1
|
Yang M, Qiu R, Jin X, Yao S, Wang W, Liu J, Liu G, Han J, Lei B. Proteomics identifies multiple retinitis pigmentosa associated proteins involved in retinal degeneration in a mouse model bearing a Pde6b mutation. Sci Rep 2024; 14:22090. [PMID: 39333705 PMCID: PMC11437026 DOI: 10.1038/s41598-024-72821-1] [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: 04/19/2024] [Accepted: 09/10/2024] [Indexed: 09/29/2024] Open
Abstract
Retinitis pigmentosa (RP) is a progressive and degenerative retinal disease resulting in severe vision loss. RP have been extensively studied for pathogenetic mechanisms and treatments. Yet there is little information about alterations of RP associated proteins in phosphodiesterase 6 beta (Pde6b) mutated model. To explore the roles of RP causing proteins, we performed a label free quantitative mass spectrometry based proteomic analysis in rd10 mouse retinas. 3737 proteins were identified at the degenerative time points in rd10 mice. 222 and 289 differentially expressed proteins (DEPs) (fold change, FC > 2, p < 0.05) were detected at 5 and 8 weeks. Based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, visual perception and phototransduction were severely affected. The downregulated DEPs were significantly enriched in cilium assembly and protein localization. 25 decreased DEPs causing autosomal recessive/dominant retinitis pigmentosa were visualized by heatmaps. Protein-protein interaction network represented 13 DEPs interacted directly with Pde6b protein. 25 DEPs causing RP were involved in phototransduction, visual perception, response to stimulus, protein localization and cilium assembly pathways. The significantly reduced expressions of DEPs were further validated by quantitative reverse transcription polymerase chain reaction (qPCR), Western blots (WB) and immunohistochemistry (IHC). This study revealed the molecular mechanisms underlying early and late stage of RP, as well as changes of RP-causing proteins.
Collapse
Affiliation(s)
- Mingzhu Yang
- Henan Provincial People's Hospital, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450003, China
| | - Ruiqi Qiu
- Henan Provincial People's Hospital, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450003, China
| | - Xiuxiu Jin
- Henan Provincial People's Hospital, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450003, China
| | - Shun Yao
- Henan Provincial People's Hospital, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450003, China
| | - Weiping Wang
- Henan Provincial People's Hospital, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450003, China
| | - Jingyang Liu
- Henan Provincial People's Hospital, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450003, China
| | - Guangming Liu
- Henan Provincial People's Hospital, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450003, China
| | - Jinfeng Han
- Henan Provincial People's Hospital, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
- Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Bo Lei
- Henan Provincial People's Hospital, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450003, China.
- Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.
| |
Collapse
|
2
|
Ruan X, Xiong Y, Li X, Yang E, Wang J. Lower ratio of IMPDH1 to IMPDH2 sensitizes gliomas to chemotherapy. Cancer Gene Ther 2024; 31:1081-1089. [PMID: 38871858 DOI: 10.1038/s41417-024-00793-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Gliomas are the most common primary tumors of the central nervous system, with approximately half of patients presenting with the most aggressive form of glioblastoma. Although several molecular markers for glioma have been identified, they are not sufficient to predict the prognosis due to the extensive genetic heterogeneity within glioma. Our study reveals that the ratio of IMPDH1 to IMPDH2 expression levels serves as a molecular indicator for glioma treatment prognosis. Patients with a higher IMPDH1/IMPDH2 ratio exhibit a worse prognosis, while those with a lower ratio display a more favorable prognosis. We further demonstrate that IMPDH1 plays a crucial role in maintaining cellular GTP/GDP levels following DNA damage compared to IMPDH2. In the absence of IMPDH1, cells experience an imbalance in the GTP/GDP ratio, impairing DNA damage repair capabilities and rendering them more sensitive to TMZ. This study not only introduces a novel prognostic indicator for glioma clinical diagnosis but also offers innovative insights for precise and stratified glioma treatment.
Collapse
Affiliation(s)
- Xiaoyu Ruan
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, 100191, Beijing, China
| | - Yundong Xiong
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, 100191, Beijing, China
| | - Xiaoman Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, 100191, Beijing, China.
| | - Ence Yang
- Department of Medical Bioinformatics, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China.
| | - Jiadong Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, 100191, Beijing, China.
- Department of Gastrointestinal Translational Research, Peking University Cancer Hospital, 100142, Beijing, China.
| |
Collapse
|
3
|
Pérez O, Stanzani A, Huang L, Schipper N, Loftsson T, Bollmark M, Marigo V. New Improved cGMP Analogues to Target Rod Photoreceptor Degeneration. J Med Chem 2024; 67:8396-8405. [PMID: 38688030 PMCID: PMC11129186 DOI: 10.1021/acs.jmedchem.4c00586] [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: 03/11/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024]
Abstract
Retinitis pigmentosa (RP) is a form of retinal degeneration affecting a young population with an unmet medical need. Photoreceptor degeneration has been associated with increased guanosine 3',5'-cyclic monophosphate (cGMP), which reaches toxic levels for photoreceptors. Therefore, inhibitory cGMP analogues attract interest for RP treatments. Here we present the synthesis of dithio-CN03, a phosphorodithioate analogue of cGMP, prepared using the H-phosphonothioate route. Two crystal modifications were identified as a trihydrate and a tetrahydrofuran monosolvates. Dithio-CN03 featured a lower aqueous solubility than its RP-phosphorothioate counterpart CN03, a drug candidate, and this characteristic might be favorable for sustained-release formulations aimed at retinal delivery. Dithio-CN03 was tested in vitro for its neuroprotective effects in photoreceptor models of RP. The comparison of dithio-CN03 to CN03 and its diastereomer SP-CN03, and to their phosphate derivative oxo-CN03 identifies dithio-CN03 as the compound with the highest efficacy in neuroprotection and thus as a promising new candidate for the treatment of RP.
Collapse
Affiliation(s)
- Oswaldo Pérez
- Chemical
Processes and Pharmaceutical Development Research Institutes of Sweden, Forskargatan 20 J, 15136 Södertälje, Sweden
- Faculty
of Pharmaceutical Sciences, University of
Iceland, Hofsvallagata
53, 107 Reykjavik, Iceland
| | - Agnese Stanzani
- Department
of Life Sciences, University of Modena and
Reggio Emilia, via Campi 287, 41125 Modena, Italy
| | - Li Huang
- Department
of Life Sciences, University of Modena and
Reggio Emilia, via Campi 287, 41125 Modena, Italy
| | - Nicolaas Schipper
- Chemical
Processes and Pharmaceutical Development Research Institutes of Sweden, Forskargatan 20 J, 15136 Södertälje, Sweden
| | - Thorsteinn Loftsson
- Faculty
of Pharmaceutical Sciences, University of
Iceland, Hofsvallagata
53, 107 Reykjavik, Iceland
| | - Martin Bollmark
- Chemical
Processes and Pharmaceutical Development Research Institutes of Sweden, Forskargatan 20 J, 15136 Södertälje, Sweden
| | - Valeria Marigo
- Department
of Life Sciences, University of Modena and
Reggio Emilia, via Campi 287, 41125 Modena, Italy
| |
Collapse
|
4
|
Pliushcheuskaya P, Kesh S, Kaufmann E, Wucherpfennig S, Schwede F, Künze G, Nache V. Similar Binding Modes of cGMP Analogues Limit Selectivity in Modulating Retinal CNG Channels via the Cyclic Nucleotide-Binding Domain. ACS Chem Neurosci 2024; 15:1652-1668. [PMID: 38579109 PMCID: PMC11027099 DOI: 10.1021/acschemneuro.3c00665] [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: 10/16/2023] [Revised: 02/28/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024] Open
Abstract
In treating retinitis pigmentosa, a genetic disorder causing progressive vision loss, selective inhibition of rod cyclic nucleotide-gated (CNG) channels holds promise. Blocking the increased Ca2+-influx in rod photoreceptors through CNG channels can potentially delay disease progression and improve the quality of life for patients. To find inhibitors for rod CNG channels, we investigated the impact of 16 cGMP analogues on both rod and cone CNG channels using the patch-clamp technique. Although modifications at the C8 position of the guanine ring did not change the ligand efficacy, modifications at the N1 and N2 positions rendered cGMP largely ineffective in activating retinal CNG channels. Notably, PET-cGMP displayed selective potential, favoring rod over cone, whereas Rp-cGMPS showed greater efficiency in activating cone over rod CNG channels. Ligand docking and molecular dynamics simulations on cyclic nucleotide-binding domains showed comparable binding energies and binding modes for cGMP and its analogues in both rod and cone CNG channels (CNGA1 vs CNGA3 subunits). Computational experiments on CNGB1a vs CNGB3 subunits showed similar binding modes albeit with fewer amino acid interactions with cGMP due to an inactivated conformation of their C-helix. In addition, no clear correlation could be observed between the computational scores and the CNG channel efficacy values, suggesting additional factors beyond binding strength determining ligand selectivity and potency. This study highlights the importance of looking beyond the cyclic nucleotide-binding domain and toward the gating mechanism when searching for selective modulators. Future efforts in developing selective modulators for CNG channels should prioritize targeting alternative channel domains.
Collapse
Affiliation(s)
- Palina Pliushcheuskaya
- Institute
for Drug Discovery, Medical Faculty, University
of Leipzig, Leipzig 04103, Germany
| | - Sandeep Kesh
- Institute
of Physiology II, University Hospital Jena, Friedrich Schiller University
Jena, Jena 07743, Germany
| | - Emma Kaufmann
- Institute
of Physiology II, University Hospital Jena, Friedrich Schiller University
Jena, Jena 07743, Germany
| | - Sophie Wucherpfennig
- Institute
of Physiology II, University Hospital Jena, Friedrich Schiller University
Jena, Jena 07743, Germany
| | - Frank Schwede
- BIOLOG
Life Science Institute GmbH & Co KG, Bremen 28199, Germany
| | - Georg Künze
- Institute
for Drug Discovery, Medical Faculty, University
of Leipzig, Leipzig 04103, Germany
- Interdisciplinary
Center for Bioinformatics, University of
Leipzig, Leipzig 04107, Germany
- Center
for Scalable Data Analytics and Artificial Intelligence, University of Leipzig, Leipzig 04105, Germany
| | - Vasilica Nache
- Institute
of Physiology II, University Hospital Jena, Friedrich Schiller University
Jena, Jena 07743, Germany
| |
Collapse
|
5
|
Bighinati A, Adani E, Stanzani A, D’Alessandro S, Marigo V. Molecular mechanisms underlying inherited photoreceptor degeneration as targets for therapeutic intervention. Front Cell Neurosci 2024; 18:1343544. [PMID: 38370034 PMCID: PMC10869517 DOI: 10.3389/fncel.2024.1343544] [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: 11/23/2023] [Accepted: 01/16/2024] [Indexed: 02/20/2024] Open
Abstract
Retinitis pigmentosa (RP) is a form of retinal degeneration characterized by primary degeneration of rod photoreceptors followed by a secondary cone loss that leads to vision impairment and finally blindness. This is a rare disease with mutations in several genes and high genetic heterogeneity. A challenging effort has been the characterization of the molecular mechanisms underlying photoreceptor cell death during the progression of the disease. Some of the cell death pathways have been identified and comprise stress events found in several neurodegenerative diseases such as oxidative stress, inflammation, calcium imbalance and endoplasmic reticulum stress. Other cell death mechanisms appear more relevant to photoreceptor cells, such as high levels of cGMP and metabolic changes. Here we review some of the cell death pathways characterized in the RP mutant retina and discuss preclinical studies of therapeutic approaches targeting the molecular outcomes that lead to photoreceptor cell demise.
Collapse
Affiliation(s)
- Andrea Bighinati
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Adani
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Agnese Stanzani
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara D’Alessandro
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Neuroscience and Neurotechnology, Modena, Italy
| |
Collapse
|
6
|
Yan J, Wang L, Yang QL, Yang QX, He X, Dong Y, Hu Z, Seeliger MW, Jiao K, Paquet-Durand F. T-type voltage-gated channels, Na +/Ca 2+-exchanger, and calpain-2 promote photoreceptor cell death in inherited retinal degeneration. Cell Commun Signal 2024; 22:92. [PMID: 38303059 PMCID: PMC10836022 DOI: 10.1186/s12964-023-01391-y] [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: 09/29/2023] [Accepted: 11/09/2023] [Indexed: 02/03/2024] Open
Abstract
Inherited retinal degenerations (IRDs) are a group of untreatable and commonly blinding diseases characterized by progressive photoreceptor loss. IRD pathology has been linked to an excessive activation of cyclic nucleotide-gated channels (CNGC) leading to Na+- and Ca2+-influx, subsequent activation of voltage-gated Ca2+-channels (VGCC), and further Ca2+ influx. However, a connection between excessive Ca2+ influx and photoreceptor loss has yet to be proven.Here, we used whole-retina and single-cell RNA-sequencing to compare gene expression between the rd1 mouse model for IRD and wild-type (wt) mice. Differentially expressed genes indicated links to several Ca2+-signalling related pathways. To explore these, rd1 and wt organotypic retinal explant cultures were treated with the intracellular Ca2+-chelator BAPTA-AM or inhibitors of different Ca2+-permeable channels, including CNGC, L-type VGCC, T-type VGCC, Ca2+-release-activated channel (CRAC), and Na+/Ca2+ exchanger (NCX). Moreover, we employed the novel compound NA-184 to selectively inhibit the Ca2+-dependent protease calpain-2. Effects on the retinal activity of poly(ADP-ribose) polymerase (PARP), sirtuin-type histone-deacetylase, calpains, as well as on activation of calpain-1, and - 2 were monitored, cell death was assessed via the TUNEL assay.While rd1 photoreceptor cell death was reduced by BAPTA-AM, Ca2+-channel blockers had divergent effects: While inhibition of T-type VGCC and NCX promoted survival, blocking CNGCs and CRACs did not. The treatment-related activity patterns of calpains and PARPs corresponded to the extent of cell death. Remarkably, sirtuin activity and calpain-1 activation were linked to photoreceptor protection, while calpain-2 activity was related to degeneration. In support of this finding, the calpain-2 inhibitor NA-184 protected rd1 photoreceptors.These results suggest that Ca2+ overload in rd1 photoreceptors may be triggered by T-type VGCCs and NCX. High Ca2+-levels likely suppress protective activity of calpain-1 and promote retinal degeneration via activation of calpain-2. Overall, our study details the complexity of Ca2+-signalling in photoreceptors and emphasizes the importance of targeting degenerative processes specifically to achieve a therapeutic benefit for IRDs. Video Abstract.
Collapse
Affiliation(s)
- Jie Yan
- Yunnan Eye Institute & Key Laboratory of Yunnan Province, Yunnan Eye Disease Clinical Medical Center, Affiliated Hospital of Yunnan University, Yunnan University, 176 Qingnian, Kunming, 650021, China
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, 72076, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, 72076, Germany
| | - Lan Wang
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, 72076, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, 72076, Germany
| | - Qian-Lu Yang
- The Third Affiliated Hospital of Kunming Medical University &Yunnan Cancer Hospital, Kunming, Yunnan, 650118, China
| | - Qian-Xi Yang
- The Third Affiliated Hospital of Kunming Medical University &Yunnan Cancer Hospital, Kunming, Yunnan, 650118, China
| | - Xinyi He
- Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, 72076, Germany
- High-resolution Functional Imaging and Test Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, 72076, Germany
| | - Yujie Dong
- Yunnan Eye Institute & Key Laboratory of Yunnan Province, Yunnan Eye Disease Clinical Medical Center, Affiliated Hospital of Yunnan University, Yunnan University, 176 Qingnian, Kunming, 650021, China
| | - Zhulin Hu
- Yunnan Eye Institute & Key Laboratory of Yunnan Province, Yunnan Eye Disease Clinical Medical Center, Affiliated Hospital of Yunnan University, Yunnan University, 176 Qingnian, Kunming, 650021, China
| | - Mathias W Seeliger
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, 72076, Germany
| | - Kangwei Jiao
- Yunnan Eye Institute & Key Laboratory of Yunnan Province, Yunnan Eye Disease Clinical Medical Center, Affiliated Hospital of Yunnan University, Yunnan University, 176 Qingnian, Kunming, 650021, China
| | - François Paquet-Durand
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, 72076, Germany.
| |
Collapse
|
7
|
Sun Y, Sun Y, Chen S, Yu Y, Ma Y, Sun F. Hypoxic preconditioned MSCs-derived small extracellular vesicles for photoreceptor protection in retinal degeneration. J Nanobiotechnology 2023; 21:449. [PMID: 38001463 PMCID: PMC10675959 DOI: 10.1186/s12951-023-02225-2] [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: 08/31/2023] [Accepted: 11/19/2023] [Indexed: 11/26/2023] Open
Abstract
Photoreceptor apoptosis is an important pathogenesis of retinal degeneration and a primary cause of vision loss with limited treatment methods. Mesenchymal stem/stromal cells-derived small extracellular vesicles (MSC-sEVs) have shown therapeutic value in various ocular disorders. Recent studies have revealed that hypoxic preconditioning can improve the effectiveness of MSC-sEVs in tissue regeneration. However, whether hypoxic preconditioned MSC-sEVs (Hyp-sEVs) exert superior effects on photoreceptor protection relative to normoxic conditioned MSC-sEVs (Nor-sEVs) remains unclear. Here, we reported that Hyp-sEVs further improved retinal structure, recovered retinal function, and suppressed photoreceptor apoptosis in N-methyl-N-nitrosourea (MNU)-induced mouse model compared with Nor-sEVs. Hyp-sEVs also exhibited enhanced anti-apoptotic roles in MNU-provoked 661 W cell injury in vitro. We then analyzed the protein profiles of Nor-sEVs and Hyp-sEVs by LC-MS/MS and found that growth-associated protein 43 (GAP43) was enriched in Hyp-sEVs. The knockdown of GAP43 abolished the retinal therapeutic effects of Hyp-sEVs. Mechanistically, hypoxic stimulation-induced hypoxia-inducible factor-1α (HIF-1α) activation was responsible for preventing tripartite motif-containing protein 25 (TRIM25)-mediated GAP43 ubiquitination and degradation, leading to the upregulation of GAP43 in Hyp-sEVs. Together, our findings uncover the efficacy and mechanism of Hyp-sEVs-based photoreceptor protection and highlight the potential of Hyp-sEVs as optimized therapeutics for retinal degeneration.
Collapse
Affiliation(s)
- Yuntong Sun
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang, China
| | - Yuntao Sun
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Shenyuan Chen
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yifan Yu
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yongjun Ma
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang, China.
| | - Fengtian Sun
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang, China.
| |
Collapse
|
8
|
Tolone A, Haq W, Fachinger A, Roy A, Kesh S, Rentsch A, Wucherpfennig S, Zhu Y, Groten J, Schwede F, Tomar T, Herberg FW, Nache V, Paquet-Durand F. The PKG Inhibitor CN238 Affords Functional Protection of Photoreceptors and Ganglion Cells against Retinal Degeneration. Int J Mol Sci 2023; 24:15277. [PMID: 37894958 PMCID: PMC10607377 DOI: 10.3390/ijms242015277] [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: 07/19/2023] [Revised: 09/27/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Hereditary retinal degeneration (RD) is often associated with excessive cGMP signalling in photoreceptors. Previous research has shown that inhibition of cGMP-dependent protein kinase G (PKG) can reduce photoreceptor loss in two different RD animal models. In this study, we identified a PKG inhibitor, the cGMP analogue CN238, which preserved photoreceptor viability and functionality in rd1 and rd10 mutant mice. Surprisingly, in explanted retinae, CN238 also protected retinal ganglion cells from axotomy-induced retrograde degeneration and preserved their functionality. Furthermore, kinase activity-dependent protein phosphorylation of the PKG target Kv1.6 was reduced in CN238-treated rd10 retinal explants. Ca2+-imaging on rd10 acute retinal explants revealed delayed retinal ganglion cell repolarization with CN238 treatment, suggesting a PKG-dependent modulation of Kv1-channels. Together, these results highlight the strong neuroprotective capacity of PKG inhibitors for both photoreceptors and retinal ganglion cells, illustrating their broad potential for the treatment of retinal diseases and possibly neurodegenerative diseases in general.
Collapse
Affiliation(s)
- Arianna Tolone
- Cell Death Mechanism Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (A.T.); (Y.Z.)
| | - Wadood Haq
- Neuroretinal Electrophysiology and Imaging, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
| | - Alexandra Fachinger
- Biochemistry Department, University of Kassel, 34132 Kassel, Germany; (A.F.); (F.W.H.)
| | - Akanksha Roy
- PamGene International B.V., 5211 ‘s-Hertogenbosch, The Netherlands; (A.R.); (J.G.); (T.T.)
| | - Sandeep Kesh
- Institute of Physiology II, University Hospital Jena, Friedrich Schiller University Jena, 07743 Jena, Germany; (S.K.); (S.W.); (V.N.)
| | - Andreas Rentsch
- Biolog Life Science Institute GmbH & Co. KG, 28199 Bremen, Germany; (A.R.); (F.S.)
| | - Sophie Wucherpfennig
- Institute of Physiology II, University Hospital Jena, Friedrich Schiller University Jena, 07743 Jena, Germany; (S.K.); (S.W.); (V.N.)
| | - Yu Zhu
- Cell Death Mechanism Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (A.T.); (Y.Z.)
| | - John Groten
- PamGene International B.V., 5211 ‘s-Hertogenbosch, The Netherlands; (A.R.); (J.G.); (T.T.)
| | - Frank Schwede
- Biolog Life Science Institute GmbH & Co. KG, 28199 Bremen, Germany; (A.R.); (F.S.)
| | - Tushar Tomar
- PamGene International B.V., 5211 ‘s-Hertogenbosch, The Netherlands; (A.R.); (J.G.); (T.T.)
| | - Friedrich W. Herberg
- Biochemistry Department, University of Kassel, 34132 Kassel, Germany; (A.F.); (F.W.H.)
| | - Vasilica Nache
- Institute of Physiology II, University Hospital Jena, Friedrich Schiller University Jena, 07743 Jena, Germany; (S.K.); (S.W.); (V.N.)
| | - François Paquet-Durand
- Cell Death Mechanism Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (A.T.); (Y.Z.)
| |
Collapse
|
9
|
Peiroten L, Zrenner E, Haq W. Artificial Vision: The High-Frequency Electrical Stimulation of the Blind Mouse Retina Decay Spike Generation and Electrogenically Clamped Intracellular Ca 2+ at Elevated Levels. Bioengineering (Basel) 2023; 10:1208. [PMID: 37892938 PMCID: PMC10604554 DOI: 10.3390/bioengineering10101208] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/05/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The electrical stimulation (stim) of retinal neurons enables blind patients to experience limited artificial vision. A rapid response outage of the stimulated ganglion cells (GCs) allows for a low visual sensation rate. Hence, to elucidate the underlying mechanism, we investigated different stim parameters and the role of the neuromodulator calcium (Ca2+). METHODS Subretinal stim was applied on retinal explants (blind rd1 mouse) using multielectrode arrays (MEAs) or single metal electrodes, and the GC activity was recorded using Ca2+ imaging or MEA, respectively. Stim parameters, including voltage, phase polarity, and frequency, were investigated using specific blockers. RESULTS At lower stim frequencies (<5 Hz), GCs responded synaptically according to the stim pulses (stim: biphasic, cathodic-first, -1.6/+1.5 V). In contrast, higher stim frequencies (≥5 Hz) also activated GCs directly and induced a rapid GC spike response outage (<500 ms, MEA recordings), while in Ca2+ imaging at the same frequencies, increased intracellular Ca2+ levels were observed. CONCLUSIONS Our study elucidated the mechanisms involved in stim-dependent GC spike response outage: sustained high-frequency stim-induced spike outage, accompanied by electrogenically clamped intracellular Ca2+ levels at elevated levels. These findings will guide future studies optimizing stim paradigms for electrical implant applications for interfacing neurons.
Collapse
Affiliation(s)
| | | | - Wadood Haq
- Neuroretinal Electrophysiology and Imaging, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (L.P.)
| |
Collapse
|
10
|
Dong Y, Yan J, Xu W, Paquet-Durand F, Hu Z, Jiao K. HDAC inhibition delays photoreceptor loss in Pde6b mutant mice of retinitis pigmentosa: insights from scRNA-seq and CUT&Tag. PeerJ 2023; 11:e15659. [PMID: 37456870 PMCID: PMC10349563 DOI: 10.7717/peerj.15659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
Purpose This research aimed to ascertain the neuroprotective effect of histone deacetylase (HDAC) inhibition on retinal photoreceptors in Pde6brd1 mice, a model of retinitis pigmentosa (RP). Methods Single-cell RNA-sequencing (scRNA-seq) explored HDAC and poly (ADP-ribose) polymerase (PARP)-related gene expression in both Pde6b-mutant rd1 and wild-type (WT) mice. The CUT&Tag method was employed to examine the functions of HDAC in rd1 mice. Organotypic retinal explant cultures from WT and rd1 mice were exposed to the HDAC inhibitor SAHA (suberoylanilide hydroxamic acid) postnatally, from day 5 to day 11. The terminal deoxynucleotidyl transferase-mediated nick-end labeling (TUNEL) assay was applied to quantify the percentage of photoreceptor loss in the outer nuclear layer (ONL). HDAC activity was confirmed to be inhibited by SAHA through an HDAC activity assay. Moreover, the study evaluated PARP activity, a key driver of the initial response to DNA damage during photoreceptor degeneration, following HDAC inhibition. Results The scRNA-seq revealed that diverse roles of HDAC and PARP isoforms in photoreceptor cell death. HDAC-related genes appeared to regulate cell death and primary immunodeficiency. Alterations in HDAC activity were consistent with the TUNEL-positive cells in the ONL at different time points. Notably, SAHA significantly postponed photoreceptor loss and decreased HDAC and PARP activity, thereby implicating both in the same degenerative pathway. Conclusions This study highlights that the interaction between HDAC inhibition and PARP can delay photoreceptor cell death, proposing a promising therapeutic approach for RP.
Collapse
Affiliation(s)
- Yujie Dong
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of Yunnan Province, Yunnan Eye Institute, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, Yunnan, China
| | - Jie Yan
- Key Laboratory of Yunnan Province, Yunnan Eye Institute, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, Yunnan, China
- Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Wenrong Xu
- Key Laboratory of Yunnan Province, Yunnan Eye Institute, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, Yunnan, China
| | - François Paquet-Durand
- Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Zhulin Hu
- Key Laboratory of Yunnan Province, Yunnan Eye Institute, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, Yunnan, China
| | - Kangwei Jiao
- Key Laboratory of Yunnan Province, Yunnan Eye Institute, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, Yunnan, China
| |
Collapse
|
11
|
Zhou J, Welinder C, Ekström P. The Phosphoproteome of the Rd1 Mouse Retina, a Model of Inherited Photoreceptor Degeneration, Changes after Protein Kinase G Inhibition. Int J Mol Sci 2023; 24:9836. [PMID: 37372984 DOI: 10.3390/ijms24129836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Retinitis pigmentosa (RP) is a frequent cause of blindness among the working population in industrial countries due to the inheritable death of photoreceptors. Though gene therapy was recently approved for mutations in the RPE65 gene, there is in general no effective treatment presently. Previously, abnormally high levels of cGMP and overactivation of its dependent protein kinase (PKG) have been suggested as causative for the fatal effects on photoreceptors, making it meaningful to explore the cGMP-PKG downstream signaling for more pathological insights and novel therapeutic target development purposes. Here, we manipulated the cGMP-PKG system in degenerating retinas from the rd1 mouse model pharmacologically via adding a PKG inhibitory cGMP-analogue to organotypic retinal explant cultures. A combination of phosphorylated peptide enrichment and mass spectrometry was then applied to study the cGMP-PKG-dependent phosphoproteome. We identified a host of novel potential cGMP-PKG downstream substrates and related kinases using this approach and selected the RAF1 protein, which may act as both a substrate and a kinase, for further validation. This showed that the RAS/RAF1/MAPK/ERK pathway may be involved in retinal degeneration in a yet unclarified mechanism, thus deserving further investigation in the future.
Collapse
Affiliation(s)
- Jiaming Zhou
- Ophthalmology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, 221 00 Lund, Sweden
| | - Charlotte Welinder
- Mass Spectrometry, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, 221 00 Lund, Sweden
| | - Per Ekström
- Ophthalmology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, 221 00 Lund, Sweden
| |
Collapse
|
12
|
Chen C, Rong Y, Zhuang Y, Tang C, Liu Q, Lin P, Li D, Zhao X, Lu F, Qu J, Liu X. RNA-Seq Analysis Reveals an Essential Role of the cGMP-PKG-MAPK Pathways in Retinal Degeneration Caused by Cep250 Deficiency. Int J Mol Sci 2023; 24:8843. [PMID: 37240188 PMCID: PMC10218315 DOI: 10.3390/ijms24108843] [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: 04/12/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Usher syndrome (USH) is characterised by degenerative vision loss known as retinitis pigmentosa (RP), sensorineural hearing loss, and vestibular dysfunction. RP can cause degeneration and the loss of rod and cone photoreceptors, leading to structural and functional changes in the retina. Cep250 is a candidate gene for atypical Usher syndrome, and this study describes the development of a Cep250 KO mouse model to investigate its pathogenesis. OCT and ERG were applied in Cep250 and WT mice at P90 and P180 to access the general structure and function of the retina. After recording the ERG responses and OCT images at P90 and P180, the cone and rod photoreceptors were visualised using an immunofluorescent stain. TUNEL assays were applied to observe the apoptosis in Cep250 and WT mice retinas. The total RNA was extracted from the retinas and executed for RNA sequencing at P90. Compared with WT mice, the thickness of the ONL, IS/OS, and whole retina of Cep250 mice was significantly reduced. The a-wave and b-wave amplitude of Cep250 mice in scotopic and photopic ERG were lower, especially the a-wave. According to the immunostaining and TUNEL stain results, the photoreceptors in the Cep250 retinas were also reduced. An RNA-seq analysis showed that 149 genes were upregulated and another 149 genes were downregulated in Cep250 KO retinas compared with WT mice retinas. A KEGG enrichment analysis indicated that cGMP-PKG signalling pathways, MAPK signalling pathways, edn2-fgf2 axis pathways, and thyroid hormone synthesis were upregulated, whereas protein processing in the endoplasmic reticulum was downregulated in Cep250 KO eyes. Cep250 KO mice experience a late-stage retinal degeneration that manifests as the atypical USH phenotype. The dysregulation of the cGMP-PKG-MAPK pathways may contribute to the pathogenesis of cilia-related retinal degeneration.
Collapse
Affiliation(s)
- Chong Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yu Rong
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Youyuan Zhuang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Cheng Tang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qian Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Peng Lin
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Dandan Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Xinyi Zhao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Fan Lu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jia Qu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Xinting Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (C.C.); (Y.R.); (Y.Z.); (C.T.); (Q.L.); (P.L.); (D.L.); (X.Z.); (F.L.)
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
13
|
Rasmussen M, Tolone A, Paquet-Durand F, Welinder C, Schwede F, Ekström P. The photoreceptor protective cGMP-analog Rp-8-Br-PET-cGMPS interacts with cGMP-interactors PKGI, PDE1, PDE6, and PKAI in the degenerating mouse retina. J Comp Neurol 2023; 531:935-951. [PMID: 36989379 DOI: 10.1002/cne.25475] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/12/2022] [Accepted: 03/06/2023] [Indexed: 03/31/2023]
Abstract
The inherited eye disease retinitis pigmentosa (RP) causes the loss of photoreceptors by a still unknown cell death mechanism. During this degeneration, cyclic guanosine-3',5'-monophosphate (cGMP) levels become elevated, leading to over-activation of the cGMP-binding protein cGMP-dependent protein kinase (PKG). cGMP analogs selectively modified to have inhibitory actions on PKG have aided in impeding photoreceptor death, and one such cGMP analog is Rp-8-Br-PET-cGMPS. However, cGMP analogs have previously been shown to interact with numerous targets, so to better understand the therapeutic action of Rp-8-Br-PET-cGMPS, it is necessary to elucidate its target-selectivity and hence what potential cellular mechanism(s) it may affect within the photoreceptors. Here, we, therefore, applied affinity chromatography together with mass spectrometry to isolate and identify Rp-8-Br-PET-cGMPS interactors from retinas derived from three different murine RP models (i.e., rd1, rd2, and rd10 mice). Our findings revealed that Rp-8-Br-PET-cGMPS bound seven known cGMP-binding proteins, including PKG1β, PDE1β, PDE1c, PDE6α, and PKA1α. Furthermore, an additional 28 proteins were found to be associated with Rp-8-Br-PET-cGMPS. This latter group included MAPK1/3, which is known to connect with cGMP/PKG in other systems. However, in organotypic retinal cultures, Rp-8-Br-PET-cGMPS had no effect on photoreceptor MAPK1/3 expression or activity. To summarize, Rp-8-Br-PET-cGMPS is more target specific compared to regular cGMP.
Collapse
Affiliation(s)
- Michel Rasmussen
- Faculty of Medicine, Department of Clinical Sciences Lund, Lund University, Ophthalmology, Lund, Sweden
| | - Arianna Tolone
- Insitute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | | | - Charlotte Welinder
- Faculty of Medicine, Department of Clinical Sciences Lund, Mass Spectrometry, Lund University, Lund, Sweden
| | - Frank Schwede
- BIOLOG Life Science Institute GmbH & Co. KG, Bremen, Germany
| | - Per Ekström
- Faculty of Medicine, Department of Clinical Sciences Lund, Lund University, Ophthalmology, Lund, Sweden
| |
Collapse
|
14
|
Dong Y, Yan J, Yang M, Xu W, Hu Z, Paquet-Durand F, Jiao K. Inherited Retinal Degeneration: Towards the Development of a Combination Therapy Targeting Histone Deacetylase, Poly (ADP-Ribose) Polymerase, and Calpain. Biomolecules 2023; 13:biom13040581. [PMID: 37189329 DOI: 10.3390/biom13040581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/05/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Inherited retinal degeneration (IRD) represents a diverse group of gene mutation-induced blinding diseases. In IRD, the loss of photoreceptors is often connected to excessive activation of histone-deacetylase (HDAC), poly-ADP-ribose-polymerase (PARP), and calpain-type proteases (calpain). Moreover, the inhibition of either HDACs, PARPs, or calpains has previously shown promise in preventing photoreceptor cell death, although the relationship between these enzyme groups remains unclear. To explore this further, organotypic retinal explant cultures derived from wild-type mice and rd1 mice as a model for IRD were treated with different combinations of inhibitors specific for HDAC, PARP, and calpain. The outcomes were assessed using in situ activity assays for HDAC, PARP, and calpain, immunostaining for activated calpain-2, and the TUNEL assay for cell death detection. We confirmed that inhibition of either HDAC, PARP, or calpain reduced rd1 mouse photoreceptor degeneration, with the HDAC inhibitor Vorinostat (SAHA) being most effective. Calpain activity was reduced by inhibition of both HDAC and PARP whereas PARP activity was only reduced by HDAC inhibition. Unexpectedly, combined treatment with either PARP and calpain inhibitors or HDAC and calpain inhibitors did not produce synergistic rescue of photoreceptors. Together, these results indicate that in rd1 photoreceptors, HDAC, PARP, and calpain are part of the same degenerative pathway and are activated in a sequence that begins with HDAC and ends with calpain.
Collapse
|
15
|
Zhou J, Ekström P. Pyruvate Kinase 2, an Energy Metabolism Related Enzyme, May Have a Neuroprotective Function in Retinal Degeneration. ASN Neuro 2023; 15:17590914231151534. [PMID: 36799552 PMCID: PMC9940218 DOI: 10.1177/17590914231151534] [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] [Indexed: 02/18/2023] Open
Abstract
Retinitis pigmentosa (RP) is an inherited disorder that results in vision impairment but general and mutation-independent therapeutic strategies are not available. However, it is widely regarded that the cGMP system, including cGMP and its interactor cGMP-dependent protein kinase (PKG), acts as a crucial effector during retinal degeneration. We have previously identified a list of cGMP-PKG-dependent genes in the context of RP, and in this study, we further validated one of these, namely pyruvate kinase 2 (PKM2), and investigated the potential role of PKM2 for the photoreceptors' well-being during RP. With the aid of organotypic retinal explant cultures, we pharmacologically manipulated the PKM2 activities in two different RP mouse models (rd2 and rd10) via the addition of TEPP-46 (a PKM2 activator) and found that activation of PKM2 alleviates the progress of photoreceptor death in the rd10 mouse model. We also noted that the expression of both PKM2 and one of its targets, glucose transporter-1 (Glut1), showed alterations depending on the degeneration state. The observations provide supportive evidence that PKM2 may serve as a novel potential molecular target in RP.
Collapse
Affiliation(s)
- Jiaming Zhou
- Ophthalmology, Department of Clinical Sciences, Lund University, Lund, Sweden,Jiaming Zhou, Biomedical Center (BMC-B11), Sölvegatan 19, SE-22362 Lund, Sweden.
| | - Per Ekström
- Ophthalmology, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
16
|
Zhou J, Rasmussen M, Ekström P. A Potential Neuroprotective Role for Pyruvate Kinase 2 in Retinal Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:479-483. [PMID: 37440075 DOI: 10.1007/978-3-031-27681-1_70] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Retinitis pigmentosa (RP) is an inherited disorder that results in vision impairment that specific therapeutic strategies are not available. However, it is widely regarded that the cGMP system, including cGMP and its interactor cGMP-dependent protein kinase (PKG), acts as a crucial effector during retinal degeneration. We have previously identified a list of cGMP-PKG-dependent genes in the context of RP, and in this study, we further validated one of the targets, namely, pyruvate kinase 2 (PKM2), and investigated the potential role of PKM2 for the photoreceptors' well-being during RP. With the aid of organotypic retinal explant cultures, we pharmacologically manipulated the PKM2 activities in different RP mouse models via the addition of TEPP-46 (a PKM2 activator) and found that activation of PKM2 alleviates the progress of photoreceptor death in the rd10 mouse model. This observation provides supportive evidence that PKM2 may serve as a novel potential molecular target in RP.
Collapse
Affiliation(s)
- Jiaming Zhou
- Ophthalmology, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - Michel Rasmussen
- Ophthalmology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Per Ekström
- Ophthalmology, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
17
|
Karamali F, Behtaj S, Babaei-Abraki S, Hadady H, Atefi A, Savoj S, Soroushzadeh S, Najafian S, Nasr Esfahani MH, Klassen H. Potential therapeutic strategies for photoreceptor degeneration: the path to restore vision. J Transl Med 2022; 20:572. [PMID: 36476500 PMCID: PMC9727916 DOI: 10.1186/s12967-022-03738-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/29/2022] [Indexed: 12/12/2022] Open
Abstract
Photoreceptors (PRs), as the most abundant and light-sensing cells of the neuroretina, are responsible for converting light into electrical signals that can be interpreted by the brain. PR degeneration, including morphological and functional impairment of these cells, causes significant diminution of the retina's ability to detect light, with consequent loss of vision. Recent findings in ocular regenerative medicine have opened promising avenues to apply neuroprotective therapy, gene therapy, cell replacement therapy, and visual prostheses to the challenge of restoring vision. However, successful visual restoration in the clinical setting requires application of these therapeutic approaches at the appropriate stage of the retinal degeneration. In this review, firstly, we discuss the mechanisms of PR degeneration by focusing on the molecular mechanisms underlying cell death. Subsequently, innovations, recent developments, and promising treatments based on the stage of disorder progression are further explored. Then, the challenges to be addressed before implementation of these therapies in clinical practice are considered. Finally, potential solutions to overcome the current limitations of this growing research area are suggested. Overall, the majority of current treatment modalities are still at an early stage of development and require extensive additional studies, both pre-clinical and clinical, before full restoration of visual function in PR degeneration diseases can be realized.
Collapse
Affiliation(s)
- Fereshteh Karamali
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sanaz Behtaj
- grid.1022.10000 0004 0437 5432Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Queensland, Australia ,grid.1022.10000 0004 0437 5432Menzies Health Institute Queensland, Griffith University, Southport, QLD 4222 Australia
| | - Shahnaz Babaei-Abraki
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hanieh Hadady
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Atefeh Atefi
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Soraya Savoj
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sareh Soroushzadeh
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Samaneh Najafian
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr Esfahani
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Henry Klassen
- grid.266093.80000 0001 0668 7243Gavin Herbert Eye Institute, Irvine, CA USA
| |
Collapse
|
18
|
Inhibition of the MAPK/c-Jun-EGR1 Pathway Decreases Photoreceptor Cell Death in the rd1 Mouse Model for Inherited Retinal Degeneration. Int J Mol Sci 2022; 23:ijms232314600. [PMID: 36498926 PMCID: PMC9740268 DOI: 10.3390/ijms232314600] [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: 08/30/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022] Open
Abstract
Retinitis pigmentosa (RP) is a group of inherited retinal dystrophies that typically results in photoreceptor cell death and vision loss. Here, we explored the effect of early growth response-1 (EGR1) expression on photoreceptor cell death in Pde6brd1 (rd1) mice and its mechanism of action. To this end, single-cell RNA-seq (scRNA-seq) was used to identify differentially expressed genes in rd1 and congenic wild-type (WT) mice. Chromatin immunoprecipitation (ChIP), the dual-luciferase reporter gene assay, and western blotting were used to verify the relationship between EGR1 and poly (ADP-ribose) polymerase-1 (PARP1). Immunofluorescence staining was used to assess PARP1 expression after silencing or overexpression of EGR1. Photoreceptor cell death was assessed using the TUNEL assay following silencing/overexpression of EGR1 or administration of MAPK/c-Jun pathway inhibitors tanzisertib and PD98059. Our results showed differential expression of ERG1 in rd1 and WT mice via scRNA-seq analysis. The ChIP assay demonstrated EGR1 binding to the PARP1 promoter region. The dual-luciferase reporter gene assay and western blotting results revealed that EGR1 upregulated PARP1 expression. Additionally, the TUNEL assay showed that silencing EGR1 effectively reduced photoreceptor cell death. Similarly, the addition of tanzisertib and PD98059 reduced the expression of c-Jun and EGR1 and decreased photoreceptor cell death. Our study revealed that inhibition of the MAPK/c-Jun pathway reduced the expression of EGR1 and PARP1 and prevented photoreceptor cell death. These results highlight the importance of EGR1 for photoreceptor cell death and identify a new avenue for therapeutic interventions in RP.
Collapse
|
19
|
Depleted Calcium Stores and Increased Calcium Entry in Rod Photoreceptors of the Cacna2d4 Mouse Model of Cone-Rod Dystrophy RCD4. Int J Mol Sci 2022; 23:ijms232113080. [DOI: 10.3390/ijms232113080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Unidentified pathogenetic mechanisms and genetic and clinical heterogeneity represent critical factors hindering the development of treatments for inherited retinal dystrophies. Frameshift mutations in Cacna2d4, which codes for an accessory subunit of voltage-gated calcium channels (VGCC), cause cone-rod dystrophy RCD4 in patients, but the underlying mechanisms remain unknown. To define its pathogenetic mechanisms, we investigated the impact of a Cacna2d4 frameshift mutation on the electrophysiological profile and calcium handling of mouse rod photoreceptors by patch-clamp recordings and calcium imaging, respectively. In mutant (MUT) rods, the dysregulation of calcium handling extends beyond the reduction in calcium entry through VGCC and surprisingly involves internal calcium stores’ depletion and upregulation of calcium entry via non-selective cationic channels (CSC). The similar dependence of CSC on basal calcium levels in WT and MUT rods suggests that the primary defect in MUT rods lies in defective calcium stores. Calcium stores’ depletion, leading to upregulated calcium and sodium influx via CSC, represents a novel and, so far, unsuspected consequence of the Cacna2d4 mutation. Blocking CSC may provide a novel strategy to counteract the well-known pathogenetic mechanisms involved in rod demise, such as the reticulum stress response and calcium and sodium overload due to store depletion.
Collapse
|
20
|
Chen Y, Dong Y, Yan J, Wang L, Yu S, Jiao K, Paquet-Durand F. Single-Cell Transcriptomic Profiling in Inherited Retinal Degeneration Reveals Distinct Metabolic Pathways in Rod and Cone Photoreceptors. Int J Mol Sci 2022; 23:12170. [PMID: 36293024 PMCID: PMC9603353 DOI: 10.3390/ijms232012170] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/28/2022] [Accepted: 10/08/2022] [Indexed: 08/31/2023] Open
Abstract
The cellular mechanisms underlying hereditary photoreceptor degeneration are still poorly understood. The aim of this study was to systematically map the transcriptional changes that occur in the degenerating mouse retina at the single cell level. To this end, we employed single-cell RNA-sequencing (scRNA-seq) and retinal degeneration-1 (rd1) mice to profile the impact of the disease mutation on the diverse retinal cell types during early post-natal development. The transcriptome data allowed to annotate 43,979 individual cells grouped into 20 distinct clusters. We further characterized cluster-specific metabolic and biological changes in individual cell types. Our results highlight Ca2+-signaling as relevant to hereditary photoreceptor degeneration. Although metabolic reprogramming in retina, known as the 'Warburg effect', has been documented, further metabolic changes were noticed in rd1 mice. Such metabolic changes in rd1 mutation was likely regulated through mitogen-activated protein kinase (MAPK) pathway. By combining single-cell transcriptomes and immunofluorescence staining, our study revealed cell type-specific changes in gene expression, as well as interplay between Ca2+-induced cell death and metabolic pathways.
Collapse
Affiliation(s)
- Yiyi Chen
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Yujie Dong
- Yunnan Eye Institute & Key Laboratory of Yunnan Province, 650021 Kunming, China
| | - Jie Yan
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Lan Wang
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Shirley Yu
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Kangwei Jiao
- Yunnan Eye Institute & Key Laboratory of Yunnan Province, 650021 Kunming, China
| | | |
Collapse
|
21
|
cGMP Analogues with Opposing Actions on CNG Channels Selectively Modulate Rod or Cone Photoreceptor Function. Pharmaceutics 2022; 14:pharmaceutics14102102. [PMID: 36297537 PMCID: PMC9612005 DOI: 10.3390/pharmaceutics14102102] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
The vertebrate retina harbors rod and cone photoreceptors. Human vision critically depends on cone photoreceptor function. In the phototransduction cascade, cGMP activates distinct rod and cone isoforms of the cyclic nucleotide-gated (CNG) channel. Excessive cGMP levels initiate a pathophysiological rollercoaster, which starts with CNG channel over-activation, typically in rod photoreceptors. This triggers cell death of rods first, and then cones, and is the root cause of many blinding retinal diseases, including Retinitis pigmentosa. While targeting of CNG channels has been proposed for therapeutic purposes, thus far, it has not been possible to inhibit rod CNG channels without compromising cone function. Here, we present a novel strategy, based on cGMP analogues with opposing actions on CNG channels, which enables the selective modulation of either rod or cone photoreceptor activity. The combined treatment with the weak rod-selective CNG-channel inhibitor (Rp-8-Br-PET-cGMPS) and the cone-selective CNG-channel activator (8-pCPT-cGMP) essentially normalized rod CNG-channel function while preserving cone functionality at physiological and pathological cGMP levels. Hence, combinations of cGMP analogues with desired properties may elegantly address the isoform-specificity problem in future pharmacological therapies. Moreover, this strategy may allow for improvements in visual performance in certain light environments.
Collapse
|
22
|
Martínez-Gil N, Maneu V, Kutsyr O, Fernández-Sánchez L, Sánchez-Sáez X, Sánchez-Castillo C, Campello L, Lax P, Pinilla I, Cuenca N. Cellular and molecular alterations in neurons and glial cells in inherited retinal degeneration. Front Neuroanat 2022; 16:984052. [PMID: 36225228 PMCID: PMC9548552 DOI: 10.3389/fnana.2022.984052] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple gene mutations have been associated with inherited retinal dystrophies (IRDs). Despite the spectrum of phenotypes caused by the distinct mutations, IRDs display common physiopathology features. Cell death is accompanied by inflammation and oxidative stress. The vertebrate retina has several attributes that make this tissue vulnerable to oxidative and nitrosative imbalance. The high energy demands and active metabolism in retinal cells, as well as their continuous exposure to high oxygen levels and light-induced stress, reveal the importance of tightly regulated homeostatic processes to maintain retinal function, which are compromised in pathological conditions. In addition, the subsequent microglial activation and gliosis, which triggers the secretion of pro-inflammatory cytokines, chemokines, trophic factors, and other molecules, further worsen the degenerative process. As the disease evolves, retinal cells change their morphology and function. In disease stages where photoreceptors are lost, the remaining neurons of the retina to preserve their function seek out for new synaptic partners, which leads to a cascade of morphological alterations in retinal cells that results in a complete remodeling of the tissue. In this review, we describe important molecular and morphological changes in retinal cells that occur in response to oxidative stress and the inflammatory processes underlying IRDs.
Collapse
Affiliation(s)
- Natalia Martínez-Gil
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | | | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Carla Sánchez-Castillo
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Isabel Pinilla
- Aragón Institute for Health Research (IIS Aragón), Zaragoza, Spain
- Department of Ophthalmology, Lozano Blesa University Hospital, Zaragoza, Spain
- Department of Surgery, University of Zaragoza, Zaragoza, Spain
- Isabel Pinilla,
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
- *Correspondence: Nicolás Cuenca,
| |
Collapse
|
23
|
Huang P, Jin W, Xu S, Jin L, Chen J, Zhang T, Mao K, Wan H, He Y. Optimization of smashing tissue and ultrasonic extraction of tanshinones and their neuroprotective effect on cerebral ischemia/reperfusion injury by inhibiting parthanatos. Food Funct 2022; 13:9658-9673. [PMID: 36040108 DOI: 10.1039/d2fo01902g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A green smashing tissue and ultrasonic (STU) extraction method, which combines smashing tissue and ultrasonic-assisted extraction, was developed for the first time. The extraction of tanshinones from Salvia miltiorrhiza Bunge (SM) was taken as an example to discuss the practicability of this method. Taking the total yield of eight tanshinones as an evaluation index, response surface methodology (RSM) and artificial neural network (ANN) models were used to optimize the extraction parameters, and these two models were also compared by investigating the extract yield of tanshinones and the antioxidant activity of the obtained SM extract. The optimal STU conditions by ANN were as follows: an ethanol concentration of 73%, a liquid/solid ratio of 30 mL g-1, a smashing tissue time of 97 s and an ultrasonic time of 40 min. Under these optimal conditions, the yield of the eight components was 0.30% ± 0.12, which was greater than 0.28% ± 0.03 optimized by RSM. The IC50 values of 1,1-diphenyl-2-picrylhydrazyl (DPPH) and 2,2'-azinobis-(3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt (ABTS) of the obtained extract were 55.25 ± 3.72 μg mL-1 and 67.33 ± 2.62 μg mL-1, respectively, which were better than those of 75.49 ± 4.33 μg mL-1 and 112.10 ± 5.98 μg mL-1, respectively, optimized by RSM. Furthermore, the SM extract was found to exert neuroprotective effects by inhibiting parthanatos in middle cerebral artery occlusion/reperfusion (MCAO/R)-induced rats. The results supported the use of the SM extract, which was obtained by STU, as a potential product in the cosmetics, medicine, and food industries.
Collapse
Affiliation(s)
- Ping Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Weifeng Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Shouchao Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Lei Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Jianzhen Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Ting Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Kunjun Mao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
24
|
A Potential Role of Cyclic Dependent Kinase 1 (CDK1) in Late Stage of Retinal Degeneration. Cells 2022; 11:cells11142143. [PMID: 35883586 PMCID: PMC9317054 DOI: 10.3390/cells11142143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 01/27/2023] Open
Abstract
Cyclin dependent kinase 1 (CDK1) has long been known to drive the cell cycle and to regulate the division and differentiation of cells. Apart from its role in mitosis regulation, it also exerts multiple functions as a protein kinase, including engagement in cell death, possibly via a cell cycle-independent mechanism. The latter is suggested, since CDK1 re-expression can be found in non-dividing and terminally differentiated neurons in several neurodegeneration models. However, the details of if and how CDK1 might be involved in the neurodegenerative condition, retinitis pigmentosa (RP), which displays progressive vision loss, are unclear. In the present study, we investigated CDK1 in degenerating RP photoreceptors of the rd1 RP model, including whether there is a link between this kinase and the cGMP-PKG system, which is regarded as a disease driver. With experiments performed using either in vivo retinal tissue or in vitro material, via organotypic retinal explants, our results showed that CDK1 appears in the photoreceptors at a late stage of their degeneration, and in such a position, it may be associated with the cGMP-PKG network.
Collapse
|
25
|
Molecular Mechanisms of Parthanatos and Its Role in Diverse Diseases. Int J Mol Sci 2022; 23:ijms23137292. [PMID: 35806303 PMCID: PMC9266317 DOI: 10.3390/ijms23137292] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Differential evolution of apoptosis, programmed necrosis, and autophagy, parthanatos is a form of cell death mediated by poly(ADP-ribose) polymerase 1 (PARP1), which is caused by DNA damage. PARP1 hyper-activation stimulates apoptosis-inducing factor (AIF) nucleus translocation, and accelerates nicotinamide adenine dinucleotide (NAD+) and adenosine triphosphate (ATP) depletion, leading to DNA fragmentation. The mechanisms of parthanatos mainly include DNA damage, PARP1 hyper-activation, PAR accumulation, NAD+ and ATP depletion, and AIF nucleus translocation. Now, it is reported that parthanatos widely exists in different diseases (tumors, retinal diseases, neurological diseases, diabetes, renal diseases, cardiovascular diseases, ischemia-reperfusion injury...). Excessive or defective parthanatos contributes to pathological cell damage; therefore, parthanatos is critical in the therapy and prevention of many diseases. In this work, the hallmarks and molecular mechanisms of parthanatos and its related disorders are summarized. The questions raised by the recent findings are also presented. Further understanding of parthanatos will provide a new treatment option for associated conditions.
Collapse
|
26
|
Calpains as mechanistic drivers and therapeutic targets for ocular disease. Trends Mol Med 2022; 28:644-661. [PMID: 35641420 PMCID: PMC9345745 DOI: 10.1016/j.molmed.2022.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022]
Abstract
Ophthalmic neurodegenerative diseases encompass a wide array of molecular pathologies unified by calpain dysregulation. Calpains are calcium-dependent proteases that perpetuate cellular death and inflammation when hyperactivated. Calpain inhibition trials in other organs have faced pharmacological challenges, but the eye offers many advantages for the development and testing of targeted molecular therapeutics, including small molecules, peptides, engineered proteins, drug implants, and gene-based therapies. This review highlights structural mechanisms underlying calpain activation, distinct cellular expression patterns, and in vivo models that link calpain hyperactivity to human retinal and developmental disease. Optimizing therapeutic approaches for calpain-mediated eye diseases can help accelerate clinically feasible strategies for treating calpain dysregulation in other diseased tissues.
Collapse
|
27
|
Rasmussen M, Zhou J, Schwede F, Ekström P. Enhanced cGMP Interactor Rap Guanine Exchange Factor 4 (EPAC2) Expression and Activity in Degenerating Photoreceptors: A Neuroprotective Response? Int J Mol Sci 2022; 23:ijms23094619. [PMID: 35563009 PMCID: PMC9103912 DOI: 10.3390/ijms23094619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/17/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Abstract
The disease retinitis pigmentosa (RP) leads to photoreceptor degeneration by a yet undefined mechanism(s). In several RP mouse models (i.e., rd mice), a high cyclic GMP (cGMP) level within photoreceptors is detected, suggesting that cGMP plays a role in degeneration. The rap guanine exchange factor 4 (EPAC2) is activated by cyclic AMP (cAMP) and is an accepted cGMP-interacting protein. It is unclear whether and how cGMP interacts with EPAC2 in degenerating photoreceptors; we therefore investigated EPAC2 expression and interactions with cGMP and cAMP in retinas of the rd1 and rd10 models for retinal degeneration. EPAC2 expression in the photoreceptor layer increased significantly during rd1 and rd10 degeneration, and an increase in EPAC2 interactions with cGMP but not cAMP in the rd1 was also seen via a proximity ligation assay on histological sections. Retinal explant cultures revealed that pharmacological inhibition of the EPAC2 activity reduced the photoreceptor layer thickness in the rd10 retina, suggesting that EPAC2 inhibition promotes degeneration. Taken together, our results support the hypothesis that high degeneration-related cGMP leads to increased EPAC2 and cGMP interactions, inhibiting EPAC2. By inference, EPAC2 could have neuroprotective capacities that may be exploited in the future.
Collapse
Affiliation(s)
- Michel Rasmussen
- Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, 22184 Lund, Sweden; (J.Z.); (P.E.)
- Correspondence:
| | - Jiaming Zhou
- Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, 22184 Lund, Sweden; (J.Z.); (P.E.)
| | - Frank Schwede
- BIOLOG Life Science Institute GmbH & Co. KG, 28199 Bremen, Germany;
| | - Per Ekström
- Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, 22184 Lund, Sweden; (J.Z.); (P.E.)
| |
Collapse
|
28
|
Belhadj S, Hermann NS, Zhu Y, Christensen G, Strasser T, Paquet-Durand F. Visualizing Cell Death in Live Retina: Using Calpain Activity Detection as a Biomarker for Retinal Degeneration. Int J Mol Sci 2022; 23:ijms23073892. [PMID: 35409251 PMCID: PMC8999672 DOI: 10.3390/ijms23073892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Calpains are a family of calcium-activated proteases involved in numerous disorders. Notably, previous studies have shown that calpain activity was substantially increased in various models for inherited retinal degeneration (RD). In the present study, we tested the capacity of the calpain-specific substrate t-BOC-Leu-Met-CMAC to detect calpain activity in living retina, in organotypic retinal explant cultures derived from wild-type mice, as well as from rd1 and RhoP23H/+ RD-mutant mice. Test conditions were refined until the calpain substrate readily detected large numbers of cells in the photoreceptor layer of RD retina but not in wild-type retina. At the same time, the calpain substrate was not obviously toxic to photoreceptor cells. Comparison of calpain activity with immunostaining for activated calpain-2 furthermore suggested that individual calpain isoforms may be active in distinct temporal stages of photoreceptor cell death. Notably, calpain-2 activity may be a relatively short-lived event, occurring only towards the end of the cell-death process. Finally, our results support the development of calpain activity detection as a novel in vivo biomarker for RD suitable for combination with non-invasive imaging techniques.
Collapse
Affiliation(s)
- Soumaya Belhadj
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (S.B.); (Y.Z.); (G.C.)
- Graduate Training Center of Neuroscience, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
| | - Nina Sofia Hermann
- Graduate Training Center of Neuroscience, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
| | - Yu Zhu
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (S.B.); (Y.Z.); (G.C.)
- Graduate Training Center of Neuroscience, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
| | - Gustav Christensen
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (S.B.); (Y.Z.); (G.C.)
- Graduate Training Center of Neuroscience, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
| | - Torsten Strasser
- Applied Vision Research Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
- University Eye Hospital Tübingen, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany
| | - François Paquet-Durand
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (S.B.); (Y.Z.); (G.C.)
- Correspondence:
| |
Collapse
|
29
|
Yan J, Günter A, Das S, Mühlfriedel R, Michalakis S, Jiao K, Seeliger MW, Paquet-Durand F. Inherited Retinal Degeneration: PARP-Dependent Activation of Calpain Requires CNG Channel Activity. Biomolecules 2022; 12:biom12030455. [PMID: 35327647 PMCID: PMC8946186 DOI: 10.3390/biom12030455] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 01/27/2023] Open
Abstract
Inherited retinal degenerations (IRDs) are a group of blinding diseases, typically involving a progressive loss of photoreceptors. The IRD pathology is often based on an accumulation of cGMP in photoreceptors and associated with the excessive activation of calpain and poly (ADP-ribose) polymerase (PARP). Inhibitors of calpain or PARP have shown promise in preventing photoreceptor cell death, yet the relationship between these enzymes remains unclear. To explore this further, organotypic retinal explant cultures derived from wild-type and IRD-mutant mice were treated with inhibitors specific for calpain, PARP, and voltage-gated Ca2+ channels (VGCCs). The outcomes were assessed using in situ activity assays for calpain and PARP and immunostaining for activated calpain-2, poly (ADP-ribose), and cGMP, as well as the TUNEL assay for cell death detection. The IRD models included the Pde6b-mutant rd1 mouse and rd1*Cngb1−/− double-mutant mice, which lack the beta subunit of the rod cyclic nucleotide-gated (CNG) channel and are partially protected from rd1 degeneration. We confirmed that an inhibition of either calpain or PARP reduces photoreceptor cell death in rd1 retina. However, while the activity of calpain was decreased by the inhibition of PARP, calpain inhibition did not alter the PARP activity. A combination treatment with calpain and PARP inhibitors did not synergistically reduce cell death. In the slow degeneration of rd1*Cngb1−/− double mutant, VGCC inhibition delayed photoreceptor cell death, while PARP inhibition did not. Our results indicate that PARP acts upstream of calpain and that both are part of the same degenerative pathway in Pde6b-dependent photoreceptor degeneration. While PARP activation may be associated with CNG channel activity, calpain activation is linked to VGCC opening. Overall, our data highlights PARP as a target for therapeutic interventions in IRD-type diseases.
Collapse
Affiliation(s)
- Jie Yan
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (J.Y.); (S.D.)
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Alexander Günter
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (A.G.); (R.M.)
| | - Soumyaparna Das
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (J.Y.); (S.D.)
| | - Regine Mühlfriedel
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (A.G.); (R.M.)
| | - Stylianos Michalakis
- Department of Ophthalmology, University Hospital, LMU Munich, 80539 München, Germany;
| | - Kangwei Jiao
- Key Laboratory of Yunnan Province, Affiliated Hospital of Yunnan University, Kunming 650051, China;
| | - Mathias W. Seeliger
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (A.G.); (R.M.)
- Correspondence: (M.W.S.); (F.P.-D.)
| | - François Paquet-Durand
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (J.Y.); (S.D.)
- Correspondence: (M.W.S.); (F.P.-D.)
| |
Collapse
|
30
|
Roy A, Tolone A, Hilhorst R, Groten J, Tomar T, Paquet-Durand F. Kinase activity profiling identifies putative downstream targets of cGMP/PKG signaling in inherited retinal neurodegeneration. Cell Death Dis 2022; 8:93. [PMID: 35241647 PMCID: PMC8894370 DOI: 10.1038/s41420-022-00897-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 11/15/2022]
Abstract
Inherited retinal diseases (IRDs) are a group of neurodegenerative disorders that lead to photoreceptor cell death and eventually blindness. IRDs are characterised by a high genetic heterogeneity, making it imperative to design mutation-independent therapies. Mutations in a number of IRD disease genes have been associated with a rise of cyclic 3’,5’-guanosine monophosphate (cGMP) levels in photoreceptors. Accordingly, the cGMP-dependent protein kinase (PKG) has emerged as a new potential target for the mutation-independent treatment of IRDs. However, the substrates of PKG and the downstream degenerative pathways triggered by its activity have yet to be determined. Here, we performed kinome activity profiling of different murine organotypic retinal explant cultures (diseased rd1 and wild-type controls) using multiplex peptide microarrays to identify proteins whose phosphorylation was significantly altered by PKG activity. In addition, we tested the downstream effect of a known PKG inhibitor CN03 in these organotypic retina cultures. Among the PKG substrates were potassium channels belonging to the Kv1 family (KCNA3, KCNA6), cyclic AMP-responsive element-binding protein 1 (CREB1), DNA topoisomerase 2-α (TOP2A), 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (F263), and the glutamate ionotropic receptor kainate 2 (GRIK2). The retinal expression of these PKG targets was further confirmed by immunofluorescence and could be assigned to various neuronal cell types, including photoreceptors, horizontal cells, and ganglion cells. Taken together, this study confirmed the key role of PKG in photoreceptor cell death and identified new downstream targets of cGMP/PKG signalling that will improve the understanding of the degenerative mechanisms underlying IRDs.
Collapse
Affiliation(s)
- Akanksha Roy
- Division of Toxicology, Wageningen University and Research, 96708 WE, Wageningen, The Netherlands.,PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands
| | - Arianna Tolone
- Cell Death Mechanism Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität, Tübingen, 72072, Germany
| | - Riet Hilhorst
- PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands
| | - John Groten
- Division of Toxicology, Wageningen University and Research, 96708 WE, Wageningen, The Netherlands.,PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands
| | - Tushar Tomar
- PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands.
| | - François Paquet-Durand
- Cell Death Mechanism Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität, Tübingen, 72072, Germany.
| |
Collapse
|
31
|
Sharrock AV, Mulligan TS, Hall KR, Williams EM, White DT, Zhang L, Emmerich K, Matthews F, Nimmagadda S, Washington S, Le KD, Meir-Levi D, Cox OL, Saxena MT, Calof AL, Lopez-Burks ME, Lander AD, Ding D, Ji H, Ackerley DF, Mumm JS. NTR 2.0: a rationally engineered prodrug-converting enzyme with substantially enhanced efficacy for targeted cell ablation. Nat Methods 2022; 19:205-215. [PMID: 35132245 PMCID: PMC8851868 DOI: 10.1038/s41592-021-01364-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/29/2021] [Indexed: 11/12/2022]
Abstract
Transgenic expression of bacterial nitroreductase (NTR) enzymes sensitizes eukaryotic cells to prodrugs such as metronidazole (MTZ), enabling selective cell-ablation paradigms that have expanded studies of cell function and regeneration in vertebrates. However, first-generation NTRs required confoundingly toxic prodrug treatments to achieve effective cell ablation, and some cell types have proven resistant. Here we used rational engineering and cross-species screening to develop an NTR variant, NTR 2.0, which exhibits ~100-fold improvement in MTZ-mediated cell-specific ablation efficacy, eliminating the need for near-toxic prodrug treatment regimens. NTR 2.0 therefore enables sustained cell-loss paradigms and ablation of previously resistant cell types. These properties permit enhanced interrogations of cell function, extended challenges to the regenerative capacities of discrete stem cell niches, and novel modeling of chronic degenerative diseases. Accordingly, we have created a series of bipartite transgenic reporter/effector resources to facilitate dissemination of NTR 2.0 to the research community.
Collapse
Affiliation(s)
- Abigail V Sharrock
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Timothy S Mulligan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Kelsi R Hall
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Elsie M Williams
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - David T White
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Liyun Zhang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Frazer Matthews
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Saumya Nimmagadda
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Selena Washington
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Katherine D Le
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Danielle Meir-Levi
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Olivia L Cox
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Meera T Saxena
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
- Luminomics, Baltimore, MD, USA
| | - Anne L Calof
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Martha E Lopez-Burks
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Arthur D Lander
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Ding Ding
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - David F Ackerley
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.
- Centre for Biodiscovery and Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington, New Zealand.
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA.
- Department of Genetic Medicine, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
32
|
Pang X, Yan R, Li L, Wang P, Zhang Y, Liu Y, Liu P, Dong W, Miao P, Mei Q. Non-doped and non-modified carbon dots with high quantum yield for the chemosensing of uric acid and living cell imaging. Anal Chim Acta 2022; 1199:339571. [DOI: 10.1016/j.aca.2022.339571] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/01/2022] [Accepted: 02/01/2022] [Indexed: 01/13/2023]
|
33
|
Jiang K, Mondal AK, Adlakha YK, Gumerson J, Aponte A, Gieser L, Kim JW, Boleda A, Brooks MJ, Nellissery J, Fox DA, Balaban R, Covian R, Swaroop A. Multiomics analyses reveal early metabolic imbalance and mitochondrial stress in neonatal photoreceptors leading to cell death in Pde6brd1/rd1 mouse model of retinal degeneration. Hum Mol Genet 2022; 31:2137-2154. [PMID: 35075486 PMCID: PMC9618164 DOI: 10.1093/hmg/ddac013] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/17/2021] [Accepted: 01/10/2022] [Indexed: 01/26/2023] Open
Abstract
Retinal diseases exhibit extensive genetic heterogeneity and complex etiology with varying onset and severity. Mutations in over 200 genes can lead to photoreceptor dysfunction and/or cell death in retinal neurodegeneration. To deduce molecular pathways that initiate and/or drive cell death, we adopted a temporal multiomics approach and examined molecular and cellular events in newborn and developing photoreceptors before the onset of degeneration in a widely-used Pde6brd1/rd1 (rd1) mouse, a model of autosomal recessive retinitis pigmentosa caused by PDE6B mutations. Transcriptome profiling of neonatal and developing rods from the rd1 retina revealed early downregulation of genes associated with anabolic pathways and energy metabolism. Quantitative proteomics of rd1 retina showed early changes in calcium signaling and oxidative phosphorylation, with specific partial bypass of complex I electron transfer, which precede the onset of cell death. Concurrently, we detected alterations in central carbon metabolism, including dysregulation of components associated with glycolysis, pentose phosphate and purine biosynthesis. Ex vivo assays of oxygen consumption and transmission electron microscopy validated early and progressive mitochondrial stress and abnormalities in mitochondrial structure and function of rd1 rods. These data uncover mitochondrial overactivation and related metabolic alterations as determinants of early pathology and implicate aberrant calcium signaling as an initiator of higher mitochondrial stress. Our studies thus provide a mechanistic framework with mitochondrial damage and metabolic disruptions as early drivers of photoreceptor cell death in retinal degeneration.
Collapse
Affiliation(s)
| | | | - Yogita K Adlakha
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892, USA,Translational Health Science and Technology Institute, National Capital Region Biotech Cluster, Faridabad, India
| | - Jessica Gumerson
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892, USA
| | - Angel Aponte
- Proteomics Core Facility, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Linn Gieser
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892, USA
| | - Jung-Woong Kim
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892, USA,Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Alexis Boleda
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892, USA,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20740, USA
| | - Matthew J Brooks
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892, USA
| | - Jacob Nellissery
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892, USA
| | - Donald A Fox
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892, USA
| | - Robert Balaban
- Laboratory of Cardiac Energetics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Raul Covian
- Laboratory of Cardiac Energetics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anand Swaroop
- To whom correspondence should be addressed. Tel: +301-435-5754; Fax: 301-480-9917;
| |
Collapse
|
34
|
Das S, Popp V, Power M, Groeneveld K, Yan J, Melle C, Rogerson L, Achury M, Schwede F, Strasser T, Euler T, Paquet-Durand F, Nache V. Redefining the role of Ca 2+-permeable channels in photoreceptor degeneration using diltiazem. Cell Death Dis 2022; 13:47. [PMID: 35013127 PMCID: PMC8748460 DOI: 10.1038/s41419-021-04482-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/07/2021] [Accepted: 11/23/2021] [Indexed: 12/18/2022]
Abstract
Hereditary degeneration of photoreceptors has been linked to over-activation of Ca2+-permeable channels, excessive Ca2+-influx, and downstream activation of Ca2+-dependent calpain-type proteases. Unfortunately, after more than 20 years of pertinent research, unequivocal evidence proving significant and reproducible photoreceptor protection with Ca2+-channel blockers is still lacking. Here, we show that both D- and L-cis enantiomers of the anti-hypertensive drug diltiazem were very effective at blocking photoreceptor Ca2+-influx, most probably by blocking the pore of Ca2+-permeable channels. Yet, unexpectedly, this block neither reduced the activity of calpain-type proteases, nor did it result in photoreceptor protection. Remarkably, application of the L-cis enantiomer of diltiazem even led to a strong increase in photoreceptor cell death. These findings shed doubt on the previously proposed links between Ca2+ and retinal degeneration and are highly relevant for future therapy development as they may serve to refocus research efforts towards alternative, Ca2+-independent degenerative mechanisms.
Collapse
Affiliation(s)
- Soumyaparna Das
- Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany
| | - Valerie Popp
- Institute of Physiology II, University Hospital Jena, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Michael Power
- Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience (CIN), University of Tübingen, 72076, Tübingen, Germany
| | - Kathrin Groeneveld
- Institute of Physiology II, University Hospital Jena, Friedrich Schiller University Jena, 07743, Jena, Germany.,Biomolecular Photonics Group, University Hospital Jena, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Jie Yan
- Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany
| | - Christian Melle
- Biomolecular Photonics Group, University Hospital Jena, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Luke Rogerson
- Werner Reichardt Centre for Integrative Neuroscience (CIN), University of Tübingen, 72076, Tübingen, Germany
| | - Marlly Achury
- Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany
| | - Frank Schwede
- BIOLOG Life Science Institute GmbH & Co KG, 28199, Bremen, Germany
| | - Torsten Strasser
- Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany
| | - Thomas Euler
- Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience (CIN), University of Tübingen, 72076, Tübingen, Germany
| | | | - Vasilica Nache
- Institute of Physiology II, University Hospital Jena, Friedrich Schiller University Jena, 07743, Jena, Germany.
| |
Collapse
|
35
|
Cha S, Choi KE, Ahn J, Yoo M, Jeong Y, Kim SW, Goo YS. Electrical response of retinal ganglion cells in an N-methyl-N-nitrosourea-induced retinal degeneration porcine model. Sci Rep 2021; 11:24135. [PMID: 34921172 PMCID: PMC8683404 DOI: 10.1038/s41598-021-03439-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/02/2021] [Indexed: 12/28/2022] Open
Abstract
Retinal prosthesis is regarded as the treatment for vision restoration in the blind with retinal degeneration (RD) due to the loss of photoreceptors. A strategy for retinal prosthesis is to electrically activate surviving neurons. The retina’s response to electrical stimulation in a larger RD model has not been studied yet. Therefore, in this study, we investigated electrically evoked retinal responses in a previously validated N-methyl-N-nitrosourea (MNU)-induced porcine RD model. Electrically evoked responses were evaluated based on the number of retinal ganglion cell (RGC) spikes via multichannel recordings. Stimulation pulses were applied to degenerative and wild-type retinas with pulse modulation. Compared to wild-type retinas, degenerative retinas showed higher threshold values of pulse amplitude and pulse duration. The rate of increase in the number of RGC spikes relative to stimulus intensity was lower in degenerative retinas than in normal retinas. In severely degenerated retinas, few RGCs showed electrically evoked spikes. Our results suggest that the degenerative porcine retina requires a higher charge than the normal porcine retina. In the early stage of RD, it is easier to induce RGC spikes through electrical stimulation using retinal prosthesis; however, when the degeneration is severe, there may be difficulty recovering patient vision.
Collapse
Affiliation(s)
- Seongkwang Cha
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, 28644, Korea
| | - Kwang-Eon Choi
- Department of Ophthalmology, Korea University College of Medicine, Seoul, 08308, Korea
| | - Jungryul Ahn
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, 28644, Korea
| | - Minsu Yoo
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, 28644, Korea
| | - Yurim Jeong
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, 28644, Korea
| | - Seong-Woo Kim
- Department of Ophthalmology, Korea University College of Medicine, Seoul, 08308, Korea.
| | - Yong Sook Goo
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, 28644, Korea.
| |
Collapse
|
36
|
Wei C, Li Y, Feng X, Hu Z, Paquet-Durand F, Jiao K. RNA Biological Characteristics at the Peak of Cell Death in Different Hereditary Retinal Degeneration Mutants. Front Genet 2021; 12:728791. [PMID: 34777465 PMCID: PMC8586524 DOI: 10.3389/fgene.2021.728791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose: The present work investigated changes in the gene expression, molecular mechanisms, and pathogenesis of inherited retinal degeneration (RD) in three different disease models, to identify predictive biomarkers for their varied phenotypes and to provide a better scientific basis for their diagnosis, treatment, and prevention. Methods: Differentially expressed genes (DEGs) between retinal tissue from RD mouse models obtained during the photoreceptor cell death peak period (Pde6b rd1 at post-natal (PN) day 13, Pde6b rd10 at PN23, Prph rd2 at PN29) and retinal tissue from C3H wild-type mice were identified using Illumina high-throughput RNA-sequencing. Co-expression gene modules were identified using a combination of GO and KEGG enrichment analyses and gene co-expression network analysis. CircRNA-miRNA-mRNA network interactions were studied by genome-wide circRNA screening. Results: Pde6b rd1 , Pde6b rd10 , and Prph rd2 mice had 1,926, 3,096, and 375 DEGs, respectively. Genes related to ion channels, stress, inflammatory processes, tumor necrosis factor (TNF) production, and microglial cell activation were up-regulated, while genes related to endoplasmic reticulum regulation, metabolism, and homeostasis were down-regulated. Differential expression of transcription factors and non-coding RNAs generally implicated in other human diseases was detected (e.g., glaucoma, diabetic retinopathy, and inherited retinal degeneration). CircRNA-miRNA-mRNA network analysis indicated that these factors may be involved in photoreceptor cell death. Moreover, excessive cGMP accumulation causes photoreceptor cell death, and cGMP-related genes were generally affected by different pathogenic gene mutations. Conclusion: We screened genes and pathways related to photoreceptor cell death. Additionally, up-stream regulatory factors, such as transcription factors and non-coding RNA and their interaction networks were analyzed. Furthermore, RNAs involved in RD were functionally annotated. Overall, this study lays a foundation for future studies on photoreceptor cell death mechanisms.
Collapse
Affiliation(s)
- Chunling Wei
- Kunming Medical University, Kunming, China.,Department of Ophthalmology, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, China
| | - Yan Li
- Department of Ophthalmology, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, China.,Key Laboratory of Yunnan Province, Yunnan Eye Institute, Kunming, China
| | - Xiaoxiao Feng
- Department of Ophthalmology, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, China.,Key Laboratory of Yunnan Province, Yunnan Eye Institute, Kunming, China
| | - Zhulin Hu
- Department of Ophthalmology, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, China.,Key Laboratory of Yunnan Province, Yunnan Eye Institute, Kunming, China
| | - François Paquet-Durand
- Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Kangwei Jiao
- Department of Ophthalmology, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, China.,Key Laboratory of Yunnan Province, Yunnan Eye Institute, Kunming, China
| |
Collapse
|
37
|
Yang M, Chen Y, Vagionitis S, Körtvely E, Ueffing M, Schmachtenberg O, Hu Z, Jiao K, Paquet-Durand F. Expression of glucose transporter-2 in murine retina: Evidence for glucose transport from horizontal cells to photoreceptor synapses. J Neurochem 2021; 160:283-296. [PMID: 34726780 DOI: 10.1111/jnc.15533] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 01/30/2023]
Abstract
The retina has the highest relative energy consumption of any tissue, depending on a steady supply of glucose from the bloodstream. Glucose uptake is mediated by specific transporters whose regulation and expression are critical for the pathogenesis of many diseases, including diabetes and diabetic retinopathy. Here, we used immunofluorescence to show that glucose transporter-2 (GLUT2) is expressed in horizontal cells of the mouse neuroretina in proximity to inner retinal capillaries. To study the function of GLUT2 in the murine retina, we used organotypic retinal explants, cultivated under entirely controlled, serum-free conditions and exposed them to streptozotocin, a cytotoxic drug transported exclusively by GLUT2. Contrary to our expectations, streptozotocin did not measurably affect horizontal cell viability, while it ablated rod and cone photoreceptors in a concentration-dependent manner. Staining for poly-ADP-ribose (PAR) indicated that the detrimental effect of streptozotocin on photoreceptors may be associated with DNA damage. The negative effect of streptozotocin on the viability of rod photoreceptors was counteracted by co-administration of either the inhibitor of connexin-formed hemi-channels meclofenamic acid or the blocker of clathrin-mediated endocytosis dynasore. Remarkably, cone photoreceptors were not protected from streptozotocin-induced degeneration by neither of the two drugs. Overall, these data suggest the existence of a GLUT2-dependent glucose transport shuttle, from horizontal cells into photoreceptor synapses. Moreover, our study points at different glucose uptake mechanisms in rod and cone photoreceptors.
Collapse
Affiliation(s)
- Ming Yang
- Affiliated Hospital of Yunnan University & 2nd People's Hospital of Yunnan Province, Kunming, China.,Yunnan Eye Institute & Key Laboratory of Yunnan Province, Kunming, China.,1st Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yiyi Chen
- Institute for Ophthalmic Research, Eberhard-Karls-Universität, Tübingen, Germany
| | - Stavros Vagionitis
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Elöd Körtvely
- Roche Pharma Research and Early Development, Immunology, Infectious Diseases and Ophthalmology (I2O), Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Marius Ueffing
- Institute for Ophthalmic Research, Eberhard-Karls-Universität, Tübingen, Germany
| | - Oliver Schmachtenberg
- CINV, Instituto de Biología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Zhulin Hu
- Affiliated Hospital of Yunnan University & 2nd People's Hospital of Yunnan Province, Kunming, China.,Yunnan Eye Institute & Key Laboratory of Yunnan Province, Kunming, China
| | - Kangwei Jiao
- Affiliated Hospital of Yunnan University & 2nd People's Hospital of Yunnan Province, Kunming, China.,Yunnan Eye Institute & Key Laboratory of Yunnan Province, Kunming, China
| | | |
Collapse
|
38
|
Biasi A, Marino V, Dal Cortivo G, Maltese PE, Modarelli AM, Bertelli M, Colombo L, Dell’Orco D. A Novel GUCA1A Variant Associated with Cone Dystrophy Alters cGMP Signaling in Photoreceptors by Strongly Interacting with and Hyperactivating Retinal Guanylate Cyclase. Int J Mol Sci 2021; 22:ijms221910809. [PMID: 34639157 PMCID: PMC8509414 DOI: 10.3390/ijms221910809] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 11/19/2022] Open
Abstract
Guanylate cyclase-activating protein 1 (GCAP1), encoded by the GUCA1A gene, is a neuronal calcium sensor protein involved in shaping the photoresponse kinetics in cones and rods. GCAP1 accelerates or slows the cGMP synthesis operated by retinal guanylate cyclase (GC) based on the light-dependent levels of intracellular Ca2+, thereby ensuring a timely regulation of the phototransduction cascade. We found a novel variant of GUCA1A in a patient affected by autosomal dominant cone dystrophy (adCOD), leading to the Asn104His (N104H) amino acid substitution at the protein level. While biochemical analysis of the recombinant protein showed impaired Ca2+ sensitivity of the variant, structural properties investigated by circular dichroism and limited proteolysis excluded major structural rearrangements induced by the mutation. Analytical gel filtration profiles and dynamic light scattering were compatible with a dimeric protein both in the presence of Mg2+ alone and Mg2+ and Ca2+. Enzymatic assays showed that N104H-GCAP1 strongly interacts with the GC, with an affinity that doubles that of the WT. The doubled IC50 value of the novel variant (520 nM for N104H vs. 260 nM for the WT) is compatible with a constitutive activity of GC at physiological levels of Ca2+. The structural region at the interface with the GC may acquire enhanced flexibility under high Ca2+ conditions, as suggested by 2 μs molecular dynamics simulations. The altered interaction with GC would cause hyper-activity of the enzyme at both low and high Ca2+ levels, which would ultimately lead to toxic accumulation of cGMP and Ca2+ in the photoreceptor outer segment, thus triggering cell death.
Collapse
Affiliation(s)
- Amedeo Biasi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy; (A.B.); (V.M.); (G.D.C.)
| | - Valerio Marino
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy; (A.B.); (V.M.); (G.D.C.)
| | - Giuditta Dal Cortivo
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy; (A.B.); (V.M.); (G.D.C.)
| | | | - Antonio Mattia Modarelli
- Department of Ophthalmology, ASST Santi Paolo e Carlo Hospital, University of Milan, 20142 Milano, Italy;
| | - Matteo Bertelli
- MAGI’S Lab s.r.l., 38068 Rovereto, Italy; (P.E.M.); (M.B.)
- MAGI Euregio, 39100 Bolzano, Italy
| | - Leonardo Colombo
- Department of Ophthalmology, ASST Santi Paolo e Carlo Hospital, University of Milan, 20142 Milano, Italy;
- Correspondence: (L.C.); (D.D.); Tel.: +39-02-81844301 (L.C.); +39-045-802-7637 (D.D.)
| | - Daniele Dell’Orco
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy; (A.B.); (V.M.); (G.D.C.)
- Correspondence: (L.C.); (D.D.); Tel.: +39-02-81844301 (L.C.); +39-045-802-7637 (D.D.)
| |
Collapse
|
39
|
Yan J, Chen Y, Zhu Y, Paquet-Durand F. Programmed Non-Apoptotic Cell Death in Hereditary Retinal Degeneration: Crosstalk between cGMP-Dependent Pathways and PARthanatos? Int J Mol Sci 2021; 22:10567. [PMID: 34638907 PMCID: PMC8508647 DOI: 10.3390/ijms221910567] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/20/2022] Open
Abstract
Programmed cell death (PCD) is a highly regulated process that results in the orderly destruction of a cell. Many different forms of PCD may be distinguished, including apoptosis, PARthanatos, and cGMP-dependent cell death. Misregulation of PCD mechanisms may be the underlying cause of neurodegenerative diseases of the retina, including hereditary retinal degeneration (RD). RD relates to a group of diseases that affect photoreceptors and that are triggered by gene mutations that are often well known nowadays. Nevertheless, the cellular mechanisms of PCD triggered by disease-causing mutations are still poorly understood, and RD is mostly still untreatable. While investigations into the neurodegenerative mechanisms of RD have focused on apoptosis in the past two decades, recent evidence suggests a predominance of non-apoptotic processes as causative mechanisms. Research into these mechanisms carries the hope that the knowledge created can eventually be used to design targeted treatments to prevent photoreceptor loss. Hence, in this review, we summarize studies on PCD in RD, including on apoptosis, PARthanatos, and cGMP-dependent cell death. Then, we focus on a possible interplay between these mechanisms, covering cGMP-signaling targets, overactivation of poly(ADP-ribose)polymerase (PARP), energy depletion, Ca2+-permeable channels, and Ca2+-dependent proteases. Finally, an outlook is given into how specific features of cGMP-signaling and PARthanatos may be targeted by therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - François Paquet-Durand
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076 Tübingen, Germany; (J.Y.); (Y.C.); (Y.Z.)
| |
Collapse
|
40
|
The 3D organisation of mitochondria in primate photoreceptors. Sci Rep 2021; 11:18863. [PMID: 34552195 PMCID: PMC8458444 DOI: 10.1038/s41598-021-98409-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/17/2021] [Indexed: 11/08/2022] Open
Abstract
Vertebrate photoreceptors contain large numbers of closely-packed mitochondria which sustain the high metabolic demands of these cells. These mitochondria populations are dynamic and undergo fusion and fission events. This activity serves to maintain the population in a healthy state. In the event of mitochondrial damage, sub-domains, or indeed whole mitochondria, can be degraded and population homeostasis achieved. If this process is overwhelmed cell death may result. Death of photoreceptors contributes to loss of vision in aging individuals and is associated with many eye diseases. In this study we used serial block face scanning electron microscopy of adult Macaca fascicularis retinae to examine the 3D structure of mitochondria in rod and cone photoreceptors. We show that healthy-looking photoreceptors contain mitochondria exhibiting a range of shapes which are associated with different regions of the cell. In some photoreceptors we observe mitochondrial swelling and other changes often associated with cellular stress. In rods and cones that appear stressed we identify elongated domains of mitochondria with densely-packed normal cristae associated with photoreceptor ciliary rootlet bundles. We observe mitochondrial fission and mitochondrion fragments localised to these domains. Swollen mitochondria with few intact cristae are located towards the periphery of the photoreceptor inner-segment in rods, whilst they are found throughout the cell in cones. Swollen mitochondria exhibit sites on the mitochondrial inner membrane which have undergone complex invagination resulting in membranous, electron-dense aggregates. Membrane contact occurs between the mitochondrion and the photoreceptor plasma membrane in the vicinity of these aggregates, and a series of subsequent membrane fusions results in expulsion of the mitochondrial aggregate from the photoreceptor. These events are primarily associated with rods. The potential fate of this purged material and consequences of its clearance by retinal pigment epithelia are discussed.
Collapse
|
41
|
Andreazzoli M, Barravecchia I, De Cesari C, Angeloni D, Demontis GC. Inducible Pluripotent Stem Cells to Model and Treat Inherited Degenerative Diseases of the Outer Retina: 3D-Organoids Limitations and Bioengineering Solutions. Cells 2021; 10:cells10092489. [PMID: 34572137 PMCID: PMC8471616 DOI: 10.3390/cells10092489] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal degenerations (IRD) affecting either photoreceptors or pigment epithelial cells cause progressive visual loss and severe disability, up to complete blindness. Retinal organoids (ROs) technologies opened up the development of human inducible pluripotent stem cells (hiPSC) for disease modeling and replacement therapies. However, hiPSC-derived ROs applications to IRD presently display limited maturation and functionality, with most photoreceptors lacking well-developed outer segments (OS) and light responsiveness comparable to their adult retinal counterparts. In this review, we address for the first time the microenvironment where OS mature, i.e., the subretinal space (SRS), and discuss SRS role in photoreceptors metabolic reprogramming required for OS generation. We also address bioengineering issues to improve culture systems proficiency to promote OS maturation in hiPSC-derived ROs. This issue is crucial, as satisfying the demanding metabolic needs of photoreceptors may unleash hiPSC-derived ROs full potential for disease modeling, drug development, and replacement therapies.
Collapse
Affiliation(s)
| | - Ivana Barravecchia
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy;
- Institute of Life Sciences, Scuola Superiore Sant’Anna, 56124 Pisa, Italy;
| | | | - Debora Angeloni
- Institute of Life Sciences, Scuola Superiore Sant’Anna, 56124 Pisa, Italy;
| | - Gian Carlo Demontis
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy;
- Correspondence: (M.A.); (G.C.D.)
| |
Collapse
|
42
|
cGMP-PKG dependent transcriptome in normal and degenerating retinas: Novel insights into the retinitis pigmentosa pathology. Exp Eye Res 2021; 212:108752. [PMID: 34478738 DOI: 10.1016/j.exer.2021.108752] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 06/25/2021] [Accepted: 08/25/2021] [Indexed: 12/26/2022]
Abstract
Retinitis Pigmentosa represents a group of genetic disorders that cause progressive vision loss via degeneration of photoreceptors, but there is in principle no treatment available. For any therapy development, a deeper comprehension of the disease-leading mechanism(s) at the molecular level is needed. Here we focused on the cGMP-PKG system, which has been suggested to be a driver in several models of the disease. To gain insights in its downstream signaling we manipulated the cGMP-PKG system with the aid of organotypic retinal explant cultures from either a mouse-based disease model, i.e. the rd1 mouse, or its healthy wild-type counterpart (wt), by adding different types of cGMP analogues to either inhibit or activate PKG in retinal explants from rd1 and wt, respectively. An RNA sequencing was then performed to study the cGMP-PKG dependent transcriptome. Expression changes of gene sets related to specific pathways or functions, that fulfilled criteria involving that the changes should match PKG activation and inhibition, were determined via bioinformatics. The analyses highlighted that several gene sets linked to oxidative phosphorylation and mitochondrial pathways were regulated by this enzyme system. Specifically, the expression of such pathway components was upregulated in the rd1 treated with PKG inhibitor and downregulated in the wt with PKG activator treatment, suggesting that cGMP-PKG act as a negative regulator in this context. Downregulation of energy production pathways may thus play an integral part in the mechanism behind the degeneration for at least several RP mutations.
Collapse
|
43
|
Technological advancements to study cellular signaling pathways in inherited retinal degenerative diseases. Curr Opin Pharmacol 2021; 60:102-110. [PMID: 34388439 DOI: 10.1016/j.coph.2021.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/02/2021] [Indexed: 01/01/2023]
Abstract
Inherited retinal degenerative diseases (IRDs) are rare neurodegenerative disorders with mutations in hundreds of genes leading to vision loss, primarily owing to photoreceptor cell death. This genetic diversity is impeding development of effective treatment options. Gene-based therapies have resulted in the first FDA-approved drug (Luxturna) for RPE65-specific IRD. Although currently explored in clinical trials, genomic medicines are mutation-dependent, hence suitable only for patients harboring a specific mutation. Better understanding of the pathways leading to photoreceptor degeneration may help to determine common targets and develop mutation-independent therapies for larger groups of patients with IRDs. In this review, we discuss the key pathways involved in photoreceptor cell death studied by transcriptomics, proteomics, and metabolomics techniques to identify potential therapeutic targets in IRDs.
Collapse
|
44
|
Quadri M, Comitato A, Palazzo E, Tiso N, Rentsch A, Pellacani G, Marconi A, Marigo V. Activation of cGMP-Dependent Protein Kinase Restricts Melanoma Growth and Invasion by Interfering with the EGF/EGFR Pathway. J Invest Dermatol 2021; 142:201-211. [PMID: 34265328 DOI: 10.1016/j.jid.2021.06.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/07/2021] [Accepted: 06/18/2021] [Indexed: 12/16/2022]
Abstract
Drug resistance mechanisms still characterize metastatic melanoma, despite the new treatments that have been recently developed. Targeting of the cGMP/protein kinase G pathway is emerging as a therapeutic approach in cancer research. In this study, we evaluated the anticancer effects of two polymeric-linked dimeric cGMP analogs able to bind and activate protein kinase G, called protein kinase G activators (PAs) 4 and 5. PA5 was identified as the most effective compound on melanoma cell lines as well as on patient-derived metastatic melanoma cells cultured as three-dimensional spheroids and in a zebrafish melanoma model. PA5 was able to significantly reduce cell viability, size, and invasion of melanoma spheroids. Importantly, PA5 showed a tumor-specific outcome because no toxic effect was observed in healthy melanocytes exposed to the cGMP analog. We defined that by triggering protein kinase G, PA5 interfered with the EGF pathway as shown by lower EGFR phosphorylation and reduction of activated, phosphorylated forms of protein kinase B and extracellular signal‒regulated kinase 1/2 in melanoma cells. Finally, PA5 significantly reduced the metastatic process in zebrafish. These studies open future perspectives for the cGMP analog PA5 as a potential therapeutic strategy for melanoma.
Collapse
Affiliation(s)
- Marika Quadri
- DermoLab, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Modena, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonella Comitato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Palazzo
- DermoLab, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Natascia Tiso
- Laboratory of Developmental Genetics, Department of Biology, University of Padua, Padua, Italy
| | - Andreas Rentsch
- BIOLOG Life Science Institute. Forschungslabor und Biochemica-Vertrieb, Bremen, Germany
| | - Giovanni Pellacani
- DermoLab, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Marconi
- DermoLab, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Modena, Italy.
| | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
45
|
Inhibition of Epigenetic Modifiers LSD1 and HDAC1 Blocks Rod Photoreceptor Death in Mouse Models of Retinitis Pigmentosa. J Neurosci 2021; 41:6775-6792. [PMID: 34193554 DOI: 10.1523/jneurosci.3102-20.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 11/21/2022] Open
Abstract
Epigenetic modifiers are increasingly being investigated as potential therapeutics to modify and overcome disease phenotypes. Diseases of the nervous system present a particular problem as neurons are postmitotic and demonstrate relatively stable gene expression patterns and chromatin organization. We have explored the ability of epigenetic modifiers to prevent degeneration of rod photoreceptors in a mouse model of retinitis pigmentosa (RP), using rd10 mice of both sexes. The histone modification eraser enzymes lysine demethylase 1 (LSD1) and histone deacetylase 1 (HDAC1) are known to have dramatic effects on the development of rod photoreceptors. In the RP mouse model, inhibitors of these enzymes blocked rod degeneration, preserved vision, and affected the expression of multiple genes including maintenance of rod-specific transcripts and downregulation of those involved in inflammation, gliosis, and cell death. The neuroprotective activity of LSD1 inhibitors includes two pathways. First, through targeting histone modifications, they increase accessibility of chromatin and upregulate neuroprotective genes, such as from the Wnt pathway. We propose that this process is going in rod photoreceptors. Second, through nonhistone targets, they inhibit transcription of inflammatory genes and inflammation. This process is going in microglia, and lack of inflammation keeps rod photoreceptors alive.SIGNIFICANCE STATEMENT Retinal degenerations are a leading cause of vision loss. RP is genetically very heterogeneous, and the multiple pathways leading to cell death are one reason for the slow progress in identifying suitable treatments for patients. Here we demonstrate that inhibition of LSD1and HDAC1 in a mouse model of RP leads to preservation of rod photoreceptors and visual function, retaining of expression of rod-specific genes, and with decreased inflammation, cell death, and Müller cell gliosis. We propose that these epigenetic inhibitors cause more open and accessible chromatin, allowing expression of neuroprotective genes. A second mechanism that allows rod photoreceptor survival is suppression of inflammation by epigenetic inhibitors in microglia. Manipulation of epigenetic modifiers is a new strategy to fight neurodegeneration in RP.
Collapse
|
46
|
Zhang L, Chen C, Fu J, Lilley B, Berlinicke C, Hansen B, Ding D, Wang G, Wang T, Shou D, Ye Y, Mulligan T, Emmerich K, Saxena MT, Hall KR, Sharrock AV, Brandon C, Park H, Kam TI, Dawson VL, Dawson TM, Shim JS, Hanes J, Ji H, Liu JO, Qian J, Ackerley DF, Rohrer B, Zack DJ, Mumm JS. Large-scale phenotypic drug screen identifies neuroprotectants in zebrafish and mouse models of retinitis pigmentosa. eLife 2021; 10:e57245. [PMID: 34184634 PMCID: PMC8425951 DOI: 10.7554/elife.57245] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/28/2021] [Indexed: 11/25/2022] Open
Abstract
Retinitis pigmentosa (RP) and associated inherited retinal diseases (IRDs) are caused by rod photoreceptor degeneration, necessitating therapeutics promoting rod photoreceptor survival. To address this, we tested compounds for neuroprotective effects in multiple zebrafish and mouse RP models, reasoning drugs effective across species and/or independent of disease mutation may translate better clinically. We first performed a large-scale phenotypic drug screen for compounds promoting rod cell survival in a larval zebrafish model of inducible RP. We tested 2934 compounds, mostly human-approved drugs, across six concentrations, resulting in 113 compounds being identified as hits. Secondary tests of 42 high-priority hits confirmed eleven lead candidates. Leads were then evaluated in a series of mouse RP models in an effort to identify compounds effective across species and RP models, that is, potential pan-disease therapeutics. Nine of 11 leads exhibited neuroprotective effects in mouse primary photoreceptor cultures, and three promoted photoreceptor survival in mouse rd1 retinal explants. Both shared and complementary mechanisms of action were implicated across leads. Shared target tests implicated parp1-dependent cell death in our zebrafish RP model. Complementation tests revealed enhanced and additive/synergistic neuroprotective effects of paired drug combinations in mouse photoreceptor cultures and zebrafish, respectively. These results highlight the value of cross-species/multi-model phenotypic drug discovery and suggest combinatorial drug therapies may provide enhanced therapeutic benefits for RP patients.
Collapse
Affiliation(s)
- Liyun Zhang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Conan Chen
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Jie Fu
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Brendan Lilley
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Cynthia Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Baranda Hansen
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Ding Ding
- Department of Biostatistics, Johns Hopkins UniversityBaltimoreUnited States
| | - Guohua Wang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Tao Wang
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- School of Chemistry, Xuzhou College of Industrial TechnologyXuzhouChina
- College of Light Industry and Food Engineering, Nanjing Forestry UniversityNanjingChina
| | - Daniel Shou
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Ying Ye
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Timothy Mulligan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- Department of Genetic Medicine, Johns Hopkins UniversityBaltimoreUnited States
| | - Meera T Saxena
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Kelsi R Hall
- School of Biological Sciences, Victoria University of WellingtonWellingtonNew Zealand
| | - Abigail V Sharrock
- Department of Biostatistics, Johns Hopkins UniversityBaltimoreUnited States
- School of Biological Sciences, Victoria University of WellingtonWellingtonNew Zealand
| | - Carlene Brandon
- Department of Ophthalmology, Medical University of South CarolinaCharlestonUnited States
| | - Hyejin Park
- Department of Neurology, Johns Hopkins UniversityBaltimoreUnited States
| | - Tae-In Kam
- Department of Neurology, Johns Hopkins UniversityBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Valina L Dawson
- Department of Neurology, Johns Hopkins UniversityBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins UniversityBaltimoreUnited States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins UniversityBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
| | - Ted M Dawson
- Department of Neurology, Johns Hopkins UniversityBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins UniversityBaltimoreUnited States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins UniversityBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
| | - Joong Sup Shim
- Faculty of Health Sciences, University of Macau, TaipaMacauChina
| | - Justin Hanes
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins UniversityBaltimoreUnited States
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins UniversityBaltimoreUnited States
- Department of Oncology, Johns Hopkins UniversityBaltimoreUnited States
| | - Jiang Qian
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - David F Ackerley
- School of Biological Sciences, Victoria University of WellingtonWellingtonNew Zealand
| | - Baerbel Rohrer
- Department of Ophthalmology, Medical University of South CarolinaCharlestonUnited States
| | - Donald J Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- Department of Genetic Medicine, Johns Hopkins UniversityBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
- Department of Molecular Biology and Genetics, Johns Hopkins UniversityBaltimoreUnited States
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- Department of Genetic Medicine, Johns Hopkins UniversityBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
| |
Collapse
|
47
|
Photoreceptor Phosphodiesterase (PDE6): Structure, Regulatory Mechanisms, and Implications for Treatment of Retinal Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1371:33-59. [PMID: 34170501 DOI: 10.1007/5584_2021_649] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The photoreceptor phosphodiesterase (PDE6) is a member of large family of Class I phosphodiesterases responsible for hydrolyzing the second messengers cAMP and cGMP. PDE6 consists of two catalytic subunits and two inhibitory subunits that form a tetrameric protein. PDE6 is a peripheral membrane protein that is localized to the signal-transducing compartment of rod and cone photoreceptors. As the central effector enzyme of the G-protein coupled visual transduction pathway, activation of PDE6 catalysis causes a rapid decrease in cGMP levels that results in closure of cGMP-gated ion channels in the photoreceptor plasma membrane. Because of its importance in the phototransduction pathway, mutations in PDE6 genes result in various retinal diseases that currently lack therapeutic treatment strategies due to inadequate knowledge of the structure, function, and regulation of this enzyme. This review focuses on recent progress in understanding the structure of the regulatory and catalytic domains of the PDE6 holoenzyme, the central role of the multi-functional inhibitory γ-subunit, the mechanism of activation by the heterotrimeric G protein, transducin, and future directions for pharmacological interventions to treat retinal degenerative diseases arising from mutations in the PDE6 genes.
Collapse
|
48
|
New In Vitro Cellular Model for Molecular Studies of Retinitis Pigmentosa. Int J Mol Sci 2021; 22:ijms22126440. [PMID: 34208617 PMCID: PMC8235468 DOI: 10.3390/ijms22126440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/04/2021] [Accepted: 06/13/2021] [Indexed: 02/05/2023] Open
Abstract
Retinitis pigmentosa (RP) is an inherited form of retinal degeneration characterized by primary rod photoreceptor cell death followed by cone loss. Mutations in several genes linked to the disease cause increased levels of cyclic guanosine monophosphate (cGMP) and calcium ion influxes. The purpose of this project was to develop a new in vitro photoreceptor degeneration model for molecular studies of RP. 661W cells were genetically modified to stably express the neural retina leucine zipper (NRL) transcription factor. One clone (661W-A11) was selected based on the expression of Nrl target genes. 661W-A11 showed a significant increase in expression of rod-specific genes but not of cone-specific genes, compared with 661W cells. Zaprinast was used to inhibit phosphodiesterase 6 (PDE6) activity to mimic photoreceptor degeneration in vitro. The activation of cell death pathways resulting from PDE6 inhibition was confirmed by detection of decreased viability and increased intracellular cGMP and calcium, as well as activation of protein kinase G (PKG) and calpains. In this new in vitro system, we validated the effects of previously published neuroprotective drugs. The 661W-A11 cells may serve as a new model for molecular studies of RP and for high-throughput drug screening.
Collapse
|
49
|
Greenwald SH, Brown EE, Scandura MJ, Hennessey E, Farmer R, Du J, Wang Y, Pierce EA. Mutant Nmnat1 leads to a retina-specific decrease of NAD+ accompanied by increased poly(ADP-ribose) in a mouse model of NMNAT1-associated retinal degeneration. Hum Mol Genet 2021; 30:644-657. [PMID: 33709122 PMCID: PMC8127407 DOI: 10.1093/hmg/ddab070] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/01/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022] Open
Abstract
Nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1) is required for nuclear nicotinamide adenine mononucleotide (NAD+) biosynthesis in all nucleated cells, and despite its functional ubiquity, mutations in this gene lead to an isolated retinal degeneration. The mechanisms underlying how mutant NMNAT1 causes disease are not well understood, nor is the reason why the pathology is confined to the retina. Using a mouse model of NMNAT1-associated retinal degeneration that harbors the p.Val9Met mutation, we tested the hypothesis that decreased function of mutant NMNAT1 has a greater effect on the levels of NAD+ in the retina than elsewhere in the body. Measurements by liquid chromatography with tandem mass spectrometry showed an early and sustained decrease of NAD+ in mutant retinas that was not observed in other tissues. To understand how consumers of nuclear NAD+ are affected by the reduced availability of NAD+ in mutant retinas, poly(ADP-ribose) polymerase (PARP) and nuclear sirtuin activity were evaluated. PARP activity was elevated during disease progression, as evidenced by overproduction of poly(ADP-ribose) (PAR) in photoreceptors, whereas histone deacetylation activity of nuclear sirtuins was not altered. We hypothesized that PARP could be activated because of elevated levels of oxidative stress; however, we did not observe oxidative DNA damage, lipid peroxidation, or a low glutathione to oxidized glutathione ratio. Terminal deoxynucleotidyl transferase dUTP nick end labeling staining revealed that photoreceptors appear to ultimately die by apoptosis, although the low NAD+ levels and overproduction of PAR suggest that cell death may include aspects of the parthanatos cell death pathway.
Collapse
Affiliation(s)
- Scott H Greenwald
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Emily E Brown
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Michael J Scandura
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Erin Hennessey
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Raymond Farmer
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506, USA
- Department of Biochemistry, West Virginia University, Morgantown, WV 26506, USA
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506, USA
- Department of Biochemistry, West Virginia University, Morgantown, WV 26506, USA
| | - Eric A Pierce
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
50
|
Das S, Chen Y, Yan J, Christensen G, Belhadj S, Tolone A, Paquet-Durand F. The role of cGMP-signalling and calcium-signalling in photoreceptor cell death: perspectives for therapy development. Pflugers Arch 2021; 473:1411-1421. [PMID: 33864120 PMCID: PMC8370896 DOI: 10.1007/s00424-021-02556-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
The second messengers, cGMP and Ca2+, have both been implicated in retinal degeneration; however, it is still unclear which of the two is most relevant for photoreceptor cell death. This problem is exacerbated by the close connections and crosstalk between cGMP-signalling and calcium (Ca2+)-signalling in photoreceptors. In this review, we summarize key aspects of cGMP-signalling and Ca2+-signalling relevant for hereditary photoreceptor degeneration. The topics covered include cGMP-signalling targets, the role of Ca2+ permeable channels, relation to energy metabolism, calpain-type proteases, and how the related metabolic processes may trigger and execute photoreceptor cell death. A focus is then put on cGMP-dependent mechanisms and how exceedingly high photoreceptor cGMP levels set in motion cascades of Ca2+-dependent and independent processes that eventually bring about photoreceptor cell death. Finally, an outlook is given into mutation-independent therapeutic approaches that exploit specific features of cGMP-signalling. Such approaches might be combined with suitable drug delivery systems for translation into clinical applications.
Collapse
Affiliation(s)
- Soumyaparna Das
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Yiyi Chen
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Jie Yan
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Gustav Christensen
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Soumaya Belhadj
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Arianna Tolone
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - François Paquet-Durand
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany.
| |
Collapse
|