Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Cells ; 13(5)2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38474365

ABSTRACT

Obesity is associated with low-grade chronic inflammation and impaired glucose metabolism, both of which are detrimental to wound healing. C-C motif chemokine receptor 2 (CCR2) plays an important role in cell recruitment during healing, and our recent studies revealed the significance of CCR2-CCL2 signaling in promoting the proliferation of pro-inflammatory monocytes/macrophages in wounds. Therefore, we sought to determine whether diet-induced obesity increases monocyte/macrophage proliferation and their accumulation in skin wounds. We first confirmed that wound closure was delayed in obese CCR2RFP/+ mice fed with a high-fat diet (HFD) compared to mice fed with a normal diet (ND). Using in vivo imaging and flow cytometry analysis, we found that HFD mice had significantly increased accumulation of CCR2+ monocytes/macrophages, particularly pro-inflammatory CCR2+Ly6C+ cells in wounds compared to their ND counterparts. Importantly, HFD mice exhibited an increased proliferation of wound CCR2+Ly6C+ compared to ND mice. Together, our data suggest that obesity leads to an increased proliferation and accumulation of pro-inflammatory CCR2+Ly6C+ monocytes/macrophages in skin wounds, which may contribute to delayed healing.


Subject(s)
Macrophages , Monocytes , Mice , Animals , Monocytes/metabolism , Macrophages/metabolism , Obesity/metabolism , Diet, High-Fat , Receptors, Chemokine/metabolism , Wound Healing , Cell Proliferation
2.
Immunometabolism (Cobham) ; 5(4): e00033, 2023 Oct.
Article in English | MEDLINE | ID: mdl-38037591

ABSTRACT

Obesity is associated with alterations in tissue composition, systemic cellular metabolism, and low-grade chronic inflammation. Macrophages are heterogenous innate immune cells ubiquitously localized throughout the body and are key components of tissue homeostasis, inflammation, wound healing, and various disease states. Macrophages are highly plastic and can switch their phenotypic polarization and change function in response to their local environments. Here, we discuss how obesity alters the intestinal microenvironment and potential key factors that can influence intestinal macrophages as well as macrophages in other organs, including adipose tissue and hematopoietic organs. As bariatric surgery can induce metabolic adaptation systemically, we discuss the potential mechanisms through which bariatric surgery reshapes macrophages in obesity.

3.
Adv Wound Care (New Rochelle) ; 11(11): 598-621, 2022 11.
Article in English | MEDLINE | ID: mdl-34353116

ABSTRACT

Significance: Emerging evidence has shown a link between the status of hematopoietic stem cells (HSCs) and wound healing responses. Thus, better understanding HSCs will contribute to further advances in wound healing research. Recent Advances: Myeloid cells such as neutrophils and monocyte-derived macrophages are critical players in the process of wound healing. HSCs actively respond to wound injury and other tissue insults, including infection and produce the effector myeloid cells, and a failing of the HSC response can result in impaired wound healing. Technological advances such as transcriptome at single-cell resolution, epigenetics, three-dimensional imaging, transgenic animals, and animal models, have provided novel concepts of myeloid generation (myelopoiesis) from HSCs, and have revealed cell-intrinsic and -extrinsic mechanisms that can impact HSC functions in the context of health conditions. Critical Issues: The newer concepts include-the programmed cellular fate at a differentiation stage that is used to be considered as the multilineage, the signaling pathways that can activate HSCs directly and indirectly, the mechanisms that can deteriorate HSCs, the roles and remodeling of the surrounding environment for HSCs and their progenitors (the niche). Future Directions: The researches on HSCs, which produce blood cells, should contribute to the development of blood biomarkers predicting a risk of chronic wounds, which may transform clinical practice of wound care with precision medicine for patients at high risk of poor healing.


