1
|
Dalvi S, Roll M, Chatterjee A, Kumar LK, Bhogavalli A, Foley N, Arduino C, Spencer W, Reuben-Thomas C, Ortolan D, Pébay A, Bharti K, Anand-Apte B, Singh R. Human iPSC-based disease modeling studies identify a common mechanistic defect and potential therapies for AMD and related macular dystrophies. Dev Cell 2024; 59:3290-3305.e9. [PMID: 39362220 PMCID: PMC11652237 DOI: 10.1016/j.devcel.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/17/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024]
Abstract
Age-related macular degeneration (AMD) and related macular dystrophies (MDs) primarily affect the retinal pigment epithelium (RPE) in the eye. A hallmark of AMD/MDs that drives later-stage pathologies is drusen. Drusen are sub-RPE lipid-protein-rich extracellular deposits, but how drusen forms and accumulates is not known. We utilized human induced pluripotent stem cell (iPSC)-derived RPE from patients with AMD and three distinct MDs to demonstrate that reduced activity of RPE-secreted matrix metalloproteinase 2 (MMP2) contributes to drusen in multiple maculopathies in a genotype-agnostic manner by instigating sterile inflammation and impaired lipid homeostasis via damage-associated molecular pattern molecule (DAMP)-mediated activation of receptor for advanced glycation end-products (RAGE) and increased secretory phospholipase 2-IIA (sPLA2-IIA) levels. Therapeutically, RPE-specific MMP2 supplementation, RAGE-antagonistic peptide, and a small molecule inhibitor of sPLA2-IIA ameliorated drusen accumulation in AMD/MD iPSC-RPE. Ultimately, this study defines a causal role of the MMP2-DAMP-RAGE-sPLA2-IIA axis in AMD/MDs.
Collapse
Affiliation(s)
- Sonal Dalvi
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Michael Roll
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Amit Chatterjee
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Lal Krishan Kumar
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Akshita Bhogavalli
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Nathaniel Foley
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Cesar Arduino
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Whitney Spencer
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Cheyenne Reuben-Thomas
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Davide Ortolan
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Bela Anand-Apte
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Ruchira Singh
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA.
| |
Collapse
|
2
|
Bernaerts E, Ahmadzadeh K, De Visscher A, Malengier-Devlies B, Häuβler D, Mitera T, Martens E, Krüger A, De Somer L, Matthys P, Vandooren J. Human monocyte-derived macrophages shift subcellular metalloprotease activity depending on their activation state. iScience 2024; 27:111171. [PMID: 39569367 PMCID: PMC11576389 DOI: 10.1016/j.isci.2024.111171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/21/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Proteases are key effectors in macrophage function during the initiation and resolution of inflammation. Recent studies have shown that some proteases, traditionally considered extracellular, also exhibit enzymatic and non-enzymatic functions within the cell. This study explores the differential protease landscapes of macrophages based on their phenotype. Human monocytes were isolated from healthy volunteers and stimulated with M-CSF (resting macrophages), LPS/IFN-γ (inflammatory macrophages), or IL-4 (immunosuppressive macrophages). IL-4-stimulated macrophages secreted higher levels of MMPs and natural protease inhibitors compared to LPS/IFN-γ-stimulated macrophages. Increased extracellular proteolytic activity was detected in LPS/IFN-γ-stimulated macrophages while IL-4 stimulation increased cell-associated proteolytic activity, particularly for MMPs. Subcellular fractionation and confocal microscopy revealed the uptake of extracellular MMP-9 and its relocation to the nucleus in IL-4-stimulated, though not in LPS/IFN-γ-stimulated macrophages. Collectively, macrophages alter the subcellular location and activity of their MMPs based on the stimuli received, suggesting another mechanism for protease regulation in macrophage biology.
Collapse
Affiliation(s)
- Eline Bernaerts
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Kourosh Ahmadzadeh
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Amber De Visscher
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Bert Malengier-Devlies
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
- Centre for Reproductive Health and Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Daniel Häuβler
- TUM School of Medicine and Health, Institute of Experimental Oncology and Therapy Research, Technical University of Munich, D-81676 Munich, Germany
| | - Tania Mitera
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Erik Martens
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Achim Krüger
- TUM School of Medicine and Health, Institute of Experimental Oncology and Therapy Research, Technical University of Munich, D-81676 Munich, Germany
| | - Lien De Somer
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
- University Hospital Leuven, Laboratory of Pediatric Immunology, 3000 Leuven, Belgium
| | - Patrick Matthys
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Jennifer Vandooren
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven Campus Kulak, 8500 Kortrijk, Belgium
| |
Collapse
|
3
|
Moustardas P, Abbasi M, Javidjam D, Asamoah CS, Schweitzer-Chaput A, Cisternino S, Bremond-Gignac D, Aberdam D, Lagali N. Duloxetine enhances PAX6 expression and suppresses innate immune responses in murine LPS-induced corneal inflammation. Ocul Surf 2024; 34:225-234. [PMID: 39127390 DOI: 10.1016/j.jtos.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/15/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND-AIM PAX6 is a key regulator of eye development and epithelial homeostasis in the cornea. When deficient, chronic corneal inflammation, neovascularization and limbal stem cell deficiency can occur. Here we investigated the potential of duloxetine, a generic serotonin reuptake inhibitor that can upregulate PAX6 in vitro, for its in vivo activity in the context of corneal inflammation. METHODS Duloxetine tolerance was tested in a human limbal stem cell line and isogenic CRISPR-knockout PAX6+/- cells. C57BL/6-Wildtype mice were administered duloxetine eye drops at concentrations of 1 μM - 2 mM and tested for toxicity and corneal PAX6 expression. In LPS-induced corneal inflammation in mice, duloxetine's effect on PAX6 expression, corneal opacification and inflammatory responses were evaluated by in vivo corneal imaging, immunostaining, and whole-transcriptome microarray analysis. RESULTS No toxicity was observed in vitro for duloxetine concentrations up to 10μΜ. In vivo, duloxetine drops were well-tolerated up to 50 μM. Duloxetine drops at 10μΜ significantly upregulated PAX6 protein levels in the cornea by 30 % within 2 days. In the LPS model, duloxetine resulted in a sustained 33 % PAX6 protein upregulation in the cornea at 7 days, and in reduced opacity within 2 days, accompanied by a significant dampening of IL-17A signaling, neutrophil degranulation, microglial activation, macrophage markers, and MMP expression, despite non-significant changes in total inflammatory cell infiltration. CONCLUSION Short-term administration of a repurposed generic drug, duloxetine, upregulates PAX6 protein levels in the cornea of mice and exerts an anti-inflammatory activity by dampening innate immune responses.
Collapse
Affiliation(s)
- Petros Moustardas
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, 581 83, Linköping, Sweden
| | - Mojdeh Abbasi
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, 581 83, Linköping, Sweden
| | - Dina Javidjam
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, 581 83, Linköping, Sweden
| | - Cindy Saah Asamoah
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, 581 83, Linköping, Sweden
| | - Arnaud Schweitzer-Chaput
- Service de Pharmacie, Hôpital Universitaire Necker - Enfants Malades, APHP, 75015, Paris, France
| | - Salvatore Cisternino
- Service de Pharmacie, Hôpital Universitaire Necker - Enfants Malades, APHP, 75015, Paris, France; Université Paris Cité, INSERM UMRS 1144, Faculté de Pharmacie, 75006, Paris, France
| | - Dominique Bremond-Gignac
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, 75270, Paris, France; Ophthalmology Department, Hôpital Universitaire Necker - Enfants Malades, 75015, Paris, France
| | - Daniel Aberdam
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, 75270, Paris, France; Université Paris Cité, 75014, Paris, France
| | - Neil Lagali
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, 581 83, Linköping, Sweden.
| |
Collapse
|
4
|
Cheung SW, Willis EF, Simmons DG, Bellingham MC, Noakes PG. Phagocytosis of aggrecan-positive perineuronal nets surrounding motor neurons by reactive microglia expressing MMP-9 in TDP-43 Q331K ALS model mice. Neurobiol Dis 2024; 200:106614. [PMID: 39067491 DOI: 10.1016/j.nbd.2024.106614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
Perineuronal nets (PNNs) are extracellular matrix structures that surround excitable neurons and their proximal dendrites. PNNs play an important role in neuroprotection against oxidative stress. Oxidative stress within motor neurons can act as a trigger for neuronal death, and this has been implicated in motor neuron degeneration in amyotrophic lateral sclerosis (ALS). We therefore characterised PNNs around alpha motor neurons and the possible contributing cellular factors in the mutant TDP-43Q331K transgenic mouse, a slow onset ALS mouse model. PNNs around alpha motor neurons showed significant loss at mid-stage disease in TDP-43Q331K mice compared to wild type strain control mice. PNN loss coincided with an increased expression of matrix metallopeptidase-9 (MMP-9), an endopeptidase known to cleave PNNs, within the ventral horn. During mid-stage disease, increased numbers of microglia and astrocytes expressing MMP-9 were present in the ventral horn of TDP-43Q331K mice. In addition, TDP-43Q331K mice showed increased levels of aggrecan, a PNN component, in the ventral horn by microglia and astrocytes during this period. Elevated aggrecan levels within glia were accompanied by an increase in fractalkine expression, a chemotaxic protein responsible for the recruitment of microglia, in alpha motor neurons of onset and mid-stage TDP-43Q331K mice. Following PNN loss, alpha motor neurons in mid-stage TDP-43Q331K mice showed increased 3-nitrotyrosine expression, an indicator of protein oxidation. Together, our observations along with previous PNN research provide suggests a possible model whereby microglia and astrocytes expressing MMP-9 degrade PNNs surrounding alpha motor neurons in the TDP-43Q331K mouse. This loss of nets may expose alpha-motor neurons to oxidative damage leading to degeneration of the alpha motor neurons in the TDP-43Q331K ALS mouse model.
Collapse
Affiliation(s)
- Sang Won Cheung
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Emily F Willis
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - David G Simmons
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Mark C Bellingham
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Peter G Noakes
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia; Queensland Brain Institute, The University of Queensland, St. Lucia, QLD 4072, Australia.
| |
Collapse
|
5
|
Butenko S, Nagalla RR, Guerrero-Juarez CF, Palomba F, David LM, Nguyen RQ, Gay D, Almet AA, Digman MA, Nie Q, Scumpia PO, Plikus MV, Liu WF. Hydrogel crosslinking modulates macrophages, fibroblasts, and their communication, during wound healing. Nat Commun 2024; 15:6820. [PMID: 39122702 PMCID: PMC11315930 DOI: 10.1038/s41467-024-50072-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 06/28/2024] [Indexed: 08/12/2024] Open
Abstract
Biomaterial wound dressings, such as hydrogels, interact with host cells to regulate tissue repair. This study investigates how crosslinking of gelatin-based hydrogels influences immune and stromal cell behavior and wound healing in female mice. We observe that softer, lightly crosslinked hydrogels promote greater cellular infiltration and result in smaller scars compared to stiffer, heavily crosslinked hydrogels. Using single-cell RNA sequencing, we further show that heavily crosslinked hydrogels increase inflammation and lead to the formation of a distinct macrophage subpopulation exhibiting signs of oxidative activity and cell fusion. Conversely, lightly crosslinked hydrogels are more readily taken up by macrophages and integrated within the tissue. The physical properties differentially affect macrophage and fibroblast interactions, with heavily crosslinked hydrogels promoting pro-fibrotic fibroblast activity that drives macrophage fusion through RANKL signaling. These findings suggest that tuning the physical properties of hydrogels can guide cellular responses and improve healing, offering insights for designing better biomaterials for wound treatment.
Collapse
Affiliation(s)
- Sergei Butenko
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Raji R Nagalla
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | | | - Francesco Palomba
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Li-Mor David
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Ronald Q Nguyen
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Denise Gay
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Axel A Almet
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
| | - Michelle A Digman
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Laboratory of Fluorescence Dynamics, The Henry Samueli School of Engineering, University of California, Irvine, CA, USA
| | - Qing Nie
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA, USA
| | - Philip O Scumpia
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Dermatology, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Wendy F Liu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California Irvine, Irvine, CA, USA.
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA.
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA.
