Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Exp Eye Res ; 242: 109881, 2024 May.
Article in English | MEDLINE | ID: mdl-38554800

ABSTRACT

The retinal ganglion cells (RGCs) serve as the critical pathway for transmitting visual information from the retina to the brain, yet they can be dramatically impacted by diseases such as glaucoma. When investigating disease processes affecting RGCs in mouse models, accurately quantifying affected cells becomes essential. However, the use of pan RGC markers like RBPMS or THY1 presents challenges in accurate total cell counting. While Brn3a serves as a reliable RGC nuclear marker for automated counting, it fails to encompass all RGC subtypes in mice. To address this limitation and enable precise automated counting, our research endeavors to develop a method for labeling nuclei in all RGC subtypes. Investigating RGC subtypes labeled with the nuclear marker POU6F2 revealed that numerous RGCs unlabeled by Brn3a were, in fact, labeled with POU6F2. We hypothesize that using antibodies against both Brn3a and POU6F2 would label virtually all RGC nuclei in the mouse retina. Our experiments confirmed that staining retinas with both markers resulted in the labeling of all RGCs. Additionally, when using the cell body marker RBPMS known to label all mouse RGCs, all RBPMS-labeled cells also exhibited Brn3a or POU6F2 labeling. This combination of Brn3a and POU6F2 antibodies provides a pan-RGC nuclear stain, facilitating accurate automated counting by labeling cell nuclei in the retina.


Subject(s)
Cell Nucleus , Mice, Inbred C57BL , Retinal Ganglion Cells , Transcription Factor Brn-3A , Animals , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Mice , Cell Count , Cell Nucleus/metabolism , Transcription Factor Brn-3A/metabolism , Staining and Labeling/methods , Biomarkers/metabolism
2.
Development ; 147(17)2020 09 11.
Article in English | MEDLINE | ID: mdl-32917668

ABSTRACT

Despite the known importance of the transcription factors ATOH1, POU4F3 and GFI1 in hair cell development and regeneration, their downstream transcriptional cascades in the inner ear remain largely unknown. Here, we have used Gfi1cre;RiboTag mice to evaluate changes to the hair cell translatome in the absence of GFI1. We identify a systematic downregulation of hair cell differentiation genes, concomitant with robust upregulation of neuronal genes in the GFI1-deficient hair cells. This includes increased expression of neuronal-associated transcription factors (e.g. Pou4f1) as well as transcription factors that serve dual roles in hair cell and neuronal development (e.g. Neurod1, Atoh1 and Insm1). We further show that the upregulated genes are consistent with the NEUROD1 regulon and are normally expressed in hair cells prior to GFI1 onset. Additionally, minimal overlap of differentially expressed genes in auditory and vestibular hair cells suggests that GFI1 serves different roles in these systems. From these data, we propose a dual mechanism for GFI1 in promoting hair cell development, consisting of repression of neuronal-associated genes as well as activation of hair cell-specific genes required for normal functional maturation.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Hair Cells, Auditory, Inner/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/genetics , Hair Cells, Auditory, Inner/cytology , Mice , Mice, Transgenic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factor Brn-3A/genetics , Transcription Factor Brn-3A/metabolism , Transcription Factors/genetics
3.
Exp Eye Res ; 226: 109310, 2023 01.
Article in English | MEDLINE | ID: mdl-36400286

ABSTRACT

Immunofluorescence is used in numerous research areas including eye research to detect specific antigens in cells and tissues. One limitation is that fluorescent signal can fade, causing detection problems if data recording was not completed in a timely manner or if additional data acquisition is required. The ability to repeat immunostaining for the same antigen after initial fluorescence has faded may require time-consuming and potentially damaging steps to remove primary antibodies. Our studies assessed whether immunofluorescence could be reapplied to previously labeled retinal ganglion cells (RGCs). To examine whether immunostaining of Brn3a, a commonly used RGC marker, could be repeated in retinas with previously faded immunostaining, retinal whole mounts were labeled with anti-Brn3a primary antibodies and green fluorescent secondary antibodies, then allowed to fade over time. Faded retinas were restained with anti-Brn3a antibody followed by secondary antibody, or with secondary antibody alone. Results show restaining with anti-Brn3a primary antibody followed by Alexa-fluor green secondary antibody is effective for RGC detection. Repeat RGC labeling improved the clarity of staining compared with original staining prior to fading, with significant reduction in the percentage of blurry/out of focus fluorescent cells (6 vs 26%); whereas, repeat application of secondary antibody alone was not effective. Preflattening retinas under a coverslip prior to initial Brn3a staining also increased the clarity of staining, and facilitated significantly more accurate automated counting of RGCs. Findings suggest Brn3a antigen remains accessible for repeat immunofluorescence labeling after original staining fades. Staining retinas after flattening tissue may enhance the clarity of staining and accuracy of automated RGC counting. Repeat immunofluorescence staining, without the need to strip off prior bound antibodies, may be useful in other tissues as well and warrants future examination.


