Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 223
Filter
1.
Nature ; 618(7967): 1072-1077, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37196676

ABSTRACT

Plasma membrane rupture (PMR) in dying cells undergoing pyroptosis or apoptosis requires the cell-surface protein NINJ11. PMR releases pro-inflammatory cytoplasmic molecules, collectively called damage-associated molecular patterns (DAMPs), that activate immune cells. Therefore, inhibiting NINJ1 and PMR may limit the inflammation that is associated with excessive cell death. Here we describe an anti-NINJ1 monoclonal antibody that specifically targets mouse NINJ1 and blocks oligomerization of NINJ1, preventing PMR. Electron microscopy studies showed that this antibody prevents NINJ1 from forming oligomeric filaments. In mice, inhibition of NINJ1 or Ninj1 deficiency ameliorated hepatocellular PMR induced with TNF plus D-galactosamine, concanavalin A, Jo2 anti-Fas agonist antibody or ischaemia-reperfusion injury. Accordingly, serum levels of lactate dehydrogenase, the liver enzymes alanine aminotransaminase and aspartate aminotransferase, and the DAMPs interleukin 18 and HMGB1 were reduced. Moreover, in the liver ischaemia-reperfusion injury model, there was an attendant reduction in neutrophil infiltration. These data indicate that NINJ1 mediates PMR and inflammation in diseases driven by aberrant hepatocellular death.


Subject(s)
Antibodies, Monoclonal , Cell Membrane , Inflammation , Liver , Nerve Growth Factors , Reperfusion Injury , Animals , Mice , Alanine Transaminase , Alarmins , Antibodies, Monoclonal/immunology , Aspartate Aminotransferases , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/immunology , Cell Adhesion Molecules, Neuronal/ultrastructure , Cell Death , Cell Membrane/pathology , Cell Membrane/ultrastructure , Concanavalin A , Galactosamine , Hepatocytes/pathology , Hepatocytes/ultrastructure , Inflammation/pathology , Lactate Dehydrogenases , Liver/pathology , Microscopy, Electron , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/deficiency , Nerve Growth Factors/immunology , Nerve Growth Factors/ultrastructure , Neutrophil Infiltration , Reperfusion Injury/pathology
2.
Invest Ophthalmol Vis Sci ; 62(7): 18, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34132748

ABSTRACT

Purpose: The cornea is richly innervated by the trigeminal ganglion (TG) and its function supported by secretions from the adjacent lacrimal (LG) and meibomian glands (MG). In this study we examined how pigment epithelium-derived factor (PEDF) gene deletion affects the cornea structure and function. Methods: We used PEDF hemizygous and homozygous knockout mice to study effects of PEDF deficiency on corneal innervation assessed by beta tubulin staining, mRNA expression of trophic factors, and PEDF receptors by adjacent supporting glands, corneal sensitivity measured using a Cochet-Bonnet esthesiometer, and tear production using phenol red cotton thread wetting. Results: Loss of PEDF was accompanied by reduced corneal innervation and sensitivity, increased corneal surface injury and tear production, thinning of the corneal stroma and loss of stromal cells. PEDF mRNA was expressed in the cornea and its supporting tissues, the TG, LG, and MG. Deletion of one or both PEDF alleles resulted in decreased expression of essential trophic support in the TG, LG, and MG including nerve growth factor, brain-derived neurotrophic growth factor, and GDNF with significantly increased levels of NT-3 in the LG and decreased EGF expression in the cornea. Decreased transcription of the putative PEDF receptors, adipose triglyceride lipase, lipoprotein receptor-related protein 6, laminin receptor, PLXDC1, and PLXDC2 was also evident in the TG, LG and MG with the first three showing increased levels in corneas of the Pedf+/- and Pedf-/- mice compared to wildtype controls. Constitutive inactivation of ERK1/2 and Akt was pronounced in the TG and cornea, although their protein levels were dramatically increased in Pedf-/- mice. Conclusions: This study highlights an essential role for PEDF in corneal structure and function and confirms the reported rescue of exogenous PEDF treatment in corneal pathologies. The pleiotropic effects of PEDF deletion on multiple trophic factors, receptors and signaling molecules are strong indications that PEDF is a key coordinator of molecular mechanisms that maintain corneal function and could be exploited in therapeutic options for several ocular surface diseases.


