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










Publication year range
1.
J Biomed Opt ; 27(8)2022 08.
Article in English | MEDLINE | ID: mdl-35982528

ABSTRACT

SIGNIFICANCE: Morphological changes in the epidermis layer are critical for the diagnosis and assessment of various skin diseases. Due to its noninvasiveness, optical coherence tomography (OCT) is a good candidate for observing microstructural changes in skin. Convolutional neural network (CNN) has been successfully used for automated segmentation of the skin layers of OCT images to provide an objective evaluation of skin disorders. Such method is reliable, provided that a large amount of labeled data is available, which is very time-consuming and tedious. The scarcity of patient data also puts another layer of difficulty to make the model more generalizable. AIM: We developed a semisupervised representation learning method to provide data augmentations. APPROACH: We used rodent models to train neural networks for accurate segmentation of clinical data. RESULT: The learning quality is maintained with only one OCT labeled image per volume that is acquired from patients. Data augmentation introduces a semantically meaningful variance, allowing for better generalization. Our experiments demonstrate the proposed method can achieve accurate segmentation and thickness measurement of the epidermis. CONCLUSION: This is the first report of semisupervised representative learning applied to OCT images from clinical data by making full use of the data acquired from rodent models. The proposed method promises to aid in the clinical assessment and treatment planning of skin diseases.


Subject(s)
Algorithms , Tomography, Optical Coherence , Animals , Epidermis/diagnostic imaging , Humans , Research Subjects , Rodentia , Tomography, Optical Coherence/methods
2.
J Biomed Opt ; 27(1)2022 01.
Article in English | MEDLINE | ID: mdl-35043611

ABSTRACT

SIGNIFICANCE: In order to elucidate therapeutic treatment to accelerate wound healing, it is crucial to understand the process underlying skin wound healing, especially re-epithelialization. Epidermis and scab detection is of importance in the wound healing process as their thickness is a vital indicator to judge whether the re-epithelialization process is normal or not. Since optical coherence tomography (OCT) is a real-time and non-invasive imaging technique that can perform a cross-sectional evaluation of tissue microstructure, it is an ideal imaging modality to monitor the thickness change of epidermal and scab tissues during wound healing processes in micron-level resolution. Traditional segmentation on epidermal and scab regions was performed manually, which is time-consuming and impractical in real time. AIM: We aim to develop a deep-learning-based skin layer segmentation method for automated quantitative assessment of the thickness of in vivo epidermis and scab tissues during a time course of healing within a rodent model. APPROACH: Five convolution neural networks were trained using manually labeled epidermis and scab regions segmentation from 1000 OCT B-scan images (assisted by its corresponding angiographic information). The segmentation performance of five segmentation architectures was compared qualitatively and quantitatively for validation set. RESULTS: Our results show higher accuracy and higher speed of the calculated thickness compared with human experts. The U-Net architecture represents a better performance than other deep neural network architectures with 0.894 at F1-score, 0.875 at mean intersection over union, 0.933 at Dice similarity coefficient, and 18.28 µm at an average symmetric surface distance. Furthermore, our algorithm is able to provide abundant quantitative parameters of the wound based on its corresponding thickness maps in different healing phases. Among them, normalized epidermal thickness is recommended as an essential hallmark to describe the re-epithelialization process of the rodent model. CONCLUSIONS: The automatic segmentation and thickness measurements within different phases of wound healing data demonstrates that our pipeline provides a robust, quantitative, and accurate method for serving as a standard model for further research into effect of external pharmacological and physical factors.


Subject(s)
Deep Learning , Tomography, Optical Coherence , Cross-Sectional Studies , Epidermis/diagnostic imaging , Neural Networks, Computer
3.
J Clin Invest ; 130(6): 3221-3237, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32191647

ABSTRACT

Neutrophilic inflammation is central to disease pathogenesis, for example, in chronic obstructive pulmonary disease, yet the mechanisms that retain neutrophils within tissues remain poorly understood. With emerging evidence that axon guidance factors can regulate myeloid recruitment and that neutrophils can regulate expression of a class 3 semaphorin, SEMA3F, we investigated the role of SEMA3F in inflammatory cell retention within inflamed tissues. We observed that neutrophils upregulate SEMA3F in response to proinflammatory mediators and following neutrophil recruitment to the inflamed lung. In both zebrafish tail injury and murine acute lung injury models of neutrophilic inflammation, overexpression of SEMA3F delayed inflammation resolution with slower neutrophil migratory speeds and retention of neutrophils within the tissues. Conversely, constitutive loss of sema3f accelerated egress of neutrophils from the tail injury site in fish, whereas neutrophil-specific deletion of Sema3f in mice resulted in more rapid neutrophil transit through the airways, and significantly reduced time to resolution of the neutrophilic response. Study of filamentous-actin (F-actin) subsequently showed that SEMA3F-mediated retention is associated with F-actin disassembly. In conclusion, SEMA3F signaling actively regulates neutrophil retention within the injured tissues with consequences for neutrophil clearance and inflammation resolution.


Subject(s)
Cell Movement/immunology , Membrane Proteins/immunology , Nerve Tissue Proteins/immunology , Neutrophils/immunology , Signal Transduction/immunology , Zebrafish Proteins/immunology , Zebrafish/immunology , Animals , Humans , Inflammation/immunology , Inflammation/pathology , Mice , Neutrophils/pathology , Up-Regulation/immunology
4.
Microbiol Spectr ; 4(3)2016 06.
Article in English | MEDLINE | ID: mdl-27337466

ABSTRACT

Cutaneous wound repair is a complex, dynamic process with the goal of rapidly sealing any breach in the skin's protective barrier. Myeloid cells compose a significant proportion of the inflammatory cells recruited to a wound site and play important roles in decontaminating the injured tissue of any invading microorganisms. Subsequently, myeloid cells are able to influence many aspects of the healing response, in part through their capacity to release a large array of signaling molecules that allow them to communicate with and regulate the behavior of other wound cells and in turn, be themselves exquisitely regulated by the wound microenvironment. Macrophages, for example, appear to play important, temporally changing roles in the initiation of scarring and subsequently in matrix remodeling to resolve fibrosis. In this way, myeloid cells seem to play both positive (e.g., pathogen killing and matrix remodeling) and negative (e.g., scarring) roles in wound repair. Further research is of course needed to elucidate the precise temporal and spatial myeloid cell phenotypes and behaviors and ultimately to design effective strategies to optimize the beneficial functions of these cells while minimizing their detrimental contributions to improve wound healing in the clinic.


Subject(s)
Cicatrix , Myeloid Cells/physiology , Skin/injuries , Wound Healing , Animals , Humans
5.
Drug Discov Today ; 19(8): 1186-92, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24993159

ABSTRACT

There is a growing appreciation of the important role of resolution mediators in the successful termination of the inflammatory response. Here, we discuss the potential importance of the lipid and peptide proresolving mediators, in particular the resolvins and chemerin-derived peptides, which mediate their effects through specific G protein-coupled receptors (GPCRs).


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Lipids/genetics , Peptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Drug Delivery Systems/methods , Humans , Inflammation/genetics , Peptides/genetics , Receptors, G-Protein-Coupled/genetics
6.
Curr Biol ; 24(12): 1406-1414, 2014 06 16.
Article in English | MEDLINE | ID: mdl-24881877

ABSTRACT

Disorders of cutaneous repair can cause disability or death given that skin functions as a protective barrier against the external environment. The inflammatory response triggered by tissue damage is thought to play both positive (e.g., pathogen-killing) and negative (e.g., scarring) roles in repair. Inflammatory resolution mediators such as chemerin15 (C15) control the magnitude and duration of the inflammatory response; however, their role in wound repair and scarring is unknown. Here, we show that the C15 precursor, chemerin, and its receptor, ChemR23, are both upregulated after skin damage and that the receptor is expressed by macrophages, neutrophils, and keratinocytes. Dynamic live-imaging studies of murine cutaneous wounds demonstrate that C15 delivery dampens the immediate intravascular inflammatory events, including platelet adhesion to neutrophils, an important event in driving leukocyte recruitment. C15 administration indirectly accelerates wound closure while altering fibroblast-mediated collagen deposition and alignment to reduce scarring. Macrophage recruitment is restricted to the immediate wound site rather than spilling extensively into the adjacent tissue as in control wounds, and macrophage phenotype in C15-treated wounds is skewed toward a less inflammatory phenotype with reduced iNOS, increased Arginase-1, and lower wound tumor necrosis factor α (TNF-α) expression. Modulation of inflammatory resolution pathways in acute and chronic wounds may therefore provide a novel therapeutic avenue to improve repair and reduce scarring.


