1
|
Ceccherini E, Michelucci E, Signore G, Coco B, Zari M, Bellini M, Brunetto MR, Cecchettini A, Rocchiccioli S. The Clinical Utility of the Saliva Proteome in Rare Diseases: A Pilot Study for Biomarker Discovery in Primary Sclerosing Cholangitis. J Clin Med 2024; 13:544. [PMID: 38256678 PMCID: PMC10816894 DOI: 10.3390/jcm13020544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/08/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Primary sclerosing cholangitis (PSC) is a rare chronic inflammatory liver disease characterized by biliary strictures and cholestasis. Due to the lack of effective serological indicators for diagnosis and prognosis, in the present study, we examined the potentiality of the saliva proteome to comprehensively screen for novel biomarkers. METHODS Saliva samples of PSC patients and healthy controls were processed and subsequently analyzed using a liquid chromatography-tandem mass spectrometry technique. A bioinformatic approach was applied to detect the differentially expressed proteins, their related biological functions and pathways, and the correlation with the clinical evidence in order to identify a possible marker for the PSC group. RESULTS We identified 25 differentially expressed proteins in PSC patients when compared to the healthy control group. Among them, eight proteins exhibited area under the curve values up to 0.800, suggesting these saliva proteins as good discriminators between the two groups. Multiple positive correlations were also identified between the dysregulated salivary proteins and increased serum alkaline phosphatase levels and the presence of ulcerative colitis. Pathway analysis revealed significant enrichments in the immune system, neutrophil degranulation, and in the interleukine-17 signaling pathway. CONCLUSION We demonstrated the potentiality of saliva as a useful biofluid to obtain a fingerprint of the pathology, suggesting disulfide-isomerase A3 and peroxiredoxin-5 as the better discriminating proteins in PSC patients. Hence, analysis of saliva proteins could become, in future, a useful tool in the screening of patients with suspected PSC.
Collapse
Affiliation(s)
- Elisa Ceccherini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.M.); (G.S.); (A.C.); (S.R.)
| | - Elena Michelucci
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.M.); (G.S.); (A.C.); (S.R.)
- Institute of Chemistry of Organometallic Compounds, National Research Council, 56124 Pisa, Italy
| | - Giovanni Signore
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.M.); (G.S.); (A.C.); (S.R.)
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
| | - Barbara Coco
- Hepatology Unit, Reference Centre of the Tuscany Region for Chronic Liver Disease and Cancer, University Hospital of Pisa, 56124 Pisa, Italy; (B.C.); (M.R.B.)
| | - Michela Zari
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.Z.); (M.B.)
| | - Massimo Bellini
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.Z.); (M.B.)
| | - Maurizia Rossana Brunetto
- Hepatology Unit, Reference Centre of the Tuscany Region for Chronic Liver Disease and Cancer, University Hospital of Pisa, 56124 Pisa, Italy; (B.C.); (M.R.B.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Antonella Cecchettini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.M.); (G.S.); (A.C.); (S.R.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Silvia Rocchiccioli
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (E.M.); (G.S.); (A.C.); (S.R.)
| |
Collapse
|
2
|
Chen S, Wang K, Wang Q. Mannose: A Promising Player in Clinical and Biomedical Applications. Curr Drug Deliv 2024; 21:1435-1444. [PMID: 38310442 DOI: 10.2174/0115672018275954231220101637] [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: 09/20/2023] [Revised: 12/02/2023] [Accepted: 12/08/2023] [Indexed: 02/05/2024]
Abstract
Mannose, an isomer of glucose, exhibits a distinct molecular structure with the same formula but a different atom arrangement, contributing to its specific biological functions. Widely distributed in body fluids and tissues, particularly in the nervous system, skin, testes, and retinas, mannose plays a crucial role as a direct precursor for glycoprotein synthesis. Glycoproteins, essential for immune regulation and glycosylation processes, underscore the significance of mannose in these physiological activities. The clinical and biomedical applications of mannose are diverse, encompassing its anti-inflammatory properties, potential to inhibit bacterial infections, role in metabolism regulation, and suggested involvement in alleviating diabetes and obesity. Additionally, mannose shows promise in antitumor effects, immune modulation, and the construction of drug carriers, indicating a broad spectrum of therapeutic potential. The article aims to present a comprehensive review of mannose, focusing on its molecular structure, metabolic pathways, and clinical and biomedical applications, and also to emphasize its status as a promising therapeutic agent.
Collapse
Affiliation(s)
- Sijing Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- The Department of Gynecologic Oncology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kana Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- The Department of Gynecologic Oncology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiao Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- The Department of Gynecologic Oncology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Albtoush N, Queisser KA, Zawerton A, Lauer ME, Beswick EJ, Petrey AC. TSG6 hyaluronan matrix remodeling dampens the inflammatory response during colitis. Matrix Biol 2023; 121:149-166. [PMID: 37391162 PMCID: PMC10530565 DOI: 10.1016/j.matbio.2023.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
In response to tissue injury, changes in the extracellular matrix (ECM) can directly affect the inflammatory response and contribute to disease progression or resolution. During inflammation, the glycosaminoglycan hyaluronan (HA) becomes modified by tumor necrosis factor stimulated gene-6 (TSG6). TSG6 covalently transfers heavy chain (HC) proteins from inter-α-trypsin inhibitor (IαI) to HA in a transesterification reaction and is to date is the only known HC-transferase. By modifying the HA matrix, TSG6 generates HC:HA complexes that are implicated in mediating both protective and pathological responses. Inflammatory bowel disease (IBD) is a lifelong chronic disorder with well-described remodeling of the ECM and increased mononuclear leukocyte influx into the intestinal mucosa. Deposition of HC:HA matrices is an early event in inflamed gut tissue that precedes and promotes leukocyte infiltration. However, the mechanisms by which TSG6 contributes to intestinal inflammation are not well understood. The aim of our study was to understand how the TSG6 and its enzymatic activity contributes to the inflammatory response in colitis. Our findings indicate that inflamed tissues of IBD patients show an elevated level of TSG6 and increased HC deposition and that levels of HA strongly associate with TSG6 levels in patient colon tissue specimens. Additionally, we observed that mice lacking TSG6 are more vulnerable to acute colitis and exhibit an aggravated macrophage-associated mucosal immune response characterized by elevated pro-inflammatory cytokines and chemokines and diminished anti-inflammatory mediators including IL-10. Surprisingly, along with significantly increased levels of inflammation in the absence of TSG6, tissue HA levels in mice were found to be significantly reduced and disorganized, absent of typical "HA-cable" structures. Inhibition of TSG6 HC-transferase activity leads to a loss of cell surface HA and leukocyte adhesion, indicating that the enzymatic functions of TSG6 are a major contributor to stability of the HA ECM during inflammation. Finally, using biochemically generated HC:HA matrices derived by TSG6, we show that HC:HA complexes can attenuate the inflammatory response of activated monocytes. In conclusion, our data suggests that TSG6 exerts a tissue-protective, anti-inflammatory effect via the generation of HC:HA complexes that become dysregulated in IBD.
Collapse
Affiliation(s)
- Nansy Albtoush
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112; Lerner Research Institute, Department of Inflammation & Immunity, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kimberly A Queisser
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112; Lerner Research Institute, Department of Inflammation & Immunity, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ash Zawerton
- Lerner Research Institute, Department of Inflammation & Immunity, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mark E Lauer
- Lerner Research Institute, Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ellen J Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Kentucky, Lexington, KY, United States
| | - Aaron C Petrey
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112; Department of Pathology, Division of Microbiology & Immunology, University of Utah School of Medicine, Salt Lake City, Utah, 84132, USA; Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA; Lerner Research Institute, Department of Inflammation & Immunity, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
4
|
Wang AJ, Ren J, Wang A, Hascall VC. Monocyte adhesive hyaluronan matrix induced by hyperglycemia in diabetic lung injuries. J Biol Chem 2023; 299:104995. [PMID: 37394007 PMCID: PMC10413281 DOI: 10.1016/j.jbc.2023.104995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023] Open
Abstract
Infiltrated pre-inflammatory monocytes and macrophages have important roles in the induction of diabetic lung injuries, but the mechanism mediating their infiltration is still unclear. Here, we showed that airway smooth muscle cells (SMCs) activated monocyte adhesion in response to hyperglycemic glucose (25.6 mM) by significantly increasing hyaluronan (HA) in the cell matrix, with concurrent 2- to 4-fold increases in adhesion of U937 monocytic-leukemic cells. The HA-based structures were attributed directly to the high-glucose and not to increased extracellular osmolality, and they required growth stimulation of SMCs by serum. Treatment of SMCs with heparin in high-glucose induces synthesis of a much larger HA matrix, consistent with our observations in the glomerular SMCs. Further, we observed increases in tumor necrosis factor-stimulated gene-6 (TSG-6) expression in high-glucose and high-glucose plus heparin cultures, and the heavy chain (HC)-modified HA structures existed on the monocyte-adhesive cable structures in high-glucose and in high-glucose plus heparin-treated SMC cultures. Interestingly, these HC-modified HA structures were unevenly distributed along the HA cables. Further, the in vitro assay with recombinant human TSG-6 and the HA14 oligo showed that heparin has no inhibitory activity on the TSG-6-induced HC-transfer to HA, consistent with the results from SMC cultures. These results support the hypothesis that hyperglycemia in airway smooth muscle induces the synthesis of a HA matrix that recruits inflammatory cells and establishes a chronic inflammatory process and fibrosis that lead to diabetic lung injuries.
Collapse
Affiliation(s)
- Andrew Jun Wang
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Juan Ren
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Aimin Wang
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Vincent C Hascall
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
5
|
Sun J, Peterson EA, Chen X, Wang J. hapln1a + cells guide coronary growth during heart morphogenesis and regeneration. Nat Commun 2023; 14:3505. [PMID: 37311876 PMCID: PMC10264374 DOI: 10.1038/s41467-023-39323-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/07/2023] [Indexed: 06/15/2023] Open
Abstract
Although several tissues and chemokines orchestrate coronary formation, the guidance cues for coronary growth remain unclear. Here, we profile the juvenile zebrafish epicardium during coronary vascularization and identify hapln1a+ cells enriched with vascular-regulating genes. hapln1a+ cells not only envelop vessels but also form linear structures ahead of coronary sprouts. Live-imaging demonstrates that coronary growth occurs along these pre-formed structures, with depletion of hapln1a+ cells blocking this growth. hapln1a+ cells also pre-lead coronary sprouts during regeneration and hapln1a+ cell loss inhibits revascularization. Further, we identify serpine1 expression in hapln1a+ cells adjacent to coronary sprouts, and serpine1 inhibition blocks vascularization and revascularization. Moreover, we observe the hapln1a substrate, hyaluronan, forming linear structures along and preceding coronary vessels. Depletion of hapln1a+ cells or serpine1 activity inhibition disrupts hyaluronan structure. Our studies reveal that hapln1a+ cells and serpine1 are required for coronary production by establishing a microenvironment to facilitate guided coronary growth.