Subject(s)
Cornea , Corneal Diseases , Eye Proteins , Nerve Growth Factors , Serpins , Tears/physiology , Trigeminal Ganglion , Animals , Cornea/innervation , Cornea/pathology , Cornea/physiopathology , Corneal Diseases/metabolism , Corneal Diseases/physiopathology , Corneal Diseases/therapy , Corneal Injuries/metabolism , Corneal Injuries/physiopathology , Eye Proteins/genetics , Eye Proteins/pharmacology , Gene Deletion , Humans , Mice , Mice, Knockout , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Protease Inhibitors/pharmacology , Receptors, Neuropeptide/metabolism , Serpins/deficiency , Serpins/genetics , Serpins/pharmacology , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/physiopathology , Tubulin/metabolism , Visual Perception/physiology
3.
Inflammation ; 44(5): 1916-1926, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33939070

ABSTRACT

Bacterial myocarditis is a key cause leading to myocardial damage and cardiac dysfunction. Mesencephalic astrocyte-derived neurotrophic factor (MANF) has been found to be an anti-inflammatory factor. This study is to explore the effect of MANF on LPS-induced myocardial inflammation and macrophage differentiation. The myocarditis mouse model was constructed by LPS treatment. Myocardial damage and serum inflammatory factors were evaluated by ELISA. RT-qPCR was used to detect mRNA of M1/M2 macrophage markers. Western blot, immunohistochemical, and immunofluorescent staining were used to examine myocardial M1/M2 macrophages and NF-κB activation. Mono-macrophage-derived MANF deficiency enhanced LPS-induced inflammatory response and increased M1 macrophages in myocardium tissues, further causing more severe myocardial injury and lower survival rate of mice. Also, LPS-induced myocardial NF-κB activation was strengthened after mono-macrophage-derived MANF knockout. Mono-macrophage-derived MANF inhibits bacterial myocarditis and myocardial M1 macrophage differentiation, which is potential to be used for bacterial myocarditis treatment clinically.


Subject(s)
Inflammation Mediators/metabolism , Macrophages/metabolism , Myocarditis/metabolism , Myocardium/metabolism , NF-kappa B/metabolism , Nerve Growth Factors/deficiency , Animals , Inflammation Mediators/antagonists & inhibitors , Lipopolysaccharides/toxicity , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/chemically induced , Myocarditis/pathology , NF-kappa B/antagonists & inhibitors
4.
Dev Cell ; 56(9): 1346-1358.e6, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33945785

ABSTRACT

Myelination is essential for central nervous system (CNS) formation, health, and function. Emerging evidence of oligodendrocyte heterogeneity in health and disease and divergent CNS gene expression profiles between mice and humans supports the development of experimentally tractable human myelination systems. Here, we developed human iPSC-derived myelinating organoids ("myelinoids") and quantitative tools to study myelination from oligodendrogenesis through to compact myelin formation and myelinated axon organization. Using patient-derived cells, we modeled a monogenetic disease of myelinated axons (Nfasc155 deficiency), recapitulating impaired paranodal axo-glial junction formation. We also validated the use of myelinoids for pharmacological assessment of myelination-both at the level of individual oligodendrocytes and globally across whole myelinoids-and demonstrated reduced myelination in response to suppressed synaptic vesicle release. Our study provides a platform to investigate human myelin development, disease, and adaptive myelination.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myelin Sheath/physiology , Organoids/physiology , Axons/metabolism , Axons/ultrastructure , Humans , Myelin Sheath/ultrastructure , Nerve Growth Factors/deficiency , Nerve Growth Factors/metabolism , Organoids/ultrastructure , Tetanus Toxin/pharmacology , Time Factors
5.
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
6.
Inflammation ; 44(2): 693-703, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33145627

ABSTRACT

The outburst of renal inflammatory response has been found to be a crucial cause of acute kidney injury (AKI). Attenuating the renal inflammation is an effective way for AKI treatment. Mesencephalic astrocyte-derived neurotrophic factor (MANF) has been proven to be an anti-inflammatory factor. However, the effect of MANF on renal inflammation induced by AKI is unknown. In this study, we have investigated the effect of mono-macrophage-derived MANF on AKI. We constructed the mono-macrophage-specific MANF knockout (Mø MANF-/-) mouse and used lipopolysaccharide (LPS) to induce AKI in wild-type (WT) and Mø MANF-/- mice. With mono-macrophage-specific MANF deficiency, Mø MANF-/- mice had a lower survival rate, more severe renal injury, and higher serum level of pro-inflammatory TNF-α after AKI was induced by LPS. Also, compared with WT mice, there were more M1 macrophages in renal tissues of Mø MANF-/- mice with LPS treatment, which might be attributed to the enhanced NF-κB activation in the renal microenvironment. Our study indicates the immunoregulatory role of mono-macrophage-derived MANF in the pathophysiological process of AKI, as well as the potential clinical application of MANF for AKI treatment.


Subject(s)
Acute Kidney Injury/immunology , Inflammation/immunology , Macrophages/immunology , Nerve Growth Factors/immunology , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Acute Kidney Injury/mortality , Animals , Biomarkers/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , Immunohistochemistry , Inflammation/etiology , Inflammation/metabolism , Inflammation/mortality , Lipopolysaccharides , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Real-Time Polymerase Chain Reaction , Severity of Illness Index
7.
Exp Eye Res ; 198: 108121, 2020 09.
Article in English | MEDLINE | ID: mdl-32721425

ABSTRACT

The SERPINF1 gene encodes pigment epithelium-derived factor (PEDF), a member of the serpin superfamily with neurotrophic and antiangiogenic properties in the retina. We hypothesized that absence of PEDF would lead to increased stress-associated retinal degeneration in Serpinf1 null mice. Accordingly, using a Serpinf1 null mouse model, we investigated the impact of PEDF absence on retinal morphology, and susceptibility to induced and inherited retinal degeneration. We studied the pattern of Serpinf1 expression in the mouse retina layers. PEDF protein was detected by western blotting. Transmission electron microscopy was performed on mouse retina. Serpinf1 null mice and wild type littermates were injected with NaIO3 (30 mg/kg body weight) intraperitonially. At post-injection day 1, 3, 4, 6 and 8 mice were euthanized, and eyes were enucleated. Serpinf1 null and rd10 double mutant mice were generated and their eyes enucleated at different time points from post-natal day 15 to post-natal day 28. Enucleated eyes were processed for hematoxylin and eosin staining and histopathological evaluations. We found that Serpinf1 was expressed in the retinal pigment epithelium, in the inner nuclear layer and in the ganglion cell layer, but undetectable in the outer nuclear layer of wild type mice. Plasma PEDF protein levels were undetectable in Serpinf1 null animals. RPE atrophy and retinal thinning were observed in NaIO3-treated wild type mice that progressed with time post-injection. NaIO3-treated Serpinf1 null mice showed comparatively better retinal morphology than wild type mice at day 4 post-injection. However, the absence of PEDF in Serpinf1 null x rd10 mice increased the susceptibility to retinal degeneration relative to that of rd10 mice. We concluded that histopathological evaluation of retinas lacking PEDF showed that removal of the Serpinf1 gene may activate PEDF-independent compensatory mechanisms to protect the retina against oxidative stress, while it increases the susceptibility to degenerate the retina in inherited retinal degeneration models.


Subject(s)
Nerve Growth Factors/deficiency , Retinal Degeneration/metabolism , Serpins/deficiency , Animals , Blotting, Western , Disease Models, Animal , Disease Progression , Eye Proteins/genetics , Eye Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Serpins/genetics , Serpins/metabolism
8.
FASEB J ; 34(6): 8702-8720, 2020 06.
Article in English | MEDLINE | ID: mdl-32385864

ABSTRACT

Disruption of colonic homeostasis caused by aberrant M1/M2 macrophage polarization and dysbiosis contributes to inflammatory bowel disease (IBD) pathogenesis. However, the molecular factors mediating colonic homeostasis are not well characterized. Here, we found that Ninjurin1 (Ninj1) limits colon inflammation by regulating macrophage polarization and microbiota composition under homeostatic conditions and during colitis development. Ninj1 deletion in mice induced hypersusceptibility to colitis, with increased prevalence of colitogenic Prevotellaceae strains and decreased immunoregulatory Lachnospiraceae strains. Upon co-housing (CoH) with WT mice, Ninj1-/- mice showed increased Lachnospiraceae and decreased Prevotellaceae abundance, with subsequent improvement of colitis. Under homeostatic conditions, M1 macrophage frequency was higher in the Ninj1-/- mouse colons than wild-type (WT) mouse colons, which may contribute to increased basal colonic inflammation and microbial imbalance. Following colitis induction, Ninj1 expression was increased in macrophages; meanwhile Ninj1-/- mice showed severe colitis development and impaired recovery, associated with decreased M2 macrophages and escalated microbial imbalance. In vitro, Ninj1 knockdown in mouse and human macrophages activated M1 polarization and restricted M2 polarization. Finally, the transfer of WT macrophages ameliorated severe colitis in Ninj1-/- mice. These findings suggest that Ninj1 mediates colonic homeostasis by modulating M1/M2 macrophage balance and preventing extensive dysbiosis, with implications for IBD prevention and therapy.


Subject(s)
Cell Adhesion Molecules, Neuronal/deficiency , Colitis/metabolism , Colitis/pathology , Gastrointestinal Microbiome/physiology , Macrophages/metabolism , Macrophages/pathology , Nerve Growth Factors/deficiency , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Colitis/microbiology , Colon/metabolism , Colon/microbiology , Colon/pathology , Disease Models, Animal , Homeostasis/physiology , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Macrophage Activation/physiology , Male , Mice , Nerve Growth Factors/metabolism , THP-1 Cells/metabolism
9.
Acta Pharmacol Sin ; 41(12): 1568-1575, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32265491

ABSTRACT

Dyslipidemia is a risk factor for cardiovascular diseases and type 2 diabetes. Several adipokines play important roles in modulation of blood lipids. Metrnl is a recently identified adipokine, and adipose Metrnl participates in regulation of blood triglyceride (TG). In this study, we generated Metrnl global, intestine-specific and liver-specific knockout mice, and explored the effects of Metrnl on serum lipid parameters. Global knockout of Metrnl had no effects on serum lipid parameters under normal chow diet, but increased blood TG by 14%, and decreased total cholesterol (TC) by 16% and high density lipoprotein cholesterol (HDL-C) by 24% under high fat diet. Nevertheless, intestine-specific knockout of Metrnl did not alter the serum lipids parameters under normal chow diet or high fat diet. Notably, liver-specific knockout of Metrnl decreased HDL-C by 24%, TC by 20% and low density lipoprotein cholesterol (LDL-C) by 16% without alterations of blood TG and nonesterified fatty acids (NEFA) under high fat diet. But deficiency of Metrnl in liver did not change VLDL secretion and expression of lipid synthetic and metabolic genes. We conclude that tissue-specific Metrnl controls different components of blood lipids. In addition to modulation of blood TG by adipose Metrnl, blood HDL-C is regulated by liver Metrnl.


Subject(s)
Cholesterol, HDL/metabolism , Nerve Growth Factors/deficiency , Triglycerides/metabolism , Animals , Cholesterol, HDL/blood , Diet, High-Fat , Gene Knockout Techniques , Mice, Inbred C57BL , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Triglycerides/blood
10.
Cell Death Dis ; 11(2): 100, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029702

ABSTRACT

The non-conventional neurotrophic factor mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER)-resident protein that promotes ER homeostasis. MANF has a cytoprotective function, shown in the central nervous system neurons and pancreatic beta cells. Here, we report that MANF is expressed in the hair cells and neurons and in selected non-sensory cells of the cochlea and that Manf inactivation triggers upregulation of the ER chaperones in these cells. However, Manf inactivation resulted in the death of only outer hair cells (OHCs), the cells responsible for sound amplification in the cochlea. All OHCs were formed in Manf-inactivated mice, but progressive OHC death started soon after the onset of hearing function. The robust OHC loss was accompanied by strongly elevated hearing thresholds. Conditional Manf inactivation demonstrated that MANF has a local function in the cochlea. Immunostainings revealed the upregulation of CHOP, the pro-apoptotic component of the unfolded protein response (UPR), in Manf-inactivated OHCs, linking the UPR to the loss of these cells. The phenotype of Manf-inactivated OHCs was distinctly dependent on the mouse strain, such that the strains characterized by early-onset age-related hearing loss (C57BL/6J and CD-1) were affected. These results suggest that Manf deficiency becomes detrimental when accompanied by gene mutations that predispose to hearing loss, by intensifying ER dyshomeostasis. Together, MANF is the first growth factor shown to antagonize ER stress-mediated OHC death. MANF might serve as a therapeutic candidate for protection against hearing loss induced by the ER-machinery-targeting stressors.


Subject(s)
Cochlea/metabolism , Endoplasmic Reticulum Stress , Hair Cells, Auditory, Outer/metabolism , Hearing Loss/metabolism , Hearing , Nerve Growth Factors/deficiency , Animals , Auditory Threshold , Cell Death , Cochlea/pathology , Cochlea/physiopathology , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory, Outer/pathology , Hearing Loss/genetics , Hearing Loss/pathology , Hearing Loss/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/genetics , Species Specificity
11.
Acta Pharmacol Sin ; 41(6): 763-770, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31949292

ABSTRACT

Metrnl is a newly identified secreted protein highly expressed in the intestinal epithelium. This study aimed to explore the role and mechanism of intestinal epithelial Metrnl in ulcerative colitis. Metrnl-/- (intestinal epithelial cell-specific Metrnl knockout) mice did not display any phenotypes of colitis under basal conditions. However, under administration of 3% dextran sodium sulfate (DSS) drinking water, colitis was more severe in Metrnl-/- mice than in WT mice, as indicated by comparisons of body weight loss, the presence of occult or gross blood per rectum, stool consistency, shrinkage in the colon, intestinal damage, and serum levels of inflammatory factors. DSS-induced colitis activated autophagy in the colon. This activation was partially inhibited by intestinal epithelial Metrnl deficiency, as indicated by a decrease in Beclin-1 and LC3-II/I and an increase in p62 in DSS-treated Metrnl-/- mice compared with WT mice. These phenomena were further confirmed by observation of autophagosomes and immunofluorescence staining for LC3 in epithelial cells. The autophagy-related AMPK-mTOR-p70S6K pathway was also activated in DSS-induced colitis, and this pathway was partially blocked by intestinal epithelial Metrnl deficiency, as indicated by a decrease in AMPK phosphorylation and an increase in mTOR and p70S6K phosphorylation in DSS-treated Metrnl-/- mice compared with WT mice. Therefore, Metrnl deficiency deteriorated ulcerative colitis at least partially through inhibition of autophagy via the AMPK-mTOR-p70S6K pathway, suggesting that Metrnl is a therapeutic target for ulcerative colitis.


Subject(s)
Autophagy , Colitis, Ulcerative/metabolism , Epithelial Cells/metabolism , Nerve Growth Factors/metabolism , Administration, Oral , Animals , Caco-2 Cells , Cells, Cultured , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Dextran Sulfate/administration & dosage , Epithelial Cells/pathology , Humans , Mice , Mice, Knockout , Mice, Transgenic , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Eur J Cell Biol ; 98(5-8): 151043, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31138438

ABSTRACT

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is the only human neurotrophic factor with an evolutionarily-conserved C. elegans homolog, Y54G2A.23 or manf-1. MANF is a small, soluble, endoplasmic-reticulum (ER)-resident protein that is secreted upon ER stress and promotes survival of target cells such as neurons. However, the role of MANF in ER stress and its mechanism of cellular protection are not clear and the function of MANF in C. elegans is only beginning to emerge. In this study, we show that depletion of C. elegans manf-1 causes a slight decrease in lifespan and brood size; furthermore, combined depletion of manf-1 and the IRE-1/XBP-1 ER stress/UPR pathway resulted in sterile animals that did not produce viable progeny. We demonstrate upregulation of markers of ER stress in L1 larval nematodes, as measured by hsp-3 and hsp-4 transcription, upon depletion of manf-1 by RNAi or mutation; however, there was no difference in tunicamycin-induced expression of hsp-3 and hsp-4 between wild-type and MANF-deficient worms. Surprisingly, larval growth arrest observed in wild-type nematodes reared on tunicamycin is completely prevented in the manf-1 (tm3603) mutant. Transcriptional microarray analysis revealed that manf-1 mutant L1 larvae exhibit a novel modulation of innate immunity genes in response to tunicamycin. The hypothesis that manf-1 negatively regulates the innate immunity pathway is supported by our finding that the development of manf-1 mutant larvae compared to wild-type larvae is not inhibited by growth on P. aeruginosa. Together, our data represent the first characterization of C. elegans MANF as a key modulator of organismal ER stress and immunity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Endoplasmic Reticulum Stress/drug effects , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Tunicamycin/pharmacology , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/metabolism , Immunity, Innate/drug effects , Larva/drug effects , Larva/immunology , Nerve Growth Factors/metabolism , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/metabolism
13.
Arterioscler Thromb Vasc Biol ; 38(10): 2358-2370, 2018 10.
Article in English | MEDLINE | ID: mdl-30354207

ABSTRACT

Objective- Angiogenesis, entire step from endothelial cells (ECs) sprouts to vascular maturation, is a critical response to ischemia. To form functional mature vessels, interactions between ECs and pericytes are essential. Ninj1 (ninjurin1) is an adhesion molecule that contributes to the pathogenesis of neuroinflammation. We recently demonstrated that Ninj1 is expressed in pericytes during angiogenesis. However, the role of Ninj1 in angiogenesis under pathophysiological ischemic conditions has not yet been elucidated. Approach and Results- Ninj1 was detected in microvessels, and its expression was enhanced in ischemic tissues after mouse hindlimb ischemia. Knockdown of Ninj1 was performed by injection of biodegradable microspheres releasing Ninj1-small interfering RNA into muscle tissues. Alternatively, pericyte-specific Ninj1 knockout was induced by tamoxifen treatment of NG2-CreERT/Ninj1-flox mice. Ninj1 knockdown/knockout reduced the formation of blood-circulating functional vessels among total CD31+ microvessels within ischemic tissues and subsequently attenuated color Doppler-assessed blood flow recovery. Ninj1 overexpression enhanced expression of Anpt (angiopoietin) 1, whereas Ninj1 knockdown enhanced the endogenous Anpt1 antagonist, Anpt2 expression in pericytes and inhibited the association of pericytes with ECs and subsequent formation of capillary-like structure, that is, EC tube surrounded with pericytes in 3-dimensional gel culture. Conclusions- Our data demonstrate that Ninj1 is involved in the formation of functional matured vessels through the association between pericytes and ECs, resulting in blood flow recovery from ischemia. These findings further the current our understanding of vascular maturation and may support the development of therapeutics for ischemic diseases.


Subject(s)
Cell Adhesion Molecules, Neuronal/deficiency , Endothelial Cells/metabolism , Gene Deletion , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Nerve Growth Factors/deficiency , Pericytes/metabolism , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Communication , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Gene Knockdown Techniques , Hindlimb , Ischemia/genetics , Ischemia/physiopathology , Male , Mice, Inbred C57BL , Nerve Growth Factors/genetics , Recovery of Function , Regional Blood Flow , Signal Transduction
14.
Mol Biotechnol ; 60(8): 539-549, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29876740

ABSTRACT

In this study, we investigated expression and dimerization of an ER-associated degradation (ERAD) substrate, a null Hong Kong variant of α-1-antitrypsin (NHK) using immunoblotting assay and a novel NanoLuc complementary reporter system called the NanoBiT (NB) assay. This NB-tagged NHK made it possible to monitor the intra- and extracellular status of NHK in living cells. The values for this NB assay fluctuated in response to distinct pharmacological stimuli and co-transfection of several ERAD-related factors. We then focused on mesencephalic astrocyte-derived neurotrophic factor (MANF), an unclarified ATF6/IRE1-downstream target, and established MANF-deficient Neuro2a (N2a) cells using CRISPR/Cas9 system. MANF-deficient N2a significantly elevated OS-9 protein after tunicamycin treatment; however, no specific differences in intra- and extracellular status of NHK protein were observed between wild-type and MANF-deficient cells. Taken together, intrinsic MANF in N2a cells is not strongly associated with the accumulation and clearance of unfolded proteins within the ER under current condition, but this novel NB assay is a useful approach for characterizing the protein status including ERAD substrates.


Subject(s)
Endoplasmic Reticulum/metabolism , Nanotechnology/methods , alpha 1-Antitrypsin/chemistry , alpha 1-Antitrypsin/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Endoplasmic Reticulum/drug effects , Gene Knockout Techniques , Luciferases/metabolism , Mice , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Protein Multimerization , Tunicamycin/pharmacology , alpha 1-Antitrypsin/genetics
15.
Am J Physiol Cell Physiol ; 314(4): C456-C472, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351407

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of vision loss among the elderly population, and is associated with severe macular degeneration and choroidal neovascularization (CNV). Although the pathogenesis of AMD is associated with choroidal dysfunction and CNV, the detailed underlying mechanisms remain unresolved. Altered production of pigment epithelium-derived factor (PEDF), a neuroprotective and antiangiogenic factor, contributes to CNV. Furthermore, exogenous PEDF mitigates angiogenesis in preclinical CNV models. How PEDF expression affects choroidal endothelial cell (ChEC) function is unknown. Here we isolated ChECs from PEDF+/+ and PEDF-deficient (PEDF-/-) mice and determined the impact of PEDF expression on the proangiogenic and pro-inflammatory properties of ChECs. We showed that PEDF expression significantly affects the proliferation, migration, adhesion, and oxidative and inflammatory state of ChECs. The PEDF-/- ChECs were, however, more sensitive to H2O2 challenge and exhibited increased rate of apoptosis and oxidative stress. We also observed a significant increase in production of cytokines with a primary role in inflammation and angiogenesis including vascular endothelial growth factor (VEGF) and osteopontin, and a reprograming of chemokines and cytokines expression profiles in PEDF-/- ChECs. Collectively, our results indicate that PEDF expression has a significant impact on oxidative and inflammatory properties of ChECs, whose alteration could contribute to pathogenesis of chronic inflammatory diseases including exudative AMD.


Subject(s)
Choroid/blood supply , Choroidal Neovascularization/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Eye Proteins/metabolism , Inflammation Mediators/metabolism , Macular Degeneration/metabolism , Nerve Growth Factors/metabolism , Oxidative Stress , Serpins/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Endothelial Cells/pathology , Eye Proteins/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Osteopontin/metabolism , Serpins/deficiency , Serpins/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Clin Chim Acta ; 478: 216-221, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29104038

ABSTRACT

BACKGROUNDS: SERPINF1 mutations caused deficiency of pigment epithelium-derived factor (PEDF) and would lead to osteogenesis imperfecta (OI) type VI. However, serum PEDF levels were unclear in Chinese OI patients who had clear molecular diagnosis. OBJECTIVE: To assess PEDF levels in different genotypes of OI, to evaluate the influencing factors of PEDF in Chinese OI patients with clear molecular diagnosis. METHODS: Known candidate genes of OI were examined by a targeted next generation sequence. Serum PEDF levels were measured by ELISA in 6 OI patients with SERPINF1 mutations, 6 carriers of one copy of the SERPINF1 mutation, 88 OI patients with COL1A1, CLO1A2, IFITM5 and other pathogenic mutations of OI and 24 healthy controls. We compared the differences in serum PEDF levels among different OI patients and normal controls. RESULTS: Serum PEDF levels were extremely low in OI patients with SERPINF1 mutations (0.66±1.60µg/ml) than in OI patients with other pathogenic mutations (4.88±1.43-7.07±2.43µg/ml), carriers of one copy of SERPINF1 mutation (4.94±2.35µg/ml), and normal controls (7.29±2.31µg/m) (P<0.001). No significant differences in serum PEDF concentrations were found among patients with OI type I, III or IV, and between patients with or without bisphosphonate treatment. Serum PEDF level was positively correlated with Z-score of weight (r=0.310, P=0.004), BMI (r=0.253, P=0.020) and alanine aminotransferase (r=0.291, P=0.007). CONCLUSIONS: Extremely low level of PEDF was demonstrated as a specific, convenient, and inexpensive diagnostic biomarker for OI patients with SERPINF1 mutations, but it could not provide information regarding the clinical severity of OI and the efficacy of bisphosphonates treatment.