Subject(s)
Hematopoietic Stem Cells , Wound Healing , Animals , Cell Differentiation , Hematopoietic Stem Cells/physiology , Myeloid Cells , Myelopoiesis , Wound Healing/physiology
4.
Adv Ther (Weinh) ; 5(10)2022 Oct.
Article in English | MEDLINE | ID: mdl-36590645

ABSTRACT

Sepsis is a complex, life-threatening hyperinflammatory syndrome associated with organ failure and high mortality due to lack of effective treatment options. Here we report a core-shell hydrogel nanoparticle with the core functionalized with telodendrimer (TD) nanotrap (NT) to control hyperinflammation in sepsis. The combination of multi-valent charged and hydrophobic moieties in TD enables effective binding with biomolecules in NT. The higher crosslinking in the shell structure of nanogel excludes the abundant large serum proteins and allows for size-selectivity in scavenging the medium-sized septic molecules (10-30 kDa), e.g., lipopolysaccharides (LPS, a potent endotoxin in sepsis), thus reducing cytokine production. At the same time, the core-shell TD NT nanogel captures the over-flowing proinflammatory cytokines effectively both in vitro and in vivo from biological fluids to further control hyperinflammation. Intraperitoneal injection of core-shell TD NT nanogel effectively attenuates NF-κB activation and cytokine production in LPS-induced septic mouse models. These results indicate the potential applications of the injectable TD NT core-shell nanogel to attenuate local or systemic inflammation.

5.
PLoS One ; 15(4): e0231962, 2020.
Article in English | MEDLINE | ID: mdl-32324828

ABSTRACT

Vascular deficits are a fundamental contributing factor of diabetes-associated diseases. Although previous studies have demonstrated that the pro-angiogenic phase of wound healing is blunted in diabetes, a comprehensive understanding of the mechanisms that regulate skin revascularization and capillary stabilization in diabetic wounds is lacking. Using a mouse model of diabetic wound healing, we performed microCT analysis of the 3-dimensional architecture of the capillary bed. As compared to wild type, vessel surface area, branch junction number, total vessel length, and total branch number were significantly decreased in wounds of diabetic mice as compared to WT mice. Diabetic mouse wounds also had significantly increased capillary permeability and decreased pericyte coverage of capillaries. Diabetic wounds exhibited significant perturbations in the expression of factors that affect vascular regrowth, maturation and stability. Specifically, the expression of VEGF-A, Sprouty2, PEDF, LRP6, Thrombospondin 1, CXCL10, CXCR3, PDGFR-ß, HB-EGF, EGFR, TGF-ß1, Semaphorin3a, Neuropilin 1, angiopoietin 2, NG2, and RGS5 were down-regulated in diabetic wounds. Together, these studies provide novel information about the complexity of the perturbation of angiogenesis in diabetic wounds. Targeting factors responsible for wound resolution and vascular pruning, as well those that affect pericyte recruitment, maturation, and stability may have the potential to improve diabetic skin wound healing.


Subject(s)
Blood Vessels/pathology , Blood Vessels/physiopathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Neovascularization, Pathologic , Wound Healing , Animals , Blood Vessels/diagnostic imaging , Blood Vessels/metabolism , Capillaries/metabolism , Capillaries/physiopathology , Diabetes Mellitus, Experimental/diagnostic imaging , Diabetes Mellitus, Experimental/metabolism , Female , Mice , Mice, Inbred C57BL , Pericytes/pathology , Permeability , X-Ray Microtomography
6.
FASEB J ; 34(4): 5838-5850, 2020 04.
Article in English | MEDLINE | ID: mdl-32124475

ABSTRACT

Chemokines and their receptors play important roles in vascular homeostasis, development, and angiogenesis. Little is known regarding the molecular signaling mechanisms activated by CCL28 chemokine via its primary receptor CCR10 in endothelial cells (ECs). Here, we test the hypothesis that CCL28/CCR10 signaling plays an important role in regulating skin wound angiogenesis through endothelial nitric oxide synthase (eNOS)-dependent Src, PI3K, and MAPK signaling. We observed nitric oxide (NO) production in human primary ECs stimulated with exogenous CCL28, which also induced direct binding of CCR10 and eNOS resulting in inhibition of eNOS activity. Knockdown of CCR10 with siRNA lead to reduced eNOS expression and tube formation suggesting the involvement of CCR10 in EC angiogenesis. Based on this interaction, we engineered a myristoylated 7 amino acid CCR10-binding domain (Myr-CBD7) peptide and showed that this can block eNOS interaction with CCR10, but not with calmodulin, resulting in upregulation of eNOS activity. Importantly, topical administration of Myr-CBD7 peptide on mouse dermal wounds not only blocked CCR10-eNOS interaction, but also enhanced expression of eNOS, CD31, and IL-4 with reduction of CCL28 and IL-6 levels associated with improved wound healing. These results point to a potential therapeutic strategy to upregulate NO bioavailability, enhance angiogenesis, and improve wound healing by disrupting CCL28-activated CCR10-eNOS interaction.