Collapse
Affiliation(s)
- Jisheng Sun
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Elizabeth A Peterson
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Xin Chen
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Jinhu Wang
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
6
|
Wang AJ, Ren J, Wang A, Hascall VC. Heparin and calreticulin interact on the surface of early G0/G1 dividing rat mesangial cells to regulate hyperglycemic glucose-induced responses. J Biol Chem 2023; 299:103074. [PMID: 36858200 PMCID: PMC10060746 DOI: 10.1016/j.jbc.2023.103074] [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: 01/11/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/02/2023] Open
Abstract
Heparin can block pathological responses associated with diabetic nephropathy in animal models and human patients. Our previous studies showed that the interaction of heparin on the surface of rat mesangial cells (RMCs) entering G1 of cell division in hyperglycemic glucose: 1) blocked glucose uptake by glucose transporter 4; 2) inhibited cytosolic uridine diphosphate-glucose elevation that would occur within 6 h from G0/G1; and 3) prevented subsequent activation of hyaluronan synthesis in intracellular compartments and subsequent inflammatory responses. However, specific proteins that interact with heparin are unresolved. Here, we showed by live cell imaging that fluorescent heparin was rapidly internalized into the cytoplasm and then into the endoplasmic reticulum, Golgi, and nuclei compartments. Biotinylated-heparin was applied onto the surface of growth arrested G0/G1 RMCs in order to extract heparin-binding protein(s). SDS-PAGE gels showed two bands at ∼70 kDa in the extract that were absent when unlabeled heparin was used to compete. Trypsin digests of the bands were analyzed by MS and identified as calreticulin and prelamin A/C. Immunostaining with their antibodies identified the presence of calreticulin on the G0/G1 RMC cell surface. Previous studies have shown that calreticulin can be on the cell surface and can interact with the LDL receptor-related protein, which has been implicated in glucose transport by interaction with glucose transporter 4. Thus, cell surface calreticulin can act as a heparin receptor through a mechanism involving LRP1, which prevents the intracellular responses in high glucose and reprograms the cells to synthesize an extracellular hyaluronan matrix after division.
Collapse
Affiliation(s)
- Andrew Jun Wang
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Juan Ren
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Aimin Wang
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Vincent C Hascall
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
7
|
Bosi A, Banfi D, Bistoletti M, Catizzone LM, Chiaravalli AM, Moretto P, Moro E, Karousou E, Viola M, Giron MC, Crema F, Rossetti C, Binelli G, Passi A, Vigetti D, Giaroni C, Baj A. Hyaluronan Regulates Neuronal and Immune Function in the Rat Small Intestine and Colonic Microbiota after Ischemic/Reperfusion Injury. Cells 2022; 11:3370. [PMID: 36359764 PMCID: PMC9657036 DOI: 10.3390/cells11213370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/12/2022] [Accepted: 10/21/2022] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Intestinal ischemia and reperfusion (IRI) injury induces acute and long-lasting damage to the neuromuscular compartment and dysmotility. This study aims to evaluate the pathogenetic role of hyaluronan (HA), a glycosaminoglycan component of the extracellular matrix, as a modulator of the enteric neuronal and immune function and of the colonic microbiota during in vivo IRI in the rat small intestine. METHODS mesenteric ischemia was induced in anesthetized adult male rats for 60 min, followed by 24 h reperfusion. Injured, sham-operated and non-injured animals were treated with the HA synthesis inhibitor, 4-methylumbelliferone (4-MU 25 mg/kg). Fecal microbiota composition was evaluated by Next Generation Sequencing. Neutrophil infiltration, HA homeostasis and toll like receptor (TLR2 and TLR4) expression in the small intestine were evaluated by immunohistochemical and biomolecular approaches (qRT-PCR and Western blotting). Neuromuscular responses were studied in vitro, in the absence and presence of the selective TLR2/4 inhibitor, Sparstolonin B (SsnB 10, 30 µM). RESULTS 4-MU significantly reduced IRI-induced enhancement of potentially harmful Escherichia and Enterococcus bacteria. After IRI, HA levels, neutrophil infiltration, and TLR2 and TLR4 expression were significantly enhanced in the muscularis propria, and were significantly reduced to baseline levels by 4-MU. In the injured, but not in the non-injured and sham-operated groups, SsnB reduced both electrical field-stimulated (EFS, 0.1-40 Hz) contractions and EFS-induced (10 Hz) non-cholinergic non-adrenergic relaxations. CONCLUSIONS enhanced HA levels after intestinal IRI favors harmful bacteria overgrowth, increases neutrophil infiltration and promotes the upregulation of bacterial target receptors, TLR2 and TLR4, in the muscularis propria, inducing a pro-inflammatory state. TLR2 and TLR4 activation may, however, underlay a provisional benefit on excitatory and inhibitory neuronal pathways underlying peristalsis.
Collapse
Affiliation(s)
- Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Davide Banfi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | | | | | - Paola Moretto
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Elisabetta Moro
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Carlo Rossetti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Giorgio Binelli
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
8
|
Parnigoni A, Viola M, Karousou E, Rovera S, Giaroni C, Passi A, Vigetti D. ROLE OF HYALURONAN IN PATHOPHYSIOLOGY OF VASCULAR1 ENDOTHELIAL AND SMOOTH MUSCLE CELLS. Am J Physiol Cell Physiol 2022; 323:C505-C519. [PMID: 35759431 DOI: 10.1152/ajpcell.00061.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the main components of the extracellular matrix (ECM) of the blood vessel is hyaluronic acid or hyaluronan (HA). It is a ubiquitous polysaccharide belonging to the family of glycosaminoglycans, but, differently from other proteoglycan-associated glycosaminoglycans, it is synthesized on the plasma membrane by a family of three HA synthases (HAS). HA can be released as a free polymer in the extracellular space or remain associated with the membrane in the pericellular space via HAS or via binding proteins. In fact, several cell surface proteins can interact with HA working as HA receptors like CD44, RHAMM, and LYVE-1. In physiological conditions, HA is localized in the glycocalyx and in the adventitia and is responsible for the loose and hydrated vascular structure favoring flexibility and allowing the stretching of vessels in response to mechanical forces. During atherogenesis, ECM undergoes dramatic alterations which have a crucial role in lipoprotein retention and in triggering multiple signaling cascades that wake up cells from their quiescent status. HA becomes highly present in the media and neointima favoring smooth muscle cells dedifferentiation, migration, and proliferation that strongly contribute to vessel wall thickening. Further, HA is able to modulate immune cell recruitment both within the vessel wall and on the endothelial cell layer. This review is focused on the effects of HA on vascular cell behavior.
Collapse
Affiliation(s)
- Arianna Parnigoni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Simona Rovera
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
9
|
Nan F, Sun Y, Liang H, Zhou J, Ma X, Zhang D. Mannose: A Sweet Option in the Treatment of Cancer and Inflammation. Front Pharmacol 2022; 13:877543. [PMID: 35645798 PMCID: PMC9136145 DOI: 10.3389/fphar.2022.877543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
As a natural sugar, mannose is a type of hexose that is abundant in many different types of fruits. Since mannose is rarely used for glycolysis in mammals, studies on the role of mannose have not attracted much attention. Glycosylation of specific proteins was thought to be the major function of mannose. Surprisingly, during the past few years, mannose was found to be effective in promoting immune tolerance and suppressing inflammatory diseases related to autoimmunity and allergy. Moreover importantly, mannose was also found to be efficient in suppressing tumors by suppressing glycolysis and enhancing chemotherapeutic agents. In this review, we summarize the recent studies of mannose on antitumor properties and anti-inflammatory characteristics. We emphasize that mannose could play a beneficial role in the treatment of a variety of diseases, including cancers and inflammatory diseases, and could be a novel therapeutic strategy that deserves continued evaluation.
Collapse
|
10
|
Bosi A, Banfi D, Bistoletti M, Moretto P, Moro E, Crema F, Maggi F, Karousou E, Viola M, Passi A, Vigetti D, Giaroni C, Baj A. Hyaluronan: A Neuroimmune Modulator in the Microbiota-Gut Axis. Cells 2021; 11:cells11010126. [PMID: 35011688 PMCID: PMC8750446 DOI: 10.3390/cells11010126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 12/22/2022] Open
Abstract
The commensal microbiota plays a fundamental role in maintaining host gut homeostasis by controlling several metabolic, neuronal and immune functions. Conversely, changes in the gut microenvironment may alter the saprophytic microbial community and function, hampering the positive relationship with the host. In this bidirectional interplay between the gut microbiota and the host, hyaluronan (HA), an unbranched glycosaminoglycan component of the extracellular matrix, has a multifaceted role. HA is fundamental for bacterial metabolism and influences bacterial adhesiveness to the mucosal layer and diffusion across the epithelial barrier. In the host, HA may be produced and distributed in different cellular components within the gut microenvironment, playing a role in the modulation of immune and neuronal responses. This review covers the more recent studies highlighting the relevance of HA as a putative modulator of the communication between luminal bacteria and the host gut neuro-immune axis both in health and disease conditions, such as inflammatory bowel disease and ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Davide Banfi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Paola Moretto
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Elisabetta Moro
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (E.M.); (F.C.)
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (E.M.); (F.C.)
| | - Fabrizio Maggi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
- Centre of Neuroscience, University of Insubria, 21100 Varese, Italy
- Correspondence: ; Tel.: +39-0332-217412; Fax: +39-0332-217111
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (M.B.); (P.M.); (F.M.); (E.K.); (M.V.); (A.P.); (D.V.); (A.B.)
| |
Collapse
|
11
|
Queisser KA, Mellema RA, Middleton EA, Portier I, Manne BK, Denorme F, Beswick EJ, Rondina MT, Campbell RA, Petrey AC. COVID-19 generates hyaluronan fragments that directly induce endothelial barrier dysfunction. JCI Insight 2021; 6:147472. [PMID: 34314391 PMCID: PMC8492325 DOI: 10.1172/jci.insight.147472] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023] Open
Abstract
Vascular injury has emerged as a complication contributing to morbidity in coronavirus disease 2019 (COVID-19). The glycosaminoglycan hyaluronan (HA) is a major component of the glycocalyx, a protective layer of glycoconjugates that lines the vascular lumen and regulates key endothelial cell functions. During critical illness, as in the case of sepsis, enzymes degrade the glycocalyx, releasing fragments with pathologic activities into circulation and thereby exacerbating disease. Here, we analyzed levels of circulating glycosaminoglycans in 46 patients with COVID-19 ranging from moderate to severe clinical severity and measured activities of corresponding degradative enzymes. This report provides evidence that the glycocalyx becomes significantly damaged in patients with COVID-19 and corresponds with severity of disease. Circulating HA fragments and hyaluronidase, 2 signatures of glycocalyx injury, strongly associate with sequential organ failure assessment scores and with increased inflammatory cytokine levels in patients with COVID-19. Pulmonary microvascular endothelial cells exposed to COVID-19 milieu show dysregulated HA biosynthesis and degradation, leading to production of pathological HA fragments that are released into circulation. Finally, we show that HA fragments present at high levels in COVID-19 patient plasma can directly induce endothelial barrier dysfunction in a ROCK- and CD44-dependent manner, indicating a role for HA in the vascular pathology of COVID-19.