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
6
|
Cheung SW, Bhavnani E, Simmons DG, Bellingham MC, Noakes PG. Perineuronal nets are phagocytosed by MMP-9 expressing microglia and astrocytes in the SOD1 G93A ALS mouse model. Neuropathol Appl Neurobiol 2024; 50:e12982. [PMID: 38742276 DOI: 10.1111/nan.12982] [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: 01/10/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024]
Abstract
AIMS Perineuronal nets (PNNs) are an extracellular matrix structure that encases excitable neurons. PNNs play a role in neuroprotection against oxidative stress. Oxidative stress within motor neurons can trigger neuronal death, which has been implicated in amyotrophic lateral sclerosis (ALS). We investigated the spatio-temporal timeline of PNN breakdown and the contributing cellular factors in the SOD1G93A strain, a fast-onset ALS mouse model. METHODS This was conducted at the presymptomatic (P30), onset (P70), mid-stage (P130), and end-stage disease (P150) using immunofluorescent microscopy, as this characterisation has not been conducted in the SOD1G93A strain. RESULTS We observed a significant breakdown of PNNs around α-motor neurons in the ventral horn of onset and mid-stage disease SOD1G93A mice compared with wild-type controls. This was observed with increased numbers of microglia expressing matrix metallopeptidase-9 (MMP-9), an endopeptidase that degrades PNNs. Microglia also engulfed PNN components in the SOD1G93A mouse. Further increases in microglia and astrocyte number, MMP-9 expression, and engulfment of PNN components by glia were observed in mid-stage SOD1G93A mice. This was observed with increased expression of fractalkine, a signal for microglia engulfment, within α-motor neurons of SOD1G93A mice. Following PNN breakdown, α-motor neurons of onset and mid-stage SOD1G93A mice showed increased expression of 3-nitrotyrosine, a marker for protein oxidation, which could render them vulnerable to death. CONCLUSIONS Our observations suggest that increased numbers of MMP-9 expressing glia and their subsequent engulfment of PNNs around α-motor neurons render these neurons sensitive to oxidative damage and eventual death in the SOD1G93A ALS model mouse.
Collapse
Affiliation(s)
- Sang Won Cheung
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Ekta Bhavnani
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - David G Simmons
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Mark C Bellingham
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Peter G Noakes
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
- Queensland Brain Institute, The University of Queensland, St. Lucia, Australia
| |
Collapse
|
7
|
Bai M, Sun R, Cao B, Feng J, Wang J. Monocyte-related cytokines/chemokines in cerebral ischemic stroke. CNS Neurosci Ther 2023; 29:3693-3712. [PMID: 37452512 PMCID: PMC10651979 DOI: 10.1111/cns.14368] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Abstract
AIMS Ischemic stroke is one of the leading causes of death worldwide and the most common cause of disability in Western countries. Multiple mechanisms contribute to the development and progression of ischemic stroke, and inflammation is one of the most important mechanisms. DISCUSSION Ischemia induces the release of adenosine triphosphate/reactive oxygen species, which activates immune cells to produce many proinflammatory cytokines that activate downstream inflammatory cascades to induce fatal immune responses. Research has confirmed that peripheral blood immune cells play a vital role in the immunological cascade after ischemic stroke. The role of monocytes has received much attention among numerous peripheral blood immune cells. Monocytes induce their effects by secreting cytokines or chemokines, including CCL2/CCR2, CCR4, CCR5, CD36, CX3CL1/CX3CR1, CXCL12(SDF-1), LFA-1/ICAM-1, Ly6C, MMP-2/9, NR4A1, P2X4R, P-selectin, CD40L, TLR2/4, and VCAM-1/VLA-4. Those factors play important roles in the process of monocyte recruitment, migration, and differentiation. CONCLUSION This review focuses on the function and mechanism of the cytokines secreted by monocytes in the process of ischemic stroke and provides novel targets for treating cerebral ischemic stroke.
Collapse
Affiliation(s)
- Meiling Bai
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ruize Sun
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bin Cao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jue Wang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
8
|
Maksimova A, Shevela E, Sakhno L, Tikhonova M, Ostanin A, Chernykh E. Human Macrophages Polarized by Interaction with Apoptotic Cells Produce Fibrosis-Associated Mediators and Enhance Pro-Fibrotic Activity of Dermal Fibroblasts In Vitro. Cells 2023; 12:1928. [PMID: 37566007 PMCID: PMC10417661 DOI: 10.3390/cells12151928] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023] Open
Abstract
Apoptosis and subsequent removal of dead cells are an essential part of wound healing. Macrophages phagocytize apoptotic cells (efferocytosis) and contribute to the resolution of inflammation. However, their participation in fibrogenesis and the mechanisms of influence on this process remain unclear. In the present study, we focused on the fibrogenic properties of human monocyte-derived macrophages polarized in the M2 direction by interaction with apoptotic cells. We studied their influence on the proliferation ([3H]-thymidine incorporation), differentiation (by the expression of α-SMA, a myofibroblast marker) and collagen-producing activity (ELISA) of dermal fibroblasts compared to classically (LPS) and alternatively (IL-4) activated macrophages. Macrophages polarized by the interaction with apoptotic cells had a unique phenotype and profile of produced factors and differed from the compared macrophage subtypes. Their conditioned media promoted the proliferation of dermal fibroblasts and the expression of α-SMA in them at the level of macrophages stimulated by IL-4, while the stimulating effect on the collagen-producing activity was more pronounced compared to that of the other macrophage subtypes. Moreover, they are characterized by the high level of production of pro-fibrotic factors such as TIMP-1, TGF-β1 and angiogenin. Taken together, M2-like macrophages polarized by efferocytosis demonstrate in vitro pro-fibrotic activity by promoting the functional activity of dermal fibroblasts and producing pro-fibrotic and pro-angiogenic factors.
Collapse
Affiliation(s)
- Aleksandra Maksimova
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (E.S.); (L.S.); (M.T.); (A.O.); (E.C.)
| | | | | | | | | | | |
Collapse
|
9
|
Soles A, Selimovic A, Sbrocco K, Ghannoum F, Hamel K, Moncada EL, Gilliat S, Cvetanovic M. Extracellular Matrix Regulation in Physiology and in Brain Disease. Int J Mol Sci 2023; 24:7049. [PMID: 37108212 PMCID: PMC10138624 DOI: 10.3390/ijms24087049] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The extracellular matrix (ECM) surrounds cells in the brain, providing structural and functional support. Emerging studies demonstrate that the ECM plays important roles during development, in the healthy adult brain, and in brain diseases. The aim of this review is to briefly discuss the physiological roles of the ECM and its contribution to the pathogenesis of brain disease, highlighting the gene expression changes, transcriptional factors involved, and a role for microglia in ECM regulation. Much of the research conducted thus far on disease states has focused on "omic" approaches that reveal differences in gene expression related to the ECM. Here, we review recent findings on alterations in the expression of ECM-associated genes in seizure, neuropathic pain, cerebellar ataxia, and age-related neurodegenerative disorders. Next, we discuss evidence implicating the transcription factor hypoxia-inducible factor 1 (HIF-1) in regulating the expression of ECM genes. HIF-1 is induced in response to hypoxia, and also targets genes involved in ECM remodeling, suggesting that hypoxia could contribute to ECM remodeling in disease conditions. We conclude by discussing the role microglia play in the regulation of the perineuronal nets (PNNs), a specialized form of ECM in the central nervous system. We show evidence that microglia can modulate PNNs in healthy and diseased brain states. Altogether, these findings suggest that ECM regulation is altered in brain disease, and highlight the role of HIF-1 and microglia in ECM remodeling.
Collapse
Affiliation(s)
- Alyssa Soles
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Adem Selimovic
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Kaelin Sbrocco
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Ferris Ghannoum
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Katherine Hamel
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Emmanuel Labrada Moncada
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Stephen Gilliat
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
10
|
Shahzad A, Rink L, Wessels I. Regulation of matrix metalloproteinase-9 during monopoiesis and zinc deficiency by chromatin remodeling. J Trace Elem Med Biol 2023; 78:127162. [PMID: 37027894 DOI: 10.1016/j.jtemb.2023.127162] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/25/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023]
Abstract
INTRODUCTION Matrix metalloproteinase-9 (MMP-9) cleaves various extracellular matrix proteins, hence significantly contributes to numerous physiological but also pathological processes. Monocytic differentiation is associated with increased MMP-9 gene expression. Interestingly, MMP-9 upregulation during monocytic differentiation is paralleled by a decline in intracellular zinc levels. Hence, an influence of zinc on the regulation of MMP-9 expression may exist. Although, previous studies suggest a vital role of zinc regarding MMP-9 activity, the possible relevance of zinc homeostasis during transcriptional regulation of MMP-9 for example via epigenetic mechanisms is rather unclear. AIM This study aims to find a correlation between zinc deficiency and MMP-9 transcriptional regulation, focusing on epigenetics as the possible mechanism behind zinc deficiency-induced changes. METHODS The effect of differentiation and zinc deficiency on MMP-9 expression and MMP9 promoter accessibility was investigated using the acute promyelocytic cell line NB4. Intracellular free zinc levels were detected by flow cytometry. MMP-9 gene expression was measured by real-time PCR and ELISA. Analysis of chromatin structures was done using chromatin accessibility by real-time PCR (CHART) assay. RESULTS During monocytic differentiation of NB4 cells, the decrease in intracellular zinc levels was paralleled by an increased production of MMP-9. Assessment of chromatin structure revealed increased accessibility of certain regions within the MMP-9 promoter in differentiated cells. Interestingly, upregulated activation-induced MMP-9 gene expression as well as a more accessible MMP-9 promoter were in zinc-deficient NB4 cells whereas zinc resupplementation reversed the effects. CONCLUSION These data demonstrate an important role of epigenetic mechanisms in regulating MMP-9 expression under zinc deficiency. This could provide an encouraging step to expand the research on using zinc for the treatment of various pathological conditions such as inflammatory, vascular and autoimmune diseases resulting from MMP-9 deregulation.
Collapse
Affiliation(s)
- Asad Shahzad
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Lothar Rink
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Inga Wessels
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, D-52074 Aachen, Germany.
| |
Collapse
|
11
|
The role of leukotriene B 4 in cow metritis. J Vet Res 2023; 67:99-104. [PMID: 37008766 PMCID: PMC10062042 DOI: 10.2478/jvetres-2023-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Abstract
Introduction
Metritis is a common postpartum disease in dairy cows. As a mast cell (MC) mediator, leukotriene B4 (LTB4) is the strongest phagocyte chemokine. It is important in inflammation for the recruitment of immune cells to resist infection. This study investigated the effect of LTB4 in metritis.
Material and Methods
Twenty Holstein cows 3 to 6 years old and at 6 to 10 days postpartum were selected, ten of which with postpartum metritis were the experimental group, and the other ten of which as healthy cows were the control group. The levels of LTB4, substance P (SP) and vasoactive intestinal peptide (VIP) were measured by ELISA, the expression of LTB4 receptor 2 (BLT2), matrix metalloproteinase (MMP)-2 and MMP-9 mRNA was measured by qPCR, and collagens I and IV were detected by immunohistochemical staining.
Results
Concentrations of SP and LTB4 were significantly higher, but those of VIP were significantly lower in the experimental group than those in the control group. The expression of BLT2, MMP-2 and MMP-9 mRNA was significantly higher in the experimental group than that in the control group. The expression of collagen Ⅰ and collagen Ⅳ was significantly lower in the experimental group than that in the control group.
Conclusion
In metritis, SP promotes the activation of MC and the synthesis and release of LTB4. Leukotriene B4 chemotactic immune cells promote the high expression of collagenase, which accelerated the hydrolysis of collagen, while the inhibitory effect of VIP on MC was weakened. This may further aggravate the damage to uterine tissue.
Collapse
|
12
|
Reichel T, Held S, Schwarz A, Hacker S, Wesemann F, Donath L, Krüger K. Acute response of biomarkers in plasma from capillary blood after a strenuous endurance exercise bout. Eur J Appl Physiol 2023; 123:179-189. [PMID: 36227377 PMCID: PMC9813032 DOI: 10.1007/s00421-022-05068-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 10/06/2022] [Indexed: 01/09/2023]
Abstract
PURPOSE The present study aims to investigate the acute response of potential exercise-sensitive biomarkers in capillary plasma to an acute incremental running test. In a second step, their concentration was compared to the changes in the venous serum. METHODS Thirty-seven active young female and male adults completed a VO2max ramp test on a treadmill. Before and after exercise, capillary blood from the earlobe and venous blood were taken and synchronized. Concentrations of Interleukin- (IL-) 1β, IL-1ra, IL-6, IL-8, IL-17A, Interferon (IFN)-y, CC-chemokine ligand (CCL)-2, Matrix metallopeptidase (MMP)-9, Secreted protein acidic and rich in cysteine (SPARC), Cluster of differentiation (CD)163, S100 Ca2+ -binding protein (S100) A8, S100A9, S100B, Brain-derived neurotrophic factor (BDNF), and Myeloperoxidase (MPO) were determined by magnetic bead-based multiplex assay. RESULTS Capillary plasma concentrations of IL-1β, IL-6, IL-8, IL-17A, IFN-y, CCL-2, MMP-9, SPARC, CD163, S100A9, S100B, and BDNF increased after exercise (p < 0.05). Comparing the values from capillary plasma and venous serum, ICCs classified as good were found for IFN-y (post), while the ICCs for IL-1β, IL-8, IL-17A, CCL-2, MMP-9 (post), SPARC, and BDNF (post) were classified as moderate. For all other parameters, only weak ICCs were found. CONCLUSION As in the venous serum, there was an increase in most markers in the capillary plasma. However, acceptable to low associations can be found in the concentration levels of these proteins between the compartments. Thus, this source of blood sampling could find some biomarker applications in sports practice.
