Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Vet Sci ; 10: 1057277, 2023.
Article in English | MEDLINE | ID: mdl-37035804

ABSTRACT

Toxoplasmosis is a parasitic disease affecting a wide range of species, including humans, and can be responsible for important clinical manifestations such as abortion and neurological signs. Sheep show a remarkable susceptibility to its causative agent, Toxoplasma gondii, and zoonotic transmission may occur in case of consumption of undercooked meat obtained from infected animals. Toxoplasma gondii seroprevalence in sheep can significantly vary on a geographical basis, as shown by numerous surveys conducted worldwide. To investigate environmental and climate conditions that may affect the likelihood of ovine infection, 405 serum samples from selected sheep raised in 91 farms were collected from two abattoirs, with each abattoir receiving animals from two regions (1/Tuscany-Latium and 2/Campania-Basilicata). The seroprevalence of infection in all examined animals was 53.8%. Young animals (n = 165) had a lower likelihood of being T. gondii positive compared to the adults (OR = 0.21), and the seropositive rate of animals slaughtered in abattoir 2 was significantly higher than that of animals slaughtered in abattoir 1 (60.5 vs. 43.2%, p < 0.01). The significant bioclimatic variables (p < 0.05) associated with the presence of T. gondii antibodies were related to areas with a lower range of temperature and higher precipitation. In conclusion, this study expands on the interpretation of serological data, with the inclusion of environmental and climatic variables, as possible risk factors in the spread of toxoplasmosis in the study area. These findings provide novel insights to support public health measures, such as risk-based control plan, and contribute to a "One Health" approach, taking into account the environmental and climatic perspectives.

2.
Life (Basel) ; 11(3)2021 Mar 21.
Article in English | MEDLINE | ID: mdl-33801080

ABSTRACT

Recent scientific literature has investigated the cardiovascular implications of COVID-19. The mechanisms of cardiovascular damage seem to involve the protein angiotensin-converting enzyme 2 (ACE2), to which severe acute respiratory syndrome (SARS) coronavirus-2 (CoV-2) binds to penetrate cells and other mechanisms, most of which are still under study. Cardiovascular sequelae of COVID-19 include heart failure, cardiomyopathy, acute coronary syndrome, arrhythmias, and venous thromboembolism. This article aims to collect scientific evidence by exploiting PubMed, Scopus, and Pedro databases to highlight the cardiovascular complications of COVID-19 and to define the physiotherapy treatment recommended for these patients. Exercise training (ET), an important part of cardiac rehabilitation, is a powerful tool in physiotherapy, capable of inducing significant changes in the cardiovascular system and functional in the recovery of endothelial dysfunction and for the containment of thromboembolic complications. In conclusion, due to the wide variety of possible exercise programs that can be obtained by combining intensity, duration, and speed in various ways, and by adjusting the program based on continuous patient monitoring, exercise training is well suited to the treatment of post-COVID patients with an impaired cardiovascular system of various degrees.

3.
Toxins (Basel) ; 12(5)2020 05 07.
Article in English | MEDLINE | ID: mdl-32392817

ABSTRACT

Ochratoxin A (OTA) is the most toxic member of ochratoxins, a group of toxic secondary metabolites produced by fungi. The most relevant species involved in OTA production in grapes is Aspergillus carbonarius. Berry infection by A. carbonarius is enhanced by damage to the skin caused by abiotic and biotic factors. Insect pests play a major role in European vineyards, and Lepidopteran species such as the European grapevine moth Lobesia botrana are undoubtedly crucial. New scenarios are also emerging due to the introduction and spread of allochthonous pests as well as climate change. Such pests may be involved in the dissemination of OTA producing fungi even if confirmation is still lacking and further studies are needed. An OTA predicting model is available, but it should be integrated with models aimed at forecasting L. botrana phenology and demography in order to improve model reliability.


