Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.928
Filter
1.
Cell ; 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38942015

ABSTRACT

Cellular homeostasis is intricately influenced by stimuli from the microenvironment, including signaling molecules, metabolites, and pathogens. Functioning as a signaling hub within the cell, mitochondria integrate information from various intracellular compartments to regulate cellular signaling and metabolism. Multiple studies have shown that mitochondria may respond to various extracellular signaling events. However, it is less clear how changes in the extracellular matrix (ECM) can impact mitochondrial homeostasis to regulate animal physiology. We find that ECM remodeling alters mitochondrial homeostasis in an evolutionarily conserved manner. Mechanistically, ECM remodeling triggers a TGF-ß response to induce mitochondrial fission and the unfolded protein response of the mitochondria (UPRMT). At the organismal level, ECM remodeling promotes defense of animals against pathogens through enhanced mitochondrial stress responses. We postulate that this ECM-mitochondria crosstalk represents an ancient immune pathway, which detects infection- or mechanical-stress-induced ECM damage, thereby initiating adaptive mitochondria-based immune and metabolic responses.

2.
Cell ; 184(26): 6313-6325.e18, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34942099

ABSTRACT

How tissues acquire complex shapes is a fundamental question in biology and regenerative medicine. Zebrafish semicircular canals form from invaginations in the otic epithelium (buds) that extend and fuse to form the hubs of each canal. We find that conventional actomyosin-driven behaviors are not required. Instead, local secretion of hyaluronan, made by the enzymes uridine 5'-diphosphate dehydrogenase (ugdh) and hyaluronan synthase 3 (has3), drives canal morphogenesis. Charged hyaluronate polymers osmotically swell with water and generate isotropic extracellular pressure to deform the overlying epithelium into buds. The mechanical anisotropy needed to shape buds into tubes is conferred by a polarized distribution of actomyosin and E-cadherin-rich membrane tethers, which we term cytocinches. Most work on tissue morphogenesis ascribes actomyosin contractility as the driving force, while the extracellular matrix shapes tissues through differential stiffness. Our work inverts this expectation. Hyaluronate pressure shaped by anisotropic tissue stiffness may be a widespread mechanism for powering morphological change in organogenesis and tissue engineering.


Subject(s)
Extracellular Space/chemistry , Hyaluronic Acid/pharmacology , Morphogenesis , Organ Specificity , Pressure , Semicircular Canals/cytology , Semicircular Canals/embryology , Actomyosin/metabolism , Animals , Anisotropy , Behavior, Animal , Extracellular Matrix/metabolism , Hyaluronic Acid/biosynthesis , Models, Biological , Morphogenesis/drug effects , Organ Specificity/drug effects , Osmotic Pressure , Semicircular Canals/diagnostic imaging , Stereotyped Behavior , Zebrafish/embryology , Zebrafish Proteins/metabolism
3.
Cell ; 172(1-2): 305-317.e10, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29328918

ABSTRACT

Phagocytic receptors must diffuse laterally to become activated upon clustering by multivalent targets. Receptor diffusion, however, can be obstructed by transmembrane proteins ("pickets") that are immobilized by interacting with the cortical cytoskeleton. The molecular identity of these pickets and their role in phagocytosis have not been defined. We used single-molecule tracking to study the interaction between Fcγ receptors and CD44, an abundant transmembrane protein capable of indirect association with F-actin, hence likely to serve as a picket. CD44 tethers reversibly to formin-induced actin filaments, curtailing receptor diffusion. Such linear filaments predominate in the trailing end of polarized macrophages, where receptor mobility was minimal. Conversely, receptors were most mobile at the leading edge, where Arp2/3-driven actin branching predominates. CD44 binds hyaluronan, anchoring a pericellular coat that also limits receptor displacement and obstructs access to phagocytic targets. Force must be applied to traverse the pericellular barrier, enabling receptors to engage their targets.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Membrane/metabolism , Hyaluronan Receptors/metabolism , Receptors, Immunologic/metabolism , Adult , Animals , Binding Sites , COS Cells , Cells, Cultured , Chlorocebus aethiops , Female , Humans , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/genetics , Hyaluronic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding
4.
Immunity ; 49(2): 326-341.e7, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30054204

