Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.423
1.
Nature ; 629(8010): 174-183, 2024 May.
Article En | MEDLINE | ID: mdl-38693412

Regular exercise promotes whole-body health and prevents disease, but the underlying molecular mechanisms are incompletely understood1-3. Here, the Molecular Transducers of Physical Activity Consortium4 profiled the temporal transcriptome, proteome, metabolome, lipidome, phosphoproteome, acetylproteome, ubiquitylproteome, epigenome and immunome in whole blood, plasma and 18 solid tissues in male and female Rattus norvegicus over eight weeks of endurance exercise training. The resulting data compendium encompasses 9,466 assays across 19 tissues, 25 molecular platforms and 4 training time points. Thousands of shared and tissue-specific molecular alterations were identified, with sex differences found in multiple tissues. Temporal multi-omic and multi-tissue analyses revealed expansive biological insights into the adaptive responses to endurance training, including widespread regulation of immune, metabolic, stress response and mitochondrial pathways. Many changes were relevant to human health, including non-alcoholic fatty liver disease, inflammatory bowel disease, cardiovascular health and tissue injury and recovery. The data and analyses presented in this study will serve as valuable resources for understanding and exploring the multi-tissue molecular effects of endurance training and are provided in a public repository ( https://motrpac-data.org/ ).


Endurance Training , Multiomics , Physical Conditioning, Animal , Physical Endurance , Animals , Female , Humans , Male , Rats , Acetylation , Blood/immunology , Blood/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Databases, Factual , Epigenome , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Internet , Lipidomics , Metabolome , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Organ Specificity/genetics , Organ Specificity/immunology , Organ Specificity/physiology , Phosphorylation , Physical Conditioning, Animal/physiology , Physical Endurance/genetics , Physical Endurance/physiology , Proteome/metabolism , Proteomics , Time Factors , Transcriptome/genetics , Ubiquitination , Wounds and Injuries/genetics , Wounds and Injuries/immunology , Wounds and Injuries/metabolism
2.
Cell Death Dis ; 15(4): 285, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38653969

Despite advances in the treatment and care of severe physical injuries, trauma remains one of the main reasons for disability-adjusted life years worldwide. Trauma patients often suffer from disturbances in energy utilization and metabolic dysfunction, including hyperglycemia and increased insulin resistance. White adipose tissue plays an essential role in the regulation of energy homeostasis and is frequently implicated in traumatic injury due to its ubiquitous body distribution but remains poorly studied. Initial triggers of the trauma response are mainly damage-associated molecular patterns (DAMPs) such as histones. We hypothesized that DAMP-induced adipose tissue inflammation contributes to metabolic dysfunction in trauma patients. Therefore, we investigated whether histone release during traumatic injury affects adipose tissue. Making use of a murine polytrauma model with hemorrhagic shock, we found increased serum levels of histones accompanied by an inflammatory response in white adipose tissue. In vitro, extracellular histones induced an inflammatory response in human adipocytes. On the molecular level, this inflammatory response was mediated via a MYD88-IRAK1-ERK signaling axis as demonstrated by pharmacological and genetic inhibition. Histones also induced lytic cell death executed independently of caspases and RIPK1 activity. Importantly, we detected increased histone levels in the bloodstream of patients after polytrauma. Such patients might benefit from a therapy consisting of activated protein C and the FDA-approved ERK inhibitor trametinib, as this combination effectively prevented histone-mediated effects on both, inflammatory gene activation and cell death in adipocytes. Preventing adipose tissue inflammation and adipocyte death in patients with polytrauma could help minimize posttraumatic metabolic dysfunction.


Adipocytes , Histones , Inflammation , Myeloid Differentiation Factor 88 , Humans , Animals , Histones/metabolism , Adipocytes/metabolism , Adipocytes/drug effects , Inflammation/pathology , Inflammation/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , Cell Death/drug effects , Male , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Wounds and Injuries/complications , Wounds and Injuries/metabolism , Wounds and Injuries/pathology , Signal Transduction/drug effects
3.
Stem Cell Rev Rep ; 19(5): 1554-1575, 2023 07.
Article En | MEDLINE | ID: mdl-37060532