Collapse
Affiliation(s)
- Thomas Reichel
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig University Giessen, Kugelberg 62, 35394 Giessen, Germany
| | - Steffen Held
- Intervention Research in Exercise Training, German Sports University, Cologne, Germany
| | - Anthony Schwarz
- Intervention Research in Exercise Training, German Sports University, Cologne, Germany
| | - Sebastian Hacker
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig University Giessen, Kugelberg 62, 35394 Giessen, Germany
| | - Fabian Wesemann
- Intervention Research in Exercise Training, German Sports University, Cologne, Germany
| | - Lars Donath
- Intervention Research in Exercise Training, German Sports University, Cologne, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig University Giessen, Kugelberg 62, 35394 Giessen, Germany
| |
Collapse
|
13
|
Peng W, Kepsch A, Kracht TO, Hasan H, Wijayarathna R, Wahle E, Pleuger C, Bhushan S, Günther S, Kauerhof AC, Planinić A, Fietz D, Schuppe HC, Wygrecka M, Loveland KL, Ježek D, Meinhardt A, Hedger MP, Fijak M. Activin A and CCR2 regulate macrophage function in testicular fibrosis caused by experimental autoimmune orchitis. Cell Mol Life Sci 2022; 79:602. [PMID: 36434305 PMCID: PMC9700630 DOI: 10.1007/s00018-022-04632-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/21/2022] [Accepted: 11/13/2022] [Indexed: 11/27/2022]
Abstract
Experimental autoimmune-orchitis (EAO), a rodent model of chronic testicular inflammation and fibrosis, replicates pathogenic changes seen in some cases of human spermatogenic disturbances. During EAO, increased levels of pro-inflammatory and pro-fibrotic mediators such as TNF, CCL2, and activin A are accompanied by infiltration of leukocytes into the testicular parenchyma. Activin A levels correlate with EAO severity, while elevated CCL2 acting through its receptor CCR2 mediates leukocyte trafficking and recruits macrophages. CCR2 + CXCR4 + macrophages producing extracellular matrix proteins contribute widely to fibrogenesis. Furthermore, testicular macrophages (TMs) play a critical role in organ homeostasis. Therefore, we aimed to investigate the role of the activin A/CCL2-CCR2/macrophage axis in the development of testicular fibrosis. Following EAO induction, we observed lower levels of organ damage, collagen deposition, and leukocyte infiltration (including fibronectin+, collagen I+ and CXCR4+ TMs) in Ccr2-/- mice than in WT mice. Furthermore, levels of Il-10, Ccl2, and the activin A subunit Inhba mRNAs were lower in Ccr2-/- EAO testes. Notably, fibronectin+ TMs were also present in biopsies from patients with impaired spermatogenesis and fibrotic alterations. Overexpression of the activin A antagonist follistatin reduced tissue damage and collagen I+ TM accumulation in WT EAO testes, while treating macrophages with activin A in vitro increased the expression of Ccr2, Fn1, Cxcr4, and Mmp2 and enhanced migration along a CCL2 gradient; these effects were abolished by follistatin. Taken together, our data indicate that CCR2 and activin A promote fibrosis during testicular inflammation by regulating macrophage function. Inhibition of CCR2 or activin A protects against damage progression, offering a promising avenue for therapeutic intervention.
Collapse
Affiliation(s)
- Wei Peng
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Artem Kepsch
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Till O Kracht
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Hiba Hasan
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Rukmali Wijayarathna
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Eva Wahle
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Christiane Pleuger
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Stefan Günther
- ECCPS Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - A Christine Kauerhof
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Ana Planinić
- Department of Histology and Embryology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Daniela Fietz
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Hans-Christian Schuppe
- Department of Urology, Paediatric Urology and Andrology, Justus Liebig University of Giessen, Giessen, Germany
| | - Małgorzata Wygrecka
- Center for Infection and Genomics of the Lung, German Center for Lung Research, University of Giessen and Marburg Lung Center, Giessen, Germany
| | - Kate L Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Davor Ježek
- Department of Histology and Embryology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Mark P Hedger
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany.
| |
Collapse
|
14
|
Lampejo AO, Hu NW, Lucas D, Lomel BM, Nguyen CM, Dominguez CC, Ren B, Huang Y, Murfee WL. A Challenge for Engineering Biomimetic Microvascular Models: How do we Incorporate the Physiology? Front Bioeng Biotechnol 2022; 10:912073. [PMID: 35795159 PMCID: PMC9252339 DOI: 10.3389/fbioe.2022.912073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
The gap between in vitro and in vivo assays has inspired biomimetic model development. Tissue engineered models that attempt to mimic the complexity of microvascular networks have emerged as tools for investigating cell-cell and cell-environment interactions that may be not easily viewed in vivo. A key challenge in model development, however, is determining how to recreate the multi-cell/system functional complexity of a real network environment that integrates endothelial cells, smooth muscle cells, vascular pericytes, lymphatics, nerves, fluid flow, extracellular matrix, and inflammatory cells. The objective of this mini-review is to overview the recent evolution of popular biomimetic modeling approaches for investigating microvascular dynamics. A specific focus will highlight the engineering design requirements needed to match physiological function and the potential for top-down tissue culture methods that maintain complexity. Overall, examples of physiological validation, basic science discoveries, and therapeutic evaluation studies will emphasize the value of tissue culture models and biomimetic model development approaches that fill the gap between in vitro and in vivo assays and guide how vascular biologists and physiologists might think about the microcirculation.
Collapse
Affiliation(s)
- Arinola O. Lampejo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Nien-Wen Hu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Daniela Lucas
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Banks M. Lomel
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Christian M. Nguyen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Carmen C. Dominguez
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Bing Ren
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, United States
| | - Yong Huang
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, United States
| | - Walter L. Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- *Correspondence: Walter L. Murfee,
| |
Collapse
|
15
|
Nong C, Guan P, Li L, Zhang H, Hu H. Tumor immunotherapy: Mechanisms and clinical applications. MEDCOMM – ONCOLOGY 2022. [DOI: 10.1002/mog2.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Cheng Nong
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Pengbo Guan
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Li Li
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Huiyuan Zhang
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Hongbo Hu
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
- Chongqing International Institution for Immunology Chongqing China
| |
Collapse
|
16
|
Yates RL, Pansieri J, Li Q, Bell JS, Yee SA, Palace J, Esiri MM, DeLuca GC. The influence of HLA-DRB1*15 on the relationship between microglia and neurons in multiple sclerosis normal appearing cortical grey matter. Brain Pathol 2021; 32:e13041. [PMID: 34904300 PMCID: PMC9245937 DOI: 10.1111/bpa.13041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/06/2022] Open
Abstract
Cortical tissue injury is common in multiple sclerosis (MS) and associates with disability progression. We have previously shown that HLA‐DRB1*15 genotype status associates with the extent of cortical inflammatory pathology. In the current study, we sought to examine the influence of HLA‐DRB1*15 on relationships between inflammation and neurodegeneration in MS. Human post‐mortem MS cases (n = 47) and controls (n = 10) were used. Adjacent sections of motor cortex were stained for microglia (Iba1+, CD68+, TMEM119+), lymphocytes (CD3+, CD8+), GFAP+ astrocytes, and neurons (NeuN+). A subset of MS cases (n = 20) and controls (n = 7) were double‐labeled for neurofilament and glutamic acid decarboxylase 65/67 (GAD+) to assess the extent of the inhibitory synaptic loss. In MS cases, microglial protein expression positively correlated with neuron density (Iba1+: r = 0.548, p < 0.001, CD68+: r = 0.498, p = 0.001, TMEM119+ r = 0.437, p = 0.003). This finding was restricted to MS cases not carrying HLA‐DRB1*15. Evidence of a 14% reduction in inhibitory synapses in MS was detected (MS: 0.299 ± 0.006 synapses/μm2 neuronal membrane versus control: 0.348 ± 0.009 synapses/μm2 neuronal membrane, p = 0.005). Neurons expressing inhibitory synapses were 24% smaller in MS cases compared to the control (MS: 403 ± 15 μm2 versus control: 531 ± 29 μm2, p = 0.001), a finding driven by HLA‐DRB1*15+ cases (15+: 376 ± 21 μm2 vs. 15−: 432 ± 22 μm2, p = 0.018). Taken together, our results demonstrate that HLA‐DRB1*15 modulates the relationship between microglial inflammation, inhibitory synapses, and neuronal density in the MS cortex.
Collapse
Affiliation(s)
- Richard L Yates
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jonathan Pansieri
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Qizhu Li
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Jack S Bell
- Salford Royal NHS Foundation Trust, Salford, UK
| | - Sydney A Yee
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Margaret M Esiri
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
17
|
Fei L, Ren X, Yu H, Zhan Y. Targeting the CCL2/CCR2 Axis in Cancer Immunotherapy: One Stone, Three Birds? Front Immunol 2021; 12:771210. [PMID: 34804061 PMCID: PMC8596464 DOI: 10.3389/fimmu.2021.771210] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/19/2021] [Indexed: 12/15/2022] Open
Abstract
CCR2 is predominantly expressed by monocytes/macrophages with strong proinflammatory functions, prompting the development of CCR2 antagonists to dampen unwanted immune responses in inflammatory and autoimmune diseases. Paradoxically, CCR2-expressing monocytes/macrophages, particularly in tumor microenvironments, can be strongly immunosuppressive. Thus, targeting the recruitment of immunosuppressive monocytes/macrophages to tumors by CCR2 antagonism has recently been investigated as a strategy to modify the tumor microenvironment and enhance anti-tumor immunity. We present here that beneficial effects of CCR2 antagonism in the tumor setting extend beyond blocking chemotaxis of suppressive myeloid cells. Signaling within the CCL2/CCR2 axis shows underappreciated effects on myeloid cell survival and function polarization. Apart from myeloid cells, T cells are also known to express CCR2. Nevertheless, tissue homing of Treg cells among T cell populations is preferentially affected by CCR2 deficiency. Further, CCR2 signaling also directly enhances Treg functional potency. Thus, although Tregs are not the sole type of T cells expressing CCR2, the net outcome of CCR2 antagonism in T cells favors the anti-tumor arm of immune responses. Finally, the CCL2/CCR2 axis directly contributes to survival/growth and invasion/metastasis of many types of tumors bearing CCR2. Together, CCR2 links to two main types of suppressive immune cells by multiple mechanisms. Such a CCR2-assoicated immunosuppressive network is further entangled with paracrine and autocrine CCR2 signaling of tumor cells. Strategies to target CCL2/CCR2 axis as cancer therapy in the view of three types of CCR2-expessing cells in tumor microenvironment are discussed.
Collapse
Affiliation(s)
- Liyang Fei
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Xiaochen Ren
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Haijia Yu
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Yifan Zhan
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| |
Collapse
|
18
|
Damian C, Ghuman H, Mauney C, Azar R, Reinartz J, Badylak SF, Modo M. Post-Stroke Timing of ECM Hydrogel Implantation Affects Biodegradation and Tissue Restoration. Int J Mol Sci 2021; 22:ijms222111372. [PMID: 34768800 PMCID: PMC8583606 DOI: 10.3390/ijms222111372] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/13/2021] [Accepted: 10/17/2021] [Indexed: 01/01/2023] Open
Abstract
Extracellular matrix (ECM) hydrogel promotes tissue regeneration in lesion cavities after stroke. However, a bioscaffold's regenerative potential needs to be considered in the context of the evolving pathological environment caused by a stroke. To evaluate this key issue in rats, ECM hydrogel was delivered to the lesion core/cavity at 7-, 14-, 28-, and 90-days post-stroke. Due to a lack of tissue cavitation 7-days post-stroke, implantation of ECM hydrogel did not achieve a sufficient volume and distribution to warrant comparison with the other time points. Biodegradation of ECM hydrogel implanted 14- and 28-days post-stroke were efficiently (80%) degraded by 14-days post-bioscaffold implantation, whereas implantation 90-days post-stroke revealed only a 60% decrease. Macrophage invasion was robust at 14- and 28-days post-stroke but reduced in the 90-days post-stroke condition. The pro-inflammation (M1) and pro-repair (M2) phenotype ratios were equivalent at all time points, suggesting that the pathological environment determines macrophage invasion, whereas ECM hydrogel defines their polarization. Neural cells (neural progenitors, neurons, astrocytes, oligodendrocytes) were found at all time points, but a 90-days post-stroke implantation resulted in reduced densities of mature phenotypes. Brain tissue restoration is therefore dependent on an efficient delivery of a bioscaffold to a tissue cavity, with 28-days post-stroke producing the most efficient biodegradation and tissue regeneration, whereas by 90-days post-stroke, these effects are significantly reduced. Improving our understanding of how the pathological environment influences biodegradation and the tissue restoration process is hence essential to devise engineering strategies that could extend the therapeutic window for bioscaffolds to repair the damaged brain.
Collapse
Affiliation(s)
- Corina Damian
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; (C.D.); (C.M.)
| | - Harmanvir Ghuman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.G.); (R.A.); (S.F.B.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Carrinton Mauney
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; (C.D.); (C.M.)
| | - Reem Azar
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.G.); (R.A.); (S.F.B.)
| | - Janina Reinartz
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Stephen F. Badylak
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.G.); (R.A.); (S.F.B.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michel Modo
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.G.); (R.A.); (S.F.B.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15260, USA;
- Correspondence: ; Tel.: +1-(412)-383-7200
| |
Collapse
|
19
|
Bissinger S, Hage C, Wagner V, Maser IP, Brand V, Schmittnaegel M, Jegg AM, Cannarile M, Watson C, Klaman I, Rieder N, González Loyola A, Petrova TV, Cassier PA, Gomez-Roca C, Sibaud V, De Palma M, Hoves S, Ries CH. Macrophage depletion induces edema through release of matrix-degrading proteases and proteoglycan deposition. Sci Transl Med 2021; 13:13/598/eabd4550. [PMID: 34135110 DOI: 10.1126/scitranslmed.abd4550] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 05/07/2021] [Indexed: 12/21/2022]
Abstract
Colony-stimulating factor 1 receptor (CSF1R) blockade abates tumor-associated macrophage (TAM) infiltrates and provides marked clinical benefits in diffuse-type tenosynovial giant cell tumors. However, facial edema is a common adverse event associated with TAM elimination in patients. In this study, we examined molecular and cellular events associated with edema formation in mice and human patients with cancer treated with a CSF1R blocking antibody. Extended antibody treatment of mice caused marked body weight gain, an indicator of enhanced body fluid retention. This was associated with an increase of extracellular matrix-remodeling metalloproteinases (MMPs), namely MMP2 and MMP3, and enhanced deposition of hyaluronan (HA) and proteoglycans, leading to skin thickening. Discontinuation of anti-CSF1R treatment or blockade of MMP activity restored unaltered body weight and normal skin morphology in the mice. In patients, edema developed at doses well below the established optimal biological dose for emactuzumab, a CSF1R dimerization inhibitor. Patients who developed edema in response to emactuzumab had elevated HA in peripheral blood. Our findings indicate that an early increase of peripheral HA can serve as a pharmacodynamic marker for edema development and suggest potential interventions based on MMP inhibition for relieving periorbital edema in patients treated with CSF1R inhibitors.
Collapse
Affiliation(s)
- Stefan Bissinger
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany.
| | - Carina Hage
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Vinona Wagner
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Ilona-Petra Maser
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Verena Brand
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Martina Schmittnaegel
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.,Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Anna-Maria Jegg
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Michael Cannarile
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany
| | | | - Irina Klaman
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Natascha Rieder
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Alejandra González Loyola
- Department of Oncology, University of Lausanne (UNIL) and Ludwig Institute for Cancer Research Lausanne (LICR), 1066 Epalinges, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne (UNIL) and Ludwig Institute for Cancer Research Lausanne (LICR), 1066 Epalinges, Switzerland
| | | | - Carlos Gomez-Roca
- Institut Claudius Regaud/Institut Universitaire du Cancer, Toulouse Oncopole, 31300 Toulouse, France
| | - Vincent Sibaud
- Institut Claudius Regaud/Institut Universitaire du Cancer, Toulouse Oncopole, 31300 Toulouse, France
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Sabine Hoves
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Carola H Ries
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, 82377 Penzberg, Germany.
| |
Collapse
|
20
|
Abstract
Fibrinolysis is of paramount importance in maintaining or regaining the patency of veins and pulmonary arteries obstructed by thrombi. Growing experimental and clinical evidence indicates that impaired fibrinolysis mediated by multiple complex mechanisms is involved in venous thromboembolism (VTE). Global plasma fibrin clot lysis markers, especially clot lysis time, have been reported to predict recurrent deep-vein thrombosis and pulmonary embolism. The current overview summarizes available data linking fibrinolysis to VTE and its long-term sequelae.
Collapse
Affiliation(s)
- Anetta Undas
- Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland.,Krakow Centre for Medical Research and Technologies, John Paul II Hospital, Krakow, Poland
| |
Collapse
|
21
|
Hermes DJ, Yadav-Samudrala BJ, Xu C, Paniccia JE, Meeker RB, Armstrong ML, Reisdorph N, Cravatt BF, Mackie K, Lichtman AH, Ignatowska-Jankowska BM, Lysle DT, Fitting S. GPR18 drives FAAH inhibition-induced neuroprotection against HIV-1 Tat-induced neurodegeneration. Exp Neurol 2021; 341:113699. [PMID: 33736974 PMCID: PMC8984429 DOI: 10.1016/j.expneurol.2021.113699] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/05/2021] [Accepted: 03/11/2021] [Indexed: 02/03/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) is known to provoke microglial immune responses which likely play a paramount role in the development of chronic neuroinflammatory conditions and neuronal damage related to HIV-1 associated neurocognitive disorders (HAND). In particular, HIV-1 Tat protein is a proinflammatory neurotoxin which predisposes neurons to synaptodendritic injury. Drugs targeting the degradative enzymes of endogenous cannabinoids have shown promise in reducing inflammation with minimal side effects in rodent models. Considering that markers of neuroinflammation can predict the extent of neuronal injury in HAND patients, we evaluated the neurotoxic effect of HIV-1 Tat-exposed microglia following blockade of fatty acid amid hydrolyze (FAAH), a catabolic enzyme responsible for degradation of endocannabinoids, e.g. anandamide (AEA). In the present study, cultured murine microglia were incubated with Tat and/or a FAAH inhibitor (PF3845). After 24 h, cells were imaged for morphological analysis and microglial conditioned media (MCM) was collected. Frontal cortex neuron cultures (DIV 7–11) were then exposed to MCM, and neurotoxicity was assessed via live cell calcium imaging and staining of actin positive dendritic structures. Results demonstrate a strong attenuation of microglial responses to Tat by PF3845 pretreatment, which is indicated by 1) microglial changes in morphology to a less proinflammatory phenotype using fractal analysis, 2) a decrease in release of neurotoxic cytokines/chemokines (MCP-1/CCL2) and matrix metalloproteinases (MMPs; MMP-9) using ELISA/multiplex assays, and 3) enhanced production of endocannabinoids (AEA) using LC/MS/MS. Additionally, PF3845’s effects on Tat-induced microglial-mediated neurotoxicity, decreased dysregulation of neuronal intracellular calcium and prevented the loss of actin-positive staining and punctate structure in frontal cortex neuron cultures. Interestingly, these observed neuroprotective effects appeared to be independent of cannabinoid receptor activity (CB1R & CB2R). We found that a purported GPR18 antagonist, CID-85469571, blocked the neuroprotective effects of PF3845 in all experiments. Collectively, these experiments increase understanding of the role of FAAH inhibition and Tat in mediating microglial neurotoxicity in the HAND condition.
Collapse
Affiliation(s)
- Douglas J Hermes
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America.
| | - Barkha J Yadav-Samudrala
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Changqing Xu
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Jacqueline E Paniccia
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Rick B Meeker
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Michael L Armstrong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Denver, CO, United States of America
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Denver, CO, United States of America
| | - Benjamin F Cravatt
- Department of Chemistry, Scripps Research Institute, La Jolla, CA, United States of America
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States of America
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States of America
| | | | - Donald T Lysle
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Sylvia Fitting
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America.
| |
Collapse
|
22
|
Byanova KL, Kunisaki KM, Vasquez J, Huang L. Chronic obstructive pulmonary disease in HIV. Expert Rev Respir Med 2021; 15:71-87. [PMID: 33167728 PMCID: PMC7856058 DOI: 10.1080/17476348.2021.1848556] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023]
Abstract
Introduction: Chronic obstructive pulmonary disease (COPD) is more prevalent in people with HIV (PWH) than in the general population and leads to an increased burden of morbidity and mortality in this population. The mechanisms behind COPD development and progression in PWH are not fully elucidated, and there are no PWH-specific guidelines for COPD management. Areas covered: The goal of this broad narrative review is to review the epidemiology of COPD in PWH globally, highlight proposed pathways contributing to increased COPD prevalence and progression in PWH, discuss structural and functional changes in the lungs in this population, assesses the excess mortality and comorbidities in PWH with COPD, and address management practices for this unique population. Expert opinion: Understanding how a chronic viral infection leads to COPD, independent of cigarette smoking, is of critical scientific importance. Further research should focus on the pathophysiology of the interaction between HIV and COPD, and determine the role of disease-modifying risk factors such as opportunistic pneumonia and air pollution, as well as generate data from randomized clinical trials on the safety and efficacy of specific therapies for this vulnerable patient population.
Collapse
Affiliation(s)
- Katerina L Byanova
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Ken M. Kunisaki
- Section of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Joshua Vasquez
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Laurence Huang
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- HIV, Infectious Diseases, and Global Medicine Division, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
23
|
Enayati M, Puchhammer S, Iturri J, Grasl C, Kaun C, Baudis S, Walter I, Schima H, Liska R, Wojta J, Toca-Herrera JL, Podesser BK, Bergmeister H. Assessment of a long-term in vitro model to characterize the mechanical behavior and macrophage-mediated degradation of a novel, degradable, electrospun poly-urethane vascular graft. J Mech Behav Biomed Mater 2020; 112:104077. [PMID: 32942230 DOI: 10.1016/j.jmbbm.2020.104077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/13/2020] [Accepted: 08/23/2020] [Indexed: 10/23/2022]
Abstract
An assessment tool to evaluate the degradation of biodegradable materials in a more physiological environment is still needed. Macrophages are critical players in host response, remodeling and degradation. In this study, a cell culture model using monocyte-derived primary macrophages was established to study the degradation, macro-/micro-mechanical behavior and inflammatory behavior of a new designed, biodegradable thermoplastic polyurethane (TPU) scaffold, over an extended period of time in vitro. For in vivo study, the scaffolds were implanted subcutaneously in a rat model for up to 36 weeks. TPU scaffolds were fabricated via the electrospinning method. This technique provided a fibrous scaffold with an average fiber diameter of 1.39 ± 0.76 μm and an average pore size of 7.5 ± 1.1 μm. The results showed that TPU scaffolds supported the attachment and migration of macrophages throughout the three-dimensional matrix. Scaffold degradation could be detected in localized areas, emphasizing the role of adherent macrophages in scaffold degradation. Weight loss, molecular weight and biomechanical strength reduction were evident in the presence of the primary macrophage cells. TPU favored the switch from initial pro-inflammatory response of macrophages to an anti-inflammatory response over time both in vitro and in vivo. Expression of MMP-2 and MMP-9 (the key enzymes in tissue remodeling based on ECM modifications) was also evident in vitro and in vivo. This study showed that the primary monocyte-derived cell culture model represents a promising tool to characterize the degradation, mechanical behavior as well as biocompatibility of the scaffolds during an extended period of observation.