Subject(s)
Aspergillus/metabolism , Food Contamination/prevention & control , Food Microbiology , Fruit/microbiology , Moths/microbiology , Ochratoxins/metabolism , Pest Control , Vitis/microbiology , Animals , Food Chain , Fruit/parasitology , Fruit and Vegetable Juices/microbiology , Vitis/parasitology , Wine/microbiology
4.
Microorganisms ; 8(1)2020 Jan 03.
Article in English | MEDLINE | ID: mdl-31947721

ABSTRACT

Dietary (co)-exposure to mycotoxins is associated with human and animal health concerns as well as economic losses. This study aims to give a data-based insight from the scientific literature on the (co-)occurrence of mycotoxins (i.e., parent and modified forms) in European core cereals, and to estimate potential patterns of co-exposure in humans and animals. Mycotoxins were mainly reported in wheat and maize showing the highest concentrations of fumonisins (FBs), deoxynivalenol (DON), aflatoxins (AFs), and zearalenone (ZEN). The maximum concentrations of FB1+FB2 were reported in maize both in feed and food and were above legal maximum levels (MLs). Similar results were observed in DON-food, whose max concentrations in wheat, barley, maize, and oat exceeded the MLs. Co-occurrence was reported in 54.9% of total records, meaning that they were co-contaminated with at least two mycotoxins. In the context of parental mycotoxins, co-occurrence of DON was frequently observed with FBs in maize and ZEN in wheat; DON + NIV and DON + T2/HT2 were frequently reported in barley and oat, respectively. Apart from the occurrence of ZEN and its phase I and phase II modified forms, only a limited number of quantified data were available for other modified forms; i.e., mainly the acetyl derivatives of DON. Data gaps are highlighted together with the need for monitoring studies on multiple mycotoxins to identify co-occurrence patterns for parent mycotoxins, metabolites, and their modified forms.

5.
J Immunol ; 184(8): 4497-509, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20231695

ABSTRACT

Inhibitor of NF-kappaB kinases beta (IKKbeta) and alpha (IKKalpha) activate distinct NF-kappaB signaling modules. The IKKbeta/canonical NF-kappaB pathway rapidly responds to stress-like conditions, whereas the IKKalpha/noncanonical pathway controls adaptive immunity. Moreover, IKKalpha can attenuate IKKbeta-initiated inflammatory responses. High mobility group box 1 (HMGB1), a chromatin protein, is an extracellular signal of tissue damage-attracting cells in inflammation, tissue regeneration, and scar formation. We show that IKKalpha and IKKbeta are each critically important for HMGB1-elicited chemotaxis of fibroblasts, macrophages, and neutrophils in vitro and neutrophils in vivo. By time-lapse microscopy we dissected different parameters of the HMGB1 migration response and found that IKKalpha and IKKbeta are each essential to polarize cells toward HMGB1 and that each kinase also differentially affects cellular velocity in a time-dependent manner. In addition, HMGB1 modestly induces noncanonical IKKalpha-dependent p52 nuclear translocation and p52/RelB target gene expression. Akin to IKKalpha and IKKbeta, p52 and RelB are also required for HMGB1 chemotaxis, and p52 is essential for cellular orientation toward an HMGB1 gradient. RAGE, a ubiquitously expressed HMGB1 receptor, is required for HMGB1 chemotaxis. Moreover, IKKbeta, but not IKKalpha, is required for HMGB1 to induce RAGE mRNA, suggesting that RAGE is at least one IKKbeta target involved in HMGB1 migration responses, and in accord with these results enforced RAGE expression rescues the HMGB1 migration defect of IKKbeta, but not IKKalpha, null cells. Thus, proinflammatory HMGB1 chemotactic responses mechanistically require the differential collaboration of both IKK-dependent NF-kappaB signaling pathways.


Subject(s)
Chemotaxis/immunology , HMGB1 Protein/physiology , I-kappa B Kinase/physiology , Animals , Cells, Cultured , Chemotaxis/genetics , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/immunology , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , Macrophages/cytology , Macrophages/enzymology , Macrophages/immunology , Mice , Mice, Knockout , Mice, Transgenic , Neutrophils/cytology , Neutrophils/enzymology , Neutrophils/immunology , Recombinant Proteins/pharmacology , Signal Transduction/genetics , Signal Transduction/immunology
6.
Autoimmunity ; 42(4): 308-10, 2009 May.
Article in English | MEDLINE | ID: mdl-19811285

ABSTRACT

High mobility group B box (HMGB) proteins are a family of chromatin proteins made up of two basic DNA binding domains, HMG box A and B, and a C-terminal acidic tail. HMGB have a highly conserved sequence, but different expression pattern: HMGB1 is almost ubiquitous, whereas the others are highly expressed in only a few tissues in adults. We previously demonstrated that HMGB1 is released by necrotic cells and has chemoattractant activity for inflammatory and stem cells, via binding to receptor for advanced glycation endproducts (RAGE). HMGB1 can be actively secreted by inflammatory cells. Here, we report that also HMGB2 can be secreted by THP-1 cells, and promotes proliferation and migration of endothelial cells. These functions of HMGB2 are exerted via engagement of RAGE, whose blockade completely abrogates cell responses. Since extracellular HMGB2 has been detected in the blood and other biological fluids, it might be necessary to target HMGB2 at the same time as HMGB1 for therapeutical efficacy.


Subject(s)
Chemotactic Factors/metabolism , HMGB1 Protein/metabolism , HMGB2 Protein/metabolism , Myeloid Cells/metabolism , Animals , Blotting, Western , Cell Movement/immunology , Cell Proliferation , Chemotactic Factors/immunology , HMGB1 Protein/immunology , HMGB2 Protein/immunology , Humans , Mice , Mice, Knockout , Myeloid Cells/immunology
7.
J Leukoc Biol ; 86(3): 617-23, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19401391

ABSTRACT

HMGB1 is a nuclear protein that signals tissue damage, as it is released by cells dying traumatically or secreted by activated innate immunity cells. Extracellular HMGB1 elicits the migration to the site of tissue damage of several cell types, including inflammatory cells and stem cells. The identity of the signaling pathways activated by extracellular HMGB1 is not known completely: We reported previously that ERK and NF-kappaB pathways are involved, and we report here that Src is also activated. The ablation of Src or inhibition with the kinase inhibitor PP2 blocks migration toward HMGB1. Src associates to and mediates the phosphorylation of FAK and the formation of focal adhesions.


Subject(s)
Cell Movement/physiology , Fibroblasts/physiology , HMGB1 Protein/metabolism , Leukocytes, Mononuclear/physiology , src-Family Kinases/physiology , 3T3 Cells , Animals , Cell Movement/drug effects , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/physiology , Coated Materials, Biocompatible/metabolism , Culture Media, Serum-Free , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibronectins/metabolism , Focal Adhesions/metabolism , HMGB1 Protein/pharmacology , Humans , Isoelectric Point , Leukocytes, Mononuclear/drug effects , Mice , Paxillin/metabolism , Phosphorylation/drug effects , Pyrimidines/pharmacology , Temperature , Time Factors , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
8.
J Invest Dermatol ; 128(6): 1545-53, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18239618

ABSTRACT

High-mobility group box 1 (HMGB1) protein is a multifunctional cytokine involved in inflammatory responses and tissue repair. In this study, it was examined whether HMGB1 plays a role in skin wound repair both in normoglycemic and diabetic mice. HMGB1 was detected in the nucleus of skin cells, and accumulated in the cytoplasm of epidermal cells in the wounded skin. Diabetic human and mouse skin showed more reduced HMGB1 levels than their normoglycemic counterparts. Topical application of HMGB1 to the wounds of diabetic mice enhanced arteriole density, granulation tissue deposition, and accelerated wound healing. In contrast, HMGB1 had no effect in normoglycemic mouse skin wounds, where endogenous HMGB1 levels may be adequate for optimal wound closure. Accordingly, inhibition of endogenous HMGB1 impaired wound healing in normal mice but had no effect in diabetic mice. Finally, HMGB1 had a chemotactic effect on skin fibroblasts and keratinoyctes in vitro. In conclusion, lower HMGB1 levels in diabetic skin may play an important role in impaired wound healing and this defect may be overcome by the topical application of HMGB1.


Subject(s)
Gene Expression Regulation , HMGB1 Protein/biosynthesis , HMGB1 Protein/physiology , Wound Healing , Animals , Chemotaxis , Cytoplasm/metabolism , Diabetes Complications/metabolism , Diabetes Complications/therapy , Epidermal Cells , Fibroblasts/metabolism , Humans , Inflammation , Keratinocytes/cytology , Mice , Models, Biological , Skin/pathology
9.
J Cell Biol ; 179(1): 33-40, 2007 Oct 08.
Article in English | MEDLINE | ID: mdl-17923528

ABSTRACT

Tissue damage is usually followed by healing, as both differentiated and stem cells migrate to replace dead or damaged cells. Mesoangioblasts (vessel-associated stem cells that can repair muscles) and fibroblasts migrate toward soluble factors released by damaged tissue. Two such factors are high mobility group box 1 (HMGB1), a nuclear protein that is released by cells undergoing unscheduled death (necrosis) but not by apoptotic cells, and stromal derived factor (SDF)-1/CXCL12. We find that HMGB1 activates the canonical nuclear factor kappaB (NF-kappaB) pathway via extracellular signal-regulated kinase phosphorylation. NF-kappaB signaling is necessary for chemotaxis toward HMGB1 and SDF-1/CXCL12, but not toward growth factor platelet-derived growth factor, formyl-met-leu-phe (a peptide that mimics bacterial invasion), or the archetypal NF-kappaB-activating signal tumor necrosis factor alpha. In dystrophic mice, mesoangioblasts injected into the general circulation ingress inefficiently into muscles if their NF-kappaB signaling pathway is disabled. These findings suggest that NF-kappaB signaling controls tissue regeneration in addition to early events in inflammation.


Subject(s)
Chemotaxis/physiology , HMGB1 Protein/metabolism , NF-kappa B/physiology , Signal Transduction , Animals , Cell Line , Chemokine CXCL12/metabolism , Fibroblasts/metabolism , Fibroblasts/physiology , Green Fluorescent Proteins/analysis , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Necrosis , Phosphorylation , Recombinant Fusion Proteins/analysis
10.
Arterioscler Thromb Vasc Biol ; 27(11): 2377-83, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17872450

ABSTRACT

OBJECTIVE: High mobility group box 1 protein (HMGB1) is a cytokine released by necrotic and inflammatory cells in response to injury. We examined the role of HMGB1 in skeletal muscle regeneration after hindlimb ischemia. METHODS AND RESULTS: Unilateral hindlimb ischemia was induced in mice by femoral artery dissection. HMGB1 levels increased in regenerating skeletal muscle and the blockade of endogenous HMGB1 by the administration of its truncated form, the BoxA, resulted in the reduction of vessel density. In contrast, intramuscular administration of HMGB1 enhanced perfusion and increased the number of regenerating fibers. To separately study the myogenic and the angiogenic effects of HMGB1, in vitro experiments were performed with isolated myoblasts and endothelial cells. Myoblasts were found to express the HMGB1 receptor RAGE and TLR4 which were downregulated during in vitro myogenic differentiation. HMGB1 was extracellularly released by differentiated myoblasts and exerted a chemotactic activity on myogenic cells. This effect was partially dependent on RAGE and was inhibited by BoxA treatment. Finally, HMGB1 stimulated tubular-like structure formation by endothelial cells through the activation of extracellular signal-regulated kinase (ERK) and JNK signal transduction pathways. CONCLUSIONS: HMGB1 plays a role in skeletal muscle regeneration modulating, in an autocrine-paracrine manner, myoblast and endothelial cell functions.


