1
|
Williams AL, Bohnsack BL. Keratin 8/18a.1 Expression Influences Embryonic Neural Crest Cell Dynamics and Contributes to Postnatal Corneal Regeneration in Zebrafish. Cells 2024; 13:1473. [PMID: 39273043 PMCID: PMC11394277 DOI: 10.3390/cells13171473] [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: 12/26/2023] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
A complete understanding of neural crest cell mechanodynamics during ocular development will provide insight into postnatal neural crest cell contributions to ophthalmic abnormalities in adult tissues and inform regenerative strategies toward injury repair. Herein, single-cell RNA sequencing in zebrafish during early eye development revealed keratin intermediate filament genes krt8 and krt18a.1 as additional factors expressed during anterior segment development. In situ hybridization and immunofluorescence microscopy confirmed krt8 and krt18a.1 expression in the early neural plate border and migrating cranial neural crest cells. Morpholino oligonucleotide (MO)-mediated knockdown of K8 and K18a.1 markedly disrupted the migration of neural crest cell subpopulations and decreased neural crest cell marker gene expression in the craniofacial region and eye at 48 h postfertilization (hpf), resulting in severe phenotypic defects reminiscent of neurocristopathies. Interestingly, the expression of K18a.1, but not K8, is regulated by retinoic acid (RA) during early-stage development. Further, both keratin proteins were detected during postnatal corneal regeneration in adult zebrafish. Altogether, we demonstrated that both K8 and K18a.1 contribute to the early development and postnatal repair of neural crest cell-derived ocular tissues.
Collapse
Affiliation(s)
- Antionette L. Williams
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave., Chicago, IL 60611, USA;
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, 645 N. Michigan Ave., Chicago, IL 60611, USA
| | - Brenda L. Bohnsack
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave., Chicago, IL 60611, USA;
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, 645 N. Michigan Ave., Chicago, IL 60611, USA
| |
Collapse
|
2
|
Sumioka T, Matsumoto KI, Reinach PS, Saika S. Tenascins and osteopontin in biological response in cornea. Ocul Surf 2023; 29:131-149. [PMID: 37209968 DOI: 10.1016/j.jtos.2023.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/01/2023] [Accepted: 05/16/2023] [Indexed: 05/22/2023]
Abstract
The structural composition, integrity and regular curvature of the cornea contribute to the maintenance of its transparency and vision. Disruption of its integrity caused by injury results in scarring, inflammation and neovascularization followed by losses in transparency. These sight compromising effects is caused by dysfunctional corneal resident cell responses induced by the wound healing process. Upregulation of growth factors/cytokines and neuropeptides affect development of aberrant behavior. These factors trigger keratocytes to first transform into activated fibroblasts and then to myofibroblasts. Myofibroblasts express extracellular matrix components for tissue repair and contract the tissue to facilitate wound closure. Proper remodeling following primary repair is critical for restoration of transparency and visual function. Extracellular matrix components contributing to the healing process are divided into two groups; a group of classical tissue structural components and matrix macromolecules that modulate cell behaviors/activities besides being integrated into the matrix structure. The latter components are designated as matricellular proteins. Their functionality is elicited through mechanisms which modulate the scaffold integrity, cell behaviors, activation/inactivation of either growth factors or cytoplasmic signaling regulation. We discuss here the functional roles of matricellular proteins in mediating injury-induced corneal tissue repair. The roles are described of major matricellular proteins, which include tenascin C, tenascin X and osteopontin. Focus is directed towards dealing with their roles in modulating individual activities of wound healing-related growth factors, e. g., transforming growth factor β (TGF β). Modulation of matricellular protein functions could encompass a potential novel strategy to improve the outcome of injury-induced corneal wound healing.
Collapse
Affiliation(s)
- Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, 641-0012, Japan.
| | - Ken-Ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Head Office for Research and Academic Information, Shimane University, 89-1 Enya-cho, Izumo, 693-8501, Japan
| | - Peter Sol Reinach
- Department of Biological. Sciences SUNY Optometry, New York, NY, 10036, USA
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, 641-0012, Japan
| |
Collapse
|
3
|
Surovtseva MA, Kim II, Bondarenko NA, Lykov AP, Krasner KY, Chepeleva EV, Bgatova NP, Trunov AN, Chernykh VV, Poveshchenko OV. Derivation of Human Corneal Keratocytes from ReLEx SMILE Lenticules for Cell Therapy and Tissue Engineering. Int J Mol Sci 2023; 24:ijms24108828. [PMID: 37240176 DOI: 10.3390/ijms24108828] [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: 04/05/2023] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Fibroblasts isolated and expanded from ReLEx SMILE lenticules can be a source of human keratocytes. Since corneal keratocytes are quiescent cells, it is difficult to expand them in vitro in suitable numbers for clinical and experimental use. In the present study, this problem was solved by isolating and growing corneal fibroblasts (CFs) with a high proliferative potential and their reversion to keratocytes in a selective serum-free medium. Fibroblasts reversed into keratocytes (rCFs) had a dendritic morphology and ultrastructural signs of activation of protein synthesis and metabolism. The cultivation of CFs in a medium with 10% FCS and their reversion into keratocytes was not accompanied by the induction of myofibroblasts. After reversion, the cells spontaneously formed spheroids and expressed keratocan and lumican markers, but not mesenchymal ones. The rCFs had low proliferative and migratory activity, and their conditioned medium contained a low level of VEGF. CF reversion was not accompanied by a change with the levels of IGF-1, TNF-alpha, SDF-1a, and sICAM-1. In the present study, it has been demonstrated that fibroblasts from ReLEx SMILE lenticules reverse into keratocytes in serum-free KGM, maintaining the morphology and functional properties of primary keratocytes. These keratocytes have a potential for tissue engineering and cell therapy of various corneal pathologies.
Collapse
Affiliation(s)
- Maria A Surovtseva
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 2 Timakova Str., 630060 Novosibirsk, Russia
| | - Irina I Kim
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 2 Timakova Str., 630060 Novosibirsk, Russia
| | - Natalia A Bondarenko
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 2 Timakova Str., 630060 Novosibirsk, Russia
| | - Alexander P Lykov
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 2 Timakova Str., 630060 Novosibirsk, Russia
| | - Kristina Yu Krasner
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 2 Timakova Str., 630060 Novosibirsk, Russia
- Novosibirsk Branch of S. Fedorov Eye Microsurgery Federal State Institution, 10 Kalkhidskaya Str., 630096 Novosibirsk, Russia
| | - Elena V Chepeleva
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 2 Timakova Str., 630060 Novosibirsk, Russia
| | - Nataliya P Bgatova
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 2 Timakova Str., 630060 Novosibirsk, Russia
| | - Alexander N Trunov
- Novosibirsk Branch of S. Fedorov Eye Microsurgery Federal State Institution, 10 Kalkhidskaya Str., 630096 Novosibirsk, Russia
| | - Valery V Chernykh
- Novosibirsk Branch of S. Fedorov Eye Microsurgery Federal State Institution, 10 Kalkhidskaya Str., 630096 Novosibirsk, Russia
| | - Olga V Poveshchenko
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 2 Timakova Str., 630060 Novosibirsk, Russia
| |
Collapse
|
4
|
Bai Y, Jiao X, Hu J, Xue W, Zhou Z, Wang W. WTAP promotes macrophage recruitment and increases VEGF secretion via N6-methyladenosine modification in corneal neovascularization. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166708. [PMID: 37019244 DOI: 10.1016/j.bbadis.2023.166708] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/03/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Corneal neovascularization (CNV) can be caused by chemical burns. Macrophages are involved in angiogenesis and lymphangiogenesis during CNV. The aim of this study was to investigate whether Wilms' tumor 1-associated protein (WTAP) is involved in macrophage recruitment and VEGF secretion via N6-methyladenosine (m6A) modification. METHODS A CNV mouse model was established by corneal alkali burn. Tumor necrosis factor alpha (TNF-α) was used to stimulate vascular endothelial cells. m6A immunoprecipitation qPCR was used to determine the enrichment of m6A levels in mRNAs. The H3K9me3 enrichment in the promoter region of CC motif chemokine ligand 2 (CCL2) was detected by chromatin immunoprecipitation assay. The WTAP inhibition in vivo was performed using the adeno-associated virus. RESULTS In the alkali burn corneal tissues, angiogenesis and lymphangiogenesis were promoted as CD31 and LYVE-1 expressions were elevated, and the number of macrophages as well as WTAP expression were increased. Under the TNF-α stimulation, WTAP promoted the recruitment of endothelial cells to macrophages by promoting CCL2 secretion. Mechanistically, WTAP affected the enrichment of H3K9me3 at the CCL2 promoter by regulating the m6A level of SUV39H1 mRNA. The in vivo experiment showed that VEGFA/C/D secretion of macrophages was reduced after WTAP interference. Mechanistically, WTAP regulated the translational efficiency of HIF-1α via m6A modification. CONCLUSION WTAP affected macrophage recruitment to endothelial cells via regulation of H3K9me3-mediated CCL2 transcription. WTAP also affected macrophage secretion of VEGFA/C/D via m6A-mediated translation regulation of HIF-1α. Both pathways were involved in the WTAP regulation of angiogenesis and lymphangiogenesis during CNV.
Collapse
|
5
|
Iwanishi H, Yamanaka O, Sumioka T, Yasuda S, Miyajima M, Saika S. Delayed regression of laser-induced choroidal neovascularization in TNFα-null mice. J Cell Mol Med 2022; 26:5315-5325. [PMID: 36127870 PMCID: PMC9575074 DOI: 10.1111/jcmm.17562] [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: 05/14/2020] [Revised: 06/30/2022] [Accepted: 09/07/2022] [Indexed: 11/28/2022] Open
Abstract
We investigated the effects of lacking TNFα on the development and regression of Argon-laser-induced choroidal neovascularization (CNV) in mice. We lasered ocular fundus for induction of CNV in both wild-type (WT) and TNFα-null (KO) mice. Fluorescence angiography was performed to examine the size of CNV lesions. Gene expression pattern of wound healing-related components was examined. The effects of exogenous TNFα on apoptosis of human retinal microvascular endothelial cells (HRMECs) and on the tube-like structure of the cells were investigated in vitro. The results showed that Argon-laser irradiation-induced CNV was significantly larger in KO mice than WT mice on Day 21, but not at other timepoints. Lacking TNFα increased neutrophil population in the lesion. The distribution of cleaved caspase3-labelled apoptotic cells was more frequently observed in the laser-irradiated tissue in a WT mouse as compared with a KO mouse. Exogenous TNFα induced apoptosis of HRMECs and accelerated regression of tube-like structure of HRMECs in cell culture. Taken together, TNFα gene knockout delays the regression of laser-induced CNV in mice. The mechanism underlying the phenotype might include the augmentation of neutrophil population in the treated tissue and attenuation of vascular endothelial cell apoptosis.