Collapse
Affiliation(s)
- Marjan Enayati
- Center for Biomedical Research, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Sarah Puchhammer
- Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Jagoba Iturri
- Institute for Biophysics, Department of Nanobiotechnology, BOKU University for Natural Resources and Life Sciences, Vienna, Austria
| | - Christian Grasl
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria
| | - Christoph Kaun
- Division of Internal Medicine II, Medical University Vienna, Austria
| | - Stefan Baudis
- Institute of Applied Synthetic Chemistry, Technische Universität Wien, Vienna, Austria
| | - Ingrid Walter
- Department of Pathobiology, Veterinary University, Vienna, Austria
| | - Heinrich Schima
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria
| | - Robert Liska
- Institute of Applied Synthetic Chemistry, Technische Universität Wien, Vienna, Austria
| | - Johann Wojta
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Division of Internal Medicine II, Medical University Vienna, Austria
| | - José Luis Toca-Herrera
- Institute for Biophysics, Department of Nanobiotechnology, BOKU University for Natural Resources and Life Sciences, Vienna, Austria
| | - Bruno K Podesser
- Center for Biomedical Research, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Helga Bergmeister
- Center for Biomedical Research, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria.
| |
Collapse
|
24
|
Xu Z, Liu A, Li S, Wang G, Ye H. Hepatopancreas immune response during molt cycle in the mud crab, Scylla paramamosain. Sci Rep 2020; 10:13102. [PMID: 32753724 PMCID: PMC7403367 DOI: 10.1038/s41598-020-70139-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 07/20/2020] [Indexed: 11/18/2022] Open
Abstract
Molt is a critical developmental process in crustaceans. Recent studies have shown that the hepatopancreas is an important source of innate immune molecules, yet hepatopancreatic patterns of gene expression during the molt cycle which may underlie changes in immune mechanism are unknown. In this study, we performed Illumina sequencing for the hepatopancreas of the mud crab, Scylla paramamosain during molt cycle (pre-molt stage, post-molt stage, and inter-molt stage). A total of 44.55 Gb high-quality reads were obtained from the normalized cDNA of hepatopancreas. A total of 70,591 transcripts were assembled; 55,167 unigenes were identified. Transcriptomic comparison revealed 948 differentially expressed genes (DEGs) in the hepatopancreas from the three molt stages. We found that genes associated with immune response patterns changed in expression during the molt cycle. Antimicrobial peptide genes, inflammatory response genes, Toll signaling pathway factors, the phenoloxidase system, antioxidant enzymes, metal-binding proteins and other immune related genes are significantly up-regulated at the post-molt stage and inter-molt stage compared with the pre-molt stage, respectively. These genes are either not expressed or are expressed at low levels at the pre-molt stage. To our knowledge, this is the first systematic transcriptome analysis of genes capable of mobilizing a hepatopancreas immune response during the molt cycle in crustaceans, and this study will contribute to a better understanding of the hepatopancreas immune system and mud crab prophylactic immune mechanisms at the post-molt stage.
Collapse
Affiliation(s)
- Zhanning Xu
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - An Liu
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Shengkang Li
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China
| | - Guizhong Wang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Haihui Ye
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
25
|
Abstract
4-hexylresorcinol (4-HR), as a derivative of phenolic lipids, has biological and pharmacological properties that are beneficial when used with a biomaterial. It has antimicrobial and antiseptic activity and can thus prevent contamination and infection of biomaterials. 4-HR suppresses the nuclear factor kappa B (NF-κB) signaling pathway related to osteoclast differentiation. The suppression of NF-κB increases the bone formation marker and contributes to new bone formation. The tumor necrosis factor-α (TNF-α) is a pro-inflammatory cytokine produced by macrophages and suppressed by 4-HR. Suppression of TNF-α decreases osteoclast activity and promotes wound healing. 4-HR increases the vascular endothelial growth factor and has an anti-thrombotic effect. When incorporated into silk vascular patches, it promotes endothelium wound healing. Recently, 4-HR has exhibited biological properties and has been successfully incorporated into various biomaterials. Consequently, it is a useful pharmacological chemical that can be used with biomaterials in the field of tissue engineering.
Collapse
|
26
|
Malik A, Ashraf MAB, Khan MW, Zahid A, Shafique H, Waquar S, Gan SH, Ashraf M. Implication of Physiological and Biochemical Variables of Prognostic Importance in Lead Exposed Subjects. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2020; 78:329-336. [PMID: 31620805 DOI: 10.1007/s00244-019-00673-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 10/09/2019] [Indexed: 06/10/2023]
Abstract
The use of leaded gasoline adversely affects cardiovascular, nervous, and immune systems. Study projects to rule out different variables of prognostic importance in lead-exposed subjects. A total of 317 traffic wardens with 5 years of outdoor experience and Hb levels < 10 µg/dl, and 100 traffic wardens with indoor duties were substituted in two groups. Levels of vitamins, cytokines, lead, iron, minerals, oxidative stress, and lipid peroxidation were estimated with help of their standard ELISA and spectrophotometric methods respectively. The present study show increased levels of lead in subjects (29.8 ± 3.8 vs. 1.5 ± 0.2 µg/dl) that may be involved in increasing oxidative stress, i.e., levels of malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and isoprostanes were increased in subjects (4.6 ± 0.5, 4.3 ± 0.6 and 37.2 ± 5.1). Moreover, levels of antioxidants, i.e., superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT), were decreased. It also exhibits reduced levels of different enzymes in anemic traffic wardens. Current study concludes that wardens exposed to environmental lead are more susceptible to develop cardiovascular and neurological disorders. It shows that toxicity of lead maybe responsible for redox imbalance and production of proinflammatory cytokines. Thus, early detection of these biomarkers may help to reduce lead toxicity and it also may help to control the dilemma of uncontrolled environmental pollution by implicating strict actions against substandard gasoline.
Collapse
Affiliation(s)
- Arif Malik
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | | | | | - Ayesha Zahid
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Hassan Shafique
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Sulayman Waquar
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Siew Hua Gan
- School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Muhammad Ashraf
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan.
| |
Collapse
|
27
|
Venkatesh S, Asha S, Krishnaveni M. Purification of Matrixins from Marine Cephalopod. Protein J 2020; 39:284-290. [PMID: 32185695 DOI: 10.1007/s10930-020-09893-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Matrixins play a major role in tissue regeneration and also in various patho-physiological processes. Discovery of matrix metallo proteins (MMPs) and their detailed structural and functional analysis would lead to the development of numerous potent synthetic inhibitors of matrixins to treat certain diseases. In the present investigation, a marine cephalopod- Octopus sp. collected from Cochin, in the south western Indian Ocean was used as animal model for purification of matrixins. The measurements, count, indices and other morphometric characters were noted down before assessing the presence of matrixins in the crude extract of Octopus samples. Purification of matrixins was carried out employing gel filtration chromatography and the purified matrixins was confirmed by gelatin zymogram. The purity of the protein was checked by both native and SDS-PAGE. The studies have provided clear indications of production of MMPs or matrixins with gelatinolytic activity in Octopus sp.
Collapse
Affiliation(s)
- S Venkatesh
- Genetic Engineering and Regenerative Biology Lab, Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu, 627012, India
| | - S Asha
- Immuno-Pharmacology Laboratory, Center for Marine Science and Technology, Manonmaniam Sundaranar University, Marina campus, Rajakkamangalam, Kanyakumari, Tamilnadu, 629502, India
| | - M Krishnaveni
- Immuno-Pharmacology Laboratory, Center for Marine Science and Technology, Manonmaniam Sundaranar University, Marina campus, Rajakkamangalam, Kanyakumari, Tamilnadu, 629502, India.
| |
Collapse
|
28
|
Du Cheyne C, Tay H, De Spiegelaere W. The complex TIE between macrophages and angiogenesis. Anat Histol Embryol 2019; 49:585-596. [PMID: 31774212 DOI: 10.1111/ahe.12518] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/30/2019] [Accepted: 11/08/2019] [Indexed: 12/20/2022]
Abstract
Macrophages are primarily known as phagocytic immune cells, but they also play a role in diverse processes, such as morphogenesis, homeostasis and regeneration. In this review, we discuss the influence of macrophages on angiogenesis, the process of new blood vessel formation from the pre-existing vasculature. Macrophages play crucial roles at each step of the angiogenic cascade, starting from new blood vessel sprouting to the remodelling of the vascular plexus and vessel maturation. Macrophages form promising targets for both pro- and anti-angiogenic treatments. However, to target macrophages, we will first need to understand the mechanisms that control the functional plasticity of macrophages during each of the steps of the angiogenic cascade. Here, we review recent insights in this topic. Special attention will be given to the TIE2-expressing macrophage (TEM), which is a subtype of highly angiogenic macrophages that is able to influence angiogenesis via the angiopoietin-TIE pathway.
Collapse
Affiliation(s)
- Charis Du Cheyne
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Hanna Tay
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Ward De Spiegelaere
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
29
|
Abstract
Brain tissue lost after a stroke is not regenerated, although a repair response associated with neurogenesis does occur. A failure to regenerate functional brain tissue is not caused by the lack of available neural cells, but rather the absence of structural support to permit a repopulation of the lesion cavity. Inductive bioscaffolds can provide this support and promote the invasion of host cells into the tissue void. The putative mechanisms of bioscaffold degradation and its pivotal role to permit invasion of neural cells are reviewed and discussed in comparison to peripheral wound healing. Key differences between regenerating and non-regenerating tissues are contrasted in an evolutionary context, with a special focus on the neurogenic response as a conditio sine qua non for brain regeneration. The pivotal role of the immune system in biodegradation and the formation of a neovasculature are contextualized with regeneration of peripheral soft tissues. The application of rehabilitation to integrate newly forming brain tissue is suggested as necessary to develop functional tissue that can alleviate behavioral impairments. Pertinent aspects of brain tissue development are considered to provide guidance to produce a metabolically and functionally integrated de novo tissue. Although little is currently known about mechanisms involved in brain tissue regeneration, this review outlines the various components and their interplay to provide a framework for ongoing and future studies. It is envisaged that a better understanding of the mechanisms involved in brain tissue regeneration will improve the design of biomaterials and the methods used for implantation, as well as rehabilitation strategies that support the restoration of behavioral functions.
Collapse
Affiliation(s)
- Michel Modo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States,Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States,Department of Radiology, University of Pittsburgh, Pittsburgh, PA, United States,*Correspondence: Michel Modo,
| |
Collapse
|
30
|
The Role of Urokinase, Tumor Necrosis Factor, and Matrix Metalloproteinase-9 in Monocyte Activation. Bull Exp Biol Med 2019; 167:492-495. [DOI: 10.1007/s10517-019-04557-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Indexed: 10/26/2022]
|
31
|
Human Immunodeficiency Virus (HIV) Infection and Use of Illicit Substances Promote Secretion of Semen Exosomes that Enhance Monocyte Adhesion and Induce Actin Reorganization and Chemotactic Migration. Cells 2019; 8:cells8091027. [PMID: 31484431 PMCID: PMC6770851 DOI: 10.3390/cells8091027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/09/2019] [Accepted: 08/15/2019] [Indexed: 12/23/2022] Open
Abstract
Semen exosomes (SE) from HIV-uninfected (HIV−) individuals potently inhibit HIV infection in vitro. However, morphological changes in target cells in response to SE have not been characterized or have the effect of HIV infection or the use of illicit substances, specifically psychostimulants, on the function of SE been elucidated. The objective of this study was to evaluate the effect of HIV infection, psychostimulant use, and both together on SE-mediated regulation of monocyte function. SE were isolated from semen of HIV− and HIV-infected (HIV+) antiretroviral therapy (ART)-naive participants who reported either using or not using psychostimulants. The SE samples were thus designated as HIV−Drug−, HIV−Drug+, HIV+Drug−, and HIV+Drug+. U937 monocytes were treated with different SEs and analyzed for changes in transcriptome, morphometrics, actin reorganization, adhesion, and chemotaxis. HIV infection and/or use of psychostimulants had minimal effects on the physical characteristics of SE. However, different SEs had diverse effects on the messenger RNA signature of monocytes and rapidly induced monocyte adhesion and spreading. SE from HIV infected or psychostimulants users but not HIV−Drug− SE, stimulated actin reorganization, leading to the formation of filopodia-like structures and membrane ruffles containing F-actin and vinculin that in some cases were colocalized. All SE stimulated monocyte chemotaxis to HIV secretome and activated the secretion of matrix metalloproteinases, a phenotype exacerbated by HIV infection and psychostimulant use. SE-directed regulation of cellular morphometrics and chemotaxis depended on the donor clinical status because HIV infection and psychostimulant use altered SE function. Although our inclusion criteria specified the use of cocaine, humans are poly-drug and alcohol users and our study participants used psychostimulants, marijuana, opiates, and alcohol. Thus, it is possible that the effects observed in this study may be due to one of these other substances or due to an interaction between different substances.
Collapse
|
32
|
Campbell WA, Deshmukh A, Blum S, Todd L, Mendonca N, Weist J, Zent J, Hoang TV, Blackshaw S, Leight J, Fischer AJ. Matrix-metalloproteinase expression and gelatinase activity in the avian retina and their influence on Müller glia proliferation. Exp Neurol 2019; 320:112984. [PMID: 31251936 DOI: 10.1016/j.expneurol.2019.112984] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/15/2019] [Accepted: 06/24/2019] [Indexed: 12/16/2022]
Abstract
Gelatinases are a class of matrix metalloproteinases (MMPs) that degrade the extracellular matrix (ECM) to regulate intercellular signaling and cell migration. Gelatinase activity is tightly regulated via proteolytic activation and through the expression of tissue inhibitors of matrix metalloproteinases (TIMPs). Gelatinase activity has been implicated in retinal pathophysiology in different animal models and human disease. However, the role of gelatinases in retinal regeneration remains uncertain. In this study we investigated the dynamic changes in gelatinase activity in response to excitotoxic damage and how this enzymatic activity influenced the formation of Müller glia progenitor cells (MGPCs) in the avian retina. This study used hydrogels containing a gelatinase-degradable fluorescent peptide to measure gelatinase activity in vitro and dye quenched gelatin to localize enzymatic activity in situ. These data were corroborated by using single cell RNA sequencing (scRNA-seq). Gelatinase mRNA, specifically MMP2, was detected in oligodendrocytes and Non-Astrocytic Inner Retinal Glia (NIRG). Total retinal gelatinase activity was reduced following NMDA-treatment, and sustained inhibition of MMP2 prior to damage or growth factor treatment increased the formation of proliferating MGPCs and c-fos signaling. We observed that microglia, Müller glia (MG), and NIRG cells were involved in regulating changes in gelatinase activity through TIMP2 and TIMP3. Collectively, these findings implicate MMP2 in reprogramming of Muller glia into MGPCs.
Collapse
Affiliation(s)
- Warren A Campbell
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States of America
| | - Ameya Deshmukh
- Department of Biomedical Engineering, College of Engineering, The comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Sydney Blum
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States of America
| | - Levi Todd
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States of America
| | - Ninoshka Mendonca
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States of America
| | - Jessica Weist
- Department of Biomedical Engineering, College of Engineering, The comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Joshua Zent
- Department of Biomedical Engineering, College of Engineering, The comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jennifer Leight
- Department of Biomedical Engineering, College of Engineering, The comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States of America.
| |
Collapse
|
33
|
Mukhopadhyay S, Johnson TA, Duru N, Buzza MS, Pawar NR, Sarkar R, Antalis TM. Fibrinolysis and Inflammation in Venous Thrombus Resolution. Front Immunol 2019; 10:1348. [PMID: 31258531 PMCID: PMC6587539 DOI: 10.3389/fimmu.2019.01348] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022] Open
Abstract
Clinical observations and accumulating laboratory evidence support a complex interplay between coagulation, inflammation, innate immunity and fibrinolysis in venous thromboembolism (VTE). VTE, which includes deep vein thrombosis (DVT) and pulmonary embolism (PE), and the subsequent complications of post-thrombotic syndrome (PTS), are significant causes of morbidity and mortality in patients. Clinical risk factors for VTE include cancer, major trauma, surgery, sepsis, inflammatory bowel disease, paralysis, prolonged periods of immobility, and aging. Abnormalities in venous blood flow or stasis initiates the activation of endothelial cells, and in concert with platelets, neutrophils and monocytes, propagates VTE in an intact vein. In addition, inflammatory cells play crucial roles in thrombus recanalization and restoration of blood flow via fibrinolysis and vascular remodeling. Faster resolution of the thrombus is key for improved disease prognosis. While in the clinical setting, anticoagulation therapy is successful in preventing propagation of venous thrombi, current therapies are not designed to inhibit inflammation, which can lead to the development of PTS. Animal models of DVT have provided many insights into the molecular and cellular mechanisms involved in the formation, propagation, and resolution of venous thrombi as well as the roles of key components of the fibrinolytic system in these processes. Here, we review the recent advances in our understanding of fibrinolysis and inflammation in the resolution of VTE.
Collapse
Affiliation(s)
- Subhradip Mukhopadhyay
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tierra A. Johnson
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nadire Duru
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marguerite S. Buzza
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nisha R. Pawar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rajabrata Sarkar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Toni M. Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
34
|
Hong SH, Shin JS, Chung H, Park CG. Galectin-4 Interaction with CD14 Triggers the Differentiation of Monocytes into Macrophage-like Cells via the MAPK Signaling Pathway. Immune Netw 2019; 19:e17. [PMID: 31281714 PMCID: PMC6597441 DOI: 10.4110/in.2019.19.e17] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/13/2019] [Accepted: 05/19/2019] [Indexed: 02/08/2023] Open
Abstract
Galectin-4 (Gal-4) is a β-galactoside-binding protein mostly expressed in the gastrointestinal tract of animals. Although intensive functional studies have been done for other galectin isoforms, the immunoregulatory function of Gal-4 still remains ambiguous. Here, we demonstrated that Gal-4 could bind to CD14 on monocytes and induce their differentiation into macrophage-like cells through the MAPK signaling pathway. Gal-4 induced the phenotypic changes on monocytes by altering the expression of various surface molecules, and induced functional changes such as increased cytokine production and matrix metalloproteinase expression and reduced phagocytic capacity. Concomitant with these changes, Gal-4-treated monocytes became adherent and showed elongated morphology with higher expression of macrophage markers. Notably, we found that Gal-4 interacted with CD14 and activated the MAPK signaling cascade. Therefore, these findings suggest that Gal-4 may exert the immunoregulatory functions through the activation and differentiation of monocytes.
Collapse
Affiliation(s)
- So-Hee Hong
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul 03080, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul 03080, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jun-Seop Shin
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul 03080, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul 03080, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyunwoo Chung
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul 03080, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Chung-Gyu Park
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul 03080, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul 03080, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
35
|
Tissue Inhibitor of Metalloproteinase-2 Polymorphisms and Risk for HIV-Associated Neurocognitive Disorder. Mediators Inflamm 2019; 2019:8278095. [PMID: 31275061 PMCID: PMC6558609 DOI: 10.1155/2019/8278095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/15/2019] [Indexed: 11/17/2022] Open
Abstract
The imbalance between MMPs and TIMPs is associated with the HIV dissemination tissue damage pathology neurodegenerative disorders, including HAND. Genetic variations in the TIMP gene may modulate the neurocognitive disorder in HIV patients. Hence, we evaluated the genetic variants of TIMP-2 (-418G/C, 303G/A) gene with the risk of HAND. Genotyping of TIMP-2 polymorphism was performed in 50 patients with HAND, 100 no HAND, and 154 healthy controls by PCR-RFLP. TIMP-2 -418GC and 303AA genotypes represented a predominant risk for HAND severity (OR = 1.55, P = 0.30; OR = 4.58, P = 0.24). The variant -418CC genotype, -418A allele, had exhibited a significant risk for the acquisition of HAND (OR = 12.55, P = 0.026; OR = 2.66, P = 0.004). TIMP-2 303GA, 303AA genotype, and 303A allele evinced a higher risk for HAND severity (OR = 1.82, P = 0.14; OR = 1.70, P = 0.63; and OR = 1.68, P = 0.12). In HIV patients, TIMP-2 -418CC genotype and -418C allele significantly occurred in comparison to healthy controls (OR = 10.10, P = 0.006; OR = 2.02, P = 0.009). In the intermediate and early HIV disease stage, TIMP-2 -418CC genotype was significantly increased compared with healthy controls (11.1% vs. 1.3%, OR = 14.63, P = 0.01; 16.9% vs. 1.3%, OR = 14.51, P = 0.002). In patients with HAND among tobacco and alcohol users, TIMP-2 -418CC genotype displayed a risk for HAND severity (OR = 3.96, P = 0.26; OR = 4.83, P = 0.19). On multivariate logistic regression, TIMP-2 303AA genotype, advanced stage, and gender had a risk for HAND severity (OR = 28.98, P = 0.02; OR = 2.35, P = 0.070; and OR = 2.36, P = 0.04). In conclusion, TIMP-2 -418G/C polymorphism independently, along with alcohol and tobacco, may have an impact on the acquisition of HAND and its severity. TIMP-2 303G/A polymorphism bare a risk for HAND severity.
Collapse
|
36
|
Duffy DM, Ko C, Jo M, Brannstrom M, Curry TE. Ovulation: Parallels With Inflammatory Processes. Endocr Rev 2019; 40:369-416. [PMID: 30496379 PMCID: PMC6405411 DOI: 10.1210/er.2018-00075] [Citation(s) in RCA: 279] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 11/18/2018] [Indexed: 12/14/2022]
Abstract
The midcycle surge of LH sets in motion interconnected networks of signaling cascades to bring about rupture of the follicle and release of the oocyte during ovulation. Many mediators of these LH-induced signaling cascades are associated with inflammation, leading to the postulate that ovulation is similar to an inflammatory response. First responders to the LH surge are granulosa and theca cells, which produce steroids, prostaglandins, chemokines, and cytokines, which are also mediators of inflammatory processes. These mediators, in turn, activate both nonimmune ovarian cells as well as resident immune cells within the ovary; additional immune cells are also attracted to the ovary. Collectively, these cells regulate proteolytic pathways to reorganize the follicular stroma, disrupt the granulosa cell basal lamina, and facilitate invasion of vascular endothelial cells. LH-induced mediators initiate cumulus expansion and cumulus oocyte complex detachment, whereas the follicular apex undergoes extensive extracellular matrix remodeling and a loss of the surface epithelium. The remainder of the follicle undergoes rapid angiogenesis and functional differentiation of granulosa and theca cells. Ultimately, these functional and structural changes culminate in follicular rupture and oocyte release. Throughout the ovulatory process, the importance of inflammatory responses is highlighted by the commonalities and similarities between many of these events associated with ovulation and inflammation. However, ovulation includes processes that are distinct from inflammation, such as regulation of steroid action, oocyte maturation, and the eventual release of the oocyte. This review focuses on the commonalities between inflammatory responses and the process of ovulation.
Collapse
Affiliation(s)
- Diane M Duffy
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - CheMyong Ko
- Department of Comparative Biosciences, University of Illinois Urbana Champaign, Urbana, Illinois
| | - Misung Jo
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, Kentucky
| | - Mats Brannstrom
- Department of Obstetrics and Gynecology, University of Gothenburg, Gothenburg, Sweden.,Stockholm IVF, Stockholm, Sweden
| | - Thomas E Curry
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
37
|
Cellular and Molecular Effects of High-Molecular-Weight Heparin on Matrix Metalloproteinase 9 Expression. Int J Mol Sci 2019; 20:ijms20071595. [PMID: 30935029 PMCID: PMC6479594 DOI: 10.3390/ijms20071595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/22/2019] [Accepted: 03/28/2019] [Indexed: 12/17/2022] Open
Abstract
Blood sampling with different anticoagulants alters matrix metalloproteinase (MMP-) 9 expression, thus influencing its concentration and diagnostic validity. Here, we aimed to evaluate the effects of different anticoagulants on MMP-9 regulation. MMP-9 expression was assessed in response to ethylenediaminetetraacetic acid, citrate, and high-/low-molecular-weight heparin (HMWH, LMWH) in co-culture experiments using THP-1, Jurkat, and HT cells (representing monocytes, T, and B cells). Triple and double cell line co-culture experiments revealed that HMWH treatment of THP-1 and Jurkat led to a significant MMP-9 induction, whereas other anticoagulants and cell type combinations had no effect. Supernatant of HMWH-treated Jurkat cells also induced MMP-9 in THP-1 suggesting monocytes as MMP-9 producers. HMWH-induced cytokine/chemokine secretion was assessed in co-culture supernatant, and the influence of cytokines/chemokines on MMP-9 production was analyzed. These experiments revealed that Jurkat-derived IL-16 and soluble intercellular adhesion molecule (sICAM-) 1 are able to induce MMP-9 and IL-8 production by THP-1. As a consequence, the increased MMP-9 expression found in HMWH blood samples may be influenced by HMWH-dependent secretion of IL-16 and sICAM-1 by T cells resulting in an increased production of MMP-9 and IL-8 by monocytes. IL-8, in turn, may support MMP-9 and its own expression in a positive autocrine feedback loop.
Collapse
|
38
|
Singh H, Nain S, Krishnaraj A, Lata S, Dhole TN. Genetic variation of matrix metalloproteinase enzyme in HIV-associated neurocognitive disorder. Gene 2019; 698:41-49. [PMID: 30825593 DOI: 10.1016/j.gene.2019.02.057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/14/2019] [Accepted: 02/17/2019] [Indexed: 01/13/2023]
Abstract
Matrix metalloproteinases (MMPs) play a key role in several diseases such as rheumatoid arthritis, HIV-associated neurological diseases (HAND), multiple sclerosis, osteoporosis, stroke, Alzheimer's disease, certain viral infections of the central nervous system, cancer, and hepatitis C virus. MMPs have been explained with regards to extracellular matrix remodeling, which occurs throughout life and ranges from tissue morphogenesis to wound healing in various processes. MMP are inhibited by endogenous tissue inhibitors of metalloproteinases (TIMPs). Matrix metalloproteases act as an interface between host's attack by Tat protein of HIV-1 virus and extracellular matrix, which causes breaches in the endothelial barriers by degrading ECM. This process initiates the dissemination of virus in tissues which can lead to an increase HIV-1 infection. MMPs are diverse and are highly polymorphic in nature, hence associated with many diseases. The main objective of this review is to study the gene expression of MMPs in HIV-related diseases and whether TIMPs and MMPs could be related with disease progression, HIV vulnerability and HAND. In this review, a brief description on the classification, regulation of MMP and TIMP, the effect of different MMPs and TIMPs gene polymorphisms and its expression on HIV-associated diseases have been provided. Previous studies have shown that MMPs polymorphism (MMP-1, MMP-2 MMP3, and MMP9) plays an important role in HIV vulnerability, disease progression and HAND. Further research is required to explore their role in pathogenesis and therapeutic perspective.