Mesenchymal stem cells (MSCs) is promising in promoting wound healing mainly due to their paracrine function. Nonetheless, the transplanted MSCs presented poor survival with cell dysfunction and paracrine problem in diabetic environment, thus limiting their therapeutic efficacy and clinical application. JAM-A, an adhesion molecule, has been reported to play multi-functional roles in diverse cells. We therefore investigated the potential effect of JAM-A on MSCs under diabetic environment and explored the underlying mechanism. Indeed, high-glucose condition inhibited MSCs viability and JAM-A expression. However, JAM-A abnormality was rescued by lentivirus transfection and JAM-A overexpression promoted MSCs proliferation, migration and adhesion under hyperglycemia. Moreover, JAM-A overexpression attenuated high-glucose-induced ROS production and MSCs apoptosis. The bio-effects of JAM-A on MSCs under hyperglycemia were confirmed by RNA-seq with enrichment analyses. Moreover, Luminex chip results showed JAM-A overexpression dramatically upregulated PDGF-BB and VEGF in the supernatant of MSCs, which was verified by RT-qPCR and western blotting. The supernatant was further found to facilitate HUVECs proliferation, migration and angiogenesis under hyperglycemia. In vivo experiments revealed JAM-A overexpression significantly enhanced MSCs survival, promoted wound angiogenesis, and thus accelerated diabetic wound closure, partially by enhancing PDGF-BB and VEGF expression. This study firstly demonstrated that JAM-A expression of MSCs was inhibited upon high-glucose stimulation. JAM-A overexpression alleviated high-glucose-induced MSCs dysfunction, enhanced their anti-oxidative capability, protected MSCs from hyperglycemia-induced apoptosis and improved their survival, thus strengthening MSCs paracrine function to promote angiogenesis and significantly accelerating diabetic wound healing, which offers a promising strategy to maximize MSCs-based therapy in diabetic wound.


Diabetes Mellitus , Hyperglycemia , Mesenchymal Stem Cells , Neovascularization, Physiologic , Wound Healing , Wounds and Injuries , Humans , Becaplermin/genetics , Becaplermin/metabolism , Cell Survival/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Glucose/pharmacology , Hyperglycemia/genetics , Hyperglycemia/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic/genetics , Paracrine Communication/genetics , Umbilical Cord/cytology , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/genetics , Wounds and Injuries/genetics , Wounds and Injuries/metabolism
4.
Shock ; 58(1): 1-13, 2022 07 01.
Article En | MEDLINE | ID: mdl-35984758

ABSTRACT: Platelets are subcellular anucleate components of blood primarily responsible for initiating and maintaining hemostasis. After injury to a blood vessel, platelets can be activated via several pathways, resulting in changed shape, adherence to the injury site, aggregation to form a plug, degranulation to initiate activation in other nearby platelets, and acceleration of thrombin formation to convert fibrinogen to fibrin before contracting to strengthen the clot. Platelet function assays use agonists to induce and measure one or more of these processes to identify alterations in platelet function that increase the likelihood of bleeding or thrombotic events. In severe trauma, these assays have revealed that platelet dysfunction is strongly associated with poor clinical outcomes. However, to date, the mechanism(s) causing clinically significant platelet dysfunction remain poorly understood. We review the pros, cons, and evidence for use of many of the popular assays in trauma, discuss limitations of their use in this patient population, and present approaches that can be taken to develop improved functional assays capable of elucidating mechanisms of trauma-induced platelet dysfunction. Platelet dysfunction in trauma has been associated with need for transfusions and mortality; however, most of the current platelet function assays were not designed for evaluating trauma patients, and there are limited data regarding their use in this population. New or improved functional assays will help define the mechanisms by which platelet dysfunction occurs, as well as help optimize future treatment.


Blood Platelets , Hemostasis , Thrombosis , Wounds and Injuries , Blood Platelets/metabolism , Fibrin/metabolism , Hemostasis/physiology , Humans , Platelet Aggregation , Platelet Function Tests/methods , Wounds and Injuries/metabolism
5.
J Wound Care ; 31(Sup7): S15-S19, 2022 Jul 01.
Article En | MEDLINE | ID: mdl-35797252