Subject(s)
Chemokines, CC/metabolism , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , Receptors, CCR10/metabolism , Skin/physiopathology , Wound Healing , Animals , Chemokines, CC/genetics , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/genetics , Receptors, CCR10/genetics , Skin/injuries
7.
J Leukoc Biol ; 107(4): 551-560, 2020 04.
Article in English | MEDLINE | ID: mdl-31777992

ABSTRACT

Monocytes and macrophages (Mo/MΦ) play critical roles in all phases of skin wound healing. The majority of these cells are thought to be recruited from blood Mo; however, the role local proliferation of Mo/MΦ in the wound has not been defined. Therefore, we tested the hypothesis that local proliferation of Mo and/or MΦ contributes to their accumulation during wound healing. Male C57Bl/6 mice (N = 4-9/group) were subjected to excisional skin wounding. Proliferating Mo/MΦ (F4/80+Ki67+) were observed in wound cryosections, peaking on day 5 post-wounding. Cell cycle analysis on cells isolated from skin tissue revealed that wounding increased both the number and percentage of inflammatory Ly6C+F4/80lo/- Mo/MΦ in the S/G2/M phases, peaking on day 6 post-wounding. In contrast, more mature Ly6C-F4/80+ cells were found predominantly in the G0 phase with less than 1% cells in S/G2/M phase following injury. In peripheral blood, Mo were very rarely found in the S/G2/M phase, suggesting that the wound environment triggered the Ly6C+F4/80lo/- Mo proliferative response. Furthermore, injury induced several potential regulators of proliferation in wounds, including IL-1ß and IL-6, and wound Mo/MΦ expressed surface receptors for these cytokines. However, wound Mo/MΦ proliferation was not altered in IL-1R1 knockout (KO) or IL-6 KO mice. In summary, our findings indicate that proliferation contributes to Mo/MΦ accumulation in wounds and, contrary to findings in other pathophysiologic conditions, Ly6C+/F4/80lo/- Mo/MΦ proliferate during skin wound healing whereas mature Ly6C-F4/80+ MΦ do not.


Subject(s)
Macrophages/cytology , Monocytes/cytology , Skin/pathology , Wound Healing , Animals , Bone Marrow Cells/cytology , Cell Proliferation , Interleukin-1/pharmacology , Interleukin-1beta/metabolism , Interleukin-6/pharmacology , Male , Mice, Inbred C57BL , Models, Biological
8.
J Pathol ; 249(4): 435-446, 2019 12.
Article in English | MEDLINE | ID: mdl-31342513

ABSTRACT

Diabetes induces dysregulation throughout the spectrum of myeloid lineage cells from progenitors to terminally differentiated cells. Another complication of diabetes is persistent inflammation, including prolonged accumulation of macrophages, which contributes to impaired wound healing. However, it remains unclear whether diabetes disrupts the response of bone marrow progenitors to peripheral injury and whether such dysregulation leads to sustained inflammation and impaired healing. Here, we demonstrated that diabetic mice (db/db, referred to here as DB) exhibit myeloid lineage bias during homeostasis and following injury. In addition, cells in the LSK (Lin- Sca-1+ cKit+ ) population of DB mice are preprogrammed towards myeloid commitment at the transcriptional level, and cultured myeloid progenitors from DB mice produce more monocytes ex vivo than their non-diabetic counterparts. We also show via bone marrow transfer between interleukin-1 receptor 1 KO (Il1r1-/- ) and DB mice that IL-1R1 signaling is likely not involved in myeloid skewing in DB mice. Furthermore, in vitro experiments indicated that macrophage colony-stimulating factor receptor signaling is not likely involved in enhanced myeloid transcription factor expression in LSK cells of DB mice. Our findings indicate that myeloid lineage commitment in bone marrow may contribute to increased macrophage numbers observed in diabetic skin wounds, and that strategies to regulate monopoiesis during homeostasis or post-wounding may improve diabetic wound healing. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cell Lineage , Diabetes Mellitus, Type 2/pathology , Macrophages/pathology , Myeloid Progenitor Cells/pathology , Skin/pathology , Wound Healing , Wounds, Penetrating/pathology , Animals , Bone Marrow Transplantation , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myeloid Progenitor Cells/metabolism , Myelopoiesis , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Signal Transduction , Skin/injuries , Skin/metabolism , Stem Cell Transplantation , Wounds, Penetrating/genetics , Wounds, Penetrating/metabolism
10.
J Immunol ; 202(9): 2720-2727, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30910860