Subject(s)
Retina , Retinal Ganglion Cells , Retinal Ganglion Cells/metabolism , Fluorescent Antibody Technique , Staining and Labeling , Transcription Factor Brn-3A/metabolism
4.
Proc Natl Acad Sci U S A ; 117(34): 20741-20752, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32788346

ABSTRACT

Unresolved inflammation can lead to tissue fibrosis and impaired organ function. Macrophage-myofibroblast transition (MMT) is one newly identified mechanism by which ongoing chronic inflammation causes progressive fibrosis in different forms of kidney disease. However, the mechanisms underlying MMT are still largely unknown. Here, we discovered a brain-specific homeobox/POU domain protein Pou4f1 (Brn3a) as a specific regulator of MMT. Interestingly, we found that Pou4f1 is highly expressed by macrophages undergoing MMT in sites of fibrosis in human and experimental kidney disease, identified by coexpression of the myofibroblast marker, α-SMA. Unexpectedly, Pou4f1 expression peaked in the early stage in renal fibrogenesis in vivo and during MMT of bone marrow-derived macrophages (BMDMs) in vitro. Mechanistically, chromatin immunoprecipitation (ChIP) assay identified that Pou4f1 is a Smad3 target and the key downstream regulator of MMT, while microarray analysis defined a Pou4f1-dependent fibrogenic gene network for promoting TGF-ß1/Smad3-driven MMT in BMDMs at the transcriptional level. More importantly, using two mouse models of progressive renal interstitial fibrosis featuring the MMT process, we demonstrated that adoptive transfer of TGF-ß1-stimulated BMDMs restored both MMT and renal fibrosis in macrophage-depleted mice, which was prevented by silencing Pou4f1 in transferred BMDMs. These findings establish a role for Pou4f1 in MMT and renal fibrosis and suggest that Pou4f1 may be a therapeutic target for chronic kidney disease with progressive renal fibrosis.


Subject(s)
Smad3 Protein/metabolism , Transcription Factor Brn-3A/genetics , Transforming Growth Factor beta1/metabolism , Animals , Female , Fibrosis/physiopathology , Gene Regulatory Networks , Humans , Inflammation/pathology , Kidney/pathology , Kidney Diseases/genetics , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism , Signal Transduction/genetics , Transcription Factor Brn-3A/metabolism , Transcription Factor Brn-3A/physiology , Transforming Growth Factor beta/metabolism , Urinary Tract/metabolism
5.
Development ; 146(16)2019 08 21.
Article in English | MEDLINE | ID: mdl-31405994

ABSTRACT

Retinal ganglion cell (RGC) degeneration is a hallmark of glaucoma, the most prevalent cause of irreversible blindness. Thus, therapeutic strategies are needed to protect and replace these projection neurons. One innovative approach is to promote de novo genesis of RGCs via manipulation of endogenous cell sources. Here, we demonstrate that the pluripotency regulator gene Krüppel-like factor 4 (Klf4) is sufficient to change the potency of lineage-restricted retinal progenitor cells to generate RGCs in vivo Transcriptome analysis disclosed that the overexpression of Klf4 induces crucial regulators of RGC competence and specification, including Atoh7 and Eya2 In contrast, loss-of-function studies in mice and zebrafish demonstrated that Klf4 is not essential for generation or differentiation of RGCs during retinogenesis. Nevertheless, induced RGCs (iRGCs) generated upon Klf4 overexpression migrate to the proper layer and project axons aligned with endogenous fascicles that reach the optic nerve head. Notably, iRGCs survive for up to 30 days after in vivo generation. We identified Klf4 as a promising candidate for reprogramming retinal cells and regenerating RGCs in the retina.This article has an associated 'The people behind the papers' interview.


Subject(s)
Kruppel-Like Transcription Factors/physiology , Neurogenesis , Retinal Ganglion Cells/physiology , Animals , Cell Cycle , Female , Homeodomain Proteins/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Regeneration , Neural Stem Cells/physiology , Rats , Transcription Factor Brn-3A/metabolism , Transcription Factor Brn-3B/metabolism , Zebrafish , Zebrafish Proteins/physiology
6.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055045

ABSTRACT

The neural crest transcription factor BRN3A is essential for the proliferation and survival of melanoma cells. It is frequently expressed in melanoma but not in normal melanocytes or benign nevi. The mechanisms underlying the aberrant expression of BRN3A are unknown. Here, we investigated the epigenetic regulation of BRN3A in melanocytes and melanoma cell lines treated with DNA methyltransferase (DNMT), histone acetyltransferase (HAT), and histone deacetylase (HDAC) inhibitors. DNMT and HAT inhibition did not significantly alter BRN3A expression levels, whereas panHDAC inhibition by trichostatin A led to increased expression. Treatment with the isoform-specific HDAC inhibitor mocetinostat, but not with PCI-34051, also increased BRN3A expression levels, suggesting that class I HDACs HDAC1, HDAC2, and HDAC3, and class IV HDAC11, were involved in the regulation of BRN3A expression. Transient silencing of HDACs 1, 2, 3, and 11 by siRNAs revealed that, specifically, HDAC2 inhibition was able to increase BRN3A expression. ChIP-Seq analysis uncovered that HDAC2 inhibition specifically increased H3K27ac levels at a distal enhancer region of the BRN3A gene. Altogether, our data suggest that HDAC2 is a key epigenetic regulator of BRN3A in melanocytes and melanoma cells. These results highlight the importance of epigenetic mechanisms in regulating melanoma oncogenes.