OBJECTIVE: Even with our best practices, we are frequently unable to prevent slow and stalled wound healing-particularly in people with impaired circulation and conditions such as diabetes. As a result, greater insight into the nature of wound healing and alternative treatment approaches is needed. An avenue that may be of particular promise is increasing understanding of the role of secretory leukocyte protease inhibitor (SLPI) as there is evidence that it enhances wound healing, its expression increases in response to inflammation and infection, and it exhibits anti-protease, anti-inflammatory, antiviral antibacterial and antifungal activities. METHOD: The response of SLPI levels to wounding and skin injury was assessed by taking punch skin biopsies from healthy volunteers and assessing the levels of SLPI at the site of injury at the time of wounding (baseline) as well as one, two, three, four, seven, nine and 12 weeks later. RESULTS: A total of 35 volunteers took part in the study. Significant elevations were found: levels of SLPI were greatly increased, 12 times that at baseline, and remained elevated at three weeks despite re-epithelialisation having occurred. CONCLUSION: These findings not only suggest that levels of SLPI rise rapidly following wounding, but that these elevations are sustained, and continue to increase even when re-epithelialisation has occurred. These results suggest that the role and potential benefits of this protease inhibitor deserve further exploration.


Secretory Leukocyte Peptidase Inhibitor , Wound Healing , Wounds and Injuries , Biopsy , Humans , Proteinase Inhibitory Proteins, Secretory/metabolism , Secretory Leukocyte Peptidase Inhibitor/metabolism , Skin/metabolism , Wounds and Injuries/metabolism
6.
Med Sci Monit ; 28: e934816, 2022 Mar 16.
Article En | MEDLINE | ID: mdl-35292616

BACKGROUND Wound healing is a dynamic and complex process that is regulated by a variety of factors and pathways. This study sought to identify the mechanisms of the four-herb Chinese medicine ANBP in enhancing wound repair. MATERIAL AND METHODS By comparing the group treated with ANBP for 6 h (Z6h) with the corresponding control group (C6h), we used the new high-throughput differential acetylation proteomics method to explore the mechanism of ANBP treatment and analyse and identify new targets of ANBP for promoting wound healing. RESULTS ANBP promoted skin wound healing in mice; the wound healing process was accelerated and the wound healing time was shortened (P<0.05). The upregulated proteins were distributed mostly in the mitochondria to nuclear respiratory chain complexes and cytoplasmic vesicles. The dominant pathways for upregulated proteins were fatty acid metabolism, pyruvate metabolism, and tricarboxylic acid cycle. Pdha1 was upregulated with the most acetylation sites, while the downregulated Ncl, and Pfkm were most acetylated. CONCLUSIONS The findings from our study showed that ANBP improved cell aerobic respiration through enhanced glycolysis, pyruvic acid oxidative decarboxylation, and the Krebs cycle to produce more ATP for energy consumption, thus accelerating wound repair of skin.


Cytokines/metabolism , Medicine, Chinese Traditional/methods , Mitochondria/metabolism , Proteomics/methods , Skin/injuries , Wound Healing , Wounds and Injuries/metabolism , Acetylation , Animals , Cells, Cultured , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mitochondria/pathology , Signal Transduction , Skin/metabolism , Skin/pathology , Up-Regulation , Wounds and Injuries/pathology
7.
Elife ; 112022 02 24.
Article En | MEDLINE | ID: mdl-35200139

The function of macrophages in vitro is linked to their metabolic rewiring. However, macrophage metabolism remains poorly characterized in situ. Here, we used two-photon intensity and lifetime imaging of autofluorescent metabolic coenzymes, nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) and flavin adenine dinucleotide (FAD), to assess the metabolism of macrophages in the wound microenvironment. Inhibiting glycolysis reduced NAD(P)H mean lifetime and made the intracellular redox state of macrophages more oxidized, as indicated by reduced optical redox ratio. We found that TNFα+ macrophages had lower NAD(P)H mean lifetime and were more oxidized compared to TNFα- macrophages. Both infection and thermal injury induced a macrophage population with a more oxidized redox state in wounded tissues. Kinetic analysis detected temporal changes in the optical redox ratio during tissue repair, revealing a shift toward a more reduced redox state over time. Metformin reduced TNFα+ wound macrophages, made intracellular redox state more reduced and improved tissue repair. By contrast, depletion of STAT6 increased TNFα+ wound macrophages, made redox state more oxidized and impaired regeneration. Our findings suggest that autofluorescence of NAD(P)H and FAD is sensitive to dynamic changes in intracellular metabolism in tissues and can be used to probe the temporal and spatial regulation of macrophage metabolism during tissue damage and repair.


Flavin-Adenine Dinucleotide/metabolism , Macrophages/metabolism , NADP/metabolism , Wounds and Injuries/metabolism , Zebrafish/metabolism , Animals , Female , Fluorescence , Glycolysis , Kinetics , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence, Multiphoton/methods , Oxidation-Reduction , Tumor Necrosis Factor-alpha/metabolism
8.
Biochem Biophys Res Commun ; 591: 68-75, 2022 02 05.
Article En | MEDLINE | ID: mdl-34999256

The regulatory mechanism of hematopoiesis and innate immunity in Drosophila is highly similar to that in mammals, and Drosophila has become a suitable model to understand vertebrate hematopoiesis and the immune response. JAK-STAT signaling pathway components are widely conserved during evolution, and contribute to hematopoiesis and multiple tissue damage and immune responses. Here, we demonstrate that Stat92E is widely expressed in the lymph gland, and the loss of jumu inhibits the maintenance of the JAK/STAT pathway in the CZ and MZ but not in the PSC of the lymph gland. Furthermore, we found that clean puncture wounding of the larval epidermis can lead to the activation of JAK/STAT signaling and the generation of lamellocytes, and Jumu is required for the activation of JAK/STAT in response to epidermal wounds.


Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epidermis/pathology , Janus Kinases/metabolism , Lymphangiogenesis , Lymphatic Vessels/metabolism , Transcription Factors/metabolism , Wounds and Injuries/metabolism , Animals , Cell Nucleus/metabolism , Drosophila Proteins/genetics , Hemocytes/metabolism , Mutation/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Transcription Factors/genetics
9.
Molecules ; 27(1)2022 Jan 01.
Article En | MEDLINE | ID: mdl-35011491

Mangifera indica can generate up to 60% of polluting by-products, including peels. However, it has been shown that flavonoids and mangiferin are mainly responsible for the antioxidant, anti-inflammatory, and antibacterial activities closely related to the wound-healing process. The chemical composition of MEMI (methanolic extract of M. indica) was analyzed by HPLC-DAD, as well as concentrations of total phenol (TPC) and flavonoids (TFC) and antioxidant activity (SA50). Wound-healing efficacy was determined by measurements of wound contraction, histological analysis, and tensiometric method; moreover, anti-inflammatory, antibacterial, and acute dermal toxicity (OECD 402) were also evaluated. Phenol, resorcinol, conjugated resorcinol, and mangiferin were detected. TPC, TFC, and SA50 were 136 mg GAE/g, 101.66 mg QE/g, and 36.33 µg/mL, respectively. Tensile strength and wound contraction closure did not show significant differences between MEMI and dexpanthenol groups. Histological analysis (after 14 days) shows a similar architecture between MEMI treatment and normal skin. MEMI exhibits a reduction in edema. Staphylococcus epidermidis had an MIC of 2 mg/mL, while Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli reached 4 mg/mL. The MEMI showed no signs of toxicity. Therefore, this study demonstrates multiple targets that flavonoids and mangiferin of MEMI may present during the healing process.


Mangifera/chemistry , Plant Extracts , Wound Healing/drug effects , Wounds and Injuries , Animals , Disease Models, Animal , Flavonoids/chemistry , Flavonoids/pharmacology , Male , Mice , Plant Extracts/chemistry , Plant Extracts/pharmacology , Wounds and Injuries/drug therapy , Wounds and Injuries/metabolism , Wounds and Injuries/microbiology , Xanthones/chemistry , Xanthones/pharmacology
10.
J Leukoc Biol ; 111(1): 93-111, 2022 01.
Article En | MEDLINE | ID: mdl-34533241

Traumatic injury is a major cause of morbidity and mortality worldwide, despite significant advances in treatments. Most deaths occur either very early, through massive head trauma/CNS injury or exsanguination (despite advances in transfusion medicine), or later after injury often through multiple organ failure and secondary infection. Extracellular vesicles (EVs) are known to increase in the circulation after trauma and have been used to limited extent as diagnostic and prognostic markers. More intriguingly, EVs are now being investigated as both causes of pathologies post trauma, such as trauma-induced coagulopathy, and as potential treatments. In this review, we highlight what is currently known about the role and effects of EVs in various aspects of trauma, as well as exploring current literature from investigators who have begun to use EVs therapeutically to alter the physiology and pathology of traumatic insults. The potential effectiveness of using EVs therapeutically in trauma is supported by a large number of experimental studies, but there is still some way to go before we understand the complex effects of EVs in what is already a complex disease process.