Subject(s)
Eye Proteins/blood , Mutation , Nerve Growth Factors/blood , Osteogenesis Imperfecta/genetics , Serpins/blood , Adult , Asian People , Biomarkers/blood , Case-Control Studies , Diphosphonates/therapeutic use , Eye Proteins/genetics , Humans , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Osteogenesis Imperfecta/blood , Osteogenesis Imperfecta/classification , Serpins/deficiency , Serpins/genetics , Young Adult
17.
Mol Psychiatry ; 23(5): 1233-1243, 2018 05.
Article in English | MEDLINE | ID: mdl-28322273

ABSTRACT

Numerous genetic and functional studies implicate variants of Neuregulin-1 (NRG1) and its neuronal receptor ErbB4 in schizophrenia and many of its endophenotypes. Although the neurophysiological and behavioral phenotypes of NRG1 mutant mice have been investigated extensively, practically nothing is known about the function of NRG2, the closest NRG1 homolog. We found that NRG2 expression in the adult rodent brain does not overlap with NRG1 and is more extensive than originally reported, including expression in the striatum and medial prefrontal cortex (mPFC), and therefore generated NRG2 knockout mice (KO) to study its function. NRG2 KOs have higher extracellular dopamine levels in the dorsal striatum but lower levels in the mPFC; a pattern with similarities to dopamine dysbalance in schizophrenia. Like ErbB4 KO mice, NRG2 KOs performed abnormally in a battery of behavioral tasks relevant to psychiatric disorders. NRG2 KOs exhibit hyperactivity in a novelty-induced open field, deficits in prepulse inhibition, hypersensitivity to amphetamine, antisocial behaviors, reduced anxiety-like behavior in the elevated plus maze and deficits in the T-maze alteration reward test-a task dependent on hippocampal and mPFC function. Acute administration of clozapine rapidly increased extracellular dopamine levels in the mPFC and improved alternation T-maze performance. Similar to mice treated chronically with N-methyl-d-aspartate receptor (NMDAR) antagonists, we demonstrate that NMDAR synaptic currents in NRG2 KOs are augmented at hippocampal glutamatergic synapses and are more sensitive to ifenprodil, indicating an increased contribution of GluN2B-containing NMDARs. Our findings reveal a novel role for NRG2 in the modulation of behaviors with relevance to psychiatric disorders.


Subject(s)
Dopamine/metabolism , Mental Disorders/metabolism , Nerve Growth Factors/deficiency , Animals , Behavior, Animal/physiology , Brain/metabolism , Clozapine/pharmacology , Dopamine/genetics , ErbB Receptors/metabolism , Male , Mental Disorders/genetics , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neuregulin-1/genetics , Neuregulin-1/metabolism , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Signal Transduction , Synapses/metabolism , Transcriptome
18.
Am J Physiol Cell Physiol ; 313(4): C405-C420, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28747334

ABSTRACT

Pigment epithelium-derived factor (PEDF) is an endogenous inhibitor of angiogenesis. Although various ocular cell types including retinal endothelial cells (EC) produce PEDF, we know very little about cell autonomous effects of PEDF in these cell types. Here we determined how PEDF expression affects retinal EC proangiogenic properties. Retinal EC were prepared from wild-type (PEDF+/+) and PEDF-deficient (PEDF-/-) mice. The identity of EC was confirmed by staining for specific markers including vascular endothelial cadherin, CD31, and B4-lectin. Retinal EC also expressed VEGF receptor 1 and endoglin, as well as ICAM-1, ICAM-2, and VCAM-1. PEDF-/- retinal EC were more proliferative, less apoptotic when challenged with H2O2, less migratory, and less adherent compared with PEDF+/+ EC. These changes could be associated, at least in part, with increased levels of tenascin-C, fibronectin, thrombospondin-1 and collagen IV, and lower amounts of osteopontin. PEDF-/- EC also exhibited alterations in expression of a number of integrins including α2, αv, ß1, ß8, and αvß3, and cell-cell adhesion molecules including CD31, zonula occluden-1, and occludin. These observations correlated with attenuation of capillary morphogenesis and increased levels of oxidative stress in PEDF-/- EC. PEDF-/- EC also produced lower levels of VEGF compared with PEDF+/+ cells. Thus, PEDF deficiency has a significant impact on retinal EC adhesion and migration, perhaps through altered production of extracellular matrix and junctional proteins in response to increased oxidative stress affecting their proangiogenic activity.