Collapse
Affiliation(s)
- HariOm Singh
- Department of Molecular Biology, National AIDS Research Institute, Pune 411026, India.
| | - Sumitra Nain
- Department of Pharmacy, University of Banasthali, Banasthali Vidyapith, Jaipur 302001, India
| | - Asha Krishnaraj
- Department of Pharmacotherapy, University of Utah, Salt Lake City, UT 84108, USA
| | - Sonam Lata
- Department of Molecular Biology, National AIDS Research Institute, Pune 411026, India
| | - T N Dhole
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
39
|
Olkowski R, Czarnowska E, Wojasiński M, Niderla-Bielińska J, Ciach T, Ratajska A. Three-dimensional nanofibrous polystyrene scaffolds modify macrophage phenotypes and activate macrophage angiogenic potential. Cell Biol Int 2019; 43:265-278. [DOI: 10.1002/cbin.11094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 12/23/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Radosław Olkowski
- Department of Pathology, Center for Biostructure; Medical University of Warsaw; Chałubińskiego 5 Warsaw 02-004 Poland
| | - Elżbieta Czarnowska
- Department of Pathology; Children's Memorial Health Institute; Warsaw Poland
| | - Michał Wojasiński
- BioMedical Engineering Laboratory; Biotechnology and Bioprocess Engineering Division; Warsaw University of Technology; Warsaw Poland
| | - Justyna Niderla-Bielińska
- Department of Histology and Embryology; Center for Biostructure; Medical University of Warsaw; Warsaw Poland
| | - Tomasz Ciach
- BioMedical Engineering Laboratory; Biotechnology and Bioprocess Engineering Division; Warsaw University of Technology; Warsaw Poland
| | - Anna Ratajska
- Department of Pathology, Center for Biostructure; Medical University of Warsaw; Chałubińskiego 5 Warsaw 02-004 Poland
| |
Collapse
|
40
|
Jiang Z, You Q, Zhang X. Medicinal chemistry of metal chelating fragments in metalloenzyme active sites: A perspective. Eur J Med Chem 2019; 165:172-197. [PMID: 30684796 DOI: 10.1016/j.ejmech.2019.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 12/22/2018] [Accepted: 01/08/2019] [Indexed: 12/25/2022]
Abstract
Numerous metal-containing enzymes (metalloenzymes) have been considered as drug targets related to diseases such as cancers, diabetes, anemia, AIDS, malaria, bacterial infection, fibrosis, and neurodegenerative diseases. Inhibitors of the metalloenzymes have been developed independently, most of which are mimics of substrates of the corresponding enzymes. However, little attention has been paid to the interactions between inhibitors and active site metal ions. This review is focused on different metal binding fragments and their chelating properties in the metal-containing active binding pockets of metalloenzymes. We have enumerated over one hundred of inhibitors targeting various metalloenzymes and identified over ten kinds of fragments with different binding patterns. Furthermore, we have investigated the inhibitors that are undergoing clinical evaluation in order to help looking for more potential scaffolds bearing metal binding fragments. This review will provide deep insights for the rational design of novel inhibitors targeting the metal-containing binding sites of specific proteins.
Collapse
Affiliation(s)
- Zhensheng Jiang
- Sate Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- Sate Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiaojin Zhang
- Sate Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
41
|
How post-translational modifications influence the biological activity of chemokines. Cytokine 2018; 109:29-51. [DOI: 10.1016/j.cyto.2018.02.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/27/2018] [Accepted: 02/28/2018] [Indexed: 12/17/2022]
|
42
|
Annamalai RT, Turner PA, Carson WF, Levi B, Kunkel S, Stegemann JP. Harnessing macrophage-mediated degradation of gelatin microspheres for spatiotemporal control of BMP2 release. Biomaterials 2018; 161:216-227. [PMID: 29421557 PMCID: PMC5831261 DOI: 10.1016/j.biomaterials.2018.01.040] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 02/07/2023]
Abstract
Biomaterials-based approaches to harnessing the immune and inflammatory responses to potentiate wound healing hold important promise. Bone fracture healing is characterized by an acute inflammatory phase, followed by a transition to a regenerative and repair phase. In this study, we developed genipin-crosslinked gelatin microspheres designed to be preferentially degraded by inflammatory (M1) macrophages. Highly crosslinked (>90%) microspheres allowed efficient incorporation of bioactive bone morphogenetic protein 2 (BMP2), a potent stimulator of osteogenesis in progenitor cells, via electrostatic interactions. Release of BMP2 was directly correlated with degradation of the gelatin matrix. Exposure of microspheres to polarized murine macrophages showed that degradation was significantly higher in the presence of M1 macrophages, relative to alternatively activated (M2) macrophages and unpolarized controls. Microsphere degradation in the presence of non-inflammatory cells resulted in very low degradation rates. The expression of matrix metalloproteinases (MMPs) and tissue inhibitors of MMP (TIMPs) by macrophages were consistent with the observed phenotype-dependent degradation rates. Indirect co-culture of BMP2-loaded microspheres and macrophages with isolated adipose-derived mesenchymal stem cells (MSC) showed that M1 macrophages produced the strongest osteogenic response, comparable to direct supplementation of the culture medium with BMP2. Controlled release systems that are synchronized with the inflammatory response have the potential to provide better spatiotemporal control of growth factor delivery and therefore may improve the outcomes of recalcitrant wounds.
Collapse
Affiliation(s)
| | - Paul A Turner
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
| | | | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, USA
| | - Steven Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
43
|
Calciolari E, Ravanetti F, Strange A, Mardas N, Bozec L, Cacchioli A, Kostomitsopoulos N, Donos N. Degradation pattern of a porcine collagen membrane in an in vivo model of guided bone regeneration. J Periodontal Res 2018; 53:430-439. [PMID: 29446096 DOI: 10.1111/jre.12530] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE Although collagen membranes have been clinically applied for guided tissue/bone regeneration for more than 30 years, their in vivo degradation pattern has never been fully clarified. A better understanding of the different stages of in vivo degradation of collagen membranes is extremely important, considering that the biology of bone regeneration requires the presence of a stable and cell/tissue-occlusive barrier during the healing stages in order to ensure a predictable result. Therefore, the aim of this study was to investigate the degradation pattern of a porcine non-cross-linked collagen membrane in an in vivo model of guided bone regeneration (GBR). MATERIAL AND METHODS Decalcified and paraffin-embedded specimens from calvarial defects of 18, 10-month-old Wistar rats were used. The defects were treated with a double layer of collagen membrane and a deproteinized bovine bone mineral particulate graft. At 7, 14 and 30 days of healing, qualitative evaluation with scanning electron microscopy and atomic force microscopy, and histomorphometric measurements were performed. Markers of collagenase activity and bone formation were investigated using an immunofluorescence technique. RESULTS A significant reduction of membrane thickness was observed from 7 to 30 days of healing, which was associated with progressive loss of collagen alignment, increased collagen remodeling and progressive invasion of woven bone inside the membranes. A limited inflammatory infiltrate was observed at all time points of healing. CONCLUSION The collagen membrane investigated was biocompatible and able to promote bone regeneration. However, pronounced signs of degradation were observed starting from day 30. Since successful regeneration is obtained only when cell occlusion and space maintenance exist for the healing time needed by the bone progenitor cells to repopulate the defect, the suitability of collagen membranes in cases where long-lasting barriers are needed needs to be further reviewed.
Collapse
Affiliation(s)
- E Calciolari
- Centre for Oral Clinical Research, Institute of Dentistry, Queen Mary University of London (QMUL), Barts and The London School of Medicine and Dentistry, London, UK.,Centre for Oral Immunobiology and Regenerative Medicine, Queen Mary University of London (QMUL), Barts and The London School of Medicine and Dentistry, London, UK
| | - F Ravanetti
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - A Strange
- Department of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, London, UK
| | - N Mardas
- Centre for Oral Immunobiology and Regenerative Medicine, Queen Mary University of London (QMUL), Barts and The London School of Medicine and Dentistry, London, UK
| | - L Bozec
- Department of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, London, UK
| | - A Cacchioli
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - N Kostomitsopoulos
- Laboratory Animal Facilities, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - N Donos
- Centre for Oral Clinical Research, Institute of Dentistry, Queen Mary University of London (QMUL), Barts and The London School of Medicine and Dentistry, London, UK.,Centre for Oral Immunobiology and Regenerative Medicine, Queen Mary University of London (QMUL), Barts and The London School of Medicine and Dentistry, London, UK
| |
Collapse
|
44
|
Xing Y, Shepherd N, Lan J, Li W, Rane S, Gupta SK, Zhang S, Dong J, Yu Q. MMPs/TIMPs imbalances in the peripheral blood and cerebrospinal fluid are associated with the pathogenesis of HIV-1-associated neurocognitive disorders. Brain Behav Immun 2017; 65:161-172. [PMID: 28487203 PMCID: PMC5793222 DOI: 10.1016/j.bbi.2017.04.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/13/2017] [Accepted: 04/30/2017] [Indexed: 10/19/2022] Open
Abstract
HIV-1-associated neurocognitive disorders (HAND) continue to be a major concern in the infected population, despite the widespread use of combined antiretroviral therapy (cART). Growing evidence suggests that an imbalance between matrix metalloproteinases (MMPs) and endogenous tissue inhibitors of MMPs (TIMPs) contributes to the pathogenesis of HAND. In our present study, we examined protein levels and enzymatic activities of MMPs and TIMPs in both plasma and cerebrospinal fluid (CSF) samples from HIV-1 patients with or without HAND and HIV-1-negative controls. Imbalances between MMPs and TIMPs with distinct patterns were revealed in both the peripheral blood and CSF of HIV-1 patients, especially those with HAND. In the peripheral blood, the protein levels of MMP-2, MMP-9, TIMP-1, TIMP-2, and the enzymatic activities of MMP-2 and MMP-9 were increased in HIV-1 patients with or without HAND when compared with HIV-1-negative controls. The enzymatic activity of MMP-2, but not MMP-9, was further increased in plasma samples of HAND patients than that of HIV-1 patients without HAND. Notably, the ratio of MMP-2/TIMP-2 in plasma was significantly increased in HAND patients, not in patients without HAND. In the CSF, MMP-2 activity was increased, but the ratio of MMP-2/TIMP-2 was not altered. De novo induction and activation of MMP-9 in the CSF of HAND patients was particularly prominent. The imbalances between MMPs and TIMPs in the blood and CSF were related to the altered profiles of inflammatory cytokines/chemokines and monocyte activation in these individuals. In addition, plasma from HIV-1 patients directly induced integrity disruption of an in vitro blood-brain barrier (BBB) model, leading to increased BBB permeability and robust transmigration of monocytes/macrophages. These results indicate that imbalances between MMPs and TIMPs are involved in BBB disruption and are implicated in the pathogenesis of neurological disorders such as HAND in HIV-1 patients.
Collapse
Affiliation(s)
- Yanyan Xing
- Department of Pathophysiology, Key Laboratory of the State Administration of Traditional Chinese Medicine, Medical College of Jinan University, Guangzhou, Guangdong 510632, China; Indiana Center for AIDS Research and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Nicole Shepherd
- Indiana Center for AIDS Research and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jie Lan
- Indiana Center for AIDS Research and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Wei Li
- Indiana Center for AIDS Research and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Sushmita Rane
- Indiana Center for AIDS Research and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Samir K Gupta
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Shanxiang Zhang
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jun Dong
- Department of Pathophysiology, Key Laboratory of the State Administration of Traditional Chinese Medicine, Medical College of Jinan University, Guangzhou, Guangdong 510632, China; Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510632, China.
| | - Qigui Yu
- Indiana Center for AIDS Research and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| |
Collapse
|
45
|
Shah A, Alhusayen R, Amini-Nik S. The critical role of macrophages in the pathogenesis of hidradenitis suppurativa. Inflamm Res 2017; 66:931-945. [PMID: 28656364 DOI: 10.1007/s00011-017-1074-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 06/21/2017] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Hidradenitis suppurativa (HS) is a painful chronic inflammatory disease with a prevalence between 1 and 4% of general population. The pathogenesis of HS long eluded scientists, but growing evidence suggests that it is a consequence of inflammatory dysregulation. FINDINGS Recent studies suggest that dysregulated immune response to skin flora and overexpression of inflammatory cytokines leads to chronic skin inflammation seen in HS. Macrophages are the most numerous inflammatory cells found in HS infiltrates and release numerous pro-inflammatory cytokines such as IL-23, and IL-1β and TNF-α, exacerbating the inflammation and contributing to the pathogenesis of HS. Furthermore, in HS, there is dysregulated function of other immune players closely associated with macrophage function including: matrix metalloproteases (MMP) 2 and 9 overexpression, toll-like receptor upregulation, impaired Notch signalling, NLRP3 inflammasome upregulation, and dysregulated keratinocyte function. Lifestyle factors including obesity and smoking also contribute to macrophage dysfunction and correlate with HS incidence. CONCLUSIONS The overexpression of pro-inflammatory cytokines and subsequent efficacy of anti-cytokine biologic therapies highlights the importance of managing macrophage dysfunction. Future therapies should target key molecular drivers of macrophage dysfunction such as TLR2 and NLRP3 overexpression.