Extracellular Vesicles , Wounds and Injuries , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Extracellular Vesicles/transplantation , Hemostasis , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Thrombosis/metabolism , Thrombosis/pathology , Thrombosis/therapy , Wounds and Injuries/metabolism , Wounds and Injuries/pathology , Wounds and Injuries/therapy
11.
J Trauma Acute Care Surg ; 92(3): 535-541, 2022 03 01.
Article En | MEDLINE | ID: mdl-34789706

BACKGROUND: Heparan sulfate is an integral component of the glycocalyx that provides an anticoagulant layer close to the endothelium. Hypoperfusion, inflammation, and sympathoadrenal activation following major trauma result in glycocalyx shedding and subsequent release of heparan sulfate into the bloodstream. The possible anticoagulant effect of this "autoheparinization" has been suggested as a potential driver of trauma-induced coagulopathy. We investigated whether thromboelastometry can be used to detect trauma-induced autoheparinization. METHODS: This study comprised three parts. First, in a retrospective clinical study of 264 major trauma patients, the clotting time (CT) in the intrinsic activation (INTEM) and intrinsic activation plus heparinase (HEPTEM) assays were evaluated upon emergency room admission. Second, in an in vivo experimental rat model of hemorrhagic-traumatic shock, the release of heparan sulfate was investigated with INTEM and HEPTEM analyses of whole blood. Third, in vitro spiking of whole blood from healthy volunteers was undertaken to assess the effects of clinically relevant quantities of heparan sulfate and heparin on CT in the INTEM and HEPTEM assays. RESULTS: In the first part, severe injury and hemorrhagic shock was not associated with any increases in INTEM CT versus HEPTEM CT. Part 2 showed that an approximate threefold increase in heparan sulfate resulting from hemorrhagic traumatic shock in rats did not prolong INTEM CT, and no significant differences between INTEM CT and HEPTEM CT were observed. Third, spiking of whole blood with heparan sulfate had no impact on INTEM CT, whereas heparin elicited significant prolongation of INTEM CT. CONCLUSION: Despite structural similarity between heparan sulfate and heparin, the amounts of heparan sulfate shed in response to trauma did not exert an anticoagulant effect that was measurable by the intrinsically activated CT in thromboelastometry. The extent to which heparan sulfate contributes to trauma-induced coagulopathy has yet to be elucidated. LEVEL OF EVIDENCE: Prognostic and Epidemiologic; Level III.


Blood Coagulation Disorders/blood , Glycocalyx/metabolism , Heparin/metabolism , Heparitin Sulfate/metabolism , Shock, Hemorrhagic/metabolism , Thrombelastography/methods , Wounds and Injuries/metabolism , Animals , Blood Coagulation Tests , Female , Heparin/pharmacology , Heparitin Sulfate/pharmacology , Humans , Male , Rats , Retrospective Studies
12.
Bioengineered ; 13(1): 789-799, 2022 01.
Article En | MEDLINE | ID: mdl-34898359

To explore the regulation of keratinocyte growth factor (KGF) in the process of repairing rat skin wounds by taspine hydrochloride (TA/HCl), 45 male Sprague-Dawley (SD) rats were purchased and divided into an experimental group, a dimethyl sulfoxide (DMSO) control group, and a basic fibroblast growth factor (bFGF) control group, each with 15 only. A back trauma model was innovatively adopted to prevent rats from biting and contaminating. The wound healing time and healing rate of the rat, and the Hydroxyproline (Hyp) and KGF expressions were observed. Morphological changes of wound tissue and the number of capillaries were observed after hematoxylin-eosin (HE) staining. The results showed that wound healing rate of experimental group and bFGF group was significantly higher than that of DMSO group (P < 0.05) after 2-15 days, and wound healing time of experimental group was 18 days, which was significantly lower than that of the DMSO group (P < 0.05). Expression levels of Hyp and KGF in the granulation tissue of rats in the experimental group were much higher than those in the DMSO control group after trauma (P < 0.05). In early stage of wound tissue repair, the number of new capillaries formed in experimental group was significantly higher than that in DMSO control group (P < 0.05). In summary, this study innovatively focused on KGF. The mechanism of TA/HCL promoting rat skin wound healing was closely related to KGF.


Alkaloids/pharmacology , Fibroblast Growth Factor 7/metabolism , Signal Transduction/drug effects , Skin , Wound Healing/drug effects , Wounds and Injuries , Animals , Male , Rats , Rats, Sprague-Dawley , Skin/injuries , Skin/metabolism , Skin/pathology , Wounds and Injuries/drug therapy , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
13.
Biomed Pharmacother ; 145: 112473, 2022 Jan.
Article En | MEDLINE | ID: mdl-34861635

Bone marrow derived-mesenchymal stem cells (BMSCs)-based therapy is an outstanding candidate for cutaneous wound healing. Melatonin (MEL) has been reported for its anti-inflammatory as well as tissue regenerative properties. Existing work aimed to explore the potential healing power of BMSCs pre-treated with MEL in a skin wound model. Adult rats were allocated into control, PIO, BMSCs (1 × 105 cells), and MEL/BMSCs groups. On the 21 days post-wounding, tissues were sampled for analysis. The results demonstrated that compared to the control group, MEL/BMSCs therapy induced noticeable decline in wound area and elevated rate of wound retraction. Furthermore, marked increases in tissue hydroxyproline, as well as tissue content and gene expression level of vascular endothelial growth factor in MEL/BMSCs treated-wounded animals. Compared to the untreated control group, marked increases were found in antioxidant enzymatic activities together with elevated GSH levels in wounded tissues after MEL/BMSCs treatment. Moreover, therapeutically handled wounds with MEL/BMSCs revealed low levels of MDA, NO and protein carbonyls. Combined therapy with MEL/BMSCs relieved the inflammation witnessed by decreasing IL-1ß, TNF-α and NF-κB levels in wounded tissues. Furthermore, noteworthy rises in levels of TGF-ß and gene expression of α-SMA were noticed after MEL/BMSCs application that reveals their anti-scarring properties. Histologically, noticeable improvement in histopathological skin lesions in wound area and elevated the collagen synthesis and deposition. Collectively, the obtained data depict that the pre-treatment of BMSCs with MEL could potentially be a successful strategy for scaling-up the wound healing outcomes more than using BMSCs monotherapy in rat models.


Melatonin , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Skin , Wound Healing , Wounds and Injuries , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Disease Models, Animal , Interleukin-1beta/metabolism , Melatonin/metabolism , Melatonin/pharmacology , NF-kappa B/metabolism , Rats , Skin/injuries , Skin/metabolism , Transforming Growth Factor beta/metabolism , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism , Wound Healing/drug effects , Wound Healing/physiology , Wounds and Injuries/metabolism , Wounds and Injuries/therapy
14.
Nat Commun ; 12(1): 7150, 2021 12 09.
Article En | MEDLINE | ID: mdl-34887411

Tissue regeneration after injury requires coordinated regulation of stem cell activation, division, and daughter cell differentiation, processes that are increasingly well understood in many regenerating tissues. How accurate stem cell positioning and localized integration of new cells into the damaged epithelium are achieved, however, remains unclear. Here, we show that enteroendocrine cells coordinate stem cell migration towards a wound in the Drosophila intestinal epithelium. In response to injury, enteroendocrine cells release the N-terminal domain of the PTK7 orthologue, Otk, which activates non-canonical Wnt signaling in intestinal stem cells, promoting actin-based protrusion formation and stem cell migration towards a wound. We find that this migratory behavior is closely linked to proliferation, and that it is required for efficient tissue repair during injury. Our findings highlight the role of non-canonical Wnt signaling in regeneration of the intestinal epithelium, and identify enteroendocrine cell-released ligands as critical coordinators of intestinal stem cell migration.


Cell Movement , Drosophila/metabolism , Enteroendocrine Cells/cytology , Intestinal Mucosa/cytology , Stem Cells/cytology , Wnt Proteins/metabolism , Wounds and Injuries/physiopathology , Animals , Drosophila/cytology , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Intestinal Mucosa/metabolism , Intestines , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Stem Cells/metabolism , Wnt Proteins/genetics , Wnt Signaling Pathway , Wounds and Injuries/genetics , Wounds and Injuries/metabolism
15.
STAR Protoc ; 2(4): 100983, 2021 12 17.
Article En | MEDLINE | ID: mdl-34927092

A key observation of tissue injury, such as stroke and burn, is a state of systemic immunosuppression characterized by loss of T cells and rise of infections. Here, we present an in vitro model for cell-cell interactions between innate (macrophages) and adaptive (T cells) immune cells. This protocol facilitates bone marrow-derived macrophages (BMDMs) and splenic T cells in a coculture model. The procedure mimics injury-induced T cell death, which is driven by inflammasome activation in macrophages. For complete details on the use and execution of this protocol, please refer to Roth et al. (2021).


