1
|
Bailey JK, Ma D, Clegg DO. Initial Characterization of WDR5B Reveals a Role in the Proliferation of Retinal Pigment Epithelial Cells. Cells 2024; 13:1189. [PMID: 39056772 PMCID: PMC11275010 DOI: 10.3390/cells13141189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
The chromatin-associated protein WDR5 has been widely studied due to its role in histone modification and its potential as a pharmacological target for the treatment of cancer. In humans, the protein with highest sequence homology to WDR5 is encoded by the retrogene WDR5B, which remains unexplored. Here, we used CRISPR-Cas9 genome editing to generate WDR5B knockout and WDR5B-FLAG knock-in cell lines for further characterization. In contrast to WDR5, WDR5B exhibits low expression in pluripotent cells and is upregulated upon neural differentiation. Loss or shRNA depletion of WDR5B impairs cell growth and increases the fraction of non-viable cells in proliferating retinal pigment epithelial (RPE) cultures. CUT&RUN chromatin profiling in RPE and neural progenitors indicates minimal WDR5B enrichment at established WDR5 binding sites. These results suggest that WDR5 and WDR5B exhibit several divergent biological properties despite sharing a high degree of sequence homology.
Collapse
Affiliation(s)
- Jeffrey K. Bailey
- Department of Molecular, Cellular and Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
- Center for Stem Cell Biology and Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Dzwokai Ma
- Department of Molecular, Cellular and Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
| | - Dennis O. Clegg
- Department of Molecular, Cellular and Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
- Center for Stem Cell Biology and Engineering, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
2
|
Jentzsch MC, Tsang SH, Koch SF. A New Preclinical Model of Retinitis Pigmentosa Due to Pde6g Deficiency. OPHTHALMOLOGY SCIENCE 2023; 3:100332. [PMID: 37363133 PMCID: PMC10285708 DOI: 10.1016/j.xops.2023.100332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 06/28/2023]
Abstract
Purpose Retinitis pigmentosa (RP) is the most common cause of inherited blindness, with onset occurring as early as 4 years of age in certain rare but severe forms caused by mutations in the gamma subunit of phosphodiesterase 6 (PDE6). Studies in humans and mice have shown that RP pathology begins with progressive photoreceptor death, which then drives changes in downstream neurons, neighboring retinal pigment epithelium (RPE), and vasculature. Here, we present the first detailed analysis of RP disease progression in Pde6g-deficient mice. Design Experimental study of an RP mouse model. Subjects We studied Pde6g-/- and Pde6g+/- mice at the age of 7, 16, 30, 44, and 56 days with n = 2 to 5 per group and time point. Methods Photoreceptor degeneration and retinal remodeling were analyzed in retinal sections by immunofluorescence. Retinal blood vessel degradation was analyzed in flat-mounted retinas immunolabeled for isolectin GS-IB4. Protein expression was measured by immunoblot. Acellular capillaries were assessed in trypsin-digested and hematoxylin-eosin-stained retinas at postnatal day (P) 44. Retinal pigment epithelium cells were delineated in flat-mounted RPE-choroid-sclera by immunolabeling for the cell-adhesion protein β-catenin. Main Outcome Measures Immunofluorescence and morphometry (quantitative analysis of outer nuclear layer, dendrite area, vessel area, acellular vessels, RPE cell size, number of nuclei per RPE cell, RPE cell eccentricity, and RPE cell solidity). Results This novel RP model exhibits early onset and rapid rod degeneration, with the vast majority gone by P16. This pathology leads to retinal remodeling, including changes of inner retinal neurons, early activation of glia cells, degradation of retinal vasculature, and structural abnormalities of the RPE. Conclusions The pathology in our Pde6g-/- mouse model precisely mirrors human RP progression. The results demonstrate the significant role of the gamma subunit in maintaining phosphodiesterase activity and provide new insights into the disease progression due to Pde6g deficiency. Financial Disclosures Proprietary or commercial disclosure may be found after the references.
Collapse
Affiliation(s)
- Michelle Carmen Jentzsch
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stephen H. Tsang
- Jonas Children’s Vision Care, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, New York
| | - Susanne Friederike Koch
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
3
|
Zhou M, Zhao Y, Weber SR, Gates C, Carruthers NJ, Chen H, Liu X, Wang H, Ford M, Swulius MT, Barber AJ, Grillo SL, Sundstrom JM. Extracellular vesicles from retinal pigment epithelial cells expressing R345W-Fibulin-3 induce epithelial-mesenchymal transition in recipient cells. J Extracell Vesicles 2023; 12:e12373. [PMID: 37855063 PMCID: PMC10585439 DOI: 10.1002/jev2.12373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/06/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
We have shown previously that expression of R345W-Fibulin-3 induces epithelial-mesenchymal transition (EMT) in retinal pigment epithelial (RPE) cells. The purpose of the current study was to determine if extracellular vesicles (EVs) derived from RPE cells expressing R345W-Fibulin-3 mutation are sufficient to induce EMT in recipient cells. ARPE-19 cells were infected with luciferase-tagged wild-type (WT)- Fibulin-3 or luciferase-tagged R345W-Fibulin-3 (R345W) using lentiviruses. EVs were isolated from the media by ultracentrifugation or density gradient ultracentrifugation. Transmission electron microscopy and cryogenic electron microscopy were performed to study the morphology of the EVs. The size distribution of EVs were determined by nanoparticle tracking analysis (NTA). EV cargo was analysed using LC-MS/MS based proteomics. EV-associated transforming growth factor beta 1 (TGFβ1) protein was measured by enzyme-linked immunosorbent assay. The capacity of EVs to stimulate RPE migration was evaluated by treating recipient cells with WT- or R345W-EVs. The role of EV-bound TGFβ was determined by pre-incubation of EVs with a pan-TGFβ blocking antibody or IgG control. EM imaging revealed spherical vesicles with two subpopulations of EVs: a group with diameters around 30 nm and a group with diameters over 100 nm, confirmed by NTA analysis. Pathway analysis revealed that members of the sonic hedgehog pathway were less abundant in R345W- EVs, while EMT drivers were enriched. Additionally, R345W-EVs had higher concentrations of TGFβ1 compared to control. Critically, treatment with R345W-EVs was sufficient to increase EMT marker expression, as well as cell migration in recipient cells. This EV-increased cell migration was significantly inhibited by pre-incubation of EVs with pan-TGFβ-neutralising antibody. In conclusion, the expression of R345W-Fibulin-3 alters the size and cargo of EVs, which are sufficient to enhance the rate of cell migration in a TGFβ dependent manner. These results suggest that EV-bound TGFβ plays a critical role in the induction of EMT in RPE cells.
Collapse
Affiliation(s)
- Mi Zhou
- Department of OphthalmologyPenn State Hershey College of MedicineHersheyPennsylvaniaUSA
| | - Yuanjun Zhao
- Department of OphthalmologyPenn State Hershey College of MedicineHersheyPennsylvaniaUSA
| | - Sarah R. Weber
- Department of OphthalmologyPenn State Hershey College of MedicineHersheyPennsylvaniaUSA
| | - Christopher Gates
- Bioinformatics Core, Biomedical Research Core FacilitiesUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Nicholas J. Carruthers
- Bioinformatics Core, Biomedical Research Core FacilitiesUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Han Chen
- Microscopy Imaging Core FacilityPenn State Hershey College of MedicineHersheyPennsylvaniaUSA
| | - Xiaoming Liu
- Department of Pediatrics, Division of Hematology and OncologyPenn State Hershey College of MedicineHersheyPennsylvaniaUSA
| | - Hong‐Gang Wang
- Department of Pediatrics, Division of Hematology and OncologyPenn State Hershey College of MedicineHersheyPennsylvaniaUSA
| | | | - Matthew T. Swulius
- Department of Biochemistry and Molecular BiologyPenn State Hershey College of MedicineHersheyPennsylvaniaUSA
| | - Alistair J. Barber
- Department of OphthalmologyPenn State Hershey College of MedicineHersheyPennsylvaniaUSA
| | - Stephanie L. Grillo
- Department of OphthalmologyPenn State Hershey College of MedicineHersheyPennsylvaniaUSA
| | - Jeffrey M. Sundstrom
- Department of OphthalmologyPenn State Hershey College of MedicineHersheyPennsylvaniaUSA
| |
Collapse
|
4
|
Gao AY, Haak AJ, Bakri SJ. In vitro laboratory models of proliferative vitreoretinopathy. Surv Ophthalmol 2023; 68:861-874. [PMID: 37209723 DOI: 10.1016/j.survophthal.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Proliferative vitreoretinopathy (PVR), the most common cause of recurrent retinal detachment, is characterized by the formation and contraction of fibrotic membranes on the surface of the retina. There are no Food and Drug Administration (FDA)-approved drugs to prevent or treat PVR. Therefore, it is necessary to develop accurate in vitro models of the disease that will enable researchers to screen drug candidates and prioritize the most promising candidates for clinical studies. We provide a summary of recent in vitro PVR models, as well as avenues for model improvement. Several in vitro PVR models were identified, including various types of cell cultures. Additionally, novel techniques that have not been used to model PVR were identified, including organoids, hydrogels, and organ-on-a-chip models. Novel ideas for improving in vitro PVR models are highlighted. Researchers may consult this review to help design in vitro models of PVR, which will aid in the development of therapies to treat the disease.
Collapse
Affiliation(s)
- Ashley Y Gao
- Mayo Clinic, Department of Ophthalmology, Rochester, Minnesota, USA
| | - Andrew J Haak
- Mayo Clinic, Department of Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Sophie J Bakri
- Mayo Clinic, Department of Ophthalmology, Rochester, Minnesota, USA.
| |
Collapse
|
5
|
Yang X, Chung JY, Rai U, Esumi N. SIRT6 overexpression in the nucleus protects mouse retinal pigment epithelium from oxidative stress. Life Sci Alliance 2023; 6:e202201448. [PMID: 37185874 PMCID: PMC10130745 DOI: 10.26508/lsa.202201448] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Retinal pigment epithelium (RPE) is essential for the survival of retinal photoreceptors. To study retinal degeneration, sodium iodate (NaIO3) has been used to cause oxidative stress-induced RPE death followed by photoreceptor degeneration. However, analyses of RPE damage itself are still limited. Here, we characterized NaIO3-induced RPE damage, which was divided into three regions: periphery with normal-shaped RPE, transitional zone with elongated cells, and center with severely damaged or lost RPE. Elongated cells in the transitional zone exhibited molecular characteristics of epithelial-mesenchymal transition. Central RPE was more susceptible to stresses than peripheral RPE. Under stresses, SIRT6, an NAD+-dependent protein deacylase, rapidly translocated from the nucleus to the cytoplasm and colocalized with stress granule factor G3BP1, leading to nuclear SIRT6 depletion. To overcome this SIRT6 depletion, SIRT6 overexpression was induced in the nucleus in transgenic mice, which protected RPE from NaIO3 and partially preserved catalase expression. These results demonstrate topological differences of mouse RPE and warrant further exploring SIRT6 as a potential target for protecting RPE from oxidative stress-induced damage.
Collapse
Affiliation(s)
- Xue Yang
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jin-Yong Chung
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Usha Rai
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Noriko Esumi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Zou L, Wang X, Han X. LncRNA MALAT 1/miR-625-3p/HIF-1α axis regulates the EMT of hypoxia-induced RPE cells by activating NF-κB/snail signaling. Exp Cell Res 2023; 429:113650. [PMID: 37209990 DOI: 10.1016/j.yexcr.2023.113650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/03/2023] [Accepted: 05/14/2023] [Indexed: 05/22/2023]
Abstract
The retina may undergo structural and functional damage as a result of hypoxia, which could lead to permanent blindness. As competing endogenous RNAs (ceRNAs), lncRNAs are essential in eye disorders. The biological function of lncRNA MALAT 1 and its potential mechanisms in hypoxic-ischemic retinal diseases are still unknown. MALAT 1 and miR-625-3p expression alterations in hypoxia-treated RPE cells were examined using qRT-PCR. The target binding relationships between MALAT 1 and miR-625-3p, as well as between miR-625-3p and HIF-1α, were identified utilizing bioinformatics analysis and dual luciferase reporter assay. We observed that si-MALAT 1 and miR-625-3p mimic both reduced apoptosis and epithelial-mesenchymal transition (EMT) in hypoxic RPE cells, whereas si-MALAT 1 was reversed by miR-625-3p inhibitor. Further, we carried out a mechanistic investigation, and rescue assays demonstrated that MALAT 1 sponging miR-625-3p influenced HIF-1α expression and consequently took part in the NF-κB/Snail signaling pathway, which regulated apoptosis and EMT. In conclusion, our research had shown that the MALAT 1/miR-625-3p/HIF-1α axis drove the progression of hypoxic-ischemic retinal disorders and may serve as a promising predictive biomarker for their therapeutic and diagnostic targets.
Collapse
Affiliation(s)
- Lirong Zou
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Eye Hospital of China Medical University, Key Lens Research Laboratory of Liaoning Province, Shenyang City, Liaoning Province, 110005, PR China
| | - Xinling Wang
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Eye Hospital of China Medical University, Key Lens Research Laboratory of Liaoning Province, Shenyang City, Liaoning Province, 110005, PR China.
| | - Xiao Han
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Eye Hospital of China Medical University, Key Lens Research Laboratory of Liaoning Province, Shenyang City, Liaoning Province, 110005, PR China.
| |
Collapse
|
7
|
Sripathi SR, Hu MW, Turaga RC, Mikeasky R, Satyanarayana G, Cheng J, Duan Y, Maruotti J, Wahlin KJ, Berlinicke CA, Qian J, Esumi N, Zack DJ. IKKβ Inhibition Attenuates Epithelial Mesenchymal Transition of Human Stem Cell-Derived Retinal Pigment Epithelium. Cells 2023; 12:1155. [PMID: 37190063 PMCID: PMC10136838 DOI: 10.3390/cells12081155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 05/17/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT), which is well known for its role in embryonic development, malignant transformation, and tumor progression, has also been implicated in a variety of retinal diseases, including proliferative vitreoretinopathy (PVR), age-related macular degeneration (AMD), and diabetic retinopathy. EMT of the retinal pigment epithelium (RPE), although important in the pathogenesis of these retinal conditions, is not well understood at the molecular level. We and others have shown that a variety of molecules, including the co-treatment of human stem cell-derived RPE monolayer cultures with transforming growth factor beta (TGF-β) and the inflammatory cytokine tumor necrosis factor alpha (TNF-α), can induce RPE-EMT; however, small molecule inhibitors of RPE-EMT have been less well studied. Here, we demonstrate that BAY651942, a small molecule inhibitor of nuclear factor kapa-B kinase subunit beta (IKKβ) that selectively targets NF-κB signaling, can modulate TGF-β/TNF-α-induced RPE-EMT. Next, we performed RNA-seq studies on BAY651942 treated hRPE monolayers to dissect altered biological pathways and signaling events. Further, we validated the effect of IKKβ inhibition on RPE-EMT-associated factors using a second IKKβ inhibitor, BMS345541, with RPE monolayers derived from an independent stem cell line. Our data highlights the fact that pharmacological inhibition of RPE-EMT restores RPE identity and may provide a promising approach for treating retinal diseases that involve RPE dedifferentiation and EMT.
Collapse
Affiliation(s)
- Srinivasa R. Sripathi
- Department of Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Henderson Ocular Stem Cell Laboratory, Retina Foundation of the Southwest, Dallas, TX 75231, USA
| | - Ming-Wen Hu
- Department of Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ravi Chakra Turaga
- Caris Life Sciences, 350 W Washington St., Tempe, AZ 85281, USA
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Rebekah Mikeasky
- Department of Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ganesh Satyanarayana
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
- Emory Eye Center, Department of Ophthalmology, Emory University, Atlanta, GA 30322, USA
| | - Jie Cheng
- Department of Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yukan Duan
- Department of Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Karl J. Wahlin
- Shiley Eye Institute, University of California, San Diego, CA 92093, USA
| | - Cynthia A. Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jiang Qian
- Department of Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Noriko Esumi
- Department of Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Donald J. Zack
- Department of Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Genetic Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Whiting School of Engineering, Baltimore, MD 21218, USA
| |
Collapse
|
8
|
Zhang J, Li W, Xiong Z, Zhu J, Ren X, Wang S, Kuang H, Lin X, Mora A, Li X. PDGF-D-induced immunoproteasome activation and cell-cell interactions. Comput Struct Biotechnol J 2023; 21:2405-2418. [PMID: 37066124 PMCID: PMC10090480 DOI: 10.1016/j.csbj.2023.03.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
Platelet-derived growth factor-D (PDGF-D) is abundantly expressed in ocular diseases. Yet, it remains unknown whether and how PDGF-D affects ocular cells or cell-cell interactions in the eye. In this study, using single-cell RNA sequencing (scRNA-seq) and a mouse model of PDGF-D overexpression in retinal pigment epithelial (RPE) cells, we found that PDGF-D overexpression markedly upregulated the key immunoproteasome genes, leading to increased antigen processing/presentation capacity of RPE cells. Also, more than 6.5-fold ligand-receptor pairs were found in the PDGF-D overexpressing RPE-choroid tissues, suggesting markedly increased cell-cell interactions. Moreover, in the PDGF-D-overexpressing tissues, a unique cell population with a transcriptomic profile of both stromal cells and antigen-presenting RPE cells was detected, suggesting PDGF-D-induced epithelial-mesenchymal transition of RPE cells. Importantly, administration of ONX-0914, an immunoproteasome inhibitor, suppressed choroidal neovascularization (CNV) in a mouse CNV model in vivo. Together, we show that overexpression of PDGF-D increased pro-angiogenic immunoproteasome activities, and inhibiting immunoproteasome pathway may have therapeutic value for the treatment of neovascular diseases.
Collapse
|
9
|
Farkas MH, Skelton LA, Ramachandra-Rao S, Au E, Fliesler SJ. Morphological, biochemical, and transcriptomic characterization of iPSC-derived human RPE cells from normal and Smith-Lemli-Opitz syndrome patients. Mol Vis 2022; 28:394-411. [PMID: 36540063 PMCID: PMC9744241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/11/2022] [Indexed: 12/30/2022] Open
Affiliation(s)
- Michael H. Farkas
- Department of Ophthalmology (Ross Eye Institute), The State University of New York- University at Buffalo, Buffalo, NY
- Department of Biochemistry and the Neuroscience Graduate Program, The State University of New York- University at Buffalo, Buffalo, NY
- Research Service, VA Western New York Healthcare System, Buffalo, NY
| | - Lara A. Skelton
- Department of Ophthalmology (Ross Eye Institute), The State University of New York- University at Buffalo, Buffalo, NY
- Department of Biochemistry and the Neuroscience Graduate Program, The State University of New York- University at Buffalo, Buffalo, NY
- Research Service, VA Western New York Healthcare System, Buffalo, NY
| | - Sriganesh Ramachandra-Rao
- Department of Ophthalmology (Ross Eye Institute), The State University of New York- University at Buffalo, Buffalo, NY
- Department of Biochemistry and the Neuroscience Graduate Program, The State University of New York- University at Buffalo, Buffalo, NY
- Research Service, VA Western New York Healthcare System, Buffalo, NY
| | - Elizabeth Au
- Department of Ophthalmology (Ross Eye Institute), The State University of New York- University at Buffalo, Buffalo, NY
| | - Steven J. Fliesler
- Department of Ophthalmology (Ross Eye Institute), The State University of New York- University at Buffalo, Buffalo, NY