ABSTRACT

The aim of this study was to determine whether skin wounding induces monocyte (Mo) expansion in bone marrow and whether IL-1R1 signaling regulates this process. Our data show that skin wounding increases myeloid lineage-committed multipotent progenitors (MPP3 subset) and Mo in bone marrow, but this expansion is not impaired in Il1r1-/- mice. We also demonstrate that M-CSF-induced differentiation of myeloid progenitors into Mo is not impaired by the loss of IL-1R1 ex vivo, indicating that IL-R1 deficiency does not abrogate myeloid progenitor differentiation potential. In addition, we observed modestly delayed wound closure in Il1r1-/- mice associated with higher frequency of Ly6Clo Mo in the circulation at baseline and in wounds early after injury. Thus, in contrast to other models of inflammation that involve IL-1R1-dependent monopoiesis, our results demonstrate that skin wounding induces Mo progenitor and Mo expansion independently of IL-1R1 signaling.


Subject(s)
Bone Marrow/immunology , Monocytes/immunology , Receptors, Interleukin-1 Type I/deficiency , Skin/immunology , Wound Healing/immunology , Wounds and Injuries/immunology , Animals , Bone Marrow/pathology , Mice , Mice, Knockout , Monocytes/pathology , Receptors, Interleukin-1 Type I/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Skin/pathology , Wound Healing/genetics , Wounds and Injuries/genetics , Wounds and Injuries/pathology
11.
J Immunol ; 201(8): 2414-2426, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30201810

ABSTRACT

Ischemic tissue damage activates hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM)-generating myeloid cells, and persistent HSPC activity may drive chronic inflammation and impair tissue recovery. Although increased reactive oxygen species in the BM regulate HSPC functions, their roles in myelopoiesis of activated HSPCs and subsequent tissue recovery during ischemic damage are not well understood. In this paper, we report that deletion of Nox2 NADPH oxidase in mice results in persistent elevations in BM HSPC activity and levels of inflammatory monocytes/macrophages in BM and ischemic tissue in a model of hindlimb ischemia. Ischemic tissue damage induces oxidants in BM such as elevations of hydrogen peroxide and oxidized phospholipids, which activate redox-sensitive Lyn kinase in a Nox2-dependent manner. Moreover, during tissue recovery after ischemic injury, this Nox2-ROS-Lyn kinase axis is induced by Nox2 in neutrophils that home to the BM, which inhibits HSPC activity and inflammatory monocyte generation and promotes tissue regeneration after ischemic damage. Thus, oxidant signaling in the BM mediated by Nox2 in neutrophils regulates myelopoiesis of HSPCs to promote regeneration of damaged tissue.


Subject(s)
Hematopoietic Stem Cells/physiology , Hindlimb/pathology , Ischemia/immunology , NADPH Oxidase 2/metabolism , Neutrophils/physiology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelopoiesis , NADPH Oxidase 2/genetics , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Regeneration , Signal Transduction , src-Family Kinases/metabolism
12.
Hum Mol Genet ; 26(24): 4951-4960, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29206970

ABSTRACT

The purpose of this study was to determine whether thrombospondin (TSP)-1 promotes macrophage activity and disease progression in dysferlinopathy. First, we found that levels of TSP-1 are elevated in blood of non-ambulant dysferlinopathy patients compared with ambulant patients and healthy controls, supporting the idea that TSP-1 levels are correlated with disease progression. We then crossed dysferlinopathic BlaJ mice with TSP-1 knockout mice and assessed disease progression longitudinally with magnetic resonance imaging (MRI). In these mice, deletion of TSP-1 ameliorated loss in volume and mass of the moderately affected gluteal muscle but not of the severely affected psoas muscle. T2 MRI parameters revealed that loss of TSP-1 modestly inhibited inflammation only in gluteal muscle of male mice. Histological assessment indicated that deletion of TSP-1 reduced inflammatory cell infiltration of muscle fibers, but only early in disease progression. In addition, flow cytometry analysis revealed that, in males, TSP-1 knockout reduced macrophage infiltration and phagocytic activity, which is consistent with TSP-1-enhanced phagocytosis and pro-inflammatory cytokine induction in cultured macrophages. In summary, TSP-1 appears to play an accessory role in modulating Mp activity in BlaJ mice in a gender, age and muscle-dependent manner, but is unlikely a primary driver of disease progression of dysferlinopathy.


Subject(s)
Muscular Dystrophies, Limb-Girdle/metabolism , Thrombospondin 1/metabolism , Adult , Animals , Disease Models, Animal , Disease Progression , Female , Humans , Inflammation/pathology , Macrophage Activation/physiology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout , Muscular Dystrophies, Limb-Girdle/blood , Muscular Dystrophies, Limb-Girdle/pathology , Phagocytosis , Thrombospondin 1/blood
13.
Cytokine ; 94: 29-36, 2017 06.
Article in English | MEDLINE | ID: mdl-28408068

ABSTRACT

Chronic inflammation plays a key role in the pathogenesis of myriad complications associated with diabetes and thus anti-inflammatory therapies may ameliorate these complications. Quercus infectoria (Qi) extract has been shown to downregulate inflammatory processes; however, the molecular mechanisms of this anti-inflammatory activity remain unclear. The hypothesis of our study was that Qi extract exerts its anti-inflammatory effect by downregulating the Set7/NF-κB pathway. Bone marrow-derived macrophages (BMM) were treated with high glucose plus palmitate medium (HG/Pa) to simulate the diabetic environment. Compared with control conditions, HG/Pa elevated expression Set7, expression and activity of NF-κB along with expression of several inflammatory cytokines. These changes were associated with increased levels of intracellular reactive oxygen species (ROS). Moreover, similar alterations were demonstrated in BMM derived from mice fed a high fat diet (HFD) compared to those from lean mice, suggesting that HFD-induced changes in BM progenitors persist throughout differentiation and culture. Importantly, Qi extract dose-dependently reduced Set7, p65 and inflammatory cytokine expression relative to vehicle controls in both HG/Pa-and HFD-treated BMM. Finally, macrophages/monocytes isolated from wounds of diabetic mice that were treated with Qi solution exhibited lower expression of the inflammatory cytokines, IL-1ß and TNF-α, compared with vehicle treated wounds, demonstrating translation to the in vivo diabetic environment. Taken together, data from this study suggests that Qi downregulates diabetes-induced activity of the Set7/NF-kB pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Macrophages/drug effects , NF-kappa B/metabolism , Plant Extracts/pharmacology , Protein Methyltransferases/metabolism , Quercus/chemistry , Signal Transduction/drug effects , Animals , Cytokines/genetics , Cytokines/metabolism , Diabetes Mellitus, Experimental/immunology , Diet, High-Fat , Glucose/pharmacology , Histone-Lysine N-Methyltransferase , Inflammation , Interleukin-1beta/metabolism , Macrophages/metabolism , Mice , Palmitates/pharmacology , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Wounds and Injuries/immunology
14.
Neuromuscul Disord ; 26(3): 240-51, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26927626

ABSTRACT

Dysferlinopathy is associated with accumulation of thrombospondin (TSP)-1 and macrophages, both of which may contribute to the pathogenesis of the disease. The purpose of this study was to determine whether TSP-1 levels can predict macrophage activity and disease progression in dysferlin deficient BlaJ mice, focusing on the early disease process. In 3 month-old BlaJ mice, muscle TSP-1 levels exhibited strong positive correlations with both accumulation of F4/80hi macrophages and with their in vivo phagocytic activity in psoas muscles as measured by magnetic resonance imaging and flow cytometry. Muscle TSP-1 levels also exhibited a strong negative correlation with muscle mass and strong positive correlations with histological measurements of muscle fiber infiltration and regeneration. Over the course of disease progression from 3 to 12 months of age, muscle TSP-1 levels showed more complicated relationships with macrophage activity and an inverse relationship with muscle mass. Importantly, blood TSP-1 levels showed strong correlations with macrophage activity and muscle degeneration, particularly early in disease progression in BlaJ mice. These data indicate that TSP-1 may contribute to a destructive macrophage response in dysferlinopathy and pose the intriguing possibility that TSP-1 levels may serve as a biomarker for disease progression.


