNF, we identified that miR-144 controls MANF expression, and miR-134 and miR-141 downregulate CDNF levels. We also demonstrated that this effect is human-specific and is executed via predicted binding sites of corresponding miRNAs. Finally, we found that miR-382 suppressed hCDNF expression indirectly. In conclusion, we demonstrate for the first time direct regulation of MANF and CDNF expression by specific miRNAs, despite the fact their binding sites are not strongly evolutionary conserved. Furthermore, we demonstrate a functional effect of miR-144 mediated regulation of MANF on ER stress response markers. These findings emphasize that (1) prediction of miRNA targets based on evolutionary conservation may miss biologically meaningful regulatory pairs; and (2) interpretation of miRNA regulatory effects in animal models should be cautiously validated.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Nerve Growth Factors/genetics , RNA Interference , 3' Untranslated Regions , Cell Line , Endoplasmic Reticulum Stress , Humans , RNA, Messenger/genetics
16.
Mol Ther ; 26(1): 238-255, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29050872

ABSTRACT

Cerebral ischemia activates endogenous reparative processes, such as increased proliferation of neural stem cells (NSCs) in the subventricular zone (SVZ) and migration of neural progenitor cells (NPCs) toward the ischemic area. However, this reparative process is limited because most of the NPCs die shortly after injury or are unable to arrive at the infarct boundary. In this study, we demonstrate for the first time that endogenous mesencephalic astrocyte-derived neurotrophic factor (MANF) protects NSCs against oxygen-glucose-deprivation-induced injury and has a crucial role in regulating NPC migration. In NSC cultures, MANF protein administration did not affect growth of cells but triggered neuronal and glial differentiation, followed by activation of STAT3. In SVZ explants, MANF overexpression facilitated cell migration and activated the STAT3 and ERK1/2 pathway. Using a rat model of cortical stroke, intracerebroventricular injections of MANF did not affect cell proliferation in the SVZ, but promoted migration of doublecortin (DCX)+ cells toward the corpus callosum and infarct boundary on day 14 post-stroke. Long-term infusion of MANF into the peri-infarct zone increased the recruitment of DCX+ cells in the infarct area. In conclusion, our data demonstrate a neuroregenerative activity of MANF that facilitates differentiation and migration of NPCs, thereby increasing recruitment of neuroblasts in stroke cortex.


Subject(s)
Cell Differentiation/genetics , Nerve Growth Factors/genetics , Nerve Regeneration/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Stroke/genetics , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Cell Death , Cell Self Renewal/genetics , Cells, Cultured , Disease Models, Animal , Doublecortin Protein , Fluorescent Antibody Technique , Gene Expression , Immunohistochemistry , Mice , Mice, Knockout , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , STAT3 Transcription Factor/metabolism , Stress, Physiological , Stroke/metabolism , Stroke/pathology
17.
Diabetologia ; 61(10): 2202-2214, 2018 10.
Article in English | MEDLINE | ID: mdl-30032427

ABSTRACT

AIMS/HYPOTHESIS: There is a great need to identify factors that could protect pancreatic beta cells against apoptosis or stimulate their replication and thus prevent or reverse the development of diabetes. One potential candidate is mesencephalic astrocyte-derived neurotrophic factor (MANF), an endoplasmic reticulum (ER) stress inducible protein. Manf knockout mice used as a model of diabetes develop the condition because of increased apoptosis and reduced proliferation of beta cells, apparently related to ER stress. Given this novel association between MANF and beta cell death, we studied the potential of MANF to protect human beta cells against experimentally induced ER stress. METHODS: Primary human islets were challenged with proinflammatory cytokines, with or without MANF. Cell viability was analysed and global transcriptomic analysis performed. Results were further validated using the human beta cell line EndoC-ßH1. RESULTS: There was increased expression and secretion of MANF in human beta cells in response to cytokines. Addition of recombinant human MANF reduced cytokine-induced cell death by 38% in human islets (p < 0.05). MANF knockdown in EndoC-ßH1 cells led to increased ER stress after cytokine challenge. Mechanistic studies showed that the protective effect of MANF was associated with repression of the NF-κB signalling pathway and amelioration of ER stress. MANF also increased the proliferation of primary human beta cells twofold when TGF-ß signalling was inhibited (p < 0.01). CONCLUSIONS/INTERPRETATION: Our studies show that exogenous MANF protein can provide protection to human beta cells against death induced by inflammatory stress. The antiapoptotic and mitogenic properties of MANF make it a potential therapeutic agent for beta cell protection.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Insulin-Secreting Cells/cytology , Nerve Growth Factors/metabolism , Astrocytes/metabolism , Cell Death/drug effects , Cell Proliferation , Cell Survival , Cells, Cultured , Cytokines/metabolism , Humans , Inflammation , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , NF-kappa B/metabolism , RNA, Small Interfering/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Transcriptome
18.
Endocr J ; 64(4): 403-410, 2017 Apr 29.
Article in English | MEDLINE | ID: mdl-28216543