ABSTRACT

The maintenance of appropriate arterial tone is critically important for normal physiological arterial function. However, the cellular and molecular mechanisms remain poorly defined. Here, we have shown that in the mouse aorta, resident macrophages prevented arterial stiffness and collagen deposition in the steady state. Using phenotyping, transcriptional profiling, and targeted deletion of Csf1r, we have demonstrated that these macrophages-which are a feature of blood vessels invested with smooth muscle cells (SMCs) in both mouse and human tissues-expressed the hyaluronan (HA) receptor LYVE-l. Furthermore, we have shown they possessed the unique ability to modulate collagen expression in SMCs by matrix metalloproteinase MMP-9-dependent proteolysis through engagement of LYVE-1 with the HA pericellular matrix of SMCs. Our study has unveiled a hitherto unknown homeostatic contribution of arterial LYVE-1+ macrophages through the control of collagen production by SMCs and has identified a function of LYVE-1 in leukocytes.


Subject(s)
Collagen/metabolism , Glycoproteins/metabolism , Hyaluronan Receptors/metabolism , Macrophages/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Vascular Stiffness/physiology , Animals , Aorta/physiology , Female , Glycoproteins/genetics , Humans , Hyaluronic Acid/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
5.
J Biol Chem ; 300(7): 107449, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38844132

ABSTRACT

Hyaluronan (HA) is a high-molecular-weight (HMW) glycosaminoglycan, which is a fundamental component of the extracellular matrix that is involved in a variety of biological processes. We previously showed that the HYBID/KIAA1199/CEMIP axis plays a key role in the depolymerization of HMW-HA in normal human dermal fibroblasts (NHDFs). However, its roles in normal human epidermal keratinocytes (NHEKs) remained unclear. HYBID mRNA expression in NHEKs was lower than that in NHDFs, and NHEKs showed no depolymerization of extracellular HMW-HA in culture, indicating that HYBID does not contribute to extracellular HA degradation. In this study, we found that the cell-free conditioned medium of NHEKs degraded HMW-HA under weakly acidic conditions (pH 4.8). This degrading activity was abolished by hyaluronidase 1 (HYAL1) knockdown but not by HYAL2 knockdown. Newly synthesized HYAL1 was mainly secreted extracellularly, and the secretion of HYAL1 was increased during differentiation, suggesting that epidermal interspace HA is physiologically degraded by HYAL1 according to pH decrease during stratum corneum formation. In HA synthesis, hyaluronan synthase 3 (HAS3) knockdown reduced HA production by NHEKs, and interferon-γ-dependent HA synthesis was correlated with increased HAS3 expression. Furthermore, HA production was increased by TMEM2 knockdown through enhanced HAS3 expression. These results indicate that NHEKs regulate HA metabolism via HYAL1 and HAS3, and TMEM2 is a regulator of HAS3-dependent HA production.

6.
Annu Rev Biomed Eng ; 26(1): 25-47, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38166186

ABSTRACT

Hyaluronan (HA) plays well-recognized mechanical and biological roles in articular cartilage and synovial fluid, where it contributes to tissue structure and lubrication. An understanding of how HA contributes to the structure of other musculoskeletal tissues, including muscle, bone, tendon, and intervertebral discs, is growing. In addition, the use of HA-based therapies to restore damaged tissue is becoming more prevalent. Nevertheless, the relationship between biomechanical stimuli and HA synthesis, degradation, and signaling in musculoskeletal tissues remains understudied, limiting the utility of HA in regenerative medicine. In this review, we discuss the various roles and significance of endogenous HA in musculoskeletal tissues. We use what is known and unknown to motivate new lines of inquiry into HA biology within musculoskeletal tissues and in the mechanobiology governing HA metabolism by suggesting questions that remain regarding the relationship and interaction between biological and mechanical roles of HA in musculoskeletal health and disease.