Subject(s)
Chemokines/genetics , Chemotactic Factors/genetics , Intercellular Signaling Peptides and Proteins/genetics , Peptide Fragments/genetics , Receptors, G-Protein-Coupled/genetics , Wound Healing , Animals , Chemokines/metabolism , Chemotactic Factors/metabolism , Cicatrix , Intercellular Signaling Peptides and Proteins/metabolism , Keratinocytes/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Peptide Fragments/metabolism , Receptors, G-Protein-Coupled/metabolism , Skin
8.
EMBO Rep ; 14(11): 999-1007, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23999103

ABSTRACT

Neutrophil activation and adhesion must be tightly controlled to prevent complications associated with excessive inflammatory responses. The role of the anti-inflammatory peptide chemerin15 (C15) and the receptor ChemR23 in neutrophil physiology is unknown. Here, we report that ChemR23 is expressed in neutrophil granules and rapidly upregulated upon neutrophil activation. C15 inhibits integrin activation and clustering, reducing neutrophil adhesion and chemotaxis in vitro. In the inflamed microvasculature, C15 rapidly modulates neutrophil physiology inducing adherent cell detachment from the inflamed endothelium, while reducing neutrophil recruitment and heart damage in a murine myocardial infarction model. These effects are mediated through ChemR23. We identify the C15/ChemR23 pathway as a new regulator and thus therapeutic target in neutrophil-driven pathologies.


Subject(s)
Blood Vessels/pathology , Chemotactic Factors/pharmacology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/pharmacology , Myocardial Reperfusion Injury/pathology , Neutrophils/pathology , Peptide Fragments/pharmacology , Peptides/pharmacology , Receptors, Chemokine/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blood Vessels/drug effects , Blood Vessels/metabolism , Cell Communication/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Microvessels/drug effects , Microvessels/metabolism , Microvessels/pathology , Myocardial Reperfusion Injury/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Up-Regulation/drug effects
9.
J Immunol ; 184(9): 5315-24, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20363975

ABSTRACT

Chemerin peptides represent a recently identified component of the endogenous anti-inflammatory network that act via the G protein-coupled receptor ChemR23. The role of the chemerin peptide/ChemR23 pathway in phagocytosis, the clearance of apoptotic cells (efferocytosis), and the resolution of inflammation is unknown. In this article, we report that low picomolar concentrations of the chemerin peptide chemerin15 (C15) enhance macrophage (MPhi) phagocytosis of microbial particles and apoptotic cells by up to 360% in vitro. These prophagocytic effects of C15 are significantly impaired in ChemR23(-/-) MPhis and are associated with increased actin polymerization and localization of F-actin to the phagocytic cup. Importantly, pharmacological inhibition of Syk activity completely abrogates the prophagocytic activities of C15 and associated changes in actin polymerization and phagocytic cup formation, suggesting that C15 promotes phagocytosis by facilitating phagocytic cup development in a Syk-dependent manner. During peritoneal inflammation, C15 administration (8 pg/mouse) enhances microbial particle clearance and apoptotic neutrophil ingestion by MPhis in wild-type but not ChemR23(-/-) mice, such that levels of apoptotic and necrotic cells at the inflammatory site are profoundly reduced. In contrast, neutralization of endogenous chemerin species during peritoneal inflammation significantly impairs MPhi ingestion of apoptotic neutrophils and zymosan. Our data identify a key role of the chemerin peptide/ChemR23 axis in the efficient clearance of foreign material, efferocytosis, and, hence, the resolution of inflammation. Manipulation of the chemerin peptide/ChemR23 axis may represent a novel therapeutic approach for the treatment of inflammatory pathologies, especially if failure to efficiently clear phagocytic targets has been implicated in their pathogenesis.


Subject(s)
Chemotactic Factors/physiology , Intercellular Signaling Peptides and Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Phagocytosis/immunology , Protein-Tyrosine Kinases/physiology , Receptors, G-Protein-Coupled/physiology , Amino Acid Sequence , Animals , Apoptosis/immunology , Cells, Cultured , Chemokines , Chemotactic Factors/deficiency , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Ligands , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Peptide Fragments/deficiency , Peptide Fragments/genetics , Peptide Fragments/physiology , Peptides/deficiency , Peptides/genetics , Peptides/physiology , Phagosomes/enzymology , Phagosomes/immunology , Phenotype , Receptors, Chemokine , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Syk Kinase
10.
Methods Enzymol ; 461: 379-96, 2009.
Article in English | MEDLINE | ID: mdl-19480928

ABSTRACT

The acute inflammatory response occurs as a result of tissue injury or infection and is characterized by the coordinated recruitment of leukocytes in response to inflammatory mediators including chemokines. This process generally resolves within a matter of days, and normal tissue architecture is restored by a process of wound healing. Failure to resolve the injury can result in chronic inflammation. Much of our understanding of the specific mediators and cell types involved in acute inflammation has come from sterile peritonitis models. The injection of a wide range of irritants into the peritoneal cavity induces the hallmarks of inflammation, including pain, leukocyte infiltration, and synthesis of inflammatory mediators. Intraperitoneal injection of zymosan, a polysaccharide cell wall component derived from Saccharomyces cerevisiae, has been widely used as a self-resolving model of acute inflammation that peaks within a few hours and is cleared within 48 to 72 h. We have used the zymosan-induced peritonitis model extensively to quantify the recruitment of monocytes and neutrophils into the peritoneal cavity and to study the effects of existing and novel antiinflammatory drugs. We discuss some of the applications and advantages of the zymosan-induced peritonitis model and describe the method for analysis of leukocyte recruitment and inflammatory mediator production in response to zymosan.


Subject(s)
Disease Models, Animal , Inflammation/chemically induced , Peritonitis/chemically induced , Peritonitis/immunology , Zymosan/pharmacology , Animals , Mice , Rats
11.
J Exp Med ; 205(4): 767-75, 2008 Apr 14.
Article in English | MEDLINE | ID: mdl-18391062

ABSTRACT

Chemerin is a chemotactic protein that binds to the G protein-coupled receptor, ChemR23. We demonstrate that murine chemerin possesses potent antiinflammatory properties that are absolutely dependent on proteolytic processing. A series of peptides was designed, and only those identical to specific C-terminal chemerin sequences exerted antiinflammatory effects at picomolar concentrations in vitro. One of these, chemerin15 (C15; A(140)-A(154)), inhibited macrophage (MPhi) activation to a similar extent as proteolyzed chemerin, but exhibited reduced activity as a MPhi chemoattractant. Intraperitoneal administration of C15 (0.32 ng/kg) to mice before zymosan challenge conferred significant protection against zymosan-induced peritonitis, suppressing neutrophil (63%) and monocyte (62%) recruitment with a concomitant reduction in proinflammatory mediator expression. Importantly, C15 was unable to ameliorate zymosan-induced peritonitis in ChemR23(-/-) mice, demonstrating that C15's antiinflammatory effects are entirely ChemR23 dependent. In addition, administration of neutralizing anti-chemerin antibody before zymosan challenge resulted in a significant exacerbation of peritoneal inflammation (up to 170%), suggesting an important endogenous antiinflammatory role for chemerin-derived species. Collectively, these results show that chemerin-derived peptides may represent a novel therapeutic strategy for the treatment of inflammatory diseases through ChemR23.


Subject(s)
Chemotactic Factors/pharmacology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/pharmacology , Peptides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Antibodies/pharmacology , Chemokines , Chemotactic Factors/therapeutic use , Chemotaxis/drug effects , Inflammation/drug therapy , Intercellular Signaling Peptides and Proteins/therapeutic use , Macrophage Activation/drug effects , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Mice , Mice, Inbred C57BL , Neutralization Tests , Peritonitis/pathology , Protein Processing, Post-Translational/drug effects , Receptors, Chemokine , Receptors, G-Protein-Coupled/deficiency , Zymosan
12.
Gastroenterology ; 133(4): 1219-28, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17919496

ABSTRACT

BACKGROUND & AIMS: We previously showed that intestinal inflammation is reduced by electrical stimulation of the efferent vagus nerve, which prevents postoperative ileus in mice. We propose that this cholinergic anti-inflammatory pathway is mediated via alpha7 nicotinic acetylcholine receptors expressed on macrophages. The aim of this study was to evaluate pharmacologic activation of the cholinergic anti-inflammatory pathway in a mouse model for postoperative ileus using the alpha7 nicotinic acetylcholine receptor-agonist AR-R17779. METHODS: Mice were pretreated with vehicle, nicotine, or AR-R17779 20 minutes before a laparotomy (L) or intestinal manipulation (IM). Twenty-four hours thereafter gastric emptying was determined using scintigraphy and intestinal muscle inflammation was quantified. Nuclear factor-kappaB transcriptional activity and cytokine production was assayed in peritoneal macrophages. RESULTS: Twenty-four hours after surgery IM led to a delayed gastric emptying compared with L (gastric retention: L(saline) 14% +/- 4% vs IM(saline) 38% +/- 10%, P = .04). Pretreatment with AR-R17779 prevented delayed gastric emptying (IM(AR-R17779) 15% +/- 4%, P = .03). IM elicited inflammatory cell recruitment (L(saline) 50 +/- 8 vs IM(saline) 434 +/- 71 cells/mm(2), P = .001) which was reduced by AR-R17779 pretreatment (IM(AR-R17779) 231 +/- 32 cells/mm(2), P = .04). An equimolar dose of nicotine was not tolerated. Subdiaphragmal vagotomy did not affect the anti-inflammatory properties of AR-R17779. In peritoneal macrophages, both nicotinic agonists reduced nuclear factor kappaB transcriptional activity and proinflammatory cytokine production, with nicotine being more effective than AR-R17779. CONCLUSIONS: AR-R17779 treatment potently prevents postoperative ileus, whereas toxicity limits nicotine administration to ineffective doses. Our data further imply that nicotinic inhibition of macrophage activation may involve other receptors in addition to alpha7 nicotinic acetylcholine receptor.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bridged-Ring Compounds/pharmacology , Gastroenteritis/prevention & control , Ileus/prevention & control , Intestines/drug effects , Macrophages, Peritoneal/drug effects , Nicotinic Agonists/pharmacology , Postoperative Complications/prevention & control , Spiro Compounds/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/toxicity , Bridged-Ring Compounds/therapeutic use , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation Therapy , Female , Gastric Emptying/drug effects , Gastroenteritis/metabolism , Gastroenteritis/physiopathology , Ileus/metabolism , Ileus/physiopathology , Intestines/innervation , Intestines/physiopathology , Intestines/surgery , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Nicotine/pharmacology , Nicotine/toxicity , Nicotinic Agonists/therapeutic use , Nicotinic Agonists/toxicity , Postoperative Complications/metabolism , Postoperative Complications/physiopathology , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Spiro Compounds/therapeutic use , Transcription, Genetic/drug effects , Vagotomy , Vagus Nerve/surgery , alpha7 Nicotinic Acetylcholine Receptor
13.
J Immunol ; 177(8): 5567-73, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015744

ABSTRACT

CC chemokines mediate mononuclear cell recruitment and activation in chronic inflammation. We have shown previously that gene transfer using recombinant adenoviruses, encoding a soluble CC chemokine-binding protein of vaccinia virus 35K, can dramatically reduce atherosclerosis and vein graft remodeling in apolipoprotein E knockout mice. In this study, we report the development of a membrane-bound form of 35K (m35K), tagged with GFP, which allows for localized, broad-spectrum CC chemokine blockade. In vitro experiments indicate that m35K-expressing cells no longer undergo CC chemokine-induced chemotaxis, and m35K-expressing cells can locally deplete the CC chemokines RANTES (CCL5) and MIP-1alpha (CCL3) from supernatant medium. This sequestration of CC chemokines can prevent chemotaxis of bystander cells to CC, but not CX(3)C chemokines. Intraperitoneal injection of mice with an adenovirus-encoding m35K leads to a significant (44%) decrease in leukocyte recruitment into the peritoneal cavity in a sterile peritonitis model. Intravenous adenovirus-encoding m35K delivery leads to m35K expression in hepatocytes, which confers significant protection against liver damage (75% reduction in liver enzymes) in a Con A-induced hepatitis model. In summary, we have generated a membrane-bound CC chemokine-binding protein (m35K) that provides localized broad-spectrum CC chemokine inhibition in vitro and in vivo. m35K may be a useful tool to study the role of CC chemokines in leukocyte trafficking and block the recruitment of monocytes in chronic inflammation.


Subject(s)
Chemokines, CC/agonists , Viral Envelope Proteins/pharmacology , Animals , Chemotaxis/drug effects , Disease Models, Animal , Green Fluorescent Proteins/genetics , Hepatitis/pathology , Hepatitis/therapy , Membrane Proteins , Mice , Peritonitis/therapy , Vaccinia virus/chemistry , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...