101
|
Huang H, Zeng J, Kuang X, He F, Yan J, Li B, Liu W, Shen H. Transcriptional patterns of human retinal pigment epithelial cells under protracted high glucose. Mol Biol Rep 2024; 51:477. [PMID: 38573426 DOI: 10.1007/s11033-024-09479-5] [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: 02/15/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND The retinal pigment epithelium (RPE) is essential for retinal homeostasis. Comprehensively exploring the transcriptional patterns of diabetic human RPE promotes the understanding of diabetic retinopathy (DR). METHODS AND RESULTS A total of 4125 differentially expressed genes (DEGs) were screened out from the human primary RPE cells subjected to prolonged high glucose (HG). The subsequent bioinformatics analysis is divided into 3 steps. In Step 1, 21 genes were revealed by intersecting the enriched genes from the KEGG, WIKI, and Reactome databases. In Step 2, WGCNA was applied and intersected with the DEGs. Further intersection based on the enrichments with the GO biological processes, GO cellular components, and GO molecular functions databases screened out 12 candidate genes. In Step 3, 13 genes were found to be simultaneously up-regulated in the DEGs and a GEO dataset involving human diabetic retinal tissues. VEGFA and ERN1 were the 2 starred genes finally screened out by overlapping the 3 Steps. CONCLUSION In this study, multiple genes were identified as crucial in the pathological process of RPE under protracted HG, providing potential candidates for future researches on DR. The current study highlights the importance of RPE in DR pathogenesis.
Collapse
Affiliation(s)
- Hao Huang
- Department of Ophthalmology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, 116 South Changjiang Road, Zhuzhou, 412000, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Jingshu Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Xielan Kuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, 510000, China
- Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 Xianlie Road, Guangzhou, 510000, China
| | - Fan He
- Amass Ophthalmology, Guangzhou, 510000, China
| | - Jianjun Yan
- Department of Ophthalmology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, 116 South Changjiang Road, Zhuzhou, 412000, China
| | - Bowen Li
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Wei Liu
- Department of Ophthalmology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, 116 South Changjiang Road, Zhuzhou, 412000, China.
| | - Huangxuan Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, 510000, China.
- Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 Xianlie Road, Guangzhou, 510000, China.
| |
Collapse
|
102
|
Park SJ, Kim YY, Han JY, Kim SW, Kim H, Ku SY. Advancements in Human Embryonic Stem Cell Research: Clinical Applications and Ethical Issues. Tissue Eng Regen Med 2024; 21:379-394. [PMID: 38502279 PMCID: PMC10987435 DOI: 10.1007/s13770-024-00627-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND The development and use of human embryonic stem cells (hESCs) in regenerative medicine have been revolutionary, offering significant advancements in treating various diseases. These pluripotent cells, derived from early human embryos, are central to modern biomedical research. However, their application is mired in ethical and regulatory complexities related to the use of human embryos. METHOD This review utilized key databases such as ClinicalTrials.gov, EU Clinical Trials Register, PubMed, and Google Scholar to gather recent clinical trials and studies involving hESCs. The focus was on their clinical application in regenerative medicine, emphasizing clinical trials and research directly involving hESCs. RESULTS Preclinical studies and clinical trials in various areas like ophthalmology, neurology, endocrinology, and reproductive medicine have demonstrated the versatility of hESCs in regenerative medicine. These studies underscore the potential of hESCs in treating a wide array of conditions. However, the field faces ethical and regulatory challenges, with significant variations in policies and perspectives across different countries. CONCLUSION The potential of hESCs in regenerative medicine is immense, offering new avenues for treating previously incurable diseases. However, navigating the ethical, legal, and regulatory landscapes is crucial for the continued advancement and responsible application of hESC research in the medical field. Considering both scientific potential and ethical implications, a balanced approach is essential for successfully integrating hESCs into clinical practice.
Collapse
Affiliation(s)
- Soo Jin Park
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Ji Yeon Han
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 101 Daehak-Ro Jongno-Gu, Seoul, 03080, Republic of Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 101 Daehak-Ro Jongno-Gu, Seoul, 03080, Republic of Korea.
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
103
|
Ashimori A, Higashijima F, Ogata T, Sakuma A, Hamada W, Sunada J, Aoki R, Mikuni M, Hayashi K, Wakuta M, Yoshimoto T, Minamoto A, Ko JA, Kimura K. HIF-1α-dependent upregulation of angiogenic factors by mechanical stimulation in retinal pigment epithelial cells. Dis Model Mech 2024; 17:dmm050640. [PMID: 38691000 PMCID: PMC11095633 DOI: 10.1242/dmm.050640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/22/2024] [Indexed: 05/03/2024] Open
Abstract
Mechanical stimulation as a mimic of drusen formation in the eye increases the expression of angiogenic factors in retinal pigment epithelial (RPE) cells, but the underlying molecular mechanisms remain unclear. We investigated and characterized the effects of mechanical stimulation on the expression of angiogenic factors in RPE cells both in vitro and in a mouse model. Mechanical stimulation increased the expression of vascular endothelial growth factor (VEGF, encoded by VEGFA) and other angiogenesis-related genes in cultured RPE1 cells. The presence of hypoxia-inducible factor 1α (HIF-1α, encoded by HIF1A) was also increased, and both knockdown of HIF-1α and treatment with the HIF-1α inhibitor CAY10585 attenuated the effect of mechanical stimulation on angiogenesis factor gene expression. Signaling by the tyrosine kinase SRC and p38 mitogen-activated protein kinase was involved in HIF-1α activation and consequent angiogenesis-related gene expression induced by mechanical stimulation. Our results suggest that SRC-p38 and HIF-1α signaling are involved in the upregulation of angiogenic factors in RPE cells by mechanical stimulation. Such in vivo suppression of upregulated expression of angiogenesis-related genes by pharmacological inhibitors of HIF-1α suggests a new potential approach to the treatment of age-related macular degeneration.
Collapse
Affiliation(s)
- Atsushige Ashimori
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Fumiaki Higashijima
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Tadahiko Ogata
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Ayano Sakuma
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Waka Hamada
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Junki Sunada
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Ren Aoki
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Masanori Mikuni
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Ken'ichiro Hayashi
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Makiko Wakuta
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Takuya Yoshimoto
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| | - Akira Minamoto
- Department of Ophthalmology and Visual Science, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Ji-Ae Ko
- Department of Ophthalmology and Visual Science, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube City, Yamaguchi 755-8505, Japan
| |
Collapse
|
104
|
Baqué-Vidal L, Main H, Petrus-Reurer S, Lederer AR, Beri NE, Bär F, Metzger H, Zhao C, Efstathopoulos P, Saietz S, Wrona A, Jaberi E, Willenbrock H, Reilly H, Hedenskog M, Moussaud-Lamodière E, Kvanta A, Villaescusa JC, La Manno G, Lanner F. Clinically compliant cryopreservation of differentiated retinal pigment epithelial cells. Cytotherapy 2024; 26:340-350. [PMID: 38349309 DOI: 10.1016/j.jcyt.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND AIMS Age-related macular degeneration (AMD) is the most common cause of blindness in elderly patients within developed countries, affecting more than 190 million worldwide. In AMD, the retinal pigment epithelial (RPE) cell layer progressively degenerates, resulting in subsequent loss of photoreceptors and ultimately vision. There is currently no cure for AMD, but therapeutic strategies targeting the complement system are being developed to slow the progression of the disease. METHODS Replacement therapy with pluripotent stem cell-derived (hPSC) RPEs is an alternative treatment strategy. A cell therapy product must be produced in accordance with Good Manufacturing Practices at a sufficient scale to facilitate extensive pre-clinical and clinical testing. Cryopreservation of the final cell product is therefore highly beneficial, as the manufacturing, pre-clinical and clinical testing can be separated in time and location. RESULTS We found that mature hPSC-RPE cells do not survive conventional cryopreservation techniques. However, replating the cells 2-5 days before cryopreservation facilitates freezing. The replated and cryopreserved hPSC-RPE cells maintained their identity, purity and functionality as characteristic RPEs, shown by cobblestone morphology, pigmentation, transcriptional profile, RPE markers, transepithelial resistance and pigment epithelium-derived factor secretion. Finally, we showed that the optimal replating time window can be tracked noninvasively by following the change in cobblestone morphology. CONCLUSIONS The possibility of cryopreserving the hPSC-RPE product has been instrumental in our efforts in manufacturing and performing pre-clinical testing with the aim for clinical translation.
Collapse
Affiliation(s)
- Laura Baqué-Vidal
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Heather Main
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Sandra Petrus-Reurer
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden; Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden; Department of Surgery, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Alex R Lederer
- Laboratory of Neurodevelopmental Systems Biology, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nefeli-Eirini Beri
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Frederik Bär
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Hugo Metzger
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Cheng Zhao
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | | | - Sarah Saietz
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | | | - Elham Jaberi
- Cell Therapy R&D, Novo Nordisk A/S, Måløv, Denmark
| | | | - Hazel Reilly
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Mona Hedenskog
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Elisabeth Moussaud-Lamodière
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Anders Kvanta
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Gioele La Manno
- Laboratory of Neurodevelopmental Systems Biology, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Fredrik Lanner
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden; Ming Wai Lau Center for Reparative Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
105
|
Bovi Dos Santos G, de Lima-Vasconcellos TH, Móvio MI, Birbrair A, Del Debbio CB, Kihara AH. New Perspectives in Stem Cell Transplantation and Associated Therapies to Treat Retinal Diseases: From Gene Editing to 3D Bioprinting. Stem Cell Rev Rep 2024; 20:722-737. [PMID: 38319527 DOI: 10.1007/s12015-024-10689-4] [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] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Inherited and non-inherited retinopathies can affect distinct cell types, leading to progressive cell death and visual loss. In the last years, new approaches have indicated exciting opportunities to treat retinopathies. Cell therapy in retinitis pigmentosa, age-related macular disease, and glaucoma have yielded encouraging results in rodents and humans. The first two diseases mainly impact the photoreceptors and the retinal pigmented epithelium, while glaucoma primarily affects the ganglion cell layer. Induced pluripotent stem cells and multipotent stem cells can be differentiated in vitro to obtain specific cell types for use in transplant as well as to assess the impact of candidate molecules aimed at treating retinal degeneration. Moreover, stem cell therapy is presented in combination with newly developed methods, such as gene editing, Müller cells dedifferentiation, sheet & drug delivery, virus-like particles, optogenetics, and 3D bioprinting. This review describes the recent advances in this field, by presenting an updated panel based on cell transplants and related therapies to treat retinopathies.
Collapse
Affiliation(s)
- Gabrieli Bovi Dos Santos
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, Santo André, SP, Brazil
| | | | - Marília Inês Móvio
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, Santo André, SP, Brazil
| | - Alexander Birbrair
- Department of Dermatology, Medical Sciences Center, University of Wisconsin-Madison, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA
| | - Carolina Beltrame Del Debbio
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo USP, São Paulo, SP, Brazil
| | - Alexandre Hiroaki Kihara
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, Santo André, SP, Brazil.
| |
Collapse
|
106
|
Vera E, Cornejo I, Henao JC, Tribiños F, Burgos J, Sepúlveda FV, Cid LP. Normal vision and development in mice with low functional expression of Kir7.1 in heterozygosis for a blindness-producing mutation inactivating the channel. Am J Physiol Cell Physiol 2024; 326:C1178-C1192. [PMID: 38406825 DOI: 10.1152/ajpcell.00597.2023] [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: 11/07/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
K+ channel Kir7.1 expressed at the apical membrane of the retinal pigment epithelium (RPE) plays an essential role in retinal function. An isoleucine-to-threonine mutation at position 120 of the protein is responsible for blindness-causing vitreo-retinal dystrophy. We have studied the molecular mechanism of action of Kir7.1-I120T in vitro by heterologous expression and in vivo in CRISPR-generated knockin mice. Full-size Kir7.1-I120T reaches the plasma membrane but lacks any activity. Analysis of Kir7.1 and the I120T mutant in mixed transfection experiments, and that of tandem tetrameric constructs made by combining wild type (WT) and mutant protomers, leads us to conclude that they do not form heterotetramers in vitro. Homozygous I120T/I120T mice show cleft palate and tracheomalacia and do not survive beyond P0, whereas heterozygous WT/I120T develop normally. Membrane conductance of RPE cells isolated from WT/WT and heterozygous WT/I120T mice is dominated by Kir7.1 current. Using Rb+ as a charge carrier, we demonstrate that the Kir7.1 current of WT/I120T RPE cells corresponds to approximately 50% of that in cells from WT/WT animals, in direct proportion to WT gene dosage. This suggests a lack of compensatory effects or interference from the mutated allele product, an interpretation consistent with results obtained using WT/- hemizygous mouse. Electroretinography and behavioral tests also show normal vision in WT/I120T animals. The hypomorphic ion channel phenotype of heterozygous Kir7.1-I120T mutants is therefore compatible with normal development and retinal function. The lack of detrimental effect of this degree of functional deficit might explain the recessive nature of Kir7.1 mutations causing human eye disease.NEW & NOTEWORTHY Human retinal pigment epithelium K+ channel Kir7.1 is affected by generally recessive mutations leading to blindness. We investigate one such mutation, isoleucine-to-threonine at position 120, both in vitro and in vivo in knockin mice. The mutated channel is inactive and in heterozygosis gives a hypomorphic phenotype with normal retinal function. Mutant channels do not interfere with wild-type Kir7.1 channels which are expressed concomitantly without hindrance, providing an explanation for the recessive nature of the disease.
Collapse
Affiliation(s)
- Erwin Vera
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | - Isabel Cornejo
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Valdivia, Chile
| | - Juan Carlos Henao
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | | | | | - Francisco V Sepúlveda
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - L Pablo Cid
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| |
Collapse
|
107
|
Liu D, Du J, Xie H, Tian H, Lu L, Zhang C, Xu GT, Zhang J. Wnt5a/β-catenin-mediated epithelial-mesenchymal transition: a key driver of subretinal fibrosis in neovascular age-related macular degeneration. J Neuroinflammation 2024; 21:75. [PMID: 38532410 DOI: 10.1186/s12974-024-03068-w] [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: 12/01/2023] [Accepted: 03/18/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Neovascular age-related macular degeneration (nAMD), accounts for up to 90% of AMD-associated vision loss, ultimately resulting in the formation of fibrotic scar in the macular region. The pathogenesis of subretinal fibrosis in nAMD involves the process of epithelial-mesenchymal transition (EMT) occurring in retinal pigment epithelium (RPE). Here, we aim to investigate the underlying mechanisms involved in the Wnt signaling during the EMT of RPE cells and in the pathological process of subretinal fibrosis secondary to nAMD. METHODS In vivo, the induction of subretinal fibrosis was performed in male C57BL/6J mice through laser photocoagulation. Either FH535 (a β-catenin inhibitor) or Box5 (a Wnt5a inhibitor) was intravitreally administered on the same day or 14 days following laser induction. The RPE-Bruch's membrane-choriocapillaris complex (RBCC) tissues were collected and subjected to Western blot analysis and immunofluorescence to examine fibrovascular and Wnt-related markers. In vitro, transforming growth factor beta 1 (TGFβ1)-treated ARPE-19 cells were co-incubated with or without FH535, Foxy-5 (a Wnt5a-mimicking peptide), Box5, or Wnt5a shRNA, respectively. The changes in EMT- and Wnt-related signaling molecules, as well as cell functions were assessed using qRT-PCR, nuclear-cytoplasmic fractionation assay, Western blot, immunofluorescence, scratch assay or transwell migration assay. The cell viability of ARPE-19 cells was determined using Cell Counting Kit (CCK)-8. RESULTS The in vivo analysis demonstrated Wnt5a/ROR1, but not Wnt3a, was upregulated in the RBCCs of the laser-induced CNV mice compared to the normal control group. Intravitreal injection of FH535 effectively reduced Wnt5a protein expression. Both FH535 and Box5 effectively attenuated subretinal fibrosis and EMT, as well as the activation of β-catenin in laser-induced CNV mice, as evidenced by the significant reduction in areas positive for fibronectin, alpha-smooth muscle actin (α-SMA), collagen I, and active β-catenin labeling. In vitro, Wnt5a/ROR1, active β-catenin, and some other Wnt signaling molecules were upregulated in the TGFβ1-induced EMT cell model using ARPE-19 cells. Co-treatment with FH535, Box5, or Wnt5a shRNA markedly suppressed the activation of Wnt5a, nuclear translocation of active β-catenin, as well as the EMT in TGFβ1-treated ARPE-19 cells. Conversely, treatment with Foxy-5 independently resulted in the activation of abovementioned molecules and subsequent induction of EMT in ARPE-19 cells. CONCLUSIONS Our study reveals a reciprocal activation between Wnt5a and β-catenin to mediate EMT as a pivotal driver of subretinal fibrosis in nAMD. This positive feedback loop provides valuable insights into potential therapeutic strategies to treat subretinal fibrosis in nAMD patients.
Collapse
Affiliation(s)
- Dandan Liu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Jingxiao Du
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, National Clinical Research Center for Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China
| | - Hai Xie
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, National Clinical Research Center for Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China
| | - Haibin Tian
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Lixia Lu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, National Clinical Research Center for Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Guo-Tong Xu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, National Clinical Research Center for Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| |
Collapse
|
108
|
Shastak Y, Pelletier W. Pet Wellness and Vitamin A: A Narrative Overview. Animals (Basel) 2024; 14:1000. [PMID: 38612239 PMCID: PMC11010875 DOI: 10.3390/ani14071000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
The health of companion animals, particularly dogs and cats, is significantly influenced by nutrition, with vitamins playing a crucial role. Vitamin A, in particular, is indispensable, with diverse roles ranging from vision to immune modulation and reproduction. Despite its importance, the metabolism and dietary requirements of vitamin A in companion animals remain complex and not fully understood. This review provides a comprehensive overview of the historical perspective, the digestion, the metabolism, the physiological roles, the deficiency, the excess, and the interactions with other micronutrients of vitamin A in companion animals. Additionally, it highlights future research directions and gaps in our understanding. Insights into the metabolism of vitamin A in companion animals, personalized nutrition strategies based on genetic variability, longitudinal studies tracking the status of vitamin A, and investigations into its immunomodulatory effects are crucial for optimizing pet health and wellness. Furthermore, understanding the stability and bioavailability of vitamin A in pet food formulations is essential for ensuring the provision of adequate micronutrients. Overall, this review underscores the importance of vitamin A in companion animal nutrition and the need for further research to enhance our understanding and to optimize dietary recommendations for pet health and well-being.
Collapse
Affiliation(s)
- Yauheni Shastak
- Nutrition & Health Division, BASF SE, 67063 Ludwigshafen am Rhein, Germany
| | | |
Collapse
|
109
|
Dontsov A, Ostrovsky M. Retinal Pigment Epithelium Pigment Granules: Norms, Age Relations and Pathology. Int J Mol Sci 2024; 25:3609. [PMID: 38612421 PMCID: PMC11011557 DOI: 10.3390/ijms25073609] [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: 02/29/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
The retinal pigment epithelium (RPE), which ensures the normal functioning of the neural retina, is a pigmented single-cell layer that separates the retina from the Bruch's membrane and the choroid. There are three main types of pigment granules in the RPE cells of the human eye: lipofuscin granules (LG) containing the fluorescent "age pigment" lipofuscin, melanoprotein granules (melanosomes, melanolysosomes) containing the screening pigment melanin and complex melanolipofuscin granules (MLG) containing both types of pigments simultaneously-melanin and lipofuscin. This review examines the functional role of pigment granules in the aging process and in the development of oxidative stress and associated pathologies in RPE cells. The focus is on the process of light-induced oxidative degradation of pigment granules caused by reactive oxygen species. The reasons leading to increased oxidative stress in RPE cells as a result of the oxidative degradation of pigment granules are considered. A mechanism is proposed to explain the phenomenon of age-related decline in melanin content in RPE cells. The essence of the mechanism is that when the lipofuscin part of the melanolipofuscin granule is exposed to light, reactive oxygen species are formed, which destroy the melanin part. As more melanolipofuscin granules are formed with age and the development of degenerative diseases, the melanin in pigmented epithelial cells ultimately disappears.
Collapse
Affiliation(s)
| | - Mikhail Ostrovsky
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow 119334, Russia;
| |
Collapse
|
110
|
Tan J, Xiao A, Yang L, Tao YL, Shao Y, Zhou Q. Diabetes and high-glucose could upregulate the expression of receptor for activated C kinase 1 in retina. World J Diabetes 2024; 15:519-529. [PMID: 38591093 PMCID: PMC10999037 DOI: 10.4239/wjd.v15.i3.519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/08/2023] [Accepted: 01/18/2024] [Indexed: 03/15/2024] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is a major ocular complication of diabetes mellitus, leading to visual impairment. Retinal pigment epithelium (RPE) injury is a key component of the outer blood retinal barrier, and its damage is an important indicator of DR. Receptor for activated C kinase 1 (RACK1) activates protein kinase C-ε (PKC-ε) to promote the generation of reactive oxygen species (ROS) in RPE cells, leading to apoptosis. Therefore, we hypothesize that the activation of RACK1 under hypoxic/high-glucose conditions may promote RPE cell apoptosis by modulating PKC-ε/ROS, thereby disrupting the barrier effect of the outer blood retinal barrier and contributing to the progression of DR. AIM To investigate the role and associated underlying mechanisms of RACK1 in the development of early DR. METHODS In this study, Sprague-Dawley rats and adult RPE cell line-19 (ARPE-19) cells were used as in vivo and in vitro models, respectively, to explore the role of RACK1 in mediating PKC-ε in early DR. Furthermore, the impact of RACK1 on apoptosis and barrier function of RPE cells was also investigated in the former model. RESULTS Streptozotocin-induced diabetic rats showed increased apoptosis and up-regulated expression of RACK1 and PKC-ε proteins in RPE cells following a prolonged modeling. Similarly, ARPE-19 cells exposed to high glucose and hypoxia displayed elevated mRNA and protein levels of RACK1 and PKC-ε, accompanied by an increases in ROS production, apoptosis rate, and monolayer permeability. However, silencing RACK1 significantly downregulated the expression of PKC-ε and ROS, reduced cell apoptosis and permeability, and protected barrier function. CONCLUSION RACK1 plays a significant role in the development of early DR and might serve as a potential therapeutic target for DR by regulating RPE apoptosis and barrier function.
Collapse
Affiliation(s)
- Jian Tan
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Ang Xiao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Lin Yang
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yu-Lin Tao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yi Shao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Qiong Zhou
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
111
|
Du X, Butler AG, Chen HY. Cell-cell interaction in the pathogenesis of inherited retinal diseases. Front Cell Dev Biol 2024; 12:1332944. [PMID: 38500685 PMCID: PMC10944940 DOI: 10.3389/fcell.2024.1332944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/06/2024] [Indexed: 03/20/2024] Open
Abstract
The retina is part of the central nervous system specialized for vision. Inherited retinal diseases (IRD) are a group of clinically and genetically heterogenous disorders that lead to progressive vision impairment or blindness. Although each disorder is rare, IRD accumulatively cause blindness in up to 5.5 million individuals worldwide. Currently, the pathophysiological mechanisms of IRD are not fully understood and there are limited treatment options available. Most IRD are caused by degeneration of light-sensitive photoreceptors. Genetic mutations that abrogate the structure and/or function of photoreceptors lead to visual impairment followed by blindness caused by loss of photoreceptors. In healthy retina, photoreceptors structurally and functionally interact with retinal pigment epithelium (RPE) and Müller glia (MG) to maintain retinal homeostasis. Multiple IRD with photoreceptor degeneration as a major phenotype are caused by mutations of RPE- and/or MG-associated genes. Recent studies also reveal compromised MG and RPE caused by mutations in ubiquitously expressed ciliary genes. Therefore, photoreceptor degeneration could be a direct consequence of gene mutations and/or could be secondary to the dysfunction of their interaction partners in the retina. This review summarizes the mechanisms of photoreceptor-RPE/MG interaction in supporting retinal functions and discusses how the disruption of these processes could lead to photoreceptor degeneration, with an aim to provide a unique perspective of IRD pathogenesis and treatment paradigm. We will first describe the biology of retina and IRD and then discuss the interaction between photoreceptors and MG/RPE as well as their implications in disease pathogenesis. Finally, we will summarize the recent advances in IRD therapeutics targeting MG and/or RPE.
Collapse
Affiliation(s)
| | | | - Holly Y. Chen
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
112
|
Bose D, Ortolan D, Farnoodian M, Sharma R, Bharti K. Considerations for Developing an Autologous Induced Pluripotent Stem Cell (iPSC)-Derived Retinal Pigment Epithelium (RPE) Replacement Therapy. Cold Spring Harb Perspect Med 2024; 14:a041295. [PMID: 37487631 PMCID: PMC10910357 DOI: 10.1101/cshperspect.a041295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Cell-replacement therapies are a new class of treatments, which include induced pluripotent stem cell (iPSC)-derived tissues that aim to replace degenerated cells. iPSCs can potentially be used to generate any cell type of the body, making them a powerful tool for treating degenerative diseases. Cell replacement for retinal degenerative diseases is at the forefront of cell therapies, given the accessibility of the eye for surgical procedures and a huge unmet medical need for retinal degenerative diseases with no current treatment options. Clinical trials are ongoing in different parts of the world using stem cell-derived retinal pigment epithelium (RPE). This review focuses on scientific and regulatory considerations when developing an iPSC-derived RPE cell therapy from the development of a robust and efficient differentiation protocol to critical quality control assays for cell validation, the choice of an appropriate animal model for preclinical testing, and the regulatory aspects that dictate the final approval for proceeding to a first-in-human clinical trial.
Collapse
Affiliation(s)
- Devika Bose
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Davide Ortolan
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Mitra Farnoodian
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
113
|
Spirig SE, Renner M. Toward Retinal Organoids in High-Throughput. Cold Spring Harb Perspect Med 2024; 14:a041275. [PMID: 37217280 PMCID: PMC10910359 DOI: 10.1101/cshperspect.a041275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Human retinal organoids recapitulate the cellular diversity, arrangement, gene expression, and functional aspects of the human retina. Protocols to generate human retinal organoids from pluripotent stem cells are typically labor intensive, include many manual handling steps, and the organoids need to be maintained for several months until they mature. To generate large numbers of human retinal organoids for therapy development and screening purposes, scaling up retinal organoid production, maintenance, and analysis is of utmost importance. In this review, we discuss strategies to increase the number of high-quality retinal organoids while reducing manual handling steps. We further review different approaches to analyze thousands of retinal organoids with currently available technologies and point to challenges that still await to be overcome both in culture and analysis of retinal organoids.
Collapse
Affiliation(s)
- Stefan Erich Spirig
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Magdalena Renner
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| |
Collapse
|
114
|
Nishikiori N, Watanabe M, Sato T, Umetsu A, Higashide M, Furuhashi M, Ohguro H. Intraocular fatty acids induce reinforcement of barrier functions on the outer blood-retinal barrier. Prostaglandins Leukot Essent Fatty Acids 2024; 202:102637. [PMID: 39128394 DOI: 10.1016/j.plefa.2024.102637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/17/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
The aim of the present study was to elucidate unknown effects of intraocular fatty acids (ioFAs) including palmitic acid (C16:0), stearic acid (C18:0), oleic acid (C18:1), linoleic acid (C18:2), arachidonic acid (C20:4), eicosapentaenoic acid (EPA, C20:5) and docosahexaenoic acid (DHA, C22:6) on the outer blood-retinal barrier (oBRB). For this purpose, human retinal pigment epithelium cell line ARPE19 was subjected to analyses for evaluating the following biological phenotypes: (1) cell viability, (2) cellular metabolic functions, (3) barrier functions by trans-epithelial electrical resistance (TEER), and (4) expression of tight junction (TJ) molecules. In the presence of 100 nM ioFAs, no significant effects on cell viability of ARPE19 cells was observed. While treatment with EPA or DHA tended to reduce non-mitochondrial oxygen consumption, most indices in mitochondrial functions were not markedly affected by treatment with ioFAs in ARPE19 cells. On the other hand, ioFAs except for palmitic acid and stearic acid significantly increased basal extracellular acidification rates, suggesting activated glycolysis or increased lactate production. Interestingly, TEER values of planar ARPE19 monolayer were significantly increased by treatment any ioFAs. Consistently, gene expression levels of TJ proteins were increased by treatment with ioFAs. Collectively, the findings presented herein suggest that ioFAs may contribute to reinforcement of barrier functions of the oBRB albeit there are some differences in biological effects depending on the type of ioFAs.
Collapse
Affiliation(s)
- Nami Nishikiori
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Megumi Watanabe
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Tatsuya Sato
- Department ofCardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Japan; Department ofCellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Japan
| | - Araya Umetsu
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Megumi Higashide
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Masato Furuhashi
- Department ofCardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Japan
| | - Hiroshi Ohguro
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan.
| |
Collapse
|
115
|
Liu B, Yang H, Song YS, Sorenson CM, Sheibani N. Thrombospondin-1 in vascular development, vascular function, and vascular disease. Semin Cell Dev Biol 2024; 155:32-44. [PMID: 37507331 PMCID: PMC10811293 DOI: 10.1016/j.semcdb.2023.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023]
Abstract
Angiogenesis is vital to developmental, regenerative and repair processes. It is normally regulated by a balanced production of pro- and anti-angiogenic factors. Alterations in this balance under pathological conditions are generally mediated through up-regulation of pro-angiogenic and/or downregulation of anti-angiogenic factors, leading to growth of new and abnormal blood vessels. The pathological manifestation of many diseases including cancer, ocular and vascular diseases are dependent on the growth of these new and abnormal blood vessels. Thrompospondin-1 (TSP1) was the first endogenous angiogenesis inhibitor identified and its anti-angiogenic and anti-inflammatory activities have been the subject of many studies. Studies examining the role TSP1 plays in pathogenesis of various ocular diseases and vascular dysfunctions are limited. Here we will discuss the recent studies focused on delineating the role TSP1 plays in ocular vascular development and homeostasis, and pathophysiology of various ocular and vascular diseases with a significant clinical relevance to human health.
Collapse
Affiliation(s)
- Bo Liu
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA.
| | - Huan Yang
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Yong-Seok Song
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Christine M Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Nader Sheibani
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
116
|
Gurubaran IS. Mitochondrial damage and clearance in retinal pigment epithelial cells. Acta Ophthalmol 2024; 102 Suppl 282:3-53. [PMID: 38467968 DOI: 10.1111/aos.16661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 03/13/2024]
Abstract
Age-related macular degeneration (AMD) is a devastating eye disease that causes permanent vision loss in the central part of the retina, known as the macula. Patients with such severe visual loss face a reduced quality of life and are at a 1.5 times greater risk of death compared to the general population. Currently, there is no cure for or effective treatment for dry AMD. There are several mechanisms thought to underlie the disease, for example, ageing-associated chronic oxidative stress, mitochondrial damage, harmful protein aggregation and inflammation. As a way of gaining a better understanding of the molecular mechanisms behind AMD and thus developing new therapies, we have created a peroxisome proliferator-activated receptor gamma coactivator 1-alpha and nuclear factor erythroid 2-related factor 2 (PGC1α/NFE2L2) double-knockout (dKO) mouse model that mimics many of the clinical features of dry AMD, including elevated levels of oxidative stress markers, damaged mitochondria, accumulating lysosomal lipofuscin and extracellular drusen-like structures in retinal pigment epithelial cells (RPE). In addition, a human RPE cell-based model was established to examine the impact of non-functional intracellular clearance systems on inflammasome activation. In this study, we found that there was a disturbance in the autolysosomal machinery responsible for clearing mitochondria in the RPE cells of one-year-old PGC1α/NFE2L2-deficient mice. The confocal immunohistochemical analysis revealed an increase in autophagosome marker microtubule-associated proteins 1A/1B light chain 3B (LC3B) as well as multiple mitophagy markers such as PTE-induced putative kinase 1 (PINK1) and E3 ubiquitin ligase (PARKIN), along with signs of damaged mitochondria. However, no increase in autolysosome formation was detected, nor was there a colocalization of the lysosomal marker LAMP2 or the mitochondrial marker, ATP synthase β. There was an upregulation of late autolysosomal fusion Ras-related protein (Rab7) in the perinuclear space of RPE cells, together with autofluorescent aggregates. Additionally, we observed an increase in the numbers of Toll-like receptors 3 and 9, while those of NOD-like receptor 3 were decreased in PGC1α/NFE2L2 dKO retinal specimens compared to wild-type animals. There was a trend towards increased complement component C5a and increased involvement of the serine protease enzyme, thrombin, in enhancing the terminal pathway producing C5a, independent of C3. The levels of primary acute phase C-reactive protein and receptor for advanced glycation end products were also increased in the PGC1α/NFE2L2 dKO retina. Furthermore, selective proteasome inhibition with epoxomicin promoted both nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondrial-mediated oxidative stress, leading to the release of mitochondrial DNA to the cytosol, resulting in potassium efflux-dependent activation of the absent in melanoma 2 (AIM2) inflammasome and the subsequent secretion of interleukin-1β in ARPE-19 cells. In conclusion, the data suggest that there is at least a relative decrease in mitophagy, increases in the amounts of C5 and thrombin and decreased C3 levels in this dry AMD-like model. Moreover, selective proteasome inhibition evoked mitochondrial damage and AIM2 inflammasome activation in ARPE-19 cells.
Collapse
Affiliation(s)
- Iswariyaraja Sridevi Gurubaran
- Department of Medicine, Clinical Medicine Unit, University of Eastern Finland Institute of Clinical Medicine, Kuopio, Northern Savonia, Finland
| |
Collapse
|
117
|
Kitahata S, Mandai M, Ichikawa H, Tanaka Y, Senba T, Kajita K, Sugita S, Kadonosono K, Takahashi M. Investigation of the effectiveness of gelatin hydrolysate in human iPS-RPE cell suspension transplantation. Regen Ther 2024; 25:238-249. [PMID: 38293586 PMCID: PMC10825282 DOI: 10.1016/j.reth.2023.12.016] [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: 10/13/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction The retinal pigment epithelium (RPE) plays essential roles in maintaining retinal functions as well as choroidal capillaries and can lead to visual disorders if dysfunctional. Transplantation of human-induced pluripotent stem cell-derived RPE (hiPSC-RPE) is a promising therapy for such RPE impaired conditions including age-related macular degeneration. The challenge with cell suspension transplantation is targeted delivery of graft cells and undesired cell reflux. Gelatin hydrolysate, a soluble variant with specific molecular weight distribution, is examined in this study for its potential use in hiPSC-RPE suspension transplantation, particularly in reducing cell reflux and enhancing RPE engraftment. Methods A retinal bleb model was created using polydimethylsiloxane (PDMS) soft lithography to quantify cellular reflux. We examined the effects of gelatin hydrolysate on the hiPSC-RPE of various aspects of cell behavior and performance such as cell viability, hypoxia reaction, morphology, induction of inflammation and immune responses. Results Gelatin hydrolysate at 5 % concentration effectively mitigated cell reflux in vitro mimic, improved cell viability, reduced cell aggregation, and had an inhibitory effect on hypoxic reactions due to cell deposition with hiPSC-RPE. Additionally, gelatin hydrolysate did not affect cell adhesion and morphology, and decreased the expression of major histocompatibility complex class II molecules, which suggests reduced immunogenicity of hiPSC-RPE. Conclusion Gelatin hydrolysate is considered a valuable and useful candidate for future regenerative therapies in hiPSC-RPE suspension transplantation.
Collapse
Affiliation(s)
- Shohei Kitahata
- Department of Ophthalmology and Micro-technology, Yokohama City University, 4-57 Urafunecho, Minami-ku, Yokohama, Japan
- Kobe City Eye Hospital, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
| | - Michiko Mandai
- Kobe City Eye Hospital, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
| | - Hinako Ichikawa
- Department of Ophthalmology and Micro-technology, Yokohama City University, 4-57 Urafunecho, Minami-ku, Yokohama, Japan
| | - Yuji Tanaka
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, Japan
| | - Toshika Senba
- Vision Care Inc., Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
| | - Keisuke Kajita
- Vision Care Inc., Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
- Department of Ophthalmology, Tokushima University, 2-24 Shinkura-cho, Tokushima, Japan
| | - Sunao Sugita
- Kobe City Eye Hospital, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
- Vision Care Inc., Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
| | - Kazuaki Kadonosono
- Department of Ophthalmology and Micro-technology, Yokohama City University, 4-57 Urafunecho, Minami-ku, Yokohama, Japan
| | - Masayo Takahashi
- Kobe City Eye Hospital, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
- Vision Care Inc., Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
| |
Collapse
|
118
|
Libreros-Jiménez HM, Manzo J, Rojas-Durán F, Aranda-Abreu GE, García-Hernández LI, Coria-Ávila GA, Herrera-Covarrubias D, Pérez-Estudillo CA, Toledo-Cárdenas MR, Hernández-Aguilar ME. On the Cranial Nerves. NEUROSCI 2024; 5:8-38. [PMID: 39483811 PMCID: PMC11523702 DOI: 10.3390/neurosci5010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 11/03/2024] Open
Abstract
The twelve cranial nerves play a crucial role in the nervous system, orchestrating a myriad of functions vital for our everyday life. These nerves are each specialized for particular tasks. Cranial nerve I, known as the olfactory nerve, is responsible for our sense of smell, allowing us to perceive and distinguish various scents. Cranial nerve II, or the optic nerve, is dedicated to vision, transmitting visual information from the eyes to the brain. Eye movements are governed by cranial nerves III, IV, and VI, ensuring our ability to track objects and focus. Cranial nerve V controls facial sensations and jaw movements, while cranial nerve VII, the facial nerve, facilitates facial expressions and taste perception. Cranial nerve VIII, or the vestibulocochlear nerve, plays a critical role in hearing and balance. Cranial nerve IX, the glossopharyngeal nerve, affects throat sensations and taste perception. Cranial nerve X, the vagus nerve, is a far-reaching nerve, influencing numerous internal organs, such as the heart, lungs, and digestive system. Cranial nerve XI, the accessory nerve, is responsible for neck muscle control, contributing to head movements. Finally, cranial nerve XII, the hypoglossal nerve, manages tongue movements, essential for speaking, swallowing, and breathing. Understanding these cranial nerves is fundamental in comprehending the intricate workings of our nervous system and the functions that sustain our daily lives.
Collapse
Affiliation(s)
| | - Jorge Manzo
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico; (J.M.); (F.R.-D.); (G.E.A.-A.); (L.I.G.-H.); (G.A.C.-Á.); (D.H.-C.); (C.A.P.-E.); (M.R.T.-C.)
| | - Fausto Rojas-Durán
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico; (J.M.); (F.R.-D.); (G.E.A.-A.); (L.I.G.-H.); (G.A.C.-Á.); (D.H.-C.); (C.A.P.-E.); (M.R.T.-C.)
| | - Gonzalo E Aranda-Abreu
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico; (J.M.); (F.R.-D.); (G.E.A.-A.); (L.I.G.-H.); (G.A.C.-Á.); (D.H.-C.); (C.A.P.-E.); (M.R.T.-C.)
| | - Luis I García-Hernández
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico; (J.M.); (F.R.-D.); (G.E.A.-A.); (L.I.G.-H.); (G.A.C.-Á.); (D.H.-C.); (C.A.P.-E.); (M.R.T.-C.)
| | - Genaro A Coria-Ávila
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico; (J.M.); (F.R.-D.); (G.E.A.-A.); (L.I.G.-H.); (G.A.C.-Á.); (D.H.-C.); (C.A.P.-E.); (M.R.T.-C.)
| | - Deissy Herrera-Covarrubias
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico; (J.M.); (F.R.-D.); (G.E.A.-A.); (L.I.G.-H.); (G.A.C.-Á.); (D.H.-C.); (C.A.P.-E.); (M.R.T.-C.)
| | - César A Pérez-Estudillo
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico; (J.M.); (F.R.-D.); (G.E.A.-A.); (L.I.G.-H.); (G.A.C.-Á.); (D.H.-C.); (C.A.P.-E.); (M.R.T.-C.)
| | - María Rebeca Toledo-Cárdenas
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico; (J.M.); (F.R.-D.); (G.E.A.-A.); (L.I.G.-H.); (G.A.C.-Á.); (D.H.-C.); (C.A.P.-E.); (M.R.T.-C.)
| | - María Elena Hernández-Aguilar
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico; (J.M.); (F.R.-D.); (G.E.A.-A.); (L.I.G.-H.); (G.A.C.-Á.); (D.H.-C.); (C.A.P.-E.); (M.R.T.-C.)
| |
Collapse
|
119
|
Liang C, Li F, Gu C, Xie L, Yan W, Wang X, Shi R, Linghu S, Liu T. Metabolomic profiling of ocular tissues in rabbit myopia: Uncovering differential metabolites and pathways. Exp Eye Res 2024; 240:109796. [PMID: 38244883 DOI: 10.1016/j.exer.2024.109796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 01/22/2024]
Abstract
To investigate the metabolic difference among tissue layers of the rabbits' eye during the development of myopia using metabolomic techniques and explore any metabolic links or cascades within the ocular wall. Ultra Performance Liquid Chromatography - Mass Spectrometry (UPLC-MS) was utilized for untargeted metabolite screening (UMS) to identify the significant differential metabolites produced between myopia (MY) and control (CT) (horizontal). Subsequently, we compared those key metabolites among tissues (Sclera, Choroid, Retina) of MY for distribution and variation (longitudinal). A total of 6285 metabolites were detected in the three tissues. The differential metabolites were screened and the metabolic pathways of these metabolites in each myopic tissue were labeled, including tryptophan and its metabolites, pyruvate, taurine, caffeine metabolites, as well as neurotransmitters like glutamate and dopamine. Our study suggests that multiple metabolic pathways or different metabolites under the same pathway, might act on different parts of the eyeball and contribute to the occurrence and development of myopia by affecting the energy supply to the ocular tissues, preventing antioxidant stress, affecting scleral collagen synthesis, and regulating various neurotransmitters mutually.
Collapse
Affiliation(s)
- Chengpeng Liang
- Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou Province, China.
| | - Fayuan Li
- Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Chengqi Gu
- Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Ling Xie
- Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Wen Yan
- Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Xiaoye Wang
- Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Rong Shi
- Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Shaorong Linghu
- Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Taixiang Liu
- Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou Province, China; Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou Province, China.
| |
Collapse
|
120
|
Hui Q, Yang N, Xiong C, Zhou S, Zhou X, Jin Q, Xu X. Isorhamnetin suppresses the epithelial-mesenchymal transition of the retinal pigment epithelium both in vivo and in vitro through Nrf2-dependent AKT/GSK-3β pathway. Exp Eye Res 2024; 240:109823. [PMID: 38331017 DOI: 10.1016/j.exer.2024.109823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Age-related macular degeneration (AMD) is a major cause of blindness in the elderly worldwide. Multiple studies have shown that epithelial-mesenchymal transition (EMT) plays a pivotal role in the pathogenesis of AMD. Isorhamnetin (Isor) is a flavonoid compound that inhibits EMT in tumor cells. However, whether it can also attenuate EMT in the retinal pigment epithelium (RPE) is unknown. Therefore, our study was designed to probe the possible impact of Isor on EMT process in both mouse retina and ARPE-19 cells. C57BL/6 mice were utilized to establish a dry AMD model. Isor and LCZ (a mixture of luteine/β-carotene/zinc gluconate) were administered orally for 3 months. The effects of Isor on the retina were evaluated using fundus autofluorescence, optical coherence tomography, and transmission electron microscopy. Transwell and wound healing assay were employed to assess ARPE-19 cell migration. Western blotting and immunofluorescence were used to measure the protein expressions associated with EMT, Nrf2 and AKT/GSK-3β pathway. The findings indicated that Isor alleviated dry AMD-like pathological changes in vehicle mice retina, inhibited the migration of Ox-LDL-treated ARPE-19 cells, and repressed the EMT processes in vivo and in vitro. Furthermore, Isor activated Nrf2 pathway and deactivated AKT/GSK-3β pathway in both vehicle mice and ARPE-19 cells. Interestingly, when Nrf2 siRNA was transfected into ARPE-19 cells, the inhibitory effect of Isor on EMT and AKT/GSK-3β pathway was attenuated. These results suggested that Isor inhibited EMT processes via Nrf2-dependent AKT/GSK-3β pathway and is a promising candidate for dry AMD treatment.
Collapse
Affiliation(s)
- Qinyi Hui
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Ning Yang
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Caijian Xiong
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Siqi Zhou
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Xin Zhou
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Qingzi Jin
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Xinrong Xu
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China.
| |
Collapse
|
121
|
Liu L, Xu Q, Song X, Zhao Y, Zhang Y, Qu Y. Evaluation of changes in macular structures after subthreshold micropulse laser therapy on chronic central serous chorioretinopathy. Lasers Med Sci 2024; 39:83. [PMID: 38418639 DOI: 10.1007/s10103-024-04030-1] [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: 03/16/2023] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE To evaluate the changes in macular structures following subthreshold micropulse laser (SHML) treatment for chronic central serous chorioretinopathy (cCSC). METHODS Data of 33 eyes from 31 cCSC patients treated with SHML and followed up for at least 6 months has been included in this retrospective study. Main outcome measurements include resolution of subretinal fluid (SRF) and pigment epithelial detachment (PED), the recovery of ellipsoid zone (EZ) continuity, and the foveal outer nuclear layer (ONL) thickness along with its ratio. RESULTS Mean observation period is 7.355 months (ranging from 6 to 24 months) and mean number of treatments administered is 1.839 (ranging from 1 to 5). 6 months after SHML treatment, there is a significant decrease in the area of SRF and PED (P < 0.001, P = 0.010, respectively). Additionally, the frequency of continuous EZ and the foveal ONL thickness reveal a significant increase (P<0.001, P = 0.005, respectively). The ratio of foveal ONL thickness is significantly higher after laser treatment, particularly in patients with a disease duration of ≤12 months (p = 0.022, P=0.036, respectively). CONCLUSION SHML treatment proves to be effective in cCSC eyes, leading to satisfactory recovery of macular structures, especially the photoreceptor layer.
Collapse
Affiliation(s)
- Lijun Liu
- Department of Geriatrics, Qilu Hospital of Shandong University, Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Qian Xu
- Department of Geriatrics, Qilu Hospital of Shandong University, Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Xian Song
- Department of Geriatrics, Qilu Hospital of Shandong University, Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Yuqing Zhao
- Department of Geriatrics, Qilu Hospital of Shandong University, Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Yu Zhang
- Department of Geriatrics, Qilu Hospital of Shandong University, Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Yi Qu
- Department of Geriatrics, Qilu Hospital of Shandong University, Wenhuaxi Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
122
|
Anderson DM, Brager DC, Kearsley AJ. Spatially-dependent model for rods and cones in the retina. J Theor Biol 2024; 579:111687. [PMID: 38103677 DOI: 10.1016/j.jtbi.2023.111687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/17/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
We develop a mathematical model for photoreceptors in the retina. We focus on rod and cone outer segment dynamics and interactions with a nutrient source associated with the retinal pigment epithelium cells. Rod and cone densities (number per unit area of retinal surface) are known to have significant spatial dependence in the retina with cones located primarily near the fovea and the rods located primarily away from the fovea. Our model accounts for this spatial dependence of the rod and cone photoreceptor density as well as for the possibility of nutrient diffusion. We present equilibrium and dynamic solutions, discuss their relation to existing models, and estimate model parameters through comparisons with available experimental measurements of both spatial and temporal photoreceptor characteristics. Our model compares well with existing data on spatially-dependent regrowth of photoreceptor outer segments in the macular region of Rhesus Monkeys. Our predictions are also consistent with existing data on the spatial dependence of photoreceptor outer segment length near the fovea in healthy human subjects. We focus primarily on the healthy eye but our model could be the basis for future efforts designed to explore various retinal pathologies, eye-related injuries, and treatments of these conditions.
Collapse
Affiliation(s)
- Daniel M Anderson
- Applied & Computational Mathematics Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, 20899, MD, USA; Department of Mathematical Sciences, George Mason University, 4400 University Drive, Fairfax, 22030, VA, USA.
| | - Danielle C Brager
- Applied & Computational Mathematics Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, 20899, MD, USA.
| | - Anthony J Kearsley
- Applied & Computational Mathematics Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, 20899, MD, USA.
| |
Collapse
|
123
|
Zhang Y, Song X, Qi T, Zhou X. Review of lipocalin-2-mediated effects in diabetic retinopathy. Int Ophthalmol 2024; 44:78. [PMID: 38351392 DOI: 10.1007/s10792-024-03015-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/09/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Studies have uncovered LCN2 as a marker of inflammation strongly related to obesity, insulin resistance, and abnormal glucose metabolism in humans, and is involved in vascular diseases, inflammatory diseases, and neurological diseases. In recent years, studies have shown that elevated levels of LCN2 have a strong association with diabetic retinopathy (DR), but the pathogenesis is unknown. Here, we reviewed the relevant literature and compiled the pathogenesis associated with LCN2-induced DR. METHODS We searched PubMed and Web of Science electronic databases using "lipocalin-2, diabetic retinopathy, retinal degeneration, diabetic microangiopathies, diabetic neuropathy and inflammation" as subject terms. RESULTS In diabetic retinal neuropathy, LCN2 causes impaired retinal photoreceptor function and retinal neurons; in retinal microangiopathy, LCN2 induces apoptosis of retinal vascular endothelial cells and promotes angiogenesis; in retinal inflammation, increased secretion of LCN2 recruits inflammatory cells and induces pro-inflammatory cytokines. Moreover, LCN2 has the potential as a biomarker for DR. Recent studies have shown that retinal damage can be attenuated by silencing LCN2, which may be associated with the inhibition of caspase-1-mediated pyroptosis, and LCN2 may be a new target for the treatment of DR. CONCLUSIONS In conclusion, LCN2, involved in the development of diabetic retinopathy, is a key factor in diabetic retinal microangiopathy, neurodegeneration, and retinal inflammation. LCN2 is likely to be a novel molecular target leading to DR, and a more in-depth study of the pathogenesis of DR caused by LCN2 may provide considerable benefits for clinical research and potential drug development.
Collapse
Affiliation(s)
- Yajuan Zhang
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, 324 Jingwu Road, Jinan, 250021, Shandong, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China
| | - Xiaojun Song
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, 324 Jingwu Road, Jinan, 250021, Shandong, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China
| | - Tianying Qi
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, 324 Jingwu Road, Jinan, 250021, Shandong, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China
| | - Xinli Zhou
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, 324 Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China.
| |
Collapse
|
124
|
Rzhanova LA, Markitantova YV, Aleksandrova MA. Recent Achievements in the Heterogeneity of Mammalian and Human Retinal Pigment Epithelium: In Search of a Stem Cell. Cells 2024; 13:281. [PMID: 38334673 PMCID: PMC10854871 DOI: 10.3390/cells13030281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
Retinal pigment epithelium (RPE) cells are important fundamentally for the development and function of the retina. In this regard, the study of the morphological and molecular properties of RPE cells, as well as their regenerative capabilities, is of particular importance for biomedicine. However, these studies are complicated by the fact that, despite the external morphological similarity of RPE cells, the RPE is a population of heterogeneous cells, the molecular genetic properties of which have begun to be revealed by sequencing methods only in recent years. This review carries out an analysis of the data from morphological and molecular genetic studies of the heterogeneity of RPE cells in mammals and humans, which reveals the individual differences in the subpopulations of RPE cells and the possible specificity of their functions. Particular attention is paid to discussing the properties of "stemness," proliferation, and plasticity in the RPE, which may be useful for uncovering the mechanisms of retinal diseases associated with pathologies of the RPE and finding new ways of treating them.
Collapse
Affiliation(s)
| | - Yuliya V. Markitantova
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (L.A.R.); (M.A.A.)
| | | |
Collapse
|
125
|
Desmettre T, Baillif S, Mathis T, Gatinel D, Mainster M. [Blue light and intraocular lenses (IOLs): Beliefs and realities]. J Fr Ophtalmol 2024; 47:104043. [PMID: 38241770 DOI: 10.1016/j.jfo.2023.104043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 01/21/2024]
Abstract
The first intraocular lenses (IOLs) used for cataract surgery transmitted both ultraviolet (UV) radiation and visible light to the retina. Colorless UV-blocking IOLs were introduced and rapidly adopted in the 1980s. Yellow-tinted blue-blocking (also known as blue-filtering) IOLs were marketed in the early 1990s. Blue-blocking IOLs were intended to simulate age-related crystalline lens yellowing to reduce the cyanopsia that some patients experienced after cataract surgery. When blue-filtering IOLs were introduced in North America, however, blue-blocking chromophores were advocated as a way to protect patients from age-related macular degeneration (AMD) despite the lack of evidence that normal environmental light exposure causes AMD. The "blue light hazard" is a term that describes the experimental finding that acute, abnormally intense light exposures are potentially more phototoxic to the retina when short rather than long wavelengths are used. Thus, in brief exposures to intense light sources such as welding arcs, ultraviolet radiation is more hazardous than blue light, which is more hazardous than longer wavelength green or red light. International commissions have cautioned that the blue light hazard does not apply to normal indoor or outdoor light exposures. Nonetheless, the hazard is used for commercial purposes to suggest misleadingly that ambient environmental light can cause acute retinal phototoxicity and increase the risk of AMD. Very large epidemiological studies show that blue-blocking IOLs do not reduce the risk or progression of AMD. Additionally, blue-filtering IOLs or spectacles cannot decrease glare disability, because they decrease image and glare illuminance in the same proportion. Blue light is essential for older adults' scotopic photoreception needed to reduce the risk of nighttime falling and related injuries. It is also critical for circadian photoreception that is essential for good health, sleep and cognitive performance. Unfortunately, age-related pupillary miosis, retinal rod and ganglion cell photoreceptor degeneration and decreased outdoor activity all reduce the amount of healthful blue light available to older adults. Blue-restricting IOLs further reduce the available blue light at a time when older adults need it most. Patients and ophthalmologists are exposed to hypothesis-based advertisements for blue-filtering optical devices that suppress short wavelength light critical for vision in dim lighting and for good physical and mental health. Spectacle and intraocular lens selections should be based on scientific fact, not conjecture. Ideal IOLs should improve photoreception rather than limit it permanently. Practice efficiency, surgical convenience and physician-manufacturer relationships may eliminate a patient's opportunity to choose between colorless blue-transmitting IOLs and yellow-tinted, blue-restricting IOLs. Cataract surgeons ultimately determine whether their patients have the opportunity to make an informed choice about their future photoreception.
Collapse
Affiliation(s)
- T Desmettre
- Centre de rétine médicale, 187, rue de Menin, 59520 Marquette-Lez-Lille, France.
| | - S Baillif
- Département d'ophtalmologie, hôpital Pasteur, 30, voie Romaine, 06000 Nice cedex 1, France
| | - T Mathis
- Service d'ophtalmologie, hôpital de la Croix-Rousse, hospices civils de Lyon, 69004 Lyon, France
| | - D Gatinel
- Service d'ophtalmologie, fondation A.-de-Rothschild, 25, rue Manin, 75940 Paris cedex 19, France
| | - M Mainster
- Department of Ophthalmology, University of Kansas School of Medicine, Prairie Village, Kansas, États-Unis
| |
Collapse
|
126
|
Shelton DA, Gefke I, Summers V, Kim YK, Yu H, Getz Y, Ferdous S, Donaldson K, Liao K, Papania JT, Chrenek MA, Boatright JH, Nickerson JM. Age-Related RPE changes in Wildtype C57BL/6J Mice between 2 and 32 Months. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.574142. [PMID: 38352604 PMCID: PMC10862734 DOI: 10.1101/2024.01.30.574142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Purpose This study provides a systematic evaluation of age-related changes in RPE cell structure and function using a morphometric approach. We aim to better capture nuanced predictive changes in cell heterogeneity that reflect loss of RPE integrity during normal aging. Using C57BL6/J mice ranging from P60-P730, we sought to evaluate how regional changes in RPE shape reflect incremental losses in RPE cell function with advancing age. We hypothesize that tracking global morphological changes in RPE is predictive of functional defects over time. Methods We tested three groups of C57BL/6J mice (young: P60-180; Middle-aged: P365-729; aged: 730+) for function and structural defects using electroretinograms, immunofluorescence, and phagocytosis assays. Results The largest changes in RPE morphology were evident between the young and aged groups, while the middle-aged group exhibited smaller but notable region-specific differences. We observed a 1.9-fold increase in cytoplasmic alpha-catenin expression specifically in the central-medial region of the eye between the young and aged group. There was an 8-fold increase in subretinal, IBA-1-positive immune cell recruitment and a significant decrease in visual function in aged mice compared to young mice. Functional defects in the RPE corroborated by changes in RPE phagocytotic capacity. Conclusions The marked increase of cytoplasmic alpha-catenin expression and subretinal immune cell deposition, and decreased visual output coincide with regional changes in RPE cell morphometrics when stratified by age. These cumulative changes in the RPE morphology showed predictive regional patterns of stress associated with loss of RPE integrity.
Collapse
Affiliation(s)
- Debresha A. Shelton
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Isabelle Gefke
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Vivian Summers
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Yong-Kyu Kim
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Department of Ophthalmology, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul, South Korea
| | - Hanyi Yu
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Department of Computer Science, Emory University, Atlanta, Georgia, United States
| | - Yana Getz
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Salma Ferdous
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Kevin Donaldson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Kristie Liao
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jack T. Papania
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - John M. Nickerson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
127
|
Souto-Neto JA, David DD, Zanetti G, Sua-Cespedes C, Freret-Meurer NV, Moraes MN, de Assis LVM, Castrucci AMDL. Light-specific wavelengths differentially affect the exploration rate, opercular beat, skin color change, opsin transcripts, and the oxi-redox system of the longsnout seahorse Hippocampus reidi. Comp Biochem Physiol A Mol Integr Physiol 2024; 288:111551. [PMID: 37972916 DOI: 10.1016/j.cbpa.2023.111551] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
Light is a strong stimulus for the sensory and endocrine systems. The opsins constitute a large family of proteins that can respond to specific light wavelengths. Hippocampus reidi is a near-threatened seahorse that has a diverse color pattern and sexual dimorphism. Over the years, H. reidi's unique characteristics, coupled with its high demand and over-exploitation for the aquarium trade, have raised concerns about its conservation, primarily due to their significant impact on wild populations. Here, we characterized chromatophore types in juvenile and adult H. reidi in captivity, and the effects of specific light wavelengths with the same irradiance (1.20 mW/cm2) on color change, growth, and survival rate. The xanthophores and melanophores were the major components of H. reidi pigmentation with differences in density and distribution between life stages and sexes. In the eye and skin of juveniles, the yellow (585 nm) wavelength induced a substantial increase in melanin levels compared to the individuals kept under white light (WL), blue (442 nm), or red (650 nm) wavelengths. In addition, blue and yellow wavelengths led to a higher juvenile mortality rate in comparison to the other treatments. Adult seahorses showed a rhythmic color change over 24 h, the highest reflectance values were obtained in the light phase, representing a daytime skin lightening for individuals under WL, blue and yellow wavelength, with changes in the acrophase. The yellow wavelength was more effective on juvenile seahorse pigmentation, while the blue wavelength exerted a stronger effect on the regulation of adult physiological color change. Dramatic changes in the opsin mRNA levels were life stage-dependent, which may infer ontogenetic opsin functions throughout seahorses' development. Exposure to specific wavelengths differentially affected the opsins mRNA levels in the skin and eyes of juveniles. In the juveniles, skin transcripts of visual (rh1, rh2, and lws) and non-visual opsins (opn3 and opn4x) were higher in individuals under yellow light. While in the juvenile's eyes, only rh1 and rh2 had increased transcripts influenced by yellow light; the lws and opn3 mRNA levels were higher in juveniles' eyes under WL. Prolonged exposure to yellow wavelength stimulates a robust increase in the antioxidant enzymes sod1 and sod2 mRNA levels. Our findings indicate that changes in the visible light spectrum alter physiological processes at different stages of life in H. reidi and may serve as the basis for a broader discussion about the implications of artificial light for aquatic species in captivity.
Collapse
Affiliation(s)
- José Araújo Souto-Neto
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil; Laboratory of Micropollutants, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Daniela Dantas David
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Giovanna Zanetti
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Cristhian Sua-Cespedes
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | | | - Maria Nathália Moraes
- Laboratory of Molecular Chronobiology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | | | - Ana Maria de Lauro Castrucci
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil; Department of Biology, University of Virginia, Charlottesville, United States.
| |
Collapse
|
128
|
Yang J, Tian M, Li J, Chen Y, Lin S, Ma X, Chen W, Hou L. Induction of human ESC-derived and adult primary multipotent limbal stem cells into retinal pigment epithelial cells and corneal stromal stem cells. Exp Eye Res 2024; 239:109778. [PMID: 38171475 DOI: 10.1016/j.exer.2023.109778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 12/20/2023] [Accepted: 12/30/2023] [Indexed: 01/05/2024]
Abstract
Human embryonic stem cell (hESC)- and human induced pluripotent stem cell (hiPSC)-derived retinal pigment epithelium (RPE) therapies are promising alternatives for the treatment of retinal degenerative diseases caused by RPE degeneration. The generation of autologous RPE cells from human adult donors, which has the advantage of avoiding immune rejection and teratoma formation, is an alternative cell resource to gain mechanistic insight into and test potential therapies for RPE degenerative diseases. Here, we found that limbal stem cells (LSCs) from hESCs and adult primary human limbus have the potential to produce RPE cells and corneal stromal stem cells (CSSCs). We showed that hESC-LSC-derived RPE cells (LSC-RPE) expressed RPE markers, had a phagocytic function, and synthesized tropical factors. Furthermore, during differentiation from LSCs to RPE cells, cells became pigmented, accompanied by a decrease in the level of LSC marker KRT15 and an increase in the level of RPE marker MITF. The Wnt signaling pathway plays a role in LSC-RPE fate transition, promotes MITF expression in the nucleus, and encourages RPE fate transition. In addition, we also showed that primary LSCs (pLSCs) from adult human limbus similar to hESC-LSC could generate RPE cells, which was supported by the co-expression of LSC and RPE cell markers (KRT15/OTX2, KRT15/MITF), suggesting the transition from pLSC to RPE cells, and typical polygonal morphology, melanization, RPE cell marker genes expression (TYR, RPE65), tight junction formation by ZO-1 expression, and the most crucial phagocytotic function. On the other hand, both hESC-LSCs and pLSCs also differentiated into CSSCs (LSC-CSSCs) that expressed stem cell markers (PAX6, NESTIN), presented MSC features, including surface marker expression and trilineage differentiation capability, like those in human CSSCs. Furthermore, the capability of pLSC-CSSC to differentiate into cells expressing keratocyte marker genes (ALDH3A1, PTGDS, PDK4) indicated the potential to induce keratocytes. These results suggest that the adult pLSC is an alternative cell resource, and its application provides a novel potential therapeutic avenue for preventing RPE dysfunction-related retinal degenerative diseases and corneal scarring.
Collapse
Affiliation(s)
- Juan Yang
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Meiyu Tian
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jinyang Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yu Chen
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Shichao Lin
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoyin Ma
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wei Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ling Hou
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
129
|
Navneet S, Wilson K, Rohrer B. Müller Glial Cells in the Macula: Their Activation and Cell-Cell Interactions in Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2024; 65:42. [PMID: 38416457 PMCID: PMC10910558 DOI: 10.1167/iovs.65.2.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/10/2024] [Indexed: 02/29/2024] Open
Abstract
Müller glia, the main glial cell of the retina, are critical for neuronal and vascular homeostasis in the retina. During age-related macular degeneration (AMD) pathogenesis, Müller glial activation, remodeling, and migrations are reported in the areas of retinal pigment epithelial (RPE) degeneration, photoreceptor loss, and choroidal neovascularization (CNV) lesions. Despite this evidence indicating glial activation localized to the regions of AMD pathogenesis, it is unclear whether these glial responses contribute to AMD pathology or occur merely as a bystander effect. In this review, we summarize how Müller glia are affected in AMD retinas and share a prospect on how Müller glial stress might directly contribute to the pathogenesis of AMD. The goal of this review is to highlight the need for future studies investigating the Müller cell's role in AMD. This may lead to a better understanding of AMD pathology, including the conversion from dry to wet AMD, which has no effective therapy currently and may shed light on drug intolerance and resistance to current treatments.
Collapse
Affiliation(s)
- Soumya Navneet
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Kyrie Wilson
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, United States
- Ralph H. Johnson VA Medical Center, Division of Research, Charleston, South Carolina, United States
| |
Collapse
|
130
|
Wang Y, Zhang YR, Ding ZQ, Zhang YC, Sun RX, Zhu HJ, Wang JN, Xu B, Zhang P, Ji JD, Liu QH, Chen X. m6A-Mediated Upregulation of Imprinted in Prader-Willi Syndrome Induces Aberrant Apical-Basal Polarization and Oxidative Damage in RPE Cells. Invest Ophthalmol Vis Sci 2024; 65:10. [PMID: 38315495 PMCID: PMC10851782 DOI: 10.1167/iovs.65.2.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
Purpose To reveal the clinical significance, pathological involvement and molecular mechanism of imprinted in Prader-Willi syndrome (IPW) in RPE anomalies that contribute to AMD. Methods IPW expression under pathological conditions were detected by microarrays and qPCR assays. In vitro cultured fetal RPE cells were used to study the pathogenicity induced by IPW overexpression and to analyze its upstream and downstream regulatory networks. Results We showed that IPW is upregulated in the macular RPE-choroid tissue of dry AMD patients and in fetal RPE cells under oxidative stress, inflammation and dedifferentiation. IPW overexpression in fetal RPE cells induced aberrant apical-basal polarization as shown by dysregulated polarized markers, disrupted tight and adherens junctions, and inhibited phagocytosis. IPW upregulation was also associated with RPE oxidative damages, as demonstrated by intracellular accumulation of reactive oxygen species, reduced cell proliferation, and accelerated cell apoptosis. Mechanically, N6-methyladenosine level of the IPW transcript regulated its stability with YTHDC1 as the reader. IPW mediated RPE features by suppressing MEG3 expression to sequester its inhibition on the AKT serine-threonine kinase (AKT)/mammalian target of rapamycin (mTOR) pathway. We also noticed that the mTOR inhibitor rapamycin suppresses the AKT/mTOR pathway to alleviate the IPW-induced RPE anomalies. Conclusions We revealed that IPW overexpression in RPE induces aberrant apical-basal polarization and oxidative damages, thus contributing to AMD progression. We also annotated the upstream and downstream regulatory networks of IPW in RPE. Our findings shed new light on the molecular mechanisms of RPE dysfunctions, and indicate that IPW blockers may be a promising option to treat RPE abnormalities in AMD.
Collapse
Affiliation(s)
- Ying Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Ye-Ran Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Zi-Qin Ding
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yi-Chen Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Ru-Xu Sun
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Hong-Jing Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Jia-Nan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Bei Xu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Ping Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Jiang-Dong Ji
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Qing-Huai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Xue Chen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| |
Collapse
|
131
|
Macura IJ, Djuricic I, Major T, Milanovic D, Sobajic S, Kanazir S, Ivkovic S. The supplementation of a high dose of fish oil during pregnancy and lactation led to an elevation in Mfsd2a expression without any changes in docosahexaenoic acid levels in the retina of healthy 2-month-old mouse offspring. Front Nutr 2024; 10:1330414. [PMID: 38328686 PMCID: PMC10847253 DOI: 10.3389/fnut.2023.1330414] [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: 11/14/2023] [Accepted: 12/12/2023] [Indexed: 02/09/2024] Open
Abstract
Introduction During fetal development, the proper development of neural and visual systems relies on the maternal supplementation of omega-3 fatty acids through placental transfer. Pregnant women are strongly advised to augment their diet with additional sources of omega-3, such as fish oil (FO). This supplementation has been linked to a reduced risk of preterm birth, pre-eclampsia, and perinatal depression. Recently, higher doses of omega-3 supplementation have been recommended for pregnant women. Considering that omega-3 fatty acids, particularly docosahexaenoic acid (DHA), play a crucial role in maintaining the delicate homeostasis required for the proper functioning of the retina and photoreceptors the effects of high-dose fish oil (FO) supplementation during pregnancy and lactation on the retina and retinal pigmented epithelium (RPE) in healthy offspring warrant better understanding. Methods The fatty acid content and the changes in the expression of the genes regulating cholesterol homeostasis and DHA transport in the retina and RPE were evaluated following the high-dose FO supplementation. Results Our study demonstrated that despite the high-dose FO treatment during pregnancy and lactation, the rigorous DHA homeostasis in the retina and RPE of the two-month-old offspring remained balanced. Another significant finding of this study is the increase in the expression levels of major facilitator superfamily domain-containing protein (Mfsd2a), a primary DHA transporter. Mfsd2a also serves as a major regulator of transcytosis during development, and a reduction in Mfsd2a levels poses a major risk for the development of leaky blood vessels. Conclusion Impairment of the blood-retinal barrier (BRB) is associated with the development of numerous ocular diseases, and a better understanding of how to manipulate transcytosis in the BRB during development can enhance drug delivery through the BRB or contribute to the repair of central nervous system (CNS) barriers.
Collapse
Affiliation(s)
- Irena Jovanovic Macura
- Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivana Djuricic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Tamara Major
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Desanka Milanovic
- Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | | | - Selma Kanazir
- Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Sanja Ivkovic
- Vinca Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
132
|
Teng M, Wang J, Su X, Tian Y, Ye X, Zhang Y. Causal associations between circulating inflammatory cytokines and blinding eye diseases: a bidirectional Mendelian randomization analysis. Front Aging Neurosci 2024; 16:1324651. [PMID: 38327497 PMCID: PMC10848324 DOI: 10.3389/fnagi.2024.1324651] [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: 10/19/2023] [Accepted: 01/09/2024] [Indexed: 02/09/2024] Open
Abstract
Background Previous studies have explored the associations between circulating inflammatory cytokines and blinding eye diseases, including glaucoma, cataract and macular degeneration. However, the causality of these associations remains controversial. This study employs a bidirectional Mendelian randomization (MR) study to investigate the causal relationships between 41 circulating inflammatory cytokines and these blinding eye diseases. Methods Summary data for glaucoma, cataract, macular degeneration and 41 circulating inflammatory cytokines were publicly available. The inverse variance weighted (IVW) method was employed as the main analysis method. Additionally, various sensitivity tests, including MR-Egger regression, weighted median, weight mode, Cochran's Q test, MR pleiotropy Residual Sum and Outlier test, and leave-one-out test, were conducted to evaluate sensitivity and stability of results. Results The IVW analysis identified six circulating inflammatory cytokines causally associated with the risk of blinding eye diseases: Monokine induced by interferon-gamma (MIG) for glaucoma, interleukin-1 receptor antagonist (IL-1ra), IL-6, IL-10, and platelet derived growth factor BB (PDGFbb) for cataract, and MIG and hepatocyte growth factor (HGF) for macular degeneration. However, it is noteworthy that none of these associations remained significant after Bonferroni correction (p < 0.0004). Reverse MR analyses indicated that cataract may lead to a decrease in vascular endothelial growth factor (VEGF) levels (OR: 3.326 × 10-04, 95% CI: 5.198 × 10-07 - 2.129 × 10-01, p = 0.0151). Conclusion This study highlights the potential roles of specific inflammatory cytokines in the development of glaucoma, cataract and macular degeneration. Moreover, it suggests that VEGF is likely to be involved in cataract development downstream. These findings offer insights for early prevention and novel therapeutic strategies for these blinding eye diseases.
Collapse
Affiliation(s)
- Menghao Teng
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiachen Wang
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xiaochen Su
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ye Tian
- Healthy Food Evaluation Research Center, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xiaomin Ye
- School of Electronic Information and Artiffcial Intelligence, Shaanxi University of Science and Technology, Xi'an, China
| | - Yingang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
133
|
Kocherlakota S, Baes M. Benefits and Caveats in the Use of Retinal Pigment Epithelium-Specific Cre Mice. Int J Mol Sci 2024; 25:1293. [PMID: 38279294 PMCID: PMC10816505 DOI: 10.3390/ijms25021293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
The retinal pigment epithelium (RPE) is an important monolayer of cells present in the outer retina, forming a major part of the blood-retina barrier (BRB). It performs many tasks essential for the maintenance of retinal integrity and function. With increasing knowledge of the retina, it is becoming clear that both common retinal disorders, like age-related macular degeneration, and rare genetic disorders originate in the RPE. This calls for a better understanding of the functions of various proteins within the RPE. In this regard, mice enabling an RPE-specific gene deletion are a powerful tool to study the role of a particular protein within the RPE cells in their native environment, simultaneously negating any potential influences of systemic changes. Moreover, since RPE cells interact closely with adjacent photoreceptors, these mice also provide an excellent avenue to study the importance of a particular gene function within the RPE to the retina as a whole. In this review, we outline and compare the features of various Cre mice created for this purpose, which allow for inducible or non-inducible RPE-specific knockout of a gene of interest. We summarize the various benefits and caveats involved in the use of such mouse lines, allowing researchers to make a well-informed decision on the choice of Cre mouse to use in relation to their research needs.
Collapse
Affiliation(s)
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
134
|
Wang W, Yang T, Chen S, Liang L, Wang Y, Ding Y, Xiong W, Ye X, Guo Y, Shen S, Chen H, Chen J. Tissue engineering RPE sheet derived from hiPSC-RPE cell spheroids supplemented with Y-27632 and RepSox. J Biol Eng 2024; 18:7. [PMID: 38229139 DOI: 10.1186/s13036-024-00405-8] [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: 10/14/2023] [Accepted: 01/08/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Retinal pigment epithelium (RPE) cell therapy is a promising way to treat many retinal diseases. However, obtaining transplantable RPE cells is time-consuming and less effective. This study aimed to develop novel strategies for generating engineered RPE patches with physiological characteristics. RESULTS Our findings revealed that RPE cells derived from human induced pluripotent stem cells (hiPSCs) successfully self-assembled into spheroids. The RPE spheroids treated with Y27632 and Repsox had increased expression of epithelial markers and RPE-specific genes, along with improved cell viability and barrier function. Transcriptome analysis indicated enhanced cell adhesion and extracellular matrix (ECM) organization in RPE spheroids. These RPE spheroids could be seeded and bioprinted on collagen vitrigel (CV) membranes to construct engineered RPE sheets. Circular RPE patches, obtained by trephining a specific section of the RPE sheet, exhibited abundant microvilli and pigment particles, as well as reduced proliferative capacity and enhanced maturation. CONCLUSIONS Our study suggests that the supplementation of small molecules and 3D spheroid culture, as well as the bioprinting technique, can be effective methods to promote RPE cultivation and construct engineered RPE sheets, which may support future clinical RPE cell therapy and the development of RPE models for research applications.
Collapse
Grants
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
Collapse
Affiliation(s)
- Wenxuan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Tingting Yang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Sihui Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Liying Liang
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Yingxin Wang
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Yin Ding
- The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| | - Wei Xiong
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Xiuhong Ye
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Yonglong Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shuhao Shen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Hang Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Jiansu Chen
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China.
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.
- Aier Eye Institute, Changsha, Hunan, China.
| |
Collapse
|
135
|
Bourauel L, Vaisband M, von der Emde L, Bermond K, Tarau IS, Heintzmann R, Holz FG, Curcio CA, Hasenauer J, Ach T. Spectral Analysis of Human Retinal Pigment Epithelium Cells in Healthy and AMD Eyes. Invest Ophthalmol Vis Sci 2024; 65:10. [PMID: 38170540 PMCID: PMC10768704 DOI: 10.1167/iovs.65.1.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Purpose Retinal pigment epithelium (RPE) cells show strong autofluorescence (AF). Here, we characterize the AF spectra of individual RPE cells in healthy eyes and those affected by age-related macular degeneration (AMD) and investigate associations between AF spectral response and the number of intracellular AF granules per cell. Methods RPE-Bruch's membrane flatmounts of 22 human donor eyes, including seven AMD-affected eyes (early AMD, three; geographic atrophy, one; neovascular, three) and 15 unaffected macula (<51 years, eight; >80 years, seven), were imaged at the fovea, perifovea, and near-periphery using confocal AF microscopy (excitation 488 nm), and emission spectra were recorded (500-710 nm). RPE cells were manually segmented with computer assistance and stratified by disease status, and emission spectra were analyzed using cubic spline transforms. Intracellular granules were manually counted and classified. Linear mixed models were used to investigate associations between spectra and the number of intracellular granules. Results Spectra of 5549 RPE cells were recorded. The spectra of RPE cells in healthy eyes showed similar emission curves that peaked at 580 nm for fovea and perifovea and at 575 and 580 nm for near-periphery. RPE spectral curves in AMD eyes differed significantly, being blue shifted by 10 nm toward shorter wavelengths. No significant association coefficients were found between wavelengths and granule counts. Conclusions This large series of RPE cell emission spectra at precisely predefined retinal locations showed a hypsochromic spectral shift in AMD. Combining different microscopy techniques, our work has identified cellular RPE spectral AF and subcellular granule properties that will inform future in vivo investigations using single-cell imaging.
Collapse
Affiliation(s)
- Leonie Bourauel
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Marc Vaisband
- Institute of Life & Medical Sciences, University of Bonn, Bonn, Germany
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute Laboratory for Immunological and Molecular Cancer Research, Paracelsus Medical University, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | | | - Katharina Bermond
- Department of Ophthalmology, Ludwigshafen Hospital, Ludwigshafen, Germany
| | - Ioana Sandra Tarau
- Department of Ophthalmology, Asklepios Klinik Nord - Heidberg, Hamburg, Germany
| | - Rainer Heintzmann
- Leibniz Institute of Photonic Technology, Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller University Jena, Jena, Germany
| | - Frank G. Holz
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Christine A. Curcio
- Department of Ophthalmology, University of Alabama at Birmingham, Alabama, Alabama, United States
| | - Jan Hasenauer
- Institute of Life & Medical Sciences, University of Bonn, Bonn, Germany
| | - Thomas Ach
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| |
Collapse
|
136
|
Agbakwuru D, Wetzel SA. The Biological Significance of Trogocytosis. Results Probl Cell Differ 2024; 73:87-129. [PMID: 39242376 PMCID: PMC11784324 DOI: 10.1007/978-3-031-62036-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Trogocytosis is the intercellular transfer of membrane and membrane-associated proteins between cells. Trogocytosis is an underappreciated phenomenon that has historically routinely been dismissed as an artefact. With a greater understanding of the process and the implications it has on biological systems, trogocytosis has the potential to become a paradigm changer. The presence on a cell of molecules they don't endogenously express can alter the biological activity of the cell and could also lead to the acquisition of new functions. To better appreciate this phenomenon, it is important to understand how these intercellular membrane exchanges influence the function and activity of the donor and the recipient cells. In this chapter, we will examine how the molecules acquired by trogocytosis influence the biology of a variety of systems including mammalian fertilization, treatment of hemolytic disease of the newborn, viral and parasitic infections, cancer immunotherapy, and immune modulation.
Collapse
Affiliation(s)
- Deborah Agbakwuru
- Center for Environmental Health Sciences, University of Montana, Missoula, MT, USA
| | - Scott A Wetzel
- Center for Environmental Health Sciences, University of Montana, Missoula, MT, USA.
- Division of Biological Sciences, University of Montana, Missoula, MT, USA.
| |
Collapse
|
137
|
Manai F, Smedowski A, Kaarniranta K, Comincini S, Amadio M. Extracellular vesicles in degenerative retinal diseases: A new therapeutic paradigm. J Control Release 2024; 365:448-468. [PMID: 38013069 DOI: 10.1016/j.jconrel.2023.11.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/03/2023] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Nanoscale extracellular vesicles (EVs), consisting of exomers, exosomes and microvesicles/ectosomes, have been extensively investigated in the last 20 years, although their biological role is still something of a mystery. EVs are involved in the transfer of lipids, nucleic acids and proteins from donor to recipient cells or distant organs as well as regulating cell-cell communication and signaling. Thus, EVs are important in intercellular communication and this is not limited to sister cells, but may also mediate the crosstalk between different cell types even over long distances. EVs play crucial functions in both cellular homeostasis and the pathogenesis of diseases, and since their contents reflect the status of the donor cell, they represent an additional valuable source of information for characterizing complex biological processes. Recent advances in isolation and analytical methods have led to substantial improvements in both characterizing and engineering EVs, leading to their use either as novel biomarkers for disease diagnosis/prognosis or even as novel therapies. Due to their capacity to carry biomolecules, various EV-based therapeutic applications have been devised for several pathological conditions, including eye diseases. In the eye, EVs have been detected in the retina, aqueous humor, vitreous body and also in tears. Experiences with other forms of intraocular drug applications have opened new ways to use EVs in the treatment of retinal diseases. We here provide a comprehensive summary of the main in vitro, in vivo, and ex vivo literature-based studies on EVs' role in ocular physiological and pathological conditions. We have focused on age-related macular degeneration, diabetic retinopathy, glaucoma, which are common eye diseases leading to permanent blindness, if not treated properly. In addition, the putative use of EVs in retinitis pigmentosa and other retinopathies is discussed. Finally, we have reviewed the potential of EVs as therapeutic tools and/or biomarkers in the above-mentioned retinal disorders. Evidence emerging from experimental disease models and human material strongly suggests future diagnostic and/or therapeutic exploitation of these biological agents in various ocular disorders with a good possibility to improve the patient's quality of life.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Adrian Smedowski
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland; GlaucoTech Co., Katowice, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland; Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - Sergio Comincini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | | |
Collapse
|
138
|
Zhang K, Cai W, Hu L, Chen S. Generating Retinas through Guided Pluripotent Stem Cell Differentiation and Direct Somatic Cell Reprogramming. Curr Stem Cell Res Ther 2024; 19:1251-1262. [PMID: 37807418 DOI: 10.2174/011574888x255496230923164547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 10/10/2023]
Abstract
Retinal degeneration diseases affect millions of people worldwide but are among the most difficult eye diseases to cure. Studying the mechanisms and developing new therapies for these blinding diseases requires researchers to have access to many retinal cells. In recent years there has been substantial advances in the field of biotechnology in generating retinal cells and even tissues in vitro, either through programmed sequential stem cell differentiation or direct somatic cell lineage reprogramming. The resemblance of these in vitro-generated retinal cells to native cells has been increasingly utilized by researchers. With the help of these in vitro retinal models, we now have a better understanding of human retinas and retinal diseases. Furthermore, these in vitro-generated retinal cells can be used as donor cells which solves a major hurdle in the development of cell replacement therapy for retinal degeneration diseases, while providing a promising option for patients suffering from these diseases. In this review, we summarize the development of pluripotent stem cell-to-retinal cell differentiation methods, the recent advances in generating retinal cells through direct somatic cell reprogramming, and the translational applications of retinal cells generated in vitro. Finally, we discuss the limitations of the current protocols and possible future directions for improvement.
Collapse
Affiliation(s)
- Ke Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510623, China
| | - Wenwen Cai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510623, China
| | - Leyi Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510623, China
| | - Shuyi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510623, China
| |
Collapse
|
139
|
Oribio-Quinto C, Herranz-Heras JC, Burgos-Blasco B, Alarcon-Garcia AD, Fernández-Vigo JI. Clinical and multimodal imaging findings in a case of serous maculopathy with absence of retinal pigment epithelium (SMARPE). Eur J Ophthalmol 2024; 34:NP66-NP69. [PMID: 37038336 DOI: 10.1177/11206721231169310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
INTRODUCTION The differential diagnosis for serous SRF can involve diseases with widely different pathogenic mechanisms that can range from vascular ocular diseases to ocular tumours and paraneoplastic syndromes. Recently, van Dijk et al. have described in three patients a new entity which they have called serous maculopathy with an absence of retinal pigment epithelium (SMARPE). We hereby describe a case of this infrequent macular disease and report its characteristic findings on multimodal imaging. CASE DESCRIPTION We present the case of a 65-year-old hyperopic woman with a three-year history of visual acuity (VA) loss in her left eye. Prior optical coherence tomography (OCT) had revealed the presence of serous subretinal fluid that had shown no response to treatment with intravitreal injections. On swept source OCT angiography scan, no macular alterations in the retinal vascular plexus structure were noted and there was no evidence of choroidal neovascularization. Ultra-widefield fluorescence angiography of the left eye revealed an early hyperfluorescent macular spot corresponding to the area of absent RPE and late fluorescein pooling. On ultra-widefield indocyanine green angiography there were no central or peripheral abnormalities of choroidal vascularization. CONCLUSION This recently described entity should be considered as a differential diagnosis in persistent serous subretinal fluid. Multimodal imaging helps differentiate SMARPE from its main differential diagnoses, and care should be taken to identify and differentiate it from similar conditions to avoid unnecessary treatment with its possible side effects and complications.
Collapse
Affiliation(s)
| | - Juan Carlos Herranz-Heras
- Department of ophthalmology, Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Madrid, Spain
| | | | | | - José Ignacio Fernández-Vigo
- Department of ophthalmology, Hospital Clínico San Carlos, Madrid, Spain
- Centro Internacional de Oftalmologia Avanzada, Madrid, Spain
| |
Collapse
|
140
|
Upadhyay M, Bonilha VL. Regulated cell death pathways in the sodium iodate model: Insights and implications for AMD. Exp Eye Res 2024; 238:109728. [PMID: 37972750 PMCID: PMC10841589 DOI: 10.1016/j.exer.2023.109728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
The sodium iodate (NaIO3) model of increased oxidative stress recapitulates dry AMD features such as patchy RPE loss, secondary photoreceptors, and underlying choriocapillaris death, allowing longitudinal evaluation of the retinal structure. Due to the time- and dose-dependent degeneration observed in diverse animal models, this preclinical model has become one of the most studied models. The events leading to RPE cell death post- NaIO3 injection have been extensively studied, and here we have reviewed different modalities of cell death, including apoptosis, necroptosis, ferroptosis, and pyroptosis with a particular focus on findings associated with in vivo and in vitro NaIO3 studies on RPE cell death. Because the fundamental cause of vision loss in patients with dry AMD is the death of these same cells affected by NaIO3, studies using NaIO3 can provide valuable insights into RPE and photoreceptor cell death mechanisms and can help understand mechanisms behind RPE degeneration in AMD.
Collapse
Affiliation(s)
- Mala Upadhyay
- Cole Eye Institute, Ophthalmic Research, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Vera L Bonilha
- Cole Eye Institute, Ophthalmic Research, Cleveland Clinic, Cleveland, OH, 44195, USA; Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.
| |
Collapse
|
141
|
Shanbagh S, Gadde SG, Shetty R, Heymans S, Abilash VG, Chaurasia SS, Ghosh A. Hyperglycemia-induced miR182-5p drives glycolytic and angiogenic response in Proliferative Diabetic Retinopathy and RPE cells via depleting FoxO1. Exp Eye Res 2024; 238:109713. [PMID: 37952722 DOI: 10.1016/j.exer.2023.109713] [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: 04/24/2023] [Revised: 09/10/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023]
Abstract
PURPOSE Diabetic Retinopathy (DR) is associated with metabolic dysfunction in cells such as retinal pigmented epithelium (RPE). Small molecular weight microRNAs can simultaneously regulate multiple gene products thus having pivotal roles in disease pathogenesis. Since miR182-5p is involved in regulating glycolysis and angiogenesis, two pathologic processes of DR, we investigated its status in DR eyes and in high glucose model in vitro. METHOD ology: Total RNA was extracted from vitreous humor of PDR (n = 48) and macular hole (n = 22) subjects followed by quantification of miR182-5p and its target genes. ARPE-19 cells, cultured in DMEM under differential glucose conditions (5 mM and 25 mM) were used for metabolic and biochemical assays. Cells were transfected with miRNA182 mimic or antagomir to evaluate the gain and loss of function effects. RESULTS PDR patient eyes had high levels of miR182-5p levels (p < 0.05). RPE cells under high glucose stress elevated miR182-5p expression with altered glycolytic pathway drivers such as HK2, PFKP and PKM2 over extended durations. Additionally, RPE cells under high glucose conditions exhibited reduced FoxO1 and enhanced Akt activation. RPE cells transfected with miR182-5p mimic phenocopied the enhanced basal and compensatory glycolytic rates observed under high glucose conditions with increased VEGF secretion. Conversely, inhibiting miR182-5p reduced Akt activation, glycolytic pathway proteins, and VEGF while stabilizing FoxO1. CONCLUSION Glycolysis-associated proteins downstream of the FoxO1-Akt axis were regulated by miR182-5p. Further, miR182-5p increased expression of VEGFR2 and VEGF levels, likely via inhibition of ZNF24. Thus, the FoxO1-Akt-glycolysis/VEGF pathway driving metabolic dysfunction with concurrent angiogenic signaling in PDR may be potentially targeted for treatment via miR182-5p modulation.
Collapse
Affiliation(s)
- Shaika Shanbagh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India; Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | | | | | | | - V G Abilash
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| | - Shyam S Chaurasia
- Ocular Immunology & Angiogenesis Lab, Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India.
| |
Collapse
|
142
|
Dujardin C, Habeler W, Monville C, Letourneur D, Simon-Yarza T. Advances in the engineering of the outer blood-retina barrier: From in-vitro modelling to cellular therapy. Bioact Mater 2024; 31:151-177. [PMID: 37637086 PMCID: PMC10448242 DOI: 10.1016/j.bioactmat.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/13/2023] [Accepted: 08/06/2023] [Indexed: 08/29/2023] Open
Abstract
The outer blood-retina barrier (oBRB), crucial for the survival and the proper functioning of the overlying retinal layers, is disrupted in numerous diseases affecting the retina, leading to the loss of the photoreceptors and ultimately of vision. To study the oBRB and/or its degeneration, many in vitro oBRB models have been developed, notably to investigate potential therapeutic strategies against retinal diseases. Indeed, to this day, most of these pathologies are untreatable, especially once the first signs of degeneration are observed. To cure those patients, a current strategy is to cultivate in vitro a mature oBRB epithelium on a custom membrane that is further implanted to replace the damaged native tissue. After a description of the oBRB and the related diseases, this review presents an overview of the oBRB models, from the simplest to the most complex. Then, we propose a discussion over the used cell types, for their relevance to study or treat the oBRB. Models designed for in vitro applications are then examined, by paying particular attention to the design evolution in the last years, the development of pathological models and the benefits of co-culture models, including both the retinal pigment epithelium and the choroid. Lastly, this review focuses on the models developed for in vivo implantation, with special emphasis on the choice of the material, its processing and its characterization, before discussing the reported pre-clinical and clinical trials.
Collapse
Affiliation(s)
- Chloé Dujardin
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science (LVTS) INSERM-U1148, 75018 Paris, France
| | - Walter Habeler
- INSERM U861, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100, Corbeil-Essonnes, France
- U861, I-Stem, AFM, Université Paris-Saclay, Université D’Evry, 91100, Corbeil-Essonnes, France
- CECS, Centre D’étude des Cellules Souches, 91100, Corbeil-Essonnes, France
| | - Christelle Monville
- INSERM U861, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100, Corbeil-Essonnes, France
- U861, I-Stem, AFM, Université Paris-Saclay, Université D’Evry, 91100, Corbeil-Essonnes, France
| | - Didier Letourneur
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science (LVTS) INSERM-U1148, 75018 Paris, France
| | - Teresa Simon-Yarza
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science (LVTS) INSERM-U1148, 75018 Paris, France
| |
Collapse
|
143
|
Różanowska MB. Lipofuscin, Its Origin, Properties, and Contribution to Retinal Fluorescence as a Potential Biomarker of Oxidative Damage to the Retina. Antioxidants (Basel) 2023; 12:2111. [PMID: 38136230 PMCID: PMC10740933 DOI: 10.3390/antiox12122111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Lipofuscin accumulates with age as intracellular fluorescent granules originating from incomplete lysosomal digestion of phagocytosed and autophagocytosed material. The purpose of this review is to provide an update on the current understanding of the role of oxidative stress and/or lysosomal dysfunction in lipofuscin accumulation and its consequences, particularly for retinal pigment epithelium (RPE). Next, the fluorescence of lipofuscin, spectral changes induced by oxidation, and its contribution to retinal fluorescence are discussed. This is followed by reviewing recent developments in fluorescence imaging of the retina and the current evidence on the prognostic value of retinal fluorescence for the progression of age-related macular degeneration (AMD), the major blinding disease affecting elderly people in developed countries. The evidence of lipofuscin oxidation in vivo and the evidence of increased oxidative damage in AMD retina ex vivo lead to the conclusion that imaging of spectral characteristics of lipofuscin fluorescence may serve as a useful biomarker of oxidative damage, which can be helpful in assessing the efficacy of potential antioxidant therapies in retinal degenerations associated with accumulation of lipofuscin and increased oxidative stress. Finally, amendments to currently used fluorescence imaging instruments are suggested to be more sensitive and specific for imaging spectral characteristics of lipofuscin fluorescence.
Collapse
Affiliation(s)
- Małgorzata B. Różanowska
- School of Optometry and Vision Sciences, College of Biomedical and Life Sciences, Cardiff University, Maindy Road, Cardiff CF24 4HQ, Wales, UK;
- Cardiff Institute for Tissue Engineering and Repair (CITER), Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK
| |
Collapse
|
144
|
Zinflou C, Rochette PJ. Indenopyrene and Blue-Light Co-Exposure Impairs the Tightly Controlled Activation of Xenobiotic Metabolism in Retinal Pigment Epithelial Cells: A Mechanism for Synergistic Toxicity. Int J Mol Sci 2023; 24:17385. [PMID: 38139215 PMCID: PMC10744144 DOI: 10.3390/ijms242417385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
High energy visible (HEV) blue light is an increasing source of concern for visual health. Polycyclic aromatic hydrocarbons (PAH), a group of compounds found in high concentrations in smokers and polluted environments, accumulate in the retinal pigment epithelium (RPE). HEV absorption by indeno [1,2,3-cd]pyrene (IcdP), a common PAH, synergizes their toxicities and promotes degenerative changes in RPE cells comparable to the ones observed in age-related macular degeneration. In this study, we decipher the processes underlying IcdP and HEV synergic toxicity in human RPE cells. We found that IcdP-HEV toxicity is caused by the loss of the tight coupling between the two metabolic phases ensuring IcdP efficient detoxification. Indeed, IcdP/HEV co-exposure induces an overactivation of key actors in phase I metabolism. IcdP/HEV interaction is also associated with a downregulation of proteins involved in phase II. Our data thus indicate that phase II is hindered in response to co-exposure and that it is insufficient to sustain the enhanced phase I induction. This is reflected by an accelerated production of endogenous reactive oxygen species (ROS) and an increased accumulation of IcdP-related bulky DNA damage. Our work raises the prospect that lifestyle and environmental pollution may be significant modulators of HEV toxicity in the retina.
Collapse
Affiliation(s)
- Corinne Zinflou
- Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec, Hôpital du Saint-Sacrement, Université Laval, Quebec, QC G1S 4L8, Canada;
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Patrick J. Rochette
- Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec, Hôpital du Saint-Sacrement, Université Laval, Quebec, QC G1S 4L8, Canada;
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Université Laval, Quebec, QC G1V 0A6, Canada
- Département d’Ophtalmologie et ORL—Chirurgie Cervico-Faciale, Université Laval, Quebec, QC G1V 0A6, Canada
| |
Collapse
|
145
|
Farjood F, Manos JD, Wang Y, Williams AL, Zhao C, Borden S, Alam N, Prusky G, Temple S, Stern JH, Boles NC. Identifying biomarkers of heterogeneity and transplantation efficacy in retinal pigment epithelial cells. J Exp Med 2023; 220:e20230913. [PMID: 37728563 PMCID: PMC10510736 DOI: 10.1084/jem.20230913] [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: 05/26/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023] Open
Abstract
Transplantation of retinal pigment epithelial (RPE) cells holds great promise for patients with retinal degenerative diseases, such as age-related macular degeneration. In-depth characterization of RPE cell product identity and critical quality attributes are needed to enhance efficacy and safety of replacement therapy strategies. Here, we characterized an adult RPE stem cell-derived (RPESC-RPE) cell product using bulk and single-cell RNA sequencing (scRNA-seq), assessing functional cell integration in vitro into a mature RPE monolayer and in vivo efficacy by vision rescue in the Royal College of Surgeons rats. scRNA-seq revealed several distinct subpopulations in the RPESC-RPE product, some with progenitor markers. We identified RPE clusters expressing genes associated with in vivo efficacy and increased cell integration capability. Gene expression analysis revealed lncRNA (TREX) as a predictive marker of in vivo efficacy. TREX knockdown decreased cell integration while overexpression increased integration in vitro and improved vision rescue in the RCS rats.
Collapse
Affiliation(s)
| | | | - Yue Wang
- Neural Stem Cell Institute, Rensselaer, NY, USA
| | | | | | | | - Nazia Alam
- Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, USA
| | - Glen Prusky
- Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, USA
| | | | | | | |
Collapse
|
146
|
Wongchaisuwat N, Wang J, Yang P, Everett L, Gregor A, Sahel JA, Nischal KK, Pennesi ME, Gillingham MB, Jia Y. Optical coherence tomography angiography of choroidal neovascularization in long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD). Am J Ophthalmol Case Rep 2023; 32:101958. [PMID: 38161518 PMCID: PMC10757195 DOI: 10.1016/j.ajoc.2023.101958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 01/03/2024] Open
Abstract
Purpose To report the clinical utility of optical coherence tomography angiography (OCTA) for demonstrating choroidal neovascularization (CNV) associated with Long-Chain 3-Hydroxyacyl-CoA Dehydrogenase Deficiency (LCHADD) retinopathy. Methods Thirty-three participants with LCHADD (age 7-36 years; median 17) were imaged with OCTA and the Center for Ophthalmic Optics & Lasers Angiography Reading Toolkit (COOL-ART) software was implemented to process OCTA scans. Results Seven participants (21 %; age 17-36 years; median 25) with LCHADD retinopathy demonstrated evidence of CNV by retinal examination or presence of CNV within outer retinal tissue on OCTA scans covering 3 × 3 and/or 6 × 6-mm. These sub-clinical CNVs are adjacent to hyperpigmented areas in the posterior pole. CNV presented at stage 2 or later of LCHADD retinopathy prior to the disappearance of RPE pigment in the macula. Conclusion OCTA can be applied as a non-invasive method to evaluate the retinal and choroidal microvasculature. OCTA can reveal CNV in LCHADD even when the clinical exam is inconclusive. These data suggest that the incidence of CNV is greater than expected and can occur even in the early stages of LCHADD retinopathy.
Collapse
Affiliation(s)
- Nida Wongchaisuwat
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
- Department of Ophthalmology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jie Wang
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
| | - Paul Yang
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
| | - Lesley Everett
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Ashley Gregor
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Jose Alain Sahel
- Vision Institute, University of Pittsburgh Medical Center and School of Medicine, Pennsylvania, USA
| | - Ken K. Nischal
- Vision Institute, University of Pittsburgh Medical Center and School of Medicine, Pennsylvania, USA
- UPMC Children's Hospital, Pennsylvania, USA
| | - Mark E. Pennesi
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Melanie B. Gillingham
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Yali Jia
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
147
|
Burri C, Salzmann S, Wandel J, Hoffmann L, Považay B, Meier C, Frenz M. Real-time OCT feedback-controlled RPE photodisruption in ex vivo porcine eyes using 8 microsecond laser pulses. BIOMEDICAL OPTICS EXPRESS 2023; 14:6328-6349. [PMID: 38420306 PMCID: PMC10898567 DOI: 10.1364/boe.503941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/24/2023] [Accepted: 11/12/2023] [Indexed: 03/02/2024]
Abstract
Selective retinal pigment epithelium (RPE) photodisruption requires reliable real-time feedback dosimetry (RFD) to prevent unwanted overexposure. In this study, optical coherence tomography (OCT) based RFD was investigated in ex vivo porcine eyes exposed to laser pulses of 8 µs duration (wavelength: 532 nm, exposure area: 90 × 90 µm2, radiant exposure: 247 to 1975 mJ/µm2). For RFD, fringe washouts in time-resolved OCT M-scans (central wavelength: 870 nm, scan rate: 85 kHz) were compared to an RPE cell viability assay. Statistical analysis revealed a moderate correlation between RPE lesion size and applied treatment energy, suggesting RFD adaptation to inter- and intraindividual RPE pigmentation and ocular transmission.
Collapse
Affiliation(s)
- Christian Burri
- optoLab, Institute for Human Centered Engineering, Bern University of Applied Sciences, Biel, Switzerland
- Biomedical Photonics Group, Institute of Applied Physics, University of Bern, Bern, Switzerland
| | - Simon Salzmann
- optoLab, Institute for Human Centered Engineering, Bern University of Applied Sciences, Biel, Switzerland
| | - Jasmin Wandel
- Institute for Optimisation and Data Analysis, Bern University of Applied Sciences, Burgdorf, Switzerland
| | - Leonie Hoffmann
- optoLab, Institute for Human Centered Engineering, Bern University of Applied Sciences, Biel, Switzerland
| | - Boris Považay
- optoLab, Institute for Human Centered Engineering, Bern University of Applied Sciences, Biel, Switzerland
| | - Christoph Meier
- optoLab, Institute for Human Centered Engineering, Bern University of Applied Sciences, Biel, Switzerland
| | - Martin Frenz
- Biomedical Photonics Group, Institute of Applied Physics, University of Bern, Bern, Switzerland
| |
Collapse
|
148
|
Feldman T, Yakovleva M, Utina D, Ostrovsky M. Short-Term and Long-Term Effects after Exposure to Ionizing Radiation and Visible Light on Retina and Retinal Pigment Epithelium of Mouse Eye. Int J Mol Sci 2023; 24:17049. [PMID: 38069372 PMCID: PMC10707529 DOI: 10.3390/ijms242317049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/25/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
A comparative in vivo study of the effects of ionizing radiation (accelerated protons) and visible light (400-700 nm) on the retina and retinal pigment epithelium (RPE) of the mouse eye was carried out. Using the methods of fluorescence spectroscopy and high-performance liquid chromatography (HPLC), we analyzed the relative composition of retinoids in chloroform extracts obtained from the retinas and RPEs immediately after exposure of animals to various types of radiation and 4.5 months after they were exposed and maintained under standard conditions throughout the period. The fluorescent properties of chloroform extracts were shown to change upon exposure to various types of radiation. This fact indicates the accumulation of retinoid oxidation and degradation products in the retina and RPE. The data from fluorescence and HPLC analyses of retinoids indicate that when exposed to ionizing radiation, retinoid oxidation processes similar to photooxidation occur. Both ionizing radiation and high-intensity visible light have been shown to be characterized by long-term effects. The action of any type of radiation is assumed to activate the mechanism of enhanced reactive oxygen species production, resulting in a long-term damaging effect.
Collapse
Affiliation(s)
- Tatiana Feldman
- Department of Biology, Lomonosov Moscow State University, Leninskiye Gory 1, Moscow 119234, Russia;
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygin Street, Moscow 119334, Russia;
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, Moscow 119334, Russia
| | - Marina Yakovleva
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygin Street, Moscow 119334, Russia;
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, Moscow 119334, Russia
| | - Dina Utina
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna 141980, Russia;
| | - Mikhail Ostrovsky
- Department of Biology, Lomonosov Moscow State University, Leninskiye Gory 1, Moscow 119234, Russia;
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygin Street, Moscow 119334, Russia;
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, Moscow 119334, Russia
| |
Collapse
|
149
|
Xia X, Guo X. Adeno-associated virus vectors for retinal gene therapy in basic research and clinical studies. Front Med (Lausanne) 2023; 10:1310050. [PMID: 38105897 PMCID: PMC10722277 DOI: 10.3389/fmed.2023.1310050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/09/2023] [Indexed: 12/19/2023] Open
Abstract
Retinal degenerative diseases, including glaucoma, age-related macular degeneration, diabetic retinopathy, and a broad range of inherited retinal diseases, are leading causes of irreversible vision loss and blindness. Gene therapy is a promising and fast-growing strategy to treat both monogenic and multifactorial retinal disorders. Vectors for gene delivery are crucial for efficient and specific transfer of therapeutic gene(s) into target cells. AAV vectors are ideal for retinal gene therapy due to their inherent advantages in safety, gene expression stability, and amenability for directional engineering. The eye is a highly compartmentalized organ composed of multiple disease-related cell types. To determine a suitable AAV vector for a specific cell type, the route of administration and choice of AAV variant must be considered together. Here, we provide a brief overview of AAV vectors for gene transfer into important ocular cell types, including retinal pigment epithelium cells, photoreceptors, retinal ganglion cells, Müller glial cells, ciliary epithelial cells, trabecular meshwork cells, vascular endothelial cells, and pericytes, via distinct injection methods. By listing suitable AAV vectors in basic research and (pre)clinical studies, we aim to highlight the progress and unmet needs of AAV vectors in retinal gene therapy.
Collapse
Affiliation(s)
| | - Xinzheng Guo
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, China
| |
Collapse
|
150
|
Lee IK, Xie R, Luz-Madrigal A, Min S, Zhu J, Jin J, Edwards KL, Phillips MJ, Ludwig AL, Gamm DM, Gong S, Ma Z. Micromolded honeycomb scaffold design to support the generation of a bilayered RPE and photoreceptor cell construct. Bioact Mater 2023; 30:142-153. [PMID: 37575875 PMCID: PMC10415596 DOI: 10.1016/j.bioactmat.2023.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/27/2023] [Accepted: 07/22/2023] [Indexed: 08/15/2023] Open
Abstract
Age-related macular degeneration (AMD) causes blindness due to loss of retinal pigment epithelium (RPE) and photoreceptors (PRs), which comprise the two outermost layers of the retina. Given the small size of the macula and the importance of direct contact between RPE and PRs, the use of scaffolds for targeted reconstruction of the outer retina in later stage AMD and other macular dystrophies is particularly attractive. We developed microfabricated, honeycomb-patterned, biodegradable poly(glycerol sebacate) (PGS) scaffolds to deliver organized, adjacent layers of RPE and PRs to the subretinal space. Furthermore, an optimized process was developed to photocure PGS, shortening scaffold production time from days to minutes. The resulting scaffolds robustly supported the seeding of human pluripotent stem cell-derived RPE and PRs, either separately or as a dual cell-layered construct. These advanced, economical, and versatile scaffolds can accelerate retinal cell transplantation efforts and benefit patients with AMD and other retinal degenerative diseases.
Collapse
Affiliation(s)
- In-Kyu Lee
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Ruosen Xie
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Agustin Luz-Madrigal
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Comparative Biomedical Sciences, University of Wisconsin–Madison, Madison, WI, 53706, USA
| | - Seunghwan Min
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Jingcheng Zhu
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jiahe Jin
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | | | - M. Joseph Phillips
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Allison L. Ludwig
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - David M. Gamm
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53705, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Comparative Biomedical Sciences, University of Wisconsin–Madison, Madison, WI, 53706, USA
| | - Shaoqin Gong
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Zhenqiang Ma
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Grainger Institute for Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|