Subject(s)
Hyaluronic Acid , Tendons , Hyaluronic Acid/chemistry , Humans , Animals , Biomechanical Phenomena , Tendons/physiology , Tendons/metabolism , Cartilage, Articular/physiology , Cartilage, Articular/metabolism , Signal Transduction , Bone and Bones/metabolism , Bone and Bones/physiology , Synovial Fluid/metabolism , Synovial Fluid/physiology , Muscle, Skeletal/physiology , Muscle, Skeletal/metabolism , Musculoskeletal System/metabolism , Regenerative Medicine/methods
7.
Semin Cell Dev Biol ; 131: 134-145, 2022 11.
Article in English | MEDLINE | ID: mdl-35534334

ABSTRACT

Morphogenesis, the process by which tissues develop into functional shapes, requires coordinated mechanical forces. Most current literature ascribes contractile forces derived from actomyosin networks as the major driver of tissue morphogenesis. Recent works from diverse species have shown that pressure derived from fluids can generate deformations necessary for tissue morphogenesis. In this review, we discuss how hydrostatic pressure is generated at the cellular and tissue level and how the pressure can cause deformations. We highlight and review findings demonstrating the mechanical roles of pressures from fluid-filled lumens and viscous gel-like components of the extracellular matrix. We also emphasise the interactions and mechanochemical feedbacks between extracellular pressures and tissue behaviour in driving tissue remodelling. Lastly, we offer perspectives on the open questions in the field that will further our understanding to uncover new principles of tissue organisation during development.


Subject(s)
Actomyosin , Hydrostatic Pressure , Morphogenesis , Viscosity
8.
J Biol Chem ; 299(9): 105120, 2023 09.
Article in English | MEDLINE | ID: mdl-37527776

ABSTRACT

Transmembrane protein 2 (TMEM2) was originally identified as a membrane-anchored protein of unknown function. We previously demonstrated that TMEM2 can degrade hyaluronan (HA). Furthermore, we showed that induced global knockout of Tmem2 in adult mice results in rapid accumulation of incompletely degraded HA in bodily fluids and organs, supporting the identity of TMEM2 as a cell surface hyaluronidase. In spite of these advances, no direct evidence has been presented to demonstrate the intrinsic hyaluronidase activity of TMEM2. Here, we directly establish the catalytic activity of TMEM2. The ectodomain of TMEM2 (TMEM2ECD) was expressed as a His-tagged soluble protein and purified by affinity and size-exclusion chromatography. Both human and mouse TMEM2ECD robustly degrade fluorescein-labeled HA into 5 to 10 kDa fragments. TMEM2ECD exhibits this HA-degrading activity irrespective of the species of TMEM2 origin and the position of epitope tag insertion. The HA-degrading activity of TMEM2ECD is more potent than that of HYAL2, a hyaluronidase which, like TMEM2, has been implicated in cell surface HA degradation. Finally, we show that TMEM2ECD can degrade not only fluorescein-labeled HA but also native high-molecular weight HA. In addition to these core findings, our study reveals hitherto unrecognized confounding factors, such as the quality of reagents and the choice of assay systems, that could lead to erroneous conclusions regarding the catalytic activity of TMEM2. In conclusion, our results demonstrate that TMEM2 is a legitimate functional hyaluronidase. Our findings also raise cautions regarding the choice of reagents and methods for performing degradation assays for hyaluronidases.


Subject(s)
Hyaluronoglucosaminidase , Membrane Proteins , Animals , Humans , Mice , Cell Membrane/metabolism , Fluoresceins , Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism
9.
J Biol Chem ; 299(4): 103074, 2023 04.
Article in English | MEDLINE | ID: mdl-36858200

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.


Subject(s)
Calreticulin , Cell Division , G1 Phase , Glucose , Heparin , Hyperglycemia , Mesangial Cells , Resting Phase, Cell Cycle , Animals , Humans , Rats , Calreticulin/metabolism , Cells, Cultured , Glomerular Mesangium/metabolism , Glucose/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Heparin/pharmacology , Heparin/metabolism , Hyaluronic Acid/metabolism , Mesangial Cells/cytology , Mesangial Cells/metabolism , Hyperglycemia/metabolism
10.
J Biol Chem ; 299(8): 104995, 2023 08.
Article in English | MEDLINE | ID: mdl-37394007

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.


Subject(s)
Diabetes Mellitus , Hyperglycemia , Lung Injury , Humans , Diabetes Mellitus/metabolism , Extracellular Matrix/metabolism , Glucose/pharmacology , Glucose/metabolism , Heparin/pharmacology , Heparin/metabolism , Hyaluronic Acid/metabolism , Hyperglycemia/metabolism , Lung Injury/metabolism , Monocytes/metabolism , Animals , Mice , Mice, Inbred BALB C
11.
J Biol Chem ; 299(6): 104826, 2023 06.
Article in English | MEDLINE | ID: mdl-37196767

ABSTRACT

Cutaneous hyaluronan (HA) is depolymerized to intermediate sizes in the extracellular matrix, and further fragmented in the regional lymph nodes. Previously, we showed that the HA-binding protein involved in HA depolymerization (HYBID), also known as KIAA1199/CEMIP, is responsible for the first step of HA depolymerization. Recently, mouse transmembrane 2 (mTMEM2) with high structural similarity to HYBID was proposed to be a membrane-bound hyaluronidase. However, we showed that the knockdown of human TMEM2 (hTMEM2) conversely promoted HA depolymerization in normal human dermal fibroblasts (NHDFs). Therefore, we examined the HA-degrading activity and function of hTMEM2 using HEK293T cells. We found that human HYBID and mTMEM2, but not hTMEM2, degraded extracellular HA, indicating that hTMEM2 does not function as a catalytic hyaluronidase. Analysis of the HA-degrading activity of chimeric TMEM2 in HEK293T cells suggested the importance of the mouse GG domain. Therefore, we focused on the amino acid residues that are conserved in active mouse and human HYBID and mTMEM2 but are substituted in hTMEM2. The HA-degrading activity of mTMEM2 was abolished when its His248 and Ala303 were simultaneously replaced by the corresponding residues of inactive hTMEM2 (Asn248 and Phe303). In NHDFs, enhancement of hTMEM2 expression by proinflammatory cytokines decreased HYBID expression and increased hyaluronan synthase 2-dependent HA production. The effects of proinflammatory cytokines were abrogated by hTMEM2 knockdown. A decreased HYBID expression by interleukin-1ß and transforming growth factor-ß was canceled by hTMEM2 knockdown. In conclusion, these results indicate that hTMEM2 is not a catalytic hyaluronidase, but a regulator of HA metabolism.


Subject(s)
Hyaluronic Acid , Hyaluronoglucosaminidase , Animals , Humans , Mice , Cytokines , HEK293 Cells , Hyaluronan Synthases/genetics , Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/metabolism
12.
Infect Immun ; 92(7): e0019924, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38842305

ABSTRACT

Enterococcus faecalis is a common cause of healthcare-acquired bloodstream infections and catheter-associated urinary tract infections (CAUTIs) in both adults and children. Treatment of E. faecalis infection is frequently complicated by multi-drug resistance. Based on protein homology, E. faecalis encodes two putative hyaluronidases, EF3023 (HylA) and EF0818 (HylB). In other Gram-positive pathogens, hyaluronidases have been shown to contribute to tissue damage and immune evasion, but the function in E. faecalis has yet to be explored. Here, we show that both hylA and hylB contribute to E. faecalis pathogenesis. In a CAUTI model, ΔhylA exhibited defects in bladder colonization and dissemination to the bloodstream, and ΔhylB exhibited a defect in kidney colonization. Furthermore, a ΔhylAΔhylB double mutant exhibited a severe colonization defect in a model of bacteremia while the single mutants colonized to a similar level as the wild-type strain, suggesting potential functional redundancy within the bloodstream. We next examined enzymatic activity, and demonstrate that HylB is capable of digesting both hyaluronic acid (HA) and chondroitin sulfate in vitro, while HylA exhibits only a very modest activity against heparin. Importantly, HA degradation by HylB provided a modest increase in cell density during the stationary phase and also contributed to dampening of lipopolysaccharide-mediated NF-κB activation. Overall, these data demonstrate that glycosaminoglycan degradation is important for E. faecalis pathogenesis in the urinary tract and during bloodstream infection.


Subject(s)
Bacteremia , Catheter-Related Infections , Enterococcus faecalis , Glycosaminoglycans , Gram-Positive Bacterial Infections , Urinary Tract Infections , Enterococcus faecalis/genetics , Enterococcus faecalis/enzymology , Enterococcus faecalis/metabolism , Urinary Tract Infections/microbiology , Bacteremia/microbiology , Catheter-Related Infections/microbiology , Animals , Gram-Positive Bacterial Infections/microbiology , Mice , Glycosaminoglycans/metabolism , Hyaluronoglucosaminidase/metabolism , Hyaluronoglucosaminidase/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Female , Humans , Hyaluronic Acid/metabolism
13.
Lab Invest ; : 102104, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38945481

ABSTRACT

The glycosaminoglycan hyaluronan (HA) plays an important role in tumor progression. However, its biological and clinical significance in papillary thyroid cancer (PTC) remains unknown. Immunohistochemistry was performed to examine HA expression in tissues from PTC patients. Two PTC cell lines were treated with HA synthesized inhibitor against HA production to assess its function. Serum HA levels from 107 PTC patients, 30 Hashimoto thyroiditis, and 45 normal controls (NC) were measured by chemiluminescence immunoassay. HA levels in FNA washouts obtained from thyroid nodules and lymph nodes (LNs) were measured by chemiluminescence immunoassay. Area under the curve (AUC) were computed to evaluate HA`s clinical value. HA was highly expressed in PTC. Reducing HA production significantly inhibited PTC cell proliferation and invasion. Importantly, serum HA levels in PTC were significantly higher than in NCs and Hashimoto thyroiditis and allowed distinguishing of thyroid cancers from NCs with high accuracy (AUC=0.782). Moreover, elevated serum HA levels in PTC correlate with LN metastasis. HA levels in fine needle aspiration (FNA) washouts from PTC patients were significantly higher than in benign controls, with a high AUC value (0.8644) for distinguishing PTC from benign controls. Furthermore, HA levels in FNA washouts from metastatic LN were significantly higher than in non-metastatic LN, with a high AUC value (0.8007) for distinguishing metastatic LNs from non-metastatic LNs. HA in serum and FNA washout exhibited a potential significance for PTC diagnosis and indicator for LN metastasis in patients with PTC.

14.
Mol Pain ; 20: 17448069241254455, 2024.
Article in English | MEDLINE | ID: mdl-38728068

ABSTRACT

Pruritis, the sensation of itch, is produced by multiple substances, exogenous and endogenous, that sensitizes specialized sensory neurons (pruriceptors and pruri-nociceptors). Unfortunately, many patients with acute and chronic pruritis obtain only partial relief when treated with currently available treatment modalities. We recently demonstrated that the topical application of high molecular weight hyaluronan (HMWH), when combined with vehicles containing transdermal transport enhancers, produce potent long-lasting reversal of nociceptor sensitization associated with inflammatory and neuropathic pain. In the present experiments we tested the hypothesis that the topical formulation of HMWH with protamine, a transdermal transport enhancer, can also attenuate pruritis. We report that this topical formulation of HMWH markedly attenuates scratching behavior at the nape of the neck induced by serotonin (5-hydroxytryptamine, 5-HT), in male and female rats. Our results support the hypothesis that topical HMWH in a transdermal transport enhancer vehicle is a strong anti-pruritic.


Subject(s)
Administration, Cutaneous , Hyaluronic Acid , Protamines , Rats, Sprague-Dawley , Animals , Hyaluronic Acid/pharmacology , Hyaluronic Acid/chemistry , Male , Female , Rats , Protamines/pharmacology , Molecular Weight , Serotonin/metabolism , Administration, Topical
15.
Biochem Biophys Res Commun ; 724: 150234, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38865812

ABSTRACT

Vasculature-on-chip (VoC) models have become a prominent tool in the study of microvasculature functions because of their cost-effective and ethical production process. These models typically use a hydrogel in which the three-dimensional (3D) microvascular structure is embedded. Thus, VoCs are directly impacted by the physical and chemical cues of the supporting hydrogel. Endothelial cell (EC) response in VoCs is critical, especially in organ-specific vasculature models, in which ECs exhibit specific traits and behaviors that vary between organs. Many studies customize the stimuli ECs perceive in different ways; however, customizing the hydrogel composition accordingly to the target organ's extracellular matrix (ECM), which we believe has great potential, has been rarely investigated. We explored this approach to organ-specific VoCs by fabricating microvessels (MVs) with either human umbilical vein ECs or human brain microvascular ECs in a 3D cylindrical VoC using a collagen hydrogel alone or one supplemented with laminin and hyaluronan, components found in the brain ECM. We characterized the physical properties of these hydrogels and analyzed the barrier properties of the MVs. Barrier function and tight junction (ZO-1) expression improved with the addition of laminin and hyaluronan in the composite hydrogel.


Subject(s)
Collagen , Human Umbilical Vein Endothelial Cells , Hyaluronic Acid , Hydrogels , Laminin , Microvessels , Tight Junctions , Humans , Hydrogels/chemistry , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Laminin/chemistry , Laminin/metabolism , Collagen/chemistry , Collagen/metabolism , Microvessels/metabolism , Microvessels/drug effects , Tight Junctions/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Lab-On-A-Chip Devices , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Cells, Cultured
16.
Development ; 148(3)2021 02 03.
Article in English | MEDLINE | ID: mdl-33462113

ABSTRACT

Macrophages are components of the innate immune system with key roles in tissue inflammation and repair. It is now evident that macrophages also support organogenesis, but few studies have characterized their identity, ontogeny and function during heart development. Here, we show that the distribution and prevalence of resident macrophages in the subepicardial compartment of the developing heart coincides with the emergence of new lymphatics, and that macrophages interact closely with the nascent lymphatic capillaries. Consequently, global macrophage deficiency led to extensive vessel disruption, with mutant hearts exhibiting shortened and mis-patterned lymphatics. The origin of cardiac macrophages was linked to the yolk sac and foetal liver. Moreover, the Cx3cr1+ myeloid lineage was found to play essential functions in the remodelling of the lymphatic endothelium. Mechanistically, macrophage hyaluronan was required for lymphatic sprouting by mediating direct macrophage-lymphatic endothelial cell interactions. Together, these findings reveal insight into the role of macrophages as indispensable mediators of lymphatic growth during the development of the mammalian cardiac vasculature.


Subject(s)
Heart/growth & development , Lymphatic Vessels , Macrophages/metabolism , Animals , CX3C Chemokine Receptor 1/genetics , Cell Adhesion , Cell Line , Endothelial Cells , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Humans , Inflammation , Lymphangiogenesis , Macrophages/immunology , Mice , Mice, Inbred C57BL , Organogenesis/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Yolk Sac
17.
Small ; 20(21): e2306207, 2024 May.
Article in English | MEDLINE | ID: mdl-38161247

ABSTRACT

Synovial fluid (SF) is the complex biofluid that facilitates the exceptional lubrication of articular cartilage in joints. Its primary lubricating macromolecules, the linear polysaccharide hyaluronic acid (HA) and the mucin-like glycoprotein proteoglycan 4 (PRG4 or lubricin), interact synergistically to reduce boundary friction. However, the precise manner in which these molecules influence the rheological properties of SF remains unclear. This study aimed to elucidate this by employing confocal microscopy and multiscale rheometry to examine the microstructure and rheology of solutions containing recombinant human PRG4 (rhPRG4) and HA. Contrary to previous assumptions of an extensive HA-rhPRG4 network, it is discovered that rhPRG4 primarily forms stiff, gel-like aggregates. The properties of these aggregates, including their size and stiffness, are found to be influenced by the viscoelastic characteristics of the surrounding HA matrix. Consequently, the rheology of this system is not governed by a single length scale, but instead responds as a disordered, hierarchical network with solid-like rhPRG4 aggregates distributed throughout the continuous HA phase. These findings provide new insights into the biomechanical function of PRG4 in cartilage lubrication and may have implications in the development of HA-based therapies for joint diseases like osteoarthritis.