Collapse
Affiliation(s)
| | | | - Elizabeth A. Middleton
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Division of General Internal Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Irina Portier
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Bhanu Kanth Manne
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Ellen J. Beswick
- Department of Pathology and
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Matthew T. Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Department of Pathology and
- Division of General Internal Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Geriatric Research, Education, and Clinical Center and
- Department of Internal Medicine, George E. Wahlen Salt Lake City Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Robert A. Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Aaron C. Petrey
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Department of Pathology and
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
12
|
Wang W, Zhang H, Hou C, Liu Q, Yang S, Zhang Z, Yang W, Yang X. Internal modulation of proteolysis in vascular extracellular matrix remodeling: role of ADAM metallopeptidase with thrombospondin type 1 motif 5 in the development of intracranial aneurysm rupture. Aging (Albany NY) 2021; 13:12800-12816. [PMID: 33934089 PMCID: PMC8148490 DOI: 10.18632/aging.202948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/16/2021] [Indexed: 12/17/2022]
Abstract
Intracranial aneurysms (IAs) are common cerebrovascular diseases that carry a high mortality rate, and the mechanisms that contribute to IA formation and rupture have not been elucidated. ADAMTS-5 (ADAM Metallopeptidase with Thrombospondin Type 1 Motif 5) is a secreted proteinase involved in matrix degradation and ECM (extracellular matrix) remodeling processes, and we hypothesized that the dysregulation of ADAMTS-5 could play a role in the pathophysiology of IA. Immunofluorescence revealed that the ADAMTS-5 levels were decreased in human and murine IA samples. The administration of recombinant protein ADAMTS-5 significantly reduced the incidence of aneurysm rupture in the experimental model of IA. IA artery tissue was collected and utilized for histology, immunostaining, and specific gene expression analysis. Additionally, the IA arteries in ADAMTS-5-administered mice showed reduced elastic fiber destruction, proteoglycan accumulation, macrophage infiltration, inflammatory response, and apoptosis. To further verify the role of ADAMTS-5 in cerebral vessels, a specific ADAMTS-5 inhibitor was used on another model animal, zebrafish, and intracranial hemorrhage was observed in zebrafish embryos. In conclusion, our findings indicate that ADAMTS-5 is downregulated in human IA, and compensatory ADAMTS-5 administration inhibits IA development and rupture with potentially important implications for treating this cerebrovascular disease.
Collapse
Affiliation(s)
- Weihan Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, China
| | - Changkai Hou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, China
| | - Quanlei Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuyuan Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhen Zhang
- Department of Neuro-Oncology and Neurosurgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Weidong Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinyu Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
13
|
Han CY, Kang I, Harten IA, Gebe JA, Chan CK, Omer M, Alonge KM, den Hartigh LJ, Gomes Kjerulf D, Goodspeed L, Subramanian S, Wang S, Kim F, Birk DE, Wight TN, Chait A. Adipocyte-Derived Versican and Macrophage-Derived Biglycan Control Adipose Tissue Inflammation in Obesity. Cell Rep 2021; 31:107818. [PMID: 32610121 DOI: 10.1016/j.celrep.2020.107818] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/20/2020] [Accepted: 06/04/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity is characterized by adipose tissue inflammation. Because proteoglycans regulate inflammation, here we investigate their role in adipose tissue inflammation in obesity. We find that adipose tissue versican and biglycan increase in obesity. Versican is produced mainly by adipocytes and biglycan by adipose tissue macrophages. Both proteoglycans are also present in adipose tissue from obese human subjects undergoing gastric bypass surgery. Deletion of adipocyte-specific versican or macrophage-specific biglycan in mice reduces macrophage accumulation and chemokine and cytokine expression, although only adipocyte-specific versican deletion leads to sustained improvement in glucose tolerance. Macrophage-derived biglycan activates inflammatory genes in adipocytes. Versican expression increases in cultured adipocytes exposed to excess glucose, and adipocyte-conditioned medium stimulates inflammation in resident peritoneal macrophages, in part because of a versican breakdown product, versikine. These findings provide insights into the role of adipocyte- and macrophage-derived proteoglycans in adipose tissue inflammation in obesity.
Collapse
Affiliation(s)
- Chang Yeop Han
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Ingrid A Harten
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - John A Gebe
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Christina K Chan
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Mohamed Omer
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Kimberly M Alonge
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Laura J den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Diego Gomes Kjerulf
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Leela Goodspeed
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Savitha Subramanian
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Shari Wang
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Francis Kim
- Division of Cardiology, University of Washington, Seattle, WA, USA
| | - David E Birk
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, USA
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Alan Chait
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA.
| |
Collapse
|
14
|
Johnson LA, Banerji S, Lagerholm BC, Jackson DG. Dendritic cell entry to lymphatic capillaries is orchestrated by CD44 and the hyaluronan glycocalyx. Life Sci Alliance 2021; 4:4/5/e202000908. [PMID: 33687996 PMCID: PMC8008951 DOI: 10.26508/lsa.202000908] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
CD44 anchors the hyaluronan glycocalyx on migrating dendritic cells to permit docking to the endothelial receptor LYVE-1, thus orchestrating lymphatic trafficking through modulating glycocalyx density. DCs play a vital role in immunity by conveying antigens from peripheral tissues to draining lymph nodes, through afferent lymphatic vessels. Critical to the process is initial docking to the lymphatic endothelial receptor LYVE-1 via its ligand hyaluronan on the DC surface. How this relatively weak binding polymer is configured for specific adhesion to LYVE-1, however, is unknown. Here, we show that hyaluronan is anchored and spatially organized into a 400–500 nm dense glycocalyx by the leukocyte receptor CD44. Using gene knockout and by modulating CD44-hyaluronan interactions with monoclonal antibodies in vitro and in a mouse model of oxazolone-induced skin inflammation, we demonstrate that CD44 is required for DC adhesion and transmigration across lymphatic endothelium. In addition, we present evidence that CD44 can dynamically control the density of the hyaluronan glycocalyx, regulating the efficiency of DC trafficking to lymph nodes. Our findings define a previously unrecognized role for CD44 in lymphatic trafficking and highlight the importance of the CD44:HA:LYVE-1 axis in its regulation.
Collapse
Affiliation(s)
- Louise A Johnson
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Suneale Banerji
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - B Christoffer Lagerholm
- Wolfson Imaging Centre Oxford, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - David G Jackson
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Kellar GG, Barrow KA, Rich LM, Debley JS, Wight TN, Ziegler SF, Reeves SR. Loss of versican and production of hyaluronan in lung epithelial cells are associated with airway inflammation during RSV infection. J Biol Chem 2021; 296:100076. [PMID: 33187989 PMCID: PMC7949086 DOI: 10.1074/jbc.ra120.016196] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 12/21/2022] Open
Abstract
Airway inflammation is a critical feature of lower respiratory tract infections caused by viruses such as respiratory syncytial virus (RSV). A growing body of literature has demonstrated the importance of extracellular matrix changes such as the accumulation of hyaluronan (HA) and versican in the subepithelial space in promoting airway inflammation; however, whether these factors contribute to airway inflammation during RSV infection remains unknown. To test the hypothesis that RSV infection promotes inflammation via altered HA and versican production, we studied an ex vivo human bronchial epithelial cell (BEC)/human lung fibroblast (HLF) coculture model. RSV infection of BEC/HLF cocultures led to decreased hyaluronidase expression by HLFs, increased accumulation of HA, and enhanced adhesion of U937 cells as would be expected with increased HA. HLF production of versican was not altered following RSV infection; however, BEC production of versican was significantly downregulated following RSV infection. In vivo studies with epithelial-specific versican-deficient mice [SPC-Cre(+) Vcan-/-] demonstrated that RSV infection led to increased HA accumulation compared with control mice, which also coincided with decreased hyaluronidase expression in the lung. SPC-Cre(+) Vcan-/- mice demonstrated enhanced recruitment of monocytes and neutrophils in bronchoalveolar lavage fluid and increased neutrophils in the lung compared with SPC-Cre(-) RSV-infected littermates. Taken together, these data demonstrate that altered extracellular matrix accumulation of HA occurs following RSV infection and may contribute to airway inflammation. In addition, loss of epithelial expression of versican promotes airway inflammation during RSV infection further demonstrating that versican's role in inflammatory regulation is complex and dependent on the microenvironment.
Collapse
Affiliation(s)
- Gerald G Kellar
- Department of Defense, United States Army, Washington, USA; Benaroya Research Institute, Seattle, Washington, USA; Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Kaitlyn A Barrow
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Lucille M Rich
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Jason S Debley
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA; Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | | | - Steven F Ziegler
- Benaroya Research Institute, Seattle, Washington, USA; Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Stephen R Reeves
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA; Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
16
|
Wei Z, Huang L, Cui L, Zhu X. Mannose: Good player and assister in pharmacotherapy. Biomed Pharmacother 2020; 129:110420. [DOI: 10.1016/j.biopha.2020.110420] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/07/2020] [Accepted: 06/13/2020] [Indexed: 10/24/2022] Open
|
17
|
Bistoletti M, Bosi A, Caon I, Chiaravalli AM, Moretto P, Genoni A, Moro E, Karousou E, Viola M, Crema F, Baj A, Passi A, Vigetti D, Giaroni C. Involvement of hyaluronan in the adaptive changes of the rat small intestine neuromuscular function after ischemia/reperfusion injury. Sci Rep 2020; 10:11521. [PMID: 32661417 PMCID: PMC7359366 DOI: 10.1038/s41598-020-67876-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal ischemia/reperfusion (I/R) injury has severe consequences on myenteric neurons, which can be irreversibly compromised resulting in slowing of transit and hindered food digestion. Myenteric neurons synthesize hyaluronan (HA) to form a well-structured perineuronal net, which undergoes derangement when myenteric ganglia homeostasis is perturbed, i.e. during inflammation. In this study we evaluated HA involvement in rat small intestine myenteric plexus after in vivo I/R injury induced by clamping a branch of the superior mesenteric artery for 60 min, followed by 24 h of reperfusion. In some experiments, 4-methylumbelliferone (4-MU, 25 mg/kg), a HA synthesis inhibitor, was intraperitoneally administered to normal (CTR), sham-operated (SH) and I/R animals for 24 h. In longitudinal muscle myenteric plexus (LMMP) whole-mount preparations, HA binding protein staining as well as HA levels were significantly higher in the I/R group, and were reduced after 4-MU treatment. HA synthase 1 and 2 (HAS1 and HAS2) labelled myenteric neurons and mRNA levels in LMMPs increased in the I/R group with respect to CTR, and were reduced by 4-MU. The efficiency of the gastrointestinal transit was significantly reduced in I/R and 4-MU-treated I/R groups with respect to CTR and SH groups. In the 4-MU-treated I/R group gastric emptying was reduced with respect to the CTR, SH and I/R groups. Carbachol (CCh) and electrical field (EFS, 0.1–40 Hz) stimulated contractions and EFS-induced (10 Hz) NANC relaxations were reduced in the I/R group with respect to both CTR and SH groups. After I/R, 4-MU treatment increased EFS contractions towards control values, but did not affect CCh-induced contractions. NANC on-relaxations after I/R were not influenced by 4-MU treatment. Main alterations in the neurochemical coding of both excitatory (tachykinergic) and inhibitory pathways (iNOS, VIPergic) were also observed after I/R, and were influenced by 4-MU administration. Overall, our data suggest that, after an intestinal I/R damage, changes of HA homeostasis in specific myenteric neuron populations may influence the efficiency of the gastrointestinal transit. We cannot exclude that modulation of HA synthesis in these conditions may ameliorate derangement of the enteric motor function preventing, at least in part, the development of dysmotility.
Collapse
Affiliation(s)
- Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Ilaria Caon
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Anna Maria Chiaravalli
- Department of Pathology, ASST-Sette Laghi, Ospedale di Circolo Viale L. Borri 57, 21100, Varese, Italy
| | - Paola Moretto
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Angelo Genoni
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Elisabetta Moro
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy.