Subject(s)
Femoral Artery/physiology , HMGB1 Protein/metabolism , Ischemia/physiopathology , Muscle, Skeletal/physiology , Regeneration/physiology , Animals , Autocrine Communication , Disease Models, Animal , Femoral Artery/injuries , Mice , Myoblasts, Skeletal/physiology , Neovascularization, Physiologic/physiology , Paracrine Communication
11.
Autoimmunity ; 40(4): 285-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17516211

ABSTRACT

When tissues are damaged, they usually heal. The cellular responses towards healing require the prior recognition that damage has occurred. High Mobility Group Box 1 protein (HMGB1) is a ubiquitous nuclear protein that is passively released by cells that have died in a traumatic, non-programmed way (necrosis). Several receptors for HMGB1 exist, and upon binding HMGB1 they alert leukocytes to extravasate from the blood into the affected tissue, trigger adaptive immunity and promote the migration and proliferation of cells (including stem cells) to repair the damaged tissue. Significantly, apoptotic cells modify their chromatin so as to bind HMGB1, which is not released. Several cell types (in particular inflammatory cells) when distressed have the ability to secrete HMGB1 actively, via a dedicated pathway, and thus produce a damage signal without dying. Because of its powerful activities, HMGB1 is involved in several disorders, including autoimmune ones.


Subject(s)
Autoimmune Diseases/immunology , HMGB1 Protein/immunology , Leukocytes/immunology , Necrosis/immunology , Regeneration/immunology , Signal Transduction/immunology , Animals , Autoimmune Diseases/pathology , Cell Movement/immunology , Cell Proliferation , Humans , Leukocytes/pathology , Necrosis/pathology , Stem Cells/immunology , Stem Cells/pathology
12.
FASEB J ; 20(14): 2565-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17060403

ABSTRACT

High mobility group box 1 protein (HMGB1) is a chromatin component leaked out by necrotic cells and actively secreted by activated myeloid cells. The extracellular protein is a potent mediator of tissue remodeling. We show here that human atherosclerotic plaques, but not normal arteries, produce extracellular HMGB1. Secreted HMGB1 originates from endothelial cells, by neointimal foam cells, and also smooth muscle cells (SMCs). SMCs are an unexpected source for secreted HMGB1, since they normally express much lower amounts of HMGB1 than other cells types, and they do not secrete it. However, cultured SMCs actively secrete HMGB1 after cholesterol loading. In turn, in response to HMGB1, SMCs proliferate, migrate, and secrete more HMGB1. Thus, SMCs are both a source and a target of HMGB1; blocking HMGB1 secretion by SMCs can be an important strategy for treatment of atherosclerotic disease and in particular restenosis.


Subject(s)
Atherosclerosis/metabolism , Cell Proliferation , HMGB1 Protein/metabolism , Myocytes, Smooth Muscle/metabolism , Cell Movement/physiology , HMGB1 Protein/genetics , Humans , Myocytes, Smooth Muscle/cytology
13.
J Thorac Cardiovasc Surg ; 131(4): 853-61, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16580444