Subject(s)
Hyaluronic Acid , Proteoglycans , Rheology , Synovial Fluid , Synovial Fluid/metabolism , Synovial Fluid/chemistry , Humans , Hyaluronic Acid/chemistry , Proteoglycans/chemistry , Proteoglycans/metabolism , Lubrication , Macromolecular Substances/chemistry , Viscosity
18.
J Neurosci Res ; 102(5): e25356, 2024 May.
Article in English | MEDLINE | ID: mdl-38773875

ABSTRACT

From the blood brain barrier to the synaptic space, astrocytes provide structural, metabolic, ionic, and extracellular matrix (ECM) support across the brain. Astrocytes include a vast array of subtypes, their phenotypes and functions varying both regionally and temporally. Astrocytes' metabolic and regulatory functions poise them to be quick and sensitive responders to injury and disease in the brain as revealed by single cell sequencing. Far less is known about the influence of the local healthy and aging microenvironments on these astrocyte activation states. In this forward-looking review, we describe the known relationship between astrocytes and their local microenvironment, the remodeling of the microenvironment during disease and injury, and postulate how they may drive astrocyte activation. We suggest technology development to better understand the dynamic diversity of astrocyte activation states, and how basal and activation states depend on the ECM microenvironment. A deeper understanding of astrocyte response to stimuli in ECM-specific contexts (brain region, age, and sex of individual), paves the way to revolutionize how the field considers astrocyte-ECM interactions in brain injury and disease and opens routes to return astrocytes to a healthy quiescent state.


Subject(s)
Astrocytes , Brain , Extracellular Matrix , Astrocytes/physiology , Astrocytes/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Humans , Animals , Brain/metabolism , Brain Injuries/pathology , Brain Injuries/metabolism
19.
J Med Virol ; 96(2): e29457, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38318772

ABSTRACT

Lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1) serves as a prominent marker for lymphatic endothelial cells (LECs) and is pivotal in the process of lymphangiogenesis, a critical factor in cancer development and metastasis. Overexpression of LYVE-1 has been observed in various cancers, where it is recognized as an adverse prognostic indicator. Targeting LYVE-1 has demonstrated inhibitory effects on tumor cell proliferation, migration, and the formation of lymph node metastases both in vitro and in vivo. While extensive research has focused on the role of LYVE-1 in cancer cells, its involvement in virus infection and associated diseases remains largely unexplored. This review consolidates recent findings regarding the expression of LYVE-1 and its functions in lymphangiogenesis during various viral infections and the development of related diseases, with a particular emphasis on Kaposi's sarcoma herpesvirus. Despite the limited available data, it is evident that further studies are essential to comprehensively understand the contribution of LYVE-1 to viral pathogenesis and oncogenesis.


Subject(s)
Neoplasms , Virus Diseases , Humans , Endothelial Cells/pathology , Hyaluronan Receptors/metabolism , Endothelium, Lymphatic/metabolism , Neoplasms/pathology , Virus Diseases/pathology
20.
Chemistry ; : e202401028, 2024 May 26.
Article in English | MEDLINE | ID: mdl-38797703

ABSTRACT

Cationic Mn(III)-meso-tetraarylporphyrin derivatives, substituted in para position with different size alkyl chains, were investigated to function as antioxidants in free-radical degradation of high-molar-mass hyaluronan by the methods of rotational viscometry and oximetry. The results of rotational viscometry showed that MnTM-4-PyP5+, MnTE-4-PyP5+, MnTPr-4-PyP5+, MnTPen-4-PyP5+ and MnTHep-4-PyP5+ showed high efficiency in decomposing H2O2, and reducing of peroxidized hyaluronan. When using oxygen electrode, MnTE-4-PyP5+, MnTPr-4-PyP5+, MnTPen-4-PyP5+, and MnTHep-4-PyP5+ applied to function as protective antioxidants in hyaluronan degradation, the uptake of dissolved oxygen from the reaction milieu was rapid, followed by continual increase in oxygen concentration up to the end of the measurement. However, when especially MnTE-4-PyP5+, MnTPr-4-PyP5+, and MnTPen-4-PyP5+ were examined as hyaluronan chain-breaking antioxidants, after short-term dissolved oxygen uptake, almost no increase in oxygen concentration was shown.

SELECTION OF CITATIONS
SEARCH DETAIL