Subject(s)
Cell Adhesion , Endothelial Cells/metabolism , Eye Proteins/metabolism , Neovascularization, Physiologic , Nerve Growth Factors/metabolism , Retinal Vessels/metabolism , Serpins/metabolism , Animals , Apoptosis , Cell Adhesion Molecules/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Endoglin/metabolism , Endothelial Cells/pathology , Extracellular Matrix/metabolism , Eye Proteins/genetics , Female , Genotype , Humans , Integrins/metabolism , Male , Mice, Knockout , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Oxidative Stress , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Retinal Vessels/pathology , Serpins/deficiency , Serpins/genetics , Signal Transduction , Tight Junction Proteins/metabolism , Time Factors , Transfection , Vascular Endothelial Growth Factor Receptor-1/metabolism
19.
Cell Death Dis ; 8(6): e2838, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28569772

ABSTRACT

Distant metastasis is the primary cause of nasopharyngeal carcinoma (NPC) treatment failure while epithelial-mesenchymal transition (EMT) is the critical process of NPC invasion and metastasis. However, tumor-suppressor genes involved in the EMT and metastasis of NPC have not been explored clearly compared with the oncogenes. In the present study, the expression of pigment epithelium-derived factor (PEDF), a potent endogenous antitumor factor, was diminished in human NPC tissues and associated with clinicopathological and EMT features. The knockdown of PEDF induced EMT in lower metastatic NPC cell lines and overexpression of PEDF restored epithelial phenotype in higher metastatic NPC cell lines with typical EMT. The inhibition of PEDF mediated NPC cell spontaneous metastasis in vivo. LRP6/GSK3ß/ß-catenin signal pathway rather than AKT/GSK3ß pathway was involved in the effects of PEDF on EMT. The expression of PEDF was directly downregulated by elevated miR-320c in NPC. In conclusion, our findings indicate for the first time that PEDF functions as tumor-suppressor gene in the occurrence of EMT and metastasis in NPC. PEDF could serve as a promising candidate for NPC diagnosis, prognosis and treatment.


Subject(s)
Carcinoma/genetics , Epithelial-Mesenchymal Transition/genetics , Eye Proteins/genetics , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/genetics , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Nasopharyngeal Neoplasms/genetics , Nerve Growth Factors/genetics , Serpins/genetics , beta Catenin/genetics , Animals , Base Sequence , Binding Sites , Carcinoma/metabolism , Carcinoma/mortality , Carcinoma/pathology , Case-Control Studies , Cell Line, Tumor , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Lymphatic Metastasis , Male , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/mortality , Nasopharyngeal Neoplasms/pathology , Neoplasm Staging , Neoplasm Transplantation , Nerve Growth Factors/deficiency , Prognosis , Serpins/deficiency , Signal Transduction , beta Catenin/metabolism
20.
Nature ; 545(7654): 350-354, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28445456

ABSTRACT

Netrin-1 is an evolutionarily conserved, secreted extracellular matrix protein involved in axon guidance at the central nervous system midline. Netrin-1 is expressed by cells localized at the central nervous system midline, such as those of the floor plate in vertebrate embryos. Growth cone turning assays and three-dimensional gel diffusion assays have shown that netrin-1 can attract commissural axons. Loss-of-function experiments further demonstrated that commissural axon extension to the midline is severely impaired in the absence of netrin-1 (refs 3, 7, 8, 9). Together, these data have long supported a model in which commissural axons are attracted by a netrin-1 gradient diffusing from the midline. Here we selectively ablate netrin-1 expression in floor-plate cells using a Ntn1 conditional knockout mouse line. We find that hindbrain and spinal cord commissural axons develop normally in the absence of floor-plate-derived netrin-1. Furthermore, we show that netrin-1 is highly expressed by cells in the ventricular zone, which can release netrin-1 at the pial surface where it binds to commissural axons. Notably, Ntn1 deletion from the ventricular zone phenocopies commissural axon guidance defects previously described in Ntn1-knockout mice. These results show that the classical view that attraction of commissural axons is mediated by a gradient of floor-plate-derived netrin-1 is inaccurate and that netrin-1 primarily acts locally by promoting growth cone adhesion.


Subject(s)
Axon Guidance , Growth Cones/metabolism , Nerve Growth Factors/metabolism , Spinal Cord/cytology , Tumor Suppressor Proteins/metabolism , Animals , Cell Adhesion , Female , Male , Mice , Mice, Knockout , Nerve Growth Factors/deficiency , Netrin-1 , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Tumor Suppressor Proteins/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...