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy.
| |
Collapse
|
18
|
Intracellular hyaluronan: Importance for cellular functions. Semin Cancer Biol 2020; 62:20-30. [DOI: 10.1016/j.semcancer.2019.07.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/25/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023]
|
19
|
Wight TN, Kang I, Evanko SP, Harten IA, Chang MY, Pearce OMT, Allen CE, Frevert CW. Versican-A Critical Extracellular Matrix Regulator of Immunity and Inflammation. Front Immunol 2020; 11:512. [PMID: 32265939 PMCID: PMC7105702 DOI: 10.3389/fimmu.2020.00512] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix (ECM) proteoglycan, versican increases along with other ECM versican binding molecules such as hyaluronan, tumor necrosis factor stimulated gene-6 (TSG-6), and inter alpha trypsin inhibitor (IαI) during inflammation in a number of different diseases such as cardiovascular and lung disease, autoimmune diseases, and several different cancers. These interactions form stable scaffolds which can act as "landing strips" for inflammatory cells as they invade tissue from the circulation. The increase in versican is often coincident with the invasion of leukocytes early in the inflammatory process. Versican interacts with inflammatory cells either indirectly via hyaluronan or directly via receptors such as CD44, P-selectin glycoprotein ligand-1 (PSGL-1), and toll-like receptors (TLRs) present on the surface of immune and non-immune cells. These interactions activate signaling pathways that promote the synthesis and secretion of inflammatory cytokines such as TNFα, IL-6, and NFκB. Versican also influences inflammation by interacting with a variety of growth factors and cytokines involved in regulating inflammation thereby influencing their bioavailability and bioactivity. Versican is produced by multiple cell types involved in the inflammatory process. Conditional total knockout of versican in a mouse model of lung inflammation demonstrated significant reduction in leukocyte invasion into the lung and reduced inflammatory cytokine expression. While versican produced by stromal cells tends to be pro-inflammatory, versican expressed by myeloid cells can create anti-inflammatory and immunosuppressive microenvironments. Inflammation in the tumor microenvironment often contains elevated levels of versican. Perturbing the accumulation of versican in tumors can inhibit inflammation and tumor progression in some cancers. Thus versican, as a component of the ECM impacts immunity and inflammation through regulating immune cell trafficking and activation. Versican is emerging as a potential target in the control of inflammation in a number of different diseases.
Collapse
Affiliation(s)
- Thomas N. Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Stephen P. Evanko
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Ingrid A. Harten
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Mary Y. Chang
- Division of Pulmonary/Critical Care Medicine, Center for Lung Biology, University of Washington School of Medicine, Seattle, WA, United States
| | - Oliver M. T. Pearce
- Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Carys E. Allen
- Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Charles W. Frevert
- Division of Pulmonary/Critical Care Medicine, Center for Lung Biology, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
20
|
Bogdani M, Speake C, Dufort MJ, Johnson PY, Larmore MJ, Day AJ, Wight TN, Lernmark Å, Greenbaum CJ. Hyaluronan deposition in islets may precede and direct the location of islet immune-cell infiltrates. Diabetologia 2020; 63:549-560. [PMID: 31907557 PMCID: PMC7002022 DOI: 10.1007/s00125-019-05066-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Substantial deposition of the extracellular matrix component hyaluronan (HA) is characteristic of insulitis in overt type 1 diabetes. We investigated whether HA accumulation is detectable in islets early in disease pathogenesis and how this affects the development of insulitis and beta cell mass. METHODS Pancreas tissue from 15 non-diabetic organ donors who were positive for islet autoantibodies (aAbs) and from 14 similarly aged aAb- control donors were examined for the amount of islet HA staining and the presence of insulitis. The kinetics of HA deposition in islets, along with the onset and progression of insulitis and changes in beta cell mass, were investigated in BioBreeding DRLyp/Lyp rats (a model of spontaneous autoimmune diabetes) from 40 days of age until diabetes onset. RESULTS Abundant islet HA deposits were observed in pancreas tissues from n = 3 single- and n = 4 double-aAb+ donors (aAb+HAhigh). In these seven tissues, the HA-stained areas in islets measured 1000 ± 240 μm2 (mean ± SEM) and were fourfold larger than those from aAb- control tissues. The aAb+HAhigh tissues also had a greater prevalence of islets that were highly rich in HA (21% of the islets in these tissues contained the largest HA-stained areas [>2000 μm2] vs less than 1% in tissues from aAb- control donors). The amount of HA staining in islets was associated with the number of aAbs (i.e. single- or double-aAb positivity) but not with HLA genotype or changes in beta cell mass. Among the seven aAb+HAhigh tissues, three from single- and one from double-aAb+ donors did not show any islet immune-cell infiltrates, indicating that HA accumulates in aAb+ donors independently of insulitis. The three aAb+HAhigh tissues that exhibited insulitis had the largest HA-stained areas and, in these tissues, islet-infiltrating immune cells co-localised with the most prominent HA deposits (i.e. with HA-stained areas >2000 μm2). Accumulation of HA in islets was evident prior to insulitis in 7-8-week-old presymptomatic DRLyp/Lyp rats, in which the islet HA-stained area measured 2370 ± 170 μm2 (mean ± SEM), which was threefold larger than in 6-week-old rats. This initial islet HA deposition was not concurrent with beta cell loss. Insulitis was first detected in 9-10-week-old rats, in which the HA-stained areas were 4980 ± 500 μm2. At this age, the rats also exhibited a 44% reduction in beta cell mass. Further enlargement of the HA-positive areas (mean ± SEM: 7220 ± 880 μm2) was associated with invasive insulitis. HA deposits remained abundant in the islets of rats with destructive insulitis, which had lost 85% of their beta cells. CONCLUSIONS/INTERPRETATION This study indicates that HA deposition in islets occurs early in type 1 diabetes and prior to insulitis, and points to a potential role of HA in triggering islet immune-cell infiltration and the promotion of insulitis.
Collapse
Affiliation(s)
- Marika Bogdani
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, 1201 9th Avenue, Seattle, WA, 98101, USA.
| | - Cate Speake
- Diabetes Research Program and Clinical Research Center, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Mathew J Dufort
- Bioinformatics Department, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Pamela Y Johnson
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, 1201 9th Avenue, Seattle, WA, 98101, USA
| | - Megan J Larmore
- Histology and Imaging Core, University of Washington, Seattle, WA, USA
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, 1201 9th Avenue, Seattle, WA, 98101, USA
| | - Åke Lernmark
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Clinical Sciences, Lund University/CRC, Skåne University Hospital, Malmö, Sweden
| | - Carla J Greenbaum
- Diabetes Research Program and Clinical Research Center, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
21
|
Reeves SR, Barrow KA, Rich LM, White MP, Shubin NJ, Chan CK, Kang I, Ziegler SF, Piliponsky AM, Wight TN, Debley JS. Respiratory Syncytial Virus Infection of Human Lung Fibroblasts Induces a Hyaluronan-Enriched Extracellular Matrix That Binds Mast Cells and Enhances Expression of Mast Cell Proteases. Front Immunol 2020; 10:3159. [PMID: 32047499 PMCID: PMC6997473 DOI: 10.3389/fimmu.2019.03159] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 12/31/2019] [Indexed: 12/14/2022] Open
Abstract
Human lung fibroblasts (HLFs) treated with the viral mimetic polyinosine-polycytidylic acid (poly I:C) form an extracellular matrix (ECM) enriched in hyaluronan (HA) that avidly binds monocytes and lymphocytes. Mast cells are important innate immune cells in both asthma and acute respiratory infections including respiratory syncytial virus (RSV); however, the effect of RSV on HA dependent mast cell adhesion and/or function is unknown. To determine if RSV infection of HLFs leads to the formation of a HA-enriched ECM that binds and enhances mast cell activity primary HLFs were infected with RSV for 48 h prior to leukocyte binding studies using a fluorescently labeled human mast cell line (LUVA). Parallel HLFs were harvested for characterization of HA production by ELISA and size exclusion chromatography. In separate experiments, HLFs were infected as above for 48 h prior to adding LUVA cells to HLF wells. Co-cultures were incubated for 48 h at which point media and cell pellets were collected for analysis. The role of the hyaladherin tumor necrosis factor-stimulated gene 6 (TSG-6) was also assessed using siRNA knockdown. RSV infection of primary HLFs for 48 h enhanced HA-dependent LUVA binding assessed by quantitative fluorescent microscopy. This coincided with increased HLF HA synthase (HAS) 2 and HAS3 expression and decreased hyaluronidase (HYAL) 2 expression leading to increased HA accumulation in the HLF cell layer and the presence of larger HA fragments. Separately, LUVAs co-cultured with RSV-infected HLFs for 48 h displayed enhanced production of the mast cell proteases, chymase, and tryptase. Pre-treatment with the HA inhibitor 4-methylumbelliferone (4-MU) and neutralizing antibodies to CD44 (HA receptor) decreased mast cell protease expression in co-cultured LUVAs implicating a direct role for HA. TSG-6 expression was increased over the 48-h infection. Inhibition of HLF TSG-6 expression by siRNA knockdown led to decreased LUVA binding suggesting an important role for this hyaladherin for LUVA adhesion in the setting of RSV infection. In summary, RSV infection of HLFs contributes to inflammation via HA-dependent mechanisms that enhance mast cell binding as well as mast cell protease expression via direct interactions with the ECM.
Collapse
Affiliation(s)
- Stephen R Reeves
- Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, Seattle, WA, United States.,Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Kaitlyn A Barrow
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, United States
| | - Lucille M Rich
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, United States
| | - Maria P White
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, United States
| | - Nicholas J Shubin
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, United States
| | - Christina K Chan
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, United States
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, United States
| | - Steven F Ziegler
- Immunology Program, Benaroya Research Institute, Seattle, WA, United States
| | - Adrian M Piliponsky
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, United States
| | - Jason S Debley
- Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, Seattle, WA, United States.,Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|
22
|
Groux-Degroote S, Cavdarli S, Uchimura K, Allain F, Delannoy P. Glycosylation changes in inflammatory diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 119:111-156. [PMID: 31997767 DOI: 10.1016/bs.apcsb.2019.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Glycosylation is one of the most important modifications of proteins and lipids, and cell surface glycoconjugates are thought to play important roles in a variety of biological functions including cell-cell and cell-substrate interactions, bacterial adhesion, cell immunogenicity and cell signaling. Alterations of glycosylation are observed in a number of inflammatory diseases. Pro-inflammatory cytokines have been shown to modulate cell surface glycosylation by regulating the expression of glycosyltransferases and sulfotransferases involved in the biosynthesis of glycan chains, inducing the expression of specific carbohydrate antigens at the cell surface that can be recognized by different types of lectins or by bacterial adhesins, contributing to the development of diseases. Glycosylation can also regulate biological functions of immune cells by recruiting leukocytes to inflammation sites with pro- or anti-inflammatory effects. Cell surface proteoglycans provide a large panel of binding sites for many mediators of inflammation, and regulate their bio-availability and functions. In this review, we summarize the current knowledge of the glycosylation changes occurring in mucin type O-linked glycans, glycosaminoglycans, as well as in glycosphingolipids, with a particular focus on cystic fibrosis and neurodegenerative diseases, and their consequences on cell interactions and disease progression.