Collapse
Affiliation(s)
- Ahmed Shah
- Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Raed Alhusayen
- Faculty of Medicine, University of Toronto, Toronto, Canada.,Division of Dermatology, University of Toronto, Toronto, Canada.,Sunnybrook Health Science Center, Sunnybrook Research Institute, Toronto, Canada
| | - Saeid Amini-Nik
- Faculty of Medicine, University of Toronto, Toronto, Canada. .,Department of Surgery, University of Toronto, Toronto, Canada. .,Department of Laboratory Medicine and Pathobiology (LMP), University of Toronto, Toronto, Canada. .,Sunnybrook Health Science Center, Sunnybrook Research Institute, Toronto, Canada.
| |
Collapse
|
46
|
Gene expression patterns associated with neurological disease in human HIV infection. PLoS One 2017; 12:e0175316. [PMID: 28445538 PMCID: PMC5405951 DOI: 10.1371/journal.pone.0175316] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/23/2017] [Indexed: 12/01/2022] Open
Abstract
The pathogenesis and nosology of HIV-associated neurological disease (HAND) remain incompletely understood. Here, to provide new insight into the molecular events leading to neurocognitive impairments (NCI) in HIV infection, we analyzed pathway dysregulations in gene expression profiles of HIV-infected patients with or without NCI and HIV encephalitis (HIVE) and control subjects. The Gene Set Enrichment Analysis (GSEA) algorithm was used for pathway analyses in conjunction with the Molecular Signatures Database collection of canonical pathways (MSigDb). We analyzed pathway dysregulations in gene expression profiles of patients from the National NeuroAIDS Tissue Consortium (NNTC), which consists of samples from 3 different brain regions, including white matter, basal ganglia and frontal cortex of HIV-infected and control patients. While HIVE is characterized by widespread, uncontrolled inflammation and tissue damage, substantial gene expression evidence of induction of interferon (IFN), cytokines and tissue injury is apparent in all brain regions studied, even in the absence of NCI. Various degrees of white matter changes were present in all HIV-infected subjects and were the primary manifestation in patients with NCI in the absence of HIVE. In particular, NCI in patients without HIVE in the NNTC sample is associated with white matter expression of chemokines, cytokines and β-defensins, without significant activation of IFN. Altogether, the results identified distinct pathways differentially regulated over the course of neurological disease in HIV infection and provide a new perspective on the dynamics of pathogenic processes in the course of HIV neurological disease in humans. These results also demonstrate the power of the systems biology analyses and indicate that the establishment of larger human gene expression profile datasets will have the potential to provide novel mechanistic insight into the pathogenesis of neurological disease in HIV infection and identify better therapeutic targets for NCI.
Collapse
|
47
|
Ariyoshi W, Okinaga T, Chaweewannakorn W, Akifusa S, Nisihara T. Mechanisms involved in enhancement of matrix metalloproteinase-9 expression in macrophages by interleukin-33. J Cell Physiol 2017; 232:3481-3495. [PMID: 28105703 DOI: 10.1002/jcp.25809] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/17/2017] [Accepted: 01/18/2017] [Indexed: 01/13/2023]
Abstract
Endothelial transmigration of macrophages is accomplished by matrix metalloproteinase (MMP)-induced degradation of the basement membrane and extracellular matrix components. Macrophages upregulate MMP-9 expression and secretion upon immunological challenges and require its activity for migration during inflammatory responses. Interleukin (IL)-33 is a recently discovered pro-inflammatory cytokine that belongs to the IL-1 family. The aim of this study was to elucidate the mechanisms underlying IL-33-induced MMP-9 expression in the mouse monocyte/macrophage line RAW264.7. IL-33 increased MMP-9 mRNA and protein expression in RAW264.7 cells. Blockage of IL-33-IL-33 receptor (ST2L) binding suppressed IL-33-mediated induction of MMP-9. IL-33 induced phosphorylation and nuclear translocation of extracellular signal-regulated kinase 1/2 (ERK1/2) and nuclear factor-kappa B (NF-κB). Chromatin immunoprecipitation indicated that IL-33 increased c-fos recruitment to the MMP-9 promoter. Reporter assay findings also revealed that IL-33 stimulated the transcriptional activity of activator protein 1 (AP-1). Pre-treatment of the cells with a specific inhibitor of ERK1/2 and NF-κB attenuated the IL-33-induced activation of AP-1 subunits, transcriptional activity of AP-1, and expression of MMP-9. We also demonstrated that ERK-dependent activation of cAMP response element binding protein (CREB) is a key step for AP-1 activation by IL-33. These results indicate an essential role of ERK/CREB and NF-κB cascades in the induction of MMP-9 in monocytes/macrophages through AP-1 activation.
Collapse
Affiliation(s)
- Wataru Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Toshinori Okinaga
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Wichida Chaweewannakorn
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, Japan.,Division of Developmental Stomatognathic Function Science, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Sumio Akifusa
- Units of Education on Healthcare Team, School of Oral Health Science, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Tatsuji Nisihara
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
48
|
Accelerated biodegradation of silk sutures through matrix metalloproteinase activation by incorporating 4-hexylresorcinol. Sci Rep 2017; 7:42441. [PMID: 28205580 PMCID: PMC5304327 DOI: 10.1038/srep42441] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/09/2017] [Indexed: 12/31/2022] Open
Abstract
Silk suture material is primarily composed of silk fibroin and regarded as a non-resorbable material. It is slowly degraded by proteolysis when it is implanted into the body. 4-Hexylresorcinol (4HR) is a well-known antiseptic. In this study, the biodegradability of 4HR-incorporated silk sutures were compared to that of untreated silk sutures and polyglactin 910 sutures, a commercially available resorbable suture. 4HR-incorporated silk sutures exhibited anti-microbial properties. Matrix metalloproteinase (MMP) can digest a wide spectrum of proteins. 4HR increased MMP-2, -3, and -9 expression in RAW264.7 cells. MMP-2, -3, and -9 were able to digest not only silk fibroin but also silk sutures. Consequently, 59.5% of the 4HR-incorporated silk suture material remained at 11 weeks after grafting, which was similar to that of polyglactin 910 degradation (56.4% remained). The residual amount of bare silk suture material at 11 weeks after grafting was 91.5%. The expression levels of MMP-2, -3 and -9 were high in the 4HR-incorporated silk suture-implanted site 12 weeks after implantation. In conclusion, 4HR-treated silk sutures exhibited anti-microbial properties and a similar level of bio-degradation to polyglactin 910 sutures and induced higher expression of MMP-2, -3, and -9 in macrophages.
Collapse
|
49
|
Sagar V, Atluri VSR, Pilakka-Kanthikeel S, Nair M. Magnetic nanotherapeutics for dysregulated synaptic plasticity during neuroAIDS and drug abuse. Mol Brain 2016; 9:57. [PMID: 27216740 PMCID: PMC4878083 DOI: 10.1186/s13041-016-0236-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 05/06/2016] [Indexed: 01/02/2023] Open
Abstract
The human immunodeficiency virus (HIV) is a neurotropic virus. It induces neurotoxicity and subsequent brain pathologies in different brain cells. Addiction to recreational drugs remarkably affects the initiation of HIV infections and expedites the progression of acquired immunodeficiency syndrome (AIDS) associated neuropathogenesis. Symptoms of HIV-associated neurocognitive disorders (HAND) are noticed in many AIDS patients. At least 50 % of HIV diagnosed cases show one or other kind of neuropathological signs or symptoms during different stages of disease progression. In the same line, mild to severe neurological alterations are seen in at least 80 % autopsies of AIDS patients. Neurological illnesses weaken the connections between neurons causing significant altercations in synaptic plasticity. Synaptic plasticity alterations during HIV infection and recreational drug abuse are mediated by complex cellular phenomena involving changes in gene expression and subsequent loss of dendritic and spine morphology and physiology. New treatment strategies with ability to deliver drugs across blood-brain barrier (BBB) are being intensively investigated. In this context, magnetic nanoparticles (MNPs) based nanoformulations have shown significant potential for target specificity, drug delivery, drug release, and bioavailability of desired amount of drugs in non-invasive brain targeting. MNPs-based potential therapies to promote neuronal plasticity during HIV infection and recreational drug abuse are being developed.
Collapse
Affiliation(s)
- Vidya Sagar
- Department of Immunology, Center for Personalized Nanomedicine/Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Venkata Subba Rao Atluri
- Department of Immunology, Center for Personalized Nanomedicine/Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Sudheesh Pilakka-Kanthikeel
- Department of Immunology, Center for Personalized Nanomedicine/Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Madhavan Nair
- Department of Immunology, Center for Personalized Nanomedicine/Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA.
| |
Collapse
|
50
|
van Strien ME, de Vries HE, Chrobok NL, Bol JGJM, Breve JJP, van der Pol SMP, Kooij G, van Buul JD, Karpuj M, Steinman L, Wilhelmus MM, Sestito C, Drukarch B, Van Dam AM. Tissue Transglutaminase contributes to experimental multiple sclerosis pathogenesis and clinical outcome by promoting macrophage migration. Brain Behav Immun 2015; 50:141-154. [PMID: 26133787 DOI: 10.1016/j.bbi.2015.06.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 06/24/2015] [Accepted: 06/27/2015] [Indexed: 12/23/2022] Open
Abstract
Multiple sclerosis is a serious neurological disorder, resulting in e.g., sensory, motor and cognitive deficits. A critical pathological aspect of multiple sclerosis (MS) is the influx of immunomodulatory cells into the central nervous system (CNS). Identification of key players that regulate cellular trafficking into the CNS may lead to the development of more selective treatment to halt this process. The multifunctional enzyme tissue Transglutaminase (TG2) can participate in various inflammation-related processes, and is known to be expressed in the CNS. In the present study, we question whether TG2 activity contributes to the pathogenesis of experimental MS, and could be a novel therapeutic target. In human post-mortem material, we showed the appearance of TG2 immunoreactivity in leukocytes in MS lesions, and particular in macrophages in rat chronic-relapsing experimental autoimmune encephalomyelitis (cr-EAE), an experimental MS model. Clinical deficits as observed in mouse EAE were reduced in TG2 knock-out mice compared to littermate wild-type mice, supporting a role of TG2 in EAE pathogenesis. To establish if the enzyme TG2 represents an attractive therapeutic target, cr-EAE rats were treated with TG2 activity inhibitors during ongoing disease. Reduction of TG2 activity in cr-EAE animals dramatically attenuated clinical deficits and demyelination. The mechanism underlying these beneficial effects pointed toward a reduction in macrophage migration into the CNS due to attenuated cytoskeletal flexibility and RhoA GTPase activity. Moreover, iNOS and TNFα levels were selectively reduced in the CNS of cr-EAE rats treated with a TG2 activity inhibitor, whereas other relevant inflammatory mediators were not affected in CNS or spleen by reducing TG2 activity. We conclude that modulating TG2 activity opens new avenues for therapeutic intervention in MS which does not affect peripheral levels of inflammatory mediators.
Collapse
Affiliation(s)
- Miriam E van Strien
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy & Neurosciences, The Netherlands
| | - Helga E de Vries
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Molecular Cell Biology and Immunology, Amsterdam, The Netherlands
| | - Navina L Chrobok
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy & Neurosciences, The Netherlands
| | - John G J M Bol
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy & Neurosciences, The Netherlands
| | - John J P Breve
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy & Neurosciences, The Netherlands
| | - Susanne M P van der Pol
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Molecular Cell Biology and Immunology, Amsterdam, The Netherlands
| | - Gijs Kooij
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Molecular Cell Biology and Immunology, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Dept. Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Lawrence Steinman
- Beckman Center for Molecular Medicine, Stanford University, Stanford, USA
| | - Micha M Wilhelmus
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy & Neurosciences, The Netherlands
| | - Claudia Sestito
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy & Neurosciences, The Netherlands
| | - Benjamin Drukarch
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy & Neurosciences, The Netherlands
| | - Anne-Marie Van Dam
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy & Neurosciences, The Netherlands.
| |
Collapse
|