ABSTRACT

OBJECTIVE: We sought to evaluate magnesium as a neuroprotectant in patients undergoing cardiac surgery with cardiopulmonary bypass. METHODS: From February 2002 to September 2003, 350 patients undergoing elective coronary artery bypass grafting, valve surgery, or both were enrolled in a randomized, blinded, placebo-controlled trial to receive either magnesium sulfate to increase plasma levels 1(1/2) to 2 times normal during cardiopulmonary bypass (n = 174) or no intervention (n = 176). Neurologic function, neuropsychologic function, and depression were assessed preoperatively, at 24 and 96 hours after extubation (neurologic) and at 3 months (neuropsychologic, depression). Neurologic scores were analyzed using ordinal longitudinal methods, and neuropsychologic and depression inventory data were summarized by principal component analysis, followed by linear regression analysis using component scores as response variables. RESULTS: Seven (2%) patients had a postoperative stroke, 2 (1%) in the magnesium and 5 (3%) in the placebo group (P = .4). Neurologic score was worse postoperatively in both groups (P < .0001); however, magnesium group patients performed better than placebo group patients (P = .0001), who had prolonged declines in short-term memory and reemergence of primitive reflexes. Three-month neuropsychologic performance and depression inventory score were generally better than preoperatively, with few differences between groups (P > .6); however, older age (P = .0006), previous stroke (P = .003), and lower education level (P = .0007) were associated with worse performance. CONCLUSIONS: Magnesium administration is safe and improves short-term postoperative neurologic function after cardiac surgery, particularly in preserving short-term memory and cortical control over brainstem functions. However, by 3 months, other factors and not administration of magnesium influence neuropsychologic and depression inventory performance.


Subject(s)
Coronary Artery Bypass , Magnesium Sulfate/therapeutic use , Neuroprotective Agents/therapeutic use , Aged , Cardiopulmonary Bypass , Depression/epidemiology , Female , Humans , Length of Stay , Magnesium Sulfate/blood , Male , Memory , Mental Status Schedule , Middle Aged , Neuropsychological Tests , Postoperative Complications/epidemiology , Principal Component Analysis , Stroke/epidemiology
14.
Circ Res ; 97(8): e73-83, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16166556

ABSTRACT

High-mobility group box 1 protein (HMGB1) is a chromatin protein that is released by inflammatory and necrotic cells. Extracellular HMGB1 signals tissue damage, stimulates the secretion of proinflammatory cytokines and chemokines, and modulates stem cell function. The present study examined exogenous HMGB1 effect on mouse left-ventricular function and myocyte regeneration after infarction. Myocardial infarction was induced in C57BL/6 mice by permanent coronary artery ligation. After 4 hours animals were reoperated and 200 ng of purified HMGB1 was administered in the peri-infarcted left ventricle. This intervention resulted in the formation of new myocytes within the infarcted portion of the wall. The regenerative process involved the proliferation and differentiation of endogenous cardiac c-kit+ progenitor cells. Circulating c-kit+ cells did not significantly contribute to HMGB1-mediated cardiac regeneration. Echocardiographic and hemodynamic parameters at 1, 2, and 4 weeks demonstrated a significant recovery of cardiac performance in HMGB1-treated mice. These effects were not observed in infarcted hearts treated either with the unrelated protein glutathione S-transferase or a truncated form of HMGB1. Thus, HMGB1 appears to be a potent inducer of myocardial regeneration following myocardial infarction.


Subject(s)
HMGB1 Protein/pharmacology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-kit/analysis , Regeneration/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Connexin 43/analysis , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/physiology , Stem Cells/cytology , Stem Cells/drug effects , Ventricular Function, Left/drug effects
15.
Biochem Pharmacol ; 68(6): 1165-70, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15313414

ABSTRACT

High mobility group box 1 (HMGB1) is a non-histone protein required to maintain chromatin architecture. Recent observations demonstrated that HMGB1 can also act as a cytokine to regulate different biological processes such as inflammation, cell migration and metastasis. We showed previously that HMGB1 can be released passively by cells that die in a traumatic and unprogrammed way, and can serve a signal of tissue damage. More recently, we showed that HMGB1 can recruit stem cells: HMGB1 induces stem cell transmigration through an endothelial barrier; moreover, when beads containing HMGB1 are implanted into healthy muscle, they recruit stem cells injected into the general circulation. The inflammatory and tissue-regenerating roles of HMGB1 may be strictly interconnected, and are discussed here.