ABSTRACT

Evidence has shown that endoplasmic reticulum (ER) stress was involved in the progression to type 2 diabetes mellitus (T2DM) and development of insulin resistance. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a novel secreted protein upregulated by ER stress. This study aimed to assess serum level of MANF in normal glucose tolerance (NGT) participants and newly diagnosed prediabetic and T2DM patients. A total of 257 participants with NGT, newly diagnosed prediabetes or T2DM were recruited from Yinchao and Hangtian communities of Chengdu, Sichuan, China. Serum MANF level was quantified by enzyme-linked immunosorbent assay (ELISA). The mean age for the 257 participants (147 females) was 62±8 years (range 44-78): 71 with NGT, 115 with newly diagnosed prediabetes and 71 with T2DM. Mean serum MANF level was significantly higher with newly diagnosed prediabetes and T2DM than NGT (2.89±1.09 and 3.03±1.73 vs 2.13±1.37 ng/mL, both p<0.001). MANF level was not correlated with insulin sensitivity indexes (homeostasis model assessment for insulin resistance [HOMA-IR], Matsuda Index and quantitative insulin sensitivity check index [QUICKI]) for NGT and T2DM participants but was correlated with such indexes for prediabetes patients. We concluded that serum MANF level was higher in patients with newly diagnosed prediabetes and T2DM than in NGT controls. MANF appears to be associated with Matsuda Index, QUICKI and HOMA-IR in prediabetes patients.


Subject(s)
Diabetes Mellitus, Type 2/blood , Insulin Resistance , Nerve Growth Factors/blood , Prediabetic State/blood , Adult , Aged , Asian People , Blood Glucose/metabolism , Case-Control Studies , China , Diabetes Mellitus, Type 2/diagnosis , Female , Glucose Intolerance/blood , Humans , Male , Middle Aged , Prediabetic State/diagnosis
19.
Immunopharmacol Immunotoxicol ; 38(3): 205-13, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27075782

ABSTRACT

Mesencephalic astrocyte-derived neurotrophic factor (MANF), a new evolutionary conserved neurotrophic factor (NTF), has been reported to protect midbrain dopaminergic neurons of neurodegenerative diseases such as Parkinson's disease (PD) model. Neural stem cells (NSCs) can play a role as the therapeutic tool in neurodegenerative diseases, but the inflammatory responses of central nervous system (CNS) appear to harm this function. Although studies have previously demonstrated the protective effect of MANF on neurons of CNS, it is lacking in making great efforts on the function of MANF on NSCs. The aim of this study was to investigate the antiinflammatory responses and signaling mechanisms of MANF on lipopolysaccharide (LPS)-induced NSCs. In the results, MANF decreased the proinflammatory cytokines of IL-1ß, TNF-α, and IFN-γ induced by LPS by regulating NF-κB and phosphorylation of p38-mitogen-activated protein kinases (MAPKs) pathways, neither p-JNK nor p-ERK signaling. These findings suggest that MANF can facilitate to protect the inflammatory responses of NSCs, and provide beneficial function for the application of NSCs in the therapy.


Subject(s)
Induced Pluripotent Stem Cells/immunology , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/immunology , NF-kappa B/immunology , Nerve Growth Factors/immunology , Neural Stem Cells/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Cells, Cultured , Induced Pluripotent Stem Cells/pathology , Inflammation/immunology , Inflammation/pathology , MAP Kinase Signaling System/drug effects , Mice
20.
J Psychiatr Res ; 176: 33-39, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38838432

ABSTRACT

BACKGROUND: Sleeping late has been associated with cognitive impairment, and insufficient sleep can affect the secretion of feeding-related cytokines. Feeding-related cytokines may contribute to cognitive deficits resulting from delayed bedtime. Glial cell line-derived neurotrophic factor (GDNF) and mesencephalic astrocyte-derived neurotrophic factor (MANF), which are feeding-related neurotrophic factors, have been associated with improved cognitive function and neuroprotective abilities. Enhanced expression of GDNF and MANF is linked to increased energy expenditure and hyperphagia, respectively. AIMS: This study aimed to investigate the association between cerebrospinal fluid (CSF) GDNF, MANF, cognition, and sleep time and to explore the moderating effects of GDNF and MANF on cognitive impairment in individuals who sleep late. METHOD: This cross-sectional study included participants (mean age 31.76 ± 10.22 years) who were categorized as ≤23 o'clock sleepers (n = 66) and >23 o'clock sleepers (n = 125) based on sleep time. Cognition was assessed using Montreal Cognitive Assessment (MoCA), and GDNF and MANF levels in CSF were measured. RESULTS: MANF may play a moderating role in the relationship between sleep time and cognition (R2 = 0.06, ß = 0.59, p = 0.031). Age showed a negative correlation with MoCA scores (R2 = 0.08, ß = -0.18), while education exhibited a positive correlation (ß = 0.17, both p < 0.05). Only ≤23 o'clock sleepers exhibited a negative correlation between MANF levels and BMI (r = -0.35, p = 0.005). CONCLUSIONS: This study provides hitherto undocumented evidence of the potential protective effect of CSF MANF on cognitive impairment of late sleepers, which suggests that maintaining a regular sleep schedule may contribute to cognition and overall health, with MANF playing a role in this process.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor , Nerve Growth Factors , Sleep , Humans , Male , Adult , Female , Cross-Sectional Studies , Glial Cell Line-Derived Neurotrophic Factor/cerebrospinal fluid , Nerve Growth Factors/cerebrospinal fluid , Sleep/physiology , Young Adult , Cognition/physiology , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/etiology , Middle Aged , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL