Leukocyte Factor XII Mediates Inflammation and Its Deficiency Promotes Wound Healing

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 616-616
Author(s):  
Evi Stavrou ◽  
Matthew J. Fullana ◽  
Gretchen LaRusch ◽  
Cindy Yushin Chang ◽  
Chao Fang ◽  
...  

Abstract Abstract 616 Background: Older investigations suggest that Factor XII (FXII) influences the inflammatory response. FXII deficient patients have reduced leukocyte migration into skin windows. In addition to bradykinin generation, FXII regulates the expression of monocyte FcγII receptor and stimulates monocytes and macrophages to release interleukin (IL)-1 and IL-6. In vitro, purified FXIIa corrects neutrophil aggregation and degranulation defects in FXII-deficient plasma. Our laboratory demonstrated that FXII signals through uPAR, β1 integrin and the EGFR to stimulate ERK1/2 and Akt phosphorylation (Blood 115:5111, 2010). The downstream consequences of this pathway are FXII-induced cell proliferation, growth and angiogenesis. Since FXII modulates wound angiogenesis, we examined the role for FXII in the inflammatory response and wound repair. Investigations: Exon 3 to 8–deleted FXII knockout mice (FXII−/−) were wounded by creating full thickness (5 mm) punch biopsies on their backs. The total surface area of inflammatory cells recruited to the injury site per 20X high power field (HPF) was determined and analyzed by Image J (NIH). We observed that on Day 1, FXII−/− mice exhibit significantly decreased recruitment of CD11 b–labeled inflammatory cells to injury sites compared with wild type mice (WT) [p=0.0136]. Similar results are observed when uPAR KO mice were compared to WT [p=0.0001], suggesting that leukocyte migration in part is mediated through this receptor. Next we determined the nature of cells in the wound sites. FXII−/− mice have reduced wound neutrophils (Gr-1 staining) and macrophages (F4-80 staining). On the thioglycolate (TG)-induced peritoneal inflammation assay, the number of peritoneal exudate cells (PECs) was measured in WT and FXII−/− mice 1 and 7 days following instillation. FXII−/− mice exhibit significantly decreased number of PEC's on days 1 and 7. Giemsa-Wright stain of peritoneal lavage fluid with manual differential counts or flow cytometry shows that there is a disproportionate decrease in neutrophil recruitment in peritoneal fluid of FXII−/− mice. Also, FXII−/− macrophages have reduced adherence to plastic. Identical assays performed on uPAR and bradykinin B2 receptor (B2R) KO mice did not reveal the same inflammatory defects; both uPAR and B2R KO mice have mild decreases in PEC cell numbers following TG instillation, but overall their inflammatory response remained intact. Investigations next determined if the leukocyte migration defect seen in FXII−/− mice is related to plasma FXII or an intrinsic leukocyte defect. On adoptive bone marrow (BM) transplantation experiments, WT bone marrow transplanted to FXII−/− mice corrects the TG-induced PEC migration defect after both 1 and 7 days from instillation. Alternatively, transplantation of FXII−/− bone marrow into WT mice produced a leukocyte migration defect. In order to further discriminate the contribution of FXII in leukocyte migration, we produced plasma FXII deficiency in WT mice using FXII siRNA. WT mice treated with FXII siRNA reduced plasma FXII activity to <10% at 24 h (T1/2= 6 h). These mice did not have a defect in PEC recruitment. In vivo infusion of purified FXII did not correct the leukocyte peritoneal migration defect in FXII−/− mice. Next we harvested BM from WT mice and isolated leukocytes to prepare bone marrow cell-derived mRNA. The mRNA was reverse-transcribed to cDNA and it demonstrates XII cDNA that shares sequence homology to hepatic XII from exons 2 through 14. Immunofluorescence staining of BM-derived leukocytes demonstrates XII-positive leukocytes with nuclear morphology resembling monocytes and neutrophils (Fig. 1). Finally although leukocyte FXII promotes their migration into wounds, FXII deficiency in leukocytes paradoxically is associated with improved rates of wound healing after punch biopsy. Conclusions: These data indicate that there is a unique pool of FXII in leukocytes distinct from plasma (hepatic) FXII. Leukocyte FXII, not plasma FXII, is responsible for leukocyte function in wounds and inflammation sites. FXII−/− mice have attenuated wound injury and, paradoxically, improved wound healing rates. Modulation of leukocyte FXII is a target for promotion of wound healing. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 368-368 ◽  
Author(s):  
Evi Stavrou ◽  
Gretchen A. LaRusch ◽  
Matthew J. Fullana ◽  
Gary E. Wnek ◽  
Cheng-Kui Qu ◽  
...  

Abstract Abstract 368 Background: Factor XII (FXII) participates in inflammation. FXII deficient patients have reduced leukocyte migration into skin windows. Recently, we have determined that FXII signals through uPAR, β1 integrin and the EGFR to stimulate ERK1/2 and Akt phosphorylation (Blood 115:5111, 2010). The downstream consequences of this pathway are FXII-induced cell proliferation and post-natal angiogenesis. Present studies examined if this pathway also influences leukocyte response to injury. Methods: Exon 3- to 8-deleted FXII knockout mice (FXII KO), generously provided by Dr. Frank Castellino, have impaired inflammatory response to injury. FXII KO were wounded by creating full thickness (5 mm) punch biopsies on their backs. The total surface area of inflammatory cells recruited at the injury site/20 × high power field (HPF) was determined and analyzed by Image J (NIH). We observed that FXII KO on Day 1 exhibit significantly decreased recruitment of CD11b-labeled inflammatory cells to injury sites compared with wild type mice (WT) [mean ± SEM, FXII KO: 40220 ± 3732 vs. WT: 59740 ± 6318 total surface area of CD11b positive cells (pixels)/HPF, p=0.0136]. Similar results are observed when uPAR KO were compared to WT mice [39380 ± 5234 vs. 73310 ± 4688, p=0.0001], suggesting that leukocyte migration is mediated through uPAR. On Day 1 of thioglycolate (TG)-induced peritonitis, FXII KO have a mean of 12.62 × 105/mL peritoneal exudate cells (PEC) vs. 25.94 × 105/mL (p=0.0072) observed in WT. Additionally on Day 7, FXII KO have 10.63 × 105/mL PEC vs. WT 25.38 × 105/mL (p< 0.0001). Likewise on Day 7, uPAR KO have 16.34 × 105/mL peritoneal leukocytes recruited vs. WT 25.38 × 105/mL (p=0.0124). These combined studies indicate that FXII influences the degree of leukocyte inflammation and the inflammatory response is partially mediated through uPAR. Studies next determined if leukocyte recruitment promotes wound healing. On Days 3–5 the rate of wound closure of back punch biopsies is faster in FXII KO than WT (p < 0.0327 by one way ANOVA). Again, we observed that FXII KO on Days 2 and 5 exhibit significantly less CD11b-labeled inflammatory cells to the injury site compared to WT (Day 2: p=0.0004, Day 5: p= 0.0075). Leukocyte subpopulation analysis reveals decreased neutrophil migration (Gr-1 positive cells) into the wound area of FXII KO on Day 2 [FXII KO 5669 ± 1844 vs. WT 40490 ± 8564 total surface area of Gr-1 positive cells (pixels)/ HPF, p=0.0018] and Day 5 [FXII KO 6216 ± 3829 vs. WT 34890 ± 7629 total surface area of Gr-1 positive cells (pixels)/HPF, p=0.0176]. Macrophage recruitment into the wound area, as determined by F4-80 antigenicity, increases over time, but remains significantly reduced in FXII KO mice on Day 2 [FXII KO 3111 ± 1115 vs. WT 19140 ± 5767 total surface area of F4-80 positive cells (pixels)/HPF, p=0.0348] and Day 5 [FXII KO 9377 ± 4772 vs. WT 28340 ± 4411 total surface area of F4-80 positive cells (pixels)/HPF, p=0.0171]. These studies indicate that, in FXII deficiency, there is less neutrophil and macrophage-mediated inflammation and this observation correlates with faster wound healing. Finally studies determined if the reduced inflammation seen in FXII KO is the result of host factors or bone marrow-derived cells. Adoptive bone marrow transplant (BMT) experiments were performed where WT or FXII KO bone marrow was transplanted into FXII KO hosts. Eight weeks following the BMT, mice were subjected to TG-induced peritonitis. KO/KO recipients, have significantly decreased number of PEC on day 1 (17.3 × 105/mL ± 3.894 × 105/mL cells) and day 7 (18.6 × 107/mL ± 4.0 × 107/mL cells) when compared to WT/KO recipients [Day 1: 68.25 × 105/mL ± 13.83 × 105/mL cells (p=0.041), Day 7: 190.5 × 107/mL ± 51.80 × 107/mL cells]. These data indicate that FXII interacts with neutrophils and macrophages to promote the inflammatory response; its absence causes decreased inflammatory cell recruitment. Conclusions: Our data indicate that FXII deficiency disrupts the leukocyte response to injury by reducing inflammatory cell recruitment in two murine models. Paradoxically, reduced leukocyte infiltration into skin wounds promotes healing. These investigations indicate a novel role for FXII in inflammation and wound healing and indicate a unique potential target for inflammation therapeutics. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 238-238
Author(s):  
Evi X. Stavrou ◽  
Chao Fang ◽  
Alona A. Merkulova ◽  
Lalitha V. Nayak ◽  
Howard Meyerson ◽  
...  

Abstract Introduction: Previous studies show that Factor XII (XII) participates in the inflammatory response. XII regulates the expression of monocyte FcγII receptor and stimulates monocytes and macrophages to release interleukin (IL)-1 and IL-6. XII deficient patients have reduced leukocyte migration into skin windows. In vitro, purified XIIa corrects neutrophil aggregation and degranulation defects in XII-deficient plasma. Recent studies show that leukocytes initiate and propagate venous thrombosis in vivo. We examined the contribution of XII in the inflammatory response and venous thrombosis. Methods & Results: Sterile punch biopsy wounds were created on wild type (WT) and F12-/- mice. On Days 2 and 5, there was a ~3-fold decrease in CD11b-stained cells in F12-/- woundsvs. WT. On the thioglycolate (TG)-induced sterile peritonitis assay, lavage fluid from F12-/- mice contained significantly less peritoneal exudative cells (PEC)] on days 1 and 7, (p<0.008). To determine the contribution of XII in WBC function, we used XII siRNA (Alnylam Pharmaceuticals) to create plasma XII deficiency in WT mice. After tail vein injection, plasma XII was reduced to < 5% within 24 h (T1/2 plasma XII: 6.7 h). In the TG assay, even though plasma XII is decreased to less than 5%, PEC migration is the same as in WT mice. These data suggest that the reduced leukocyte migration observed in F12-/- mice is related to altered leukocyte function. On adoptive bone marrow (BM) transplantation (BMT) experiments, WT BM transplanted into KO hosts corrects the leukocyte migration defect on the TG assay. These data suggest that there is a pool of XII associated with BM cells that is functionally distinct than plasma-derived, hepatic XII. F12 cDNA is found in leukocytes and shares sequence homology to hepatic XII. Immunofluorescence confirms XII antigen on murine BM-derived and human peripheral blood WBCs. No XII antigen is observed in BM-derived leukocytes from F12-/- mice. When WBC are activated with fMLF, XII antigen translocates to the external membrane. F12-/- PMNs have reduced chemotaxis to fMLF and adherence to several integrin-binding glycoproteins. pAktS473 mediates neutrophil cell migration, integrin activation, and cytoskeletal assembly. Normal and F12-/- PMNs exhibit pAktS473 in response to fMLF and XII. Histologically, F12-/- wounds show a smaller wound gap and a greater percentage of wound re-epithelialization than WT controls. Inferior vena cava (IVC) thrombosis induced by 90% restriction to flow at 24h contains a smaller thrombus in F12-/- than WT mice (p<0.04). Histologically, IVC thrombi from WT mice contain abundant neutrophils that are adherent to the wall and trapped within a dense fibrin network (Fig 1). siRNA treatment results in less-occlusive thrombi (n.s) with an adequate neutrophil content but a finer fibrin network (Fig 1). F12-/- thrombi are non-occlusive and contain significantly less adherent neutrophils (Fig 1). XII itself is integrally a part of neutrophil extracellular traps (NETs) in the forming thrombus and F12-/- mice have reduced NETs at sites of occlusion. WT BM transplanted into F12-/- hosts corrects the thrombus weight and degree of inflammation in F12-/- mice to normal. Likewise, F12-/- BM into WT hosts, reduces thrombus weight and degree of inflammation. Conclusions: Leukocyte XII has a dual role in neutrophil function. We hypothesize that signaling by leukocyte XII contributes to neutrophil trafficking in sites of inflammation and venous stasis. At these sites, neutrophils become indispensable for activation of both the extrinsic and intrinsic pathways of coagulation during the early formation of intraluminal fibrin and for subsequent thrombus propagation by NETs and the activation of circulating XII. Defining the signaling pathway of XII in leukocytes will further our understanding as to the mechanism(s) by which these cells cooperate to initiate and propagate venous thrombosis in vivo. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4480-4480
Author(s):  
Jason M Aliotta ◽  
Michael Del Tatto ◽  
Mark Dooner ◽  
Mandy Pereira ◽  
Peter J. Quesenberry

Abstract Abstract 4480 Objective Pulmonary complications are common in bone marrow transplant (BMT) recipients and often of an infectious etiology or attributed to intensive conditioning regimens. Cases of pulmonary arterial hypertension have been described in the setting of autologous BMT but are believed to be the result of acquired infections or previously-administered chemotherapy. Whether transplanted marrow cells themselves are toxic to the recipient's lungs is unknown. Methods To address this, non-irradiated female BALB/C mice were transplanted with 5-6×107 whole bone marrow (WBM) cells from male BALB/C mice daily for four days on week zero, then again on week eight (4.5×108 cells total/mouse), or an equal volume of diluent (control). On week 24, transplanted mice received 1000 cGy of chest-only radiation or no radiation. On week 32, histochemical and immunohistochemical analyses were performed on recipient's lungs. Results Bone marrow chimerism was not significantly different in the irradiated and non-irradiated cohorts at the time of sacrifice (average 45.11+6.25% Y chromosome+, all mice). Recipient lungs contained few non-hematopoietic donor marrow-derived cells. These cells were exclusively epithelial (Y+/cytokeratin+), primarily type II pneumocytes (Y+/prosurfactant C+), while no endothelial (Y+/von Willebrand+) or vascular smooth muscle (Y+/alpha-actin+) cells were identified. Irradiated and non-irradiated cohorts had similar quantities of these cells (0.80+0.22% vs. 0.51+0.08% Y+/cytokeratin+; 0.37+0.08% vs. 0.32+0.12% Y+/prosurfactant C+, p>0.2). Pulmonary vessel wall thickness-to-blood vessel diameter ratios (PVWT/D) were significantly increased in the non-irradiated cohort compared with controls and these ratios were further increased in the irradiated cohort (141+5.75%, 161+5.34% of control, p<0.05 comparing all groups). Conclusions These data indicated that marrow cell infusion alone results in pulmonary vascular remodeling and this effect is augmented by radiation injury. These changes are independent of transplanted marrow cell conversion to pulmonary vascular endothelial, smooth muscle cells. These studies suggest that transplanted cells may be lung toxic entities in the setting clinical BMT. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 339-339
Author(s):  
Markus Bender ◽  
Silvia Giannini ◽  
Terese Jönsson ◽  
Renata Grozovsky ◽  
Hilary Christensen ◽  
...  

Abstract Dynamins are large and highly conserved GTPases involved in endocytosis and vesicle trafficking. Mutations K562E/del in the ubiquitous dynamin 2 (DNM2) have been associated with thrombocytopenia in humans. To determine the role of DNM2 in megakaryopoiesis we generated Dnm2fl/fl Pf4-Cre mice specifically lacking DNM2 in the megakaryocyte (MK) lineage. Dnm2fl/fl Pf4-Cre mice were viable, but had severe macrothrombocytopenia with moderately accelerated platelet clearance and prolonged bleeding due to poorly functional platelets. Dnm2-null bone marrow MKs had altered demarcation membrane system, appearing at times as a compact, narrow twisting membrane system of clathrin-coated vesicles. Fetal liver cell derived Dnm2-null MKs formed proplatelets poorly in vitro, showing that DNM2 plays a major role in MK membrane formation and thrombopoiesis. Both endogenous DNM2 and overexpressed DNM2 WT, but not DNM2 K562E/del mutants localized with the early endosome in bone marrow MKs. The endocytic pathway was disrupted in Dnm2-null MKs, as evidenced by severely reduced early endosome EEA1 and APPL1 staining and weak LysoTracker internalization. Endocytosis of the thrombopoietin (TPO) receptor Mpl was impaired in Dnm2-null platelets, causing constitutive phosphorylation of the tyrosine kinase JAK2 and elevated circulating TPO levels. MK-specific DNM2 deletion severely disrupted bone marrow homeostasis, as reflected by massive MK hyperplasia and myelofibrosis, and consequent extramedullary hematopoiesis and splenomegaly. However, additional Mpl genetic deletion failed to rescue the severe splenomegaly of Dnm2fl/fl Pf4-Cre mice, and Mpl-/- Dnm2fl/fl Pf4-Cre mice instead died at 4-5 weeks of age. Taken together, our data demonstrates that unrestrained MK growth and proliferation results in rapid myelofibrosis independently of Mpl expression and other bone marrow cell types, and establishes a previously unrecognized role for DNM2-dependent endocytosis in megakaryopoiesis, thrombopoiesis and bone marrow homeostasis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3563-3563
Author(s):  
Lyla A Youssef ◽  
Stuart Phillip Weisberg ◽  
Sheila Bandyopadhyay ◽  
Eldad A. Hod ◽  
Steven L Spitalnik

Abstract Introduction Red blood cell (RBC) transfusions are a common therapy, with ~15 million RBC units administered annually in the United States. Studies in mice and dogs identified increased circulating levels of multiple inflammatory cytokines after transfusions of refrigerator storage-damaged RBCs. One chemokine identified was monocyte chemoattractant protein 1 (MCP-1; also known as CCL2). MCP-1 is the ligand for CCR2, which is expressed by several cell types including Ly6Chi (i.e., inflammatory) monocytes. One reserve site of Ly6Chi monocytes is the bone marrow, from which they emigrate into the circulation in a CCR2-dependent manner to traffic to sites of inflammation. Because the spleen is a primary site of RBC clearance, we aimed to identify splenic cells responsible for producing MCP-1 following transfusions of storage-damaged or antibody-coated RBCs. Methods Leukoreduced murine RBCs were prepared from wild-type (WT) C57BL/6 donors and refrigerator stored for 12-13 days. MCP-1 transgenic reporter mice were transfused with fresh, or storage-damaged ("old"), or fresh anti-RBC antibody-coated RBCs. Spleen, bone marrow, and blood were collected post-transfusion and cells were analyzed by multi-parameter flow cytometry. WT C57BL/6 mice were also transfused with fresh or old RBCs; splenocytes isolated post-transfusion were flow sorted followed by RNA isolation and RT-PCR. Results Splenic red pulp macrophages (VCAM1hi, F4/80hi, CD11blo) were primarily responsible for erythrophagocytosis after transfusions with old or antibody-coated RBCs. However, in each case, Ly6C hi, CD11b+ splenic inflammatory monocytes in MCP-1 reporter mice expressed MCP-1. MCP-1 expression by these cells was also confirmed in WT recipients by RT-PCR after flow sorting. Interestingly, only a small percentage of inflammatory monocytes ingested transfused RBCs. Furthermore, circulating inflammatory monocyte levels increased following transfusion of old or antibody-coated RBCs, accompanied by reduced levels in the bone marrow. Conclusions Although red pulp macrophages were the major cell type responsible for clearing transfused refrigerator-damaged or antibody-coated RBCs, a different splenic cell population (i.e., inflammatory monocytes) produced MCP-1. Thus, the splenic reserve of inflammatory monocytes produced an inflammatory response following phagocytosis-mediated acute hemolytic events. Increased numbers of circulating inflammatory monocytes and reduced numbers in the bone marrow suggest that these cells emigrated from the bone marrow, perhaps in response to MCP-1 signaling. Thus, one possible mechanism explaining our results is that erythrophagocytosis in the spleen induces an inflammatory response, triggering splenic inflammatory monocytes to synthesize MCP-1 and release it into the circulation; MCP-1 then binds CCR2 on bone marrow inflammatory monocytes, causing their egress into the circulation. The additional signals involved in these phenomena, along with their clinical relevance to transfusion medicine, require additional investigation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. SCI-23-SCI-23
Author(s):  
David Stegner ◽  
Judith van Eeuwijk ◽  
Maximilian G Gorelashvili ◽  
Oguzhan Angay ◽  
Mike Friedrich ◽  
...  

