Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Rep ; 27(8): 2304-2312.e6, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31116977

ABSTRACT

Mechanisms that govern transcriptional regulation of inflammation in atherosclerosis remain largely unknown. Here, we identify the nuclear transcription factor c-Myb as an important mediator of atherosclerotic disease in mice. Atherosclerosis-prone animals fed a diet high in cholesterol exhibit increased levels of c-Myb in the bone marrow. Use of mice that either harbor a c-Myb hypomorphic allele or where c-Myb has been preferentially deleted in B cell lineages revealed that c-Myb potentiates atherosclerosis directly through its effects on B lymphocytes. Reduced c-Myb activity prevents the expansion of atherogenic B2 cells yet associates with increased numbers of IgM-producing antibody-secreting cells (IgM-ASCs) and elevated levels of atheroprotective oxidized low-density lipoprotein (OxLDL)-specific IgM antibodies. Transcriptional profiling revealed that c-Myb has a limited effect on B cell function but is integral in maintaining B cell progenitor populations in the bone marrow. Thus, targeted disruption of c-Myb beneficially modulates the complex biology of B cells in cardiovascular disease.


Subject(s)
Antibody-Producing Cells/immunology , Atherosclerosis/genetics , Atherosclerosis/immunology , Immunoglobulin M/metabolism , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/immunology , Animals , Antibody-Producing Cells/metabolism , Atherosclerosis/pathology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Genes, myb , Male , Mice
2.
Nat Immunol ; 17(2): 159-68, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26642357

ABSTRACT

Resident macrophages densely populate the normal arterial wall, yet their origins and the mechanisms that sustain them are poorly understood. Here we use gene-expression profiling to show that arterial macrophages constitute a distinct population among macrophages. Using multiple fate-mapping approaches, we show that arterial macrophages arise embryonically from CX3CR1(+) precursors and postnatally from bone marrow-derived monocytes that colonize the tissue immediately after birth. In adulthood, proliferation (rather than monocyte recruitment) sustains arterial macrophages in the steady state and after severe depletion following sepsis. After infection, arterial macrophages return rapidly to functional homeostasis. Finally, survival of resident arterial macrophages depends on a CX3CR1-CX3CL1 axis within the vascular niche.


Subject(s)
Cell Self Renewal , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/metabolism , Receptors, Chemokine/metabolism , Animals , CX3C Chemokine Receptor 1 , Cell Survival , Chemokine CX3CL1/metabolism , Cluster Analysis , Female , Gene Expression Profiling , Immunophenotyping , Macrophages/immunology , Macrophages/microbiology , Male , Mice , Mice, Transgenic , Phenotype , Protein Binding , Stem Cell Niche , Transcriptome
3.
Nat Med ; 19(9): 1166-72, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23933982

ABSTRACT

During the inflammatory response that drives atherogenesis, macrophages accumulate progressively in the expanding arterial wall. The observation that circulating monocytes give rise to lesional macrophages has reinforced the concept that monocyte infiltration dictates macrophage buildup. Recent work has indicated, however, that macrophage accumulation does not depend on monocyte recruitment in some inflammatory contexts. We therefore revisited the mechanism underlying macrophage accumulation in atherosclerosis. In murine atherosclerotic lesions, we found that macrophages turn over rapidly, after 4 weeks. Replenishment of macrophages in these experimental atheromata depends predominantly on local macrophage proliferation rather than monocyte influx. The microenvironment orchestrates macrophage proliferation through the involvement of scavenger receptor A (SR-A). Our study reveals macrophage proliferation as a key event in atherosclerosis and identifies macrophage self-renewal as a therapeutic target for cardiovascular disease.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Proliferation , Macrophages/physiology , Animals , Cells, Cultured , Inflammation , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/physiology
4.
PLoS One ; 6(12): e28457, 2011.
Article in English | MEDLINE | ID: mdl-22163019

ABSTRACT

BACKGROUND: Cigarette smoking is the main risk factor for the development of chronic obstructive pulmonary disease (COPD), a major cause of morbidity and mortality worldwide. Despite this, the cellular and molecular mechanisms that contribute to COPD pathogenesis are still poorly understood. METHODOLOGY AND PRINCIPAL FINDINGS: The objective of this study was to assess IL-1 α and ß expression in COPD patients and to investigate their respective roles in perpetuating cigarette smoke-induced inflammation. Functional studies were pursued in smoke-exposed mice using gene-deficient animals, as well as blocking antibodies for IL-1α and ß. Here, we demonstrate an underappreciated role for IL-1α expression in COPD. While a strong correlation existed between IL-1α and ß levels in patients during stable disease and periods of exacerbation, neutrophilic inflammation was shown to be IL-1α-dependent, and IL-1ß- and caspase-1-independent in a murine model of cigarette smoke exposure. As IL-1α was predominantly expressed by hematopoietic cells in COPD patients and in mice exposed to cigarette smoke, studies pursued in bone marrow chimeric mice demonstrated that the crosstalk between IL-1α+ hematopoietic cells and the IL-1R1+ epithelial cells regulates smoke-induced inflammation. IL-1α/IL-1R1-dependent activation of the airway epithelium also led to exacerbated inflammatory responses in H1N1 influenza virus infected smoke-exposed mice, a previously reported model of COPD exacerbation. CONCLUSIONS AND SIGNIFICANCE: This study provides compelling evidence that IL-1α is central to the initiation of smoke-induced neutrophilic inflammation and suggests that IL-1α/IL-1R1 targeted therapies may be relevant for limiting inflammation and exacerbations in COPD.


Subject(s)
Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin-1alpha/biosynthesis , Neutrophils/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Smoking , Animals , Biopsy , Caspase 1/metabolism , Humans , Inflammation , Interleukin-1beta/metabolism , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Smoke , Sputum/metabolism
5.
PLoS One ; 5(10): e13251, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20967263

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease is a progressive lung disease that is punctuated by periods of exacerbations (worsening of symptoms) that are attributable to viral infections. While rhinoviruses are most commonly isolated viruses during episodes of exacerbation, influenza viruses have the potential to become even more problematic with the increased likelihood of an epidemic. METHODOLOGY AND PRINCIPAL FINDINGS: This study examined the impact of current and potential pharmacological targets namely the systemic corticosteroid dexamethasone and the peroxisome proliferator-activated receptor-gamma agonist pioglitazone on the outcome of infection in smoke-exposed mice. C57BL/6 mice were exposed to room air or cigarette smoke for 4 days and subsequently inoculated with an H1N1 influenza A virus. Interventions were delivered daily during the course of infection. We show that smoke-exposed mice have an exacerbated inflammatory response following infection. While smoke exposure did not compromise viral clearance, precision cut lung slices from smoke-exposed mice showed greater expression of CC (MCP-1, -3), and CXC (KC, MIP-2, GCP-2) chemokines compared to controls when stimulated with a viral mimic or influenza A virus. While dexamethasone treatment partially attenuated the inflammatory response in the broncho-alveolar lavage of smoke-exposed, virally-infected animals, viral-induced neutrophilia was steroid insensitive. In contrast to controls, dexamethasone-treated smoke-exposed influenza-infected mice had a worsened health status. Pioglitazone treatment of virally-infected smoke-exposed mice proved more efficacious than the steroid intervention. Further mechanistic evaluation revealed that a deficiency in CCR2 did not improve the inflammatory outcome in smoke-exposed, virally-infected animals. CONCLUSIONS AND SIGNIFICANCE: This animal model of cigarette smoke and H1N1 influenza infection demonstrates that smoke-exposed animals are differentially primed to respond to viral insult. While providing a platform to test pharmacological interventions, this model demonstrates that treating viral exacerbations with alternative anti-inflammatory drugs, such as PPAR-gamma agonists should be further explored since they showed greater efficacy than systemic corticosteroids.


Subject(s)
Disease Models, Animal , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/complications , Pulmonary Disease, Chronic Obstructive/physiopathology , Smoking , Animals , Chemokines/metabolism , Humans , Influenza, Human/drug therapy , Influenza, Human/metabolism , Influenza, Human/virology , Mice , Mice, Inbred C57BL , PPAR gamma/agonists , Pioglitazone , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/virology , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use
6.
J Immunol ; 184(4): 2001-13, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20065113

ABSTRACT

Influenza virus infection is a leading cause of death and disability throughout the world. Influenza-infected hosts are vulnerable to secondary bacterial infection, however, and an ensuing bacterial pneumonia is actually the predominant cause of influenza-attributed deaths during pandemics. A number of mechanisms have been proposed by which influenza may predispose to superinfection with an unrelated or heterologous pathogen, but the subsequent interaction between the host, virus, and bacteria remains an understudied area. In this study, we develop and examine a novel model of heterologous pulmonary infection in which an otherwise subclinical Bordetella parapertussis infection synergizes with an influenza virus infection to yield a life-threatening secondary pneumonia. Despite a profound pulmonary inflammatory response and unaltered viral clearance, bacterial clearance was significantly impaired in heterologously infected mice. No deficits were observed in pulmonary or systemic adaptive immune responses or the viability or function of infiltrating inflammatory cells to explain this phenomenon, and we provide evidence that the onset of severe pulmonary inflammation actually precedes the increased bacterial burden, suggesting that exacerbated inflammation is independent of bacterial burden. To that end, neutralization of the ELR(+) inflammatory chemokine MIP-2 (CXCL2/GRO-beta) attenuated the inflammation, weight loss, and clinical presentation of heterologously infected mice without impacting bacterial burden. These data suggest that pulmonary inflammation, rather than pathogen burden, is the key threat during bacterial superinfection of influenza and that selective chemokine antagonists may be a novel therapeutic intervention in cases of bacterial superinfection of influenza.


Subject(s)
Bordetella Infections/immunology , Bordetella parapertussis/immunology , Chemokine CXCL2/biosynthesis , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Superinfection/immunology , Animals , Bordetella Infections/pathology , Bordetella Infections/therapy , Cell Line , Chemokine CXCL2/antagonists & inhibitors , Chemokine CXCL2/physiology , Dogs , Female , Gene Expression Regulation, Bacterial/immunology , Gene Expression Regulation, Viral/immunology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/therapy , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/virology , Pneumonia, Viral/immunology , Pneumonia, Viral/microbiology , Pneumonia, Viral/pathology , Receptors, Interleukin-8B/antagonists & inhibitors , Superinfection/microbiology , Superinfection/virology
7.
Am J Respir Crit Care Med ; 179(8): 666-75, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19179487

ABSTRACT

RATIONALE: The pathogenesis of chronic obstructive pulmonary disease is associated with acute episodes of bacterial exacerbations. The most commonly isolated bacteria during episodes of exacerbation is nontypeable Haemophilus influenzae (NTHI). OBJECTIVES: In this study, we investigated the in vivo consequences of cigarette smoke exposure on the inflammatory response to an NTHI challenge. METHODS: C57BL/6 and BALB/c mice were exposed to cigarette smoke for 8 weeks and subsequently challenged intranasally with NTHI. MEASUREMENTS AND MAIN RESULTS: We observed increased pulmonary inflammation and lung damage in cigarette smoke-exposed NTHI-challenged mice as compared with control NTHI-challenged mice. Furthermore, although NTHI challenge in control mice was marked by increases in tumor necrosis factor-alpha, IL-6, MIP-2, and KC/GROalpha, NTHI challenge in cigarette smoke-exposed mice led to a prominent up-regulation of a different subset of inflammatory mediators, most notably MCP-1, -3, and -5, IP-10, and MIP-1gamma. This skewed inflammatory mediator expression was also observed after ex vivo NTHI stimulation of alveolar macrophages, signifying their importance to this altered response. Importantly, corticosteroids attenuated inflammation after NTHI challenge in both cigarette smoke-exposed and control mice; however, this was associated with significantly increased bacterial burden. CONCLUSIONS: Collectively, these data suggest that cigarette smoke exacerbates the inflammatory response to a bacterial challenge via skewed inflammatory mediator expression.


Subject(s)
Haemophilus influenzae/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Tobacco Smoke Pollution/adverse effects , Administration, Intranasal , Animals , Disease Progression , Female , Inflammation/etiology , Inflammation/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
8.
Cell Immunol ; 253(1-2): 38-44, 2008.
Article in English | MEDLINE | ID: mdl-18533139

ABSTRACT

Although its direct effects cannot be discounted, tobacco's effects on the immune system have been proposed to play a key role in mediating its deleterious health impact. Studies in rats using high levels of smoke exposure have suggested that tobacco smoke exhausts cellular signal transduction cascades, making lymphocytes unresponsive to stimulation. In the present study, we show that purified B or T cells, and total lymphocytes from the lungs, lymph nodes and spleens of smoke-exposed mice fluxed calcium, proliferated, and secreted immunoglobulin or IFN-gamma similarly to control mice when stimulated with ligands including anti-IgM, and anti-CD3. Importantly, we recapitulated these findings in PBMCs from human smokers; cells from long-term smokers and never-smokers proliferated equivalently when stimulated ex vivo. Previous reports of lymphocyte unresponsiveness in rats are inconsistent with these findings, and may reflect a phenomenon observed only at levels of smoke exposure well above those seen in actual human smokers.


Subject(s)
B-Lymphocytes/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Subsets/drug effects , Nicotiana/adverse effects , Smoke/adverse effects , T-Lymphocytes/drug effects , Animals , B-Lymphocytes/immunology , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Lung/cytology , Lung/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Rats , Smoking/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Nicotiana/immunology
9.
J Interferon Cytokine Res ; 28(3): 167-79, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18338949

ABSTRACT

The objective of this study was to investigate the impact of cigarette smoke on innate antiviral defense mechanisms; specifically, we examined the effects of cigarette smoke on the induction of type I interferon (IFN). We observed a dose-dependent decrease in the ability of human lung fibroblast and epithelial cells to elicit an antiviral response against a viral double-strand RNA (dsRNA) mimic, polyI:C, in the presence of cigarette smoke-conditioned medium (SCM). Mechanistically, SCM decreases the expression of IFN-stimulated gene 15 (ISG15) and IFN regulatory factor-7 (IRF-7) transcripts and suppresses the nuclear translocation of key transcription factors, nuclear factor-kappaB (NF-kappaB) and IRF-3, after polyI:C stimulation. Furthermore, we provide evidence that the intercellular defense strategy against viral infection is also impaired. We observed a decrease in the ability of fibroblasts to elicit an antiviral state in response to IFN-beta stimulation. This was associated with decreased nuclear translocation of phosphorylated Stat1 in response to IFN-beta treatment. The effects elicited by SCM are reversible and are almost entirely abrogated in the presence of an antioxidant, such as glutathione. Our findings suggest that cigarette smoke affects the immediate-early, inductive, and amplification phases of the type I IFN response.


Subject(s)
Epithelial Cells/immunology , Fibroblasts/immunology , Interferon Type I/immunology , Smoke/adverse effects , Antiviral Agents/immunology , Antiviral Agents/metabolism , Cell Line , Cell Nucleus/metabolism , Culture Media, Conditioned , Cytokines/metabolism , Epithelial Cells/metabolism , Fibroblasts/metabolism , Glutathione/pharmacology , Humans , Immunity, Innate , Interferon Inducers/pharmacology , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Type I/metabolism , Lung/immunology , Lung/metabolism , Poly I-C/pharmacology , Protein Serine-Threonine Kinases/metabolism , STAT1 Transcription Factor/metabolism , Nicotiana , Ubiquitins/metabolism , NF-kappaB-Inducing Kinase
10.
Am J Respir Cell Mol Biol ; 38(2): 218-26, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17872497

ABSTRACT

Alveolar macrophages (aMs) play a central role in respiratory host defense by sensing microbial antigens and initiating immune-inflammatory responses early in the course of an infection. The purpose of this study was to investigate the effect of cigarette smoke exposure on aMs after stimulation of innate pattern recognition receptors (PRRs) in a murine model. To accomplish this, C57BL/6 mice were exposed for 8 weeks using two models of cigarette smoke exposure, nose-only or whole-body exposure, and aMs isolated from the bronchoalveolar lavage. After stimulation of aMs with pI:C, a mimic of viral replication, and bacterial cell-wall constituent LPS, aMs from cigarette smoke-exposed mice produced significantly attenuated levels of the inflammatory cytokines TNF-alpha and IL-6, and the chemokine RANTES. This attenuation was specific to the aM compartment, and not related to changes in aM viability or expression of Toll-like receptor (TLR)3 or TLR4 between groups. Furthermore, aMs from smoke-exposed mice had decreased cytokine RNA as compared with aMs from sham-exposed mice. Mechanistically, this was associated with decreased nuclear translocation of the proinflammatory transcription factor NF-kappaB, and increased activator protein-1 nuclear translocation, in aMs from smoke-exposed mice. Attenuated cytokine production was reversible after smoking cessation. Cigarette smoke exposure also attenuated TNF-alpha production after stimulation with nucleotide-oligomerization domain-like receptor agonists, showing that the effect applies more broadly to other PRR pathways. Our data demonstrate that cigarette smoke exposure attenuates aM responses after innate stimulation, including pathways typically associated with bacterial and viral infections.


