Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
2.
Wound Repair Regen ; 22(4): 521-6, 2014.
Article in English | MEDLINE | ID: mdl-25041621

ABSTRACT

Age-related changes in the extracellular matrix contribute to delayed wound repair in aging. Hyaluronan, a linear nonsulfated glycosaminoglycan, promotes synthesis and assembly of key extracellular matrix components, such as the interstitial collagens, during wound healing. The biological effects of hyaluronan are mediated, in part, by hyaluronan size. We have previously determined that dermal wounds in aged mice, relative to young mice, have deficits in the generation of lower molecular weight hyaluronan (defined as <300 kDa). Here, we tested the effect of exogenous hyaluronan of 2, 250, or 1,000 kDa sizes on full-thickness excisional wounds in aged mice. Only wounds treated with 250 kDa hyaluronan (HA250) were significantly improved over wounds that received carrier (water) alone. Treatment with HA250 was associated with increased expression of transcripts for the hyaluronan receptors CD44 and RHAMM, as well as collagens III and I. Analyses of dermal protein content by mass spectrometry and Western blotting confirmed significantly increased expression of collagen III in wounds treated with HA250 relative to control wounds. In summary, we find that HA250 improves wound repair and increases the synthesis of collagen III in aged dermal wounds.


Subject(s)
Collagen Type III/drug effects , Collagen Type III/metabolism , Hyaluronic Acid/pharmacology , Soft Tissue Injuries/metabolism , Wound Healing/drug effects , Aging/metabolism , Animals , Blotting, Western , Dermis/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Hyaluronan Receptors/drug effects , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Soft Tissue Injuries/drug therapy
3.
Proc Natl Acad Sci U S A ; 108(19): 7938-43, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21518860

ABSTRACT

We describe a role for ECM as a biosensor for inflammatory microenvironments that plays a critical role in peripheral immune tolerance. We show that hyaluronan (HA) promotes induction of Foxp3- IL-10-producing regulatory T cells (TR1) from conventional T-cell precursors in both murine and human systems. This is, to our knowledge, the first description of an ECM component inducing regulatory T cells. Intact HA, characteristic of healing tissues, promotes induction of TR1 capable of abrogating disease in an IL-10-dependent mouse colitis model whereas fragmentary HA, typical of inflamed tissues, does not, indicating a decisive role for tissue integrity in this system. The TR1 precursor cells in this system are CD4(+)CD62L(-)FoxP3(-), suggesting that effector memory cells assume a regulatory phenotype when they encounter their cognate antigen in the context of intact HA. Matrix integrity cues might thereby play a central role in maintaining peripheral tolerance. This TR1 induction is mediated by CD44 cross-linking and signaling through p38 and ERK1/2. This induction is suppressed, also in a CD44-dependent manner, by osteopontin, a component of chronically inflamed ECM, indicating that CD44 signaling serves as a nexus for fate decisions regarding TR1 induction. Finally, we demonstrate that TR1 induction signals can be recapitulated using synthetic matrices. These results reveal important roles for the matrix microenvironment in immune regulation and suggest unique strategies for immunomodulation.


Subject(s)
Extracellular Matrix/immunology , Interleukin-10/biosynthesis , Precursor Cells, T-Lymphoid/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Forkhead Transcription Factors/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Hyaluronan Receptors/immunology , Hyaluronic Acid/immunology , Immunologic Memory , In Vitro Techniques , Interleukin-2/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Osteopontin/immunology , T-Lymphocyte Subsets/immunology
4.
JCI Insight ; 7(12)2022 06 22.
Article in English | MEDLINE | ID: mdl-35730564

ABSTRACT

Thick, viscous respiratory secretions are a major pathogenic feature of COVID-19, but the composition and physical properties of these secretions are poorly understood. We characterized the composition and rheological properties (i.e., resistance to flow) of respiratory secretions collected from intubated COVID-19 patients. We found the percentages of solids and protein content were greatly elevated in COVID-19 compared with heathy control samples and closely resembled levels seen in cystic fibrosis, a genetic disease known for thick, tenacious respiratory secretions. DNA and hyaluronan (HA) were major components of respiratory secretions in COVID-19 and were likewise abundant in cadaveric lung tissues from these patients. COVID-19 secretions exhibited heterogeneous rheological behaviors, with thicker samples showing increased sensitivity to DNase and hyaluronidase treatment. In histologic sections from these same patients, we observed increased accumulation of HA and the hyaladherin versican but reduced tumor necrosis factor-stimulated gene-6 staining, consistent with the inflammatory nature of these secretions. Finally, we observed diminished type I interferon and enhanced inflammatory cytokines in these secretions. Overall, our studies indicated that increases in HA and DNA in COVID-19 respiratory secretion samples correlated with enhanced inflammatory burden and suggested that DNA and HA may be viable therapeutic targets in COVID-19 infection.


Subject(s)
COVID-19 , Interferon Type I , Humans , Lung , SARS-CoV-2 , Sputum
5.
medRxiv ; 2022 Apr 04.
Article in English | MEDLINE | ID: mdl-35411348

ABSTRACT

Thick, viscous respiratory secretions are a major pathogenic feature of COVID-19 disease, but the composition and physical properties of these secretions are poorly understood. We characterized the composition and rheological properties (i.e. resistance to flow) of respiratory secretions collected from intubated COVID-19 patients. We find the percent solids and protein content are greatly elevated in COVID-19 compared to heathy control samples and closely resemble levels seen in cystic fibrosis, a genetic disease known for thick, tenacious respiratory secretions. DNA and hyaluronan (HA) are major components of respiratory secretions in COVID-19 and are likewise abundant in cadaveric lung tissues from these patients. COVID-19 secretions exhibit heterogeneous rheological behaviors with thicker samples showing increased sensitivity to DNase and hyaluronidase treatment. In histologic sections from these same patients, we observe increased accumulation of HA and the hyaladherin versican but reduced tumor necrosis factorâ€"stimulated gene-6 (TSG6) staining, consistent with the inflammatory nature of these secretions. Finally, we observed diminished type I interferon and enhanced inflammatory cytokines in these secretions. Overall, our studies indicate that increases in HA and DNA in COVID-19 respiratory secretion samples correlate with enhanced inflammatory burden and suggest that DNA and HA may be viable therapeutic targets in COVID-19 infection.

6.
J Histochem Cytochem ; 69(6): 415-428, 2021 06.
Article in English | MEDLINE | ID: mdl-34080894

ABSTRACT

Although many studies have focused on a role for hyaluronan (HA) of interstitial extracellular matrix (presumably produced by non-vascular "stromal" cells) in regulating vascular growth, we herein examine the influence of "autocrine HA" produced by vascular endothelial cells themselves on tubulogenesis, using human umbilical vein endothelial cells (HUVECs) in angiogenic and vasculogenic three-dimensional collagen gel cultures. Relative to unstimulated controls, tubulogenic HUVECs upregulated HAS2 mRNA and increased the synthesis of cell-associated HA (but not HA secreted into media). Confocal microscopy/immunofluorescence on cultures fixed with neutral-buffered 10% formalin (NBF) revealed cytoplasmic HAS2 in HUVEC cords and tubes. Cultures fixed with NBF (with cetylpyridinium chloride added to retain HA), stained for HA using "affinity fluorescence" (biotinylated HA-binding protein with streptavidin-fluor), and viewed by confocal microscopy showed HA throughout tube lumens, but little/no HA on the abluminal sides of the tubes or in the surrounding collagen gel. Lumen formation in angiogenic and vasculogenic cultures was strongly suppressed by metabolic inhibitors of HA synthesis (mannose and 4-methylumbelliferone). Hyaluronidase strongly inhibited lumen formation in angiogenic cultures, but not in vasculogenic cultures (where developing lumens are not open to culture medium). Collectively, our results point to a role for autocrine, luminal HA in microvascular sprouting and lumen development. (J Histochem Cytochem 69: 415-428, 2021).


Subject(s)
Endothelial Cells/metabolism , Hyaluronic Acid/metabolism , Neovascularization, Physiologic , Cell Culture Techniques , Collagen/metabolism , Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells , Humans , Hyaluronan Synthases/genetics , Hyaluronan Synthases/metabolism , Up-Regulation
7.
Matrix Biol Plus ; 9: 100052, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33718858

ABSTRACT

The extracellular matrix glycosaminoglycan hyaluronan (HA) accumulates in human and mouse islets during the onset of autoimmune type 1 diabetes (T1D). HA plays a critical role in T1D pathogenesis, as spontaneous disease is blocked in mice fed the HA synthesis inhibitor 4-methylumbelliferone (4MU). The present study demonstrates the involvement of HA in T cell-mediated autoimmune responses to transplanted islets and in in vivo and in vitro T cell activation. Scaffolded islet implants (SIs) loaded with RIP-mOVA mouse islets expressing chicken ovalbumin (OVA) on their ß cells were grafted into T and B cell-deficient RIP-mOVA mice, which subsequently received CD4+ T cells from DO11.10 transgenic mice bearing OVA peptide-specific T cell receptors (TcRs), followed by injection of OVA peptide to induce an immune response to the OVA-expressing islets. By affinity histochemistry (AHC), HA was greatly increased in grafted islets with T cell infiltrates (compared to islets grafted into mice lacking T cells) and a portion of this HA co-localized with the infiltrating T cells. Transferred T cells underwent HA synthase (HAS) isoform switching - T cells isolated from the SI grafts strongly upregulated HAS1 and HAS2 mRNAs and downregulated HAS3 mRNA, in contrast to T cells from graft-draining mesenteric lymph nodes, which expressed HAS3 mRNA only. Expression of HAS1 and HAS2 proteins by T cells in SI infiltrates was confirmed by immunohistochemistry (IHC). DO11.10 mice fed 4MU had suppressed in vivo T cell immune priming (measured as a reduced recall response to OVA peptide) compared to T cells from control mice fed a normal diet. In co-cultures of naïve DO11.10 T cells and OVA peptide-loaded antigen-presenting cells (APCs), pre-exposure of the T cells (but not pre-exposure of APCs) to 4MU inhibited early T cell activation (CD69 expression). In addition, T cells exposed to 4MU during activation in vitro with anti-CD3/CD28 antibodies had inhibited phosphorylation of the CD3ζ subunit of the TcR, a very early event in TcR signaling. Collectively, our results demonstrate that T cell-derived HA plays a significant role in T cell immune responses, and that expression of T cell HAS isoforms changes in a locale-specific manner during in vivo priming and functional phases of the T cell response.

8.
Matrix Biol ; 96: 69-86, 2021 02.
Article in English | MEDLINE | ID: mdl-33290836

ABSTRACT

A coat of pericellular hyaluronan surrounds mature dendritic cells (DC) and contributes to cell-cell interactions. We asked whether 4-methylumbelliferone (4MU), an oral inhibitor of HA synthesis, could inhibit antigen presentation. We find that 4MU treatment reduces pericellular hyaluronan, destabilizes interactions between DC and T-cells, and prevents T-cell proliferation in vitro and in vivo. These effects were observed only when 4MU was added prior to initial antigen presentation but not later, consistent with 4MU-mediated inhibition of de novo antigenic responses. Building on these findings, we find that 4MU delays rejection of allogeneic pancreatic islet transplant and allogeneic cardiac transplants in mice and suppresses allogeneic T-cell activation in human mixed lymphocyte reactions. We conclude that 4MU, an approved drug, may have benefit as an adjunctive agent to delay transplantation rejection.


Subject(s)
Dendritic Cells/cytology , Graft Rejection/prevention & control , Hyaluronic Acid/biosynthesis , Hymecromone/administration & dosage , T-Lymphocytes, Regulatory/cytology , Animals , Antigen Presentation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Disease Models, Animal , Graft Rejection/immunology , Heart Transplantation/adverse effects , Humans , Hymecromone/pharmacology , Leukocytes/cytology , Leukocytes/drug effects , Leukocytes/immunology , Mice , Pancreas Transplantation/adverse effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous
9.
J Histochem Cytochem ; 68(11): 797-811, 2020 11.
Article in English | MEDLINE | ID: mdl-33064036

ABSTRACT

Hyaluronan and proteoglycan link protein 1 (HAPLN1) stabilizes interactions between two important extracellular matrix (ECM) macromolecules, versican and hyaluronan, which facilitate proliferation of fibroblasts and their conversion to myofibroblasts. However, the role of HAPLN1 in these events has not been studied. Using immunocytochemistry, cellular and ECM locations of HAPLN1 were evaluated in cultured human lung fibroblasts during proliferation and conversion to myofibroblasts. HAPLN1 localized to pericellular matrices, associating with both versican and hyaluronan in the ECM and on the cell surface. Nuclear and total HAPLN1 immunostaining increased after myofibroblast induction. Confocal microscopy showed HAPLN1 predominant in the ECM under cells while versican predominated above cells. Versican and HAPLN1 were also juxtaposed in columnar inclusions in the cytoplasm and nucleus. Nuclear HAPLN1 staining in interphase cells redistributed to the cytosol during mitosis. In the absence of TGF-ß1, addition of exogenous bovine HAPLN1 (together with aggrecan G1) facilitated myofibroblast formation, as seen by significant upregulation of α-smooth muscle actin (SMA) staining, while adding full-length bovine versican had no effect. Increased compaction of hyaluronan-rich ECM suggests that HAPLN1 plus G1 addition affects hyaluronan networks and myofibroblast formation. These observations demonstrate changes in both extracellular and intracellular localization of HAPLN1 during fibroblast proliferation and myofibroblast conversion suggesting a possible role in fibrotic remodeling.


Subject(s)
Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Lung/cytology , Proteoglycans/metabolism , Cell Proliferation , Cells, Cultured , Fibroblasts/cytology , Humans , Phenotype , Versicans/metabolism
10.
Fluids Barriers CNS ; 17(1): 60, 2020 Sep 29.
Article in English | MEDLINE | ID: mdl-32993718

ABSTRACT

BACKGROUND: The microvasculature (MV) of brains with Alzheimer's disease neuropathologic change (ADNC) and cerebral amyloid angiopathy (CAA), in the absence of concurrent pathologies (e.g., infarctions, Lewy bodies), is incompletely understood. OBJECTIVE: To analyze microvascular density, diameter and extracellular matrix (ECM) content in association with ADNC and CAA. METHODS: We examined samples of cerebral cortex and isolated brain microvasculature (MV) from subjects with the National Institute on Aging-Alzheimer's Association (NIA-AA) designations of not-, intermediate-, or high ADNC and from subjects with no CAA and moderate-severe CAA. Cases for all groups were selected with no major (territorial) strokes, ≤ 1 microinfarct in screening sections, and no Lewy body pathology. MV density and diameter were measured from cortical brain sections. Levels of basement membrane (BM) ECM components, the protein product of TNF-stimulated gene-6 (TSG-6), and the ubiquitous glycosaminoglycan hyaluronan (HA) were assayed by western blots or HA ELISA of MV lysates. RESULTS: We found no significant changes in MV density or diameter among any of the groups. Levels of BM laminin and collagen IV (col IV) were lower in MV isolated from the high ADNC vs. not-ADNC groups. In contrast, BM laminin was significantly higher in MV from the moderate-severe CAA vs. the no CAA groups. TSG-6 and HA content were higher in the presence of both high ADNC and CAA, whereas levels of BM fibronectin and perlecan were similar among all groups. CONCLUSIONS: Cortical MV density and diameter are not appreciably altered by ADNC or CAA. TSG-6 and HA are increased in both ADNC and CAA, with laminin and col IV decreased in the BM of high ADNC, but laminin increased in moderate-severe CAA. These results show that changes in the ECM occur in AD and CAA, but independently of one another, and likely reflect on the regional functioning of the brain microvasculature.


Subject(s)
Alzheimer Disease , Basement Membrane , Cerebral Amyloid Angiopathy , Cerebral Cortex , Extracellular Matrix , Microvessels , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Basement Membrane/blood supply , Basement Membrane/metabolism , Basement Membrane/pathology , Cell Adhesion Molecules/metabolism , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Collagen/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Humans , Hyaluronic Acid/metabolism , Laminin/metabolism , Male , Microvessels/pathology , Tissue Banks
11.
J Alzheimers Dis ; 67(1): 91-102, 2019.
Article in English | MEDLINE | ID: mdl-30507579

ABSTRACT

Little is known about the extracellular matrix (ECM) during progression of AD pathology. Brain ECM is abundant in hyaluronan (HA), a non-sulfated glycosaminoglycan synthesized by HA synthases (HAS) 1-3 in a high molecular weight (MW) form that is degraded into lower MW fragments. We hypothesized that pathologic severity of AD is associated with increases in HA and HA-associated ECM molecules. To test this hypothesis, we assessed HA accumulation and size; HA synthases (HAS) 1-3; and the HA-stabilizing hyaladherin, TSG-6 in parietal cortex samples from autopsied research subjects with not AD (CERAD = 0, Braak = 0- II, n = 12-21), intermediate AD (CERAD = 2, Braak = III-IV, n = 13-18), and high AD (CERAD = 3, Braak = V-VI, n = 32-40) neuropathologic change. By histochemistry, HA was associated with deposits of amyloid and tau, and was also found diffusely in brain parenchyma, with overall HA quantity (measured by ELSA) significantly greater in brains with high AD neuropathology. Mean HA MW was similar among the samples. HAS2 and TSG-6 mRNA expression, and TSG-6 protein levels were significantly increased in high AD and both molecules were present in vasculature, NeuN-positive neurons, and Iba1-positive microglia. These results did not change when accounting for gender, advanced age (≥ 90 years versus <90 years), or the clinical diagnosis of dementia. Collectively, our results indicate a positive correlation between HA accumulation and AD neuropathology, and suggest a possible role for HA synthesis and metabolism in AD progression.


Subject(s)
Alzheimer Disease/pathology , Cell Adhesion Molecules/analysis , Hyaluronic Acid/analysis , Aged , Aged, 80 and over , Alzheimer Disease/blood , Amyloid beta-Peptides/analysis , Autopsy , Disease Progression , Extracellular Matrix Proteins/analysis , Female , Humans , Male , Parietal Lobe/chemistry , RNA, Messenger/analysis , tau Proteins/analysis
12.
Mater Sci Eng C Mater Biol Appl ; 93: 390-398, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30274071

ABSTRACT

Immunomodulatory monoclonal antibodies (IM-mAbs) are a cornerstone of modern immunotherapy; however, when administered systemically (i.e., via injection), these agents can generate a variety of negative side effects. For many diseases, systemic delivery of IM-mAbs is the most effective mode of treatment, but in instances where the cellular target occupies a limited, well-defined space (e.g., solid tumors or cellularized implants) local, controlled release of IM-mAbs might be desirable. Antibodies are highly sensitive to a variety of environmental conditions, which limit the kinds of polymers suitable for antibody retention and controlled release. The present study evaluates the release of antibodies from biocompatible, 2-mm diameter alginate spheres coated with poly-l-lysine and a thin outer layer of alginate (APA spheres). In vitro, rates of antibody release (including IM-mAbs) could be incrementally decreased and made linear by incrementally increasing the quantity of poly-l-lysine deposited on the alginate, with linear release lasting in one scenario for at least 46 days. To evaluate the bioactivity in vivo of IM-mAbs, APA spheres loaded with either anti-CD3ε or anti-CD95 mAb were incorporated into scaffolded islet implant (SI) test-beds and the SIs implanted into a mouse model of autoimmune (type 1) diabetes. Release of mAbs within the implanted SIs resulted in reduced autoimmune responses to both transplanted and native islets. Notably, mice implanted with APA spheres loaded with quantities of anti-CD95 mAb that would be lethal if given systemically showed immunomodulation with no toxic side effects. Collectively, our results indicate that APA spheres are a relatively simple means to evaluate the effects of local, controlled release of IM-mAbs in a way that preserves mAb function and limits systemic toxicity.


Subject(s)
Alginates , Antibodies, Monoclonal , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Immunologic Factors , Polylysine , Alginates/chemistry , Alginates/pharmacokinetics , Alginates/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Drug Implants , Immunologic Factors/chemistry , Immunologic Factors/pharmacokinetics , Immunologic Factors/pharmacology , Mice , Mice, Inbred BALB C , Mice, Knockout , Polylysine/chemistry , Polylysine/pharmacokinetics , Polylysine/pharmacology
13.
Cell Transplant ; 27(3): 531-541, 2018 03.
Article in English | MEDLINE | ID: mdl-29756517

ABSTRACT

Islet transplantation remains the only alternative to daily insulin therapy for control of type 1 diabetes (T1D) in humans. To avoid the drawbacks of intrahepatic islet transplantation, we are developing a scaffolded islet implant to transplant islets into nonhepatic sites. The implant test bed, sized for mice, consists of a limited (2-mm) thickness, large-pore polymeric sponge scaffold perforated with peripheral cavities that contain islets suspended in a collagen hydrogel. A central cavity in the scaffold holds a 2-mm diameter alginate sphere for controlled release of the angiogenic cytokine vascular endothelial growth factor ( VEGF). Host microvessels readily penetrate the scaffold and collagen gel to vascularize the islets. Here, we evaluate the performance of the implant in a subcutaneous (SC) graft site. Implants incorporating 500 syngeneic islets reversed streptozotocin-induced diabetes in mice approximately 30 d after SC placement. Controlled release of a modest quantity (20 ng) of VEGF within the implant significantly reduced the time to normoglycemia compared to control implants lacking VEGF. Investigation of underlying causes for this effect revealed that inclusion of 20 ng of VEGF in the implants significantly reduced central necrosis of islets 24 h after grafting and increased implant vascularization (measured 12 d after grafting). Collectively, our results demonstrate (1) that the scaffolded islet implant design can reverse diabetes in SC sites in the absence of prevascularization of the graft site and (2) that relatively low quantities of VEGF, delivered by controlled release within the implant, can be a useful approach to limit islet stress after grafting.


Subject(s)
Islets of Langerhans Transplantation/methods , Vascular Endothelial Growth Factor A/metabolism , Alginates/chemistry , Animals , Delayed-Action Preparations , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/therapy , Graft Survival , Immunohistochemistry , Islets of Langerhans/cytology , Mice , Mice, Inbred C57BL , Necrosis/metabolism
14.
J Histochem Cytochem ; 66(10): 697-707, 2018 10.
Article in English | MEDLINE | ID: mdl-29782809

ABSTRACT

The brain changes in volume and composition with normal aging. Cellular components of the brain are supported by an extracellular matrix (ECM) comprised largely of hyaluronan (HA) and HA-associated members of the lectican family of chondroitin sulfate proteoglycans (CSPGs). We examined regional differences in microvascular density, neuronal and glial markers, and accumulation of HA and CSPGs in mouse brains during normal aging. The cortex, hippocampus, dentate gyrus, and cerebellum of young (4 months), middle-aged (14 months), and aged (24-26 months) brains were analyzed. Microvascular density decreased in cerebral cortex and cerebellum with age. There were no detectable differences in neuronal density. There was an increase in astrocytes in the hippocampus with aging. HA accumulation was higher in aged brain relative to young brain in the cerebral cortex and cerebellum, but not in other regions examined. In contrast, CSPGs did not change with aging in any of the brain regions examined. HA and CSPGs colocalized with a subset of neuronal cell bodies and astrocytes, and at the microvasculature. Differences in accumulation of ECM in the aging brain, in the setting of decreased microvascular density and/or increased glial activation, might contribute to age-related regional differences in vulnerability to injury and ischemia.


Subject(s)
Aging , Brain/physiology , Brain/ultrastructure , Chondroitin Sulfate Proteoglycans/metabolism , Hyaluronic Acid/metabolism , Animals , Chondroitin Sulfate Proteoglycans/analysis , Fluorescent Antibody Technique/methods , Hippocampus/physiology , Hippocampus/ultrastructure , Hyaluronic Acid/analysis , Image Processing, Computer-Assisted/methods , Male , Mice, Inbred C57BL , Microscopy, Fluorescence/methods
15.
J Gerontol A Biol Sci Med Sci ; 72(6): 740-746, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28482035

ABSTRACT

The microvasculature of the aged brain is less dense and more vulnerable to dysfunction than that of the young brain. Brain microvasculature is supported by its surrounding extracellular matrix, which is comprised largely of hyaluronan (HA). HA is continually degraded into lower molecular weight forms that induce neuroinflammation. We examined HA associated with microvessels (MV) of the cerebral cortex of young (4 months), middle-aged (14 months), and aged (24-26 months) mice. We confirmed that the density of cortical MV decreased with age. Perivascular HA levels increased with age, but there was no age-associated change in HA molecular weight profile. MV isolated from aged cortex had more HA than MV from young cortex. Examination of mechanisms that might account for elevated HA levels with aging showed increased HA synthase 2 (HAS2) mRNA and protein in aged MV relative to young MV. In contrast, mRNAs for HA-degrading hyaluronidases or hyaladherins that mitigate HA degradation showed no changes with age. Corresponding to increased HAS2, aged MV synthesized significantly more HA (of all molecular weight classes) in vitro than young MV. We propose that increased HA synthesis and accumulation in brain MV contributes to neuroinflammation and reduced MV density and function in aging.


Subject(s)
Aging/metabolism , Cerebral Cortex/metabolism , Hyaluronic Acid/biosynthesis , Microvessels/metabolism , Animals , Cerebral Cortex/blood supply , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Expression , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Hyaluronan Synthases , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/metabolism , Immunohistochemistry , Mice, Inbred C57BL , RNA, Messenger , Real-Time Polymerase Chain Reaction , Up-Regulation
16.
In Vivo ; 20(6B): 801-6, 2006.
Article in English | MEDLINE | ID: mdl-17203771

ABSTRACT

BACKGROUND: It is unclear whether delays in wound repair due to the age of the host persist into the later stages of healing. Late stage healing of dermal wounds and myocardial infarcts in rodents was examined to determine if aged animals "catch up" to their younger counterparts. MATERIALS AND METHODS: Excisional dermal wounds (5 mm) were made by punch biopsy and myocardial infarctions were produced by ligation of the left anterior descending coronary artery in young and aged mice and rats, respectively. Dermal wounds at day 11 and myocardial infarctions at day 14 were analyzed for wound area, angiogenesis, deposition of basement membrane proteins, and remodeling of collagen. RESULTS: Analyses demonstrated that wound areas, the deposition of basement membrane proteins and angiogenic responses were similar in young and aged rodents at late stages of wound repair. The dermal wounds of young mice had larger quantities of mature, compacted collagen fibers relative to aged mice, but immature collagen fibers predominated in myocardial infarcts in both young and aged rats. CONCLUSION: These results show that, with the exception of dermal collagen remodeling, aged animals catch up to their young counterparts with respect to many features of tissue repair. Consequently, therapies that target age-related deficiencies in healing will be most effective when administered shortly after the initial insult.


Subject(s)
Aging/physiology , Myocardial Infarction/physiopathology , Skin/physiopathology , Wound Healing/physiology , Animals , Blood Vessels/metabolism , Blood Vessels/physiopathology , Collagen/metabolism , Heart/physiopathology , Immunohistochemistry , Mice , Mice, Inbred Strains , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Inbred F344 , Skin/injuries , Skin/metabolism , Time Factors
17.
Diabetes ; 51(4): 1194-200, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11916944

ABSTRACT

Cardiovascular diseases represent a significant cause of morbidity and mortality in diabetes. Of the many animal models used in the study of non-insulin-dependent (type 2) diabetes, the JCR:LA-cp rat is unique in that it develops insulin resistance in the presence of obesity and manifests both peripheral and coronary vasculopathies. In this animal model, arterial vascular smooth muscle cells (VSMCs) from homozygous obese (cp/cp) rats, but not from age-matched healthy (+/+ or + /cp, collectively defined +/?) littermates, display an " activated" phenotype in vitro and in vivo and have an elevated level of cAMP phosphodiesterase (PDE) activity. In this report, we confirm that cp/cp rat aortic VSMCs have an elevated level of PDE3 activity and show that only particulate PDE3 (PDE3B) activity is elevated. In marked contrast to results obtained in + /? VSMCs, simultaneous activation of adenylyl cyclase and inhibition of PDE3 activity in cp/cp VSMCs synergistically increased cAMP. Although PDE3 inhibition did not potentiate the antimigratory effects of forskolin on +/? VSMCs, PDE3 inhibition did markedly potentiate the forskolin-induced inhibition of migration of cp/cp-derived VSMCs. Although PDE3 activity was elevated in cp/cp rat aortic VSMCs, levels of expression of cytosolic PDE3 (PDE3A) and PDE3B in +/? and cp/cp VSMCs, as well as activation of these enzymes following activation of the cAMP-protein kinase A signaling cascade, were not different. Our data are consistent with an increased role for PDE3 in regulating cAMP-dependent signaling in cp/cp VSMCs and identify PDE3 as a cellular activity potentially responsible for the phenotype of cp/cp VSMCs.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cyclic AMP/metabolism , Diabetes Mellitus, Type 2/physiopathology , Muscle, Smooth, Vascular/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , 4-(3-Butoxy-4-methoxybenzyl)-2-imidazolidinone/pharmacology , Animals , Aorta/physiology , Cell Membrane/enzymology , Cell Movement , Cells, Cultured , Colforsin/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Cytosol/enzymology , Hydrolysis , Obesity/physiopathology , Phenotype , Phosphodiesterase Inhibitors/pharmacology , Quinolones/pharmacology , Rats , Rats, Mutant Strains
18.
J Histochem Cytochem ; 53(5): 571-81, 2005 May.
Article in English | MEDLINE | ID: mdl-15872050

ABSTRACT

Secreted protein acidic and rich in cysteine (SPARC) and thrombospondin-2 (TSP-2) are structurally unrelated matricellular proteins that have important roles in cell-extracellular matrix (ECM) interactions and tissue repair. SPARC-null mice exhibit accelerated wound closure, and TSP-2-null mice show an overall enhancement in wound healing. To assess potential compensation of one protein for the other, we examined cutaneous wound healing and fibrovascular invasion of subcutaneous sponges in SPARC-TSP-2 (ST) double-null and wild-type (WT) mice. Epidermal closure of cutaneous wounds was found to occur significantly faster in ST-double-null mice, compared with WT animals: histological analysis of dermal wound repair revealed significantly more mature phases of healing at 1, 4, 7, 10, and 14 days after wounding, and electron microscopy showed disrupted ECM at 14 days in these mice. ST-double-null dermal fibroblasts displayed accelerated migration, relative to WT fibroblasts, in a wounding assay in vitro, as well as enhanced contraction of native collagen gels. Zymography indicated that fibroblasts from ST-double-null mice also produced higher levels of matrix metalloproteinase (MMP)-2. These data are consistent with the increased fibrovascular invasion of subcutaneous sponge implants seen in the double-null mice. The generally accelerated wound healing of ST-double-null mice reflects that described for the single-null animals. Importantly, the absence of both proteins results in elevated MMP-2 levels. SPARC and TSP-2 therefore perform similar functions in the regulation of cutaneous wound healing, but fine-tuning with respect to ECM production and remodeling could account for the enhanced response seen in ST-double-null mice.


Subject(s)
Neovascularization, Physiologic , Osteonectin/physiology , Thrombospondins/physiology , Wound Healing , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Collagen/physiology , Extracellular Matrix/ultrastructure , Fibroblasts/physiology , Gels , Mice , Mice, Knockout , Osteonectin/genetics , Polyvinyl Alcohol , Skin/injuries , Skin/pathology , Thrombospondins/genetics
19.
Biomaterials ; 26(16): 3131-40, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15603808

ABSTRACT

For several years, microgrooved substrates have been evaluated as a means to orient cells in engineered tissues. Recently, we fabricated thin (0.1-5.3 microm) planar and tubular collagen membranes (CMs) from air-dried hydrogels of native, fibrillar type I collagen (Vernon et al., Biomaterials 2004;26:1109-17). The CMs were strong, stable, and permeable and, hence, of potential use as scaffolds for tissue engineering. In the present study, planar CMs supported a robust attachment, spreading, and proliferation of human dermal fibroblasts (HDFs) and human umbilical artery smooth muscle cells (HUASMCs). Collagen hydrogels were air-dried onto microgrooved templates and subsequently removed in the form of grooved CMs with the potential to align cells. The grooved CMs were highly effective at inducing HDFs and HUASMCs to elongate and align, as revealed by scanning electron microscopy and by assays of f-actin and nuclear orientation. Alignment of cells was maintained at high cell densities. CMs with grooves of substantially different widths and depths were similarly effective in causing cell alignment; however, cells aligned poorly on CMs that had grooves less than 1 microm in depth. Grooved CMs with the capability to align cells might be of considerable use in the fabrication of tissue substitutes.


Subject(s)
Biocompatible Materials/chemistry , Fibrillar Collagens/chemistry , Actins/chemistry , Cell Adhesion , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Collagen Type I/chemistry , Endothelium, Vascular/cytology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Hydrogels/chemistry , Microscopy, Electron, Scanning , Myocytes, Smooth Muscle/cytology , Surface Properties , Time Factors , Tissue Engineering , Umbilical Veins/cytology
20.
Biomaterials ; 26(10): 1109-17, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15451630

ABSTRACT

Fibrillar type I collagen is nontoxic, biocompatible, and possesses considerable strength and stability. In a study of scaffolds for use in laminated tissue substitutes, we examined the properties of membranes made from air-dried hydrogels of collagen fibrils that were polymerized from native, monomeric collagen. Planar collagen membranes (CMs) of 0.1-5.3 microm dry thickness were made by variation of the collagen concentration and/or the volume of the hydrogel. The planar CMs, which were comprised of a dense feltwork of long collagen fibrils 70-100 nm in diameter, showed considerable resistance to rupture and retained their membranous character after 6 weeks in tissue culture medium at 37 degrees C. CMs that were relatively thick when dry exhibited a greater proportional increase in rehydrated thickness and a greater diffusivity (when rehydrated) to 4.3 kDa dextran than did CMs that were relatively thin when dry. Hollow, tubular CMs of several configurations were prepared by embedment of solid, removable forms into collagen hydrogels prior to drying. By use of special fixtures, a planar CM that incorporated multiple, parallel tubes was fabricated. In summary, hydrogels of fibrillar collagen can be transformed into membranous structures suitable for tissue engineering applications.


Subject(s)
Cell Culture Techniques/methods , Collagen Type I/chemistry , Collagen Type I/ultrastructure , Membranes, Artificial , Nanotechnology/methods , Tissue Engineering/methods , Biocompatible Materials/chemistry , Hydrogels/chemistry , Materials Testing , Porosity , Tensile Strength
SELECTION OF CITATIONS
SEARCH DETAIL