Subject(s)
Cell Movement/physiology , Cues , HMGB1 Protein/physiology , Stem Cells/cytology , Animals , Cytokines/physiology , Forecasting , Humans , Inflammation Mediators , Stem Cells/physiology
16.
J Cell Biol ; 164(3): 441-9, 2004 Feb 02.
Article in English | MEDLINE | ID: mdl-14744997

ABSTRACT

High mobility group box 1 (HMGB1) is an abundant chromatin protein that acts as a cytokine when released in the extracellular milieu by necrotic and inflammatory cells. Here, we show that extracellular HMGB1 and its receptor for advanced glycation end products (RAGE) induce both migration and proliferation of vessel-associated stem cells (mesoangioblasts), and thus may play a role in muscle tissue regeneration. In vitro, HMGB1 induces migration and proliferation of both adult and embryonic mesoangioblasts, and disrupts the barrier function of endothelial monolayers. In living mice, mesoangioblasts injected into the femoral artery migrate close to HMGB1-loaded heparin-Sepharose beads implanted in healthy muscle, but are unresponsive to control beads. Interestingly, alpha-sarcoglycan null dystrophic muscle contains elevated levels of HMGB1; however, mesoangioblasts migrate into dystrophic muscle even if their RAGE receptor is disabled. This implies that the HMGB1-RAGE interaction is sufficient, but not necessary, for mesoangioblast homing; a different pathway might coexist. Although the role of endogenous HMGB1 in the reconstruction of dystrophic muscle remains to be clarified, injected HMGB1 may be used to promote tissue regeneration.


Subject(s)
Cell Division/physiology , Cell Movement/physiology , Endothelium, Vascular/metabolism , HMGB1 Protein/metabolism , Stem Cells/physiology , Animals , Cattle , Cell Transplantation , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Embryo, Mammalian/physiology , Endothelium, Vascular/cytology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Regeneration/physiology , Sarcoglycans , Stem Cells/cytology
17.
Arterioscler Thromb Vasc Biol ; 22(3): 405-11, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11884282

ABSTRACT

In the present study, we analyzed the effect of conditioned media (CM) from bovine aortic endothelial cells exposed to laminar shear stress (SS) of 5 dyne/cm2 (SS5) or 15 dyne/cm2 (SS15) for 16 hours on smooth muscle cell (SMC) migration. In response to CM from bovine aortic endothelial cells exposed to SS5 (CMSS5) and SS15 (CMSS15), migration was 45 +/- 5.5 and 30 +/- 1.5 cells per field, respectively (P<0.05). Similar results were obtained with SS of 2 versus 20 dyne/cm2 and also when SS of 5 and 15 dyne/cm2 lasted 24 hours. Platelet-derived growth factor (PDGF)-AA levels in CMSS5 and CMSS15 were 9 +/- 7 and 18 +/- 5 ng/10(6) cells for 16 hours, respectively (P<0.05); PDGF-BB levels in CMSS5 and CMSS15 were 38 +/- 10 and 53 +/- 10 ng/10(6) cells for 16 hours, respectively (P<0.05). PDGF receptor alpha (PDGFRalpha) and PDGF receptor beta (PDGFRbeta) in SMCs were phosphorylated by CMSS15>CMSS5. In response to CMSS15, a neutralizing antibody against PDGF-AA enhanced SMC migration to a level comparable to that of CMSS5; in contrast, antibodies against PDGF-BB abolished SMC migration. Transfection of SMCs with a dominant-negative PDGFRalpha or PDGFRbeta increased or inhibited, respectively, SMC migration in response to CMSS15. Overexpression of wild-type PDGFRalpha inhibited SMC migration in response to CMSS5, CMSS15, or recombinant PDGF-BB (P<0.001). These results suggest that the ability of high SS to inhibit arterial wall thickening in vivo may be related to enhanced activation of PDGFRalpha in SMCs by PDGF isoforms secreted by the endothelium.


Subject(s)
Chemotaxis , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Platelet-Derived Growth Factor/biosynthesis , Animals , Becaplermin , Cattle , Culture Media, Conditioned/pharmacology , Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/drug effects , Mutation , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/physiology , Stress, Mechanical , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...