Subject(s)
Cytokines/biosynthesis , Macrophages, Alveolar/metabolism , Nicotiana , Smoke/adverse effects , Animals , Carboxyhemoglobin/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Pulmonary Disease, Chronic Obstructive/etiology , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors/metabolism
11.
Contrib Microbiol ; 14: 126-141, 2007.
Article in English | MEDLINE | ID: mdl-17684337

ABSTRACT

Modeling acute exacerbations of chronic obstructive pulmonary disease (AECOPD) in animals has proven challenging due to the clinical and pathological complexity of the underlying disease. This has hindered the progress in understanding the cellular and molecular mechanisms that lie beneath AECOPD. In this chapter, we will address modeling possibilities of AECOPD that may be drawn from the current knowledge of factors that cause exacerbations. Importantly, since it is widely accepted that the most common causes of AECOPD are viral and bacterial infections, animal models of AECOPD should incorporate both the causative agents of exacerbation: viruses and bacteria. However, other factors that are also believed to determine both progression of COPD, as well as the frequency and severity of AECOPD, such as proteolytic enzymes, cigarette smoke or other noxious stimuli must also be considered. Such animal models will provide mechanistic insight into the etiology of AECOPD and will prove invaluable in furthering our understanding of key events in disease pathogenesis.


Subject(s)
Disease Models, Animal , Pulmonary Disease, Chronic Obstructive/etiology , Animals , Humans , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Pulmonary Disease, Chronic Obstructive/virology
12.
J Immunol ; 178(2): 936-43, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17202355

ABSTRACT

In this study, we investigated the impact of cigarette smoke on tumor immune surveillance and its consequences to lung tumor burden in a murine lung metastasis model. Cigarette smoke exposure significantly increased the numbers of lung metastases following B16-MO5 melanoma challenge. This effect was reversible; we observed significantly fewer tumor nodules following smoking cessation. Using RAG2(-/-) and RAG2(-/-)gamma(c)(-/-) mice, we provide strong evidence that increased tumor incidence was NK cell dependent. Furthermore, we show that cigarette smoke suppressed NK activation and attenuated NK CTL activity, without apparent effect on activating or inhibitory receptor expression. Finally, activation of NK cells through bone marrow-derived dendritic cells conferred protection against lung metastases in smoke-exposed mice; however, protection was not as efficacious as in sham-exposed mice. To our knowledge, this is the first experimental evidence showing that cigarette smoke impairs NK cell-dependent tumor immune surveillance and that altered immunity is associated with increased tumor burden. Our findings suggest that altered innate immunity may contribute to the increased risk of cancer in smokers.


Subject(s)
Immunologic Surveillance/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Nicotiana/adverse effects , Smoke/adverse effects , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/immunology , Female , Immunologic Surveillance/drug effects , Interleukin-12/deficiency , Interleukin-12/genetics , Interleukin-12/metabolism , Lectins, C-Type , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lymphocyte Activation/immunology , Mice , Mice, Knockout , T-Lymphocytes, Cytotoxic/immunology
13.
Purinergic Signal ; 2(4): 651-61, 2006 Nov.
Article in English | MEDLINE | ID: mdl-18404468

ABSTRACT

Wound healing is a complex sequence of cellular and molecular processes that involves multiple cell types and biochemical mediators. Several growth factors have been identified that regulate tissue repair, including the neurotrophin nerve growth factor (NGF). As non-adenine based purines (NABPs) are known to promote cell proliferation and the release of growth factors, we investigated whether NABPs had an effect on wound healing. Full-thickness, excisional wound healing in healthy BALB/c mice was significantly accelerated by daily topical application of NABPs such as guanosine (50% closure by days 2.5-2.8). Co-treatment of wounds with guanosine plus anti-NGF reversed the guanosine-promoted acceleration of wound healing, indicating that this effect of guanosine is mediated, at least in part, by NGF. Selective inhibitors of the NGF-inducible serine/threonine protein kinase (protein kinase N), such as 6-methylmercaptopurine riboside abolished the acceleration of wound healing caused by guanosine, confirming that activation of this enzyme is required for this effect of guanosine. Treatment of genetically diabetic BKS.Cg-m+/+lepr db mice, which display impaired wound healing, with guanosine led to accelerated healing of skin wounds (25% closure by days 2.8-3.0). These results provide further confirmation that the NABP-mediated acceleration of cutaneous wound healing is mediated via an NGF-dependent mechanism. Thus, NABPs may offer an alternative and viable approach for the treatment of wounds in a clinical setting.

SELECTION OF CITATIONS
SEARCH DETAIL
...