Collapse
Affiliation(s)
- Hiroki Iwanishi
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Osamu Yamanaka
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Shingo Yasuda
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Masayasu Miyajima
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
6
|
Lack of transient receptor potential ankyrin 1 (TRPA1) retards cutaneous wound healing in mice: A preliminary study. Biochem Biophys Rep 2022; 31:101322. [PMID: 35990578 PMCID: PMC9382432 DOI: 10.1016/j.bbrep.2022.101322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/21/2022] Open
Abstract
Wound healing is an important process in various diseases, and elucidating the underlying mechanism is essential for developing therapeutic strategies. We investigated whether the loss of transient receptor potential ankyrin 1 (TRPA1) affects the cutaneous wound healing process in mice. We assessed the formation of granulation tissue by myofibroblasts and macrophages, re-epithelialization, and related gene expression. TRPA1-null (KO) and wild-type (WT) C57BL/6 mice were used for establishing the wound model. Two round full-thickness excision wounds (diameter, 5.0 mm) were produced in the dorsal skin of mice under general anesthesia. After specific intervals, healing was evaluated using macroscopic observation, histology, immunohistochemistry, and real-time reverse transcription-polymerase chain reaction (RT-PCR). TRPA1 KO retarded the formation of granulation tissue and re-epithelialization in the healing of cutaneous wound. Furthermore, TRPA1 KO suppressed the appearance of myofibroblasts, macrophage infiltration, and mRNA expression of αSMA, F4/80, and Col-1α2. These findings indicate that TRPA1 is required for cutaneous wound healing in mice. The lack of TRPA1 retards macrophage infiltration and the subsequent fibrotic tissue formation, which might further impair the fibrogenic behavior of fibroblasts. Transient receptor potential ankyrin 1 (TRPA1) is essential for wound healing. Early cutaneous wound healing was delayed in TRPA-1-deficient mice. Loss of TRPA1 retards formation of new granulation tissue in the wound. Lack of TRPA1 inhibits macrophage infiltration and myofibroblast formation. TRPA1 deficiency inhibits expression of TGF-β1, αSMA, F4/80, and collagen 1α2 mRNA.
Collapse
|
7
|
Understanding Drivers of Ocular Fibrosis: Current and Future Therapeutic Perspectives. Int J Mol Sci 2021; 22:ijms222111748. [PMID: 34769176 PMCID: PMC8584003 DOI: 10.3390/ijms222111748] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 01/10/2023] Open
Abstract
Ocular fibrosis leads to severe visual impairment and blindness worldwide, being a major area of unmet need in ophthalmology and medicine. To date, the only available treatments are antimetabolite drugs that have significant potentially blinding side effects, such as tissue damage and infection. There is thus an urgent need to identify novel targets to prevent/treat scarring and postsurgical fibrosis in the eye. In this review, the latest progress in biological mechanisms underlying ocular fibrosis are discussed. We also summarize the current knowledge on preclinical studies based on viral and non-viral gene therapy, as well as chemical inhibitors, for targeting TGFβ or downstream effectors in fibrotic disorders of the eye. Moreover, the role of angiogenetic and biomechanical factors in ocular fibrosis is discussed, focusing on related preclinical treatment approaches. Moreover, we describe available evidence on clinical studies investigating the use of therapies targeting TGFβ-dependent pathways, angiogenetic factors, and biomechanical factors, alone or in combination with other strategies, in ocular tissue fibrosis. Finally, the recent progress in cell-based therapies for treating fibrotic eye disorders is discussed. The increasing knowledge of these disorders in the eye and the promising results from testing of novel targeted therapies could offer viable perspectives for translation into clinical use.
Collapse
|
8
|
Sumioka T, Iwanishi H, Okada Y, Miyajima M, Ichikawa K, Reinach PS, Matsumoto KI, Saika S. Impairment of corneal epithelial wound healing is association with increased neutrophil infiltration and reactive oxygen species activation in tenascin X-deficient mice. J Transl Med 2021; 101:690-700. [PMID: 33782532 PMCID: PMC8137452 DOI: 10.1038/s41374-021-00576-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 01/14/2023] Open
Abstract
The purpose of the study was to uncover the role of tenascin X in modulation of healing in mouse corneas subjected to epithelium debridement. Healing in corneas with an epithelial defect was evaluated at the levels of gene and protein expression. Wound healing-related mediators and inflammatory cell infiltration were detected by histology, immunohistochemistry and real-time RT-PCR. Tenascin X protein was upregulated in the wounded wild-type (WT) corneal epithelium. The lack of tenascin X impaired closure of an epithelial defect and accelerated infiltration of neutrophils into the wound periphery as compared to the response in WT tissue. Expression of wound healing-related proinflammatory and reparative components, i.e., interleukin-6, transforming growth factor β, matrix metalloproteinases, were unaffected by the loss of tenascin X expression. Marked accumulation of malondialdehyde (a lipid peroxidation-derived product) was observed in KO healing epithelia as compared with its WT counterpart. Neutropenia induced by systemic administration of a specific antibody rescued the impairment of epithelial healing in KO corneas, with reduction of malondialdehyde levels in the epithelial cells. Finally, we showed that a chemical scavenging reactive oxygen species reversed the impairment of attenuation of epithelial repair with a reduction of tissue levels of malondialdehyde. In conclusion, loss of tenascin X prolonged corneal epithelial wound healing and increased neutrophilic inflammatory response to debridement in mice. Tenascin X contributes to the control of neutrophil infiltration needed to support the regenerative response to injury and prevent the oxidative stress mediators from rising to cytotoxic levels.
Collapse
Affiliation(s)
- Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan.
| | - Hiroki Iwanishi
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Masayasu Miyajima
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Kana Ichikawa
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Peter S Reinach
- Department of Ophthalmology, Wenzhou Medical University, Wenzhou, P. R. China
| | - Ken-Ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Enya-cho, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| |
Collapse
|
9
|
Maier AKB, Reichhart N, Gonnermann J, Kociok N, Riechardt AI, Gundlach E, Strauß O, Joussen AM. Effects of TNFα receptor TNF-Rp55- or TNF-Rp75- deficiency on corneal neovascularization and lymphangiogenesis in the mouse. PLoS One 2021; 16:e0245143. [PMID: 33835999 PMCID: PMC8034740 DOI: 10.1371/journal.pone.0245143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/22/2020] [Indexed: 02/02/2023] Open
Abstract
Tumor necrosis factor (TNF)α is an inflammatory cytokine likely to be involved in the process of corneal inflammation and neovascularization. In the present study we evaluate the role of the two receptors, TNF-receptor (TNF-R)p55 and TNF-Rp75, in the mouse model of suture-induced corneal neovascularization and lymphangiogenesis. Corneal neovascularization and lymphangiogenesis were induced by three 11-0 intrastromal corneal sutures in wild-type (WT) C57BL/6J mice and TNF-Rp55-deficient (TNF-Rp55d) and TNF-Rp75-deficient (TNF-Rp75d) mice. The mRNA expression of VEGF-A, VEGF-C, Lyve-1 and TNFα and its receptors was quantified by qPCR. The area covered with blood- or lymphatic vessels, respectively, was analyzed by immunohistochemistry of corneal flatmounts. Expression and localization of TNFα and its receptors was assessed by immunohistochemistry of sagittal sections and Western Blot. Both receptors are expressed in the murine cornea and are not differentially regulated by the genetic alteration. Both TNF-Rp55d and TNF-Rp75d mice showed a decrease in vascularized area compared to wild-type mice 14 days after suture treatment. After 21 days there were no differences detectable between the groups. The number of VEGF-A-expressing macrophages did not differ when comparing WT to TNF-Rp55d and TNF-Rp75d. The mRNA expression of lymphangiogenic markers VEGF-C or LYVE-1 does not increase after suture in all 3 groups and lymphangiogenesis showed a delayed effect only for TNF-Rp75d. TNFα mRNA and protein expression increased after suture treatment but showed no difference between the three groups. In the suture-induced mouse model, TNFα and its ligands TNF-Rp55 and TNF-Rp75 do not play a significant role in the pathogenesis of neovascularisation and lymphangiogenesis.
Collapse
MESH Headings
- Animals
- Cornea/metabolism
- Cornea/pathology
- Corneal Neovascularization/genetics
- Corneal Neovascularization/pathology
- Gene Deletion
- Humans
- Lymphangiogenesis
- Mice, Inbred C57BL
- RNA, Messenger/genetics
- Receptors, Tumor Necrosis Factor, Type I/analysis
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type II/analysis
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Mice
Collapse
Affiliation(s)
- Anna-Karina B. Maier
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Nadine Reichhart
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Johannes Gonnermann
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Norbert Kociok
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Aline I. Riechardt
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Enken Gundlach
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Olaf Strauß
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Antonia M. Joussen
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
10
|
Yasuda S, Sumioka T, Iwanishi H, Okada Y, Miyajima M, Ichikawa K, Reinach PS, Saika S. Loss of sphingosine 1-phosphate receptor 3 gene function impairs injury-induced stromal angiogenesis in mouse cornea. J Transl Med 2021; 101:245-257. [PMID: 33199821 PMCID: PMC7815507 DOI: 10.1038/s41374-020-00505-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/11/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid generated through sphingosine kinase1 (SPK1)-mediated phosphorylation of sphingosine. We show here that injury-induced S1P upregulation increases corneal neovascularization through stimulating S1PR3, a cognate receptor. since this response was suppressed in S1PR3-knockout mice. Furthermore, Cayman10444, a selective S1PR3 inhibitor, reduced this response in WT mice. Such reductions in neovascularization were associated with reduced vascular endothelial growth factor A (VEGF-A) mRNA expression levels in WT TKE2 corneal epithelial cells and macrophages treated with CAY10444 as well as macrophages isolated from S1PR3 KO mice. S1P increased tube-like vessel formation in human vascular endothelial cells (HUVEC) and human retinal microvascular endothelial cells (HRMECs) cells expressing S1PR3. In S1PR3 KO mice, TGFβ1-induced increases in αSMA gene expression levels were suppressed relative to those in the WT counterparts. In S1PR3 deficient macrophages, VEGF-A expression levels were lower than in WT macrophages. Transforming growth factor β1(TGFβ1) upregulated SPK1 expression levels in ocular fibroblasts and TKE2 corneal epithelial cells. CAY10444 blocked S1P-induced increases in VEGF-A mRNA expression levels in TKE2 corneal epithelial cells. Endogenous S1P signaling upregulated VEGF-A and VE-cadherin mRNA expression levels in HUVEC. Unlike in TKE2 cells, SIS3 failed to block TGFβ1-induced VEGF-A upregulation in ocular fibroblasts. Taken together, these results indicate that injury-induced TGFβ1 upregulation increases S1P generation through increases in SPK1 activity. The rise in S1P formation stimulates the S1PR3-linked signaling pathway, which in turn increases VEGF-A expression levels and angiogenesis in mouse corneas.
Collapse
Affiliation(s)
- Shingo Yasuda
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, 641-0012, Japan.
| | - Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Hiroki Iwanishi
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Masayasu Miyajima
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Kana Ichikawa
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Peter S Reinach
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| |
Collapse
|
11
|
Zhang N, Luo X, Zhang S, Liu R, Liang L, Su W, Liang D. Subconjunctival injection of tumor necrosis factor-α pre-stimulated bone marrow-derived mesenchymal stem cells enhances anti-inflammation and anti-fibrosis in ocular alkali burns. Graefes Arch Clin Exp Ophthalmol 2020; 259:929-940. [PMID: 33237391 DOI: 10.1007/s00417-020-05017-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 11/02/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
PURPOSE To investigate the therapeutic effect of subconjunctival injection of tumor necrosis factor-α (TNF-α) pre-stimulated bone marrow-derived mesenchymal stem cells (BMMSCs) on ocular alkali burns in a rat model. METHODS After applying a 6 mm filter paper soaking in 1 N NaOH on the cornea of rats, the suspension of TNF-α pre-stimulated BMMSCs, BMMSCs and PBS were given subconjunctivally and respectively. Corneal epithelial defect, corneal opacity, inflammation as well as PTGS2 and TSG-6 expression on day 7 and fibrosis on day 14 were compared. RESULTS TNF-α pre-stimulated BMMSCs group had a more predominate effect on promoting corneal epithelial repairing, decreasing corneal opacity, reducing inflammatory cells and CD68 + macrophages on day 7 and suppressing fibrosis on day 14 compared to BMMSCs group. Besides, it had significant increased expressions of PTGS2 and TSG-6 in vitro. Pre-treated with Indomethacin revealed a reverse effect on above-mentioned changes. CONCLUSION Subconjunctival injection of TNF-α pre-stimulated BMMSCs enhanced anti-inflammatory and anti-fibrotic effect in ocular alkali burns, which was possibly though up regulation of PTGS2 and TSG-6 expression.
Collapse
Affiliation(s)
- Nuan Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xiaohui Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Shiyao Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Ren Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Lingyi Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Dan Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
12
|
Kao WWY. Keratin expression by corneal and limbal stem cells during development. Exp Eye Res 2020; 200:108206. [PMID: 32882212 DOI: 10.1016/j.exer.2020.108206] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
Keratins are the forming units of intermediate filaments (IF) that provide mechanical support, and formation of desmosomes between cells and hemi desmosomes with basement membranes for epithelium integrity. Keratin IF are polymers of obligate heterodimer consisting one type I keratin and one type II keratin molecules. There are 54 functional keratin genes in human genome, which are classified into three major groups, i.e., epithelial keratins, hair follicle cell-specific epithelial keratins and hair keratins. Their expression is cell type-specific and developmentally regulated. Corneal epithelium expresses a subgroup of keratins similar to those of epidermal epithelium. Limbal basal stem cells express K5/K14, and K8/K18 and K8/K19 IF suggesting that there probably are two populations of limbal stem cells (LSCs). In human, LSCs at limbal basal layer can directly stratify and differentiate to limbal suprabasal cells that express K3/K12 IF, or centripetally migrate then differentiate to corneal basal transient amplifying cells (TAC) that co-express both K3/K12 and K5/K14 prior to moving upward and assuming suprabasal cells phenotype of only K3/K12 expression that signifies corneal type epithelium differentiation. In rodent, the differentiated cornea epithelial cells express K5/K12 in lieu of K3/K12, because K3 allele exists as a pseudogene and does not encode a functional K3 protein. The basal corneal cells of new-born mice originate from surface ectoderm during embryonic development slowly commit to differentiation of becoming TAC co-expressing K5/K12 and K5/K14 IF. However, the centripetal migration may still occur at a slower rate in young mice, which is accelerated during wound healing. In this review, we will discuss and compare the cornea-specific keratins expression patterns between corneal and epidermal epithelial cells during mouse development, and between human and mouse during development and homeostasis in adult, and pathology caused by a mutation of keratins.
Collapse
Affiliation(s)
- Winston W-Y Kao
- Departments of Ophthalmology, University of Cincinnati, Cincinnati, OH, 45267-0838, USA.
| |
Collapse
|
13
|
Pratsinis H, Mavrogonatou E, Kletsas D. Scarless wound healing: From development to senescence. Adv Drug Deliv Rev 2019; 146:325-343. [PMID: 29654790 DOI: 10.1016/j.addr.2018.04.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/29/2018] [Accepted: 04/09/2018] [Indexed: 12/21/2022]
Abstract
An essential element of tissue homeostasis is the response to injuries, cutaneous wound healing being the most studied example. In the adults, wound healing aims at quickly restoring the barrier function of the skin, leading however to scar, a dysfunctional fibrotic tissue. On the other hand, in fetuses a scarless tissue regeneration takes place. During ageing, the wound healing capacity declines; however, in the absence of comorbidities a higher quality in tissue repair is observed. Senescent cells have been found to accumulate in chronic unhealed wounds, but more recent reports indicate that their transient presence may be beneficial for tissue repair. In this review data on skin wound healing and scarring are presented, covering the whole spectrum from early embryonic development to adulthood, and furthermore until ageing of the organism.
Collapse
|
14
|
Yu H, Sun L, Cui J, Li Y, Yan Y, Wei X, Wang C, Song F, Jiang W, Liu Y, Ge H, Qian H, Li X, Tang X, Liu P. Three kinds of corneal host cells contribute differently to corneal neovascularization. EBioMedicine 2019; 44:542-553. [PMID: 31126890 PMCID: PMC6604366 DOI: 10.1016/j.ebiom.2019.05.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/25/2019] [Accepted: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
Background Corneal neovascularization (angiogenesis and lymphangiogenesis) compromises corneal transparency and transplant survival, however, the molecular mechanisms of corneal host epithelial and stromal cells in neovascularization have not yet been fully elucidated. Furthermore, the contribution and mechanism of corneal host endothelial cells involved in neovascularization are largely unexplored. Methods Liquid chromatography-mass spectrometry, immunoblotting, and ELISA were used to screen and identify potential neovascularization-related factors in human full-thickness vascularized corneal tissues. Lipopolysaccharide was used to induce inflammation in three kinds of corneal host cells in vitro, including corneal epithelial, stromal, and endothelial cells. Fungus was used to establish an animal model of corneal neovascularization in vivo. Tube formation and spheroid sprouting assays were used to evaluate the contribution of three kinds of corneal host cells to the degree of neovascularization under various stimuli. Matrix metalloproteinase (MMP)-2, alpha-crystallin A chain (CRYAA), galectin-8, Bcl-2, neuropilin-2, MMP-9 plasmids, and recombinant human fibronectin were used to identify the key proteins of corneal host cells involved in corneal inflammatory neovascularization. Findings All three kinds of corneal host cells influenced corneal neovascularization to varying degrees. MMP-9 in human corneal epithelial cells, MMP-2, and CRYAA in human corneal stromal cells, and MMP-2 and galectin-8 in human corneal endothelial cells are potential key proteins that participate in corneal inflammatory neovascularization. Interpretation Our data indicated that both the effects of key proteins and corneal host cells involved should be considered for the treatment of corneal inflammatory neovascularization.
Collapse
Affiliation(s)
- Haiyang Yu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University and Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150081, China
| | - Liyao Sun
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University and Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150081, China
| | - Jing Cui
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China
| | - Yan Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Yu Yan
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University and Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150081, China
| | - Xi Wei
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University and Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150081, China
| | - Chao Wang
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University and Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150081, China
| | - Fanqian Song
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University and Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150081, China
| | - Wentong Jiang
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China
| | - Yifan Liu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China
| | - Hongyan Ge
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China
| | - Hua Qian
- Department of Pharmacology, College of Pharmacy, Harbin Medical University and Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150081, China
| | - Xiaoguang Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University and Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin 150081, China
| | - Xianling Tang
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China.
| | - Ping Liu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, 23 Youzheng Street, Harbin 150001, China.
| |
Collapse
|
15
|
Usui-Kusumoto K, Iwanishi H, Ichikawa K, Okada Y, Sumioka T, Miyajima M, Liu CY, Reinach PS, Saika S. Suppression of neovascularization in corneal stroma in a TRPA1-null mouse. Exp Eye Res 2019; 181:90-97. [PMID: 30633924 DOI: 10.1016/j.exer.2019.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 12/19/2018] [Accepted: 01/02/2019] [Indexed: 12/16/2022]
Abstract
Corneal neovascularization and inflammatory fibrosis induced by severe injury or infection leads to tissue opacification and even blindness. Transient receptor potential (TRP) channel subtypes contribute to mediating these maladaptive responses through their interactions with other receptors. TRPV1 is one of the contributing channel isoforms inducing neovascularization in an alkali burn mouse wound healing model. VEGF-A upregulation contributes to neovascularization through interaction with its cognate receptors (VEGFR). Since the TRP isoform in this tissue, TRPA1, is also involved, we determined here if one of the pathways mediating neovascularization and immune cell infiltration involve an interaction between VEGFR and TRPA1 in a cauterization corneal mouse wound healing model. Localization of TRPA1 and endothelial cell (EC) CD31 immunostaining pattern intensity determined if TRPA1 expression was EC delimited during cauterization induced angiogenesis. Quantitative RT-PCR evaluated the effects of the absence of TRPA1 function on VEGF-A and TGF-β1 mRNA expression during this process. Macrophage infiltration increased based on rises in F4/80 antigen immunoreactivity. TRPA1 immunostaining was absent on CD31-immunostained EC cells undergoing neovascularization, but it was present on other cell type(s) adhering to EC in vivo. Absence of TRPA1 expression suppressed both stromal neovascularization and inhibited macrophage infiltration. Similarly, the increases occurring in both VEGF-A and TGF-β1 mRNA expression levels in WT tissue were blunted in the TRPA1-/- counterpart. On the other hand, in the macrophages their levels were invariant and their infiltration was inhibited. To determine if promotion by TRPA1 of angiogenesis was dependent on its expression on other unidentified cell types, the effects were compared of pharmacological manipulation of TRPA1 activity on EC proliferation tube formation and migration. In the presence and absence of a fibroblast containing feeder layer. Neither VEGF-induced increases in human vascular endothelial cell (HUVEC) proliferation nor migration were changed by a TRPA1 antagonist HC-030031 in the absence of a feeder layer. However, on a fibroblast feeder layer this antagonist suppressed HUVEC tube formation. In conclusion, during corneal wound healing transactivation by VEGFR of TRPA1 contributes to mediating neovascularization and macrophage infiltration. Such crosstalk is possible because of close proximity between VEGFR delimited expression on EC and TRPA1 expression restricted to cell types adhering to EC.
Collapse
Affiliation(s)
- Keiko Usui-Kusumoto
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Hiroki Iwanishi
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Kana Ichikawa
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan.
| | - Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Masayasu Miyajima
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | | | - Peter S Reinach
- Wenzhou Medical University School of Ophthalmology and Optometry, Wenzhou, PR China
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| |
Collapse
|
16
|
Inoue M, Naritani M, Raju R, Miyagi M, Oshima M, Inoue M, Matsuka Y. Effect of Short-term Tumour Necrosis Factor-alpha (TNF-α) -stimulation on the Growth and Differentiation of MC3T3-E1 Osteoblast-like Cells. J HARD TISSUE BIOL 2018. [DOI: 10.2485/jhtb.27.213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Miho Inoue
- Department of Stomatognathic Function and Occlusal Reconstruction, Tokushima University
| | - Mio Naritani
- Department of Stomatognathic Function and Occlusal Reconstruction, Tokushima University
| | - Resmi Raju
- Department of Stomatognathic Function and Occlusal Reconstruction, Tokushima University
| | - Mayu Miyagi
- Department of Stomatognathic Function and Occlusal Reconstruction, Tokushima University
| | - Masamitsu Oshima
- Department of Stomatognathic Function and Occlusal Reconstruction, Tokushima University
| | - Masahisa Inoue
- Laboratories for Structure and Function Research, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Yoshizo Matsuka
- Department of Stomatognathic Function and Occlusal Reconstruction, Tokushima University
| |
Collapse
|
17
|
Sumioka T, Iwanishi H, Okada Y, Nidegawa Y, Miyajima M, Matsumoto KI, Saika S. Loss of tenascin X gene function impairs injury-induced stromal angiogenesis in mouse corneas. J Cell Mol Med 2017; 22:948-956. [PMID: 29160014 PMCID: PMC5783828 DOI: 10.1111/jcmm.13397] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 08/26/2017] [Indexed: 12/18/2022] Open
Abstract
To determine the contribution by tenascin X (Tnx) gene expression to corneal stromal angiogenesis, the effects were determined of its loss on this response in TNX knockout (KO) mice. In parallel, the effects of such a loss were evaluated on vascular endothelial growth factor (VEGF) and transforming growth factor β1 (TGFβ1) gene and protein expression in fibroblasts and macrophages in cell culture. Histological, immunohistochemical and quantitative RT‐PCR changes determined if Tnx gene ablation on angiogenic gene expression, inflammatory cell infiltration and neovascularization induced by central corneal stromal cauterization. The role was determined of Tnx function in controlling VEGF‐A or TGFβ1 gene expression by comparing their expression levels in ocular fibroblasts and macrophages obtained from wild‐type (WT) and body‐wide Tnx KO mice. Tnx was up‐regulated in cauterized cornea. In Tnx KO, macrophage invasion was attenuated, VEGF‐A and its cognate receptor mRNA expression along with neovascularization were lessened in Tnx KOs relative to the changes occurring in their WT counterpart. Loss of Tnx instead up‐regulated in vivo mRNA expression of anti‐angiogenic VEGF‐B but not VEGF‐A. On the other hand, TGFβ1 mRNA expression declined in Tnx KO cultured ocular fibroblasts. Loss of Tnx gene expression caused VEGF‐A expression to decline in macrophages. Tnx gene expression contributes to promoting TGFβ1 mRNA expression in ocular fibroblasts and VEGF‐A in macrophages, macrophage invasion, up‐regulation of VEGF‐A expression and neovascularization in an injured corneal stroma. On the other hand, it suppresses anti‐angiogenic VEGF‐B mRNA expression in vivo.
Collapse
Affiliation(s)
- Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Hiroki Iwanishi
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Yuka Nidegawa
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Masayasu Miyajima
- Animal Center, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Ken-Ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| |
Collapse
|
18
|
Mizoguchi S, Iwanishi H, Kokado M, Sumioka T, Parfitt GJ, Xie Y, Arita R, Shirakawa R, Yamanaka O, Okada Y, Jester JV, Saika S. Ocular surface alkali injury damages meibomian glands in mice. Ocul Surf 2017; 15:713-722. [PMID: 28442381 DOI: 10.1016/j.jtos.2017.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 04/20/2017] [Accepted: 04/20/2017] [Indexed: 11/16/2022]
Abstract
PURPOSE To examine effects of alkali injury of the ocular surface on meibomian gland pathology in mice. METHODS Three μL of 1 N NaOH were applied under general anesthesia to the right eye of 10-week-old BALB/c (n = 54) mice to produce a total ocular surface alkali burn. The meibomian gland morphology was examined at days 1, 2, 5, 10, and 20 by stereomicroscopy and non-contact infrared meibography. Mice were then sacrificed and eyelids processed for histology with hematoxylin-eosin and immunohistochemistry for ELOVL4, PPARγ, myeloperoxidase (a neutrophil marker) and F4/80 macrophage antigen, as well as TUNEL staining. Another set of specimens was processed for cryosectioning and Oil red O staining. RESULTS Alkali injury to the ocular surface produced cellular apoptosis, infiltration of neutrophils and macrophages, degeneration of the meibomian gland, and ductal dilation. Inflammation in and destruction of acunal stricture seemed more prominent in the lower eyelid, while duct dilation was more frequently observed in the upper eyelid during healing. Surviving acinar cells were labeled for ELOVL4 and PPARγ. Oil red O staining showed that the substance in the dilated duct contained predominantly neutral lipid. CONCLUSIONS Alkali injury to the ocular surface results in damage and destruction of the eyelid meibomian glands. The pattern of the tissue damage differs between glands of the upper and lower eyelids.
Collapse
Affiliation(s)
- Shin Mizoguchi
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Hiroki Iwanishi
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan.
| | - Masahide Kokado
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | | | - Yilu Xie
- Gavin Herbert Eye Institute, University of California, Irvine, USA
| | | | - Rika Shirakawa
- Department of Ophthalmology, The University of Tokyo, Tokyo, Japan
| | - Osamu Yamanaka
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - James V Jester
- Gavin Herbert Eye Institute, University of California, Irvine, USA
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
19
|
Impaired Healing of a Cutaneous Wound in an Inducible Nitric Oxide Synthase-Knockout Mouse. Dermatol Res Pract 2017; 2017:2184040. [PMID: 28487726 PMCID: PMC5406723 DOI: 10.1155/2017/2184040] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/28/2017] [Indexed: 01/30/2023] Open
Abstract
Background. We investigated the effects of loss of inducible nitric oxide synthase (iNOS) on the healing process of cutaneous excisional injury by using iNOS-null (KO) mice. Population of granulation tissue-related cell types, that is, myofibroblasts and macrophages, growth factor expression, and reepithelialization were evaluated. Methods. KO and wild type (WT) mice of C57BL/6 background were used. Under general anesthesia two round full-thickness excision wounds of 5.0 mm in diameter were produced in dorsal skin. After specific intervals of healing, macroscopic observation, histology, immunohistochemistry, and real-time reverse transcription-polymerase chain reaction (RT-PCR) were employed to evaluate the healing process. Results. The loss of iNOS retards granulation tissue formation and reepithelialization in excision wound model in mice. Detailed analyses showed that myofibroblast appearance, macrophage infiltration, and mRNA expression of transforming growth factor b and of collagen 1α2 were all suppressed by lacking iNOS. Conclusions. iNOS is required in the process of cutaneous wound healing. Lacking iNOS retards macrophage invasion and its expression of fibrogenic components that might further impair fibrogenic behaviors of fibroblasts.
Collapse
|
20
|
Eisenman ST, Gibbons SJ, Verhulst PJ, Cipriani G, Saur D, Farrugia G. Tumor necrosis factor alpha derived from classically activated "M1" macrophages reduces interstitial cell of Cajal numbers. Neurogastroenterol Motil 2017; 29:10.1111/nmo.12984. [PMID: 27781339 PMCID: PMC5367986 DOI: 10.1111/nmo.12984] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/20/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Delayed gastric emptying in diabetic mice and humans is associated with changes in macrophage phenotype and loss of interstitial cells of Cajal (ICC) in the gastric muscle layers. In diabetic mice, classically activated M1 macrophages are associated with delayed gastric emptying, whereas alternatively activated M2 macrophages are associated with normal gastric emptying. This study aimed to determine if secreted factors from M1 macrophages could injure mouse ICC in primary culture. METHODS Cultures of gastric ICC were treated with conditioned medium (CM) from activated bone marrow-derived macrophages (BMDMs) and the effect of CM was quantified by counting ICC per high-powered field. KEY RESULTS Bone marrow-derived macrophages were activated to a M1 or M2 phenotype confirmed by qRT-PCR. Conditioned medium from M1 macrophages reduced ICC numbers by 41.1%, whereas M2-CM had no effect as compared to unconditioned, control media. Immunoblot analysis of 40 chemokines/cytokines found 12 that were significantly increased in M1-CM, including tumor necrosis factor alpha (TNF-α). ELISA detected 0.697±0.03 ng mL-1 TNF-α in M1-CM. Recombinant mouse TNF-α reduced Kit expression and ICC numbers in a concentration-dependent manner (EC50 = 0.817 ng mL-1 ). Blocking M1-CM TNF-α with a neutralizing antibody preserved ICC numbers. The caspase inhibitor Z-VAD.fmk partly preserved ICC numbers (cells/field; 6.63±1.04, 9.82±1.80 w/Z-VAD.fmk, n=6, P<.05). CONCLUSIONS & INFERENCES This work demonstrates that TNF-α secreted from M1 macrophages can result in Kit loss and directly injure ICC in vitro partly through caspase-dependent apoptosis and may play an important role in ICC depletion in diabetic gastroparesis.
Collapse
Affiliation(s)
| | | | | | | | - Dieter Saur
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | | |
Collapse
|
21
|
Chinnery HR, McMenamin PG, Dando SJ. Macrophage physiology in the eye. Pflugers Arch 2017; 469:501-515. [DOI: 10.1007/s00424-017-1947-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/29/2017] [Accepted: 01/31/2017] [Indexed: 10/20/2022]
|
22
|
Loss of TRPV4 Function Suppresses Inflammatory Fibrosis Induced by Alkali-Burning Mouse Corneas. PLoS One 2016; 11:e0167200. [PMID: 28030558 PMCID: PMC5193391 DOI: 10.1371/journal.pone.0167200] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 11/10/2016] [Indexed: 01/21/2023] Open
Abstract
In humans suffering from pulmonary disease and a mouse model, transient receptor potential vanilloid 4 (TRPV4) channel activation contributes to fibrosis. As a corneal alkali burn induces the same response, we determined if such an effect is also attributable to TRPV4 activation in mice. Accordingly, we determined if the alkali burn wound healing responses in wild-type (WT) mice are different than those in their TRPV4-null (KO) counterpart. Stromal opacification due to fibrosis in KO (n = 128) mice was markedly reduced after 20 days relative to that in WT (n = 157) mice. Immunohistochemistry revealed that increases in polymorphonuclear leukocytes and macrophage infiltration declined in KO mice. Semi-quantitative real time RT-PCR of ocular KO fibroblast cultures identified increases in proinflammatory and monocyte chemoattractant protein-1 chemoattractant gene expression after injury. Biomarker gene expression of fibrosis, collagen1a1 and α-smooth muscle actin were attenuated along with macrophage release of interleukin-6 whereas transforming growth factor β, release was unchanged. Tail vein reciprocal bone marrow transplantation between WT and KO chimera mouse models mice showed that reduced scarring and inflammation in KO mice are due to loss of TRPV4 expression on both corneal resident immune cells, fibroblasts and infiltrating polymorphonuclear leukocytes and macrophages. Intraperitoneal TRPV4 receptor antagonist injection of HC-067047 (10 mg/kg, daily) into WT mice reproduced the KO-phenotype. Taken together, alkali-induced TRPV4 activation contributes to inducing fibrosis and inflammation since corneal transparency recovery was markedly improved in KO mice.
Collapse
|
23
|
Saika S, Yamanaka O, Okada Y, Sumioka T. Modulation of Smad signaling by non-TGFβ components in myofibroblast generation during wound healing in corneal stroma. Exp Eye Res 2016; 142:40-8. [PMID: 26675402 DOI: 10.1016/j.exer.2014.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/05/2014] [Accepted: 12/26/2014] [Indexed: 10/22/2022]
Abstract
Corneal scarring/fibrosis disturbs normal transparency and curvature of the tissue and thus impairs vision. The lesion is characterized by appearance of myofibroblasts, the key player of the fibrogenic reaction, and excess accumulation of extracellular matrix. Inflammatory/fibrogenic growth factors or cytokines expressed in inflammatory cells that infiltrate into injured tissues play a pivotal role in fibrotic tissue formation. In this article the pathogenesis of fibrosis/scarring in the corneal stroma is reviewed focusing on the roles of myofibroblast, the key player in corneal stromal wound healing and fibrosis, and cytoplasmic signals activated by the fibrogenic cytokine, transforming growth factor β (TGFβ). Although it is established that TGFβ/Smad signal is essential to the process of keratocyte-myofibroblast transformation in a healing corneal stroma post-injury. This article emphasizes the involvement of non-TGFβ molecular mechanisms in modulating Smad signal. We focus on the roles of matricellular proteins, i.e., osteopontin and tenascin C, and as cellular components, the roles of transient receptor potential (TRP) cation channel receptors are discussed. Our intent is to draw attention to the possibility of signal transduction cascade modulation (e.g., Smad signal and mitogen-activated protein kinases, by gene transfer and other related technology) as being beneficial in a clinical setting to reduce or even prevent corneal stromal tissue fibrosis/scarring and inflammation.
Collapse
Affiliation(s)
- Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-0012, Japan.
| | - Osamu Yamanaka
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-0012, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-0012, Japan
| | - Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-0012, Japan
| |
Collapse
|
24
|
Chen L, Mongan M, Meng Q, Wang Q, Kao W, Xia Y. Corneal Wound Healing Requires IKB kinase β Signaling in Keratocytes. PLoS One 2016; 11:e0151869. [PMID: 26987064 PMCID: PMC4795706 DOI: 10.1371/journal.pone.0151869] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 03/04/2016] [Indexed: 11/19/2022] Open
Abstract
IkB kinase β (IKKβ) is a key signaling kinase for inflammatory responses, but it also plays diverse cell type-specific roles that are not yet fully understood. Here we investigated the role of IKKβ in the cornea using IkkβΔCS mice in which the Ikkβ gene was specifically deleted in the corneal stromal keratocytes. The IkkβΔCS corneas had normal morphology, transparency and thickness; however, they did not heal well from mild alkali burn injury. In contrast to the IkkβF/F corneas that restored transparency in 2 weeks after injury, over 50% of the IkkβΔCS corneas failed to fully recover. They instead developed recurrent haze with increased stromal thickness, severe inflammation and apoptosis. This pathogenesis correlated with sustained myofibroblast transformation with increased α smooth muscle actin (α-SMA) expression, higher levels of senescence β-Gal activity and scar tissue formation at the late stage of wound healing. In addition, the IkkβΔCS corneas displayed elevated expression of hemo-oxygenase-1 (HO-1), a marker of oxidative stress, and activation of stress signaling pathways with increased JNK, c-Jun and SMAD2/3 phosphorylation. These data suggest that IKKβ in keratocytes is required to repress oxidative stress and attenuate fibrogenesis and senescence in corneal wound healing.
Collapse
Affiliation(s)
- Liang Chen
- Department of Environmental Health and Center of Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267–0056, United States of America
| | - Maureen Mongan
- Department of Environmental Health and Center of Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267–0056, United States of America
| | - Qinghang Meng
- Department of Environmental Health and Center of Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267–0056, United States of America
| | - Qin Wang
- Department of Environmental Health and Center of Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267–0056, United States of America
| | - Winston Kao
- Department of Ophthalmology, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267–0056, United States of America
| | - Ying Xia
- Department of Environmental Health and Center of Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267–0056, United States of America
- * E-mail:
| |
Collapse
|
25
|
Bone marrow-derived cells in ocular neovascularization: contribution and mechanisms. Angiogenesis 2016; 19:107-18. [PMID: 26880135 PMCID: PMC4819501 DOI: 10.1007/s10456-016-9497-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 02/02/2016] [Indexed: 01/16/2023]
Abstract
Ocular neovascularization often leads to severe vision loss. The role of bone marrow-derived cells (BMCs) in the development of ocular neovascularization, and its significance, is increasingly being recognized. In this review, we discuss their contribution and the potential mechanisms that mediate the effect of BMCs on the progression of ocular neovascularization. The sequence of events by which BMCs participate in ocular neovascularization can be roughly divided into four phases, i.e., mobilization, migration, adhesion and differentiation. This process is delicately regulated and liable to be affected by multiple factors. Cytokines such as vascular endothelial growth factor, granulocyte colony-stimulating factor and erythropoietin are involved in the mobilization of BMCs. Studies have also demonstrated a key role of cytokines such as stromal cell-derived factor-1, tumor necrosis factor-α, as well as vascular endothelial growth factor, in regulating the migration of BMCs. The adhesion of BMCs is mainly regulated by vascular cell adhesion molecule-1, intercellular adhesion molecule-1 and vascular endothelial cadherin. However, the mechanisms regulating the differentiation of BMCs are largely unknown at present. In addition, BMCs secrete cytokines that interact with the microenvironment of ocular neovascularization; their contribution to ocular neovascularization, especially choroidal neovascularization, can be aggravated by several risk factors. An extensive regulatory network is thought to modulate the role of BMCs in the development of ocular neovascularization. A comprehensive understanding of the involved mechanisms will help in the development of novel therapeutic strategies related to BMCs. In this review, we have limited the discussion to the recent progress in this field, especially the research conducted at our laboratory.
Collapse
|
26
|
Suppression of In Vivo Neovascularization by the Loss of TRPV1 in Mouse Cornea. J Ophthalmol 2015; 2015:706404. [PMID: 26491553 PMCID: PMC4600561 DOI: 10.1155/2015/706404] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/06/2015] [Accepted: 03/16/2015] [Indexed: 11/18/2022] Open
Abstract
To investigate the effects of loss of transient receptor potential vanilloid receptor 1 (TRPV1) on the development of neovascularization in corneal stroma in mice. Blocking TRPV1 receptor did not affect VEGF-dependent neovascularization in cell culture. Lacking TRPV1 inhibited neovascularization in corneal stroma following cauterization. Immunohistochemistry showed that immunoreactivity for active form of TGFβ1 and VEGF was detected in subepithelial stroma at the site of cauterization in both genotypes of mice, but the immunoreactivity seemed less marked in mice lacking TRPV1. mRNA expression of VEGF and TGFβ1 in a mouse cornea was suppressed by the loss of TRPV1. TRPV1 gene ablation did not affect invasion of neutrophils and macrophage in a cauterized mouse cornea. Blocking TRPV1 signal does not affect angiogenic effects by HUVECs in vitro. TRPV1 signal is, however, involved in expression of angiogenic growth factors in a cauterized mouse cornea and is required for neovascularization in the corneal stroma in vivo.
Collapse
|
27
|
Tumor necrosis factor-α inhibitors as a treatment of corneal hemangiogenesis and lymphangiogenesis. Eye Contact Lens 2015; 41:72-6. [PMID: 25503908 DOI: 10.1097/icl.0000000000000071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The cornea is normally devoid of blood and lymphatic vessels; however, a number of infectious/inflammatory diseases can induce corneal neovascularization (CNV). Tumor necrosis factor (TNF)-α, a well known pro-inflammatory cytokine, acts on the vascular endothelium by promoting vasodilatation, edema, and leukocyte recruitment, which are all commonly associated with the development of CNV. Corneal neovascularization is the second cause of blindness worldwide; hence, pharmacological TNF-α inhibition might represent an attractive therapeutic option. Although none of the existing TNF-α antagonists has been registered as a CNV inhibitor, three of them (etanercept, adalimumab, and infliximab) have been proposed to control ocular inflammation. More specifically, it has been demonstrated that infliximab is also effective in reducing hemangiogenesis and lymphangiogenesis in different animal models of CNV. In this article, we review the role of TNF-α on the ocular surface and, in particular, its specific role in the process of CNV. Moreover, we review existing literature and speculate on the potential role of TNF-α inhibitors in the treatment of CNV.
Collapse
|
28
|
Strojny C, Boyle M, Bartholomew A, Sundivakkam P, Alapati S. Interferon Gamma-treated Dental Pulp Stem Cells Promote Human Mesenchymal Stem Cell Migration In Vitro. J Endod 2015; 41:1259-64. [PMID: 26051078 DOI: 10.1016/j.joen.2015.02.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 01/15/2015] [Accepted: 02/14/2015] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Chronic inflammation disrupts dental pulp regeneration by disintegrating the recruitment process of progenitors for repair. Bone marrow-derived mesenchymal stem cells (BM-MSCs) share the common features with dental pulp stem cells (DPSCs). The aim of the study was to investigate the migration of BM-MSCs toward DPSCs in response to inflammatory chemoattractants. Additionally, our studies also delineated the signaling mechanisms from BM-MSCs in mediating the proliferation and differentiation of DPSCs in vitro. METHODS Human DPSCs and BM-MSCs between passages 2 and 4 were used and were grown in odontogenic differentiation medium. Mineralization was determined by alizarin red staining analysis. Migration was assessed using crystal violet staining in cells grown in Boyden chamber Transwell inserts (Corning Inc Foundation, Tewksbury, MA). The mineralization potential of DPSCs was evaluated using alkaline phosphatase activity assay. Real-time polymerase chain reaction analysis was performed to assess the gene expression profile of chemokine (C-X-C motif) ligand (Cxcl) 3, 5, 6, 10, 11, 12, 14, and 16; stromal cell-derived factor (SDF) α; vascular endothelial growth factor; and fibroblast growth factor. RESULTS Interferon gamma (FN-γ) treatment significantly abrogated the differentiation potential of DPSCs as shown by using alizarin red and alkaline phosphatase activity analysis. An increase in the migration of BM-MSCs was documented when cocultured with IFN-γ-treated DPSCs. RNA expression studies showed an increase in the levels of Cxcl6 and Cxcl12 in BM-MSCs when cocultured with IFN-γ-treated DPSCs. Additionally, an up-regulation of proangiogenic factors vascular endothelial growth factor and fibroblast growth factor were observed in DPSCs exposed to IFN-γ. CONCLUSIONS Our findings indicate that inflamed IFN-γ-treated DPSCs release factors (presumably Cxcl6 and 12) that contribute to the homing of MSCs. This model might provide a potential research tool for studying MSC-DPSC cross talk and for future studies involving the recruitment and sustainability of progenitor stem cells sustaining the inflammatory cascade to treat pulp inflammation.
Collapse
Affiliation(s)
- Chelsee Strojny
- Department of Endodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Michael Boyle
- Department of Endodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Amelia Bartholomew
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Premanand Sundivakkam
- Department of Endodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois; Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Satish Alapati
- Department of Endodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois; Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
29
|
Strand DW, Liang YY, Yang F, Barron DA, Ressler SJ, Schauer IG, Feng XH, Rowley DR. TGF-β induction of FGF-2 expression in stromal cells requires integrated smad3 and MAPK pathways. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2014; 2:239-248. [PMID: 25374926 PMCID: PMC4219310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 09/30/2014] [Indexed: 06/04/2023]
Abstract
Transforming Growth Factor-β (TGF-β) regulates the reactive stroma microenvironment associated with most carcinomas and mediates expression of many stromal derived factors important for tumor progression, including FGF-2 and CTGF. TGF-β is over-expressed in most carcinomas, and FGF-2 action is important in tumor-induced angiogenesis. The signaling mechanisms of how TGF-β regulates FGF-2 expression in the reactive stroma microenvironment are not understood. Accordingly, we have assessed key signaling pathways that mediate TGF-β1-induced FGF-2 expression in prostate stromal fibroblasts and mouse embryo fibroblasts (MEFs) null for Smad2 and Smad3. TGF-β1 induced phosphorylation of Smad2, Smad3, p38 and ERK1/2 proteins in both control MEFs and prostate fibroblasts. Of these, Smad3, but not Smad2 was found to be required for TGF-β1 induction of FGF-2 expression in stromal cells. ChIP analysis revealed a Smad3/Smad4 complex was associated with the -1.9 to -2.3 kb upstream proximal promoter of the FGF-2 gene, further suggesting a Smad3-specific regulation. In addition, chemical inhibition of p38 or ERK1/2 MAPK activity also blocked TGF-β1-induced FGF-2 expression in a Smad3-independent manner. Conversely, inhibition of JNK signaling enhanced FGF-2 expression. Together, these data indicate that expression of FGF-2 in fibroblasts in the tumor stromal cell microenvironment is coordinately dependent on both intact Smad3 and MAP kinase signaling pathways. These pathways and key downstream mediators of TGF-β action in the tumor reactive stroma microenvironment, may evolve as putative targets for therapeutic intervention.
Collapse
Affiliation(s)
- Douglas W Strand
- Department of Molecular and Cellular Biology, Baylor College of MedicineHouston, TX, USA
- Current address: Department of Urology, University of Texas Southwestern Medical CenterDallas, TX 75390-9110
| | - Yao-Yun Liang
- Department of Molecular and Cellular Biology, Baylor College of MedicineHouston, TX, USA
- Department of Surgery, Baylor College of MedicineHouston, TX, USA
| | - Feng Yang
- Department of Molecular and Cellular Biology, Baylor College of MedicineHouston, TX, USA
| | - David A Barron
- Department of Molecular and Cellular Biology, Baylor College of MedicineHouston, TX, USA
- Current address: Department of Radiation Oncology, Memorial Sloan-Kettering Cancer CenterNew York City, NY 10065
| | - Steven J Ressler
- Department of Molecular and Cellular Biology, Baylor College of MedicineHouston, TX, USA
- Current address: Aptalis Pharmaceuticals100 Somerset Corporate Blvd, Bridgewater, NJ 08807, USA
| | - Isaiah G Schauer
- Department of Molecular and Cellular Biology, Baylor College of MedicineHouston, TX, USA
- Current address: Department of Mathematics and Life Sciences, Brazosport CollegeLake Jackson, TX 77566
| | - Xin-Hua Feng
- Department of Molecular and Cellular Biology, Baylor College of MedicineHouston, TX, USA
- Department of Surgery, Baylor College of MedicineHouston, TX, USA
| | - David R Rowley
- Department of Molecular and Cellular Biology, Baylor College of MedicineHouston, TX, USA
| |
Collapse
|
30
|
Okada Y, Shirai K, Reinach PS, Kitano-Izutani A, Miyajima M, Flanders KC, Jester JV, Tominaga M, Saika S. TRPA1 is required for TGF-β signaling and its loss blocks inflammatory fibrosis in mouse corneal stroma. J Transl Med 2014; 94:1030-41. [PMID: 25068659 PMCID: PMC5919187 DOI: 10.1038/labinvest.2014.85] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 05/28/2014] [Accepted: 05/29/2014] [Indexed: 12/19/2022] Open
Abstract
We examined whether the loss of transient receptor potential ankyrin 1 (TRPA1), an irritant-sensing ion channel, or TRPA1 antagonist treatment affects the severity inflammation and scarring during tissue wound healing in a mouse cornea injury model. In addition, the effects of the absence of TRPA1 on transforming growth factor β1 (TGF-β1)-signaling activation were studied in cell culture. The lack of TRPA1 in cultured ocular fibroblasts attenuated expression of TGF-β1, interleukin-6, and α-smooth muscle actin, a myofibroblast the marker, but suppressed the activation of Smad3, p38 MAPK, ERK, and JNK. Stroma of the healing corneas of TRPA1(-/-) knockout (KO) mice appeared more transparent compared with those of wild-type mice post-alkali burn. Eye globe diameters were measured from photographs. An examination of the corneal surface and eye globes suggested the loss of TRPA1 suppressed post-alkali burn inflammation and fibrosis/scarring, which was confirmed by histology, immunohistochemistry, and gene expression analysis. Reciprocal bone marrow transplantation between mice showed that KO corneal tissue resident cells, but not KO bone marrow-derived cells, are responsible for KO mouse wound healing with reduced inflammation and fibrosis. Systemic TRPA1 antagonists reproduced the KO phenotype of healing. In conclusion, a loss or blocking of TRPA1 in mice reduces inflammation and fibrosis/scarring in the corneal stroma during wound healing following an alkali burn. The responsible mechanism may include the inhibition of TGF-β1-signaling cascades in fibroblasts by attenuated TRPA1 signaling. Inflammatory cells are considered to have a minimum involvement in the exhibition of the KO phenotype after injury.
Collapse
Affiliation(s)
- Yuka Okada
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Kumi Shirai
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Peter S Reinach
- Wenzhou Medical University School of Ophthalmology and Optometry, Wenzhou, China
| | - Ai Kitano-Izutani
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Masayasu Miyajima
- Laboratory Animal Center, Wakayama Medical University, Wakayama, Japan
| | - Kathleen C Flanders
- Laboratory of Cell Regulation and Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Makoto Tominaga
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, National Institute of Natural Science, Okazaki, Aichi, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
31
|
Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab 2014; 20:103-18. [PMID: 24930973 PMCID: PMC4079756 DOI: 10.1016/j.cmet.2014.05.005] [Citation(s) in RCA: 487] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/09/2014] [Accepted: 04/28/2014] [Indexed: 02/08/2023]
Abstract
Chronic inflammation constitutes an important link between obesity and its pathophysiological sequelae. In contrast to the belief that inflammatory signals exert a fundamentally negative impact on metabolism, we show that proinflammatory signaling in the adipocyte is in fact required for proper adipose tissue remodeling and expansion. Three mouse models with an adipose tissue-specific reduction in proinflammatory potential were generated that display a reduced capacity for adipogenesis in vivo, while the differentiation potential is unaltered in vitro. Upon high-fat-diet exposure, the expansion of visceral adipose tissue is prominently affected. This is associated with decreased intestinal barrier function, increased hepatic steatosis, and metabolic dysfunction. An impaired local proinflammatory response in the adipocyte leads to increased ectopic lipid accumulation, glucose intolerance, and systemic inflammation. Adipose tissue inflammation is therefore an adaptive response that enables safe storage of excess nutrients and contributes to a visceral depot barrier that effectively filters gut-derived endotoxin.
Collapse
|
32
|
Papaconstantinou I, Zeglinas C, Gazouli M, Nastos K, Yiallourou A, Papalois A, Tzathas C. The impact of peri-operative anti-TNF treatment on anastomosis-related complications in Crohn's disease patients. A critical review. J Gastrointest Surg 2014; 18:1216-24. [PMID: 24599780 DOI: 10.1007/s11605-014-2487-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 02/13/2014] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Tumor necrosis factor (TNF)α is a cytokine exerting pleiotropic effects on critical cell functions and, most importantly, is the main regulator of pro-inflammatory cytokine production and a key player in the pathophysiology of numerous autoimmune diseases, including Crohn's disease. METHODS TNFα became a therapeutic target and TNFα blocking agents are currently used in the treatment of inflammatory diseases. Beyond the therapeutic benefits deriving from TNFα neutralization, amendments in the cellular functions of the immune system may as well induce potent immunosuppressive effects. An attenuated immune response may compromise the intestinal healing ability, thus leading to weaker anastomosis and increased risk of anastomotic leak and septic complications. RESULTS This hypothesis raises great concerns about the safety of perioperative administration of anti-TNF and has been the endpoint of numerous studies. CONCLUSION The aim of this review is to critically evaluate the evidence regarding the role of TNFα in anastomosis related complications after abdominal surgery in Crohn's disease patients.
Collapse
Affiliation(s)
- Ioannis Papaconstantinou
- Second Department of Surgery, Aretaieion Hospital, Medical School, University of Athens, Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
33
|
A short-term treatment with tumor necrosis factor-alpha enhances stem cell phenotype of human dental pulp cells. Stem Cell Res Ther 2014; 5:31. [PMID: 24580841 PMCID: PMC4055131 DOI: 10.1186/scrt420] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 02/17/2014] [Indexed: 01/09/2023] Open
Abstract
Introduction During normal pulp tissue healing, inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) or interleukins, act in the initial 48 hours (inflammatory phase) and play important roles not only as chemo-attractants of inflammatory cells and stem/progenitor cells but also in inducing a cascade of reactions toward tissue regeneration or reparative dentin formation or both. Previous reports have shown that inflammatory cytokines regulate the differentiation capacity of dental pulp stem/progenitor cells (DPCs), but none has interrogated the impact of these cytokines on the stem cell phenotype of stem/progenitor cells. This study investigated the effects of a short-term treatment with TNF-α on the stem cell phenotype and differentiation ability of human DPCs. Methods An in vivo mouse model of pulp exposure was performed for analysis of expression of the mesenchymal stem cell marker CD146 in DPCs during the initial stage of inflammatory response. For in vitro studies, human DPCs were isolated and incubated with TNF-α for 2 days and passaged to eliminate TNF-α completely. Analysis of stem cell phenotype was performed by quantification of cells positive for mesenchymal stem cell markers SSEA-4 (stage-specific embryonic antigen 4) and CD146 by flow cytometry as well as by quantitative analysis of telomerase activity and mRNA levels of OCT-4 and NANOG. Cell migration, colony-forming ability, and differentiation toward odontogenesis and adipogenesis were also investigated. Results The pulp exposure model revealed a strong staining for CD146 during the initial inflammatory response, at 2 days after pulp exposure. In vitro experiments demonstrated that a short-term (2-day) treatment of TNF-α increased by twofold the percentage of SSEA-4+ cells. Accordingly, STRO-1, CD146, and SSEA-4 protein levels as well as OCT-4 and NANOG mRNA levels were also significantly upregulated upon TNF-α treatment. A short-term TNF-α treatment also enhanced DPC function, including the ability to form cell colonies, to migrate, and to differentiate into odontogenic and adipogenic lineages. Conclusions A short-term treatment with TNF-α enhanced the stem cell phenotype, migration, and differentiation ability of DPCs.
Collapse
|
34
|
Chen TC, Chang SW, Wang TY. Moxifloxacin modifies corneal fibroblast-to-myofibroblast differentiation. Br J Pharmacol 2013; 168:1341-54. [PMID: 23072440 DOI: 10.1111/bph.12015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Revised: 09/24/2012] [Accepted: 09/28/2012] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Fibroblast-to-myofibroblast differentiation is associated with scarring, an important issue in corneal surgery. Moxifloxacin (MOX), commonly applied to prevent post-operative infection, would benefit more if it modifies fibroblast-to-myofibroblast differentiation other than antimicrobial activity. Our purpose was to explore whether MOX has anti-fibrotic effect in human corneal fibroblasts (HCFs). EXPERIMENTAL APPROACH HCFs were incubated in MOX-containing medium concurrently with TGF-β1 (co-treatment), before (pretreatment) or after (post-treatment) adding TGF-β1. HCF contractility was evaluated with a type I collagen gel contraction assay. Expression of α-smooth muscle actin (α-SMA), Smad2, phospho-Smad2-Ser467, Smad4 and Smad7 was determined by immunoblotting. Formation of α-SMA-positive filaments and distribution of active Smad2 were observed under confocal microscopy. Expression of TGF-β receptor types I (TGFBR1) and II (TGFBR2) was assessed with flow cytometry. KEY RESULTS MOX did not affect gel contractility or α-SMA filament formation in HCFs without TGF-β1 stimulation. MOX did, however, retard HCF-containing gel contractility and α-SMA filament formation following TGF-β1 stimulation in the pretreatment and co-treatment groups but not in the post-treatment group. MOX blocked the expression of Smad2, phospho-Smad2-Ser467 and TGFBR1 under TGF-β1 incubation. Additionally, MOX enhanced Smad7 expression in TGF-β1-incubated HCFs, but did not interfere with TGF-β-triggered Smad2 nuclear translocation or Smad4 expression. CONCLUSIONS AND IMPLICATIONS MOX inhibited TGF-β-induced fibroblast-to-myofibroblast differentiation via blocking TGFBR1 and enhancing Smad7 expression. MOX should be used before or during surgery to achieve these effects. These results suggest a de novo mechanism by which MOX participates in corneal wound healing.
Collapse
Affiliation(s)
- T C Chen
- Department of Ophthalmology, Far Eastern Memorial Hospital, Banqiao District, New Taipei City, Taiwan
| | | | | |
Collapse
|
35
|
Arancibia R, Oyarzún A, Silva D, Tobar N, Martínez J, Smith PC. Tumor Necrosis Factor-α Inhibits Transforming Growth Factor-β–Stimulated Myofibroblastic Differentiation and Extracellular Matrix Production in Human Gingival Fibroblasts. J Periodontol 2013; 84:683-93. [DOI: 10.1902/jop.2012.120225] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
36
|
Abstract
We investigated the effects of loss of tenascin C on the healing of the stroma using incision-injured mice corneas. Tenascin C was upregulated in the stroma following incision injury to the cornea. Wild-type (WT) and tenascin C-null (knockout (KO)) mice on a C57BL/6 background were used. Cell culture experiments were also conducted to determine the effects of the lack of tenascin C on fibrogenic gene expression in ocular fibroblasts. Histology, immunohistochemistry and real-time reverse transcription PCR were employed to evaluate the healing process in the stroma. The difference in the incidence of wound closure was statistically analyzed in hematoxylin and eosin-stained samples between WT and KO mice in addition to qualitative observation. Healing of incision injury in corneal stroma was delayed, with less appearance of myofibroblasts, less invasion of macrophages and reduction in expression of collagen Iα1, fibronectin and transforming growth factor β1 (TGFβ1) in KO mice compared with WT mice. In vitro experiments showed that the loss of tenascin C counteracted TGFβ1 acceleration of mRNA expression of TGFβ1, and of collagen Iα1 and of myofibroblast conversion in ocular fibroblasts. These results indicate that tenascin C modulates wound healing-related fibrogenic gene expression in ocular fibroblasts and is required for primary healing of the corneal stroma.
Collapse
|
37
|
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases with the potential to degrade all types of extracellular matrix. The ADAM (a disintegrin and metalloproteinase) family of peptidases was recently identified as cleaving the extracellular domain of transmembrane proteins. This was termed ectodomain shedding. We investigated the MMP expression in patients with corneal diseases and the potential role of ADAMs in corneal pathophysiology. We detected upregulation of the active form of MMP-2 and MMP-9 in the tear fluid from patients with corneal melting or recurrent corneal erosion. Using human corneal epithelial cells, we observed ADAM17-dependent ectodomain shedding of soluble tumor necrosis factor receptor 1 and soluble interleukin-6 (IL-6) receptor (sIL-6R). The production of sIL-6R was also induced by messenger RNA splicing in the human corneal epithelial cells. IL-6/sIL-6R-induced signal transducer and activator of transcription 3 phosphorylation was observed in cultured human corneal fibroblasts, suggesting that IL-6 trans-signaling induced inflammatory cellular signaling in the human corneal fibroblasts. We demonstrated that MMPs are significantly upregulated in collagen-destructive disorders of the cornea. Additionally, we observed that ectodomain shedding by ADAMs in corneal epithelial cells mediated the production of soluble cytokine receptors. Trans-signaling of IL-6 can induce an inflammatory response in corneal stroma, indicating the significance of IL-6 trans-signaling in ocular surface inflammation. Thus, MMPs and ADAMs play an important role in the pathophysiology of corneal diseases.
Collapse
|
38
|
Kiguchi N, Kobayashi Y, Maeda T, Tominaga S, Nakamura J, Fukazawa Y, Ozaki M, Kishioka S. Activation of nicotinic acetylcholine receptors on bone marrow-derived cells relieves neuropathic pain accompanied by peripheral neuroinflammation. Neurochem Int 2012; 61:1212-9. [PMID: 22989685 DOI: 10.1016/j.neuint.2012.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 07/31/2012] [Accepted: 09/01/2012] [Indexed: 01/21/2023]
Abstract
Emerging evidence indicates that chronic neuroinflammation plays a pivotal role in neuropathic pain. We explored whether activation of the nicotinic acetylcholine receptor (nAChRs) pathway on peripheral immune cells improves neuropathic pain. Mice were subjected to partial sciatic nerve ligation (PSL). Enhanced green fluorescent protein (EGFP)-chimeric mice were generated by transplantation of EGFP(+) bone marrow (BM) cells from EGFP-transgenic mice into wild-type mice. EGFP(+) BM-derived cells infiltrated the injured sciatic nerve (SCN) of EGFP-chimeric mice, and these cells were found to be F4/80(+) macrophages and Ly6G(+) neutrophils. The protein expression of nAChR subunit α4 and α7 were up-regulated in the injured SCN. Increased α4 and α7 subunits were localized on both BM-derived macrophages and neutrophils. When nicotine (20nmol) was perineurally administered once a day for 4days (days 0-3), PSL-induced tactile allodynia and thermal hyperalgesia were significantly prevented. Relieving effects of nicotine on neuropathic pain were reversed by co-administration of mecamylamine (20nmol), a non-selective antagonist for nAChRs. PSL-induced up-regulation of inflammatory cytokines and chemokines was suppressed by perineural administration of nicotine. Taken together, the expression of α4β2 and α7 subtypes of nAChRs may be increased on circulating macrophages and neutrophils in injured peripheral nerves. Activation of nAChRs on immune cells may relieve neuropathic pain accompanied by the suppression of neuroinflammation.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Pharmacology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-0012, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Okada Y, Reinach PS, Shirai K, Kitano A, Kao WWY, Flanders KC, Miyajima M, Liu H, Zhang J, Saika S. TRPV1 involvement in inflammatory tissue fibrosis in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2654-64. [PMID: 21641388 DOI: 10.1016/j.ajpath.2011.02.043] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 02/06/2011] [Accepted: 02/10/2011] [Indexed: 10/18/2022]
Abstract
We examined whether absence or blocking of transient receptor potential vanilloid subtype 1 (TRPV1) affects the level of inflammation and fibrosis/scarring during healing of injured tissue using an alkali burn model of cornea in mice. A cornea burn was produced with 1 N NaOH instilled into one eye of TRPV1-/- (KO) (n = 88) or TRPV1+/+ (n = 94) mice. Examinations of the corneal surface and eye globe size suggested that the loss of TRPV1 suppressed inflammation and fibrosis/scarring after alkali burn, and this was confirmed by histology, IHC, and gene expression analysis. The loss of TRPV1 inhibited inflammatory cell invasion and myofibroblast generation in association with reduction of expression of proinflammatory and profibrogenic components. Experiments of bone marrow transplantation between either genotype of mice showed that KO corneal tissue resident cells, but not KO bone marrow-derived cells, are responsible for KO-type wound healing with reduced inflammation and fibrosis. The absence of TRPV1 attenuated expression of transforming growth factor β 1 (TGFβ1) and other proinflammatory gene expression in cultured ocular fibroblasts, but did not affect TGFβ1 expression in macrophages. Loss of TRPV1 inhibited myofibroblast transdifferentiation in cultured fibroblasts. Systemic TRPV1 antagonists reproduced the KO type of healing. In conclusion, absence or blocking of TRPV1 suppressed inflammation and fibrosis/scarring during healing of alkali-burned mouse cornea. TRPV1 is a potential drug target for improving the outcome of inflammatory/fibrogenic wound healing.
Collapse
Affiliation(s)
- Yuka Okada
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Current concepts in scar evolution and control. Aesthetic Plast Surg 2011; 35:628-35. [PMID: 21136250 DOI: 10.1007/s00266-010-9635-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Accepted: 11/08/2010] [Indexed: 01/05/2023]
Abstract
The basic principles influencing scar expression and outcome have long been defined. Although these were relatively clear at the time, the exact events at a molecular level were poorly defined. The past decade has delineated the myriad of events that occur in the run-up to scar evolution far more clearly, although the intricate details have yet to be elucidated. What is clear is that a series of conversations and crosstalk takes place in the cell cytosol, in the cellular nucleus, and outside the cell within in the extracellular matrix. This interaction or "dynamic reciprocity" takes place via a series of signals, protein activation, ionic translocations, and receptor transactions. Marrying the previously defined principles with current described cellular/extracellular matrix (ECM) interactions enables us to describe more accurately the crosstalk occurring in scar evolution and possibly to influence the "wording" of that crosstalk to improve scar outcome. Thus, the principles of mechanostimulation and scar support, hydration occlusion, controlled inflammation, and collagen/extracellular remodeling are discussed with possible interventions in each category.
Collapse
|
41
|
Abstract
The principles of scar evolution and control are recognized and defined. Further clarity has been shed on these principles with the elucidation and elaboration of the sequence of events occurring at a molecular level. Cellular cross-talk among structures in the cell cytosol, in the cellular nucleus, and outside the cell within in the extracellular matrix is continuous and controlling in nature. This interaction or "dynamic reciprocity" takes place via a series of signals, ionic messenger shifts, protein activation, and receptor transactions. The described principles are now able to be defined in terms of cellular/extracellular matrix interactions and the identification of the cross-talk involved in scar evolution and maturation presents the possibility of influencing the "wording" of this cross-talk to improve scar outcome. The principles of mechanostimulation and scar support, hydration occlusion, controlled inflammation, and collagen/extracellular remodeling are discussed with possible interventions in each category.