Subject(s)
Disease Progression , Macrophages/physiology , Membrane Proteins/genetics , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Thrombospondin 1/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Disease Models, Animal , Dysferlin , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/physiopathology , Phagocytosis , Psoas Muscles/metabolism , Psoas Muscles/pathology , Thrombospondin 1/blood
15.
Microvasc Res ; 106: 57-66, 2016 07.
Article in English | MEDLINE | ID: mdl-27009591

ABSTRACT

Properly regulated angiogenesis and arteriogenesis are essential for effective wound healing. Tissue injury induces robust new vessel formation and subsequent vessel maturation, which involves vessel regression and remodeling. Although formation of functional vasculature is essential for healing, alterations in vascular structure over the time course of skin wound healing are not well understood. Here, using high-resolution ex vivo X-ray micro-computed tomography (microCT), we describe the vascular network during healing of skin excisional wounds with highly detailed three-dimensional (3D) reconstructed images and associated quantitative analysis. We found that relative vessel volume, surface area and branching number are significantly decreased in wounds from day 7 to days 14 and 21. Segmentation and skeletonization analysis of selected branches from high-resolution images as small as 2.5µm voxel size show that branching orders are decreased in the wound vessels during healing. In histological analysis, we found that the contrast agent fills mainly arterioles, but not small capillaries nor large veins. In summary, high-resolution microCT revealed dynamic alterations of vessel structures during wound healing. This technique may be useful as a key tool in the study of the formation and regression of wound vessels.


Subject(s)
Computed Tomography Angiography/methods , Neovascularization, Physiologic , Skin/blood supply , Skin/diagnostic imaging , Wound Healing , Wounds and Injuries/diagnostic imaging , X-Ray Microtomography , Animals , Arterioles/diagnostic imaging , Arterioles/physiopathology , Disease Models, Animal , Imaging, Three-Dimensional , Male , Mice, Inbred C57BL , Predictive Value of Tests , Radiographic Image Interpretation, Computer-Assisted , Time Factors , Wounds and Injuries/physiopathology
16.
Arterioscler Thromb Vasc Biol ; 36(1): 86-96, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26586661

ABSTRACT

OBJECTIVE: Comprehensive understanding of the mechanisms regulating angiogenesis might provide new strategies for angiogenic therapies for treating diverse physiological and pathological ischemic conditions. The E-twenty six (ETS) factor Ets variant 2 (ETV2; aka Ets-related protein 71) is essential for the formation of hematopoietic and vascular systems. Despite its indispensable function in vessel development, ETV2 role in adult angiogenesis has not yet been addressed. We have therefore investigated the role of ETV2 in vascular regeneration. APPROACH AND RESULTS: We used endothelial Etv2 conditional knockout mice and ischemic injury models to assess the role of ETV2 in vascular regeneration. Although Etv2 expression was not detectable under steady-state conditions, its expression was readily observed in endothelial cells after injury. Mice lacking endothelial Etv2 displayed impaired neovascularization in response to eye injury, wounding, or hindlimb ischemic injury. Lentiviral Etv2 expression in ischemic hindlimbs led to improved recovery of blood perfusion with enhanced vessel formation. After injury, fetal liver kinase 1 (Flk1), aka VEGFR2, expression and neovascularization were significantly upregulated by Etv2, whereas Flk1 expression and vascular endothelial growth factor response were significantly blunted in Etv2-deficient endothelial cells. Conversely, enforced Etv2 expression enhanced vascular endothelial growth factor-mediated endothelial sprouting from embryoid bodies. Lentiviral Flk1 expression rescued angiogenesis defects in endothelial Etv2 conditional knockout mice after hindlimb ischemic injury. Furthermore, Etv2(+/-); Flk1(+/-) double heterozygous mice displayed a more severe hindlimb ischemic injury response compared with Etv2(+/-) or Flk1(+/-) heterozygous mice, revealing an epistatic interaction between ETV2 and FLK1 in vascular regeneration. CONCLUSIONS: Our study demonstrates a novel obligatory role for the ETV2 in postnatal vascular repair and regeneration.


Subject(s)
Angiogenic Proteins/metabolism , Endothelial Cells/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Regeneration , Transcription Factors/metabolism , Angiogenic Proteins/deficiency , Angiogenic Proteins/genetics , Animals , Cells, Cultured , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/physiopathology , Disease Models, Animal , Endothelial Cells/pathology , Gene Expression Regulation , Gene Transfer Techniques , Genetic Vectors , Heterozygote , Hindlimb , Ischemia/genetics , Ischemia/pathology , Ischemia/physiopathology , Ischemia/therapy , Lentivirus/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Phenotype , Recovery of Function , Signal Transduction , Skin/blood supply , Time Factors , Transcription Factors/deficiency , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing
17.
Sci Rep ; 5: 14780, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26437801

ABSTRACT

Copper (Cu), an essential micronutrient, plays a fundamental role in inflammation and angiogenesis; however, its precise mechanism remains undefined. Here we uncover a novel role of Cu transport protein Antioxidant-1 (Atox1), which is originally appreciated as a Cu chaperone and recently discovered as a Cu-dependent transcription factor, in inflammatory neovascularization. Atox1 expression is upregulated in patients and mice with critical limb ischemia. Atox1-deficient mice show impaired limb perfusion recovery with reduced arteriogenesis, angiogenesis, and recruitment of inflammatory cells. In vivo intravital microscopy, bone marrow reconstitution, and Atox1 gene transfer in Atox1(-/-) mice show that Atox1 in endothelial cells (ECs) is essential for neovascularization and recruitment of inflammatory cells which release VEGF and TNFα. Mechanistically, Atox1-depleted ECs demonstrate that Cu chaperone function of Atox1 mediated through Cu transporter ATP7A is required for VEGF-induced angiogenesis via activation of Cu enzyme lysyl oxidase. Moreover, Atox1 functions as a Cu-dependent transcription factor for NADPH oxidase organizer p47phox, thereby increasing ROS-NFκB-VCAM-1/ICAM-1 expression and monocyte adhesion in ECs inflamed with TNFα in an ATP7A-independent manner. These findings demonstrate a novel linkage between Atox1 and NADPH oxidase involved in inflammatory neovascularization and suggest Atox1 as a potential therapeutic target for treatment of ischemic disease.


Subject(s)
Adenosine Triphosphatases/genetics , Cation Transport Proteins/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Ischemia/genetics , Metallochaperones/genetics , NADPH Oxidases/genetics , Neovascularization, Pathologic/genetics , Protein-Lysine 6-Oxidase/genetics , Adenosine Triphosphatases/metabolism , Animals , Cation Transport Proteins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Copper Transport Proteins , Copper-Transporting ATPases , Gene Expression Regulation , Hindlimb , Human Umbilical Vein Endothelial Cells/cytology , Humans , Ischemia/metabolism , Ischemia/pathology , Leg/blood supply , Leg/pathology , Metallochaperones/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones , Monocytes/metabolism , Monocytes/pathology , NADPH Oxidases/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Protein-Lysine 6-Oxidase/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
J Pathol ; 236(4): 433-44, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25875529

ABSTRACT

Macrophages undergo a transition from pro-inflammatory to healing-associated phenotypes that is critical for efficient wound healing. However, the regulation of this transition during normal and impaired healing remains to be elucidated. In our studies, the switch in macrophage phenotypes during skin wound healing was associated with up-regulation of the peroxisome proliferator-activated receptor (PPAR)γ and its downstream targets, along with increased mitochondrial content. In the setting of diabetes, up-regulation of PPARγ activity was impaired by sustained expression of IL-1ß in both mouse and human wounds. In addition, experiments with myeloid-specific PPARγ knockout mice indicated that loss of PPARγ in macrophages is sufficient to prolong wound inflammation and delay healing. Furthermore, PPARγ agonists promoted a healing-associated macrophage phenotype both in vitro and in vivo, even in the diabetic wound environment. Importantly, topical administration of PPARγ agonists improved healing in diabetic mice, suggesting an appealing strategy for down-regulating inflammation and improving the healing of chronic wounds.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Leg Ulcer/metabolism , Macrophages/metabolism , PPAR gamma/metabolism , Skin/metabolism , Wound Healing , Administration, Cutaneous , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Female , Humans , Interleukin-1beta/metabolism , Leg Ulcer/drug therapy , Leg Ulcer/genetics , Leg Ulcer/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/agonists , PPAR gamma/deficiency , PPAR gamma/genetics , Phenotype , Prostaglandin D2/administration & dosage , Prostaglandin D2/analogs & derivatives , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/genetics , Rosiglitazone , Skin/drug effects , Skin/pathology , Thiazolidinediones/administration & dosage , Time Factors , Wound Healing/drug effects
19.
Arterioscler Thromb Vasc Biol ; 35(4): 877-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25675998

ABSTRACT

OBJECTIVE: Transient receptor potential melastatin-2 (TRPM2) channel is a nonselective cation channel that mediates influx of Ca(2+) and Na(+) with relative permeability of PCa:PNa ≈0.6 in response to cellular oxidative stress. As angiogenesis and ischemic neovascularization are both significantly dependent on oxidant signaling, here we investigated the possible role of vascular endothelial growth factor (VEGF)-induced reactive oxygen species production in activating TRPM2-dependent Ca(2+) signaling and in the mechanism of angiogenesis and ischemic neovascularization. APPROACH AND RESULTS: We observed that VEGF stimulation rapidly induced the association of TRPM2 and cellular Src kinase with vascular endothelial-cadherin forming a signalplex at vascular endothelial-cadherin junctions in endothelial cells. Using endothelial cells isolated from TRPM2(-/-) mice or after small interfering RNA depletion of TRPM2, we demonstrated that TRPM2-activated Ca(2+) signaling was required for cellular Src kinase-induced phosphorylation of vascular endothelial-cadherin at Y658 and Y731, the crucial sites involved in vascular endothelial-cadherin internalization in response to VEGF. VEGF-induced reactive oxygen species generation activated TRPM2-induced Ca(2+) entry, whereas the reactive oxygen species-insensitive TRPM2 mutant (C1008→A) showed impaired Ca(2+) entry. Endothelial cells depleted of TRPM2 also displayed significantly perturbed migratory phenotype and impaired activation of cellular Src in response to VEGF. TRPM2(-/-) mice reconstituted with wild-type myeloid cells demonstrated aberrant angiogenesis and neovascularization in the hindlimb ischemia model as compared with wild-type mice. CONCLUSIONS: VEGF-induced angiogenesis and postischemic neovascularization in mice required reactive oxygen species generation in endothelial cells and resultant TRPM2 activation. Thus, our findings provide novel insight into the role of TRPM2 in mechanism of angiogenesis and ischemic neovascularization.


Subject(s)
Endothelial Cells/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Reactive Oxygen Species/metabolism , TRPM Cation Channels/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Calcium/metabolism , Calcium Signaling , Cell Movement , Cells, Cultured , Disease Models, Animal , Electric Impedance , Hindlimb , Humans , Ischemia/genetics , Ischemia/physiopathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Mutation , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA Interference , Signal Transduction , TRPM Cation Channels/deficiency , TRPM Cation Channels/genetics , Time Factors , Transfection , Vascular Endothelial Growth Factor A/metabolism
20.
Stem Cells ; 32(6): 1538-52, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24496925

ABSTRACT

Endothelial cell (EC) dedifferentiation in relation to neovascularization is a poorly understood process. In this report, we addressed the role of Wnt signaling in the mechanisms of neovascularization in adult tissues. Here, we show that a low-dose of 6-bromoindirubin-3'-oxime (BIO), a competitive inhibitor of glycogen synthase kinase-3ß, induced the stabilization of ß-catenin and its subsequent direct interaction with the transcription factor NANOG in the nucleus of ECs. This event induced loss of VE-cadherin from the adherens junctions, increased EC proliferation accompanied by asymmetric cell division (ACD), and formed cellular aggregates in hanging drop assays indicating the acquisition of a dedifferentiated state. In a chromatin immunoprecipitation assay, nuclear NANOG protein bound to the NANOG- and VEGFR2-promoters in ECs, and the addition of BIO activated the NANOG-promoter-luciferase reporter system in a cell-based assay. Consequently, NANOG-knockdown decreased BIO-induced NOTCH-1 expression, thereby decreasing cell proliferation, ACD, and neovascularization. In a Matrigel plug assay, BIO induced increased neovascularization, secondary to the presence of vascular endothelial growth factor (VEGF). Moreover, in a mouse model of hind limb ischemia, BIO augmented neovascularization that was coupled with increased expression of NOTCH-1 in ECs and increased smooth muscle α-actin(+) cell recruitment around the neovessels. Thus, these results demonstrate the ability of a low-dose of BIO to augment neovascularization secondary to VEGF, a process that was accompanied by a partial dedifferentiation of ECs via ß-catenin and the NANOG signaling pathway.


Subject(s)
Cell Dedifferentiation/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Indoles/pharmacology , Neovascularization, Physiologic/drug effects , Oximes/pharmacology , Angiogenesis Inducing Agents/metabolism , Animals , Cell Aggregation/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Fetal Proteins/genetics , Hindlimb/blood supply , Hindlimb/pathology , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Indoles/administration & dosage , Ischemia/pathology , Mice , Nanog Homeobox Protein , Oximes/administration & dosage , Phenotype , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Stability/drug effects , T-Box Domain Proteins/genetics , Vascular Endothelial Growth Factor A/pharmacology , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...