Cell Death/physiology , Coculture Techniques/methods , Macrophages/cytology , Models, Immunological , T-Lymphocytes/cytology , Animals , Female , Inflammasomes/immunology , Inflammasomes/metabolism , Male , Mice , Mice, Inbred C57BL , Spleen/cytology , Wounds and Injuries/immunology , Wounds and Injuries/metabolism
16.
Emerg Microbes Infect ; 10(1): 2326-2339, 2021 Dec.
Article En | MEDLINE | ID: mdl-34821529

ABSTRACTMelioidosis is a serious infectious disease endemic in Southeast Asia, Northern Australia and has been increasingly reported in other tropical and subtropical regions in the world. Percutaneous inoculation through cuts and wounds on the skin is one of the major modes of natural transmission. Despite cuts in skin being a major route of entry, very little is known about how the causative bacterium Burkholderia pseudomallei initiates an infection at the skin and the disease manifestation at the skin known as cutaneous melioidosis. One key issue is the lack of suitable and relevant infection models. Employing an in vitro 2D keratinocyte cell culture, a 3D skin equivalent fibroblast-keratinocyte co-culture and ex vivo organ culture from human skin, we developed infection models utilizing surrogate model organism Burkholderia thailandensis to investigate Burkholderia-skin interactions. Collectively, these models show that the bacterial infection was largely limited at the wound's edge. Infection impedes wound closure, triggers inflammasome activation and cellular extrusion in the keratinocytes as a potential way to control bacterial infectious load at the skin. However, extensive infection over time could result in the epidermal layer being sloughed off, potentially contributing to formation of skin lesions.


Burkholderia pseudomallei/physiology , Burkholderia/physiology , Epidermis/microbiology , Inflammasomes/metabolism , Keratinocytes/microbiology , Melioidosis/microbiology , Skin/microbiology , Wounds and Injuries/microbiology , Cells, Cultured , Epidermis/metabolism , Humans , Keratinocytes/metabolism , Melioidosis/metabolism , Melioidosis/pathology , Models, Biological , Skin/metabolism , Skin/pathology , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
17.
J Tissue Eng Regen Med ; 15(12): 1131-1143, 2021 12.
Article En | MEDLINE | ID: mdl-34551191

Volumetric muscle loss (VML) is traumatic or surgical loss of skeletal muscle with resultant functional impairment. Skeletal muscle's innate capacity for regeneration is lost with VML due to a critical loss of stem cells, extracellular matrix, and neuromuscular junctions. Consequences of VML include permanent disability or delayed amputations of the affected limb. Currently, a successful clinical therapy has not been identified. Mesenchymal stem cells (MSCs) possess regenerative and immunomodulatory properties and their three-dimensional aggregation can further enhance therapeutic efficacy. In this study, MSC aggregation into spheroids was optimized in vitro based on cellular viability, spheroid size, and trophic factor secretion. The regenerative potential of the optimized MSC spheroid therapy was then investigated in a murine model of VML injury. Experimental groups included an untreated VML injury control, intramuscular injection of MSC spheroids, and MSC spheroids encapsulated in a fibrin-laminin hydrogel. Compared to the untreated VML group, the spheroid encapsulating hydrogel group enhanced myogenic marker (i.e., MyoD and myogenin) protein expression, improved muscle mass, increased presence of centrally nucleated myofibers as well as small fibers (<500 µm2 ), modulated pro- and anti-inflammatory macrophage marker expression (i.e., iNOS and Arginase), and increased the presence of CD146+ pericytes and CD31+ endothelial cells in the VML injured muscles. Future studies will evaluate the extent of functional recovery with the spheroid encapsulating hydrogel therapy.


Cells, Immobilized , Fibrin/chemistry , Hydrogels/chemistry , Laminin/chemistry , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Muscle, Skeletal , Regeneration , Spheroids, Cellular , Wounds and Injuries , Animals , Cells, Immobilized/metabolism , Cells, Immobilized/transplantation , Male , Mice , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Spheroids, Cellular/metabolism , Spheroids, Cellular/transplantation , Wounds and Injuries/metabolism , Wounds and Injuries/therapy
18.
Nat Commun ; 12(1): 4939, 2021 08 16.
Article En | MEDLINE | ID: mdl-34400627

Pain is a central feature of soft tissue trauma, which under certain contexts, results in aberrant osteochondral differentiation of tissue-specific stem cells. Here, the role of sensory nerve fibers in this abnormal cell fate decision is investigated using a severe extremity injury model in mice. Soft tissue trauma results in NGF (Nerve growth factor) expression, particularly within perivascular cell types. Consequently, NGF-responsive axonal invasion occurs which precedes osteocartilaginous differentiation. Surgical denervation impedes axonal ingrowth, with significant delays in cartilage and bone formation. Likewise, either deletion of Ngf or two complementary methods to inhibit its receptor TrkA (Tropomyosin receptor kinase A) lead to similar delays in axonal invasion and osteochondral differentiation. Mechanistically, single-cell sequencing suggests a shift from TGFß to FGF signaling activation among pre-chondrogenic cells after denervation. Finally, analysis of human pathologic specimens and databases confirms the relevance of NGF-TrkA signaling in human disease. In sum, NGF-mediated TrkA-expressing axonal ingrowth drives abnormal osteochondral differentiation after soft tissue trauma. NGF-TrkA signaling inhibition may have dual therapeutic use in soft tissue trauma, both as an analgesic and negative regulator of aberrant stem cell differentiation.


Cell Differentiation , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Signal Transduction , Wounds and Injuries/metabolism , Animals , Axons/metabolism , Cartilage/metabolism , Mice , Mice, Inbred C57BL , Nerve Growth Factor/genetics , Osteogenesis , Stem Cells/metabolism , Wounds and Injuries/pathology
19.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Article En | MEDLINE | ID: mdl-34272284

As collective cell migration is essential in biological processes spanning development, healing, and cancer progression, methods to externally program cell migration are of great value. However, problems can arise if the external commands compete with strong, preexisting collective behaviors in the tissue or system. We investigate this problem by applying a potent external migratory cue-electrical stimulation and electrotaxis-to primary mouse skin monolayers where we can tune cell-cell adhesion strength to modulate endogenous collectivity. Monolayers with high cell-cell adhesion showed strong natural coordination and resisted electrotactic control, with this conflict actively damaging the leading edge of the tissue. However, reducing preexisting coordination in the tissue by specifically inhibiting E-cadherin-dependent cell-cell adhesion, either by disrupting the formation of cell-cell junctions with E-cadherin-specific antibodies or rapidly dismantling E-cadherin junctions with calcium chelators, significantly improved controllability. Finally, we applied this paradigm of weakening existing coordination to improve control and demonstrate accelerated wound closure in vitro. These results are in keeping with those from diverse, noncellular systems and confirm that endogenous collectivity should be considered as a key quantitative design variable when optimizing external control of collective migration.


Cell Movement , Keratinocytes/physiology , Animals , Cadherins/metabolism , Calcium/metabolism , Cell Adhesion , Cell Line , Humans , Intercellular Junctions/metabolism , Keratinocytes/chemistry , Mice , Skin/chemistry , Skin/cytology , Wound Healing , Wounds and Injuries/metabolism , Wounds and Injuries/physiopathology
20.
Dev Cell ; 56(15): 2160-2175.e5, 2021 08 09.
Article En | MEDLINE | ID: mdl-34273275

The presence of a wound triggers surrounding cells to initiate repair mechanisms, but it is not clear how cells initially detect wounds. In epithelial cells, the earliest known wound response, occurring within seconds, is a dramatic increase in cytosolic calcium. Here, we show that wounds in the Drosophila notum trigger cytoplasmic calcium increase by activating extracellular cytokines, Growth-blocking peptides (Gbps), which initiate signaling in surrounding epithelial cells through the G-protein-coupled receptor Methuselah-like 10 (Mthl10). Latent Gbps are present in unwounded tissue and are activated by proteolytic cleavage. Using wing discs, we show that multiple protease families can activate Gbps, suggesting that they act as a generalized protease-detector system. We present experimental and computational evidence that proteases released during wound-induced cell damage and lysis serve as the instructive signal: these proteases liberate Gbp ligands, which bind to Mthl10 receptors on surrounding epithelial cells, and activate downstream release of calcium.


Epithelium/metabolism , Receptors, G-Protein-Coupled/metabolism , Wound Healing/physiology , Animals , Calcium/metabolism , Calcium Signaling , Cytosol/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epithelial Cells/metabolism , Epithelium/physiology , Peptides/metabolism , Proteolysis , Wounds and Injuries/metabolism
...