Subject(s)
Gene Expression Regulation , Histone Deacetylase 2/metabolism , Melanocytes/metabolism , Melanoma/etiology , Melanoma/metabolism , Transcription Factor Brn-3A/genetics , Cell Line , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation/drug effects , Gene Silencing , Histone Deacetylase 2/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Melanocytes/pathology , Melanoma/pathology , Transcription Factor Brn-3A/metabolism
7.
Diabetologia ; 64(3): 693-706, 2021 03.
Article in English | MEDLINE | ID: mdl-33319325

ABSTRACT

AIMS/HYPOTHESIS: Diabetic retinopathy is characterised by retinal neurodegeneration and retinal vascular abnormalities, affecting one third of diabetic patients with disease duration of more than 10 years. Accumulated evidence suggests that serine racemase (SR) and D-serine are correlated with the pathogenesis of diabetic retinopathy and the deletion of the Srr gene reverses neurovascular pathologies in diabetic mice. Since D-serine content is balanced by SR synthesis and D-amino acid oxidase (DAAO) degradation, we examined the roles of DAAO in diabetic retinopathy and further explored relevant therapy. METHODS: Rats were used as a model of diabetes by i.p. injection of streptozotocin at the age of 2 months and blood glucose was monitored with a glucometer. Quantitative real-time PCR was used to examine Dao mRNA and western blotting to examine targeted proteins in the retinas. Bisulphite sequencing was used to examine the methylation of Dao mRNA promoter in the retinas. Intravitreal injection of DAAO-expressing adenovirus (AAV8-DAAO) was conducted one week before streptozotocin administration. Brain specific homeobox/POU domain protein 3a (Brn3a) immunofluorescence was conducted to indicate retinal ganglion cells at 3 months after virus injection. The permeability of the blood-retinal barrier was examined by Evans blue leakage from retinal capillaries. Periodic acid-Schiff staining and haematoxylin counterstaining were used to indicate retinal vasculature, which was further examined with double immunostaining at 7 months after virus injection. RESULTS: At the age of 12 months, DAAO mRNA and protein levels in retinas from diabetic animals were reduced to 66.2% and 70.4% of those from normal (control) animals, respectively. The Dao proximal promoter contained higher levels of methylation in diabetic than in normal retinas. Consistent with the observation, DNA methyltransferase 1 was increased in diabetic retinas. Injection of DAAO-expressing virus completely prevented the loss of retinal ganglion cells and the disruption of blood-retinal barrier in diabetic rats. Diabetic retinas contained retinal ganglion cells at a density of 54 ± 4/mm2, which was restored to 68 ± 9/mm2 by DAAO overexpression, similar to the levels in normal retinas. The ratio between the number of endothelial cells and pericytes in diabetic retinas was 6.06 ± 1.93/mm2, which was reduced to 3.42 ± 0.55/mm2 by DAAO overexpression; the number of acellular capillaries in diabetic retinas was 10 ± 5/mm2, which was restored to 6 ± 2/mm2 by DAAO overexpression, similar to the levels in normal retinas. Injection of the DAAO-expressing virus increased the expression of occludin and reduced gliosis, which were examined to probe the mechanism by which the disrupted blood-retinal barrier in diabetic rats was rescued and retinal neurodegeneration was prevented. CONCLUSIONS/INTERPRETATION: Altogether, overexpression of DAAO before the onset of diabetes protects against neurovascular abnormalities in retinas from diabetic rats, which suggests a novel strategy for preventing diabetic retinopathy. Graphical abstract.


Subject(s)
Blood-Retinal Barrier/enzymology , D-Amino-Acid Oxidase/biosynthesis , Diabetic Retinopathy/prevention & control , Retinal Ganglion Cells/enzymology , Animals , Blood-Retinal Barrier/pathology , Capillary Permeability , D-Amino-Acid Oxidase/genetics , DNA Methylation , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/enzymology , Diabetic Retinopathy/enzymology , Diabetic Retinopathy/etiology , Diabetic Retinopathy/pathology , Enzyme Induction , Male , Nerve Degeneration , Promoter Regions, Genetic , Rats, Sprague-Dawley , Retinal Ganglion Cells/pathology , Transcription Factor Brn-3A/genetics , Transcription Factor Brn-3A/metabolism
8.
Exp Eye Res ; 210: 108694, 2021 09.
Article in English | MEDLINE | ID: mdl-34245756

ABSTRACT

PURPOSE: To analyze responses of different RGC populations to left intraorbital optic nerve transection (IONT) and intraperitoneal (i.p.) treatment with 7,8-Dihydroxyflavone (DHF), a potent selective TrkB agonist. METHODS: Adult albino Sprague-Dawley rats received, following IONT, daily i.p. injections of vehicle (1%DMSO in 0.9%NaCl) or DHF. Group-1 (n = 58) assessed at 7days (d) the optimal DHF amount (1-25 mg/kg). Group-2, using freshly dissected naïve or treated retinas (n = 28), investigated if DHF treatment was associated with TrkB activation using Western-blotting at 1, 3 or 7d. Group-3 (n = 98) explored persistence of protection and was analyzed at survival intervals from 7 to 60d after IONT. Groups 2-3 received daily i.p. vehicle or DHF (5 mg/kg). Retinal wholemounts were immunolabelled for Brn3a and melanopsin to identify Brn3a+RGCs and m+RGCs, respectively. RESULTS: Optimal neuroprotection was achieved with 5 mg/kg DHF and resulted in TrkB phosphorylation. The percentage of surviving Brn3a+RGCs in vehicle treated rats was 60, 28, 18, 13, 12 or 8% of the original value at 7, 10, 14, 21, 30 or 60d, respectively, while in DHF treated retinas was 94, 70, 64, 17, 10 or 9% at the same time intervals. The percentages of m+RGCs diminished by 7d-13%, and recovered by 14d-38% in vehicle-treated and to 48% in DHF-treated retinas, without further variations. CONCLUSIONS: DHF neuroprotects Brn3a + RGCs and m + RGCs; its protective effects for Brn3a+RGCs are maximal at 7 days but still significant at 21d, whereas for m+RGCs neuroprotection was significant at 14d and permanent.