- Department of Biochemistry and the Neuroscience Graduate Program, The State University of New York- University at Buffalo, Buffalo, NY
- Research Service, VA Western New York Healthcare System, Buffalo, NY
| |
Collapse
|
10
|
Indrischek H, Hammer J, Machate A, Hecker N, Kirilenko B, Roscito J, Hans S, Norden C, Brand M, Hiller M. Vision-related convergent gene losses reveal SERPINE3's unknown role in the eye. eLife 2022; 11:77999. [PMID: 35727138 PMCID: PMC9355568 DOI: 10.7554/elife.77999] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022] Open
Abstract
Despite decades of research, knowledge about the genes that are important for development and function of the mammalian eye and are involved in human eye disorders remains incomplete. During mammalian evolution, mammals that naturally exhibit poor vision or regressive eye phenotypes have independently lost many eye-related genes. This provides an opportunity to predict novel eye-related genes based on specific evolutionary gene loss signatures. Building on these observations, we performed a genome-wide screen across 49 mammals for functionally uncharacterized genes that are preferentially lost in species exhibiting lower visual acuity values. The screen uncovered several genes, including SERPINE3, a putative serine proteinase inhibitor. A detailed investigation of 381 additional mammals revealed that SERPINE3 is independently lost in 18 lineages that typically do not primarily rely on vision, predicting a vision-related function for this gene. To test this, we show that SERPINE3 has the highest expression in eyes of zebrafish and mouse. In the zebrafish retina, serpine3 is expressed in Müller glia cells, a cell type essential for survival and maintenance of the retina. A CRISPR-mediated knockout of serpine3 in zebrafish resulted in alterations in eye shape and defects in retinal layering. Furthermore, two human polymorphisms that are in linkage with SERPINE3 are associated with eye-related traits. Together, these results suggest that SERPINE3 has a role in vertebrate eyes. More generally, by integrating comparative genomics with experiments in model organisms, we show that screens for specific phenotype-associated gene signatures can predict functions of uncharacterized genes.
Collapse
Affiliation(s)
- Henrike Indrischek
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Juliane Hammer
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Anja Machate
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Nikolai Hecker
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Juliana Roscito
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Stefan Hans
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Caren Norden
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Michael Brand
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | | |
Collapse
|
11
|
Sinha T, Ikelle L, Makia MS, Crane R, Zhao X, Kakakhel M, Al-Ubaidi MR, Naash MI. Riboflavin deficiency leads to irreversible cellular changes in the RPE and disrupts retinal function through alterations in cellular metabolic homeostasis. Redox Biol 2022; 54:102375. [PMID: 35738087 PMCID: PMC9233280 DOI: 10.1016/j.redox.2022.102375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 10/25/2022] Open
Abstract
Ariboflavinosis is a pathological condition occurring as a result of riboflavin deficiency. This condition is treatable if detected early enough, but it lacks timely diagnosis. Critical symptoms of ariboflavinosis include neurological and visual manifestations, yet the effects of flavin deficiency on the retina are not well investigated. Here, using a diet induced mouse model of riboflavin deficiency, we provide the first evidence of how retinal function and metabolism are closely intertwined with riboflavin homeostasis. We find that diet induced riboflavin deficiency causes severe decreases in retinal function accompanied by structural changes in the neural retina and retinal pigment epithelium (RPE). This is preceded by increased signs of cellular oxidative stress and metabolic disorder, in particular dysregulation in lipid metabolism, which is essential for both photoreceptors and the RPE. Though many of these deleterious phenotypes can be ameliorated by riboflavin supplementation, our data suggests that some patients may continue to suffer from multiple pathologies at later ages. These studies provide an essential cellular and mechanistic foundation linking defects in cellular flavin levels with the manifestation of functional deficiencies in the visual system and paves the way for a more in-depth understanding of the cellular consequences of ariboflavinosis.
Collapse
Affiliation(s)
- Tirthankar Sinha
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Larissa Ikelle
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Mustafa S Makia
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Ryan Crane
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Xue Zhao
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Mashal Kakakhel
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Muayyad R Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
12
|
Chang S, Singh L, Thaker K, Abedi S, Singh MK, Patel TH, Chwa M, Atilano SR, Udar N, Bota D, Kenney MC. Altered Retrograde Signaling Patterns in Breast Cancer Cells Cybrids with H and J Mitochondrial DNA Haplogroups. Int J Mol Sci 2022; 23:6687. [PMID: 35743133 PMCID: PMC9224519 DOI: 10.3390/ijms23126687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022] Open
Abstract
The aim of this study was to determine the role of retrograde signaling (mitochondria to nucleus) in MCF7 breast cancer cells. Therefore, in the present study, MCF7-H and MCF7-J cybrids were produced using the mitochondria from the same H and J individuals that were already used in our non-diseased retinal pigment epithelium (ARPE19) cybrids. MCF7 cybrids were treated with cisplatin and analyzed for cell viability, mitochondrial membrane potential, ROS, and expression levels of genes associated with the cGAS-STING and cancer-related pathways. Results showed that unlike the ARPE19-H and ARPE19-J cybrids, the untreated MCF7-H and MCF7-J cybrids had similar levels of ATP, lactate, and OCR: ECAR ratios. After cisplatin treatment, MCF7-H and MCF7-J cybrids showed similar (a) decreases in cell viability and ROS levels; (b) upregulation of ABCC1, BRCA1 and CDKN1A/P21; and (c) downregulation of EGFR. Cisplatin-treated ARPE19-H and ARPE19-J cybrids showed increased expression of six cGAS-STING pathway genes, while two were increased for MCF7-J cybrids. In summary, the ARPE19-H and ARPE19-J cybrids behave differentially from each other with or without cisplatin. In contrast, the MCF7-H and MCF7-J cybrids had identical metabolic/bioenergetic profiles and cisplatin responses. Our findings suggest that cancer cell nuclei might have a diminished ability to respond to the modulating signaling of the mtDNA that occurs via the cGAS-STING pathway.
Collapse
Affiliation(s)
- Steven Chang
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (L.S.); (K.T.); (S.A.); (M.K.S.); (T.H.P.); (M.C.); (S.R.A.); (N.U.)
| | - Lata Singh
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (L.S.); (K.T.); (S.A.); (M.K.S.); (T.H.P.); (M.C.); (S.R.A.); (N.U.)
| | - Kunal Thaker
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (L.S.); (K.T.); (S.A.); (M.K.S.); (T.H.P.); (M.C.); (S.R.A.); (N.U.)
| | - Sina Abedi
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (L.S.); (K.T.); (S.A.); (M.K.S.); (T.H.P.); (M.C.); (S.R.A.); (N.U.)
| | - Mithalesh K. Singh
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (L.S.); (K.T.); (S.A.); (M.K.S.); (T.H.P.); (M.C.); (S.R.A.); (N.U.)
| | - Tej H. Patel
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (L.S.); (K.T.); (S.A.); (M.K.S.); (T.H.P.); (M.C.); (S.R.A.); (N.U.)
| | - Marilyn Chwa
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (L.S.); (K.T.); (S.A.); (M.K.S.); (T.H.P.); (M.C.); (S.R.A.); (N.U.)
| | - Shari R. Atilano
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (L.S.); (K.T.); (S.A.); (M.K.S.); (T.H.P.); (M.C.); (S.R.A.); (N.U.)
| | - Nitin Udar
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (L.S.); (K.T.); (S.A.); (M.K.S.); (T.H.P.); (M.C.); (S.R.A.); (N.U.)
| | - Daniela Bota
- Department of Neurology, Neuro-Oncology Division, University of California Irvine, Irvine, CA 92697, USA;
| | - Maria Cristina Kenney
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA; (S.C.); (L.S.); (K.T.); (S.A.); (M.K.S.); (T.H.P.); (M.C.); (S.R.A.); (N.U.)
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|