Collapse
Affiliation(s)
- Sophie Groux-Degroote
- University Lille, CNRS, UMR 8576 - UGSF - Unite de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Sumeyye Cavdarli
- University Lille, CNRS, UMR 8576 - UGSF - Unite de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Kenji Uchimura
- University Lille, CNRS, UMR 8576 - UGSF - Unite de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Fabrice Allain
- University Lille, CNRS, UMR 8576 - UGSF - Unite de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Philippe Delannoy
- University Lille, CNRS, UMR 8576 - UGSF - Unite de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| |
Collapse
|
23
|
Petrey AC, Obery DR, Kessler SP, Zawerton A, Flamion B, de la Motte CA. Platelet hyaluronidase-2 regulates the early stages of inflammatory disease in colitis. Blood 2019; 134:765-775. [PMID: 31262781 PMCID: PMC6716076 DOI: 10.1182/blood.2018893594] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/12/2019] [Indexed: 12/31/2022] Open
Abstract
Platelets are specialized cells essential for hemostasis that also function as crucial effectors capable of mediating inflammatory and immune responses. These sentinels continually survey their environment and discriminate between homeostatic and danger signals such as modified components of the extracellular matrix. The glycosaminoglycan hyaluronan (HA) is a major extracellular matrix component that coats the vascular lumen and, under normal conditions, restricts access of inflammatory cells. In response to tissue damage, the endothelial HA matrix enhances leukocyte recruitment and regulates the early stages of the inflammatory response. We have shown that platelets can degrade HA from the surface of activated endothelial cells via the enzyme hyaluronidase-2 (HYAL2) and that HYAL2 is deficient in platelets isolated from patients with inflammatory bowel disease (IBD). Platelets are known to be involved in the pathogenesis of several chronic disease states, including IBD, but they have been largely overlooked in the context of intestinal inflammation. We therefore wanted to define the mechanism by which platelet HYAL2 regulates the inflammatory response during colitis. In this study, we provide evidence that HA catabolism is disrupted in human intestinal microvascular endothelial cells isolated from patients with IBD. Furthermore, mice deficient in HYAL2 are more susceptible to an acute model of colitis, and this increased susceptibility is abrogated by transfusion of HYAL2-competent platelets. Finally, we show that platelets, via HYAL2-dependent degradation of endothelial HA, regulate the early stages of inflammation in colitis by limiting leukocyte extravasation.
Collapse
Affiliation(s)
- Aaron C Petrey
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH; and
| | - Dana R Obery
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH; and
| | - Sean P Kessler
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH; and
| | - Ash Zawerton
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH; and
| | - Bruno Flamion
- Molecular Physiology Research Unit, Namur Research Institute for Life Sciences, University of Namur, Namur, Belgium
| | - Carol A de la Motte
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH; and
| |
Collapse
|
24
|
Hascall VC. The journey of hyaluronan research in the Journal of Biological Chemistry. J Biol Chem 2019; 294:1690-1696. [PMID: 30710015 DOI: 10.1074/jbc.tm118.005836] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hyaluronan has a very simple structure. It is a linear glycosaminoglycan composed of disaccharide units of GlcNAc and d-glucuronic acid with alternating β-1,4 and β-1,3 glycosidic bonds that can be repeated 20,000 or more times, a molecular mass >8 million Da, and a length >20 μm. However, it has a very complex biology. It is a major, ubiquitous component of extracellular matrices involved in everything from fertilization, development, inflammations, to cancer. This JBC Review highlights some of these processes that were initiated through publications in the Journal of Biological Chemistry.
Collapse
Affiliation(s)
- Vincent C Hascall
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195.
| |
Collapse
|
25
|
Petrey AC, de la Motte CA. Hyaluronan in inflammatory bowel disease: Cross-linking inflammation and coagulation. Matrix Biol 2019; 78-79:314-323. [PMID: 29574062 PMCID: PMC6150849 DOI: 10.1016/j.matbio.2018.03.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/13/2018] [Accepted: 03/14/2018] [Indexed: 12/14/2022]
Abstract
Hyaluronan, a major extracellular matrix component, is an active participant in many disease states, including inflammatory bowel disease (IBD). The synthesis of this dynamic polymer is increased at sites of inflammation. Hyaluronan together with the enzymes responsible for its synthesis, degradation, and its binding proteins, directly modulates the promotion and resolution of disease by controlling recruitment of immune cells, by release of inflammatory cytokines, and by balancing hemostasis. This review discusses the functional significance of hyaluronan in the cells and tissues involved in inflammatory bowel disease pathobiology.
Collapse
Affiliation(s)
- Aaron C Petrey
- Department of Pathobiology, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Carol A de la Motte
- Department of Pathobiology, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States.
| |
Collapse
|
26
|
Wang AJ, Ren J, Abbadi A, Wang A, Hascall VC. Heparin affects cytosolic glucose responses of hyperglycemic dividing mesangial cells. J Biol Chem 2019; 294:6591-6597. [PMID: 30723159 DOI: 10.1074/jbc.ra119.007395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/30/2019] [Indexed: 11/06/2022] Open
Abstract
Mesangial expansion underlies diabetic nephropathy, leading to sclerosis and renal failure. The glycosaminoglycan heparin inhibits mesangial cell growth, but the molecular mechanism is unclear. Here, rat mesangial cells (RMCs) were growth-arrested in the G0/G1 phase of cell division, stimulated to divide in normal glucose (5.6 mm) or high glucose (25.6 mm) with or without heparin, and analyzed for glucose uptake. We observed that RMCs entering the G1 phase in normal glucose with or without heparin rapidly cease glucose uptake. RMCs entering G1 in high glucose sustained glucose uptake for the first 3 h, and high-glucose exposure of RMCs only in the first 8 h of G1 induced the formation of an extracellular monocyte-adhesive hyaluronan matrix after cell division was completed. Moreover, a low heparin concentration under high-glucose conditions blocked glucose uptake by 1 h into G1 Of note, glucose transporter 4 (glut4) localized on the RMC surface at G0/G1 and was internalized into G1 cells under normal glucose conditions with or without heparin within 30 min. We also noted that, under high-glucose conditions, glut4 remained on the RMC surface for at least 2 h into G1 and was internalized by 4 h without heparin and within 1 h with heparin. These results provide evidence that the influx of glucose in hyperglycemic dividing RMCs initiates intermediate glucose metabolism, leading to increased cytosolic UDP sugars, and induces abnormal intracellular hyaluronan synthesis during the S phase of cell division.
Collapse
Affiliation(s)
- Andrew Jun Wang
- From the Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195
| | - Juan Ren
- From the Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195
| | - Amina Abbadi
- From the Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195
| | - Aimin Wang
- From the Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195
| | - Vincent C Hascall
- From the Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
27
|
Wight TN. A role for proteoglycans in vascular disease. Matrix Biol 2018; 71-72:396-420. [PMID: 29499356 PMCID: PMC6110991 DOI: 10.1016/j.matbio.2018.02.019] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 12/15/2022]
Abstract
The content of proteoglycans (PGs) is low in the extracellular matrix (ECM) of vascular tissue, but increases dramatically in all phases of vascular disease. Early studies demonstrated that glycosaminoglycans (GAGs) including chondroitin sulfate (CS), dermatan sulfate (DS), keratan sulfate (KS) and heparan sulfate (HS) accumulate in vascular lesions in both humans and in animal models in areas of the vasculature that are susceptible to disease initiation (such as at branch points) and are frequently coincident with lipid deposits. Later studies showed the GAGs were covalently attached to specific types of core proteins that accumulate in vascular lesions. These molecules include versican (CSPG), biglycan and decorin (DS/CSPGs), lumican and fibromodulin (KSPGs) and perlecan (HSPG), although other types of PGs are present, but in lesser quantities. While the overall molecular design of these macromolecules is similar, there is tremendous structural diversity among the different PG families creating multiple forms that have selective roles in critical events that form the basis of vascular disease. PGs interact with a variety of different molecules involved in disease pathogenesis. For example, PGs bind and trap serum components that accumulate in vascular lesions such as lipoproteins, amyloid, calcium, and clotting factors. PGs interact with other ECM components and regulate, in part, ECM assembly and turnover. PGs interact with cells within the lesion and alter the phenotypes of both resident cells and cells that invade the lesion from the circulation. A number of therapeutic strategies have been developed to target specific PGs involved in key pathways that promote vascular disease. This review will provide a historical perspective of this field of research and then highlight some of the evidence that defines the involvement of PGs and their roles in the pathogenesis of vascular disease.
Collapse
Affiliation(s)
- Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, United States.
| |
Collapse
|
28
|
Reeves SR, Kang I, Chan CK, Barrow KA, Kolstad TK, White MP, Ziegler SF, Wight TN, Debley JS. Asthmatic bronchial epithelial cells promote the establishment of a Hyaluronan-enriched, leukocyte-adhesive extracellular matrix by lung fibroblasts. Respir Res 2018; 19:146. [PMID: 30071849 PMCID: PMC6090698 DOI: 10.1186/s12931-018-0849-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/23/2018] [Indexed: 02/07/2023] Open
Abstract
Background Airway inflammation is a hallmark of asthma. Alterations in extracellular matrix (ECM) hyaluronan (HA) content have been shown to modulate the recruitment and retention of inflammatory cells. Bronchial epithelial cells (BECs) regulate the activity of human lung fibroblasts (HLFs); however, their contribution in regulating HLF production of HA in asthma is unknown. In this study, we tested the hypothesis that BECs from asthmatic children promote the generation of a pro-inflammatory, HA-enriched ECM by HLFs, which promotes the retention of leukocytes. Methods BECs were obtained from well-characterized asthmatic and healthy children ages 6–18 years. HLFs were co-cultured with BECs for 96 h and samples were harvested for analysis of gene expression, synthesis and accumulation of HA, and subjected to a leukocyte adhesion assay with U937 monocytes. Results We observed increased expression of HA synthases HAS2 and HAS3 in HLFs co-cultured with asthmatic BECs. Furthermore, we demonstrated greater total accumulation and increased synthesis of HA by HLFs co-cultured with asthmatic BECs compared to healthy BEC/HLF co-cultures. ECM generated by HLFs co-cultured with asthmatic BECs displayed increased HA-dependent adhesion of leukocytes in a separate in vitro binding assay. Conclusions Our findings demonstrate that BEC regulation of HA production by HLFs is altered in asthma, which may in turn promote the establishment of a more leukocyte-permissive ECM promoting airway inflammation in this disease. Electronic supplementary material The online version of this article (10.1186/s12931-018-0849-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stephen R Reeves
- Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA, 98105, USA. .,Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA. .,Department of Pediatrics, University of Washington, Seattle, WA, USA.
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Christina K Chan
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Kaitlyn A Barrow
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Tessa K Kolstad
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Maria P White
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Steven F Ziegler
- Immunology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Jason S Debley
- Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA, 98105, USA.,Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
29
|
Majors AK, Chakravarti R, Ruple LM, Leahy R, Stuehr DJ, Lauer M, Erzurum SC, Janocha A, Aronica MA. Nitric oxide alters hyaluronan deposition by airway smooth muscle cells. PLoS One 2018; 13:e0200074. [PMID: 29966020 PMCID: PMC6028120 DOI: 10.1371/journal.pone.0200074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 06/19/2018] [Indexed: 12/04/2022] Open
Abstract
Asthma is a chronic inflammatory disease that is known to cause changes in the extracellular matrix, including changes in hyaluronan (HA) deposition. However, little is known about the factors that modulate its deposition or the potential consequences. Asthmatics with high levels of exhaled nitric oxide (NO) are characterized by greater airway reactivity and greater evidence of airway inflammation. Based on these data and our previous work we hypothesized that excessive NO promotes the pathologic production of HA by airway smooth muscle cells (SMCs). Exposure of cultured SMCs to various NO donors results in the accumulation of HA in the form of unique, cable-like structures. HA accumulates rapidly after exposure to NO and can be seen as early as one hour after NO treatment. The cable-like HA in NO-treated SMC cultures supports the binding of leukocytes. In addition, NO produced by murine macrophages (RAW cells) and airway epithelial cells also induces SMCs to produce HA cables when grown in co-culture. The modulation of HA by NO appears to be independent of soluble guanylate cyclase. Taken together, NO-induced production of leukocyte-binding HA by SMCs provides a new potential mechanism for the non-resolving airway inflammation in asthma and suggests a key role of non-immune cells in driving the chronic inflammation of the submucosa. Modulation of NO, HA and the consequent immune cell interactions may serve as potential therapeutic targets in asthma.
Collapse
Affiliation(s)
- Alana K. Majors
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ritu Chakravarti
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Lisa M. Ruple
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Rachel Leahy
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Dennis J. Stuehr
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Mark Lauer
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Serpil C. Erzurum
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Allison Janocha
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Mark A. Aronica
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| |
Collapse
|
30
|
Jackson DG. Hyaluronan in the lymphatics: The key role of the hyaluronan receptor LYVE-1 in leucocyte trafficking. Matrix Biol 2018; 78-79:219-235. [PMID: 29425695 DOI: 10.1016/j.matbio.2018.02.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 12/25/2022]
Abstract
LYVE-1, a close relative of the leucocyte receptor, CD44, is the main receptor for hyaluronan (HA) in lymphatic vessel endothelium and a widely used marker for distinguishing between blood and lymphatic vessels. Enigmatic for many years because of its anomalous HA-binding characteristics, the function of LYVE-1 has just recently been identified as that of a lymphatic docking receptor for dendritic cells, selectively engaging with their surface HA glycocalyx to regulate entry to peripheral lymphatics and migration to downstream lymph nodes for immune activation. Furthermore, LYVE-1 mediates the trafficking of macrophages, and is also exploited by HA-encapsulated Group A streptococci for lymphatic invasion and host dissemination. Consistent with a role in lymphatic trafficking, the interaction of LYVE-1 with HA and its degradation products can also activate intracellular signalling pathways for endothelial junctional retraction and lymphatic endothelial proliferation. Here we outline the latest findings on the receptor in the context of its peculiar biochemical properties and speculate on how the interaction of LYVE-1 with different HA sizes and conformations might variably influence cell function as a consequence of avidity and receptor crosslinking. Finally, we evaluate evidence that LYVE-1 can also bind growth factors and associate with kinase-linked growth factor receptors and conclude on how the LYVE-1·HA axis may be exploited as a target to either block inflammation or tissue allograft rejection, or potentiate vaccine and drug delivery.
Collapse
Affiliation(s)
- David G Jackson
- University of Oxford, MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK.
| |
Collapse
|
31
|
Heldin P, Lin CY, Kolliopoulos C, Chen YH, Skandalis SS. Regulation of hyaluronan biosynthesis and clinical impact of excessive hyaluronan production. Matrix Biol 2018; 78-79:100-117. [PMID: 29374576 DOI: 10.1016/j.matbio.2018.01.017] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 10/25/2022]
Abstract
The tightly regulated biosynthesis and catabolism of the glycosaminoglycan hyaluronan, as well as its role in organizing tissues and cell signaling, is crucial for the homeostasis of tissues. Overexpression of hyaluronan plays pivotal roles in inflammation and cancer, and markedly high serum and tissue levels of hyaluronan are noted under such pathological conditions. This review focuses on the complexity of the regulation at transcriptional and posttranslational level of hyaluronan synthetic enzymes, and the outcome of their aberrant expression and accumulation of hyaluronan in clinical conditions, such as systemic B-cell cancers, aggressive breast carcinomas, metabolic diseases and virus infection.
Collapse
Affiliation(s)
- Paraskevi Heldin
- Department Medical Biochemistry and Microbiology, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden.
| | - Chun-Yu Lin
- Department Medical Biochemistry and Microbiology, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden; Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Constantinos Kolliopoulos
- Department Medical Biochemistry and Microbiology, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden
| | - Yen-Hsu Chen
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsin Chu, Taiwan
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| |
Collapse
|
32
|
Filpa V, Bistoletti M, Caon I, Moro E, Grimaldi A, Moretto P, Baj A, Giron MC, Karousou E, Viola M, Crema F, Frigo G, Passi A, Giaroni C, Vigetti D. Changes in hyaluronan deposition in the rat myenteric plexus after experimentally-induced colitis. Sci Rep 2017; 7:17644. [PMID: 29247178 PMCID: PMC5732300 DOI: 10.1038/s41598-017-18020-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 12/05/2017] [Indexed: 12/27/2022] Open
Abstract
Myenteric plexus alterations hamper gastrointestinal motor function during intestinal inflammation. Hyaluronan (HA), an extracellular matrix glycosaminoglycan involved in inflammatory responses, may play a role in this process. In the colon of control rats, HA-binding protein (HABP), was detected in myenteric neuron soma, perineuronal space and ganglia surfaces. Prominent hyaluronan synthase 2 (HAS2) staining was found in myenteric neuron cytoplasm, suggesting that myenteric neurons produce HA. In the myenteric plexus of rats with 2, 4-dinitrobenzene sulfonic (DNBS)-induced colitis HABP staining was altered in the perineuronal space, while both HABP staining and HA levels increased in the muscularis propria. HAS2 immunopositive myenteric neurons and HAS2 mRNA and protein levels also increased. Overall, these observations suggest that inflammation alters HA distribution and levels in the gut neuromuscular compartment. Such changes may contribute to alterations in the myenteric plexus.
Collapse
Affiliation(s)
- Viviana Filpa
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Ilaria Caon
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Elisabetta Moro
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Annalisa Grimaldi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Paola Moretto
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Gianmario Frigo
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy.
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| |
Collapse
|
33
|
Varol C, Sagi I. Phagocyte-extracellular matrix crosstalk empowers tumor development and dissemination. FEBS J 2017; 285:734-751. [DOI: 10.1111/febs.14317] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/01/2017] [Accepted: 10/31/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Chen Varol
- The Research Center for Digestive Tract and Liver Diseases; Tel-Aviv Sourasky Medical Center; Sackler Faculty of Medicine; Tel-Aviv University; Israel
- Department of Clinical Microbiology and Immunology; Sackler Faculty of Medicine; Tel Aviv University; Israel
| | - Irit Sagi
- Department of Biological Regulation; Weizmann Institute of Science; Rehovot Israel
| |
Collapse
|
34
|
Hyaluronan-mediated mononuclear leukocyte binding to gingival fibroblasts. Clin Oral Investig 2017; 22:1063-1070. [PMID: 28801807 DOI: 10.1007/s00784-017-2188-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 07/20/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Binding of mononuclear leukocytes to hyaluronan cable structures is a well-known pathomechanism in several chronic inflammatory diseases, but has not yet described for chronic oral inflammations. The aim of this study was to evaluate if and how binding of mononuclear leukocytes to pathologic hyaluronan cable structures can be induced in human gingival fibroblasts. MATERIAL AND METHODS Experiments were performed with human gingival fibroblasts and peripheral blood mononuclear cells (PBMCs) from three healthy blood donors. Gingival fibroblasts were stimulated with (1) tunicamycin, (2) polyinosinic/polycytidylic acid (Poly:IC), and (3) lipopolysaccharides (LPS) to simulate (1) ER stress and (2) viral and (3) bacterial infections, respectively. Fibroblasts were then co-incubated with PBMCs, and the number of bound and fluorescently labeled PBMCs was assessed using a fluorescence reader and microscopy. For data analysis, a linear mixed model was used. RESULTS Hyaluronan-mediated binding of PBMCs to gingival fibroblasts was increased by tunicamycin and Poly(I:C) but not by LPS. Hyaluronidase treatment and co-incubation with hyaluronan transport inhibitors reduced this binding. CONCLUSIONS Results suggest that hyaluronan-mediated binding of blood cells might play a role in oral inflammations. A potential superior role of viruses needs to be confirmed in further clinical studies. CLINICAL RELEVANCE The linkage between pathological hyaluronan matrices and oral infections opens up potential applications of hyaluronan transport inhibitors in the treatment of chronic oral inflammations.
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW The extracellular matrix (ECM) is a frequently overlooked component of the pathogenesis of inflammatory bowel disease (IBD). However, the functional and clinically significant interactions between immune as well as nonimmune cells with the ECM have important implications for disease pathogenesis. In this review, we discuss how the ECM participates in process associated with IBD that involves diverse cell types of the intestine. RECENT FINDINGS Remodeling of the ECM is a consistent feature of IBD, and studies have implicated key ECM molecules in IBD pathogenesis. While the majority of prior studies have focused on ECM degradation by proteases, more recent studies have uncovered additional degrading enzymes, identified fragments of ECM components as potential biomarkers, and revealed that ECM synthesis is increased in IBD. These new studies support the notion that the ECM, rather than acting as a passive element, is an active participant in promoting inflammation. SUMMARY New studies have offered exciting clues about the function of the ECM during IBD pathogenesis. The balance of ECM synthesis and turnover is altered in IBD, and the molecules involved exhibit discreet biological functions that regulate inflammation on the basis of specific cell type and matrix molecule.
Collapse
|
36
|
Gaucherand L, Falk BA, Evanko SP, Workman G, Chan CK, Wight TN. Crosstalk Between T Lymphocytes and Lung Fibroblasts: Generation of a Hyaluronan-Enriched Extracellular Matrix Adhesive for Monocytes. J Cell Biochem 2017; 118:2118-2130. [PMID: 27982477 DOI: 10.1002/jcb.25842] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/14/2016] [Indexed: 01/13/2023]
Abstract
In immunity and inflammation, T cells are often associated with stromal mesenchymal cells such as fibroblasts. Hyaluronan and proteins that associate with hyaluronan such as versican and tumor necrosis factor-inducible gene-6 (TSG-6) are extracellular matrix (ECM) components that promote leukocyte adhesion, accumulation, and activation. However, the factors responsible for producing this specialized ECM and its impact on inflammatory events are not well understood. In this study, we explored the role of T cells in stimulating lung fibroblasts to produce an ECM that impacts monocyte adhesion. We found that CD3/CD28-activated human CD4+ T cells when co-cultured with human lung fibroblasts stimulated the expression of mRNA for hyaluronan synthase 2 (HAS2) and decreased the expression of hyaluronidase 2 (HYAL2). This led to an increase in the deposition of hyaluronan that formed cable-like structures within the ECM. Co-culturing activated T cells with fibroblasts also led to increased expression and accumulation of TSG-6. Surprisingly, addition of activated CD4+ T cells to the fibroblasts reduced the expression of mRNA for versican, and increased the expression of enzymes that degrade versican, such as ADAMTS4 and ADAMTS9 (a disintegrin and metalloproteinase with a thrombospondin type-1 motif) leading to a decrease in versican in the ECM of the co-cultures. Furthermore, addition of human monocytes to these co-cultures resulted in elevated monocyte adhesion to the cable-like structures in the ECM when compared to controls. These results illustrate the importance of crosstalk between T cells and fibroblasts in promoting the generation of a matrix that is adhesive for monocytes. J. Cell. Biochem. 118: 2118-2130, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Léa Gaucherand
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington
| | - Ben A Falk
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington
| | - Stephen P Evanko
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington
| | - Gail Workman
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington
| | - Christina K Chan
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington
| |
Collapse
|
37
|
Smigiel KS, Parks WC. Matrix Metalloproteinases and Leukocyte Activation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:167-195. [PMID: 28413028 DOI: 10.1016/bs.pmbts.2017.01.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
As their name implies, matrix metalloproteinases (MMPs) are thought to degrade extracellular matrix proteins, a function that is indeed performed by some members. However, regardless of their cell source, matrix degradation is not the only function of these enzymes. Rather, individual MMPs have been shown to regulate specific immune processes, such as leukocyte influx and migration, antimicrobial activity, macrophage activation, and restoration of barrier function, typically by processing a range of nonmatrix protein substrates. Indeed, MMP expression is low under steady-state conditions but is markedly induced during inflammatory processes including infection, wound healing, and cancer. Increasing research is showing that MMPs are not just a downstream consequence of a generalized inflammatory process, but rather are critical factors in the overall regulation of the pattern, type, and duration of immune responses. This chapter outlines the role of leukocytes in tissue remodeling and describes recent progress in our understanding of how MMPs alter leukocyte activity.
Collapse
Affiliation(s)
- Kate S Smigiel
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - William C Parks
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
38
|
Wight TN, Frevert CW, Debley JS, Reeves SR, Parks WC, Ziegler SF. Interplay of extracellular matrix and leukocytes in lung inflammation. Cell Immunol 2017; 312:1-14. [PMID: 28077237 PMCID: PMC5290208 DOI: 10.1016/j.cellimm.2016.12.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022]
Abstract
During inflammation, leukocytes influx into lung compartments and interact with extracellular matrix (ECM). Two ECM components, versican and hyaluronan, increase in a range of lung diseases. The interaction of leukocytes with these ECM components controls leukocyte retention and accumulation, proliferation, migration, differentiation, and activation as part of the inflammatory phase of lung disease. In addition, bronchial epithelial cells from asthmatic children co-cultured with human lung fibroblasts generate an ECM that is adherent for monocytes/macrophages. Macrophages are present in both early and late lung inflammation. Matrix metalloproteinase 10 (MMP10) is induced in alveolar macrophages with injury and infection and modulates macrophage phenotype and their ability to degrade collagenous ECM components. Collectively, studies outlined in this review highlight the importance of specific ECM components in the regulation of inflammatory events in lung disease. The widespread involvement of these ECM components in the pathogenesis of lung inflammation make them attractive candidates for therapeutic intervention.
Collapse
Affiliation(s)
- Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| | - Charles W Frevert
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Jason S Debley
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, and Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Stephen R Reeves
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, and Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Steven F Ziegler
- Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
39
|
Abstract
Simple and complex carbohydrates (glycans) have long been known to play major metabolic, structural and physical roles in biological systems. Targeted microbial binding to host glycans has also been studied for decades. But such biological roles can only explain some of the remarkable complexity and organismal diversity of glycans in nature. Reviewing the subject about two decades ago, one could find very few clear-cut instances of glycan-recognition-specific biological roles of glycans that were of intrinsic value to the organism expressing them. In striking contrast there is now a profusion of examples, such that this updated review cannot be comprehensive. Instead, a historical overview is presented, broad principles outlined and a few examples cited, representing diverse types of roles, mediated by various glycan classes, in different evolutionary lineages. What remains unchanged is the fact that while all theories regarding biological roles of glycans are supported by compelling evidence, exceptions to each can be found. In retrospect, this is not surprising. Complex and diverse glycans appear to be ubiquitous to all cells in nature, and essential to all life forms. Thus, >3 billion years of evolution consistently generated organisms that use these molecules for many key biological roles, even while sometimes coopting them for minor functions. In this respect, glycans are no different from other major macromolecular building blocks of life (nucleic acids, proteins and lipids), simply more rapidly evolving and complex. It is time for the diverse functional roles of glycans to be fully incorporated into the mainstream of biological sciences.
Collapse
Affiliation(s)
- Ajit Varki
- Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center, University of California at San Diego, La Jolla, CA 92093-0687, USA
| |
Collapse
|
40
|
Hyaluronan mediates the adhesion of porcine peripheral blood mononuclear cells to poly (I:C)-treated intestinal cells and modulates their cytokine production. Vet Immunol Immunopathol 2016; 184:8-17. [PMID: 28166932 DOI: 10.1016/j.vetimm.2016.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/18/2016] [Accepted: 12/22/2016] [Indexed: 02/07/2023]
Abstract
Hyaluronan (HA), a major component of the extracellular matrix (ECM), has been increasingly recognized as a regulator of inflammation. Its role is complex since it has pro- and anti-inflammatory actions by modulating the expression of inflammatory genes, the recruitment of inflammatory cells and the production of inflammatory cytokines, but also by attenuating the course of inflammation and providing protection against tissue damage. Certain viruses and other inflammatory stimuli induce organization of HA into cable-like structures, which may be responsible for leukocyte recruitment and, on the other hand, low molecular weight fragments of HA have been shown to activate various inflammatory responses. The aim of the present study was to analyze the effects of a simulated infection with the viral mimetic Poly (I:C) on HA deposition on different porcine intestinal cells (primary colonic muscular smooth muscle cells (SMC), and epithelial IPEC-J2 and IPI-2I cell lines) and on the recruitment of peripheral blood mononuclear cells (PBMC) to intestinal cell layers. We show that Poly (I:C) treatment induces the formation of an HA-based pericellular matrix coat in muscular SMC and in intestinal epithelial cells (IECs) and that, on differentiated IPEC-J2 cells, HA accumulates in the basolateral membrane. Porcine PBMCs bind to Poly (I:C)-treated cells and this binding is dependent on HA, since the increase in adhesion is abolished by hyaluronidase treatment of the cell layers. A second goal was to study the effect of different molecular weight HA forms on the production of pro-inflammatory cytokines and chemokines (TNF-α, IL-1β and IL-8) by porcine PBMCs. Low molecular weight HA fragments (100-150kDa), in contrast to high molecular weight HA (2500kDa), stimulate the release of these pro-inflammatory mediators by porcine PBMCs. Our results suggest that HA is involved in the inflammatory response against pathogenic insults to the porcine gut.
Collapse
|
41
|
Wight TN. Provisional matrix: A role for versican and hyaluronan. Matrix Biol 2016; 60-61:38-56. [PMID: 27932299 DOI: 10.1016/j.matbio.2016.12.001] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 11/22/2016] [Accepted: 12/01/2016] [Indexed: 12/19/2022]
Abstract
Hyaluronan and versican are extracellular matrix (ECM) components that are enriched in the provisional matrices that form during the early stages of development and disease. These two molecules interact to create pericellular "coats" and "open space" that facilitate cell sorting, proliferation, migration, and survival. Such complexes also impact the recruitment of leukocytes during development and in the early stages of disease. Once thought to be inert components of the ECM that help hold cells together, it is now quite clear that they play important roles in controlling cell phenotype, shaping tissue response to injury and maintaining tissue homeostasis. Conversion of hyaluronan-/versican-enriched provisional matrix to collagen-rich matrix is a "hallmark" of tissue fibrosis. Targeting the hyaluronan and versican content of provisional matrices in a variety of diseases including, cardiovascular disease and cancer, is becoming an attractive strategy for intervention.
Collapse
Affiliation(s)
- Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, 1201 9th Avenue, Seattle, WA 98101, United States.
| |
Collapse
|
42
|
Petrey AC, de la Motte CA. Thrombin Cleavage of Inter-α-inhibitor Heavy Chain 1 Regulates Leukocyte Binding to an Inflammatory Hyaluronan Matrix. J Biol Chem 2016; 291:24324-24334. [PMID: 27679489 DOI: 10.1074/jbc.m116.755660] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Indexed: 02/06/2023] Open
Abstract
Dynamic alterations of the extracellular matrix in response to injury directly modulate inflammation and consequently the promotion and resolution of disease. During inflammation, hyaluronan (HA) is increased at sites of inflammation where it may be covalently modified with the heavy chains (HC) of inter-α-trypsin inhibitor. Deposition of this unique, pathological form of HA (HC-HA) leads to the formation of cable-like structures that promote adhesion of leukocytes. Naive mononuclear leukocytes bind specifically to inflammation-associated HA matrices but do not adhere to HA constitutively expressed under homeostatic conditions. In this study, we have directly investigated a role for the blood-coagulation protease thrombin in regulating the adhesion of monocytic cells to smooth muscle cells producing an inflammatory matrix. Our data demonstrate that the proteolytic activity of thrombin negatively regulates the adhesion of monocytes to an inflammatory HC-HA complex. This effect is independent of protease-activated receptor activation but requires proteolytic activity toward a novel substrate. Components of HC-HA complexes were predicted to contain conserved thrombin-susceptible cleavage sites based on sequence analysis, and heavy chain 1 (HC1) was confirmed to be a substrate of thrombin. Thrombin treatment is sufficient to cleave HC1 associated with either cell-surface HA or serum inter-α-trypsin inhibitor. Furthermore, thrombin treatment of the inflammatory matrix leads to dissolution of HC-HA cable structures and abolishes leukocyte adhesion. These data establish a novel mechanism whereby thrombin cleavage of HC1 regulates the adhesive properties of an inflammatory HA matrix.
Collapse
Affiliation(s)
- Aaron C Petrey
- From the Department of Pathobiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195
| | - Carol A de la Motte
- From the Department of Pathobiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195.
| |
Collapse
|
43
|
McNamee EN, Rivera-Nieves J. Ectopic Tertiary Lymphoid Tissue in Inflammatory Bowel Disease: Protective or Provocateur? Front Immunol 2016; 7:308. [PMID: 27579025 PMCID: PMC4985530 DOI: 10.3389/fimmu.2016.00308] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/29/2016] [Indexed: 12/15/2022] Open
Abstract
Organized lymphoid tissues like the thymus first appeared in jawed vertebrates around 500 million years ago and have evolved to equip the host with a network of specialized sites, strategically located to orchestrate strict immune-surveillance and efficient immune responses autonomously. The gut-associated lymphoid tissues maintain a mostly tolerant environment to dampen our responses to daily dietary and microbial products in the intestine. However, when this homeostasis is perturbed by chronic inflammation, the intestine is able to develop florid organized tertiary lymphoid tissues (TLT), which heralds the onset of regional immune dysregulation. While TLT are a pathologic hallmark of Crohn's disease (CD), their role in the overall process remains largely enigmatic. A critical question remains; are intestinal TLT generated by the immune infiltrated intestine to modulate immune responses and rebuild tolerance to the microbiota or are they playing a more sinister role by generating dysregulated responses that perpetuate disease? Herein, we discuss the main theories of intestinal TLT neogenesis and focus on the most recent findings that open new perspectives to their role in inflammatory bowel disease.
Collapse
Affiliation(s)
- Eóin N McNamee
- Mucosal Inflammation Program, Department of Anesthesiology, School of Medicine, University of Colorado - Anschutz Medical Campus , Aurora, CO , USA
| | - Jesús Rivera-Nieves
- Division of Gastroenterology, Inflammatory Bowel Disease Center, San Diego VAMC, University of California San Diego , La Jolla, CA , USA
| |
Collapse
|
44
|
McMahan RS, Birkland TP, Smigiel KS, Vandivort TC, Rohani MG, Manicone AM, McGuire JK, Gharib SA, Parks WC. Stromelysin-2 (MMP10) Moderates Inflammation by Controlling Macrophage Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:899-909. [PMID: 27316687 PMCID: PMC4955757 DOI: 10.4049/jimmunol.1600502] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/24/2016] [Indexed: 02/06/2023]
Abstract
Several members of the matrix metalloproteinase (MMP) family control a range of immune processes, such as leukocyte influx and chemokine activity. Stromelysin-2 (MMP10) is expressed by macrophages in numerous tissues after injury; however, little is known of its function. In this study, we report that MMP10 is expressed by macrophages in human lungs from patients with cystic fibrosis and induced in mouse macrophages in response to Pseudomonas aeruginosa infection both in vivo and by isolated resident alveolar and bone marrow-derived macrophages (BMDM). Our data indicates that macrophage MMP10 serves a beneficial function in response to acute infection. Whereas wild-type mice survived infection with minimal morbidity, 50% of Mmp10(-/-) mice died and all showed sustained weight loss (morbidity). Although bacterial clearance and neutrophil influx did not differ between genotypes, macrophage numbers were ∼3-fold greater in infected Mmp10(-/-) lungs than in wild-types. Adoptive transfer of wild-type BMDM normalized infection-induced morbidity in Mmp10(-/-) recipients to wild-type levels, demonstrating that the protective effect of MMP10 was due to its production by macrophages. Both in vivo and in cultured alveolar macrophages and BMDM, expression of several M1 macrophage markers was elevated, whereas M2 markers were reduced in Mmp10(-/-) tissue and cells. Global gene expression analysis revealed that infection-mediated transcriptional changes persisted in Mmp10(-/-) BMDM long after they were downregulated in wild-type cells. These results indicate that MMP10 serves a beneficial role in response to acute infection by moderating the proinflammatory response of resident and infiltrating macrophages.
Collapse
Affiliation(s)
- Ryan S McMahan
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105
| | - Timothy P Birkland
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Medicine, University of Washington, Seattle, WA 98195
| | - Kate S Smigiel
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Tyler C Vandivort
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105; Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Maryam G Rohani
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Medicine, University of Washington, Seattle, WA 98195; Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Anne M Manicone
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Medicine, University of Washington, Seattle, WA 98195
| | - John K McGuire
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Pediatrics, University of Washington, Seattle, WA 98195
| | - Sina A Gharib
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Medicine, University of Washington, Seattle, WA 98195
| | - William C Parks
- Center for Lung Biology, University of Washington, Seattle, WA 98109; Department of Medicine, University of Washington, Seattle, WA 98195; Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| |
Collapse
|
45
|
Merrilees MJ, Zuo N, Evanko SP, Day AJ, Wight TN. G1 Domain of Versican Regulates Hyaluronan Organization and the Phenotype of Cultured Human Dermal Fibroblasts. J Histochem Cytochem 2016; 64:353-63. [PMID: 27126822 PMCID: PMC4888412 DOI: 10.1369/0022155416643913] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/17/2016] [Indexed: 12/21/2022] Open
Abstract
Variants of versican have wide-ranging effects on cell and tissue phenotype, impacting proliferation, adhesion, pericellular matrix composition, and elastogenesis. The G1 domain of versican, which contains two Link modules that bind to hyaluronan (HA), may be central to these effects. Recombinant human G1 (rhG1) with an N-terminal 8 amino acid histidine (His) tag, produced in Nicotiana benthamiana, was applied to cultures of dermal fibroblasts, and effects on proliferation and pericellular HA organization determined. rhG1 located to individual strands of cell surface HA which aggregated into structures resembling HA cables. On both individual and aggregated strands, the spacing of attached rhG1 was similar (~120 nm), suggesting interaction between rhG1 molecules. Endogenous V0/V1, present on HA between attached rhG1, did not prevent cable formation, while treatment with V0/V1 alone, which also bound to HA, did not induce cables. A single treatment with rhG1 suppressed cell proliferation for an extended period. Treating cells for 4 weeks with rhG1 resulted in condensed layers of elongated, differentiated α actin-positive fibroblasts, with rhG1 localized to cell surfaces, and a compact extracellular matrix including both collagen and elastin. These results demonstrate that the G1 domain of versican can regulate the organization of pericellular HA and affect phenotype.
Collapse
Affiliation(s)
- Mervyn J. Merrilees
- Mervyn J. Merrilees, Department of Anatomy
and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health
Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New
Zealand. E-mail:
| | | | | | | | | |
Collapse
|
46
|
Poukka M, Bykachev A, Siiskonen H, Tyynelä-Korhonen K, Auvinen P, Pasonen-Seppänen S, Sironen R. Decreased expression of hyaluronan synthase 1 and 2 associates with poor prognosis in cutaneous melanoma. BMC Cancer 2016; 16:313. [PMID: 27184066 PMCID: PMC4867536 DOI: 10.1186/s12885-016-2344-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 05/08/2016] [Indexed: 12/16/2022] Open
Abstract
Background Hyaluronan is a large extracellular matrix molecule involved in several biological processes such as proliferation, migration and invasion. In many cancers, hyaluronan synthesis is altered, which implicates disease progression and metastatic potential. We have previously shown that synthesis of hyaluronan and expression of its synthases 1–2 (HAS1-2) decrease in cutaneous melanoma, compared to benign melanocytic lesions. Methods In the present study, we compared immunohistological staining results of HAS1 and HAS2 with clinical and histopathological parameters to investigate whether HAS1 or HAS2 has prognostic value in cutaneous melanoma. The specimens consisted of 129 tissue samples including superficial (Breslow ≤ 1 mm) and deep (Breslow > 4 mm) melanomas and lymph node metastases. The differences in immunostainings were analysed with non-parametric Mann–Whitney U test. Associations between immunohistological staining results and clinical parameters were determined with the χ2 test. Survival between patient groups was compared by the Kaplan-Meier method using log rank test and Cox’s regression model was used for multivariate analyses. Results The expression of HAS1 and HAS2 was decreased in deep melanomas and metastases compared to superficial melanomas. Decreased immunostaining of HAS2 in melanoma cells was significantly associated with several known unfavourable histopathologic prognostic markers like increased mitotic count, absence of tumor infiltrating lymphocytes and the nodular subtype. Furthermore, reduced HAS1 and HAS2 immunostaining in the melanoma cells was associated with increased recurrence of melanoma (p = 0.041 and p = 0.006, respectively) and shortened disease- specific survival (p = 0.013 and p = 0.001, respectively). Conclusions This study indicates that reduced expression of HAS1 and HAS2 is associated with melanoma progression and suggests that HAS1 and HAS2 have a prognostic significance in cutaneous melanoma. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2344-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mari Poukka
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
| | | | - Hanna Siiskonen
- Department of Dermatology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | | | - Päivi Auvinen
- Cancer Center, Kuopio University Hospital, Kuopio, Finland
| | - Sanna Pasonen-Seppänen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| | - Reijo Sironen
- Institute of Clinical Medicine/Clinical Pathology, University of Eastern Finland, Kuopio, Finland.,Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland.,Cancer Center of Eastern Finland, Kuopio, Finland
| |
Collapse
|
47
|
Reeves SR, Kaber G, Sheih A, Cheng G, Aronica MA, Merrilees MJ, Debley JS, Frevert CW, Ziegler SF, Wight TN. Subepithelial Accumulation of Versican in a Cockroach Antigen-Induced Murine Model of Allergic Asthma. J Histochem Cytochem 2016; 64:364-80. [PMID: 27126823 DOI: 10.1369/0022155416642989] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/12/2016] [Indexed: 01/13/2023] Open
Abstract
The extracellular matrix (ECM) is an important contributor to the asthmatic phenotype. Recent studies investigating airway inflammation have demonstrated an association between hyaluronan (HA) accumulation and inflammatory cell infiltration of the airways. The ECM proteoglycan versican interacts with HA and is important in the recruitment and activation of leukocytes during inflammation. We investigated the role of versican in the pathogenesis of asthmatic airway inflammation. Using cockroach antigen (CRA)-sensitized murine models of allergic asthma, we demonstrate increased subepithelial versican in the airways of CRA-treated mice that parallels subepithelial increases in HA and leukocyte infiltration. During the acute phase, CRA-treated mice displayed increased gene expression of the four major versican isoforms, as well as increased expression of HA synthases. Furthermore, in a murine model that examines both acute and chronic CRA exposure, versican staining peaked 8 days following CRA challenge and preceded subepithelial leukocyte infiltration. We also assessed versican and HA expression in differentiated primary human airway epithelial cells from asthmatic and healthy children. Increases in the expression of versican isoforms and HA synthases in these epithelial cells were similar to those of the murine model. These data indicate an important role for versican in the establishment of airway inflammation in asthma.
Collapse
Affiliation(s)
- Stephen R Reeves
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington (SRR, JSD),Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington (SRR, JSD)
| | - Gernot Kaber
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington (GK, TNW)
| | - Alyssa Sheih
- Immunology Program, Benaroya Research Institute, Seattle, Washington (AS, SFZ)
| | - Georgiana Cheng
- Department of Pathobiology, the Respiratory Institute, and Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio (GC, MAA)
| | - Mark A Aronica
- Department of Pathobiology, the Respiratory Institute, and Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio (GC, MAA)
| | - Mervyn J Merrilees
- Department of Anatomy and Medical Imaging, School of Medical Sciences, University of Auckland, Auckland, New Zealand (MJM)
| | - Jason S Debley
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington (SRR, JSD),Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington (SRR, JSD)
| | - Charles W Frevert
- Department of Comparative Medicine and Center for Lung Biology, University of Washington, Seattle, Washington (CWF)
| | - Steven F Ziegler
- Immunology Program, Benaroya Research Institute, Seattle, Washington (AS, SFZ)
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington (GK, TNW)
| |
Collapse
|
48
|
Maytin EV. Hyaluronan: More than just a wrinkle filler. Glycobiology 2016; 26:553-9. [PMID: 26964566 DOI: 10.1093/glycob/cww033] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/02/2016] [Indexed: 11/13/2022] Open
Abstract
Dermatology is a field that strives not only to alleviate skin disease (therapeutics) but also to improve the perception of wellness (cosmetics). Thus, in this special issue of Glycobiology, it seems appropriate to discuss the biology of a glycosaminoglycan, called hyaluronic acid (hyaluronan, or HA), that has become the most popular agent today for intradermal injections to improve wrinkles and other cosmetic defects. HA is a simple linear polymer in which a simple disaccharide is repeated thousands of time, thereby creating a huge hydrophilic molecule that confers a large volume of hydration and contributes to the turgor and flexibility of healthy skin. Beyond cosmetic considerations, however, HA also has important biological and physiological functions that were largely under-appreciated until recently. New research has confirmed that HA is dynamically produced by most skin cells, not only fibroblasts (the cells that make most of the skin's extracellular matrix) but also by keratinocytes in the outer protective layer (epidermis). For both fibroblasts and keratinocytes, HA plays a regulatory role in controlling cell physiology through interaction of extracellular HA with a major cell-surface receptor, CD44. This interaction mediates intracellular signaling both directly and indirectly, through CD44 interactions with the cytoskeleton and with EGF and TGFβ receptors. Furthermore, degradation of HA by specific hyaluronidase enzymes produces HA fragments that can help to regulate inflammatory processes. In this review, current knowledge about the role of HA in skin inflammation and wound healing are reviewed and possible future applications of such knowledge discussed.
Collapse
Affiliation(s)
- Edward V Maytin
- Department of Dermatology Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
49
|
Liang J, Jiang D, Noble PW. Hyaluronan as a therapeutic target in human diseases. Adv Drug Deliv Rev 2016; 97:186-203. [PMID: 26541745 PMCID: PMC4753080 DOI: 10.1016/j.addr.2015.10.017] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 02/07/2023]
Abstract
Accumulation and turnover of extracellular matrix is a hallmark of tissue injury, repair and remodeling in human diseases. Hyaluronan is a major component of the extracellular matrix and plays an important role in regulating tissue injury and repair, and controlling disease outcomes. The function of hyaluronan depends on its size, location, and interactions with binding partners. While fragmented hyaluronan stimulates the expression of an array of genes by a variety of cell types regulating inflammatory responses and tissue repair, cell surface hyaluronan provides protection against tissue damage from the environment and promotes regeneration and repair. The interactions of hyaluronan and its binding proteins participate in the pathogenesis of many human diseases. Thus, targeting hyaluronan and its interactions with cells and proteins may provide new approaches to developing therapeutics for inflammatory and fibrosing diseases. This review focuses on the role of hyaluronan in biological and pathological processes, and as a potential therapeutic target in human diseases.
Collapse
Affiliation(s)
- Jiurong Liang
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dianhua Jiang
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul W Noble
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
50
|
Hyaluronan-positive plasma membrane protrusions exist on mesothelial cells in vivo. Histochem Cell Biol 2016; 145:531-44. [DOI: 10.1007/s00418-016-1405-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2016] [Indexed: 11/27/2022]
|