Subject(s)
Flavones/administration & dosage , Neuroprotective Agents/administration & dosage , Receptor, trkB/metabolism , Retinal Ganglion Cells/drug effects , Animals , Axotomy , Blotting, Western , Cell Survival/physiology , Female , Immunohistochemistry , Injections, Intraperitoneal , Neuroprotection , Optic Nerve/physiopathology , Optic Nerve/surgery , Phosphorylation , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Rod Opsins/metabolism , Transcription Factor Brn-3A/metabolism
9.
Exp Eye Res ; 213: 108853, 2021 12.
Article in English | MEDLINE | ID: mdl-34800481

ABSTRACT

PURPOSE: The roles of vascular dysfunction and chronic stress have been extensively discussed in the pathophysiology of glaucoma. Our aim was to test whether chronic stress causes retinal vascular dysfunction and therewith induces retinal ganglion cells (RGCs) loss. METHODS: Twelve mice underwent chronic social defeat (CSD) stress, while 12 mice received control treatment only. Intraocular pressure (IOP) was measured with a rebound tonometer. Blood plasma corticosterone concentration and adrenal gland weight were used to assess stress levels. Brn-3a staining in retinas and PPD staining in optic nerve cross sections were conducted to assess the survival of RGCs and axons respectively. The ET-1 and α-SMA levels were determined in retina. Retinal vascular autoregulation, functional response to various vasoactive agents and vascular mechanics were measured using video microscopy. RESULTS: No significant difference in IOP levels was observed during and after CSD between CSD mice and controls. CSD stress caused hypercortisolemia 2 days post-CSD. However, increased corticosterone levels went back to normal 8 months after CSD. CSD-exposed mice developed adrenal hyperplasia 3 days post-CSD, which was normalized by 8 months. RGC and axon survival were similar between CSD mice and controls. However, CSD stress caused irreversible, impaired autoregulation and vascular dysfunction of retinal arterioles in CSD mice. In addition, impaired maximal dilator capacity of retinal arterioles was observed 8 months post-CSD rather than 3 days post-CSD. Remarkably, ET-1 levels were increased 3 days post-CSD while α-SMA levels were decreased 8 months post-CSD. CONCLUSIONS: We found that CSD stress does not cause IOP elevation, nor loss of RGCs and their axons. However, it strikingly causes irreversible impaired autoregulation and endothelial function in murine retinal arterioles. In addition, CSD changed vascular mechanics on a long-term basis. Increased ET-1 levels and loss of pericytes in retina vessels may involve in this process.


Subject(s)
Retinal Artery/physiopathology , Retinal Diseases/physiopathology , Retinal Ganglion Cells/pathology , Social Defeat , Stress, Psychological/physiopathology , Actins/metabolism , Adrenal Hyperplasia, Congenital/physiopathology , Animals , Cell Survival , Chronic Disease , Corticosterone/blood , Disease Models, Animal , Disorder of Sex Development, 46,XY/physiopathology , Endothelin-1/metabolism , Intraocular Pressure/physiology , Male , Mice , Mice, Inbred C57BL , Ocular Hypertension/physiopathology , Optic Nerve/physiopathology , Retinal Artery/metabolism , Retinal Diseases/metabolism , Retinal Ganglion Cells/metabolism , Stress, Psychological/metabolism , Tonometry, Ocular , Transcription Factor Brn-3A/metabolism , Video Recording
10.
Exp Cell Res ; 396(1): 112159, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32652081

ABSTRACT

Traumatic optic neuropathy is a common clinical problem. Damage to the optic nerve leads to shear stress and triggers secondary swelling within the optic canal. The study aims to explore the role of the inflammatory response following optic nerve injury (ONI) in toll-like receptor-9 knockout mice (TLR-9-/-) compared to wild-type mice (WT). At first, TLR-9-/- and WT mice were subjected to ONI. We then found that ONI significantly up-regulated TLR-9 expression levels in retinal tissues of WT mice. The retinal degeneration after ONI was alleviated in TLR-9-/- mice, as evidenced by the increased number of retinal ganglion cells (RGCs) and thickness of inner retinal layer (IRL). TUNEL staining and immunofluorescence staining of BRN3A indicated that TLR-9 knockout effectively improved the survival of RGCs. ONI-enhanced expression of Iba-1 and TMEM119 was markedly reduced in TLR-9-/- mice, indicating the suppression of microglial activation. Moreover, production of pro-inflammatory regulators, including inducible nitric oxide synthase (iNOS), macrophage chemo-attractant protein (MCP)-1, cyclooxygenase-2 (COX-2), interleukin (IL)-1ß, IL-18 and tumor necrosis factor-α (TNF-α), was significantly decreased in TLR-9-/- mice following ONI. TLR-9 knockout-attenuated inflammation was mainly through repressing myeloid differentiation factor 88 (MyD88) and IL-1 receptor-associated kinase 4 (IRAK4). Furthermore, ONI greatly up-regulated the protein expression levels of phosphorylated (p)-IKKα, p-IκBα and p-nuclear factor (NF)-κB, whereas being repressed in TLR-9-/- mice. The effects of TLR-9 on ONI were verified in lipopolysaccharide (LPS)-stimulated retinal microglial cells transfected with small interfering RNA TLR-9 (siTLR-9). As expected, promoting TLR-9 with its agonist markedly restored inflammation in TLR-9 knockdown cells stimulated by LPS. Therefore, all findings above suggested that suppressing TLR-9 showed neuroprotective effects against ONI through reducing inflammatory response, and TILR-9 might be a promising therapeutic target to develop effective strategies for the treatment of optic neuropathies.


