1
|
Bai Y, Wang R, Jin C, Wang L, Tang Y, Wang H, Ding K, Cai S. Impacts of sulfate polysaccharide JCS1S2 on retinal neovascularization in oxygen-induced retinopathy rats. Front Pharmacol 2025; 16:1499420. [PMID: 40271076 PMCID: PMC12014746 DOI: 10.3389/fphar.2025.1499420] [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: 09/20/2024] [Accepted: 03/19/2025] [Indexed: 04/25/2025] Open
Abstract
Retinal neovascularization, a pathological form of angiogenesis, is a leading cause of blindness. This study investigated the effects of Dendrobium sulfate polysaccharide JCS1S2, derived from Dendrobium chrysogenum, on oxygen-induced retinopathy (OIR) in rat models and Müller cells of the rat retina. To this end, we established an OIR rat model and divided the rats into three primary groups, namely, Group I (control), Group II (OIR), and Group III (OIR + JCS1S2). Group III was further subdivided into three subgroups treated with different concentrations of JCS1S2 (10 μg/μL, 20 μg/μL, and 40 μg/μL). After finding the optimal concentration of JCS1S2 by ADP and HE, PCR, Western blot and transcriptome sequencing were used to analyze the role of JCS1S2 in Müller cells of OIR rats and rat retinas. ADP and hematoxylin and eosin (HE) staining revealed that JCS1S2 dose-dependently inhibited retinal neovascularization. Quantitative polymerase chain reaction (qPCR) and Western blot analyses showed significant downregulation of vascular endothelial growth factor A (VEGF-A), VEGF-B, VEGF-D, VEGF receptor 1 (VEGF-R1), and VEGF receptor 2 (VEGF-R2) following JCS1S2 treatment. Transcriptome analysis suggested that JCS1S2 may suppress the activation of the Toll-like receptor (TLR) signaling pathway, regulate the expression of genes associated with endothelial activation and angiogenesis, and participate in the inflammatory and metabolic pathways of the retina. Western blotting data indicate that JCS1S2 can markedly reduce abnormal retinal angiogenesis and Müller cell activation in OIR rats through the TLR4/p-NF-κB/VEGF pathway, JCS1S2 may have the potential as a therapeutic agent for retinal neovascularization.
Collapse
Affiliation(s)
- Yang Bai
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Rui Wang
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Can Jin
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, Zhongshan, China
- Glycochemistry and Glycobiology Lab, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Lili Wang
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Yun Tang
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Han Wang
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Kan Ding
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, Zhongshan, China
- Glycochemistry and Glycobiology Lab, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shanjun Cai
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
2
|
Dobariya KH, Goyal D, Kumar H. Molecular signature-based labeling techniques for vascular endothelial cells. Acta Histochem 2025; 127:152222. [PMID: 39644518 DOI: 10.1016/j.acthis.2024.152222] [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: 07/13/2024] [Revised: 11/14/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
Vascular endothelial cells (VECs) play a crucial role in the development and maintenance of vascular biology specific to the tissue types. Molecular signature-based labeling and imaging of VECs help researchers understand potential mechanisms linking VECs to disease pathology, serving as valuable biomarkers in clinical settings and trials. Labeling techniques involve selectively tagging or marking VECs for visualization. Immunolabeled employs antibodies that specifically bind to VECs markers, while fluorescent tracers or dyes can directly label VECs for imaging. Some techniques use specific carbohydrate residues on cell surface, while others employ endothelial-specific promoters to express fluorescent proteins. Additionally, VEC can be labeled with contrast agents, radiolabeled tracers, and nanoparticles. The choice of labeling technique depends on study context, including whether it involves animal models, in vitro cell cultures, or clinical applications. Herein, we discussed the various labeling methods utilized to label VECs and the techniques to visualize them.
Collapse
Affiliation(s)
- Krutika H Dobariya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Divya Goyal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
3
|
Saito R, Inagaki A, Nakamura Y, Imura T, Kanai N, Mitsugashira H, Endo Kumata Y, Katano T, Suzuki S, Tokodai K, Kamei T, Unno M, Watanabe K, Tabata Y, Goto M. A Gelatin Hydrogel Nonwoven Fabric Combined With Adipose Tissue-Derived Stem Cells Enhances Subcutaneous Islet Engraftment. Cell Transplant 2024; 33:9636897241251621. [PMID: 38756050 PMCID: PMC11102670 DOI: 10.1177/09636897241251621] [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: 02/12/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 05/18/2024] Open
Abstract
Subcutaneous islet transplantation is a promising treatment for severe diabetes; however, poor engraftment hinders its prevalence. We previously revealed that a gelatin hydrogel nonwoven fabric (GHNF) markedly improved subcutaneous islet engraftment. We herein investigated whether the addition of adipose tissue-derived stem cells (ADSCs) to GHNF affected the outcome. A silicone spacer sandwiched between two GHNFs with (AG group) or without (GHNF group) ADSCs, or a silicone spacer alone (Silicone group) was implanted into the subcutaneous space of healthy mice at 6 weeks before transplantation, then diabetes was induced 7 days before transplantation. Syngeneic islets were transplanted into the pretreated space. Intraportal transplantation (IPO group) was also performed to compare the transplant efficiency. Blood glucose, intraperitoneal glucose tolerance, immunohistochemistry, and inflammatory mediators were evaluated. The results in the subcutaneous transplantation were compared using the Silicone group as a control. The results of the IPO group were also compared with those of the AG group. The AG group showed significantly better blood glucose changes than the Silicone and the IPO groups. The cure rate of AG group (72.7%) was the highest among the groups (GHNF; 40.0%, IPO; 40.0%, Silicone; 0%). The number of vWF-positive vessels in the subcutaneous space of the AG group was significantly higher than that in other groups before transplantation (P < 0.01). Lectin angiography also showed that the same results (P < 0.05). According to the results of the ADSCs tracing, ADSCs did not exist at the transplant site (6 weeks after implantation). The positive rates for laminin and collagen III constructed around the transplanted islets did not differ among groups. Inflammatory mediators were higher in the Silicone group, followed by the AG and GHNF groups. Pretreatment using bioabsorbable scaffolds combined with ADSCs enhanced neovascularization in subcutaneous space, and subcutaneous islet transplantation using GHNF with ADSCs was superior to intraportal islet transplantation.
Collapse
Affiliation(s)
- Ryusuke Saito
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Norifumi Kanai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Mitsugashira
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukiko Endo Kumata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takumi Katano
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shoki Suzuki
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuaki Tokodai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kimiko Watanabe
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiko Tabata
- Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
4
|
Bhutto IA, McLeod DS, Thomson BR, Lutty GA, Edwards MM. Visualization of choroidal vasculature in pigmented mouse eyes from experimental models of AMD. Exp Eye Res 2024; 238:109741. [PMID: 38056552 PMCID: PMC10872330 DOI: 10.1016/j.exer.2023.109741] [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: 10/12/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 12/08/2023]
Abstract
A variety of techniques exist to investigate retinal and choroidal vascular changes in experimental mouse models of human ocular diseases. While all have specific advantages, a method for evaluating the choroidal vasculature in pigmented mouse eyes has been more challenging especially for whole mount visualization and morphometric analysis. Here we report a simple, reliable technique involving bleaching pigment prior to immunostaining the vasculature in whole mounts of pigmented mouse choroids. Eyes from healthy adult pigmented C57BL/6J mice were used to establish the methodology. The retina and anterior segment were separated from the choroid. The choroid with retinal pigment epithelial cells (RPE) and sclera was soaked in 1% ethylenediaminetetraacetic acid (EDTA) to remove the RPE. Tissues were fixed in 2% paraformaldehyde (PFA) in phosphate-buffered saline (PBS). Choroids were subjected to melanin bleaching with 10% hydrogen peroxide (H2O2) at 55 °C for 90 min, washed in PBS and then immunostained with anti-podocalyxin antibody to label vascular endothelium followed by Cy3-AffiniPure donkey anti-goat IgG at 4 °C overnight. Images of immunostained bleached choroids were captured using a Zeiss 710 confocal microscope. In addition to control eyes, this method was used to analyze the choroids from subretinal sodium iodate (NaIO3) RPE atrophy and laser-induced choroidal neovascularization (CNV) mouse models. The H2O2 pretreatment effectively bleached the melanin, resulting in a transparent choroid. Immunolabeling with podocalyxin antibody following bleaching provided excellent visualization of choroidal vasculature in the flat perspective. In control choroids, the choriocapillaris (CC) displayed different anatomical patterns in peripapillary (PP), mid peripheral (MP) and far peripheral (FP) choroid. Morphometric analysis of the vascular area (VA) revealed that the CC was most dense in the PP region (87.4 ± 4.3% VA) and least dense in FP (79.9 ± 6.7% VA). CC diameters also varied depending on location from 11.4 ± 1.97 mm in PP to 15.1 ± 3.15 mm in FP. In the NaIO3-injected eyes, CC density was significantly reduced in the RPE atrophic regions (50.7 ± 5.8% VA in PP and 45.8 ± 6.17% VA in MP) compared to the far peripheral non-atrophic regions (82.8 ± 3.8% VA). CC diameters were significantly reduced in atrophic regions (6.35 ± 1.02 mm in PP and 6.5 ± 1.2 mm in MP) compared to non-atrophic regions (14.16 ± 2.12 mm). In the laser-induced CNV model, CNV area was 0.26 ± 0.09 mm2 and luminal diameters of CNV vessels were 4.7 ± 0.9 mm. Immunostaining on bleached choroids with anti-podocalyxin antibody provides a simple and reliable tool for visualizing normal and pathologic choroidal vasculature in pigmented mouse eyes for quantitative morphometric analysis. This method will be beneficial for examining and evaluating the effects of various treatment modalities on the choroidal vasculature in mouse models of ocular diseases such as age-related macular degeneration, and degenerative genetic diseases.
Collapse
Affiliation(s)
- Imran A Bhutto
- Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - D Scott McLeod
- Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Benjamin R Thomson
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg SOM, Chicago, IL, USA
| | - Gerard A Lutty
- Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Malia M Edwards
- Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
5
|
Saito R, Inagaki A, Nakamura Y, Imura T, Kanai N, Mitsugashira H, Endo Kumata Y, Katano T, Suzuki S, Tokodai K, Kamei T, Unno M, Watanabe K, Tabata Y, Goto M. A Gelatin Hydrogel Nonwoven Fabric Enhances Subcutaneous Islet Engraftment in Rats. Cells 2023; 13:51. [PMID: 38201255 PMCID: PMC10777905 DOI: 10.3390/cells13010051] [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: 11/26/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Although subcutaneous islet transplantation has many advantages, the subcutaneous space is poor in vessels and transplant efficiency is still low in animal models, except in mice. Subcutaneous islet transplantation using a two-step approach has been proposed, in which a favorable cavity is first prepared using various materials, followed by islet transplantation into the preformed cavity. We previously reported the efficacy of pretreatment using gelatin hydrogel nonwoven fabric (GHNF), and the length of the pretreatment period influenced the results in a mouse model. We investigated whether the preimplantation of GHNF could improve the subcutaneous islet transplantation outcomes in a rat model. GHNF sheets sandwiching a silicone spacer (GHNF group) and silicone spacers without GHNF sheets (control group) were implanted into the subcutaneous space of recipients three weeks before islet transplantation, and diabetes was induced seven days before islet transplantation. Syngeneic islets were transplanted into the space where the silicone spacer was removed. Blood glucose levels, glucose tolerance, immunohistochemistry, and neovascularization were evaluated. The GHNF group showed significantly better blood glucose changes than the control group (p < 0.01). The cure rate was significantly higher in the GHNF group (p < 0.05). The number of vWF-positive vessels was significantly higher in the GHNF group (p < 0.01), and lectin angiography showed the same tendency (p < 0.05). The expression of laminin and collagen III around the transplanted islets was also higher in the GHNF group (p < 0.01). GHNF pretreatment was effective in a rat model, and the main mechanisms might be neovascularization and compensation of the extracellular matrices.
Collapse
Affiliation(s)
- Ryusuke Saito
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (R.S.)
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Graduate School of Medicine, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Norifumi Kanai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (R.S.)
| | - Hiroaki Mitsugashira
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (R.S.)
| | - Yukiko Endo Kumata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (R.S.)
| | - Takumi Katano
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Shoki Suzuki
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (R.S.)
| | - Kazuaki Tokodai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (R.S.)
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (R.S.)
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (R.S.)
| | - Kimiko Watanabe
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences (LiMe), Kyoto University, Kyoto 606-8507, Japan
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (R.S.)
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| |
Collapse
|
6
|
Jeong JH, Ojha U, Jang H, Kang S, Lee S, Lee YM. Dual anti-angiogenic and anti-metastatic activity of myriocin synergistically enhances the anti-tumor activity of cisplatin. Cell Oncol (Dordr) 2023; 46:117-132. [PMID: 36329364 DOI: 10.1007/s13402-022-00737-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] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
PURPOSE Tumor microenvironment consists of various kind of cells, forming complex interactions and signal transductions for tumor growth. Due to this complexity, targeting multiple kinases could yield improved clinical outcomes. In this study, we aimed to investigate the potential of myriocin, from Mycelia sterilia, as a novel dual-kinase inhibitor and suggest myriocin as a candidate for combined chemotherapy. METHODS We initially evaluated the anti-tumor and anti-metastatic effect of myriocin in mouse allograft tumor models. We examined the effects of myriocin on angiogenesis and tumor vasculature using in vitro, in vivo, and ex vivo models, and also tested the anti-migration effect of myriocin in in vitro models. Next, we explored the effects of myriocin alone and in combination with cisplatin on tumor growth and vascular normalization in mouse models. RESULTS We found that myriocin inhibited tumor growth and lung metastasis in mouse allograft tumor models. Myriocin induced normalization of the tumor vasculature in the mouse models. We also found that myriocin suppressed angiogenesis through the VEGFR2/PI3K/AKT pathway in endothelial cells (ECs), as well as cancer cell migration by blocking the IκBα/NF-κB(p65)/MMP-9 pathway. Finally, we found that myriocin enhanced the drug delivery efficacy of cisplatin by increasing the integrity of tumor vasculature in the mouse models, which synergistically increased the anti-tumor activity of cisplatin. CONCLUSION We suggest that myriocin is a novel potent anti-cancer agent that dually targets both VEGFR2 in ECs and IκBα in cancer cells, and exerts more pronounced anti-tumor effects than with either kinase being inhibited alone.
Collapse
Affiliation(s)
- Ji-Hak Jeong
- Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
- National Basic Research Lab. of Vascular Homeostasis Regulation, College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
| | - Uttam Ojha
- Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
- National Basic Research Lab. of Vascular Homeostasis Regulation, College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
| | - Hyeonha Jang
- Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
- National Basic Research Lab. of Vascular Homeostasis Regulation, College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
| | - Soohyun Kang
- National Basic Research Lab. of Vascular Homeostasis Regulation, College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
| | - Sunhee Lee
- Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
| | - You Mie Lee
- Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea.
- National Basic Research Lab. of Vascular Homeostasis Regulation, College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea.
- College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea.
| |
Collapse
|
7
|
Bei J, Miranda-Morales EG, Gan Q, Qiu Y, Husseinzadeh S, Liew JY, Chang Q, Krishnan B, Gaitas A, Yuan S, Felicella M, Qiu WQ, Fang X, Gong B. Circulating Exosomes from Alzheimer's Disease Suppress Vascular Endothelial-Cadherin Expression and Induce Barrier Dysfunction in Recipient Brain Microvascular Endothelial Cell. J Alzheimers Dis 2023; 95:869-885. [PMID: 37661885 DOI: 10.3233/jad-230347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
BACKGROUND Blood-brain barrier (BBB) breakdown is a crucial aspect of Alzheimer's disease (AD) progression. Dysfunction in BBB is primarily caused by impaired tight junction and adherens junction proteins in brain microvascular endothelial cells (BMECs). The role of adherens junctions in AD-related BBB dysfunction remains unclear. Exosomes from senescent cells have unique characteristics and contribute to modulating the phenotype of recipient cells. However, it remains unknown if and how these exosomes cause BMEC dysfunction in AD. OBJECTIVE This study aimed to investigate the impact of AD circulating exosomes on brain endothelial dysfunction. METHODS Exosomes were isolated from sera of AD patients and age- and sex-matched cognitively normal controls using size-exclusion chromatography. The study measured the biomechanical nature of BMECs' endothelial barrier, the lateral binding forces between live BMECs. Paracellular expressions of the key adherens junction protein vascular endothelial (VE)-cadherin were visualized in BMEC cultures and a 3D BBB model using human BMECs and pericytes. VE-cadherin signals were also examined in brain tissues from AD patients and normal controls. RESULTS Circulating exosomes from AD patients reduced VE-cadherin expression levels and impaired barrier function in recipient BMECs. Immunostaining analysis demonstrated that AD exosomes damaged VE-cadherin integrity in a 3D microvascular tubule formation model. The study found that AD exosomes weakened BBB integrity depending on their RNA content. Additionally, diminished microvascular VE-cadherin expression was observed in AD brains compared to controls. CONCLUSION These findings highlight the significant role of circulating exosomes from AD patients in damaging adherens junctions of recipient BMECs, dependent on exosomal RNA.
Collapse
Affiliation(s)
- Jiani Bei
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ernesto G Miranda-Morales
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Qini Gan
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Yuan Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sorosh Husseinzadeh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jia Yi Liew
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Qing Chang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Balaji Krishnan
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Angelo Gaitas
- The Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Subo Yuan
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Michelle Felicella
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Wei Qiao Qiu
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Xiang Fang
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
8
|
Yang J, Chang S, Chen IA, Kura S, Rosen GA, Saltiel NA, Huber BR, Varadarajan D, Balbastre Y, Magnain C, Chen SC, Fischl B, McKee AC, Boas DA, Wang H. Volumetric Characterization of Microvasculature in Ex Vivo Human Brain Samples By Serial Sectioning Optical Coherence Tomography. IEEE Trans Biomed Eng 2022; 69:3645-3656. [PMID: 35560084 PMCID: PMC9888394 DOI: 10.1109/tbme.2022.3175072] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Serial sectioning optical coherence tomography (OCT) enables accurate volumetric reconstruction of several cubic centimeters of human brain samples. We aimed to identify anatomical features of the ex vivo human brain, such as intraparenchymal blood vessels and axonal fiber bundles, from the OCT data in 3D, using intrinsic optical contrast. METHODS We developed an automatic processing pipeline to enable characterization of the intraparenchymal microvascular network in human brain samples. RESULTS We demonstrated the automatic extraction of the vessels down to a 20 μm in diameter using a filtering strategy followed by a graphing representation and characterization of the geometrical properties of microvascular network in 3D. We also showed the ability to extend this processing strategy to extract axonal fiber bundles from the volumetric OCT image. CONCLUSION This method provides a viable tool for quantitative characterization of volumetric microvascular network as well as the axonal bundle properties in normal and pathological tissues of the ex vivo human brain.
Collapse
|
9
|
Zaitoun IS, Song YS, Zaitoun HB, Sorenson CM, Sheibani N. Assessment of Choroidal Vasculature and Innate Immune Cells in the Eyes of Albino and Pigmented Mice. Cells 2022; 11:3329. [PMID: 36291198 PMCID: PMC9600292 DOI: 10.3390/cells11203329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/06/2022] [Accepted: 10/17/2022] [Indexed: 12/05/2022] Open
Abstract
The visualization of choroidal vasculature and innate immune cells in the eyes of pigmented mice has been challenging due to the presence of a retinal pigment epithelium (RPE) layer separating the choroid and retina. Here, we established methods for visualizing the choroidal macrophages, mast cells, and vasculature in eyes of albino and pigmented mice using cell type-specific staining. We were able to visualize the choroidal arterial and venous systems. An arterial circle around the optic nerve was found in mice similar to the Zinn-Haller arterial circle that exists in humans and primates. Three different structural patterns of choriocapillaris were observed throughout the whole choroid: honeycomb-like, maze-like, and finger-like patterns. Choroidal mast cells were relatively few but dense around the optic nerve. Mast cell distribution in the middle and periphery was different among strains. Macrophages were found in all layers of the choroid. Thus, utilizing the simple and reliable methods described herein will allow the evaluation of transgenic and preclinical mouse models of ocular diseases that affect the choroid, including age-related macular degeneration (AMD), diabetic choroidopathy, and retinopathy of prematurity. These studies will advance our understanding of the pathophysiology, and molecular and cellular mechanisms that can be targeted therapeutically, in these diseases.
Collapse
Affiliation(s)
- Ismail S. Zaitoun
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
- McPherson Eye Research Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
| | - Yong-Seok Song
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
- McPherson Eye Research Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
| | - Hammam B. Zaitoun
- Faculty of Medicine, Yarmouk University in Irbid, Irbid 21163, Jordan
| | - Christine M. Sorenson
- McPherson Eye Research Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
- McPherson Eye Research Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
10
|
Jeon J, Kim SH, Kong E, Kim SJ, Yang JM, Lee JY, Lee J, Kim YM, Kim P. Establishment of the reproducible branch retinal artery occlusion mouse model and intravital longitudinal imaging of the retinal CX3CR1-GFP+ cells after spontaneous arterial recanalization. Front Med (Lausanne) 2022; 9:897800. [PMID: 35911406 PMCID: PMC9334526 DOI: 10.3389/fmed.2022.897800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
Animal models of retinal artery occlusion (RAO) have been widely used in many studies. However, most of these studies prefer using a central retinal artery occlusion (CRAO) which is a typical global ischemia model of the retina, due to the technical limitation of producing single vessel targeted modeling with real-time imaging. A focal ischemia model, such as branch retinal artery occlusion (BRAO), is also needed for explaining interactions, including the immunological reaction between the ischemic retina and adjacent healthy retina. Accordingly, a relevant model for clinical RAO patients has been demanded to understand the pathophysiology of the RAO disease. Herein, we establish a convenient BRAO mouse model to research the focal reaction of the retina. As a photo-thrombotic agent, Rose bengal was intravenously injected into 7 week-old transgenic mice (CX3CR1-GFP) for making embolism occlusion, which causes pathology similarly to clinical cases. In an optimized condition, a 561 nm laser (13.1 mw) was projected to a targeted vessel to induce photo-thrombosis for 27 s by custom-built retinal confocal microscopy. Compared to previous BRAO models, the procedures of thrombosis generation were naturally and minimal invasively generated with real-time retinal imaging. In addition, by utilizing the self-remission characteristics of Rose bengal thrombus, a reflow of the BRAO with immunological reactions of the CX3CR1-GFP+ inflammatory cells such as the retinal microglia and monocytes was monitored and analyzed. In this models, reperfusion began on day 3 after modeling. Simultaneously, the activation of CX3CR1-GFP+ inflammatory cells, including the increase of activation marker and morphologic change, was confirmed by immunohistochemical (IHC) staining and quantitative real-time PCR. CD86 and Nox2 were prominently expressed on day 3 after the modeling. At day 7, blood flow was almost restored in the large vessels. CX3CR1-GFP+ populations in both superficial and deep layers of the retina also increased around even in the BRAO peri-ischemic area. In summary, this study successfully establishes a reproducible BRAO modeling method with convenient capabilities of easily controllable time points and selection of a specific single vessel. It can be a useful tool to analyze the behavior of inflammatory cell after spontaneous arterial recanalization in BRAO and further investigate the pathophysiology of BRAO.
Collapse
Affiliation(s)
- Jehwi Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Sang-Hoon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Eunji Kong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Soo Jin Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jee Myung Yang
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Dongguk University Ilsan Hospital, Ilsan, South Korea
| | - Joo Yong Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Junyeop Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - You-Me Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Pilhan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- *Correspondence: Pilhan Kim,
| |
Collapse
|
11
|
Huo F, Zhang Y, Li Y, Bu H, Zhang Y, Li W, Guo Y, Wang L, Jia R, Huang T, Zhang W, Li P, Ding L, Yan C. Mannose-targeting Concanavalin A-Epirubicin Conjugate for Targeted Intravesical Chemotherapy of Bladder Cancer. Chem Asian J 2022; 17:e202200342. [PMID: 35713953 DOI: 10.1002/asia.202200342] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/10/2022] [Indexed: 12/26/2022]
Abstract
Intravesical instillation of chemotherapeutic drugs such as epirubicin (EPI) is routinely used to prevent tumor recurrence and progression after transurethral resection of bladder tumor. However, the lack of tumor selectivity often causes severe damage to normal bladder urothelium leading to intolerable side effects. Here, we analyzed abnormal changes in glycosylation in bladder cancer and identified mannose as the most aberrantly expressed glycan on the surface of bladder cancer cell lines and human bladder tumor tissues. We then constructed a lectin-drug conjugate by linking concanavalin A (ConA) - a lectin that specifically binds to mannose, with EPI through a pH-sensitive linker. This ConA-EPI conjugate conferred EPI with mannose-targeting ability and selectively internalized cancer cells in vitro. This conjugate showed selective cytotoxicity to cancer cells in vitro and better antitumor activity in an orthotopic mouse model of bladder cancer. Our lectin-drug conjugation strategy makes targeted intravesical chemotherapy of bladder cancer possible.
Collapse
Affiliation(s)
- Fan Huo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Huagang Bu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Yaliang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Wei Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Yuna Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Lan Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Ru Jia
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Tengfei Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Weiyi Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Pengchao Li
- Department of Urology, the First Affiliated Hospital with Nanjing Medical University, Jiangsu Province Hospital), Nanjing, Jiangsu, 210023, P. R. China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China.,Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China.,Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu, 210023, P. R. China.,Engineering Research Center of Protein and Peptide Medicine, Ministry of Education, P. R. China
| |
Collapse
|
12
|
An CZ, Li CQ, Song LB, He YF, Chen W, Liu B, Zhao YD. A simple fluorescent strategy for liver capillary labeling with carbon quantum dot-lectin nanoprobe. Analyst 2022; 147:1952-1960. [DOI: 10.1039/d1an02364k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Based on lycopersicon esculentum lectin that can target vascular endothelial cells and carbon quantum dots, we designed a carbon-based probe for the fluorescence labeling and imaging of hepatic blood vessels of liver tissue sections.
Collapse
Affiliation(s)
- Chang-Zhi An
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China
| | - Chao-Qing Li
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China
| | - Lai-Bo Song
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China
| | - Yan-Fei He
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China
| | - Wei Chen
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China
| | - Bo Liu
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China
| | - Yuan-Di Zhao
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China
- Key Laboratory of Biomedical Photonics (HUST), Ministry of Education, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China
| |
Collapse
|
13
|
Battistella R, Kritsilis M, Matuskova H, Haswell D, Cheng AX, Meissner A, Nedergaard M, Lundgaard I. Not All Lectins Are Equally Suitable for Labeling Rodent Vasculature. Int J Mol Sci 2021; 22:ijms222111554. [PMID: 34768985 PMCID: PMC8584019 DOI: 10.3390/ijms222111554] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 11/16/2022] Open
Abstract
The vascular system is vital for all tissues and the interest in its visualization spans many fields. A number of different plant-derived lectins are used for detection of vasculature; however, studies performing direct comparison of the labeling efficacy of different lectins and techniques are lacking. In this study, we compared the labeling efficacy of three lectins: Griffonia simplicifolia isolectin B4 (IB4); wheat germ agglutinin (WGA), and Lycopersicon esculentum agglutinin (LEA). The LEA lectin was identified as being far superior to the IB4 and WGA lectins in histological labeling of blood vessels in brain sections. A similar signal-to-noise ratio was achieved with high concentrations of the WGA lectin injected during intracardial perfusion. Lectins were also suitable for labeling vasculature in other tissues, including spinal cord, dura mater, heart, skeletal muscle, kidney, and liver tissues. In uninjured tissues, the LEA lectin was as accurate as the Tie2–eGFP reporter mice and GLUT-1 immunohistochemistry for labeling the cerebral vasculature, validating its specificity and sensitivity. However, in pathological situations, e.g., in stroke, the sensitivity of the LEA lectin decreases dramatically, limiting its applicability in such studies. This work can be used for selecting the type of lectin and labeling method for various tissues.
Collapse
Affiliation(s)
- Roberta Battistella
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22362 Lund, Sweden; (R.B.); (M.K.); (H.M.); (A.M.)
- WCMM Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, 22362 Lund, Sweden
| | - Marios Kritsilis
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22362 Lund, Sweden; (R.B.); (M.K.); (H.M.); (A.M.)
- WCMM Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, 22362 Lund, Sweden
| | - Hana Matuskova
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22362 Lund, Sweden; (R.B.); (M.K.); (H.M.); (A.M.)
- WCMM Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, 22362 Lund, Sweden
- German Center for Neurodegenerative Diseases, 53127 Bonn, Germany
- Department of Neurology, Division of Vascular Neurology, University Hospital Bonn, 53127 Bonn, Germany
| | - Douglas Haswell
- Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (D.H.); (A.X.C.); (M.N.)
| | - Anne Xiaoan Cheng
- Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (D.H.); (A.X.C.); (M.N.)
| | - Anja Meissner
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22362 Lund, Sweden; (R.B.); (M.K.); (H.M.); (A.M.)
- WCMM Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, 22362 Lund, Sweden
- German Center for Neurodegenerative Diseases, 53127 Bonn, Germany
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (D.H.); (A.X.C.); (M.N.)
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Iben Lundgaard
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22362 Lund, Sweden; (R.B.); (M.K.); (H.M.); (A.M.)
- WCMM Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, 22362 Lund, Sweden
- Correspondence:
| |
Collapse
|
14
|
Zhang N, Zhang X, Girardot PE, Chrenek MA, Sellers JT, Li Y, Kim YK, Summers VR, Ferdous S, Shelton DA, Boatright JH, Nickerson JM. Electrophysiologic and Morphologic Strain Differences in a Low-Dose NaIO3-Induced Retinal Pigment Epithelium Damage Model. Transl Vis Sci Technol 2021; 10:10. [PMID: 34251426 PMCID: PMC8287050 DOI: 10.1167/tvst.10.8.10] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose We aimed to explore differences in the NaIO3-elicited responses of retinal pigment epithelium (RPE) and other retinal cells associated with mouse strains and dosing regimens. Methods One dose of NaIO3 at 10 or 15 mg/kg was given intravenously to adult male C57BL/6J and 129/SV-E mice. Control animals were injected with PBS. Morphologic and functional changes were characterized by spectral domain optical coherence tomography, electroretinography, histologic, and immunofluorescence techniques. Results Injection with 10 mg/kg of NaIO3 did not cause consistent RPE or retinal changes in either strain. Administration of 15 mg/kg of NaIO3 initially induced a large transient increase in scotopic electroretinography a-, b-, and c-wave amplitudes within 12 hours of injection, followed by progressive structural and functional degradation at 3 days after injection in C57BL/6J mice and at 1 week after injection in 129/SV-E mice. RPE cell loss occurred in a large posterior-central lesion with a ring-like transition zone of abnormally shaped cells starting 12 hours after NaIO3 treatment. Conclusions NaIO3 effects depended on the timing, dosage, and mouse strain. The RPE in the periphery was spared from damage compared with the central RPE. The large transient increase in the electroretinography was remarkable. Translational Relevance This study is a phase T1 translational research study focusing on the development and validation of a mouse model of RPE damage. It provides a detailed foundation for future research, informing choices of mouse strain, dosage, and time points to establish NaIO3-induced RPE damage.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA.,Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xian Zhang
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA.,Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Preston E Girardot
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Micah A Chrenek
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Jana T Sellers
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Ying Li
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Yong-Kyu Kim
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA.,Department of Ophthalmology, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul, South Korea
| | - Vivian R Summers
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Salma Ferdous
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Debresha A Shelton
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Jeffrey H Boatright
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA.,Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur, GA, USA
| | - John M Nickerson
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|