Abstract Blood platelets play key roles in hemostasis and thrombosis and are the second most abundant cell type in the circulation. Due to their short life span of only a few days, anuclear platelets are continuously replenished and thus provide a classic system to study hematopoiesis. In mammals, platelets are produced by megakaryocytes (MKs) that are predominantly residing in the bone marrow (BM). MKs originate from hematopoietic stem cells and are thought to migrate from an endosteal niche towards the vascular sinusoids during their maturation. Unfortunately, previous studies on megakaryopoiesis were often limited by 2D imaging and cutting artefacts when analyzing bone sections, potentially resulting in underestimation of MK-to-vessel contacts and MK volumes. We studied megakaryopoiesis by visualizing MKs in their 3D environment. To this end, murine bones were simultaneously stained for MKs and endothelial cells, fixed, chemically cleared and imaged by Light Sheet Fluorescence Microscopy (LSFM). Thus, we achieved 3D-reconstructions of the complete and intact bone with subcellular resolution. Through imaging of MKs in the intact BM, we show that MKs can be found within the entire BM, without a bias towards bone-distant regions. We developed and compared different image processing pipelines and simulation scenarios for precise identification of MKs in 3D light-sheet fluorescence microscopy of uncut murine bones. By combining in vivo two-photon microscopy and in situ LSFM with computational simulations, we reveal surprisingly slow MK migration, limited intervascular space, and a vessel-biased MK pool. To complement limited imaging approaches computational simulations represent an important, well-controllable tool. Typically, simulation studies use artificial meshes as templates to minimize the computational effort or due to the lack of experimental data. Unfortunately, such simplified artificial templates for MKs and the vasculature can bias simulations and lead to misinterpretations as we show here. Using the segmented cell and vessel objects of true 3D images can overcome those limitations providing a simulation framework that has the prerequisites to maximally reflect the physiological situation. Thus, imaging and simulations go hand in hand when the respective 3D cell and vessel objects perfectly serve as biological templates for advanced simulations. We demonstrate reliable whole-bone analysis in silico, and found that MKs influence neutrophil and HSC migration as biomechanical restrainers modulating cell mobility and extravasation. These data challenge the current thrombopoiesis model of MK migration and support a modified model, where MKs at sinusoids are replenished by sinusoidal precursors rather than cells from a distant periostic niche (1). Furthermore, we identify MKs as biomechanical restraints for bone marrow cell mobilization. As MKs themselves do not need to migrate to reach the vessel, therapies to increase MK numbers might be sufficient to raise platelet counts. (1) Stegner D, van Eeuwijk JMM, Angay O, Gorelashvili MG, Semeniak D, Pinnecker J, Schmithausen P, Meyer I, Friedrich M, Dütting S, Brede C, Beilhack A, Schulze H, Nieswandt B, Heinze KG. Thrombopoiesis is spatially regulated by the bone marrow vasculature, Nat Commun. 2017 8(1):127. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 2019 ◽  
pp. 1-16 ◽  
Author(s):  
Xiaoning He ◽  
Zhiwei Dong ◽  
Yina Cao ◽  
Han Wang ◽  
Shiyu Liu ◽  
...  

Mesenchymal stem cell transplantation (MSCT) promotes cutaneous wound healing. Numerous studies have shown that the therapeutic effects of MSCT appear to be mediated by paracrine signaling. However, the cell-cell interaction during MSCT between MSCs and macrophages in the region of cutaneous wound healing is still unknown. In this study, early depletion of macrophages delayed the wound repair with MSC injection, which suggested that MSC-mediated wound healing required macrophages. Moreover, we demonstrated that systemically infused bone marrow MSCs (BMMSCs) and jaw bone marrow MSCs (JMMSCs) could translocate to the wound site, promote macrophages toward M2 polarization, and enhance wound healing.In vitrococulture of MSCs with macrophages enhanced their M2 polarization. Mechanistically, we found that exosomes derived from MSCs induced macrophage polarization and depletion of exosomes of MSCs reduced the M2 phenotype of macrophages. Infusing MSCs without exosomes led to lower number of M2 macrophages at the wound site along with delayed wound repair. We further showed that the miR-223, derived from exosomes of MSCs, regulated macrophage polarization by targeting pknox1. These findings provided the evidence that MSCT elicits M2 polarization of macrophages and may accelerate wound healing by transferring exosome-derived microRNA.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1922-1922
Author(s):  
Alessandro Moscetti ◽  
Francesca Saltarelli ◽  
Maria Paola Bianchi ◽  
Guglielmo Bruno ◽  
Gerardo Salerno ◽  
...  

Abstract Abstract 1922 AL amyloidosis is a pathology characterised by the deposition of fibrillary aggregates of immunoglobuline light chains with β-sheet conformation. The light chains are synthetized by neoplastic plasma cell and fibrils deposition can infiltrate tissues leading to multi systemic organ damage. To evaluate if vascular modifications are involved in AL amyloidosis, inflammatory activity of cytokines as MCP-1 and VEGF was investigated. MCP-1 is a chemokine that activates mononuclear phagocytes by promoting leukocyte-endothelium binding and migration to sites of inflammation, while VEGF is an endothelial cell mitogen and permeability factor that is potently angiogenic in bone marrow of AL amyloidosis patients. Aim of this study is to evaluate serum cytokines MCP-1 and VEGF levels in patients with systemic or localized AL amyloidosis at presentation to find out potential differences useful to define a characteristic inflammatory pattern. Blood samples were collected from 8 patients with systemic amyloidosis and from 4 patients with localized amyloidosis and analyzed for serum MCP-1 and VEGF levels. Mann-Whitney test and Spearman correlation were used to compare results. MCP-1 level was significantly higher in the serum of patients with systemic disease (350.52±58.70 pg/ml) compared to the group of patients with localized amyloidosis (147.82±26.03; p=0.004); VEGF was also significantly increased in systemic disease group (p= 0.028). In addition, a positive correlation between MCP-1 and VEGF (r2= 0.755; p=0.031) has been found in the group of patients with systemic amyloidosis. Results seems to suggest a difference in serum cytokine MCP-1 and VEGF levels between AL systemic and localized amyloidosis. In systemic amyloidosis the neoplastic plasma cells interact with bone marrow microenvironment resulting in VEGF release leading to a new angiogenesis also supported by an inflammatory cells increase. The MCP-1 activates and promotes leukocyte-endothelium binding increasing the inflammatory process. The high correlation between MCP-1 and VEGF suggests a positive relationship between a new angiogenesis and a migration of inflammatory cells in the bone marrow stroma. On the basis of our results, MCP-1 and VEGF chemokines can be used to evaluate the inflammatory process in patient with systemic or localized AL amyloidosis. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 2019 ◽  
pp. 1-12 ◽  
Author(s):  
Jianing Ding ◽  
Xin Wang ◽  
Bi Chen ◽  
Jieyuan Zhang ◽  
Jianguang Xu

The exosomes are derived from mesenchymal stem cells (MSCs) and may be potentially used as an alternative for cell therapy, for treating diabetic wounds, and aid in angiogenesis. This study, aimed to investigate whether exosomes originated from bone marrow-derived MSCs (BMSCs) preconditioned by deferoxamine (DFO-Exos) exhibited superior proangiogenic property in wound repair and to explore the underlying mechanisms involved. Human umbilical vein endothelial cells (HUVECs) were used for assays involving cell proliferation, scratch wound healing, and tube formation. To test the effects in vivo, streptozotocin-induced diabetic rats were established. Two weeks after the procedure, histological analysis was used to measure wound-healing effects, and the neovascularization was evaluated as well. Our findings demonstrated that DFO-Exos activate the PI3K/AKT signaling pathway via miR-126 mediated PTEN downregulation to stimulate angiogenesis in vitro. This contributed to enhanced wound healing and angiogenesis in streptozotocin-induced diabetic rats in vivo. Our results suggest that, in cell-free therapies, exosomes derived from DFO preconditioned stem cells manifest increased proangiogenic ability.


Sign in / Sign up

Export Citation Format

Share Document