Subject(s)
Microglia/metabolism , Myeloid Differentiation Factor 88/genetics , Optic Nerve Injuries/genetics , Optic Nerve/metabolism , Retinal Ganglion Cells/metabolism , Toll-Like Receptor 9/genetics , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Count , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Gene Expression Regulation , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Inflammation , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/pathology , Myeloid Differentiation Factor 88/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Optic Nerve/pathology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/pathology , Signal Transduction , Toll-Like Receptor 9/deficiency , Transcription Factor Brn-3A/genetics , Transcription Factor Brn-3A/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
11.
J Neurosci ; 39(27): 5284-5298, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31085606

ABSTRACT

Acoustic signals are relayed from the ear to the brain via spiral ganglion neurons (SGNs) that receive auditory information from the cochlear inner hair cells (IHCs) and transmit that information to the cochlear nucleus of the brainstem. Physiologically distinct classes of SGNs have been characterized by their spontaneous firing rate and responses to sound and those physiological distinctions are thought to correspond to stereotyped synaptic positions on the IHC. More recently, single-cell profiling has identified multiple groups of SGNs based on transcriptional profiling; however, correlations between any of these groups and distinct neuronal physiology have not been determined. In this study, we show that expression of the POU (Pit-Oct-Unc) transcription factor Pou4f1 in type I SGNs in mice of both sexes correlates with a synaptic location on the modiolar side of IHCs. Conditional deletion of Pou4f1 in SGNs beginning in mice at embryonic day 13 rescues the early path-finding and apoptotic phenotypes reported for germline deletion of Pou4f1, resulting in a phenotypically normal development of SGN patterning. However, conditional deletion of Pou4f1 in SGNs alters the activation of Ca2+ channels in IHCs primarily by increasing their voltage sensitivity. Moreover, the modiolar to pillar gradient of active zone Ca2+ influx strength is eliminated. These results demonstrate that a subset of modiolar-targeted SGNs retain expression of Pou4f1 beyond the onset of hearing and suggest that this transcription factor plays an instructive role in presynaptic Ca2+ signaling in IHCs.SIGNIFICANCE STATEMENT Physiologically distinct classes of type I spiral ganglion neurons (SGNs) are necessary to encode sound intensities spanning the audible range. Although anatomical studies have demonstrated structural correlates for some physiologically defined classes of type I SGNs, an understanding of the molecular pathways that specify each type is only now emerging. Here, we demonstrate that expression of the transcription factor Pou4f1 corresponds to a distinct subgroup of type I SGNs that synapse on the modiolar side of inner hair cells. The conditional deletion of Pou4f1 after SGN formation does not disrupt ganglion size or morphology, change the distribution of IHC synaptic locations, or affect the creation of synapses, but it does influence the voltage dependence and strength of Ca2+ influx at presynaptic active zones in inner hair cells.


Subject(s)
Calcium Signaling , Hearing/physiology , Neurons/metabolism , Presynaptic Terminals/metabolism , Spiral Ganglion/metabolism , Transcription Factor Brn-3A/metabolism , Animals , Evoked Potentials, Auditory, Brain Stem , Female , Hair Cells, Auditory, Inner , Male , Mice, Inbred C57BL , Spiral Ganglion/cytology
12.
Biochem Biophys Res Commun ; 533(3): 533-539, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32988584

ABSTRACT

Over-expression of the human epidermal growth factor receptor-2 (HER2) is related to aggressive tumors and poor prognosis in breast cancer. Trastuzumab (TRA) resistance leads to tumor recurrence and metastasis, resulting in poor prognosis in HER2-positive breast cancer. POU Class 4 Homeobox 1 (POU4F1) is a member of the POU domain family transcription factors, and has a key role in regulating cancers. However, its effects on TRA-resistant HER2-positive breast cancer are still vague. In the present study, we found that POU4F1 expression was dramatically increased in clinical breast cancer specimens with TRA resistance. Higher POU4F1 was also detected in HER2-positive breast cancer cells with TRA resistance than that of the parental ones. Poor prognosis was detected in breast cancer patients with high POU4F1 expression. Under TRA treatment, POU4F1 knockdown significantly reduced the proliferative capacity of HER2-positive breast cancer cells with TRA resistance. POU4F1 silence also sensitized resistant HER-positive breast cancer cells to TRA treatment in vivo using a xenograft mouse model, along with the markedly reduced tumor growth rate and tumor weight. Moreover, we found that POU4F1 deletion greatly decreased the activation of mitogen-activated or extracellular signal-regulated protein kinase kinases 1 and 2 (MEK1/2) and extracellular-regulated kinase 1/2 (ERK1/2) signaling pathways in breast cancer cells with TRA resistance. Migration and invasion were also effectively hindered by POU4F1 knockdown in TRA-resistant HER2-positive breast cancer cells. Notably, we found that POU4F1 deletion-improved chemosensitivity of HER2-positive breast cancer cells with drug-resistance to TRA treatment was closely associated with the blockage of ERK1/2 signaling. Collectively, our findings reported a critical role of POU4F1 in regulating TRA resistance, and demonstrated the underlying molecular mechanisms in HER2-positive breast cancer. Thus, POU4F1 may be a promising prognostic and therapeutic target to develop effective treatment for overcoming TRA resistance.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/metabolism , MAP Kinase Signaling System , Transcription Factor Brn-3A/metabolism , Trastuzumab/therapeutic use , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Female , Humans , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Receptor, ErbB-2/analysis , Transcription Factor Brn-3A/genetics , Transcription Factor Brn-3A/physiology
13.
Exp Eye Res ; 188: 107784, 2019 11.
Article in English | MEDLINE | ID: mdl-31476280

ABSTRACT

Ischaemia/reperfusion contributes to the pathophysiological process of many retinal diseases. Previous studies have shown that retinal ischaemia/reperfusion mainly results in neuronal degeneration, including thinning of the retina, retinal ganglion cell death and reductions in electroretinography. A high-salt diet contributes to the inflammatory response and tissue hypoperfusion and may be associated with ischaemia/reperfusion injury. In the present study, we investigated the influence of a high-salt diet on retinal ischaemia/reperfusion injury and explored the potential mechanism in a rat model. The results revealed that the high-salt diet aggravated ischaemia/reperfusion-induced thinning of the retina. A TUNEL assay and Brn-3a staining revealed substantially more severe cell death and loss of retinal ganglion cells, and electroretinography confirmed worse retinal function in the ischaemia/reperfusion eyes of rats fed the high-salt diet. These effects may be associated with upregulation of Caspase-3, Bax, Interleukin-1ß and Interleukin-6 and decreased expression of nitric oxide. In summary, a high-salt diet aggravates ischaemia/reperfusion-induced retinal neuronal impairment by activating pro-apoptotic and pro-inflammatory signalling pathways and inhibiting vasodilation.


Subject(s)
Reperfusion Injury/metabolism , Retinal Diseases/metabolism , Sodium Chloride, Dietary/administration & dosage , Animals , Blotting, Western , Caspase 3/metabolism , Cell Count , Electroretinography , Enzyme-Linked Immunosorbent Assay , In Situ Nick-End Labeling , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Nitric Oxide/metabolism , Rats , Rats, Wistar , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Retinal Diseases/pathology , Retinal Diseases/physiopathology , Retinal Ganglion Cells/pathology , Transcription Factor Brn-3A/metabolism , bcl-2-Associated X Protein/metabolism
14.
Exp Eye Res ; 188: 107781, 2019 11.
Article in English | MEDLINE | ID: mdl-31473259

ABSTRACT

To study the effect of taurine depletion induced by ß-alanine supplementation in the retinal nerve fiber layer (RNFL), and retinal ganglion cell (RGC) survival and axonal transport. Albino Sprague-Dawley rats were divided into two groups: one group received ß-alanine supplementation (3%) in the drinking water during 2 months to induce taurine depletion, and the other group received regular water. After one month, half of the rats from each group were exposed to light. Retinas were analyzed in-vivo using Spectral-Domain Optical Coherence Tomography (SD-OCT). Prior to processing, RGCs were retrogradely traced with fluorogold (FG) applied to both superior colliculi, to assess the state of their retrograde axonal transport. Retinas were dissected as wholemounts, surviving RGCs were immunoidentified with Brn3a, and the RNFL with phosphorylated high-molecular-weight subunit of the neurofilament triplet (pNFH) antibodies. ß-alanine supplementation decreases significantly taurine plasma levels and causes a significant reduction of the RNFL thickness that is increased after light exposure. An abnormal pNFH immunoreactivity in some RGC bodies, their proximal dendrites and axons, and a further diminution of the mean number of FG-traced RGCs compared with Brn3a+RGCs, indicate that their retrograde axonal transport is affected. In conclusion, taurine depletion causes RGC loss and axonal transport impairment. Finally, our results suggest that care should be taken when ingesting ß-alanine supplements due to the limited understanding of their potential adverse effects.


Subject(s)
Axonal Transport/drug effects , Light/adverse effects , Nerve Fibers/drug effects , Retinal Degeneration/etiology , Retinal Ganglion Cells/drug effects , Taurine/deficiency , beta-Alanine/toxicity , Animals , Nerve Fibers/metabolism , Nerve Fibers/pathology , Neurofilament Proteins/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Taurine/blood , Tomography, Optical Coherence , Transcription Factor Brn-3A/metabolism
15.
Int J Mol Sci ; 20(13)2019 Jun 29.
Article in English | MEDLINE | ID: mdl-31261943

ABSTRACT

BACKGROUND: To induce a moderate chronic ocular hypertension (OHT) by injecting polidocanol, a foamed sclerosant drug, in the aqueous humor outflow pathway. METHODS: Intraocular pressure (IOP) was monitored for up to 6 months. Pattern and full-field electroretinogram (PERG and ERG) were recorded and retinal ganglion cells (RGC) and retinal nerve fiber layer (RNFL) thickness were assessed in vivo with optical coherence tomography (OCT) and ex vivo using Brn3a immunohistochemistry. RESULTS: In the first 3 weeks post-injection, a significant IOP elevation was observed in the treated eyes (18.47 ± 3.36 mmHg) when compared with the control fellow eyes (12.52 ± 2.84 mmHg) (p < 0.05). At 8 weeks, 65% (11/17) of intervention eyes had developed an IOP increase >25% over the baseline. PERG responses were seen to be significantly reduced in the hypertensive eyes (2.25 ± 0.24 µV) compared to control eyes (1.44 ± 0.19 µV) (p < 0.01) at week 3, whereas the ERG components (photoreceptor a-wave and bipolar cell b-wave) remained unaltered. By week 24, RNFL thinning and cell loss in the ganglion cell layer was first detected (2/13, 15.3%) as assessed by OCT and light microscopy. CONCLUSIONS: This novel OHT rat model, with moderate levels of chronically elevated IOP, and abnormal PERG shows selective functional impairment of RGC.


Subject(s)
Disease Models, Animal , Glaucoma/etiology , Polidocanol/toxicity , Sclerosing Solutions/toxicity , Animals , Glaucoma/metabolism , Glaucoma/pathology , Injections, Intraocular , Intraocular Pressure , Male , Rats , Rats, Wistar , Transcription Factor Brn-3A/metabolism
16.
Int J Mol Sci ; 20(12)2019 Jun 20.
Article in English | MEDLINE | ID: mdl-31226772

ABSTRACT

We studied short- and long-term effects of intravitreal injection of N-methyl-d-aspartate (NMDA) on melanopsin-containing (m+) and non-melanopsin-containing (Brn3a+) retinal ganglion cells (RGCs). In adult SD-rats, the left eye received a single intravitreal injection of 5µL of 100nM NMDA. At 3 and 15 months, retinal thickness was measured in vivo using Spectral Domain-Optical Coherence Tomography (SD-OCT). Ex vivo analyses were done at 3, 7, or 14 days or 15 months after damage. Whole-mounted retinas were immunolabelled for brain-specific homeobox/POU domain protein 3A (Brn3a) and melanopsin (m), the total number of Brn3a+RGCs and m+RGCs were quantified, and their topography represented. In control retinas, the mean total numbers of Brn3a+RGCs and m+RGCs were 78,903 ± 3572 and 2358 ± 144 (mean ± SD; n = 10), respectively. In the NMDA injected retinas, Brn3a+RGCs numbers diminished to 49%, 28%, 24%, and 19%, at 3, 7, 14 days, and 15 months, respectively. There was no further loss between 7 days and 15 months. The number of immunoidentified m+RGCs decreased significantly at 3 days, recovered between 3 and 7 days, and were back to normal thereafter. OCT measurements revealed a significant thinning of the left retinas at 3 and 15 months. Intravitreal injections of NMDA induced within a week a rapid loss of 72% of Brn3a+RGCs, a transient downregulation of melanopsin expression (but not m+RGC death), and a thinning of the inner retinal layers.


Subject(s)
N-Methylaspartate/toxicity , Retinal Ganglion Cells/drug effects , Rod Opsins/metabolism , Animals , Cell Count , Female , Intravitreal Injections , N-Methylaspartate/administration & dosage , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Rod Opsins/analysis , Transcription Factor Brn-3A/analysis , Transcription Factor Brn-3A/metabolism
17.
J Neurosci ; 37(8): 2125-2136, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28115479

ABSTRACT

O-GlcNAc transferase (OGT) regulates a wide range of cellular processes through the addition of the O-GlcNAc sugar moiety to thousands of protein substrates. Because nutrient availability affects the activity of OGT, its role has been broadly studied in metabolic tissues. OGT is enriched in the nervous system, but little is known about its importance in basic neuronal processes in vivo Here, we show that OGT is essential for sensory neuron survival and maintenance in mice. Sensory neuron-specific knock-out of OGT results in behavioral hyposensitivity to thermal and mechanical stimuli accompanied by decreased epidermal innervation and cell-body loss in the dorsal root ganglia. These effects are observed early in postnatal development and progress as animals age. Cultured sensory neurons lacking OGT also exhibit decreased axonal outgrowth. The effects on neuronal health in vivo are not solely due to disruption of developmental processes, because inducing OGT knock-out in the sensory neurons of adult mice results in a similar decrease in nerve fiber endings and cell bodies. Significant nerve-ending loss occurs before a decrease in cell bodies; this phenotype is indicative of axonal dieback that progresses to neuronal death. Our findings demonstrate that OGT is important in regulating axonal maintenance in the periphery and the overall health and survival of sensory neurons.SIGNIFICANCE STATEMENT We show the importance of O-GlcNAc transferase (OGT) for sensory neuron health and survival in vivo This study is the first to find that loss of OGT results in neuronal cell death. Moreover, it suggests that aberrant O-GlcNAc signaling can contribute to the development of neuropathy. The sensory neurons lie outside of the blood-brain barrier and therefore, compared to central neurons, may have a greater need for mechanisms of metabolic sensing and compensation. Peripheral sensory neurons in particular are subject to degeneration in diabetes. Our findings provide a foundation for understanding the role of OGT under normal physiological conditions in the peripheral nervous system. This knowledge will be important for gaining greater insight into such disease states as diabetic neuropathy.


Subject(s)
N-Acetylglucosaminyltransferases/metabolism , Sensory Receptor Cells/physiology , Animals , Body Weight/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Ganglia, Spinal/cytology , Gene Expression Regulation/genetics , Glucose Tolerance Test , Locomotion/genetics , Male , Mental Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Strength/genetics , N-Acetylglucosaminyltransferases/deficiency , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Neuronal Plasticity/genetics , Thermosensing/genetics , Transcription Factor Brn-3A/genetics , Transcription Factor Brn-3A/metabolism
18.
Glia ; 66(11): 2366-2384, 2018 11.
Article in English | MEDLINE | ID: mdl-30375063

ABSTRACT

Excitotoxicity is well known in the neuronal death in the brain and is also linked to neuronal damages in the retina. Recent accumulating evidence show that microglia greatly affect excitotoxicity in the brain, but their roles in retina have received only limited attention. Here, we report that retinal excitotoxicity is mediated by microglia. To this end, we employed three discrete methods, that is, pharmacological inhibition of microglia by minocycline, pharmacological ablation by an antagonist for colony stimulating factor 1 receptor (PLX5622), and genetic ablation of microglia using Iba1-tTA::DTAtetO/tetO mice. Intravitreal injection of NMDA increased the number of apoptotic retinal ganglion cells (RGCs) followed by reduction in the number of RGCs. Although microglia did not respond to NMDA directly, they became reactive earlier than RGC damages. Inhibition or ablation of microglia protected RGCs against NMDA. We found up-regulation of proinflammatory cytokine genes including Il1b, Il6 and Tnfa, among which Tnfa was selectively blocked by minocycline. PLX5622 also suppressed Tnfa expression. Tumor necrosis factor α (TNFα) signals were restricted in microglia at very early followed by spreading into other cell types. TNFα up-regulation in microglia and other cells were significantly attenuated by minocycline and PLX5622, suggesting a central role of microglia for TNFα induction. Both inhibition of TNFα and knockdown of TNF receptor type 1 by siRNA protected RGCs against NMDA. Taken together, our data demonstrate that a phenotypic change of microglia into a neurotoxic one is a critical event for the NMDA-induced degeneration of RGCs, suggesting an importance of non-cell-autonomous mechanism in the retinal neuronal excitotoxicity.