Collapse
Affiliation(s)
- Alan D Widgerow
- Plastic Surgery Department, University of Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
42
|
Abstract
OBJECTIVE To review the use of genetically modified mouse lines for elucidating corneal morphogenesis during embryonic development and diseases. METHODS Transgenesis and gene-targeting techniques were used to create doxycycline-inducible mouse models (tet-On) to express transgenes or ablation of LoxP-modified genes or both in corneal cells, e.g., epithelial cells, and keratocytes and periocular mesenchymal cells of neural crest origin. RESULTS Two driver mouse lines, i.e., Krt12-rtTA and Kera-rtTA, were created, which express reverse tetracycline transcription activator (rtTA) in corneal epithelial cells and keratocytes, respectively. Bitransgenic (Krt12-rtTA/tet-o-FGF7) and triple transgenic mice (Krt12rtTA/tet-o-Cre/Ctnnb1 and Kera-rtTA/tet-o-Cre/Ctnnb1) were obtained through cross-breeding tet-o-FGF7, tet-o-Cre, and Ctnnb1 mice. On doxycycline induction, overexpression of FGF7 by corneal epithelial cells of bitransgenic Krt12-rtTA/tet-o-FGF7 mice caused nuclear translocation of beta-catenin and epithelium hyperplasia resembling human ocular surface squamous neoplasia; in triple transgenic mice (Krt12rtTA/tet-o-Cre/Ctnnb1), constitutive nuclear translocation of mutant beta-catenin (loss of exon 3) leads to hyper proliferation of corneal epithelial cells; in comparison of expression of beta-catenin mutant protein by migrating, periocular mesenchymal cells of Kera-rtTA/tet-o-Cre/Ctnnb1 caused eyelid malformation. CONCLUSIONS Use of genetically modified mice is of great value to study the pathophysiology of ocular surface defects resulting from genetic mutations.
Collapse
|
43
|
|
44
|
Kitano A, Okada Y, Yamanka O, Shirai K, Mohan RR, Saika S. Therapeutic potential of trichostatin A to control inflammatory and fibrogenic disorders of the ocular surface. Mol Vis 2010; 16:2964-73. [PMID: 21203344 PMCID: PMC3013068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2009] [Accepted: 12/26/2010] [Indexed: 10/29/2022] Open
Abstract
PURPOSE To examine the effects of a histone deacetylase inhibitor, Trichostatin A (TSA), on the behavior of macrophages and subconjunctival fibroblasts in vitro and on ocular surface inflammation and scarring in vivo using an alkali burn wound healing model. METHODS Effects of TSA on expression of inflammation-related growth factors or collagen I were examined by real-time RT-PCR or immunoassay in mouse macrophages or human subconjunctival fibroblasts. Effects of TSA on trans forming growth factor β (TGFβ)/Smad signaling were evaluated with western blotting and/or immunocytochemistry. Alkali-burn injuries on the eyes of mice were performed with three µl of 0.5 N NaOH under general and topical anesthesia. TSA (600 µg/Kg daily) or vehicle was administered to animals via intraperitoneal (i.p.) injection. Histology and real-time RT-PCR investigations evaluated the effects of TSA on the healing process of the cornea. RESULTS TSA inhibited TGFβ 1 and vascular endothelial growth factor (VEGF) expression in macrophages, and TGFβ1 and collagen I in ocular fibroblasts. It elevated the expression of 5'-TG-3'-interacting factor (TGIF) and Smad7 in fibroblasts and blocked nuclear translocation of phospho-Smad2. Real-time PCR and immunocytochemistry studies showed that systemic administration of TSA suppressed the inflammation and fibrotic response in the stroma and accelerated epithelial healing in the alkali-burned mouse cornea. CONCLUSIONS Systemic administration of TSA reduces inflammatory and fibrotic responses in the alkali-burned mouse ocular surface in vivo. The mechanisms of action involve attenuation of Smad signal in mesenchymal cells and reduction in the activation and recruitment of macrophages. TSA has the potential to treat corneal scarring in vivo.
Collapse
Affiliation(s)
- Ai Kitano
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Osamu Yamanka
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Kumi Shirai
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Rajiv R. Mohan
- Mason Eye Institute, University of Missouri-Columbia, Columbia, MO
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
45
|
Kao WWY, Liu CY. Corneal morphogenesis during development and wound healing. Jpn J Ophthalmol 2010; 54:206-10. [PMID: 20577853 DOI: 10.1007/s10384-010-0800-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 02/04/2010] [Indexed: 11/30/2022]
Affiliation(s)
- Winston W-Y Kao
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267-0838, USA.
| | | |
Collapse
|
46
|
Zhang F, Yang H, Pan Z, Wang Z, Wolosin JM, Gjorstrup P, Reinach PS. Dependence of resolvin-induced increases in corneal epithelial cell migration on EGF receptor transactivation. Invest Ophthalmol Vis Sci 2010; 51:5601-9. [PMID: 20538990 DOI: 10.1167/iovs.09-4468] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE To determine whether resolvin E1 (RvE1), an endogenous oxygenation product of eicosapentaenoic acid (EPA), induces increases in migration in human corneal epithelial cells (HCECs) and to identify signal pathways mediating this response. METHODS Migration was measured with the scratch wound assay. Western blot analysis identified changes in the phosphorylation status of prospective intracellular signal transduction mediators. Immunocytochemistry probed for intracellular paxillin localization and actin reorganization. RESULTS RvE1 enhanced HCEC migratory rates to levels comparable to those induced by epidermal growth factor (EGF). These increases were accompanied by increases in the phosphorylation status of epidermal growth factor receptor (EGFR), Akt, p38 MAPK, GSK-3α/β, and paxillin, which essentially persisted for up to 60 minutes. The EGFR inhibitor AG1478 blocked the subsequent effects of RvE1 to induce increases in phosphorylation status and cell migration. The PI3-K inhibitor LY294002 or wortmannin or the p38 inhibitor BIRB796 blocked resolvin-induced increases in cell migration. Either the matrix metalloproteinase (MMP) inhibitor GM6001 or the specific heparin-bound EGF-like growth factor inhibitor CRM197 suppressed RvE1-induced stimulation of EGFR/PI3-K/Akt phosphorylation and cell migration. CONCLUSIONS RvE1 enhances HCEC migration through MMP and sheddase-mediated EGFR transactivation. This response is dependent on PI3-K and p38-linked signaling eliciting paxillin (Tyr118) phosphorylation.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Biological Sciences, State University of New York, College of Optometry, New York, New York 10036, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Gilliver SC. Sex steroids as inflammatory regulators. J Steroid Biochem Mol Biol 2010; 120:105-15. [PMID: 20045727 DOI: 10.1016/j.jsbmb.2009.12.015] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 12/15/2009] [Accepted: 12/23/2009] [Indexed: 12/21/2022]
Abstract
It is becoming increasingly clear that endogenous sex steroids are key players in a range of inflammatory contexts. Androgens and estrogens have been shown to have a profound influence on the function of inflammatory cells including macrophages and on the secretion and activation of a range of plasma-borne inflammatory mediators. The menopause and polymorphisms in estrogen receptor genes have separately been shown to affect the incidence of a range of inflammatory disorders. Sex steroids themselves have been shown to be protective in certain conditions; harmful in others. This review will summarize their documented effects on inflammatory processes, with particular focus on two areas that have received much recent attention: the antiatherosclerotic properties of estrogens in females and the wound healing effects of sex steroids.
Collapse
|
48
|
Kenchegowda S, Bazan HEP. Significance of lipid mediators in corneal injury and repair. J Lipid Res 2010; 51:879-91. [PMID: 19965607 PMCID: PMC2853455 DOI: 10.1194/jlr.r001347] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Revised: 11/03/2009] [Indexed: 12/23/2022] Open
Abstract
Corneal injury induces an inflammatory reaction and damages the sensory nerves that exert trophic influences in the corneal epithelium. Alterations in normal healing disrupt the integrity and function of the tissue with undesirable consequences, ranging from dry eye and loss of transparency to ulceration and perforation. Lipids play important roles in this complex process. Whereas lipid mediators such as platelet activating factor (PAF) and cyclooxygenease-2 metabolites contribute to tissue damage and neovascularization, other mediators, such as the lipoxygenase (LOX) derivatives from arachidonic acid, 12- and 15-hydroxy/hydroperoxyeicosatetraenoic acids, and lipoxin A4, act as second messengers for epidermal growth factor to promote proliferation and repair. Stimulation of the cornea with pigment epithelial derived factor in the presence of docosahexaenoic acid gives rise to the synthesis of neuroprotectin D1, a derivative of LOX activity, and increases regeneration of corneal nerves. More knowledge about the role that lipids play in corneal wound healing can provide insight into the development of new therapeutic approaches for treating corneal injuries. PAF antagonists, lipoxins, and neuroprotectins can be effective therapeutic tools for maintaining the integrity of the cornea.
Collapse
Affiliation(s)
| | - Haydee E. P. Bazan
- Neuroscience Center of Excellence and Department of Ophthalmology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| |
Collapse
|
49
|
Esquenazi S, Bazan HEP. Role of platelet-activating factor in cell death signaling in the cornea: A review. Mol Neurobiol 2010; 42:32-8. [PMID: 20431963 DOI: 10.1007/s12035-010-8129-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 04/05/2010] [Indexed: 11/28/2022]
Abstract
Platelet-activating factor (PAF) is a potent bioactive lipid generated in the cornea after injury whose actions are mediated through specific receptors. Studies from our laboratory have shown that PAF interactions with its receptors activate several transmembrane signals involved in apoptosis. Continuous exposure to PAF during prolonged inflammation increases keratocyte apoptosis and inhibition of epithelial adhesion to the basement membrane. As a consequence, there is a marked delay in wound healing, which is not countered by the action of growth factors. While apoptosis of stroma cells is rapid and potent, epithelial cells as well as myofibroblasts, which appear in the stroma during the repair phase, are resistant to apoptosis. However, PAF accelerates apoptosis of corneal epithelial cells exposed to oxidative stress by stimulating phospholipase A2, producing an early release of cytochrome C from mitochondria and activating caspase-3. In myofibroblasts, PAF has a synergistic action with tumor necrosis factor-alpha (TNF-alpha), increasing apoptosis of the cells to 85%. PAF antagonists block the effects of PAF and could have a therapeutic role in maintaining a healthy and transparent cornea.
Collapse
Affiliation(s)
- Salomon Esquenazi
- Neuroscience Center of Excellence and Department of Ophthalmology, LSU Health Sciences Center, New Orleans, LA, USA
| | | |
Collapse
|
50
|
Safvati A, Cole N, Hume E, Willcox M. Mediators of neovascularization and the hypoxic cornea. Curr Eye Res 2009; 34:501-14. [PMID: 19899985 DOI: 10.1080/02713680902919557] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The maintenance of corneal avascularity is essential to vision. The mechanisms by which the cornea becomes vascularized in response to inflammation or hypoxic stress are beginning to be elucidated. A detailed understanding of the molecular responses of the cornea to hypoxia is critical for prevention and development of novel treatments for neovascularization in a range of disease states. Here, we have examined the current literature on the major mediators of angiogenesis, which have previously been reported during hypoxia in the cornea in order to better understand the mechanisms by which corneal angiogenesis occurs in circumstances where the available oxygen is reduced. The normal cornea produces angiogenic factors that are regulated by the production of anti-angiogenic molecules. The various cell types of the cornea respond differentially to inflammatory and hypoxic stimuli. An understanding of the factors that may predispose patients to development of corneal blood vessels may provide an opportunity to develop novel prophylactic strategies. The difficulties with extrapolating data from other cell types and animal models to the cornea are also examined.
Collapse
Affiliation(s)
- Aidin Safvati
- Vision Cooperative Research Centre and School of Optometry and Vision Science, The University of New South Wales, Sydney, NSW, Australia
| | | | | | | |
Collapse
|