1
|
Tan J, Duron A, Sucov HM, Makita T. Placode and neural crest origins of congenital deafness in mouse models of Waardenburg-Shah syndrome. iScience 2025; 28:111680. [PMID: 39868048 PMCID: PMC11762213 DOI: 10.1016/j.isci.2024.111680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/19/2024] [Accepted: 12/20/2024] [Indexed: 01/28/2025] Open
Abstract
Mutations in the human genes encoding the endothelin ligand-receptor pair EDN3 and EDNRB cause Waardenburg-Shah syndrome (WS4), which includes congenital hearing impairment. The current explanation for auditory dysfunction is defective migration of neural crest-derived melanocytes to the inner ear. We explored the role of endothelin signaling in auditory development in mice using neural crest-specific and placode-specific Ednrb mutation plus related genetic resources. On an outbred strain background, we find a normal representation of melanocytes in hearing-impaired mutant mice. Instead, our results in neural crest-specific Ednrb mutants implicate a previously unrecognized role for glial support of synapse assembly between auditory neurons and cochlear hair cells. Placode-specific Ednrb mutation also caused impaired hearing, resulting from deficient synaptic transmission. Our observations demonstrate the significant influence of genetic modifiers in auditory development, and invoke independent and separable roles for endothelin signaling in the neural crest and placode lineages to create a functional auditory circuitry.
Collapse
Affiliation(s)
- Jaime Tan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Alicia Duron
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Henry M. Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Takako Makita
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
2
|
Poltavski DM, Cunha AT, Tan J, Sucov HM, Makita T. Lineage-specific intersection of endothelin and GDNF signaling in enteric nervous system development. eLife 2024; 13:RP96424. [PMID: 39641974 PMCID: PMC11623925 DOI: 10.7554/elife.96424] [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] [Indexed: 12/07/2024] Open
Abstract
Two major ligand-receptor signaling axes - endothelin Edn3 and its receptor Ednrb, and glial-derived neurotrophic factor (GDNF) and its receptor Ret - are required for migration of enteric nervous system (ENS) progenitors to the hindgut. Mutations in either component cause colonic aganglionosis, also called Hirschsprung disease. Here, we have used Wnt1Cre and Pax2Cre in mice to show that these driver lines label distinct ENS lineages during progenitor migration and in their terminal hindgut fates. Both Cre lines result in Hirschsprung disease when combined with conditional Ednrb or conditional Ret alleles. In vitro explant assays and analysis of lineage-labeled mutant embryos show that GDNF but not Edn3 is a migration cue for cells of both lineages. Instead, Edn3-Ednrb function is required in both for GDNF responsiveness albeit in different ways: by expanding the Ret+ population in the Pax2Cre lineage, and by supporting Ret function in Wnt1Cre-derived cells. Our results demonstrate that two distinct lineages of progenitors give rise to the ENS, and that these divergently utilize endothelin signaling to support migration to the hindgut.
Collapse
Affiliation(s)
- Denise M Poltavski
- Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharlestonUnited States
| | - Alexander T Cunha
- Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharlestonUnited States
| | - Jaime Tan
- Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharlestonUnited States
| | - Henry M Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharlestonUnited States
| | - Takako Makita
- Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharlestonUnited States
| |
Collapse
|
3
|
Folkert IW, Molina Arocho WA, To TKJ, Devalaraja S, Molina IS, Shoush J, Mohei H, Zhai L, Akhtar MN, Kochat V, Arslan E, Lazar AJ, Wani K, Israel WP, Zhang Z, Chaluvadi VS, Norgard RJ, Liu Y, Fuller AM, Dang MT, Roses RE, Karakousis GC, Miura JT, Fraker DL, Eisinger-Mathason TK, Simon MC, Weber K, Tan K, Fan Y, Rai K, Haldar M. An iron-rich subset of macrophages promotes tumor growth through a Bach1-Ednrb axis. J Exp Med 2024; 221:e20230420. [PMID: 39347789 PMCID: PMC11457473 DOI: 10.1084/jem.20230420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/04/2024] [Accepted: 08/07/2024] [Indexed: 10/01/2024] Open
Abstract
We define a subset of macrophages in the tumor microenvironment characterized by high intracellular iron and enrichment of heme and iron metabolism genes. These iron-rich tumor-associated macrophages (iTAMs) supported angiogenesis and immunosuppression in the tumor microenvironment and were conserved between mice and humans. iTAMs comprise two additional subsets based on gene expression profile and location-perivascular (pviTAM) and stromal (stiTAM). We identified the endothelin receptor type B (Ednrb) as a specific marker of iTAMs and found myeloid-specific deletion of Ednrb to reduce tumor growth and vascular density. Further studies identified the transcription factor Bach1 as a repressor of the iTAM transcriptional program, including Ednrb expression. Heme is a known inhibitor of Bach1, and, correspondingly, heme exposure induced Ednrb and iTAM signature genes in macrophages. Thus, iTAMs are a distinct macrophage subset regulated by the transcription factor Bach1 and characterized by Ednrb-mediated immunosuppressive and angiogenic functions.
Collapse
Affiliation(s)
- Ian W. Folkert
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William A. Molina Arocho
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tsun Ki Jerrick To
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samir Devalaraja
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Irene S. Molina
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason Shoush
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hesham Mohei
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Zhai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Md Naushad Akhtar
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Veena Kochat
- Department of Genomic Medicine and MDACC Epigenomics Therapy Initiative, MD Anderson Cancer Center, Houston, TX, USA
| | - Emre Arslan
- Department of Genomic Medicine and MDACC Epigenomics Therapy Initiative, MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander J. Lazar
- Department of Genomic Medicine and MDACC Epigenomics Therapy Initiative, MD Anderson Cancer Center, Houston, TX, USA
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Khalida Wani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William P. Israel
- Department of Genomic Medicine and MDACC Epigenomics Therapy Initiative, MD Anderson Cancer Center, Houston, TX, USA
| | - Zhan Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Venkata S. Chaluvadi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert J. Norgard
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying Liu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley M. Fuller
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mai T. Dang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Washington University in St. Louis Schoold of Medicine, St. Louis, MO, USA
| | - Robert E. Roses
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Giorgos C. Karakousis
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John T. Miura
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas L. Fraker
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - T.S. Karin Eisinger-Mathason
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M. Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristy Weber
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kai Tan
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi Fan
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kunal Rai
- Department of Genomic Medicine and MDACC Epigenomics Therapy Initiative, MD Anderson Cancer Center, Houston, TX, USA
| | - Malay Haldar
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
4
|
Shigesada N, Shikada N, Shirai M, Toriyama M, Higashijima F, Kimura K, Kondo T, Bessho Y, Shinozuka T, Sasai N. Combination of blockade of endothelin signalling and compensation of IGF1 expression protects the retina from degeneration. Cell Mol Life Sci 2024; 81:51. [PMID: 38252153 PMCID: PMC10803390 DOI: 10.1007/s00018-023-05087-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 12/01/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
Retinitis pigmentosa (RP) and macular dystrophy (MD) cause severe retinal dysfunction, affecting 1 in 4000 people worldwide. This disease is currently assumed to be intractable, because effective therapeutic methods have not been established, regardless of genetic or sporadic traits. Here, we examined a RP mouse model in which the Prominin-1 (Prom1) gene was deficient and investigated the molecular events occurring at the outset of retinal dysfunction. We extracted the Prom1-deficient retina subjected to light exposure for a short time, conducted single-cell expression profiling, and compared the gene expression with and without stimuli. We identified the cells and genes whose expression levels change directly in response to light stimuli. Among the genes altered by light stimulation, Igf1 was decreased in rod photoreceptor cells and astrocytes under the light-stimulated condition. Consistently, the insulin-like growth factor (IGF) signal was weakened in light-stimulated photoreceptor cells. The recovery of Igf1 expression with the adeno-associated virus (AAV) prevented photoreceptor cell death, and its treatment in combination with the endothelin receptor antagonist led to the blockade of abnormal glial activation and the promotion of glycolysis, thereby resulting in the improvement of retinal functions, as assayed by electroretinography. We additionally demonstrated that the attenuation of mammalian/mechanistic target of rapamycin (mTOR), which mediates IGF signalling, leads to complications in maintaining retinal homeostasis. Together, we propose that combinatorial manipulation of distinct mechanisms is useful for the maintenance of the retinal condition.
Collapse
Affiliation(s)
- Naoya Shigesada
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan
| | - Naoya Shikada
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan
| | - Manabu Shirai
- Omics Research Center (ORC), National Cerebral and Cardiovascular Center, Suita, Osaka, 564-8565, Japan
| | - Michinori Toriyama
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, 669-1337, Japan
| | - Fumiaki Higashijima
- Department of Ophthalmology, Graduate School of Medicine, Yamaguchi University, Ube, 755-0046, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Graduate School of Medicine, Yamaguchi University, Ube, 755-0046, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
| | - Yasumasa Bessho
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan
| | - Takuma Shinozuka
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan
| | - Noriaki Sasai
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan.
| |
Collapse
|
5
|
Alarcon-Martinez L, Shiga Y, Villafranca-Baughman D, Cueva Vargas JL, Vidal Paredes IA, Quintero H, Fortune B, Danesh-Meyer H, Di Polo A. Neurovascular dysfunction in glaucoma. Prog Retin Eye Res 2023; 97:101217. [PMID: 37778617 DOI: 10.1016/j.preteyeres.2023.101217] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Retinal ganglion cells, the neurons that die in glaucoma, are endowed with a high metabolism requiring optimal provision of oxygen and nutrients to sustain their activity. The timely regulation of blood flow is, therefore, essential to supply firing neurons in active areas with the oxygen and glucose they need for energy. Many glaucoma patients suffer from vascular deficits including reduced blood flow, impaired autoregulation, neurovascular coupling dysfunction, and blood-retina/brain-barrier breakdown. These processes are tightly regulated by a community of cells known as the neurovascular unit comprising neurons, endothelial cells, pericytes, Müller cells, astrocytes, and microglia. In this review, the neurovascular unit takes center stage as we examine the ability of its members to regulate neurovascular interactions and how their function might be altered during glaucomatous stress. Pericytes receive special attention based on recent data demonstrating their key role in the regulation of neurovascular coupling in physiological and pathological conditions. Of particular interest is the discovery and characterization of tunneling nanotubes, thin actin-based conduits that connect distal pericytes, which play essential roles in the complex spatial and temporal distribution of blood within the retinal capillary network. We discuss cellular and molecular mechanisms of neurovascular interactions and their pathophysiological implications, while highlighting opportunities to develop strategies for vascular protection and regeneration to improve functional outcomes in glaucoma.
Collapse
Affiliation(s)
- Luis Alarcon-Martinez
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada; Centre for Eye Research Australia, University of Melbourne, Melbourne, Australia
| | - Yukihiro Shiga
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Jorge L Cueva Vargas
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Isaac A Vidal Paredes
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Heberto Quintero
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Healthy, Portland, OR, USA
| | - Helen Danesh-Meyer
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada.
| |
Collapse
|
6
|
MUSTHAFA AHMAD, RYANTO GUSTYRIZKYTEGUH, SURAYA RATOE, NAGANO TATSUYA, SUZUKI YOKO, HARA TETSUYA, HIRATA KENICHI, EMOTO NORIAKI. Acute Amelioration of Inflammatory Activity Caused by Endothelin-2 Deficiency during Acute Lung Injury. THE KOBE JOURNAL OF MEDICAL SCIENCES 2023; 69:E96-E105. [PMID: 37941117 PMCID: PMC10695096 DOI: 10.24546/0100483406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 11/10/2023]
Abstract
In acute lung injury (ALI), a severe insult induces a hyperinflammatory state in the lungs. The mortality rate of severe ALI remains high, and novel mechanistic insights are required to improve therapeutic strategies. Endothelin-2 (Edn2), the least studied isoform of endothelin, is involved in lung physiology and development and can be affected by various factors. One of them is inflammation, and another isoform of endothelin, endothelin-1 (Edn1), affects lung inflammatory responses. Considering the importance of Edn2 in the lungs and how Edn2 works through the same receptors as Edn1, we postulated that Edn2 may affect inflammatory responses that are central to ALI pathophysiology. In this study, we performed 24 hours intratracheal lipopolysaccharide (LPS) instillation or PBS control as an in vivo ALI model in eight-week-old conditional Edn2 knockout mice (Edn2-iKO), with Edn2-floxed mice as controls. Bronchoalveolar lavage (BAL) fluid and tissue were collected after exsanguination and analyzed for its cellular, molecular, functional, and histological inflammatory phenotypes. We found that Edn2-iKO mice displayed a reduced pro-neutrophilic inflammatory phenotype even after acute LPS treatment, shown by the reduction in the overall protein concentration and neutrophil count in bronchoalveolar lavage fluids. Further investigation revealed a reduction in mRNA interferon gamma (IFNγ) level of Edn2-iKO lungs and suppression of its downstream signaling, including phosphorylated level of STAT1 and IL-1β secretion, leading to reduced NFĸB activation. To conclude, Edn2 deletion suppressed acute lung inflammation by reducing neutrophil-mediated IFNγ/STAT1/IL-1β/NFĸB signaling cascade. Targeting Edn2 signaling may be beneficial for the development of novel treatment options for ALI.
Collapse
Affiliation(s)
- AHMAD MUSTHAFA
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | - RATOE SURAYA
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - TATSUYA NAGANO
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - YOKO SUZUKI
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan
| | - TETSUYA HARA
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan
| | - KEN-ICHI HIRATA
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - NORIAKI EMOTO
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan
| |
Collapse
|
7
|
Damle SR, Krzyzanowska AK, Korsun MK, Morse KW, Gilbert S, Kim HJ, Boachie-Adjei O, Rawlins BA, van der Meulen MCH, Greenblatt MB, Hidaka C, Cunningham ME. Inducing Angiogenesis in the Nucleus Pulposus. Cells 2023; 12:2488. [PMID: 37887332 PMCID: PMC10605635 DOI: 10.3390/cells12202488] [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: 07/31/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Bone morphogenetic protein (BMP) gene delivery to Lewis rat lumbar intervertebral discs (IVDs) drives bone formation anterior and external to the IVD, suggesting the IVD is inhospitable to osteogenesis. This study was designed to determine if IVD destruction with a proteoglycanase, and/or generating an IVD blood supply by gene delivery of an angiogenic growth factor, could render the IVD permissive to intra-discal BMP-driven osteogenesis and fusion. Surgical intra-discal delivery of naïve or gene-programmed cells (BMP2/BMP7 co-expressing or VEGF165 expressing) +/- purified chondroitinase-ABC (chABC) in all permutations was performed between lumbar 4/5 and L5/6 vertebrae, and radiographic, histology, and biomechanics endpoints were collected. Follow-up anti-sFlt Western blotting was performed. BMP and VEGF/BMP treatments had the highest stiffness, bone production and fusion. Bone was induced anterior to the IVD, and was not intra-discal from any treatment. chABC impaired BMP-driven osteogenesis, decreased histological staining for IVD proteoglycans, and made the IVD permissive to angiogenesis. A soluble fragment of VEGF Receptor-1 (sFlt) was liberated from the IVD matrix by incubation with chABC, suggesting dysregulation of the sFlt matrix attachment is a possible mechanism for the chABC-mediated IVD angiogenesis we observed. Based on these results, the IVD can be manipulated to foster vascular invasion, and by extension, possibly osteogenesis.
Collapse
Affiliation(s)
- Sheela R. Damle
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Agata K. Krzyzanowska
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Maximilian K. Korsun
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Kyle W. Morse
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Susannah Gilbert
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Han Jo Kim
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Oheneba Boachie-Adjei
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Bernard A. Rawlins
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Marjolein C. H. van der Meulen
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Meinig School of Biomedical Engineering and Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Chisa Hidaka
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Department of Genetic Medicine and Belfer Gene Therapy Core Facility, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Matthew E. Cunningham
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| |
Collapse
|
8
|
Su Z, Qin F, Zhang H, Huang Z, Guan K, Zheng M, Dai Z, Song W, Li X. Evaluation of developmental toxicity of safinamide in zebrafish larvae (Danio rerio). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115284. [PMID: 37556957 DOI: 10.1016/j.ecoenv.2023.115284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 08/11/2023]
Abstract
Monoamine oxidase-B (MAO-B), as a principal metabolizing enzyme, plays important roles in the metabolism of catecholamines and xenobiotics in the central nervous system and peripheral tissues. Safinamide, the third-generation reversible MAO-B inhibitor, has potential to alleviate many neurological diseases such as Parkinson's disease (PD) and depression. Exposure to clinical psychotropic drugs often has adverse effects on fetuses. Currently, a variety of studies of safinamide focus on its curative effect and pharmacological effect, while its side effect of embryonic development is barely studied. In this study, we used zebrafish as a model to evaluate the embryonic developmental toxicity of safinamide. Our results revealed that higher concentrations (30 μM) of safinamide treatment caused a decrease in hatching rate and an increase in malformation and mortality in zebrafish larvae. Meanwhile, we observed that lower safinamide exposure (10 μM) increased the body length of zebrafish larvae and resulted in hyperactivity-like behaviors. In addition, an increased trend in dopamine (DA) level was found in 3.3 μM and 10 μM safinamide-exposed groups. Transcriptome analysis identified that safinamide exposure may disturb a variety of physiological processes such as neuroactive ligand-receptor interaction signaling pathway. In summary, our study reveals that safinamide may cause developmental defects in zebrafish larvae and provides insights into its toxic reactions in early develoment.
Collapse
Affiliation(s)
- Zhengkang Su
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China
| | - Fengqing Qin
- Guangxi College and University Key Laboratory of High-value Utilization of Seafood and Prepared Food in Beibu Gulf
| | - Hai Zhang
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang, 325035, China
| | - Zhengwei Huang
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China
| | - Kaiyu Guan
- Wenzhou Seventh People's Hospital, Wenzhou, Zhejiang 325000, China
| | - Miaomiao Zheng
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China
| | - Ziru Dai
- Guangxi College and University Key Laboratory of High-value Utilization of Seafood and Prepared Food in Beibu Gulf
| | - Weihong Song
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China; Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xi Li
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China; College of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
9
|
Song Z, Chen B, Tsai CH, Wu D, Liu E, Hawkins IS, Phan A, Auman JT, Tao Y, Mei H. Differentiation Trajectory of Limbal Stem and Progenitor Cells under Normal Homeostasis and upon Corneal Wounding. Cells 2022; 11:cells11131983. [PMID: 35805068 PMCID: PMC9266118 DOI: 10.3390/cells11131983] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/06/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
Limbal stem cells (LSCs) reside discretely at limbus surrounded by niche cells and progenitor cells. The aim of this study is to identify the heterogeneous cell populations at limbus under normal homeostasis and upon wounding using single-cell RNA sequencing in a mouse model. Two putative LSC types were identified which showed a differentiation trajectory into limbal progenitor cell (LPC) types under normal homeostasis and during wound healing. They were designated as “putative active LSCs” and “putative quiescent LSCs”, respectively, because the former type actively divided upon wounding while the later type stayed at a quiescent status upon wounding. The “putative quiescent LSCs” might contribute to a barrier function due to their characteristic markers regulating vascular and epithelial barrier and growth. Different types of LPCs at different proliferative statuses were identified in unwounded and wounded corneas with distinctive markers. Four maturation markers (Aldh3, Slurp1, Tkt, and Krt12) were screened out for corneal epithelium, which showed an increased expression along the differentiation trajectory during corneal epithelial maturation. In conclusion, our study identified two different types of putative LSCs and several types of putative LPCs under normal homeostasis and upon wounding, which will facilitate the understanding of corneal epithelial regeneration and wound healing.
Collapse
Affiliation(s)
- Zhenwei Song
- Department of Ophthalmology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Z.S.); (C.-H.T.); (E.L.); (I.S.H.)
- School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha 410081, China
| | - Brian Chen
- Department of Biostatistics, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (B.C.); (D.W.)
| | - Chi-Hao Tsai
- Department of Ophthalmology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Z.S.); (C.-H.T.); (E.L.); (I.S.H.)
| | - Di Wu
- Department of Biostatistics, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (B.C.); (D.W.)
- Division of Oral and Craniofacial Health Research, Adams School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Emily Liu
- Department of Ophthalmology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Z.S.); (C.-H.T.); (E.L.); (I.S.H.)
| | - Isha Sharday Hawkins
- Department of Ophthalmology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Z.S.); (C.-H.T.); (E.L.); (I.S.H.)
| | - Andrew Phan
- Department of Psychology and Neuroscience, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - James Todd Auman
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.T.A.); (Y.T.)
| | - Yazhong Tao
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.T.A.); (Y.T.)
| | - Hua Mei
- Department of Ophthalmology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Z.S.); (C.-H.T.); (E.L.); (I.S.H.)
- Department of Cell Biology and Physiology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence:
| |
Collapse
|
10
|
Fan Y, Chen W, Wei R, Qiang W, Pearson JD, Yu T, Bremner R, Chen D. Mapping transgene insertion sites reveals the α-Cre transgene expression in both developing retina and olfactory neurons. Commun Biol 2022; 5:411. [PMID: 35505181 PMCID: PMC9065156 DOI: 10.1038/s42003-022-03379-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 04/18/2022] [Indexed: 02/05/2023] Open
Abstract
The Tg(Pax6-cre,GFP)2Pgr (α-Cre) mouse is a commonly used Cre line thought to be retinal-specific. Using targeted locus amplification (TLA), we mapped the insertion site of the transgene, and defined primers useful to deduce zygosity. Further analyses revealed four tandem copies of the transgene. The insertion site mapped to clusters of vomeronasal and olfactory receptor genes. Using R26R and Ai14 Cre reporter mice, we confirmed retinal Cre activity, but also detected expression in Gα0+ olfactory neurons. Most α-Cre+ olfactory neurons do not express Pax6, implicating the influence of neighboring regulatory elements. RT-PCR and buried food pellet test did not detect any effects of the transgene on flanking genes in the nasal mucosa and retina. Together, these data precisely map α-Cre, show that it does not affect surrounding loci, but reveal previously unanticipated transgene expression in olfactory neurons. The α-Cre mouse can be a valuable tool in both retinal and olfactory research. The Pax6-α-Cre mouse line used in retinal studies actually contains four transgene insertion within gene clusters of olfactory and vomeronasal receptors, leading to expression in not just retinal, but also olfactory and vomeronasal sensory neurons.
Collapse
Affiliation(s)
- Yimeng Fan
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Wenyue Chen
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Ran Wei
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Qiang
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Joel D Pearson
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Departments of Ophthalmology and Visual Science, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tao Yu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Departments of Ophthalmology and Visual Science, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Rod Bremner
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Departments of Ophthalmology and Visual Science, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Danian Chen
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China. .,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China. .,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Departments of Ophthalmology and Visual Science, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Marola OJ, Howell GR, Libby RT. Vascular derived endothelin receptor A controls endothelin-induced retinal ganglion cell death. Cell Death Discov 2022; 8:207. [PMID: 35429992 PMCID: PMC9013356 DOI: 10.1038/s41420-022-00985-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/13/2022] [Accepted: 03/23/2022] [Indexed: 11/29/2022] Open
Abstract
Endothelin (EDN, also known as ET) signaling has been suggested to be an important mediator of retinal ganglion cell (RGC) death in glaucoma. Antagonism of EDN receptors (EDNRA and EDNRB, also known as ET-A and ET-B) prevented RGC death in mouse models of chronic ocular hypertension, and intravitreal injection of EDN ligand was sufficient to drive RGC death. However, it remains unclear which cell types EDN ligands directly affect to elicit RGC death. Multiple cell types in the retina and optic nerve express EDNRA and EDNRB and thus could respond to EDN ligands in the context of glaucoma. Here, we systematically deleted Edn receptors from specific cell types to identify the critical EDN receptor mediating RGC death in vivo. Deletion of both Ednra and Ednrb from retinal neurons (including RGCs) and macroglia did not prevent RGC loss after exposure to EDN1 ligands, suggesting EDN1 ligands cause RGC death via an indirect mechanism involving a secondary cell type. Deletion of Ednra from the full body, and then specifically from vascular mural cells, prevented EDN1-induced vasoconstriction and RGC death. Together, these data suggest EDN ligands cause RGC death via a mechanism initiated by vascular mural cells. It is possible RGC death is a consequence of vascular mural cell-induced vasoconstriction and its pathological sequelae. These results highlight the potential importance of neurovascular dysfunction in glaucoma.
Collapse
Affiliation(s)
- Olivia J Marola
- Department of Ophthalmology, Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY, USA
- Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
- The Center for Visual Sciences, University of Rochester, Rochester, NY, USA
| | - Gareth R Howell
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Richard T Libby
- Department of Ophthalmology, Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY, USA.
- The Center for Visual Sciences, University of Rochester, Rochester, NY, USA.
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
12
|
Syc-Mazurek SB, Yang HS, Marola OJ, Howell GR, Libby RT. Transcriptional control of retinal ganglion cell death after axonal injury. Cell Death Dis 2022; 13:244. [PMID: 35296661 PMCID: PMC8927149 DOI: 10.1038/s41419-022-04666-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 01/19/2022] [Accepted: 02/09/2022] [Indexed: 11/25/2022]
Abstract
Injury to the axons of retinal ganglion cells (RGCs) is a key pathological event in glaucomatous neurodegeneration. The transcription factors JUN (the target of the c-Jun N-terminal kinases, JNKs) and DDIT3/CHOP (a mediator of the endoplasmic reticulum stress response) have been shown to control the majority of proapoptotic signaling after mechanical axonal injury in RGCs and in other models of neurodegeneration. The downstream transcriptional networks controlled by JUN and DDIT3, which are critical for RGC death, however, are not well defined. To determine these networks, RNA was isolated from the retinas of wild-type mice and mice deficient in Jun, Ddit3, and both Jun and Ddit3 three days after mechanical optic nerve crush injury (CONC). RNA-sequencing data analysis was performed and immunohistochemistry was used to validate potential transcriptional signaling changes after axonal injury. This study identified downstream transcriptional changes after injury including both neuronal survival and proinflammatory signaling that were attenuated to differing degrees by loss of Ddit3, Jun, and Ddit3/Jun. These data suggest proinflammatory signaling in the retina might be secondary to activation of pro-death pathways in RGCs after acute axonal injury. These results determine the downstream transcriptional networks important for apoptotic signaling which may be important for ordering and staging the pro-degenerative signals after mechanical axonal injury.
Collapse
Affiliation(s)
- Stephanie B Syc-Mazurek
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY, USA
- Medical Scientist Training Program, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Olivia J Marola
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY, USA
- Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
- The Center for Visual Sciences, University of Rochester, Rochester, NY, USA
| | | | - Richard T Libby
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY, USA.
- The Center for Visual Sciences, University of Rochester, Rochester, NY, USA.
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
13
|
SORAYA ARISTIINTAN, SUZUKI YOKO, MORIMOTO MITSURU, KO CHEMYONGJAY, IKEDA KOJI, HIRATA KENICHI, EMOTO NORIAKI. Protective Effects of Endothelin-2 Expressed in Epithelial Cells on Bleomycin-Induced Pulmonary Fibrosis in Mice. THE KOBE JOURNAL OF MEDICAL SCIENCES 2021; 67:E61-E70. [PMID: 34795157 PMCID: PMC8622215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/05/2021] [Indexed: 06/13/2023]
Abstract
Initially, endothelin (ET)-2 was described as an endothelium-derived vasoconstrictor. However, accumulating evidence suggests the involvement of ET-2 in non-cardiovascular physiology and disease pathophysiology. The deficiency of ET-2 in mice can be lethal, and such mice exhibit a distinct developmental abnormality in the lungs. Nonetheless, the definite role of ET-2 in the lungs remains unclear. The ET-2 isoform, ET-1, promotes pulmonary fibrosis in mice. Although endothelin receptor antagonists (ERAs) show improvements in bleomycin-induced pulmonary fibrosis in mouse models, clinical trials examining ERAs for pulmonary fibrosis treatment have been unsuccessful, even showing harmful effects in patients. We hypothesized that ET-2, which activates the same receptor as ET-1, plays a distinct role in pulmonary fibrosis. In this study, we showed that ET-2 is expressed in the lung epithelium, and ET-2 deletion in epithelial cells of mice results in the exacerbation of bleomycin-induced pulmonary fibrosis. ET-2 knockdown in lung epithelial cell lines resulted in increased apoptosis mediated via oxidative stress induction. In contrast to the effects of ET-1, which induced fibroblast activation, ET-2 hampered fibroblast activation in primary mouse lung fibroblast cells by inhibiting the TGF-β-SMAD2/3 pathway. Our results demonstrated the divergent roles of ET-1 and ET-2 in pulmonary fibrosis pathophysiology and suggested that ET-2, expressed in epithelial cells, exerts protective effects against the development of pulmonary fibrosis in mice.
Collapse
Affiliation(s)
- ARISTI INTAN SORAYA
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, 650-0017, Japan
| | - YOKO SUZUKI
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, 658-8558, Japan
| | - MITSURU MORIMOTO
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - CHEMYONG JAY KO
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL 61802, USA
| | - KOJI IKEDA
- Department of Epidemiology for Longevity and Regional Health, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - KEN-ICHI HIRATA
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, 650-0017, Japan
| | - NORIAKI EMOTO
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, 650-0017, Japan
| |
Collapse
|
14
|
Kobayashi Y, Watanabe S, Ong ALC, Shirai M, Yamashiro C, Ogata T, Higashijima F, Yoshimoto T, Hayano T, Asai Y, Sasai N, Kimura K. Early manifestations and differential gene expression associated with photoreceptor degeneration in Prom1-deficient retina. Dis Model Mech 2021; 14:272527. [PMID: 34664634 PMCID: PMC8628633 DOI: 10.1242/dmm.048962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Retinitis pigmentosa (RP) and macular dystrophy (MD) are characterized by gradual photoreceptor death in the retina and are often associated with genetic mutations, including those in the prominin-1 (Prom1) gene. Prom1-knockout (KO) mice recapitulate key features of these diseases including light-dependent retinal degeneration and constriction of retinal blood vessels. The mechanisms underlying such degeneration have remained unclear, however. We here analysed early events associated with retinal degeneration in Prom1-KO mice. We found that photoreceptor cell death and glial cell activation occur between 2 and 3 weeks after birth. Whereas gene expression was not affected at 2 weeks, the expression of several genes was altered at 3 weeks in the Prom1-KO retina, with the expression of that for endothelin-2 (Edn2) being markedly upregulated. Expression of Edn2 was also induced by light stimulation in Prom1-KO mice reared in the dark. Treatment with endothelin receptor antagonists attenuated photoreceptor cell death, gliosis and retinal vessel stenosis in Prom1-KO mice. Our findings thus reveal early manifestations of retinal degeneration in a model of RP/MD and suggest potential therapeutic agents for these diseases. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Yuka Kobayashi
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Shizuka Watanabe
- Developmental Biomedical Science, Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Agnes Lee Chen Ong
- Developmental Biomedical Science, Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Manabu Shirai
- Omics Research Center (ORC), National Cerebral and Cardiovascular Center, 6-1 Kishibe Shinmachi, Suita, Osaka 564-8565, Japan
| | - Chiemi Yamashiro
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Tadahiko Ogata
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Fumiaki Higashijima
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Takuya Yoshimoto
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Takahide Hayano
- Department of Systems Bioinformatics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Yoshiyuki Asai
- Department of Systems Bioinformatics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Noriaki Sasai
- Developmental Biomedical Science, Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| |
Collapse
|
15
|
Wang Y, Smallwood PM, Williams J, Nathans J. A mouse model for kinesin family member 11 (Kif11)-associated familial exudative vitreoretinopathy. Hum Mol Genet 2021; 29:1121-1131. [PMID: 31993640 DOI: 10.1093/hmg/ddaa018] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 12/07/2019] [Accepted: 01/22/2020] [Indexed: 12/30/2022] Open
Abstract
During mitosis, Kif11, a kinesin motor protein, promotes bipolar spindle formation and chromosome movement, and during interphase, Kif11 mediates diverse trafficking processes in the cytoplasm. In humans, inactivating mutations in KIF11 are associated with (1) retinal hypovascularization with or without microcephaly and (2) multi-organ syndromes characterized by variable combinations of lymphedema, chorioretinal dysplasia, microcephaly and/or mental retardation. To explore the pathogenic basis of KIF11-associated retinal vascular disease, we generated a Kif11 conditional knockout (CKO) mouse and investigated the consequences of early postnatal inactivation of Kif11 in vascular endothelial cells (ECs). The principal finding is that postnatal EC-specific loss of Kif11 leads to severely stunted growth of the retinal vasculature, mildly stunted growth of the cerebellar vasculature and little or no effect on the vasculature elsewhere in the central nervous system (CNS). Thus, in mice, Kif11 function in early postnatal CNS ECs is most significant in the two CNS regions-the retina and cerebellum-that exhibit the most rapid rate of postnatal growth, which may sensitize ECs to impaired mitotic spindle function. Several lines of evidence indicate that these phenotypes are not caused by reduced beta-catenin signaling in ECs, despite the close resemblance of the Kif11 CKO phenotype to that caused by EC-specific reductions in beta-catenin signaling. Based on prior work, defective beta-catenin signaling had been the only known mechanism responsible for monogenic human disorders of retinal hypovascularization. The present study implies that retinal hypovascularization can arise from a second and mechanistically distinct cause.
Collapse
Affiliation(s)
- Yanshu Wang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Philip M Smallwood
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - John Williams
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
16
|
Jain F, Longakit A, Huang JLY, Van Raamsdonk CD. Endothelin signaling promotes melanoma tumorigenesis driven by constitutively active GNAQ. Pigment Cell Melanoma Res 2020; 33:834-849. [PMID: 32453908 DOI: 10.1111/pcmr.12900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 05/15/2020] [Accepted: 05/17/2020] [Indexed: 12/20/2022]
Abstract
The G-protein-coupled receptor, endothelin receptor B (EDNRB), is an important regulator of melanocyte survival and proliferation. It acts by stimulating downstream heterotrimeric G proteins, such as Gαq and Gα1 . Constitutively active, oncogenic versions of Gαq and Gα11 drive melanomagenesis, but the role of Ednrb in the context of these mutant G proteins has not been previously examined. In this paper, we used a knock-in mouse allele at the Rosa26 locus to force oncogenic GNAQQ209L expression in melanocytes in combination with Ednrb gene knockout. The resulting pathological analysis revealed that every aspect of melanomagenesis driven by GNAQQ209L was inhibited. We conclude that even in the presence of oncogenic Gαq , the Ednrb receptor activates normal Gαq and Gα11 proteins. This likely promotes tumorigenesis by activating phospholipase C-beta, the immediate effector of Gαq/11 . These findings suggest that it might be possible to target upstream receptors to offset the effects of hyperactive G proteins, recognized as the cause of a growing number of human disorders.
Collapse
Affiliation(s)
- Fagun Jain
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Anne Longakit
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Jenny Li-Ying Huang
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Catherine D Van Raamsdonk
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Impacts of ciliary neurotrophic factor on the retinal transcriptome in a mouse model of photoreceptor degeneration. Sci Rep 2020; 10:6593. [PMID: 32313077 PMCID: PMC7171121 DOI: 10.1038/s41598-020-63519-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/01/2020] [Indexed: 01/13/2023] Open
Abstract
Ciliary neurotrophic factor (CNTF) has been tested in clinical trials for human retinal degeneration due to its potent neuroprotective effects in various animal models. To decipher CNTF-triggered molecular events in the degenerating retina, we performed high-throughput RNA sequencing analyses using the Rds/Prph2 (P216L) transgenic mouse as a preclinical model for retinitis pigmentosa. In the absence of CNTF treatment, transcriptome alterations were detected at the onset of rod degeneration compared with wild type mice, including reduction of key photoreceptor transcription factors Crx, Nrl, and rod phototransduction genes. Short-term CNTF treatments caused further declines of photoreceptor transcription factors accompanied by marked decreases of both rod- and cone-specific gene expression. In addition, CNTF triggered acute elevation of transcripts in the innate immune system and growth factor signaling. These immune responses were sustained after long-term CNTF exposures that also affected neuronal transmission and metabolism. Comparisons of transcriptomes also uncovered common pathways shared with other retinal degeneration models. Cross referencing bulk RNA-seq with single-cell RNA-seq data revealed the CNTF responsive cell types, including Müller glia, rod and cone photoreceptors, and bipolar cells. Together, these results demonstrate the influence of exogenous CNTF on the retinal transcriptome landscape and illuminate likely CNTF impacts in degenerating human retinas.
Collapse
|
18
|
A putative silencer variant in a spontaneous canine model of retinitis pigmentosa. PLoS Genet 2020; 16:e1008659. [PMID: 32150541 PMCID: PMC7082071 DOI: 10.1371/journal.pgen.1008659] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 03/19/2020] [Accepted: 02/06/2020] [Indexed: 01/19/2023] Open
Abstract
Retinitis pigmentosa (RP) is the leading cause of blindness with nearly two million people affected worldwide. Many genes have been implicated in RP, yet in 30–80% of the RP patients the genetic cause remains unknown. A similar phenotype, progressive retinal atrophy (PRA), affects many dog breeds including the Miniature Schnauzer. We performed clinical, genetic and functional experiments to identify the genetic cause of PRA in the breed. The age of onset and pattern of disease progression suggested that at least two forms of PRA, types 1 and 2 respectively, affect the breed, which was confirmed by genome-wide association study that implicated two distinct genomic loci in chromosomes 15 and X, respectively. Whole-genome sequencing revealed a fully segregating recessive regulatory variant in type 1 PRA. The associated variant has a very recent origin based on haplotype analysis and lies within a regulatory site with the predicted binding site of HAND1::TCF3 transcription factor complex. Luciferase assays suggested that mutated regulatory sequence increases expression. Case-control retinal expression comparison of six best HAND1::TCF3 target genes were analyzed with quantitative reverse-transcriptase PCR assay and indicated overexpression of EDN2 and COL9A2 in the affected retina. Defects in both EDN2 and COL9A2 have been previously associated with retinal degeneration. In summary, our study describes two genetically different forms of PRA and identifies a fully penetrant variant in type 1 form with a possible regulatory effect. This would be among the first reports of a regulatory variant in retinal degeneration in any species, and establishes a new spontaneous dog model to improve our understanding of retinal biology and gene regulation while the affected breed will benefit from a reliable genetic testing. Retinitis pigmentosa (RP) is a blinding eye disease that affects nearly two million people worldwide. Several genes and variants have been associated with the disease, but still 30–80% of the patients lack genetic diagnosis. There is currently no standard treatment for RP, and much is expected from gene therapy. A similar disease, called progressive retinal atrophy (PRA), affects many dog breeds. We performed clinical, genetic and functional analyses to find the genetic cause for PRA in Miniature Schnauzers. We discovered two forms of PRA in the breed, named type 1 and 2, and show that they are genetically distinct as they map to different chromosomes, 15 and X, respectively. Further genetic, bioinformatic and functional analyses discovered a fully penetrant recessive variant in a putative silencer region for type 1 PRA. Silencer regions are important for gene regulation and we found that two of its predicted target genes, EDN2 and COL9A2, were overexpressed in the retina of the affected dog. Defects in both EDN2 and COL9A2 have been associated with retinal degeneration. This study provides new insights to retinal biology while the genetic test guides better breeding choices.
Collapse
|
19
|
Zasada M, Madetko-Talowska A, Revhaug C, Rognlien AGW, Baumbusch LO, Książek T, Szewczyk K, Grabowska A, Bik-Multanowski M, Józef Pietrzyk J, Kwinta P, Saugstad OD. Short- and long-term impact of hyperoxia on the blood and retinal cells' transcriptome in a mouse model of oxygen-induced retinopathy. Pediatr Res 2020; 87:485-493. [PMID: 31578039 PMCID: PMC7033041 DOI: 10.1038/s41390-019-0598-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/04/2019] [Accepted: 09/06/2019] [Indexed: 01/25/2023]
Abstract
BACKGROUND We aimed to identify global blood and retinal gene expression patterns in murine oxygen-induced retinopathy (OIR), a common model of retinopathy of prematurity, which may allow better understanding of the pathogenesis of this severe ocular prematurity complication and identification of potential blood biomarkers. METHODS A total of 120 C57BL/6J mice were randomly divided into an OIR group, in which 7-day-old pups were maintained in 75% oxygen for 5 days, or a control group. RNA was extracted from the whole-blood mononuclear cells and retinal cells on days 12, 17, and 28. Gene expression in the RNA samples was evaluated with mouse gene expression microarrays. RESULTS There were 38, 1370 and 111 genes, the expression of which differed between the OIR and control retinas on days 12, 17, and 28, respectively. Gene expression in the blood mononuclear cells was significantly altered only on day 17. Deptor and Nol4 genes showed reduced expression both in the blood and retinal cells on day 17. CONCLUSION There are sustained marked changes in the global pattern of gene expression in the OIR mice retinas. An altered expression of Deptor and Nol4 genes in the blood mononuclear cells requires further investigation as they may indicate retinal neovascularization.
Collapse
Affiliation(s)
- Magdalena Zasada
- 0000 0001 2162 9631grid.5522.0Department of Paediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Madetko-Talowska
- 0000 0001 2162 9631grid.5522.0Department of Medical Genetics, Jagiellonian University Medical College, Krakow, Poland
| | - Cecilie Revhaug
- 0000 0004 0389 8485grid.55325.34Department of Paediatric Research, Oslo University Hospital Rikshospitalet, Oslo, Norway ,0000 0004 1936 8921grid.5510.1University of Oslo, Oslo, Norway
| | - Anne Gro W. Rognlien
- 0000 0004 0389 8485grid.55325.34Department of Paediatric Research, Oslo University Hospital Rikshospitalet, Oslo, Norway ,0000 0004 1936 8921grid.5510.1University of Oslo, Oslo, Norway
| | - Lars O. Baumbusch
- 0000 0004 0389 8485grid.55325.34Department of Paediatric Research, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Teofila Książek
- 0000 0001 2162 9631grid.5522.0Department of Medical Genetics, Jagiellonian University Medical College, Krakow, Poland
| | - Katarzyna Szewczyk
- 0000 0001 2162 9631grid.5522.0Department of Medical Genetics, Jagiellonian University Medical College, Krakow, Poland
| | - Agnieszka Grabowska
- 0000 0001 2162 9631grid.5522.0Department of Medical Genetics, Jagiellonian University Medical College, Krakow, Poland
| | - Miroslaw Bik-Multanowski
- 0000 0001 2162 9631grid.5522.0Department of Medical Genetics, Jagiellonian University Medical College, Krakow, Poland
| | - Jacek Józef Pietrzyk
- 0000 0001 2162 9631grid.5522.0Department of Paediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Przemko Kwinta
- Department of Paediatrics, Jagiellonian University Medical College, Krakow, Poland.
| | - Ola Didrik Saugstad
- 0000 0004 0389 8485grid.55325.34Department of Paediatric Research, Oslo University Hospital Rikshospitalet, Oslo, Norway ,0000 0004 1936 8921grid.5510.1University of Oslo, Oslo, Norway
| |
Collapse
|
20
|
Rattner A, Williams J, Nathans J. Roles of HIFs and VEGF in angiogenesis in the retina and brain. J Clin Invest 2019; 129:3807-3820. [PMID: 31403471 DOI: 10.1172/jci126655] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 06/13/2019] [Indexed: 12/17/2022] Open
Abstract
Vascular development in the mammalian retina is a paradigm for CNS vascular development in general, and its study is revealing fundamental mechanisms that explain the efficacy of antiangiogenic therapies in retinal vascular disease. During development of the mammalian retina, hypoxic astrocytes are hypothesized to secrete VEGF, which attracts growing endothelial cells as they migrate radially from the optic disc. However, published tests of this model using astrocyte-specific deletion of Vegf in the developing mouse retina appear to contradict this theory. Here, we report that selectively eliminating Vegf in neonatal retinal astrocytes with a Gfap-Cre line that recombines with approximately 100% efficiency had no effect on proliferation or radial migration of astrocytes, but completely blocked radial migration of endothelial cells, strongly supporting the hypoxic astrocyte model. Using additional Cre driver lines, we found evidence for essential and partially redundant actions of retina-derived (paracrine) and astrocyte-derived (autocrine) VEGF in controlling astrocyte proliferation and migration. We also extended previous studies by showing that HIF-1α in retinal neurons and HIF-2α in Müller glia play distinct roles in retinal vascular development and disease, adding to a growing body of data that point to the specialization of these 2 hypoxia-sensing transcription factors.
Collapse
Affiliation(s)
| | - John Williams
- Department of Molecular Biology and Genetics.,Howard Hughes Medical Institute
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics.,Howard Hughes Medical Institute.,Department of Neuroscience, and.,Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Elshaer SL, Evans W, Pentecost M, Lenin R, Periasamy R, Jha KA, Alli S, Gentry J, Thomas SM, Sohl N, Gangaraju R. Adipose stem cells and their paracrine factors are therapeutic for early retinal complications of diabetes in the Ins2 Akita mouse. Stem Cell Res Ther 2018; 9:322. [PMID: 30463601 PMCID: PMC6249931 DOI: 10.1186/s13287-018-1059-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/05/2018] [Accepted: 10/23/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Early-stage diabetic retinopathy (DR) is characterized by neurovascular defects. In this study, we hypothesized that human adipose-derived stem cells (ASCs) positive for the pericyte marker CD140b, or their secreted paracrine factors, therapeutically rescue early-stage DR features in an Ins2Akita mouse model. METHODS Ins2Akita mice at 24 weeks of age received intravitreal injections of CD140b-positive ASCs (1000 cells/1 μL) or 20× conditioned media from cytokine-primed ASCs (ASC-CM, 1 μL). Age-matched wildtype mice that received saline served as controls. Visual function experiments and histological analyses were performed 3 weeks post intravitreal injection. Biochemical and molecular analyses assessed the ASC-CM composition and its biological effects. RESULTS Three weeks post-injection, Ins2Akita mice that received ASCs had ameliorated decreased b-wave amplitudes and vascular leakage but failed to improve visual acuity, whereas Ins2Akita mice that received ASC-CM demonstrated amelioration of all aforementioned visual deficits. The ASC-CM group demonstrated partial amelioration of retinal GFAP immunoreactivity and DR-related gene expression but the ASC group did not. While Ins2Akita mice that received ASCs exhibited occasional (1 in 8) hemorrhagic retinas, mice that received ASC-CM had no adverse complications. In vitro, ASC-CM protected against TNFα-induced retinal endothelial permeability as measured by transendothelial electrical resistance. Biochemical and molecular analyses demonstrated several anti-inflammatory proteins including TSG-6 being highly expressed in cytokine-primed ASC-CM. CONCLUSIONS ASCs or their secreted factors mitigate retinal complications of diabetes in the Ins2Akita model. Further investigation is warranted to determine whether ASCs or their secreted factors are safe and effective therapeutic modalities long-term as current locally delivered therapies fail to effectively mitigate the progression of early-stage DR. Nonetheless, our study sheds new light on the therapeutic mechanisms of adult stem cells, with implications for assessing relative risks/benefits of experimental regenerative therapies for vision loss.
Collapse
Affiliation(s)
- Sally L. Elshaer
- Ophthalmology, University of Tennessee Health Science Center, 930 Madison Ave, Suite#768, Memphis, TN 38163 USA
- Pharmacology & Toxicology Department, College of Pharmacy, Mansoura University, Mansoura, Egypt
| | - William Evans
- Ophthalmology, University of Tennessee Health Science Center, 930 Madison Ave, Suite#768, Memphis, TN 38163 USA
| | | | - Raji Lenin
- Ophthalmology, University of Tennessee Health Science Center, 930 Madison Ave, Suite#768, Memphis, TN 38163 USA
| | - Ramesh Periasamy
- Ophthalmology, University of Tennessee Health Science Center, 930 Madison Ave, Suite#768, Memphis, TN 38163 USA
| | - Kumar Abhiram Jha
- Ophthalmology, University of Tennessee Health Science Center, 930 Madison Ave, Suite#768, Memphis, TN 38163 USA
| | - Shanta Alli
- Ophthalmology, University of Tennessee Health Science Center, 930 Madison Ave, Suite#768, Memphis, TN 38163 USA
| | - Jordy Gentry
- Ophthalmology, University of Tennessee Health Science Center, 930 Madison Ave, Suite#768, Memphis, TN 38163 USA
| | - Samuel M. Thomas
- Ophthalmology, University of Tennessee Health Science Center, 930 Madison Ave, Suite#768, Memphis, TN 38163 USA
| | - Nicolas Sohl
- Cell Care Therapeutics, Inc., Monrovia, CA 91016 USA
| | - Rajashekhar Gangaraju
- Ophthalmology, University of Tennessee Health Science Center, 930 Madison Ave, Suite#768, Memphis, TN 38163 USA
- Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163 USA
| |
Collapse
|
22
|
Sabbagh MF, Heng JS, Luo C, Castanon RG, Nery JR, Rattner A, Goff LA, Ecker JR, Nathans J. Transcriptional and epigenomic landscapes of CNS and non-CNS vascular endothelial cells. eLife 2018; 7:36187. [PMID: 30188322 PMCID: PMC6126923 DOI: 10.7554/elife.36187] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 08/21/2018] [Indexed: 02/06/2023] Open
Abstract
Vascular endothelial cell (EC) function depends on appropriate organ-specific molecular and cellular specializations. To explore genomic mechanisms that control this specialization, we have analyzed and compared the transcriptome, accessible chromatin, and DNA methylome landscapes from mouse brain, liver, lung, and kidney ECs. Analysis of transcription factor (TF) gene expression and TF motifs at candidate cis-regulatory elements reveals both shared and organ-specific EC regulatory networks. In the embryo, only those ECs that are adjacent to or within the central nervous system (CNS) exhibit canonical Wnt signaling, which correlates precisely with blood-brain barrier (BBB) differentiation and Zic3 expression. In the early postnatal brain, single-cell RNA-seq of purified ECs reveals (1) close relationships between veins and mitotic cells and between arteries and tip cells, (2) a division of capillary ECs into vein-like and artery-like classes, and (3) new endothelial subtype markers, including new validated tip cell markers.
Collapse
Affiliation(s)
- Mark F Sabbagh
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Jacob S Heng
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Chongyuan Luo
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, United States.,Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, United States
| | - Rosa G Castanon
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - Joseph R Nery
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - Amir Rattner
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Loyal A Goff
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Joseph R Ecker
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, United States.,Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, United States
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
23
|
Human notochordal cell transcriptome unveils potential regulators of cell function in the developing intervertebral disc. Sci Rep 2018; 8:12866. [PMID: 30150762 PMCID: PMC6110784 DOI: 10.1038/s41598-018-31172-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 08/01/2018] [Indexed: 11/08/2022] Open
Abstract
The adult nucleus pulposus originates from the embryonic notochord, but loss of notochordal cells with skeletal maturity in humans is thought to contribute to the onset of intervertebral disc degeneration. Thus, defining the phenotype of human embryonic/fetal notochordal cells is essential for understanding their roles and for development of novel therapies. However, a detailed transcriptomic profiling of human notochordal cells has never been achieved. In this study, the notochord-specific marker CD24 was used to specifically label and isolate (using FACS) notochordal cells from human embryonic and fetal spines (7.5–14 weeks post-conception). Microarray analysis and qPCR validation identified CD24, STMN2, RTN1, PRPH, CXCL12, IGF1, MAP1B, ISL1, CLDN1 and THBS2 as notochord-specific markers. Expression of these markers was confirmed in nucleus pulposus cells from aged and degenerate discs. Ingenuity pathway analysis revealed molecules involved in inhibition of vascularisation (WISP2, Noggin and EDN2) and inflammation (IL1-RN) to be master regulators of notochordal genes. Importantly, this study has, for the first time, defined the human notochordal cell transcriptome and suggests inhibition of inflammation and vascularisation may be key roles for notochordal cells during intervertebral disc development. The molecules and pathways identified in this study have potential for use in developing strategies to retard/prevent disc degeneration, or regenerate tissue.
Collapse
|
24
|
Alrashdi SF, Deliyanti D, Wilkinson-Berka JL. Intravitreal administration of endothelin type A receptor or endothelin type B receptor antagonists attenuates hypertensive and diabetic retinopathy in rats. Exp Eye Res 2018; 176:1-9. [PMID: 29944850 DOI: 10.1016/j.exer.2018.06.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/25/2018] [Accepted: 06/22/2018] [Indexed: 12/17/2022]
Abstract
Hypertension is an independent risk factor for diabetic retinopathy, yet anti-hypertensive medications such as blockade of angiotensin II do not completely protect against vision-threatening vascular disease. We hypothesized that the potent vasoactive factor, endothelin (ET), is up-regulated in diabetic retinopathy and antagonism of the ET type A receptor (ETRA) or ET type B receptor (ETRB) ameliorates retinal vascular leakage independently of any blood pressure lowering effects. Spontaneously hypertensive rats (SHR) and their normotensive and genetic controls, Wistar Kyoto rats, were randomized to become diabetic or non-diabetic and studied for 8 weeks. Rats were further randomized to receive by intravitreal injection the ETRA antagonist, BQ123, the ETRB antagonist, BQ788, or vehicle, 5 days after the induction of streptozotocin diabetes and 4 weeks later. The treatments had no effect on systolic blood pressure which remained elevated in SHR. ET-1, ET-2, ETRA and ETRB were expressed in retina and retinal pigment epithelium (RPE)/choroid and increased by hypertension or diabetes. BQ123 reduced ET-1 and ET-2 expression in retina and RPE/choroid, while BQ788 had a similar effect but did not influence the mRNA levels of ET-1 in retina. Retinal vascular leakage and Müller cell stress as well as vascular endothelial growth factor (VEGF) expression in retina and RPE/choroid, were increased by hypertension or diabetes and there was an additive effect of these conditions. Treatment with BQ123 or BQ788 effectively reduced these events as well as the elevated levels of inflammatory factors in the retina. Our findings indicate that local ET systems exist in the retina and RPE/choroid that are up-regulated by hypertension and diabetes. The ability of locally delivered ET receptor antagonists to supress these overactive ET systems and reduce retinal vascular leakage and VEGF in the presence of hypertension indicate the potential of these approaches for the treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- Saeed F Alrashdi
- Department of Diabetes, The Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Devy Deliyanti
- Department of Diabetes, The Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | | |
Collapse
|
25
|
Cho HJ, Heo W, Han JW, Lee YH, Park JM, Kang MJ, Yoon JH, Lee MG, Kim CH, Kim JY. Chronological Change of Right Ventricle by Chronic Intermittent Hypoxia in Mice. Sleep 2017. [PMID: 28637196 DOI: 10.1093/sleep/zsx103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Study Objective No studies have investigated sequential changes in the heart on magnetic resonance imaging (MRI), along with observation of functional lung phenotypes and genetics, over the duration of chronic intermittent hypoxia (CIH). We investigated chronological changes in heart and lung phenotypes after CIH using a mouse model to provide new insights into the pathophysiology of sleep apnea-induced cardiovascular disease. Methods C57BL/6J adult male mice were randomized to 4 or 8 weeks of CIH. Cardiac cine-MRI images were analyzed to assess functional parameters of right ventricle (RV). Histopathological features of myocytes and pulmonary vessels, as well as genes involved in the endothelin (ET) system, were investigated. Results Function of the RV reduced significantly at 4 weeks and continuously decreased following another 4 weeks of CIH, although the rate of decrease was attenuated. Notably, persistence of reduced ejection fraction and end-systole RV wall thickness (WT) and increases in the ET system of the lungs and blood strongly implied the development of pulmonary hypertension after 8 weeks of CIH. Conclusions RV dysfunction with reduced end-systole RV WT could be a late phenotype in long-standing CIH and possibly also in obstructive sleep apnea.
Collapse
Affiliation(s)
- Hyung-Ju Cho
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea.,The Airway Mucus Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Woon Heo
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Woo Han
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Hyuk Lee
- Research Center for Human Natural Defense System, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Myung Park
- Research Center for Human Natural Defense System, Yonsei University College of Medicine, Seoul, Korea
| | - Min Jung Kang
- Research Center for Human Natural Defense System, Yonsei University College of Medicine, Seoul, Korea
| | - Joo-Heon Yoon
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea.,The Airway Mucus Institute, Yonsei University College of Medicine, Seoul, Korea.,Research Center for Human Natural Defense System, Yonsei University College of Medicine, Seoul, Korea
| | - Min Goo Lee
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Chang-Hoon Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea.,The Airway Mucus Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Joo Young Kim
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
26
|
Cho C, Smallwood PM, Nathans J. Reck and Gpr124 Are Essential Receptor Cofactors for Wnt7a/Wnt7b-Specific Signaling in Mammalian CNS Angiogenesis and Blood-Brain Barrier Regulation. Neuron 2017; 95:1056-1073.e5. [PMID: 28803732 PMCID: PMC5586543 DOI: 10.1016/j.neuron.2017.07.031] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/14/2017] [Accepted: 07/26/2017] [Indexed: 10/19/2022]
Abstract
Reck, a GPI-anchored membrane protein, and Gpr124, an orphan GPCR, have been implicated in Wnt7a/Wnt7b signaling in the CNS vasculature. We show here that vascular endothelial cell (EC)-specific reduction in Reck impairs CNS angiogenesis and that EC-specific postnatal loss of Reck, combined with loss of Norrin, impairs blood-brain barrier (BBB) maintenance. The most N-terminal domain of Reck binds to the leucine-rich repeat (LRR) and immunoglobulin (Ig) domains of Gpr124, and weakening this interaction by targeted mutagenesis reduces Reck/Gpr124 stimulation of Wnt7a signaling in cell culture and impairs CNS angiogenesis. Finally, a soluble Gpr124(LRR-Ig) probe binds to cells expressing Frizzled, Wnt7a or Wnt7b, and Reck, and a soluble Reck(CC1-5) probe binds to cells expressing Frizzled, Wnt7a or Wnt7b, and Gpr124. These experiments indicate that Reck and Gpr124 are part of the cell surface protein complex that transduces Wnt7a- and Wnt7b-specific signals in mammalian CNS ECs to promote angiogenesis and regulate the BBB.
Collapse
Affiliation(s)
- Chris Cho
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Philip M Smallwood
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
27
|
Shah RS, Soetikno BT, Yi J, Liu W, Skondra D, Zhang HF, Fawzi AA. Visible-Light Optical Coherence Tomography Angiography for Monitoring Laser-Induced Choroidal Neovascularization in Mice. Invest Ophthalmol Vis Sci 2017; 57:OCT86-95. [PMID: 27409510 PMCID: PMC4968775 DOI: 10.1167/iovs.15-18891] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose This study sought to determine the earliest time-point at which evidence of choroidal neovascularization (CNV) could be detected with visible-light optical coherence tomography angiography (vis-OCTA) in a mouse model of laser-induced CNV. Methods Visible light-OCTA was used to study laser-induced CNV at different time-points after laser injury to monitor CNV development and measure CNV lesion size. Measurements obtained from vis-OCTA angiograms were compared with histopathologic measurements from isolectin-stained choroidal flatmounts. Results Choroidal neovascularization area measurements between the vis-OCTA system and isolectin-stained choroidal flatmounts were significantly different in area for days 2 to 4 postlaser injury, and were not significantly different in area for days 5, 7, and 14. Choroidal neovascularization area measurements taken from the stained flatmounts were larger than their vis-OCTA counterparts for all time-points. Both modalities showed a similar trend of CNV size increasing from the day of laser injury until a peak of day 7 postlaser injury and subsequently decreasing by day 14. Conclusions The earliest vis-OCTA can detect the presence of aberrant vessels in a mouse laser-induced CNV model is 5 days after laser injury. Visible light-OCTA was able to visualize the maximum of the CNV network 7 days postlaser injury, in accordance with choroidal flatmount immunostaining. Visible light-OCTA is a reliable tool in both detecting the presence of CNV development, as well as accurately determining the size of the lesion in a mouse laser-induced CNV model.
Collapse
Affiliation(s)
- Ronil S Shah
- Department of Ophthalmology Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Brian T Soetikno
- Department of Ophthalmology Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States 2Functional Optical Imaging Laboratory, Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States 3Med
| | - Ji Yi
- Functional Optical Imaging Laboratory, Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| | - Wenzhong Liu
- Functional Optical Imaging Laboratory, Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| | - Dimitra Skondra
- Department of Ophthalmology Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Hao F Zhang
- Department of Ophthalmology Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States 2Functional Optical Imaging Laboratory, Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| | - Amani A Fawzi
- Department of Ophthalmology Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| |
Collapse
|
28
|
Cacioppo JA, Lin PCP, Hannon PR, McDougle DR, Gal A, Ko C. Granulosa cell endothelin-2 expression is fundamental for ovulatory follicle rupture. Sci Rep 2017; 7:817. [PMID: 28400616 PMCID: PMC5429765 DOI: 10.1038/s41598-017-00943-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/20/2017] [Indexed: 12/22/2022] Open
Abstract
Ovulation is dependent upon numerous factors mediating follicular growth, vascularization, and ultimately oocyte release via follicle rupture. Endothelin-2 (EDN2) is a potent vasoconstrictor that is transiently produced prior to follicle rupture by granulosa cells of periovulatory follicles and induces ovarian contraction. To determine the role of Edn2 expression, surgical transplant and novel conditional knockout mice were super-ovulated and analyzed. Conditional knockout mice utilized a new iCre driven by the Esr2 promoter to selectively remove Edn2. Follicle rupture and fertility were significantly impaired in the absence of ovarian Edn2 expression. When ovaries of Edn2KO mice were transplanted in wild type recipients, significantly more corpora lutea containing un-ovulated oocytes were present after hormonal stimulation (1.0 vs. 5.4, p = 0.010). Following selective ablation of Edn2 in granulosa cells, Esr2-Edn2KO dams had reduced oocytes ovulated (3.8 vs. 16.4 oocytes/ovary) and smaller litters (4.29 ± l.02 vs. 8.50 pups/dam). However, the number of pregnancies per pairing was not different and the reproductive axis remained intact. Esr2-Edn2KO ovaries had a higher percentage of antral follicles and fewer corpora lutea; follicles progressed to the antral stage but many were unable to rupture. Conditional loss of endothelin receptor A in granulosa cells also decreased ovulation but did not affect fecundity. These data demonstrate that EDN2-induced intraovarian contraction is a critical trigger of normal ovulation and subsequent fecundity.
Collapse
Affiliation(s)
- Joseph A Cacioppo
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Po-Ching Patrick Lin
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Patrick R Hannon
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA.,Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY, 40536, USA
| | - Daniel R McDougle
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Arnon Gal
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA.,Department of Small Animal Internal Medicine, Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Palmerston North, 4442, New Zealand
| | - CheMyong Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA.
| |
Collapse
|
29
|
Abstract
Müller glia (MG) are the only glial cell type produced by the neuroepithelial progenitor cells that generate the vertebrate retina. MG are required to maintain retinal homeostasis and support the survival of retinal neurons. Furthermore, in certain vertebrate classes, MG function as adult stem cells, mediating retinal regeneration in response to injury. However, the mechanisms that regulate MG development are poorly understood because there is considerable overlap in gene expression between retinal progenitor cells and differentiated MG. We show that the LIM homeodomain transcription factor Lhx2 is required for the development of MG in the mouse retina. Temporally controlled knock-out studies reveal a requirement for Lhx2 during all stages of MG development, ranging from the proliferation of gliocompetent retinal progenitors, activation of Müller-specific gene expression, and terminal differentiation of MG morphological features. We show that Lhx2 regulates gliogenesis in part by regulating directly the expression of Notch pathway genes including Notch1, Dll1, and Dll3 and gliogenic transcription factors such as Hes1, Hes5, Sox8, and Rax. Conditional knock-out of Lhx2 resulted in a rapid downregulation of Notch pathway genes and loss of Notch signaling. We further demonstrate that Müller gliogenesis induced by misexpression of the potently gliogenic Notch pathway transcriptional effector Hes5 requires Lhx2 expression. These results indicate that Lhx2 not only directly regulates expression of Notch signaling pathway components, but also acts together with the gliogenic Notch pathway to drive MG specification and differentiation.
Collapse
|
30
|
Zhou Y, Williams J, Smallwood PM, Nathans J. Sox7, Sox17, and Sox18 Cooperatively Regulate Vascular Development in the Mouse Retina. PLoS One 2015; 10:e0143650. [PMID: 26630461 PMCID: PMC4667919 DOI: 10.1371/journal.pone.0143650] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 11/06/2015] [Indexed: 02/06/2023] Open
Abstract
Vascular development and maintenance are controlled by a complex transcriptional program, which integrates both extracellular and intracellular signals in endothelial cells. Here we study the roles of three closely related SoxF family transcription factors–Sox7, Sox17, and Sox18 –in the developing and mature mouse vasculature using targeted gene deletion on a mixed C57/129/CD1 genetic background. In the retinal vasculature, each SoxF gene exhibits a distinctive pattern of expression in different classes of blood vessels. On a mixed genetic background, vascular endothelial-specific deletion of individual SoxF genes has little or no effect on vascular architecture or differentiation, a result that can be explained by overlapping function and by reciprocal regulation of gene expression between Sox7 and Sox17. By contrast, combined deletion of Sox7, Sox17, and Sox18 at the onset of retinal angiogenesis leads to a dense capillary plexus with a nearly complete loss of radial arteries and veins, whereas the presence of a single Sox17 allele largely restores arterial identity, as determined by vascular smooth muscle cell coverage. In the developing retina, expression of all three SoxF genes is reduced in the absence of Norrin/Frizzled4-mediated canonical Wnt signaling, but SoxF gene expression is unaffected by reduced VEGF signaling in response to deletion of Neuropilin1 (Npn1). In adulthood, Sox7, Sox17, and Sox18 act in a largely redundant manner to maintain blood vessel function, as adult onset vascular endothelial-specific deletion of all three SoxF genes leads to massive edema despite nearly normal vascular architecture. These data reveal critical and partially redundant roles for Sox7, Sox17 and Sox18 in vascular growth, differentiation, and maintenance.
Collapse
Affiliation(s)
- Yulian Zhou
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - John Williams
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Philip M. Smallwood
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Jeremy Nathans
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- * E-mail:
| |
Collapse
|
31
|
Nrf2 in ischemic neurons promotes retinal vascular regeneration through regulation of semaphorin 6A. Proc Natl Acad Sci U S A 2015; 112:E6927-36. [PMID: 26621751 DOI: 10.1073/pnas.1512683112] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Delayed revascularization of ischemic neural tissue is a major impediment to preservation of function in central nervous system (CNS) diseases including stroke and ischemic retinopathies. Therapeutic strategies allowing rapid revascularization are greatly needed to reduce ischemia-induced cellular damage and suppress harmful pathologic neovascularization. However, key mechanisms governing vascular recovery in ischemic CNS, including regulatory molecules governing the transition from tissue injury to tissue repair, are largely unknown. NF-E2-related factor 2 (Nrf2) is a major stress-response transcription factor well known for its cell-intrinsic cytoprotective function. However, its role in cell-cell crosstalk is less appreciated. Here we report that Nrf2 is highly activated in ischemic retina and promotes revascularization by modulating neurons in their paracrine regulation of endothelial cells. Global Nrf2 deficiency strongly suppresses retinal revascularization and increases pathologic neovascularization in a mouse model of ischemic retinopathy. Conditional knockout studies demonstrate a major role for neuronal Nrf2 in vascular regrowth into avascular retina. Deletion of neuronal Nrf2 results in semaphorin 6A (Sema6A) induction in hypoxic/ischemic retinal ganglion cells in a hypoxia-inducible factor-1 alpha (HIF-1α)-dependent fashion. Sema6A expression increases in avascular inner retina and colocalizes with Nrf2 in human fetal eyes. Extracellular Sema6A leads to dose-dependent suppression of the migratory phenotype of endothelial cells through activation of Notch signaling. Lentiviral-mediated delivery of Sema6A small hairpin RNA (shRNA) abrogates the defective retinal revascularization in Nrf2-deficient mice. Importantly, pharmacologic Nrf2 activation promotes reparative angiogenesis and suppresses pathologic neovascularization. Our findings reveal a unique function of Nrf2 in reprogramming ischemic tissue toward neurovascular repair via Sema6A regulation, providing a potential therapeutic strategy for ischemic retinal and CNS diseases.
Collapse
|
32
|
Hua ZL, Emiliani FE, Nathans J. Rac1 plays an essential role in axon growth and guidance and in neuronal survival in the central and peripheral nervous systems. Neural Dev 2015; 10:21. [PMID: 26395878 PMCID: PMC4580344 DOI: 10.1186/s13064-015-0049-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 09/04/2015] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Rac1 is a critical regulator of cytoskeletal dynamics in multiple cell types. In the nervous system, it has been implicated in the control of cell proliferation, neuronal migration, and axon development. RESULTS To systematically investigate the role of Rac1 in axon growth and guidance in the developing nervous system, we have examined the phenotypes associated with deleting Rac1 in the embryonic mouse forebrain, in cranial and spinal motor neurons, in cranial sensory and dorsal root ganglion neurons, and in the retina. We observe a widespread requirement for Rac1 in axon growth and guidance and a cell-autonomous defect in axon growth in Rac1 (-/-) motor neurons in culture. Neuronal death, presumably a secondary consequence of the axon growth and/or guidance defects, was observed in multiple locations. Following deletion of Rac1 in the forebrain, thalamocortical axons were misrouted inferiorly, with the majority projecting to the contralateral thalamus and a minority projecting ipsilaterally to the ventral cortex, a pattern of misrouting that is indistinguishable from the pattern previously observed in Frizzled3 (-/-) and Celsr3 (-/-) forebrains. In the limbs, motor-neuron-specific deletion of Rac1 produced a distinctive stalling of axons within the dorsal nerve of the hindlimb but a much milder loss of axons in the ventral hindlimb and forelimb nerves, a pattern that is virtually identical to the one previously observed in Frizzled3 (-/-) limbs. CONCLUSIONS The similarities in axon growth and guidance phenotypes caused by Rac1, Frizzled3, and Celsr3 loss-of-function mutations suggest a mechanistic connection between tissue polarity/planar cell polarity signaling and Rac1-dependent cytoskeletal regulation.
Collapse
Affiliation(s)
- Zhong L Hua
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Present address: Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA.
| | - Francesco E Emiliani
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
33
|
Li H, Gong Y, Qian H, Chen T, Liu Z, Jiang Z, Wei S. Brain-derived neurotrophic factor is a novel target gene of the has-miR-183/96/182 cluster in retinal pigment epithelial cells following visible light exposure. Mol Med Rep 2015; 12:2793-9. [PMID: 25955435 DOI: 10.3892/mmr.2015.3736] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 12/09/2014] [Indexed: 11/06/2022] Open
Abstract
Light-induced retinal injury is clinically and experimentally well-documented. It may be categorized into three types: Photothermal, photomechanical and photochemical injuries. To date, the variation in the hsa-miR-183/96/182 cluster and its potential target genes in human primary retinal pigment epithelial (RPE) cells, following visible light exposure, has not been reported. In the present study, RPE cells were exposed to 4 h of constant visible light. The expression of the hsa-miR-183/96/182 cluster was determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and its potential target genes were investigated. Additionally, hsa-miR-183, hsa-miR-96, hsa-miR-182 and has-miR-183/96/182 mimics were designed and synthesized in vitro, and transfected into the RPE cells. Subsequently, the expression of brain-derived neurotrophic factor (BDNF) mRNA and protein was measured, using RT-qPCR and western blotting, respectively. The regulation of miRNAs to the BDNF gene were then validated using a dual luciferase reporter gene assay system. The expression of hsa-miR-183, hsa-miR-96 and hsa-miR-182 significantly increased in RPE cells following 4 h of visible light exposure, compared with RPE cells that had been exposed to dark conditions (P<0.01). Following RPE cell transfection with mimics, BDNF mRNA and protein expression in the RPE cells was significantly downregulated compared with control RPE cells (P<0.05, P<0.01, respectively). Similarly, the ratio of Renilla luciferase/firefly luciferase significantly decreased in the RPE cells of the mimic + wild type (WT) group compared with cells of the psiCHECK(TM)-2 (a vector lacking the sequence of the BDNF gene), wild type and mimic + mutation groups (P<0.05, P<0.01). The present study suggests that BDNF is a target gene of the has-miR-183-96-182 cluster in RPE cells. The present study suggests an underlying protective mechanism against retinal light injury and may provide a novel target for the prevention and treatment of light-induced retinal injury.
Collapse
Affiliation(s)
- Hongyang Li
- Department of Opthalmology, The Chinese People's Liberation Army General Hospital, Beijing 100000, P.R. China
| | - Yan Gong
- Department of Opthalmology, The Chinese People's Liberation Army General Hospital, Beijing 100000, P.R. China
| | - Haiyan Qian
- Department of Opthalmology, The Chinese People's Liberation Army General Hospital, Beijing 100000, P.R. China
| | - Tingjun Chen
- Department of Opthalmology, The Chinese People's Liberation Army General Hospital, Beijing 100000, P.R. China
| | - Zihao Liu
- Department of Opthalmology, The Chinese People's Liberation Army General Hospital, Beijing 100000, P.R. China
| | - Zhaocai Jiang
- Department of Opthalmology, The Chinese People's Liberation Army General Hospital, Beijing 100000, P.R. China
| | - Shihui Wei
- Department of Opthalmology, The Chinese People's Liberation Army General Hospital, Beijing 100000, P.R. China
| |
Collapse
|
34
|
Cacioppo JA, Koo Y, Lin PCP, Gal A, Ko C. Generation and characterization of an endothelin-2 iCre mouse. Genesis 2015; 53:245-56. [PMID: 25604013 DOI: 10.1002/dvg.22845] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/14/2015] [Accepted: 01/15/2015] [Indexed: 12/16/2022]
Abstract
A novel transgenic mouse line that expresses codon-improved Cre recombinase (iCre) under regulation of the Endothelin-2 gene (edn2) promoter was developed for the conditional deletion of genes in Endothelin-2 lineage cells and for the spatial and temporal localization of Endothelin-2 expression. Endothelin-2 (EDN2, ET-2, previously VIC) is a transcriptionally regulated 21 amino acid peptide implicated in vascular homeostasis, and more recently in female reproduction, gastrointestinal function, immunology, and cancer pathogenesis that acts through membrane receptors and G-protein signaling. A cassette (edn2-iCre) was constructed that contained iCre, a polyadenylation sequence, and a neomycin selection marker in front of the endogenous start codon of the edn2 gene in a mouse genome BAC clone. The cassette was introduced into the C57BL/6 genome by pronuclear injection, and two lines of edn2-iCre positive mice were produced. The edn2-iCre mice were bred with ROSA26-lacZ and Ai9 reporter mice to visualize areas of functional iCre expression. Strong expression was seen in the periovulatory ovary, stomach and small intestine, and colon. Uniquely, we report punctate expression in the corneal epithelium, the liver, the lung, the pituitary, the uterus, and the heart. In the embryo, expression is localized in developing hair follicles and the dermis. Therefore, edn2-iCre mice will serve as a novel line for conditional gene deletion in these tissues.
Collapse
Affiliation(s)
- Joseph A Cacioppo
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | | | | | | | | |
Collapse
|
35
|
Maguire JJ, Davenport AP. Endothelin@25 - new agonists, antagonists, inhibitors and emerging research frontiers: IUPHAR Review 12. Br J Pharmacol 2014; 171:5555-72. [PMID: 25131455 PMCID: PMC4290702 DOI: 10.1111/bph.12874] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/22/2014] [Accepted: 07/25/2014] [Indexed: 12/16/2022] Open
Abstract
Since the discovery of endothelin (ET)-1 in 1988, the main components of the signalling pathway have become established, comprising three structurally similar endogenous 21-amino acid peptides, ET-1, ET-2 and ET-3, that activate two GPCRs, ETA and ETB . Our aim in this review is to highlight the recent progress in ET research. The ET-like domain peptide, corresponding to prepro-ET-193-166 , has been proposed to be co-synthesized and released with ET-1, to modulate the actions of the peptide. ET-1 remains the most potent vasoconstrictor in the human cardiovascular system with a particularly long-lasting action. To date, the major therapeutic strategy to block the unwanted actions of ET in disease, principally in pulmonary arterial hypertension, has been to use antagonists that are selective for the ETA receptor (ambrisentan) or that block both receptor subtypes (bosentan). Macitentan represents the next generation of antagonists, being more potent than bosentan, with longer receptor occupancy and it is converted to an active metabolite; properties contributing to greater pharmacodynamic and pharmacokinetic efficacy. A second strategy is now being more widely tested in clinical trials and uses combined inhibitors of ET-converting enzyme and neutral endopeptidase such as SLV306 (daglutril). A third strategy based on activating the ETB receptor, has led to the renaissance of the modified peptide agonist IRL1620 as a clinical candidate in delivering anti-tumour drugs and as a pharmacological tool to investigate experimental pathophysiological conditions. Finally, we discuss biased signalling, epigenetic regulation and targeting with monoclonal antibodies as prospective new areas for ET research.
Collapse
Affiliation(s)
- J J Maguire
- Clinical Pharmacology Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
36
|
Rattner A, Wang Y, Zhou Y, Williams J, Nathans J. The role of the hypoxia response in shaping retinal vascular development in the absence of Norrin/Frizzled4 signaling. Invest Ophthalmol Vis Sci 2014; 55:8614-25. [PMID: 25414188 DOI: 10.1167/iovs.14-15693] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE To define the role of hypoxia and vascular endothelial growth factor (VEGF) in modifying the pattern, density, and permeability of the retinal vasculature in mouse models in which Norrin/Frizzled4 signaling is impaired. METHODS Retinal vascular structure was analyzed in mice with mutation of Ndp (the gene coding for Norrin) or Frizzle4 (Fz4) with or without three additional perturbations: (1) retinal hyperoxia and reduction of VEGF, (2) reduced induction of VEGF in response to hypoxia, or (3) reduced responsiveness of vascular endothelial cells (ECs) to VEGF. These perturbations were produced, respectively, by (1) genetic ablation of rod photoreceptors in the retinal degeneration 1 (rd1) mutant background, (2) conditional deletion of the gene coding for hypoxia-inducible factor (HIF)-2alpha either in all neural retina cells or specifically in Müller glia, and (3) conditional deletion of the VEGF coreceptor neuropilin1 (NRP1) in ECs. RESULTS All three conditions reduced vascular proliferation. Eliminating HIF2-alpha in Müller glia blocked VEGF induction in the inner nuclear layer, identifying HIF2-alpha as the transcription factor responsible for the hypoxia response in these cells. When Norrin/Frizzled4 signaling was eliminated, a secondary elevation in VEGF levels was required to compromise the barrier to transendothelial movement of high molecular weight compounds. CONCLUSIONS In the absence of Norrin or Frizzled4, the vascular phenotype is determined by the primary defect in Norrin/Frizzled4 signaling (i.e., canonical Wnt signaling) and compensatory responses resulting from hypoxia. This work may be useful in guiding therapeutic strategies for the treatment of familial exudative vitreoretinopathy (FEVR).
Collapse
Affiliation(s)
- Amir Rattner
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Yanshu Wang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Yulian Zhou
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - John Williams
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
37
|
Activation of the endothelin system mediates pathological angiogenesis during ischemic retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3040-51. [PMID: 25203536 DOI: 10.1016/j.ajpath.2014.07.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/10/2014] [Accepted: 07/15/2014] [Indexed: 02/08/2023]
Abstract
Retinopathy of prematurity adversely affects premature infants because of oxygen-induced damage of the immature retinal vasculature, resulting in pathological neovascularization (NV). Our pilot studies using the mouse model of oxygen-induced retinopathy (OIR) showed marked increases in angiogenic mediators, including endothelins and endothelin receptor (EDNR) A. We hypothesized that activation of the endothelin system via EDNRA plays a causal role in pathological angiogenesis and up-regulation of angiogenic mediators, including vascular endothelial growth factor A (VEGFA) in OIR. Mice were exposed to 75% oxygen from post-natal day P7 to P12, treated with either vehicle or EDNRA antagonist BQ-123 or EDNRB antagonist BQ-788 on P12, and kept at room air from P12 to P17 (ischemic phase). RT-PCR analysis revealed increased levels of EDN2 and EDNRA mRNA, and Western blot analysis revealed increased EDN2 expression during the ischemic phase. EDNRA inhibition significantly increased vessel sprouting, resulting in enhanced physiological angiogenesis and decreased pathological NV, whereas EDNRB inhibition modestly improved vascular repair. OIR triggered significant increases in VEGFA protein and mRNA for delta-like ligand 4, apelin, angiopoietin-2, and monocyte chemoattractant protein-1. BQ-123 treatment significantly reduced these alterations. EDN2 expression was localized to retinal glia and pathological NV tufts of the OIR retinas. EDN2 also induced VEGFA protein expression in cultured astrocytes. In conclusion, inhibition of the EDNRA during OIR suppresses pathological NV and promotes physiological angiogenesis.
Collapse
|
38
|
Venous endothelin guides sympathetic innervation of the developing mouse heart. Nat Commun 2014; 5:3918. [PMID: 24875861 PMCID: PMC4080092 DOI: 10.1038/ncomms4918] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 04/18/2014] [Indexed: 01/09/2023] Open
Abstract
The mechanisms responsible for establishing correct target innervation during organ development are largely unknown. Sympathetic nerves traverse or follow blood vessels to reach their end-organs, suggesting the existence of vascular guidance cues that direct axonal extension. The sinoatrial node and the ventricle of the heart receive sympathetic innervation from the stellate ganglia (STG). Here we show that STG axons follow veins, specifically the superior vena cavae and sinus venosus, to reach these targets. We find that the election of these routes is determined by venous endothelium-derived endothelin-1, acting through its specific receptor Ednra expressed within a subpopulation of STG neurons. Furthermore, we demonstrate that Edn1-Ednra signaling is essential for functional regulation of the heart by sympathetic nerves. Our findings present venous Edn1 as a sympathetic guidance cue, and show how axon guidance mechanisms are coordinated with end-organ morphogenesis.
Collapse
|
39
|
Cacioppo JA, Oh SW, Kim HY, Cho J, Lin PCP, Yanagisawa M, Ko C. Loss of function of endothelin-2 leads to reduced ovulation and CL formation. PLoS One 2014; 9:e96115. [PMID: 24763822 PMCID: PMC3999112 DOI: 10.1371/journal.pone.0096115] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 04/03/2014] [Indexed: 12/17/2022] Open
Abstract
Endothelin-2 (EDN2), a potent vasoconstrictive peptide, is transiently produced by periovulatory follicles at the time of ovulation when corpus luteum (CL) formation begins. EDN2 induces contraction of ovarian smooth muscles ex vivo via an endothelin receptor A-mediated pathway. In this study, we aimed to determine if EDN2 is required for normal ovulation and subsequent CL formation in?vivo. In the ovaries of a mouse model that globally lacks the Edn2 gene (Edn2 knockout mouse; Edn2KO), histology showed that post-pubertal Edn2KO mice possess follicles of all developmental stages, but no corpora lutea. When exogenous gonadotropins were injected to induce super-ovulation, Edn2KO mice exhibited significantly impaired ovulation and CL formation compared to control littermates. Edn2KO ovaries that did ovulate in response to gonadotropins did not contain histologically and functionally identifiable CL. Intra-ovarian injection of EDN2 peptide results suggest partial induction of ovulation in Edn2KO mice. Endothelin receptor antagonism in wild type mice similarly disrupted ovulation, CL formation, and progesterone secretion. Overall, this study suggests that EDN2 is necessary for normal ovulation and CL formation.
Collapse
Affiliation(s)
- Joseph A. Cacioppo
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Champaign, Illinois, United States of America
| | - Sang Wook Oh
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Champaign, Illinois, United States of America
- Department of Biology Education, Institute of Fusion Science, Chonbuk National University, Jeonju, South Korea
| | - Hey-young Kim
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Champaign, Illinois, United States of America
| | - Jongki Cho
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Champaign, Illinois, United States of America
- College of Veterinary Medicine, Research Institute of Veterinary Medicine, Chungnam National University, Daejon, South Korea
| | - Po-Ching Patrick Lin
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Champaign, Illinois, United States of America
| | - Masashi Yanagisawa
- Howard Hughes Medical Institute and Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - CheMyong Ko
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Champaign, Illinois, United States of America
- * E-mail:
| |
Collapse
|