Subject(s)
Cell Death/physiology , Cytokines/metabolism , Microglia/physiology , Retinal Ganglion Cells/physiology , Aminopyridines/pharmacology , Animals , Animals, Newborn , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Death/drug effects , Cells, Cultured , Cytokines/genetics , Excitatory Amino Acid Agonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/ultrastructure , N-Methylaspartate/pharmacology , Nerve Degeneration/chemically induced , Optic Nerve Injuries/chemically induced , Organic Chemicals/pharmacology , Pyrroles/pharmacology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/ultrastructure , Signal Transduction/drug effects , Transcription Factor Brn-3A/genetics , Transcription Factor Brn-3A/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Cell Physiol Biochem ; 50(6): 2314-2328, 2018.
Article in English | MEDLINE | ID: mdl-30423551

ABSTRACT

BACKGROUND/AIMS: Chronic Lymphocytic leukemia (CLL) is characterized by accumulation of cells in the G0/G1 phase of the cell cycle and resistance to apoptosis due to gene mutation or abnormal gene expression. In our previous study, constitutively photomorphogenic 1 (COP1) was shown to be upregulated in Binet C-phase CLL patients. Based on the negative regulation of COP1 in the repair of DNA damage, we further studied the function of COP1 in CLL cell apoptosis induced by fludarabine in vitro and in vivo. METHODS: We analyzed the sensitivity of primary CLL cells to the fludarabine by CCK-8, and detected the expression of p53 in cells after drug treatment by western blot. Next, we constructed COP1 overexrpessing CLL cell line HG3, and analyzed the effect of COP1 overexpression on the HG3 cell's apoptosis, and HG3 transplant mice survival with drug treatment. RESULTS: Here, we found that primary CLL cells with high expression of COP1 showed low sensitivity to the drug and presented delayed enrichment of p53 protein than cells with low COP1 expressed. COP1 overexpression reduced HG3 cell sensitivity to the fludarabine treatment and inhibited cell apoptosis, and also retarded itself via autoubiquitination. The further study showed that COP1 promoted ubiquitin-dependent p53 degradation, which further disrupts the formation of the p53-Brn-3a complex and activation of Bcl-2 transcription. Moreover, mice engrafted with cells overexpressing COP1 showed a shortened survival, increased tumor cells burden in spleen and bone marrow (BM), and reduced tumor cell apoptosis even when fludarabine combined cyclophosphamide (F+C) therapy was administered. CONCLUSION: This study demonstrates that COP1 contributes to drug resistance of CLL cells to the fludarabine treatment in vitro and in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Vidarabine/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Bone Marrow/pathology , Cell Line, Tumor , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Mice , Mice, Inbred NOD , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Spleen/pathology , Survival Rate , Transcription Factor Brn-3A/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Vidarabine/pharmacology , Vidarabine/therapeutic use
20.
Mol Vis ; 24: 574-586, 2018.
Article in English | MEDLINE | ID: mdl-30174387

ABSTRACT

Purpose: To investigate the relationship between intraocular pressure (IOP) and GABA receptors within the arcuate nucleus (ARC). Methods: In the chronic high IOP rat model, ibotenic acid (IBO) was injected to induce impairment of the ARC, and IOP was measured at the 0, 1, 2, 3, and 4 week time points with a Tono-Pen. To assess the expression of GABA-A/B receptors within the ARC under persistent high IOP, we performed immunofluorescence (IF) and immunohistochemical (IHC) staining at 2 weeks and 4 weeks. Furthermore, we treated the ARC with GABA-A/B receptor antagonists separately, and IOP was evaluated, as well as retinal ganglion cell apoptosis in the chronic high IOP rat model. In the following induced high IOP animal model, the expression of GABA-A/B receptors within the ARC was evaluated in DBA/2J mice which developed progressive eye abnormalities spontaneously that closely mimic human hereditary glaucoma. Results: Compared with the control group, statistically significant downregulation of IOP was noted due to the IBO injection into the ARC at the 2, 3, and 4 week time points (p<0.05). Persistent high IOP elicited increased expression of the GABA-A/B receptors in the ARC compared with the control group (p<0.01). In addition, treatment with GABA-A/B receptor antagonists separately caused a decrease in the IOP, along with reduced retinal ganglion cell apoptosis (p<0.01). In the DBA/2J mice, the expression of the GABA receptors was statistically significantly increased (p<0.01). Conclusions: GABA-A/B receptors in the ARC may be involved in regulation of IOP, and pathologically high IOP affects the expression of GABA-A/B receptors in the ARC.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Disease Models, Animal , Intraocular Pressure/physiology , Ocular Hypertension/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-B/metabolism , Animals , Apoptosis , Arcuate Nucleus of Hypothalamus/drug effects , Excitatory Amino Acid Agonists/pharmacology , Fluorescent Antibody Technique, Indirect , GABA-A Receptor Antagonists/pharmacology , GABA-B Receptor Antagonists/pharmacology , Ibotenic Acid/pharmacology , Immunoenzyme Techniques , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/pathology , Tonometry, Ocular , Transcription Factor Brn-3A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL