1
|
Hu Z, Cano I, Lei F, Liu J, Bossardi Ramos R, Gordon H, Paschalis EI, Saint-Geniez M, Ng YSE, D'Amore PA. Loss of the Endothelial Glycocalyx Component EMCN Leads to Glomerular Impairment. Circ Res 2025; 136:59-74. [PMID: 39584795 DOI: 10.1161/circresaha.124.325218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND EMCN (endomucin), an endothelial-specific glycocalyx component, was found to be highly expressed by the endothelium of the renal glomerulus. We reported an anti-inflammatory role of EMCN and its involvement in the regulation of VEGF (vascular endothelial growth factor) activity through modulating VEGFR2 (VEGF receptor 2) endocytosis. The goal of this study is to investigate the phenotypic and functional effects of EMCN deficiency using the first global EMCN knockout mouse model. METHODS Global EMCN knockout mice were generated by crossing EMCN-floxed mice with ROSA26-Cre mice. Flow cytometry was used to analyze infiltrating myeloid cells in the kidneys. The ultrastructure of the glomerular filtration barrier was examined by transmission electron microscopy, whereas urinary albumin, creatinine, and total protein levels were analyzed from freshly collected urine samples. Expression and localization of EMCN, EGFP (enhanced green fluorescent protein), CD45 (cluster of differentiation 45), CD31, CD34, podocin, and albumin were examined by immunohistochemistry. Mice were weighed regularly, and their systemic blood pressure was measured using a noninvasive tail-cuff system. Glomerular endothelial cells and podocytes were isolated by fluorescence-activated cell sorting for RNA sequencing. Transcriptional profiles were analyzed to identify differentially expressed genes in both endothelium and podocytes, followed by gene ontology analysis. Protein levels of EMCN, albumin, and podocin were quantified by Western blot. RESULTS The EMCN-/- mice exhibited increased infiltration of CD45+ cells, with an increased proportion of Ly6GhighLy6Chigh myeloid cells and higher VCAM-1 (vascular cell adhesion molecule 1) expression. EMCN-/- mice displayed albuminuria with increased albumin in the Bowman's space compared with the EMCN+/+ littermates. Glomeruli in EMCN-/- mice revealed fused and effaced podocyte foot processes and disorganized endothelial fenestrations. We found no significant difference in blood pressure between EMCN knockout mice and their wild-type littermates. RNA sequencing of glomerular endothelial cells revealed downregulation of cell-cell adhesion and MAPK (mitogen-activated protein kinase)/ERK (extracellular signal-regulated kinase) pathways, along with glycocalyx and extracellular matrix remodeling. In podocytes, we observed reduced VEGF signaling and alterations in cytoskeletal organization. Notably, there was a significant decrease in both mRNA and protein levels of podocin, a key component of the slit diaphragm. CONCLUSION Our study demonstrates a critical role of the endothelial marker EMCN in supporting normal glomerular filtration barrier structure and function by maintaining glomerular endothelial tight junction and homeostasis and podocyte function through endothelial-podocyte crosstalk.
Collapse
Affiliation(s)
- Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Issahy Cano
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Fengyang Lei
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Jie Liu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Ramon Bossardi Ramos
- Department of Molecular and Cellular Physiology, Albany Medical Center, NY (R.B.R.)
| | - Harper Gordon
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Eleftherios I Paschalis
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Magali Saint-Geniez
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
- Now with Biomedical Research, Novartis, Cambridge, MA (M.S.-G.)
| | - Yin Shan Eric Ng
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
- Now with EyeBiotech Limited, a subsidiary of Merck & Co, Inc, Rahway, NJ (Y.S.E.N.)
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
- Department of Pathology (P.A.D.), Harvard Medical School, Boston, MA
| |
Collapse
|
2
|
Ronning KE, Burns ME, Sennlaub F. Monocytes in Retinal Degeneration: Little Cells with a Big Impact. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1468:133-137. [PMID: 39930185 DOI: 10.1007/978-3-031-76550-6_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Inflammation has been strongly implicated in retinal degenerative disorders, including inherited retinal degenerations (IRDs) and age-related macular degeneration (AMD). Microglia are the only immune cells in the retina during normal function, but during damage and disease, monocytes are able to invade the retina. Despite similarities to microglia, monocyte-derived cells (MdCs) may play a distinct and often pathogenic role in disease. Recent technological advances are rapidly improving our ability to investigate monocytic cells, yet many questions remain. Still, it is clear monocytes play an important role during retinal degenerative disorders and they are an exciting target for the development of therapeutic interventions.
Collapse
Affiliation(s)
- Kaitryn E Ronning
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, Paris, France.
| | - Marie E Burns
- Center for Neuroscience, University of California Davis, Davis, CA, USA
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA, USA
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, CA, USA
| | - Florian Sennlaub
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, Paris, France
| |
Collapse
|
3
|
Zhou C, Lei F, Mittermaier M, Dana R, Dohlman CH, Chodosh J, Paschalis EI. TNF-α Suppression Attenuates Limbal Stem Cell Damage in Ocular Injury. Cornea 2024; 44:762-771. [PMID: 39626088 PMCID: PMC12052064 DOI: 10.1097/ico.0000000000003738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 12/12/2024]
Abstract
PURPOSE Ocular chemical injuries often cause uveal inflammation, upregulation of TNF-α at the limbus, and subsequent limbal stem cell (LSC) damage. In this study, we investigate the protective role of TNF-α suppression in LSC survival. METHODS Corneal alkali injuries were performed using NaOH as previously described by our group. Anterior chamber pH elevation in the absence of corneal alkali exposure was achieved by cannulation. A CX3CR1 +/EGFP ::CCR2 +/RFP bone marrow chimera was used to study the role of innate immune cells in LSC damage, which was assessed by TUNEL assay, ABCB5, cytokeratin 12 and 13 staining, flow cytometry, in situ hybridization, and qPCR. Corneal neovascularization and conjunctivalization were evaluated by light microscopy. Intraperitoneal injection of 6.25 mg/kg infliximab was administered after irrigation. A TNFR1/2 knockout mouse was used to confirm the findings by a second method. RESULTS Systemic administration of 6.25 mg/kg infliximab suppressed uveal inflammation after anterior chamber pH elevation or corneal alkali injury and led to reduction of TNF-α secreting CCR2 + and CX3CR1 + monocytes in the basal limbal tissue. In turn, this led to LSC survival ( P < 0.01) and allowed reestablishment of K12 + epithelium ( P < 0.05) on the injured cornea. Moreover, it led to less corneal neovascularization, conjunctivalization, and scarring, as compared with untreated animals. The protective effect of TNF-α suppression was confirmed in TNFR1/2 knockout mice. CONCLUSIONS Prompt systemic administration of TNF-α inhibitor prevents LSC deficiency and facilitates corneal reepithelialization after alkali burn. TNF-α suppression may benefit the outcomes of other ocular injuries that cause LSC deficiency.
Collapse
Affiliation(s)
- Chengxin Zhou
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA
- Department of Ophthalmology, Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
- Department of Ophthalmology, Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA; and
| | - Fengyang Lei
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA
- Department of Ophthalmology, Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
- Department of Ophthalmology, Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA; and
| | - Mirja Mittermaier
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA
| | - Reza Dana
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA
| | - Claes H. Dohlman
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA
- Department of Ophthalmology, Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA
- Department of Ophthalmology, Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
- Department of Ophthalmology, Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA; and
- Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, NM
| | - Eleftherios I. Paschalis
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA
- Department of Ophthalmology, Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
- Department of Ophthalmology, Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA; and
| |
Collapse
|
4
|
Mohamed SH, Vanhoffelen E, Shun Fu M, Hei Lau P, Hain S, Seldeslachts L, Cosway E, Anderson G, McCulloch L, Vande Velde G, Drummond RA. CSF1R inhibition by PLX5622 reduces pulmonary fungal infection by depleting MHCII hi interstitial lung macrophages. Mucosal Immunol 2024; 17:1256-1272. [PMID: 39168451 DOI: 10.1016/j.mucimm.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Abstract
PLX5622 is a small molecular inhibitor of the CSF1 receptor (CSF1R) and is widely used to deplete macrophages within the central nervous system (CNS). We investigated the impact of PLX5622 treatment in wild-type C57BL/6 mice and discovered that one-week treatment with PLX5622 was sufficient to deplete interstitial macrophages in the lung and brain-infiltrating Ly6Clow patrolling monocytes, in addition to CNS-resident macrophages. These cell types were previously indicated to act as infection reservoirs for the pathogenic fungus Cryptococcus neoformans. We found that PLX5622-treated mice had significantly reduced fungal lung infection and reduced extrapulmonary dissemination to the CNS but not to the spleen or liver. Fungal lung infection mapped to MHCIIhi interstitial lung macrophages, which underwent significant expansion during infection following monocyte replenishment and not local division. Although PLX5622 depleted CNS infiltrating patrolling monocytes, these cells did not accumulate in the fungal-infected CNS following pulmonary infection. In addition, Nr4a1-deficient mice, which lack patrolling monocytes, had similar control and dissemination of C. neoformans infection to wild-type controls. PLX5622 did not directly affect CD4 T-cell responses, or significantly affect production of antibody in the lung during infection. However, we found that mice lacking lymphocytes had reduced numbers of MHCIIhi interstitial macrophages in the lung, which correlated with reduced infection load. Accordingly, PLX5622 treatment did not alter fungal burdens in the lungs of lymphocyte-deficient mice. Our data demonstrate that PLX5622 may help reduce lung burden of pathogenic fungi that utilise CSF1R-dependent myeloid cells as infection reservoirs, an effect which is dependent on the presence of lymphocytes.
Collapse
Affiliation(s)
- Sally H Mohamed
- Institute of Immunology & Immunotherapy, University of Birmingham, UK
| | - Eliane Vanhoffelen
- Department of Imaging and Pathology, Biomedical MRI/MoSAIC, KU Leuven, Leuven, Belgium
| | - Man Shun Fu
- Institute of Immunology & Immunotherapy, University of Birmingham, UK
| | - Pui Hei Lau
- Institute of Immunology & Immunotherapy, University of Birmingham, UK
| | - Sofia Hain
- Institute of Immunology & Immunotherapy, University of Birmingham, UK
| | - Laura Seldeslachts
- Department of Imaging and Pathology, Biomedical MRI/MoSAIC, KU Leuven, Leuven, Belgium
| | - Emilie Cosway
- Institute of Immunology & Immunotherapy, University of Birmingham, UK
| | - Graham Anderson
- Institute of Immunology & Immunotherapy, University of Birmingham, UK
| | - Laura McCulloch
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Greetje Vande Velde
- Department of Imaging and Pathology, Biomedical MRI/MoSAIC, KU Leuven, Leuven, Belgium
| | - Rebecca A Drummond
- Institute of Immunology & Immunotherapy, University of Birmingham, UK; Institute of Microbiology & Infection, University of Birmingham, UK.
| |
Collapse
|
5
|
Liu J, Lei F, Yan B, Cui N, Sharma J, Correa V, Roach L, Nicolaou S, Pitts K, Chodosh J, Maidana DE, Vavvas D, Margeta MA, Zhang H, Weitz D, Mostoslavsky R, Paschalis EI. Epigenetic adaptation drives monocyte differentiation into microglia-like cells upon engraftment into the retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612126. [PMID: 39314467 PMCID: PMC11419019 DOI: 10.1101/2024.09.09.612126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The identification of specific markers for microglia has been a long-standing challenge. Recently, markers such as P2ry12, TMEM119, and Fcrls have been proposed as microglia-specific and widely used to explore microglial functions within various central nervous system (CNS) contexts. The specificity of these markers was based on the assumption that circulating monocytes retain their distinct signatures even after infiltrating the CNS. However, recent findings reveal that infiltrating monocytes can adopt microglia-like characteristics while maintaining a pro-inflammatory profile upon permanent engraftment in the CNS.In this study, we utilize bone marrow chimeras, single-cell RNA sequencing, ATAC-seq, flow cytometry, and immunohistochemistry to demonstrate that engrafted monocytes acquire expression of established microglia markers-P2ry12, TMEM119, Fcrls-and the pan-myeloid marker Iba1, which has been commonly mischaracterized as microglia-specific. These changes are accompanied by alterations in chromatin accessibility and shifts in chromatin binding motifs that are indicative of microglial identity. Moreover, we show that engrafted monocytes dynamically regulate the expression of CX3CR1, CCR2, Ly6C, and transcription factors PU.1, CTCF, RUNX, AP-1, CEBP, and IRF2, all of which are crucial for shaping microglial identity. This study is the first to illustrate that engrafted monocytes in the retina undergo both epigenetic and transcriptional changes, enabling them to express microglia-like signatures. These findings highlight the need for future research to account for these changes when assessing the roles of monocytes and microglia in CNS pathology.
Collapse
Affiliation(s)
- Jie Liu
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Fengyang Lei
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Bin Yan
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Naiwen Cui
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA, USA
| | - Jyoti Sharma
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Victor Correa
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Lara Roach
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Savvas Nicolaou
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Kristen Pitts
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - James Chodosh
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Daniel E. Maidana
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL
| | - Demetrios Vavvas
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Milica A Margeta
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Huidan Zhang
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA, USA
| | - David Weitz
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA, USA
| | - Raul Mostoslavsky
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Eleftherios I. Paschalis
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
6
|
Hu Z, Cano I, Lei F, Liu J, Ramos RB, Gordon H, Paschalis EI, Saint-Geniez M, Ng YSE, D'Amore PA. Deletion of the endothelial glycocalyx component endomucin leads to impaired glomerular structure and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603749. [PMID: 39071302 PMCID: PMC11275787 DOI: 10.1101/2024.07.16.603749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background Endomucin (EMCN), an endothelial-specific glycocalyx component, was found to be highly expressed by the endothelium of the renal glomerulus. We reported an anti-inflammatory role of EMCN and its involvement in the regulation of vascular endothelial growth factor (VEGF) activity through modulating VEGF receptor 2 (VEGFR2) endocytosis. The goal of this study is to investigate the phenotypic and functional effects of EMCN deficiency using the first global EMCN knockout mouse model. Methods Global EMCN knockout mice were generated by crossing EMCN-floxed mice with ROSA26-Cre mice. Flow cytometry was employed to analyze infiltrating myeloid cells in the kidneys. The ultrastructure of the glomerular filtration barrier was examined by transmission electron microscopy, while urinary albumin, creatinine, and total protein levels were analyzed from freshly collected urine samples. Expression and localization of EMCN, EGFP, CD45, CD31, CD34, podocin, albumin, and α-smooth muscle actin were examined by immunohistochemistry. Mice were weighed regularly, and their systemic blood pressure was measured using a non-invasive tail-cuff system. Glomerular endothelial cells and podocytes were isolated by fluorescence-activated cell sorting for RNA-seq. Transcriptional profiles were analyzed to identify differentially expressed genes in both endothelium and podocytes, followed by gene ontology analysis of up- and down-regulated genes. Protein levels of EMCN, albumin, and podocin were quantified by Western blot. Results EMCN -/- mice were viable with no gross anatomical defects in kidneys. The EMCN -/- mice exhibited increased infiltration of CD45 + cells, with an increased proportion of Ly6G high Ly6C high myeloid cells and higher VCAM-1 expression. EMCN -/- mice displayed albuminuria with increased albumin in the Bowman's space compared to the EMCN +/+ littermates. Glomeruli in EMCN -/- mice revealed fused and effaced podocyte foot processes and disorganized endothelial fenestrations. We found no significant difference in blood pressure between EMCN knockout mice and their wild-type littermates. RNA-seq of glomerular endothelial cells revealed downregulation of cell-cell adhesion and MAPK/ERK pathways, along with glycocalyx and extracellular matrix remodeling. In podocytes, we observed reduced VEGF signaling and alterations in cytoskeletal organization. Notably, there was a significant decrease in both mRNA and protein levels of podocin, a key component of the slit diaphragm. Conclusion Our study demonstrates a critical role of the endothelial marker EMCN in supporting normal glomerular filtration barrier structure and function by maintaining glomerular endothelial tight junction and homeostasis and podocyte function through endothelial-podocyte crosstalk.
Collapse
|
7
|
Shahror RA, Shosha E, Morris C, Wild M, Mu S, Csanyi G, Boerma M, Rusch NJ, Fouda AY. Deletion of myeloid HDAC3 promotes efferocytosis to ameliorate retinal ischemic injury. J Neuroinflammation 2024; 21:170. [PMID: 38997746 PMCID: PMC11241909 DOI: 10.1186/s12974-024-03159-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Ischemia-induced retinopathy is a hallmark finding of common visual disorders including diabetic retinopathy (DR) and central retinal artery and vein occlusions. Treatments for ischemic retinopathies fail to improve clinical outcomes and the design of new therapies will depend on understanding the underlying disease mechanisms. Histone deacetylases (HDACs) are an enzyme class that removes acetyl groups from histone and non-histone proteins, thereby regulating gene expression and protein function. HDACs have been implicated in retinal neurovascular injury in preclinical studies in which nonspecific HDAC inhibitors mitigated retinal injury. Histone deacetylase 3 (HDAC3) is a class I histone deacetylase isoform that plays a central role in the macrophage inflammatory response. We recently reported that myeloid cells upregulate HDAC3 in a mouse model of retinal ischemia-reperfusion (IR) injury. However, whether this cellular event is an essential contributor to retinal IR injury is unknown. In this study, we explored the role of myeloid HDAC3 in ischemia-induced retinal neurovascular injury by subjecting myeloid-specific HDAC3 knockout (M-HDAC3 KO) and floxed control mice to retinal IR. The M-HDAC3 KO mice were protected from retinal IR injury as shown by the preservation of inner retinal neurons, vascular integrity, and retinal thickness. Electroretinography confirmed that this neurovascular protection translated to improved retinal function. The retinas of M-HDAC3 KO mice also showed less proliferation and infiltration of myeloid cells after injury. Interestingly, myeloid cells lacking HDAC3 more avidly engulfed apoptotic cells in vitro and after retinal IR injury in vivo compared to wild-type myeloid cells, suggesting that HDAC3 hinders the reparative phagocytosis of dead cells, a process known as efferocytosis. Further mechanistic studies indicated that although HDAC3 KO macrophages upregulate the reparative enzyme arginase 1 (A1) that enhances efferocytosis, the inhibitory effect of HDAC3 on efferocytosis is not solely dependent on A1. Finally, treatment of wild-type mice with the HDAC3 inhibitor RGFP966 ameliorated the retinal neurodegeneration and thinning caused by IR injury. Collectively, our data show that HDAC3 deletion enhances macrophage-mediated efferocytosis and protects against retinal IR injury, suggesting that inhibiting myeloid HDAC3 holds promise as a novel therapeutic strategy for preserving retinal integrity after ischemic insult.
Collapse
Affiliation(s)
- Rami A Shahror
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
| | - Esraa Shosha
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
- Clinical Pharmacy Department, Cairo University, Cairo, Egypt
| | - Carol Morris
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
| | - Melissa Wild
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
| | - Shengyu Mu
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
| | - Gabor Csanyi
- Department of Pharmacology and Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Marjan Boerma
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA
| | - Abdelrahman Y Fouda
- Department of Pharmacology and Toxicology College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, Office, AR, 72205, USA.
- Clinical Pharmacy Department, Cairo University, Cairo, Egypt.
| |
Collapse
|
8
|
Colella P, Sayana R, Suarez-Nieto MV, Sarno J, Nyame K, Xiong J, Pimentel Vera LN, Arozqueta Basurto J, Corbo M, Limaye A, Davis KL, Abu-Remaileh M, Gomez-Ospina N. CNS-wide repopulation by hematopoietic-derived microglia-like cells corrects progranulin deficiency in mice. Nat Commun 2024; 15:5654. [PMID: 38969669 PMCID: PMC11226701 DOI: 10.1038/s41467-024-49908-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 06/17/2024] [Indexed: 07/07/2024] Open
Abstract
Hematopoietic stem cell transplantation can deliver therapeutic proteins to the central nervous system (CNS) through transplant-derived microglia-like cells. However, current conditioning approaches result in low and slow engraftment of transplanted cells in the CNS. Here we optimized a brain conditioning regimen that leads to rapid, robust, and persistent microglia replacement without adverse effects on neurobehavior or hematopoiesis. This regimen combines busulfan myeloablation and six days of Colony-stimulating factor 1 receptor inhibitor PLX3397. Single-cell analyses revealed unappreciated heterogeneity of microglia-like cells with most cells expressing genes characteristic of homeostatic microglia, brain-border-associated macrophages, and unique markers. Cytokine analysis in the CNS showed transient inductions of myeloproliferative and chemoattractant cytokines that help repopulate the microglia niche. Bone marrow transplant of progranulin-deficient mice conditioned with busulfan and PLX3397 restored progranulin in the brain and eyes and normalized brain lipofuscin storage, proteostasis, and lipid metabolism. This study advances our understanding of CNS repopulation by hematopoietic-derived cells and demonstrates its therapeutic potential for treating progranulin-dependent neurodegeneration.
Collapse
Affiliation(s)
- Pasqualina Colella
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Ruhi Sayana
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | - Jolanda Sarno
- Hematology, Oncology, Stem Cell Transplant, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900, Monza, Italy
| | - Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, 94305, USA
| | - Jian Xiong
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, 94305, USA
| | | | | | - Marco Corbo
- MedGenome, Inc, 348 Hatch Dr, Foster City, CA, 94404, USA
| | - Anay Limaye
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- MedGenome, Inc, 348 Hatch Dr, Foster City, CA, 94404, USA
| | - Kara L Davis
- Hematology, Oncology, Stem Cell Transplant, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, 94305, USA
| | - Natalia Gomez-Ospina
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
9
|
Maidana DE, Gonzalez-Buendia L, Pastor-Puente S, Naqvi A, Paschalis E, Kazlauskas A, Miller JW, Vavvas DG. Peripheral monocytes and neutrophils promote photoreceptor cell death in an experimental retinal detachment model. Cell Death Dis 2023; 14:834. [PMID: 38102109 PMCID: PMC10724298 DOI: 10.1038/s41419-023-06350-6] [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: 06/19/2023] [Revised: 10/31/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023]
Abstract
Photoreceptor cell death and immune cell infiltration are two major events that contribute to retinal degeneration. However, the relationship between these two events has not been well delineated, primarily because of an inadequate understanding of the immunological processes involved in photoreceptor degeneration, especially that of peripheral leukocytes that infiltrate the subretinal space and retinal tissues. In this work, we characterized the role of leukocyte infiltration within the detached retina. We observed that CD45+ CD11b+ Ly6G+ neutrophils and CD45+ CD11b+ Ly6G- Ly6C+ monocytes are the predominant peripheral immune cell populations that infiltrate the retinal and subretinal space after detachment. Selective depletion of monocytes or neutrophils using cell-specific targeting is neuroprotective for photoreceptors. These results indicate that peripheral innate immune cells contribute to photoreceptor degeneration, and targeting these immune cell populations could be therapeutic during retinal detachment.
Collapse
Affiliation(s)
- Daniel E Maidana
- Retina Service, Angiogenesis Lab, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Lucia Gonzalez-Buendia
- Retina Service, Angiogenesis Lab, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Sara Pastor-Puente
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Afsar Naqvi
- Mucosal Immunology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Eleftherios Paschalis
- Retina Service, Angiogenesis Lab, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Andrius Kazlauskas
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Joan W Miller
- Retina Service, Angiogenesis Lab, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Demetrios G Vavvas
- Retina Service, Angiogenesis Lab, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Colella P, Sayana R, Suarez-Nieto MV, Sarno J, Nyame K, Xiong J, Vera LNP, Basurto JA, Corbo M, Limaye A, Davis KL, Abu-Remaileh M, Gomez-Ospina N. CNS Repopulation by Hematopoietic-Derived Microglia-Like Cells Corrects Progranulin deficiency. RESEARCH SQUARE 2023:rs.3.rs-3263412. [PMID: 37790525 PMCID: PMC10543302 DOI: 10.21203/rs.3.rs-3263412/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Hematopoietic stem cell transplantation can deliver therapeutic proteins to the CNS through donor-derived hematopoietic cells that become microglia-like cells. However, using standard conditioning approaches, hematopoietic stem cell transplantation is currently limited by low and slow engraftment of microglia-like cells. We report an efficient conditioning regimen based on Busulfan and a six-day course of microglia depletion using the colony-stimulating factor receptor 1 inhibitor PLX3397. Combining Busulfan-myeloablation and transient microglia depletion results in robust, rapid, and persistent microglia replacement by bone marrow-derived microglia-like cells throughout the CNS. Adding PLX3397 does not affect neurobehavior or has adverse effects on hematopoietic reconstitution. Through single-cell RNA sequencing and high-dimensional CyTOF mass cytometry, we show that microglia-like cells are a heterogeneous population and describe six distinct subpopulations. Though most bone-marrow-derived microglia-like cells can be classified as homeostatic microglia, their gene signature is a hybrid of homeostatic/embryonic microglia and border associated-macrophages. Busulfan-myeloablation and transient microglia depletion induce specific cytokines in the brain, ultimately combining myeloid proliferative and chemo-attractive signals that act locally to repopulate microglia from outside the niche. Importantly, this conditioning approach demonstrates therapeutic efficacy in a mouse model of GRN deficiency. Transplanting wild-type bone marrow into Grn-/- mice conditioned with Busulfan plus PLX3397 results in high engraftment of microglia-like cells in the brain and retina, restoring GRN levels and normalizing lipid metabolism.
Collapse
Affiliation(s)
- Pasqualina Colella
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305
| | - Ruhi Sayana
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305
| | | | - Jolanda Sarno
- Hematology, Oncology, Stem Cell Transplant, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, 94305
| | - Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
- Department of Genetics, Stanford University, Stanford, CA 94305
| | - Jian Xiong
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
- Department of Genetics, Stanford University, Stanford, CA 94305
| | | | | | - Marco Corbo
- MedGenome, Inc, 348 Hatch Dr, Foster City, CA 94404
| | - Anay Limaye
- MedGenome, Inc, 348 Hatch Dr, Foster City, CA 94404
| | - Kara Lynn Davis
- Hematology, Oncology, Stem Cell Transplant, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, 94305
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
- Department of Genetics, Stanford University, Stanford, CA 94305
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, USA
| | - Natalia Gomez-Ospina
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305
| |
Collapse
|
11
|
Lei F, Cui N, Zhou C, Chodosh J, Vavvas DG, Paschalis EI. CSF1R inhibition is not specific to innate immune cells but also affects T-helper cell differentiation independently of microglia depletion. RESEARCH SQUARE 2023:rs.3.rs-3308220. [PMID: 37720036 PMCID: PMC10503844 DOI: 10.21203/rs.3.rs-3308220/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Colony-stimulating factor 1 receptor (CSF1R) inhibition has been proposed as a specific method for microglia depletion. However, recent work revealed that in addition to microglia, CSF1R inhibition also affects other innate immune cells, such as peripheral monocytes and tissue-resident macrophages of the lung, liver, spleen, and peritoneum. Here, we show that this effect is not restricted to innate immune cells only but extends to the adaptive immune compartment. CSF1R inhibition alters the transcriptional profile of bone marrow cells that control T helper cell activation. In vivo or ex vivo inhibition of CSF1R profoundly changes the transcriptional profile of CD4+ cells and suppresses Th1 and Th2 differentiation in directionally stimulated and unstimulated cells and independently of microglia depletion. Given that T cells also contribute in CNS pathology, these effects may have practical implications in the interpretation of relevant experimental data.
Collapse
Affiliation(s)
- Fengyang Lei
- Massachusetts Eye and Ear, Harvard Medical School
| | | | | | | | | | | |
Collapse
|
12
|
Zhou C, Lei F, Mittermaier M, Ksander B, Dana R, Dohlman CH, Vavvas DG, Chodosh J, Paschalis EI. Opposing Roles of Blood-Borne Monocytes and Tissue-Resident Macrophages in Limbal Stem Cell Damage after Ocular Injury. Cells 2023; 12:2089. [PMID: 37626899 PMCID: PMC10453077 DOI: 10.3390/cells12162089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Limbal stem cell (LSC) deficiency is a frequent and severe complication after chemical injury to the eye. Previous studies have assumed this is mediated directly by the caustic agent. Here we show that LSC damage occurs through immune cell mediators, even without direct injury to LSCs. In particular, pH elevation in the anterior chamber (AC) causes acute uveal stress, the release of inflammatory cytokines at the basal limbal tissue, and subsequent LSC damage and death. Peripheral C-C chemokine receptor type 2 positive/CX3C motif chemokine receptor 1 negative (CCR2+ CX3CR1-) monocytes are the key mediators of LSC damage through the upregulation of tumor necrosis factor-alpha (TNF-α) at the limbus. In contrast to peripherally derived monocytes, CX3CR1+ CCR2- tissue-resident macrophages have a protective role, and their depletion prior to injury exacerbates LSC loss and increases LSC vulnerability to TNF-α-mediated apoptosis independently of CCR2+ cell infiltration into the tissue. Consistently, repopulation of the tissue by new resident macrophages not only restores the protective M2-like phenotype of macrophages but also suppresses LSC loss after exposure to inflammatory signals. These findings may have clinical implications in patients with LSC loss after chemical burns or due to other inflammatory conditions.
Collapse
Affiliation(s)
- Chengxin Zhou
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Fengyang Lei
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Mirja Mittermaier
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
| | - Bruce Ksander
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
| | - Reza Dana
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
| | - Claes H. Dohlman
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA
| | - Demetrios G. Vavvas
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA;
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, NM 87108, USA
| | - Eleftherios I. Paschalis
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
13
|
Zhou C, Lei F, Sharma J, Hui PC, Wolkow N, Dohlman CH, Vavvas DG, Chodosh J, Paschalis EI. Sustained Inhibition of VEGF and TNF-α Achieves Multi-Ocular Protection and Prevents Formation of Blood Vessels after Severe Ocular Trauma. Pharmaceutics 2023; 15:2059. [PMID: 37631272 PMCID: PMC10458495 DOI: 10.3390/pharmaceutics15082059] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
PURPOSE This study aimed to develop a clinically feasible and practical therapy for multi-ocular protection following ocular injury by using a thermosensitive drug delivery system (DDS) for sustained delivery of TNF-α and VEGF inhibitors to the eye. METHODS A thermosensitive, biodegradable hydrogel DDS (PLGA-PEG-PLGA triblock polymer) loaded with 0.7 mg of adalimumab and 1.4 mg of aflibercept was injected subconjunctivally into Dutch-belted pigmented rabbits after corneal alkali injury. Control rabbits received 2 mg of IgG-loaded DDS or 1.4 mg of aflibercept-loaded DDS. Animals were followed for 3 months and assessed for tolerability and prevention of corneal neovascularization (NV), improvement of corneal re-epithelialization, inhibition of retinal ganglion cell (RGC) and optic nerve axon loss, and inhibition of immune cell infiltration into the cornea. Drug-release kinetics was assessed in vivo using an aqueous humor protein analysis. RESULTS A single subconjunctival administration of dual anti-TNF-α/anti-VEGF DDS achieved a sustained 3-month delivery of antibodies to the anterior chamber, iris, ciliary body, and retina. Administration after corneal alkali burn suppressed CD45+ immune cell infiltration into the cornea, completely inhibited cornea NV for 3 months, accelerated corneal re-epithelialization and wound healing, and prevented RGC and optic nerve axon loss at 3 months. In contrast, anti-VEGF alone or IgG DDS treatment led to persistent corneal epithelial defect (combined: <1%; anti-VEGF: 15%; IgG: 10%, of cornea area), increased infiltration of CD45+ immune cells into the cornea (combined: 28 ± 20; anti-VEGF: 730 ± 178; anti-IgG: 360 ± 186, cells/section), and significant loss of RGCs (combined: 2.7%; anti-VEGF: 63%; IgG: 45%) and optic nerve axons at 3 months. The aqueous humor protein analysis showed first-order release kinetics without adverse effects at the injection site. CONCLUSIONS Concomitant inhibition of TNF-α and VEGF prevents corneal neovascularization and ameliorates subsequent irreversible damage to the retina and optic nerve after severe ocular injury. A single subconjunctival administration of this therapy, using a biodegradable, slow-release thermosensitive DDS, achieved the sustained elution of therapeutic levels of antibodies to all ocular tissues for 3 months. This therapeutic approach has the potential to dramatically improve the outcomes of severe ocular injuries in patients and improve the therapeutic outcomes in patients with retinal vascular diseases.
Collapse
Affiliation(s)
- Chengxin Zhou
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Fengyang Lei
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Jyoti Sharma
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Pui-Chuen Hui
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Natalie Wolkow
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- David G. Cogan Laboratory of Eye Pathology and Ophthalmic Plastic Surgery Service, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Claes H. Dohlman
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Demetrios G. Vavvas
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Angiogenesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - James Chodosh
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, NM 87108, USA
| | - Eleftherios I. Paschalis
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| |
Collapse
|
14
|
Maidana DE, Gonzalez-Buendia L, Miller JW, Vavvas DG. RIPK necrotic cell death pathway in both donor photoreceptor and host immune cells synergize to affect photoreceptor graft survival. FASEB J 2023; 37:e22847. [PMID: 36862516 PMCID: PMC10590064 DOI: 10.1096/fj.202201137r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 11/29/2022] [Accepted: 02/15/2023] [Indexed: 03/03/2023]
Abstract
Photoreceptor transplant has been put forward as a repair strategy to tackle degenerated retinas. Nonetheless, cell death and immune rejection seriously limit the success of this strategy, with only a small fraction of transplanted cells surviving. Improving the survival of transplanted cells is of critical importance. Recent evidence has identified receptor-interacting protein kinase 3 (RIPK3) as a molecular trigger controlling necroptotic cell death and inflammation. However, its role in photoreceptor transplantation and regenerative medicine has not been studied. We hypothesized that modulation of RIPK3 to address both cell death and immunity could have advantageous effects on photoreceptor survival. In a model of inherited retinal degeneration, deletion of RIPK3 in donor photoreceptor precursors significantly increases the survival of transplanted cells. Simultaneous RIPK3 deletion in donor photoreceptors and recipients maximizes graft survival. Lastly, to discern the role of RIPK3 in the host immune response, bone marrow transplant experiments demonstrated that peripheral immune cell RIPK3 deficiency is protective for both donor and host photoreceptor survival. Interestingly, this finding is independent of photoreceptor transplantation, as the peripheral protective effect is also observed in an additional retinal detachment photoreceptor degeneration model. Altogether, these results indicate that immunomodulatory and neuroprotective strategies targeting the RIPK3 pathway can aid regenerative therapies of photoreceptor transplantation.
Collapse
Affiliation(s)
- Daniel E. Maidana
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lucia Gonzalez-Buendia
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Joan W. Miller
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Demetrios G. Vavvas
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
15
|
Pitts KM, Margeta MA. Myeloid masquerade: Microglial transcriptional signatures in retinal development and disease. Front Cell Neurosci 2023; 17:1106547. [PMID: 36779012 PMCID: PMC9909491 DOI: 10.3389/fncel.2023.1106547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
Microglia are dynamic guardians of neural tissue and the resident immune cells of the central nervous system (CNS). The disease-associated microglial signature (DAM), also known as the microglial neurodegenerative phenotype (MGnD), has gained significant attention in recent years as a fundamental microglial response common to various neurodegenerative disease pathologies. Interestingly, this signature shares many features in common with developmental microglia, suggesting the existence of recycled gene programs which play a role both in early neural circuit formation as well as in response to aging and disease. In addition, recent advances in single cell RNA sequencing have revealed significant heterogeneity within the original DAM signature, with contributions from both yolk sac-derived microglia as well as bone marrow-derived macrophages. In this review, we examine the role of the DAM signature in retinal development and disease, highlighting crosstalk between resident microglia and infiltrating monocytes which may critically contribute to the underlying mechanisms of age-related neurodegeneration.
Collapse
Affiliation(s)
- Kristen M. Pitts
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
- Schepens Eye Research Institute of Mass, Eye and Ear, Boston, MA, United States
| | - Milica A. Margeta
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
- Schepens Eye Research Institute of Mass, Eye and Ear, Boston, MA, United States
| |
Collapse
|
16
|
Ronning KE, Karlen SJ, Burns ME. Structural and functional distinctions of co-resident microglia and monocyte-derived macrophages after retinal degeneration. J Neuroinflammation 2022; 19:299. [PMID: 36510226 PMCID: PMC9743742 DOI: 10.1186/s12974-022-02652-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Both resident microglia and invading peripheral immune cells can respond to injury and degeneration in the central nervous system. However, after dead and dying neurons have been cleared and homeostasis is re-established, it is unknown whether resident immune cells fully resume normal functions and to what degree the peripheral immune cells take up residence. METHODS Using flow cytometry, in vivo retinal imaging, immunohistochemistry, and single-cell mRNA sequencing, we assess resident microglia and monocyte-derived macrophages in the retina during and after the loss of photoreceptors in the Arr1-/- mouse model of inducible degeneration. RESULTS We find that photoreceptor loss results in a small, sustained increase in mononuclear phagocytes and, after degeneration wanes, these cells re-establish a spatial mosaic reminiscent of healthy retinas. Transcriptomic analysis revealed the population remained unusually heterogeneous, with several subpopulations expressing gene patterns consistent with mildly activated phenotypes. Roughly a third of "new resident" cells expressed markers traditionally associated with both microglial and monocytic lineages, making their etiology ambiguous. Using an inducible Cre-based fluorescent lineage tracing paradigm to confirm the origins of new resident immune cells, we found approximately equal numbers of microglia and monocyte-derived macrophages after degeneration had subsided. In vivo retinal imaging and immunohistochemical analysis showed that both subpopulations remained functionally responsive to sites of local damage, though on average the monocyte-derived cells had less morphological complexity, expressed higher levels of MHCII, and had less migratory activity than the native resident population. CONCLUSIONS Monocytic cells that infiltrate the retina during degeneration differentiate into monocyte-derived macrophages that can remain in the retina long-term. These monocyte-derived macrophages adopt ramified morphologies and microglia-like gene expression. However, they remain distinguishable in morphology and gene expression from resident microglia and appear to differ functionally, showing less responsiveness to subsequent retinal injuries. These findings support the idea that persistent changes in the local immune population that occur in response to cell loss in aging and progressive retinal diseases may include the establishment of subpopulations of bone marrow-derived cells whose ability to respond to subsequent insults wanes over time.
Collapse
Affiliation(s)
- Kaitryn E Ronning
- Center for Neuroscience, University of California, 1544 Newton Court, Davis, CA, 95618, USA
| | - Sarah J Karlen
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95616, USA
| | - Marie E Burns
- Center for Neuroscience, University of California, 1544 Newton Court, Davis, CA, 95618, USA. .,Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95616, USA. .,Department of Ophthalmology & Vision Science, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
17
|
McPherson SW, Heuss ND, Abedin M, Roehrich H, Pierson MJ, Gregerson DS. Parabiosis reveals the correlation between the recruitment of circulating antigen presenting cells to the retina and the induction of spontaneous autoimmune uveoretinitis. J Neuroinflammation 2022; 19:295. [PMID: 36494807 PMCID: PMC9733026 DOI: 10.1186/s12974-022-02660-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Characterizing immune cells and conditions that govern their recruitment and function in autoimmune diseases of the nervous system or in neurodegenerative processes is an area of active investigation. We sought to analyze the origin of antigen presenting cells associated with the induction of retinal autoimmunity using a system that relies on spontaneous autoimmunity, thus avoiding uncertainties associated with immunization with adjuvants at remotes sites or adoptive transfer of in vitro activated T cells. METHODS R161H mice (B10.RIII background), which spontaneously and rapidly develop severe spontaneous autoimmune uveoretinitis (SAU), were crossed to CD11cDTR/GFP mice (B6/J) allowing us to track the recruitment to and/or expansion within the retina of activated, antigen presenting cells (GFPhi cells) in R161H+/- × CD11cDTR/GFP F1 mice relative to the course of SAU. Parabiosis between R161H+/- × CD11cDTR/GFP F1 mice and B10.RIII × B6/J F1 (wild-type recipient) mice was done to explore the origin and phenotype of antigen presenting cells crucial for the induction of autoimmunity. Analysis was done by retinal imaging, flow cytometry, and histology. RESULTS Onset of SAU in R161H+/- × CD11cDTR/GFP F1 mice was delayed relative to B10.RIII-R161H+/- mice revealing a disease prophase prior to frank autoimmunity that was characterized by expansion of GFPhi cells within the retina prior to any clinical or histological evidence of autoimmunity. Parabiosis between mice carrying the R161H and CD11cDTR/GFP transgenes and transgene negative recipients showed that recruitment of circulating GFPhi cells into retinas was highly correlative with the occurrence of SAU. CONCLUSIONS Our results here contrast with our previous findings showing that retinal antigen presenting cells expanding in response to either sterile mechanical injury or neurodegeneration were derived from myeloid cells within the retina or optic nerve, thus highlighting a unique facet of retinal autoimmunity.
Collapse
Affiliation(s)
- Scott W. McPherson
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| | - Neal D. Heuss
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| | - Md. Abedin
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| | - Heidi Roehrich
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| | - Mark J. Pierson
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| | - Dale S. Gregerson
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| |
Collapse
|
18
|
Abstract
Millions of people worldwide are bilaterally blind due to corneal diseases including infectious etiologies, trauma, and chemical injuries. While corneal transplantation can successfully restore sight in many, corneal graft survival decreases in eyes with chronic inflammation and corneal vascularization. Additionally, the availability of donor cornea material can be limited, especially in underdeveloped countries where corneal blindness may also be highly prevalent. Development of methods to create and implant an artificial cornea (keratoprosthesis)may be the only option for patients whose eye disease is not suitable for corneal transplantation or who live in regions where corneal transplantation is not possible. The Boston keratoprosthesis (B-KPro) is the most commonly implanted keratoprosthesis worldwide, having restored vision in thousands of patients. This article describes the initial design of the B-KPro and the modifications that have been made over many years. Additionally, some of the complications of surgical implantation and long-term care challenges, particularly complicating inflammation and glaucoma, are discussed. Expected final online publication date for the Annual Review of Vision Science, Volume 8 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Claes Dohlman
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
19
|
Neuroprotection for Age-Related Macular Degeneration. OPHTHALMOLOGY SCIENCE 2022; 2:100192. [PMID: 36570623 PMCID: PMC9767822 DOI: 10.1016/j.xops.2022.100192] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 12/27/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness worldwide. Early to intermediate AMD is characterized by the accumulation of lipid- and protein-rich drusen. Late stages of the disease are characterized by the development of choroidal neovascularization, termed "exudative" or "neovascular AMD," or retinal pigment epithelium (RPE) cell and photoreceptor death, termed "geographic atrophy" (GA) in advanced nonexudative AMD. Although we have effective treatments for exudative AMD in the form of anti-VEGF agents, they have no role for patients with GA. Neuroprotection strategies have emerged as a possible way to slow photoreceptor degeneration and vision loss in patients with GA. These approaches include reduction of oxidative stress, modulation of the visual cycle, reduction of toxic molecules, inhibition of pathologic protein activity, prevention of cellular apoptosis or programmed necrosis (necroptosis), inhibition of inflammation, direct activation of neurotrophic factors, delivery of umbilical tissue-derived cells, and RPE replacement. Despite active investigation in this area and significant promise based on preclinical studies, many clinical studies have not yielded successful results. We discuss selected past and current neuroprotection trials for AMD, highlight the lessons learned from these past studies, and discuss our perspective regarding remaining questions that must be answered before neuroprotection can be successfully applied in the field of AMD research.
Collapse
Key Words
- AD, Alzheimer disease
- ALA, alpha lipoic acid
- AMD, age-related macular degeneration
- AREDS, Age-Related Eye Disease Study
- AREDS2, Age-Related Eye Disease Study 2
- Age-related macular degeneration
- CFH, complement factor H
- CNTF, ciliary neurotrophic factor
- GA, geographic atrophy
- HTRA1, high-temperature requirement A1
- IOP, intraocular pressure
- Neuroprotection
- RBP, retinol-binding protein
- RGC, retinal ganglion cell
- RIPK3, receptor-interacting serine/threonine-protein kinase 3
- ROS, reactive oxygen species
- RPE, retinal pigment epithelium
- Retinal degeneration
- VA, visual acuity
- iPSC, induced pluripotent stem cell
Collapse
|
20
|
Gong L, Gu Y, Han X, Luan C, Liu C, Wang X, Sun Y, Zheng M, Fang M, Yang S, Xu L, Sun H, Yu B, Gu X, Zhou S. Spatiotemporal Dynamics of the Molecular Expression Pattern and Intercellular Interactions in the Glial Scar Response to Spinal Cord Injury. Neurosci Bull 2022; 39:213-244. [PMID: 35788904 PMCID: PMC9905408 DOI: 10.1007/s12264-022-00897-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022] Open
Abstract
Nerve regeneration in adult mammalian spinal cord is poor because of the lack of intrinsic regeneration of neurons and extrinsic factors - the glial scar is triggered by injury and inhibits or promotes regeneration. Recent technological advances in spatial transcriptomics (ST) provide a unique opportunity to decipher most genes systematically throughout scar formation, which remains poorly understood. Here, we first constructed the tissue-wide gene expression patterns of mouse spinal cords over the course of scar formation using ST after spinal cord injury from 32 samples. Locally, we profiled gene expression gradients from the leading edge to the core of the scar areas to further understand the scar microenvironment, such as neurotransmitter disorders, activation of the pro-inflammatory response, neurotoxic saturated lipids, angiogenesis, obstructed axon extension, and extracellular structure re-organization. In addition, we described 21 cell transcriptional states during scar formation and delineated the origins, functional diversity, and possible trajectories of subpopulations of fibroblasts, glia, and immune cells. Specifically, we found some regulators in special cell types, such as Thbs1 and Col1a2 in macrophages, CD36 and Postn in fibroblasts, Plxnb2 and Nxpe3 in microglia, Clu in astrocytes, and CD74 in oligodendrocytes. Furthermore, salvianolic acid B, a blood-brain barrier permeation and CD36 inhibitor, was administered after surgery and found to remedy fibrosis. Subsequently, we described the extent of the scar boundary and profiled the bidirectional ligand-receptor interactions at the neighboring cluster boundary, contributing to maintain scar architecture during gliosis and fibrosis, and found that GPR37L1_PSAP, and GPR37_PSAP were the most significant gene-pairs among microglia, fibroblasts, and astrocytes. Last, we quantified the fraction of scar-resident cells and proposed four possible phases of scar formation: macrophage infiltration, proliferation and differentiation of scar-resident cells, scar emergence, and scar stationary. Together, these profiles delineated the spatial heterogeneity of the scar, confirmed the previous concepts about scar architecture, provided some new clues for scar formation, and served as a valuable resource for the treatment of central nervous system injury.
Collapse
Affiliation(s)
- Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaoxiao Han
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chengcheng Luan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yufeng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengru Zheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengya Fang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shuhai Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Lai Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
21
|
Intravenous route to choroidal neovascularization by macrophage-disguised nanocarriers for mTOR modulation. Acta Pharm Sin B 2022; 12:2506-2521. [PMID: 35646523 PMCID: PMC9136612 DOI: 10.1016/j.apsb.2021.10.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/20/2021] [Accepted: 10/15/2021] [Indexed: 11/22/2022] Open
Abstract
Retinal pigment epithelial (RPE) is primarily impaired in age-related macular degeneration (AMD), leading to progressive loss of photoreceptors and sometimes choroidal neovascularization (CNV). mTOR has been proposed as a promising therapeutic target, while the usage of its specific inhibitor, rapamycin, was greatly limited. To mediate the mTOR pathway in the retina by a noninvasive approach, we developed novel biomimetic nanocomplexes where rapamycin-loaded nanoparticles were coated with cell membrane derived from macrophages (termed as MRaNPs). Taking advantage of the macrophage-inherited property, intravenous injection of MRaNPs exhibited significantly enhanced accumulation in the CNV lesions, thereby increasing the local concentration of rapamycin. Consequently, MRaNPs effectively downregulated the mTOR pathway and attenuate angiogenesis in the eye. Particularly, MRaNPs also efficiently activated autophagy in the RPE, which was acknowledged to rescue RPE in response to deleterious stimuli. Overall, we design and prepare macrophage-disguised rapamycin nanocarriers and demonstrate the therapeutic advantages of employing biomimetic cell membrane materials for treatment of AMD.
Collapse
|
22
|
Pichi F, Neri P, Hay S, Parrulli S, Zicarelli F, Invernizzi A. An en face swept source optical coherence tomography study of the vitreous in eyes with anterior uveitis. Acta Ophthalmol 2022; 100:e820-e826. [PMID: 34219383 DOI: 10.1111/aos.14965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/17/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE To analyse vitreous anatomy patients with anterior uveitis (AU) using swept source optical coherence tomography (SS-OCT). METHODS Patients with newly diagnosed AU and healthy participants were scanned using 16-mm SS-OCT B-scans and 12 × 12 mm cube centred at the fovea. Linear SS-OCT scans were evaluated to identify the premacular bursa (PB) above the macula and the Cloquet's canal above the optic disc. A dynamic evaluation of the 12 × 12 mm cube enabled en face measurement of the vertical and horizontal diameters of the PB and the presence/absence of communication between the PB and Cloquet's canal. RESULTS One hundred and thirty-five subjects were included in the study. Forty-five patients were healthy (mean age 33.47 years). En face measurements of the horizontal and vertical PB mean diameters were 5.2 mm and 4.7 mm. In 90 subjects with AU (mean age 30.10 years), the mean horizontal and vertical diameter of the PB were significantly larger (p < 0.01). Thirty-seven eyes with AU presented with cystoid macular oedema (CME). The mean diameters of the PB in this subgroup were significantly larger (p < 0.01). In 75% of the entire cohort (51% of eyes in the control group, 87% of eyes with AU and 100% of eyes with AU and CME), a channel connecting the PB and Cloquet's canal could be identified. CONCLUSIONS SS-OCT identification of a connecting channel between the PB and the Cloquet's canal suggests that inflammatory cytokines may drain from the anterior chamber through this system of channels, thus increasing the risk of CME.
Collapse
Affiliation(s)
- Francesco Pichi
- Cleveland Clinic Abu Dhabi Eye Institute Abu Dhabi UAE
- Cleveland Clinic Lerner College of Medicine Case Western Reserve University Cleveland OH USA
| | - Piergiorgio Neri
- Cleveland Clinic Abu Dhabi Eye Institute Abu Dhabi UAE
- Cleveland Clinic Lerner College of Medicine Case Western Reserve University Cleveland OH USA
| | - Steven Hay
- Cleveland Clinic Abu Dhabi Eye Institute Abu Dhabi UAE
| | - Salvatore Parrulli
- Eye Clinic Department of Biomedical and Clinical Science 'Luigi Sacco' Luigi Sacco Hospital University of Milan Milan Italy
| | - Federico Zicarelli
- Eye Clinic Department of Biomedical and Clinical Science 'Luigi Sacco' Luigi Sacco Hospital University of Milan Milan Italy
| | - Alessandro Invernizzi
- Eye Clinic Department of Biomedical and Clinical Science 'Luigi Sacco' Luigi Sacco Hospital University of Milan Milan Italy
- Save Sight Institute University of Sydney Sydney NSW Australia
| |
Collapse
|
23
|
Colony-stimulating factor 1 receptor signaling in the central nervous system and the potential of its pharmacological inhibitors to halt the progression of neurological disorders. Inflammopharmacology 2022; 30:821-842. [PMID: 35290551 DOI: 10.1007/s10787-022-00958-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/24/2022] [Indexed: 02/07/2023]
Abstract
Colony Stimulating Factor-1 (CSF-1)/Colony Stimulating Factor-1 Receptor (CSF-1R) signaling axis plays an essential role in the development, maintenance, and proliferation of macrophage lineage cells. Within the central nervous system, CSF-1R signaling primarily maintains microglial homeostasis. Microglia, being the resident macrophage and first responder to any neurological insults, plays critical importance in overall health of the human brain. Aberrant and sustained activation of microglia along with continued proliferation and release of neurotoxic proinflammatory cytokines have been reported in various neurological and neurodegenerative diseases. Therefore, halting the neuroinflammatory pathway via targeting microglial proliferation, which depends on CSF-1R signaling, has emerged as a potential therapeutic target for neurological disorders. However, apart from regulating the microglial function, recently it has been discovered that CSF-1R has much broader role in central nervous system. These findings limit the therapeutic utility of CSF-1R inhibitors but also highlight the need for a complete understanding of CSF-1R function within the central nervous system. Moreover, it has been found that selective inhibitors of CSF-1R may be more efficient in avoiding non-specific targeting and associated side effects. Short-term depletion of microglial population in diseased conditions have also been found to be beneficial; however, the dose and therapeutic window for optimum effects may need to be standardized further.This review summarizes the present understanding of CSF-1R function within the central nervous system. We discuss the CSF-1R signaling in the context of microglia function, crosstalk between microglia and astroglia, and regulation of neuronal cell function. We also discuss a few of the neurological disorders with a focus on the utility of CSF-1R inhibitors as potential therapeutic strategy for halting the progression of neurological diseases.
Collapse
|
24
|
Funatsu J, Murakami Y, Shimokawa S, Nakatake S, Fujiwara K, Okita A, Fukushima M, Shibata K, Yoshida N, Koyanagi Y, Akiyama M, Notomi S, Nakao S, Hisatomi T, Takeda A, Paschalis EI, Vavvas DG, Ikeda Y, Sonoda KH. Circulating inflammatory monocytes oppose microglia and contribute to cone cell death in retinitis pigmentosa. PNAS NEXUS 2022; 1. [PMID: 35529318 DOI: 10.1093/pnasnexus/pgac003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Retinitis pigmentosa (RP) is an intractable inherited disease that primarily affects the rods through gene mutations followed by secondary cone degeneration. This cone-related dysfunction can lead to impairment of daily life activities, and ultimately blindness in patients with RP. Paradoxically, microglial neuroinflammation contributes to both protection against and progression of RP, but it is unclear which population(s) - tissue-resident microglia and/or peripheral monocyte-derived macrophages (mφ) - are implicated in the progression of the disease. Here we show that circulating blood inflammatory monocytes (IMo) are key effector cells that mediate cone cell death in RP. Attenuation of IMo and peripherally engrafted mφ by Ccl2 deficiency or immune modulation via intravenous nano-particle treatment suppressed cone cell death in rd10 mice, an animal model of RP. In contrast, the depletion of resident microglia by a colony-stimulating factor 1 receptor inhibitor exacerbated cone cell death in the same model. In human patients with RP, IMo was increased and correlated with disease progression. These results suggest that peripheral IMo is a potential target to delay cone cell death and prevent blindness in RP.
Collapse
Affiliation(s)
- Jun Funatsu
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Shotaro Shimokawa
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Shunji Nakatake
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Kohta Fujiwara
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Ayako Okita
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Masatoshi Fukushima
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Kensuke Shibata
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan.,Department of Genomics and Molecular Analysis, Yamaguchi University School of Medicine, Yamaguchi 755-8505, Japan
| | - Noriko Yoshida
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan.,Department of Ophthalmology, Fukuoka Dental College Medical and Dental Hospital, Fukuoka 814-0193, Japan
| | - Yoshito Koyanagi
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Masato Akiyama
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan.,Department of Ocular Pathology and Imaging Science, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Shoji Notomi
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Shintaro Nakao
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Toshio Hisatomi
- Department of Ophthalmology, Chikushi Hospital, Fukuoka University, Fukuoka 818-8502, Japan
| | - Atsunobu Takeda
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Eleftherios I Paschalis
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA.,Boston Keratoprosthesis Laboratory, Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA.,Disruptive Technology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Demetrios G Vavvas
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA.,Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Yasuhiro Ikeda
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan.,Department of Ophthalmology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
25
|
Cuadros MA, Sepulveda MR, Martin-Oliva D, Marín-Teva JL, Neubrand VE. Microglia and Microglia-Like Cells: Similar but Different. Front Cell Neurosci 2022; 16:816439. [PMID: 35197828 PMCID: PMC8859783 DOI: 10.3389/fncel.2022.816439] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Microglia are the tissue-resident macrophages of the central nervous parenchyma. In mammals, microglia are thought to originate from yolk sac precursors and posteriorly maintained through the entire life of the organism. However, the contribution of microglial cells from other sources should also be considered. In addition to “true” or “bona-fide” microglia, which are of embryonic origin, the so-called “microglia-like cells” are hematopoietic cells of bone marrow origin that can engraft the mature brain mainly under pathological conditions. These cells implement great parts of the microglial immune phenotype, but they do not completely adopt the “true microglia” features. Because of their pronounced similarity, true microglia and microglia-like cells are usually considered together as one population. In this review, we discuss the origin and development of these two distinct cell types and their differences. We will also review the factors determining the appearance and presence of microglia-like cells, which can vary among species. This knowledge might contribute to the development of therapeutic strategies aiming at microglial cells for the treatment of diseases in which they are involved, for example neurodegenerative disorders like Alzheimer’s and Parkinson’s diseases.
Collapse
Affiliation(s)
- Miguel A Cuadros
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - M Rosario Sepulveda
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - David Martin-Oliva
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - José L Marín-Teva
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - Veronika E Neubrand
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| |
Collapse
|
26
|
Sanchez JMS, DePaula-Silva AB, Doty DJ, Hanak TJ, Truong A, Libbey JE, Fujinami RS. The CSF1R-Microglia Axis Has Protective Host-Specific Roles During Neurotropic Picornavirus Infection. Front Immunol 2021; 12:621090. [PMID: 34566948 PMCID: PMC8458822 DOI: 10.3389/fimmu.2021.621090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 08/16/2021] [Indexed: 01/19/2023] Open
Abstract
Viral encephalitis is a major cause of morbidity and mortality, but the manifestation of disease varies greatly between individuals even in response to the same virus. Microglia are professional antigen presenting cells that reside in the central nervous system (CNS) parenchyma that are poised to respond to viral insults. However, the role of microglia in initiating and coordinating the antiviral response is not completely understood. Utilizing Theiler's murine encephalomyelitis virus (TMEV), a neurotropic picornavirus, and PLX5622, a small molecule inhibitor of colony-stimulating factor 1 receptor (CSF1R) signaling that can deplete microglia in the CNS; we investigated the role of the CSF1R-microglia axis in neurotropic picornavirus infection of C57BL/6J and SJL/J mice. These mouse strains differ in their ability to clear TMEV and exhibit different neurological disease in response to TMEV infection. CSF1R antagonism in C57BL/6J mice, which normally clear TMEV in the CNS, led to acute fatal encephalitis. In contrast, CSF1R antagonism in SJL/J mice, which normally develop a chronic CNS TMEV infection, did not result in acute encephalitis, but exacerbated TMEV-induced demyelination. Immunologically, inhibition of CSF1R in C57BL/6J mice reduced major histocompatibility complex II expression in microglia, decreased the proportion of regulatory T cells in the CNS, and upregulated proinflammatory pathways in CNS T cells. Acute CSF1R inhibition in SJL/J mice had no effect on microglial MHC-II expression and upregulated anti-inflammatory pathways in CNS T cells, however chronic CSF1R inhibition resulted in broad immunosuppression. Our results demonstrate strain-specific effects of the CSF1R-microglia axis in the context of neurotropic viral infection as well as inherent differences in microglial antigen presentation and subsequent T cell crosstalk that contribute to susceptibility to neurotropic picornavirus infection.
Collapse
Affiliation(s)
- John Michael S. Sanchez
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | | | - Daniel J. Doty
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Tyler J. Hanak
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Amanda Truong
- Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Jane E. Libbey
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Robert S. Fujinami
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
27
|
Hohsfield LA, Najafi AR, Ghorbanian Y, Soni N, Crapser J, Figueroa Velez DX, Jiang S, Royer SE, Kim SJ, Henningfield CM, Anderson A, Gandhi SP, Mortazavi A, Inlay MA, Green KN. Subventricular zone/white matter microglia reconstitute the empty adult microglial niche in a dynamic wave. eLife 2021; 10:66738. [PMID: 34423781 PMCID: PMC8425950 DOI: 10.7554/elife.66738] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 08/22/2021] [Indexed: 02/06/2023] Open
Abstract
Microglia, the brain’s resident myeloid cells, play central roles in brain defense, homeostasis, and disease. Using a prolonged colony-stimulating factor 1 receptor inhibitor (CSF1Ri) approach, we report an unprecedented level of microglial depletion and establish a model system that achieves an empty microglial niche in the adult brain. We identify a myeloid cell that migrates from the subventricular zone and associated white matter areas. Following CSF1Ri, these amoeboid cells migrate radially and tangentially in a dynamic wave filling the brain in a distinct pattern, to replace the microglial-depleted brain. These repopulating cells are enriched in disease-associated microglia genes and exhibit similar phenotypic and transcriptional profiles to white-matter-associated microglia. Our findings shed light on the overlapping and distinct functional complexity and diversity of myeloid cells of the CNS and provide new insight into repopulating microglia function and dynamics in the mouse brain.
Collapse
Affiliation(s)
- Lindsay A Hohsfield
- Department of Neurobiology and Behavior, Irvine, United States.,Institute for Memory Impairments and Neurological Disorders, Irvine, United States
| | - Allison R Najafi
- Department of Neurobiology and Behavior, Irvine, United States.,Institute for Memory Impairments and Neurological Disorders, Irvine, United States
| | - Yasamine Ghorbanian
- Sue and Bill Gross Stem Cell Research Center, Irvine, United States.,Department of Molecular Biology and Biochemistry, Irvine, United States
| | - Neelakshi Soni
- Department of Neurobiology and Behavior, Irvine, United States.,Institute for Memory Impairments and Neurological Disorders, Irvine, United States
| | - Joshua Crapser
- Department of Neurobiology and Behavior, Irvine, United States.,Institute for Memory Impairments and Neurological Disorders, Irvine, United States
| | | | - Shan Jiang
- Department of Developmental and Cell Biology, Irvine, United States
| | - Sarah E Royer
- Department of Neurobiology and Behavior, Irvine, United States.,Sue and Bill Gross Stem Cell Research Center, Irvine, United States.,Department of Anatomy and Neurobiology, Irvine, United States
| | - Sung Jin Kim
- Department of Neurobiology and Behavior, Irvine, United States.,Institute for Memory Impairments and Neurological Disorders, Irvine, United States
| | - Caden M Henningfield
- Department of Neurobiology and Behavior, Irvine, United States.,Institute for Memory Impairments and Neurological Disorders, Irvine, United States
| | - Aileen Anderson
- Department of Neurobiology and Behavior, Irvine, United States.,Institute for Memory Impairments and Neurological Disorders, Irvine, United States.,Sue and Bill Gross Stem Cell Research Center, Irvine, United States.,Department of Anatomy and Neurobiology, Irvine, United States.,Department of Physical Medicine & Rehabilitation, University of California, Irvine, Irvine, United States
| | - Sunil P Gandhi
- Department of Neurobiology and Behavior, Irvine, United States
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, Irvine, United States
| | - Matthew A Inlay
- Department of Neurobiology and Behavior, Irvine, United States.,Sue and Bill Gross Stem Cell Research Center, Irvine, United States.,Department of Molecular Biology and Biochemistry, Irvine, United States
| | - Kim N Green
- Department of Neurobiology and Behavior, Irvine, United States.,Institute for Memory Impairments and Neurological Disorders, Irvine, United States
| |
Collapse
|
28
|
Houser MC, Caudle WM, Chang J, Kannarkat GT, Yang Y, Kelly SD, Oliver D, Joers V, Shannon KM, Keshavarzian A, Tansey MG. Experimental colitis promotes sustained, sex-dependent, T-cell-associated neuroinflammation and parkinsonian neuropathology. Acta Neuropathol Commun 2021; 9:139. [PMID: 34412704 PMCID: PMC8375080 DOI: 10.1186/s40478-021-01240-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
Background The etiology of sporadic Parkinson’s disease (PD) remains uncertain, but genetic, epidemiological, and physiological overlap between PD and inflammatory bowel disease suggests that gut inflammation could promote dysfunction of dopamine-producing neurons in the brain. Mechanisms behind this pathological gut-brain effect and their interactions with sex and with environmental factors are not well understood but may represent targets for therapeutic intervention. Methods We sought to identify active inflammatory mechanisms which could potentially contribute to neuroinflammation and neurological disease in colon biopsies and peripheral blood immune cells from PD patients. Then, in mouse models, we assessed whether dextran sodium sulfate-mediated colitis could exert lingering effects on dopaminergic pathways in the brain and whether colitis increased vulnerability to a subsequent exposure to the dopaminergic neurotoxicant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We assessed the involvement of inflammatory mechanisms identified in the PD patients in colitis-related neurological dysfunction in male and female mice, utilizing mice lacking the Regulator of G-Protein Signaling 10 (RGS10)—an inhibitor of nuclear factor kappa B (NFκB)—to model enhanced NFκB activity, and mice in which CD8+ T-cells were depleted. Results High levels of inflammatory markers including CD8B and NFκB p65 were found in colon biopsies from PD patients, and reduced levels of RGS10 were found in immune cells in the blood. Male mice that experienced colitis exhibited sustained reductions in tyrosine hydroxylase but not in dopamine as well as sustained CD8+ T-cell infiltration and elevated Ifng expression in the brain. CD8+ T-cell depletion prevented colitis-associated reductions in dopaminergic markers in males. In both sexes, colitis potentiated the effects of MPTP. RGS10 deficiency increased baseline intestinal inflammation, colitis severity, and neuropathology. Conclusions This study identifies peripheral inflammatory mechanisms in PD patients and explores their potential to impact central dopaminergic pathways in mice. Our findings implicate a sex-specific interaction between gastrointestinal inflammation and neurologic vulnerability that could contribute to PD pathogenesis, and they establish the importance of CD8+ T-cells in this process in male mice. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s40478-021-01240-4.
Collapse
|
29
|
Geoffrion D, Robert MC, Di Polo A, Koenekoop RK, Agoumi Y, Harissi-Dagher M. Tear Film Cytokine Profile of Patients With the Boston Keratoprosthesis Type 1: Comparing Patients With and Without Glaucoma. Invest Ophthalmol Vis Sci 2021; 62:20. [PMID: 33856415 PMCID: PMC8054627 DOI: 10.1167/iovs.62.4.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Inflammatory cytokines are involved in glaucoma pathogenesis. The purpose is to compare cytokine levels in the tear film of Boston keratoprosthesis (KPro) patients with and without glaucoma, relative to controls, and correlate levels with clinical parameters. Methods This cross-sectional study enrolled 58 eyes (58 patients): 41 KPro eyes with glaucoma, 7 KPro eyes without glaucoma, and 10 healthy controls. Twenty-seven cytokines were measured by multiplex bead immunoassay. Intraocular pressure (IOP), cup-to-disk ratio (CDR), retinal nerve fiber layer, visual acuity, topical medications, and angle closure were assessed in all KPro eyes. Cytokine levels between groups were analyzed by nonparametric tests, and correlations with clinical parameters by Spearman's test. Results Levels of TNF-ɑ, IL-1β, FGF-basic, and IFN-ɣ were significantly higher in KPro with glaucoma compared to KPro without (P = 0.020; 0.008; 0.043; 0.018, respectively). KPro groups had similar characteristics and topical antibiotic/steroid regimen. Levels of IL-1Ra, IL-15, VEGF, and RANTES were significantly higher in KPro with glaucoma compared to controls (P < 0.001; = 0.034; < 0.001; = 0.001, respectively). IL-1β and IFN-ɣ levels were positively correlated with CDR (r = 0.309, P = 0.039 and r = 0.452, P = 0.006, respectively) and IOP (r = 0.292, P = 0.047 and r = 0.368, P = 0.023, respectively). TNF-α and FGF-basic levels were positively correlated with CDR (r = 0.348, P = 0.022 and r = 0.344, P = 0.021, respectively). Conclusions TNF-α, IL-1β, FGF-basic, IFN-ɣ are elevated in tears of KPro patients with glaucoma and correlate with CDR and IOP. These results show, for the first time in humans, concordance with documented elevations of TNF-α and IL-1β in the murine KPro model. Ocular surface inflammation may reflect inflammatory processes of KPro glaucoma.
Collapse
Affiliation(s)
- Dominique Geoffrion
- Department of Ophthalmology, Université de Montréal, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Department of Experimental Surgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Marie-Claude Robert
- Department of Ophthalmology, Université de Montréal, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Adriana Di Polo
- Department of Ophthalmology, Université de Montréal, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Department of Neurosciences, Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Robert K Koenekoop
- Departments of Experimental Surgery, Paediatric Surgery, Adult Ophthalmology, and Human Genetics, Montreal Children's Hospital, McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Younes Agoumi
- Department of Ophthalmology, Université de Montréal, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Mona Harissi-Dagher
- Department of Ophthalmology, Université de Montréal, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
30
|
Atkinson SP. A preview of selected articles. Stem Cells Transl Med 2021. [PMCID: PMC7980205 DOI: 10.1002/sctm.21-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
31
|
Ochocka N, Kaminska B. Microglia Diversity in Healthy and Diseased Brain: Insights from Single-Cell Omics. Int J Mol Sci 2021; 22:3027. [PMID: 33809675 PMCID: PMC8002227 DOI: 10.3390/ijms22063027] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) that have distinct ontogeny from other tissue macrophages and play a pivotal role in health and disease. Microglia rapidly react to the changes in their microenvironment. This plasticity is attributed to the ability of microglia to adapt a context-specific phenotype. Numerous gene expression profiling studies of immunosorted CNS immune cells did not permit a clear dissection of their phenotypes, particularly in diseases when peripheral cells of the immune system come to play. Only recent advances in single-cell technologies allowed studying microglia at high resolution and revealed a spectrum of discrete states both under homeostatic and pathological conditions. Single-cell technologies such as single-cell RNA sequencing (scRNA-seq) and mass cytometry (Cytometry by Time-Of-Flight, CyTOF) enabled determining entire transcriptomes or the simultaneous quantification of >30 cellular parameters of thousands of individual cells. Single-cell omics studies demonstrated the unforeseen heterogeneity of microglia and immune infiltrates in brain pathologies: neurodegenerative disorders, stroke, depression, and brain tumors. We summarize the findings from those studies and the current state of knowledge of functional diversity of microglia under physiological and pathological conditions. A precise definition of microglia functions and phenotypes may be essential to design future immune-modulating therapies.
Collapse
Affiliation(s)
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02-093 Warsaw, Poland;
| |
Collapse
|
32
|
Reply to Green and Hume: Nonmicroglia peripheral immune effects of short-term CSF1R inhibition with PLX5622. Proc Natl Acad Sci U S A 2021; 118:2020660118. [PMID: 33446487 PMCID: PMC7848750 DOI: 10.1073/pnas.2020660118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
33
|
Tribble JR, Kokkali E, Otmani A, Plastino F, Lardner E, Vohra R, Kolko M, André H, Morgan JE, Williams PA. When Is a Control Not a Control? Reactive Microglia Occur Throughout the Control Contralateral Pathway of Retinal Ganglion Cell Projections in Experimental Glaucoma. Transl Vis Sci Technol 2021; 10:22. [PMID: 33510961 PMCID: PMC7804521 DOI: 10.1167/tvst.10.1.22] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose Animal models show retinal ganglion cell (RGC) injuries that replicate features of glaucoma and the contralateral eye is commonly used as an internal control. There is significant crossover of RGC axons from the ipsilateral to the contralateral side at the level of the optic chiasm, which may confound findings when damage is restricted to one eye. The effect of unilateral glaucoma on neuroinflammatory damage to the contralateral pathway of RGC projections has largely been unexplored. Methods Ocular hypertensive glaucoma was induced unilaterally or bilaterally in the rat and RGC neurodegenerative events were assessed. Neuroinflammation was quantified in the retina, optic nerve head, optic nerve, lateral geniculate nucleus, and superior colliculus by high-resolution imaging, and in the retina by flow cytometry and protein arrays. Results After ocular hypertensive stress, peripheral monocytes enter the retina and microglia become reactive. This effect is more marked in animals with bilateral ocular hypertensive glaucoma. In rats where glaucoma was induced unilaterally, there was significant microglia activation in the contralateral (control) eye. Microglial activation extended into the optic nerve and terminal visual thalami, where it was similar across hemispheres in unilateral ocular hypertension. Conclusions These data suggest that caution is warranted when using the contralateral eye as a control and in comparing visual thalami in unilateral models of glaucoma. Translational Relevance The use of a contralateral eye as a control may confound the discovery of human-relevant mechanism and treatments in animal models. We also identify neuroinflammatory protein responses that warrant further investigation as potential disease-modifiable targets.
Collapse
Affiliation(s)
- James R. Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Eirini Kokkali
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
| | - Amin Otmani
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Flavia Plastino
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Emma Lardner
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Rupali Vohra
- Department of Veterinary and Animal Sciences, Pathobiological Sciences, University of Copenhagen, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Ophthalmology, Rigshospitalet-Glostrup, Copenhagen, Denmark
| | - Helder André
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - James E. Morgan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
- School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Pete A. Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Bruzelius A, Hidalgo I, Boza-Serrano A, Hjelmér AG, Tison A, Deierborg T, Bengzon J, Ramos-Moreno T. The human bone marrow harbors a CD45 - CD11B + cell progenitor permitting rapid microglia-like cell derivative approaches. Stem Cells Transl Med 2020; 10:582-597. [PMID: 33295698 PMCID: PMC7980218 DOI: 10.1002/sctm.20-0127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 09/23/2020] [Accepted: 10/25/2020] [Indexed: 12/21/2022] Open
Abstract
Microglia, the immune sentinel of the central nervous system (CNS), are generated from yolk sac erythromyeloid progenitors that populate the developing CNS. Interestingly, a specific type of bone marrow-derived monocyte is able to express a yolk sac microglial signature and populate CNS in disease. Here we have examined human bone marrow (hBM) in an attempt to identify novel cell sources for generating microglia-like cells to use in cell-based therapies and in vitro modeling. We demonstrate that hBM stroma harbors a progenitor cell that we name stromal microglial progenitor (STR-MP). STR-MP single-cell gene analysis revealed the expression of the consensus genetic microglial signature and microglial-specific genes present in development and CNS pathologies. STR-MPs can be expanded and generate microglia-like cells in vitro, which we name stromal microglia (STR-M). STR-M cells show phagocytic ability, classically activate, and survive and phagocyte in human brain tissue. Thus, our results reveal that hBM harbors a source of microglia-like precursors that can be used in patient-centered fast derivative approaches.
Collapse
Affiliation(s)
- Andreas Bruzelius
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden.,Department of Experimental Medical Science and Lund Stem Cell Center BMC, Lund University, Lund, Sweden
| | - Isabel Hidalgo
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden.,Institution for Laboratory Medicine, Division of Molecular Hematology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Antonio Boza-Serrano
- Departamento de Bioquimica y Biologia Molecular, Facultad de Farmacia e Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain.,Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Bio Medical Center (BMC)., Lund University, Lund, Sweden
| | - Anna-Giorgia Hjelmér
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden
| | - Amelie Tison
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Bio Medical Center (BMC)., Lund University, Lund, Sweden
| | - Johan Bengzon
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden
| | - Tania Ramos-Moreno
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden.,Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Bio Medical Center (BMC)., Lund University, Lund, Sweden
| |
Collapse
|
35
|
Hui PC, Pereira LA, Dore R, Chen S, Taniguchi E, Chodosh J, Dohlman CH, Paschalis EI. Intrinsic Optical Properties of Boston Keratoprosthesis. Transl Vis Sci Technol 2020; 9:10. [PMID: 33200051 PMCID: PMC7645245 DOI: 10.1167/tvst.9.12.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/07/2020] [Indexed: 12/03/2022] Open
Abstract
Purpose To benchmark the optical performance of Boston Keratoprosthesis (B-KPro). Methods Back focal lengths (BFL) of B-KPros for various eye axial lengths were measured using an optical bench, International Organization for Standardization–certified for intraocular lens characterization, and compared against manufacturer's specification. The modulation transfer function (MTF) and the resolution efficiencies were measured. The theoretical geometry-dependent higher-order aberrations (HOA) were calculated. The devices were characterized with optical profilometry for estimating the surface scattering. Aberration correction and subsequent image quality improvement were simulated in CODE-V. Natural scene-imaging was performed in a mock ocular environment. Retrospective analysis of 15 B-KPro recipient eyes were presented to evaluate the possibility of achieving 20/20 best-corrected visual acuity (BCVA). Results BFL measurements were in excellent agreement with the manufacturer-reported values (r = 0.999). The MTF specification exceeded what is required for achieving 20/20 visual acuity. Astigmatism and field curvature, correctable in simulations, were the primary aberrations limiting imaging performance. Profilometry of the anterior surface revealed nanoscale roughness (root-mean-square amplitude, 30–50 nm), contributing negligibly to optical scattering. Images of natural scenes obtained with a simulated B-KPro eye demonstrated good central vision, with 10/10 visual acuity (equivalent to 20/20). Full restoration of 20/20 BCVA was obtainable for over 9 years in some patients. Conclusions Theoretical and experimental considerations demonstrate that B-KPro has the optical capacity to restore 20/20 BCVA in patients. Further image quality improvement can be anticipated through correction of HOAs. Translational Relevance We establish an objective benchmark to characterize the optics of the B-KPro and other keratoprosthesis and propose design changes to allow improved vision in B-KPro patients.
Collapse
Affiliation(s)
- Pui-Chuen Hui
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.,Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Leonardo A Pereira
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.,CAPES Foundation, Ministry of Education of Brazil, Brasilia, Brazil
| | - Renald Dore
- University of Rochester, Institute of Optics, Rochester, NY, USA
| | - Shengtong Chen
- University of Rochester, Institute of Optics, Rochester, NY, USA
| | - Elise Taniguchi
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.,Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Claes H Dohlman
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.,Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Eleftherios I Paschalis
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.,Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
36
|
Karlen SJ, Miller EB, Burns ME. Microglia Activation and Inflammation During the Death of Mammalian Photoreceptors. Annu Rev Vis Sci 2020; 6:149-169. [PMID: 32936734 PMCID: PMC10135402 DOI: 10.1146/annurev-vision-121219-081730] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Photoreceptors are highly specialized sensory neurons with unique metabolic and physiological requirements. These requirements are partially met by Müller glia and cells of the retinal pigment epithelium (RPE), which provide essential metabolites, phagocytose waste, and control the composition of the surrounding microenvironment. A third vital supporting cell type, the retinal microglia, can provide photoreceptors with neurotrophic support or exacerbate neuroinflammation and hasten neuronal cell death. Understanding the physiological requirements for photoreceptor homeostasis and the factors that drive microglia to best promote photoreceptor survival has important implications for the treatment and prevention of blinding degenerative diseases like retinitis pigmentosa and age-related macular degeneration.
Collapse
Affiliation(s)
- Sarah J. Karlen
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, California 95616, USA
| | - Eric B. Miller
- Center for Neuroscience, University of California, Davis, Davis, California 95616, USA
| | - Marie E. Burns
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, California 95616, USA
- Center for Neuroscience, University of California, Davis, Davis, California 95616, USA
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, California 95616, USA
| |
Collapse
|
37
|
CSF1R inhibition by a small-molecule inhibitor is not microglia specific; affecting hematopoiesis and the function of macrophages. Proc Natl Acad Sci U S A 2020; 117:23336-23338. [PMID: 32900927 PMCID: PMC7519218 DOI: 10.1073/pnas.1922788117] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Colony-stimulating factor 1 receptor (CSF1R) inhibition has been proposed as a method for microglia depletion, with the assumption that it does not affect peripheral immune cells. Here, we show that CSF1R inhibition by PLX5622 indeed affects the myeloid and lymphoid compartments, causes long-term changes in bone marrow-derived macrophages by suppressing interleukin 1β, CD68, and phagocytosis but not CD208, following exposure to endotoxin, and also reduces the population of resident and interstitial macrophages of peritoneum, lung, and liver but not spleen. Thus, small-molecule CSF1R inhibition is not restricted to microglia, causing strong effects on circulating and tissue macrophages that perdure long after cessation of the treatment. Given that peripheral monocytes repopulate the central nervous system after CSF1R inhibition, these changes have practical implications for relevant experimental data.
Collapse
|
38
|
Green KN, Crapser JD, Hohsfield LA. To Kill a Microglia: A Case for CSF1R Inhibitors. Trends Immunol 2020; 41:771-784. [PMID: 32792173 PMCID: PMC7484341 DOI: 10.1016/j.it.2020.07.001] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Microglia, the brain's immune sentinels, have garnered much attention in recent years. Researchers have begun to identify the manifold roles that these cells play in the central nervous system (CNS), and this work has been greatly facilitated by microglial depletion paradigms. The varying degrees of spatiotemporal manipulation afforded by such techniques allow microglial ablation before, during, and/or following insult, injury, or disease. We review the major methods of microglial depletion, including toxin-based, genetic, and pharmacological approaches, which differ in key factors including depletion onset, duration, and off-target effects. We conclude that pharmacological CSF1R inhibitors afford the most extensive versatility in manipulating microglia, making them ideal candidates for future studies investigating microglial function in health and disease.
Collapse
Affiliation(s)
- Kim N Green
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA.
| | - Joshua D Crapser
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Lindsay A Hohsfield
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| |
Collapse
|
39
|
Chen X, Lei F, Zhou C, Chodosh J, Wang L, Huang Y, Dohlman CH, Paschalis EI. Glaucoma after Ocular Surgery or Trauma: The Role of Infiltrating Monocytes and Their Response to Cytokine Inhibitors. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2056-2066. [PMID: 32693061 DOI: 10.1016/j.ajpath.2020.07.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/21/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022]
Abstract
Glaucoma is a frequent and devastating long-term complication following ocular trauma, including corneal surgery, open globe injury, chemical burn, and infection. Postevent inflammation and neuroglial remodeling play a key role in subsequent ganglion cell apoptosis and glaucoma. To this end, this study was designed to investigate the amplifying role of monocyte infiltration into the retina. By using three different ocular injury mouse models (corneal suture, penetrating keratoplasty, and globe injury) and monocyte fate mapping techniques, we show that ocular trauma or surgery can cause robust infiltration of bone marrow-derived monocytes into the retina and subsequent neuroinflammation by up-regulation of Tnf, Il1b, and Il6 mRNA within 24 hours. This is accompanied by ganglion cell apoptosis and neurodegeneration. Prompt inhibition of tumor necrosis factor-α or IL-1β markedly suppresses monocyte infiltration and ganglion cell loss. Thus, acute ocular injury (surgical or trauma) can lead to rapid neuroretinal inflammation and subsequent ganglion cell loss, the hallmark of glaucoma. Infiltrating monocytes play a central role in this process, likely amplifying the inflammatory cascade, aiding in the activation of retinal microglia. Prompt administration of cytokine inhibitors after ocular injury prevents this infiltration and ameliorates the damage to the retina-suggesting that it may be used prophylactically for neuroprotection against post-traumatic glaucoma.
Collapse
Affiliation(s)
- Xiaoniao Chen
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Fengyang Lei
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Chengxin Zhou
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - James Chodosh
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Liqiang Wang
- Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Yifei Huang
- Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Claes H Dohlman
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts.
| | - Eleftherios I Paschalis
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
40
|
Takeda A, Yanai R, Murakami Y, Arima M, Sonoda KH. New Insights Into Immunological Therapy for Retinal Disorders. Front Immunol 2020; 11:1431. [PMID: 32719682 PMCID: PMC7348236 DOI: 10.3389/fimmu.2020.01431] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/03/2020] [Indexed: 12/24/2022] Open
Abstract
In the twentieth century, a conspicuous lack of effective treatment strategies existed for managing several retinal disorders, including age-related macular degeneration; diabetic retinopathy (DR); retinopathy of prematurity (ROP); retinitis pigmentosa (RP); uveitis, including Behçet's disease; and vitreoretinal lymphoma (VRL). However, in the first decade of this century, advances in biomedicine have provided new treatment strategies in the field of ophthalmology, particularly biologics that target vascular endothelial growth factor or tumor necrosis factor (TNF)-α. Furthermore, clinical trials on gene therapy specifically for patients with autosomal recessive or X-linked RP have commenced. The overall survival rates of patients with VRL have improved, owing to earlier diagnoses and better treatment strategies. However, some unresolved problems remain such as primary or secondary non-response to biologics or chemotherapy, and the lack of adequate strategies for treating most RP patients. In this review, we provide an overview of the immunological mechanisms of the eye under normal conditions and in several retinal disorders, including uveitis, DR, ROP, RP, and VRL. In addition, we discuss recent studies that describe the inflammatory responses that occur during the course of these retinal disorders to provide new insights into their diagnosis and treatment.
Collapse
Affiliation(s)
- Atsunobu Takeda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Ophthalmology, Clinical Research Institute, Kyushu Medical Center, National Hospital Organization, Fukuoka, Japan
| | - Ryoji Yanai
- Department of Ophthalmology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mitsuru Arima
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
41
|
Hui PC, Shtyrkova K, Zhou C, Chen X, Chodosh J, Dohlman CH, Paschalis EI. Implantable self-aligning fiber-optic optomechanical devices for in vivo intraocular pressure-sensing in artificial cornea. JOURNAL OF BIOPHOTONICS 2020; 13:e202000031. [PMID: 32246524 DOI: 10.1002/jbio.202000031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 06/11/2023]
Abstract
Artificial cornea is an effective treatment of corneal blindness. Yet, intraocular pressure (IOP) measurements for glaucoma monitoring remain an urgent unmet need. Here, we present the integration of a fiber-optic Fabry-Perot pressure sensor with an FDA-approved keratoprosthesis for real-time IOP measurements using a novel strategy based on optical-path self-alignment with micromagnets. Additionally, an alternative noncontact sensor-interrogation approach is demonstrated using a bench-top optical coherence tomography system. We show stable pressure readings with low baseline drift (<2.8 mm Hg) for >4.5 years in vitro and efficacy in IOP interrogation in vivo using fiber-optic self-alignment, with good initial agreement with the actual IOP. Subsequently, IOP drift in vivo was due to retroprosthetic membrane (RPM) formation on the sensor secondary to surgical inflammation (more severe in the current pro-fibrotic rabbit model). This study paves the way for clinical adaptation of optical pressure sensors with ocular implants, highlighting the importance of controlling RPM in clinical adaptation.
Collapse
Affiliation(s)
- Pui-Chuen Hui
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Katia Shtyrkova
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States
| | - Chengxin Zhou
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Xiaoniao Chen
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - James Chodosh
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Claes H Dohlman
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Eleftherios I Paschalis
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
42
|
Abstract
To review clinical aspects and cellular and molecular steps in the development of long-term glaucoma after corneal surgery or acute trauma—especially the pivotal role of tumor necrosis factor alpha (TNF-α), the rapidity of the secondary damage to the retinal ganglion cells, and the clinical promise of early antiinflammatory intervention.
Collapse
|
43
|
Moos WH, Faller DV, Glavas IP, Harpp DN, Kanara I, Mavrakis AN, Pernokas J, Pernokas M, Pinkert CA, Powers WR, Sampani K, Steliou K, Vavvas DG, Zamboni RJ, Kodukula K, Chen X. Klotho Pathways, Myelination Disorders, Neurodegenerative Diseases, and Epigenetic Drugs. Biores Open Access 2020; 9:94-105. [PMID: 32257625 PMCID: PMC7133426 DOI: 10.1089/biores.2020.0004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this review we outline a rationale for identifying neuroprotectants aimed at inducing endogenous Klotho activity and expression, which is epigenetic action, by definition. Such an approach should promote remyelination and/or stimulate myelin repair by acting on mitochondrial function, thereby heralding a life-saving path forward for patients suffering from neuroinflammatory diseases. Disorders of myelin in the nervous system damage the transmission of signals, resulting in loss of vision, motion, sensation, and other functions depending on the affected nerves, currently with no effective treatment. Klotho genes and their single-pass transmembrane Klotho proteins are powerful governors of the threads of life and death, true to the origin of their name, Fates, in Greek mythology. Among its many important functions, Klotho is an obligatory co-receptor that binds, activates, and/or potentiates critical fibroblast growth factor activity. Since the discovery of Klotho a little over two decades ago, it has become ever more apparent that when Klotho pathways go awry, oxidative stress and mitochondrial dysfunction take over, and age-related chronic disorders are likely to follow. The physiological consequences can be wide ranging, potentially wreaking havoc on the brain, eye, kidney, muscle, and more. Central nervous system disorders, neurodegenerative in nature, and especially those affecting the myelin sheath, represent worthy targets for advancing therapies that act upon Klotho pathways. Current drugs for these diseases, even therapeutics that are disease modifying rather than treating only the symptoms, leave much room for improvement. It is thus no wonder that this topic has caught the attention of biomedical researchers around the world.
Collapse
Affiliation(s)
- Walter H. Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, San Francisco, California
- ShangPharma Innovation, Inc., South San Francisco, California
| | - Douglas V. Faller
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts
| | - Ioannis P. Glavas
- Department of Ophthalmology, New York University School of Medicine, New York, New York
| | - David N. Harpp
- Department of Chemistry, McGill University, Montreal, Canada
| | | | - Anastasios N. Mavrakis
- Department of Medicine, Tufts University School of Medicine, St. Elizabeth's Medical Center, Boston, Massachusetts
| | - Julie Pernokas
- Advanced Dental Associates of New England, Woburn, Massachusetts
| | - Mark Pernokas
- Advanced Dental Associates of New England, Woburn, Massachusetts
| | - Carl A. Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Whitney R. Powers
- Department of Health Sciences, Boston University, Boston, Massachusetts
- Department of Anatomy, Boston University School of Medicine, Boston, Massachusetts
| | - Konstantina Sampani
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Beetham Eye Institute, Joslin Diabetes Center, Boston, Massachusetts
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts
- PhenoMatriX, Inc., Natick, Massachusetts
| | - Demetrios G. Vavvas
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | | | | | - Xiaohong Chen
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| |
Collapse
|
44
|
Fouda AY, Xu Z, Narayanan SP, Caldwell RW, Caldwell RB. Utility of LysM-cre and Cdh5-cre Driver Mice in Retinal and Brain Research: An Imaging Study Using tdTomato Reporter Mouse. Invest Ophthalmol Vis Sci 2020; 61:51. [PMID: 32232350 PMCID: PMC7405957 DOI: 10.1167/iovs.61.3.51] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/22/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose The lysozyme 2 (Lyz2 or LysM) cre mouse is extensively used to achieve genetic manipulation in myeloid cells and it has been widely employed in retinal research. However, LysM has been recently described to be expressed in brain neurons and there is a debate on whether it is also expressed by resident microglia in addition to infiltrating macrophages. Methods We examined LysM-cre recombination in retinal tissue using a LysM-cre/tdTomato reporter mouse together with immunolabeling for several retinal cell markers. We further compared LysM-cre tdTomato recombination with that of Cdh5-cre driver, which is expressed in both endothelial and hematopoietic cells. Results LysM-cre was strongly expressed in most microglia/resident macrophages in neonatal retinas (P8) and to a lesser extent in microglia of adult retinas. In addition, there was some neuronal recombination (8 %) of LysM-cre specifically in adult retinal ganglion cells and amacrine cells. After retinal ischemia-reperfusion injury, LysM-cre was strongly expressed in microglia/infiltrating macrophages. Cdh5-cre was expressed in endothelial and myeloid cells of P8 pups retinas. Unexpectedly, Cdh5 showed additional expression in adult mouse retinal ganglion cells and brain neurons. Conclusions LysM-cre is expressed in macrophages and a subset of microglia together with a small but significant recombination of LysM-cre in the retinal neurons of adult mice. Cdh5 also showed some neuronal expression in both retina and brain of adult mice. These findings should be taken into consideration when interpreting results from central nervous system research using LysM-cre and Cdh5-cre mice.
Collapse
Affiliation(s)
- Abdelrahman Y. Fouda
- Vascular Biology Center, Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Zhimin Xu
- Vascular Biology Center, Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - S. Priya Narayanan
- Vascular Biology Center, Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, Georgia, United States
| | - R. William Caldwell
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Pharmacology and Toxicology, Augusta University, Augusta, Georgia, United States
| | - Ruth B. Caldwell
- Vascular Biology Center, Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Cellular Biology & Anatomy, Augusta University, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| |
Collapse
|
45
|
Chaudhary R, Scott RAH, Wallace G, Berry M, Logan A, Blanch RJ. Inflammatory and Fibrogenic Factors in Proliferative Vitreoretinopathy Development. Transl Vis Sci Technol 2020; 9:23. [PMID: 32742753 PMCID: PMC7357815 DOI: 10.1167/tvst.9.3.23] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Proliferative vitreoretinopathy (PVR) occurs in 5%-10% of rhegmatogenous retinal detachment cases and is the principle cause for failure of retinal reattachment surgery. Although there are a number of surgical adjunctive agents available for preventing the development of PVR, all have limited efficacy. Discovering predictive molecular biomarkers to determine the probability of PVR development after retinal reattachment surgery will allow better patient stratification for more targeted drug evaluations. Methods Narrative literature review. Results We provide a summary of the inflammatory and fibrogenic factors found in ocular fluid samples during the development of retinal detachment and PVR and discuss their possible use as molecular PVR predictive biomarkers. Conclusions Studies monitoring the levels of the above factors have found that few if any have predictive biomarker value, suggesting that widening the phenotype of potential factors and a combinatorial approach are required to determine predictive biomarkers for PVR. Translational Relevance The identification of relevant biomarkers relies on an understanding of disease signaling pathways derived from basic science research. We discuss the extent to which those molecules identified as biomarkers and predictors of PVR relate to disease pathogenesis and could function as useful disease predictors. (http://www.umin.ac.jp/ctr/ number, UMIN000005604).
Collapse
Affiliation(s)
- Rishika Chaudhary
- Academic Unit of Ophthalmology, Birmingham and Midland Eye Centre, Birmingham, UK.,Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,NIHR Surgical Reconstruction and Microbiology Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Graham Wallace
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Martin Berry
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Ann Logan
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,NIHR Surgical Reconstruction and Microbiology Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Richard J Blanch
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,NIHR Surgical Reconstruction and Microbiology Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Department of Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Academic Unit of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, UK
| |
Collapse
|
46
|
Schwarzer P, Kokona D, Ebneter A, Zinkernagel MS. Effect of Inhibition of Colony-Stimulating Factor 1 Receptor on Choroidal Neovascularization in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 190:412-425. [PMID: 31783006 DOI: 10.1016/j.ajpath.2019.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 07/09/2019] [Accepted: 10/21/2019] [Indexed: 12/15/2022]
Abstract
Neovascular age-related macular degeneration is one of the leading causes of blindness. Microglia and macrophages play a critical role in choroidal neovascularization (CNV) and may, therefore, be potential targets to modulate the disease course. This study evaluated the effect of the colony-stimulating factor-1 receptor inhibitor PLX5622 on experimental laser-induced CNV. A 98% reduction of retinal microglia cells was observed in the retina 1 week after initiation of PLX5622 treatment, preventing accumulation of macrophages within the laser site and leading to a reduction of leukocytes within the choroid after CNV induction. Mice treated with PLX5622 had a significantly faster decrease of the CNV lesion size, as revealed by in vivo imaging and immunohistochemistry from day 3 to day 14 compared with untreated mice. Several inflammatory modulators, such as chemokine (C-C motif) ligand 9, granulocyte-macrophage colony-stimulating factor, soluble tumor necrosis factor receptor-I, IL-1α, and matrix metallopeptidase-2, were elevated in the acute phase of the disease when microglia were ablated with PLX5622, whereas other cytokines (eg, interferon-γ, IL-4, and IL-10) were reduced. Our results suggest that colony-stimulating factor-1 receptor inhibition may be a novel therapeutic target in patients with neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Petra Schwarzer
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern; and the Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Despina Kokona
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern; and the Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Andreas Ebneter
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern; and the Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern; and the Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
47
|
Jovanovic J, Liu X, Kokona D, Zinkernagel MS, Ebneter A. Inhibition of inflammatory cells delays retinal degeneration in experimental retinal vein occlusion in mice. Glia 2019; 68:574-588. [PMID: 31652020 PMCID: PMC7003783 DOI: 10.1002/glia.23739] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022]
Abstract
The role of microglia in retinal inflammation is still ambiguous. Branch retinal vein occlusion initiates an inflammatory response whereby resident microglia cells are activated. They trigger infiltration of neutrophils that exacerbate blood–retina barrier damage, regulate postischemic inflammation and irreversible loss of neuroretina. Suppression of microglia‐mediated inflammation might bear potential for mitigating functional impairment after retinal vein occlusion (RVO). To test this hypothesis, we depleted microglia by PLX5622 (a selective tyrosine kinase inhibitor that targets the colony‐stimulating factor‐1 receptor) in fractalkine receptor reporter mice (Cx3cr1gfp/+) subjected to various regimens of PLX5622 treatment and experimental RVO. Effectiveness of microglia suppression and retinal outcomes including retinal thickness as well as ganglion cell survival were compared to a control group of mice with experimental vein occlusion only. PLX5622 caused dramatic suppression of microglia. Despite vein occlusion, reappearance of green fluorescent protein positive cells was strongly impeded with continuous PLX5622 treatment and significantly delayed after its cessation. In depleted mice, retinal proinflammatory cytokine signaling was diminished and retinal ganglion cell survival improved by almost 50% compared to nondepleted animals 3 weeks after vein occlusion. Optical coherence tomography suggested delayed retinal degeneration in depleted mice. In summary, findings indicate that suppression of cells bearing the colony‐stimulating factor‐1 receptor, mainly microglia and monocytes, mitigates ischemic damage and salvages retinal ganglion cells. Blood–retina barrier breakdown seems central in the disease mechanism, and complex interactions between different cell types composing the blood–retina barrier as well as sustained hypoxia might explain why the protective effect was only partial.
Collapse
Affiliation(s)
- Joël Jovanovic
- Department of Ophthalmology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Xuan Liu
- Department of Ophthalmology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland.,Department of Ophthalmology, 1st Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi, China
| | - Despina Kokona
- Department of Ophthalmology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Andreas Ebneter
- Department of Ophthalmology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
48
|
In vivo imaging reveals transient microglia recruitment and functional recovery of photoreceptor signaling after injury. Proc Natl Acad Sci U S A 2019; 116:16603-16612. [PMID: 31350349 PMCID: PMC6697899 DOI: 10.1073/pnas.1903336116] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Microglia, the resident macrophages of the central nervous system, are critical for synaptic pruning and maintenance and for mitigating injury and neurodegeneration. Determining whether microglia–neuron interactions are beneficial in specific instances has been difficult, largely because of the local and transient nature of the interactions. Using simultaneous optical coherence tomography/scanning laser ophthalmoscopy (SLO) and adaptive optics SLO retinal imaging in mice, we show interactions of microglia and photoreceptors over time scales from seconds to months during injury, degeneration, and repair. In vivo optical assessment of photoreceptor signaling in a large neuronal field encompassing the injured area allows us to relate the time course of these microglia movements to that of the tissue remodeling and functional recovery. Microglia respond to damage and microenvironmental changes within the central nervous system by morphologically transforming and migrating to the lesion, but the real-time behavior of populations of these resident immune cells and the neurons they support have seldom been observed simultaneously. Here, we have used in vivo high-resolution optical coherence tomography (OCT) and scanning laser ophthalmoscopy with and without adaptive optics to quantify the 3D distribution and dynamics of microglia in the living retina before and after local damage to photoreceptors. Following photoreceptor injury, microglia migrated both laterally and vertically through the retina over many hours, forming a tight cluster within the area of visible damage that resolved over 2 wk. In vivo OCT optophysiological assessment revealed that the photoreceptors occupying the damaged region lost all light-driven signaling during the period of microglia recruitment. Remarkably, photoreceptors recovered function to near-baseline levels after the microglia had departed the injury locus. These results demonstrate the spatiotemporal dynamics of microglia engagement and restoration of neuronal function during tissue remodeling and highlight the need for mechanistic studies that consider the temporal and structural dynamics of neuron–microglia interactions in vivo.
Collapse
|
49
|
Anderson SR, Roberts JM, Zhang J, Steele MR, Romero CO, Bosco A, Vetter ML. Developmental Apoptosis Promotes a Disease-Related Gene Signature and Independence from CSF1R Signaling in Retinal Microglia. Cell Rep 2019; 27:2002-2013.e5. [PMID: 31091440 PMCID: PMC6544177 DOI: 10.1016/j.celrep.2019.04.062] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/21/2019] [Accepted: 04/11/2019] [Indexed: 12/19/2022] Open
Abstract
Microglia have important remodeling functions in neurodevelopment, aging, and disease, with evidence for molecular diversity. However, the signaling pathways and environmental cues that drive diverse states of microglia are incompletely understood. We profiled microglia of a discrete developing CNS region, the murine retina. We found distinct transcriptional signatures for retinal microglia across development and peak postnatal density of a population that resembles aging and disease-associated microglia (DAM) and CD11c+ microglia of developing white matter. While TREM2 signaling modulates the expression of select genes, the DAM-related signature is significantly reduced in retinas lacking Bax, a proapoptotic factor required for neuronal death. Furthermore, we found postnatal retinal microglia highly expressing CD11c are resistant to loss or inhibition of colony stimulating factor 1 receptor (CSF1R), while most microglia can be eliminated in Bax knockout retina. Thus, developmental apoptosis promotes a microglia gene signature linked to CSF1R independence that shares features with microglia in developing white matter and in disease.
Collapse
Affiliation(s)
- Sarah R Anderson
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Jacqueline M Roberts
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Jianmin Zhang
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Michael R Steele
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Cesar O Romero
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Alejandra Bosco
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Monica L Vetter
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
50
|
Paschalis EI, Taniguchi EV, Chodosh J, Pasquale LR, Colby K, Dohlman CH, Shen LQ. Blood Levels of Tumor Necrosis Factor Alpha and Its Type 2 Receptor Are Elevated in Patients with Boston Type I Keratoprosthesis. Curr Eye Res 2019; 44:599-606. [PMID: 30632412 DOI: 10.1080/02713683.2019.1568500] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Purpose: Boston keratoprosthesis (KPro) patients are prone to glaucoma even with well-controlled intraocular pressure (IOP). Recent experimental data have shown that soluble tumor necrosis factor alpha (TNF-α) after ocular injury may contribute to progressive retinal damage and subsequent glaucoma. This study evaluates the blood plasma levels of soluble TNF-α, TNF receptors 1 (TNFR1) and 2 (TNFR2), and leptin in patients with Boston type I KPro. Methods: Venous blood samples were collected from KPro patients with glaucoma (KPro G, n = 19), KPro patients without glaucoma (KPro NoG, n = 12), primary angle closure glaucoma without KPro (PACG, n = 13), and narrow angles without glaucoma or KPro (NA, n = 21). TNF-α, TNFR1, TNFR2, and leptin levels were quantified using the enzyme-linked immunosorbent assay. Erythrocyte sedimentation rate (ESR) was assessed using the Westergren test. Patients with underlying autoimmune conditions or diabetes were excluded from the study. Results: All groups had similar age, body mass index (BMI), IOP, and ESR (p ≥ 0.11). The mean time from KPro surgery to blood draw was 5.3 ± 3.7 years. Compared to NA patients (0.72 ± 0.3 pg/ml), KPro G and KPro NoG patients had higher blood plasma levels of TNF-α (1.18 ± 0.58 pg/ml, p = 0.006; 1.16 ± 0.50 pg/ml, p = 0.04, respectively). Similarly, KPro G patients had higher blood plasma levels of TNFR2 (2768 ± 1368 pg/ml) than NA patients (2020 ± 435 pg/ml, p = 0.048). In multivariate analysis, KPro status remained positively associated with TNF-α levels (β = 0.36; 95% confidence intervals [CI]: 0.14-0.58; p = 0.002) and TNFR2 levels (β = 458.3; 95% CI: 32.8-883.7; p = 0.035) after adjusting for age, gender, BMI, glaucoma status, and ESR. TNFR1 and leptin levels were not significantly different in the study groups. Conclusions: We detected elevated serum levels of TNF-α and TNFR2 in KPro patients. Longitudinal studies are needed to establish TNF-α and TNFR2 as serum biomarkers related to KPro surgery. Abbreviations: BCVA: best corrected visual acuity; BMI: body mass index; CDR: cup-to-disc ratio; EDTA: ethylenediaminetetraacetic acid; ELISA: enzyme-linked immunosorbent assay; ESR: erythrocyte sedimentation rate; HVF: Humphrey visual field; IOP: intraocular pressure; KPro G: keratoprosthesis with glaucoma; KPro NoG: keratoprosthesis without glaucoma; KPro: keratoprosthesis; MD: mean deviation; NA: narrow angle; non-KPro: without keratoprosthesis; PACG: primary angle closure glaucoma; RNFL: retinal nerve fiber layer; TNF-α: tumor necrosis factor alpha; TNFR1: tumor necrosis factor receptor 1; TNFR2: tumor necrosis factor receptor 2.
Collapse
Affiliation(s)
- Eleftherios I Paschalis
- a Department of Ophthalmology , Harvard Medical School, Massachusetts Eye and Ear , Boston , MA , USA.,b Boston Keratoprosthesis Laboratory , Massachusetts Eye and Ear - Schepens Eye Research Institute, Harvard Medical School , Boston , MA , USA.,c Disruptive Technology Laboratory, Department of Ophthalmology , Massachusetts Eye and Ear, Harvard Medical School , Boston , MA , USA
| | - Elise V Taniguchi
- a Department of Ophthalmology , Harvard Medical School, Massachusetts Eye and Ear , Boston , MA , USA.,b Boston Keratoprosthesis Laboratory , Massachusetts Eye and Ear - Schepens Eye Research Institute, Harvard Medical School , Boston , MA , USA.,d Department of Ophthalmology , Universidade Federal de Sao Paulo , Sao Paulo , Brazil
| | - James Chodosh
- a Department of Ophthalmology , Harvard Medical School, Massachusetts Eye and Ear , Boston , MA , USA.,b Boston Keratoprosthesis Laboratory , Massachusetts Eye and Ear - Schepens Eye Research Institute, Harvard Medical School , Boston , MA , USA.,c Disruptive Technology Laboratory, Department of Ophthalmology , Massachusetts Eye and Ear, Harvard Medical School , Boston , MA , USA
| | - Louis R Pasquale
- e Department of Ophthalmology , Icahn School of Medicine, Mount Sinai Hospital , New York , NY , USA
| | - Kathryn Colby
- f Department of Ophthalmology , University of Chicago , Chicago , IL , USA
| | - Claes H Dohlman
- a Department of Ophthalmology , Harvard Medical School, Massachusetts Eye and Ear , Boston , MA , USA.,b Boston Keratoprosthesis Laboratory , Massachusetts Eye and Ear - Schepens Eye Research Institute, Harvard Medical School , Boston , MA , USA
| | - Lucy Q Shen
- a Department of Ophthalmology , Harvard Medical School, Massachusetts Eye and Ear , Boston , MA , USA
| |
Collapse
|