Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Small ; 18(1): e2104229, 2022 01.
Article in English | MEDLINE | ID: mdl-34791802

ABSTRACT

The treatment of diabetic wounds remains a major challenge in clinical practice, with chronic wounds characterized by multiple drug-resistant bacterial infections, angiopathy, and oxidative damage to the microenvironment. Herein, a novel in situ injectable HA@MnO2 /FGF-2/Exos hydrogel is introduced for improving diabetic wound healing. Through a simple local injection, this hydrogel is able to form a protective barrier covering the wound, providing rapid hemostasis and long-term antibacterial protection. The MnO2 /ε-PL nanosheet is able to catalyze the excess H2 O2 produced in the wound, converting it to O2 , thus not only eliminating the harmful effects of H2 O2 but also providing O2 for wound healing. Moreover, the release of M2-derived Exosomes (M2 Exos) and FGF-2 growth factor stimulates angiogenesis and epithelization, respectively. These in vivo and in vitro results demonstrate accelerated healing of diabetic wounds with the use of the HA@MnO2 /FGF-2/Exos hydrogel, presenting a viable strategy for chronic diabetic wound repair.


Subject(s)
Diabetes Mellitus , Exosomes , Exosomes/metabolism , Fibroblast Growth Factors/metabolism , Humans , Hydrogels , Manganese Compounds , Oxidative Stress , Oxides , Wound Healing
2.
Cell Commun Signal ; 20(1): 165, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36284303

ABSTRACT

BACKGROUND: Postmenopausal bone loss, mainly caused by excessive bone resorption mediated by osteoclasts, has become a global public health burden. Metformin, a hypoglycemic drug, has been reported to have beneficial effects on maintaining bone health. However, the role and underlying mechanism of metformin in ovariectomized (OVX)-induced bone loss is still vague. RESULTS: In this study, we demonstrated for the first time that metformin administration alleviated bone loss in postmenopausal women and ovariectomized mice, based on reduced bone resorption markers, increased bone mineral density (BMD) and improvement of bone microstructure. Then, osteoclast precursors administered metformin in vitro and in vivo were collected to examine the differentiation potential and autophagical level. The mechanism was investigated by infection with lentivirus-mediated BNIP3 or E2F1 overexpression. We observed a dramatical inhibition of autophagosome synthesis and osteoclast formation and activity. Treatment with RAPA, an autophagy activator, abrogated the metformin-mediated autophagy downregulation and inhibition of osteoclastogenesis. Additionally, overexpression of E2F1 demonstrated that reduction of OVX-upregulated autophagy mediated by metformin was E2F1 dependent. Mechanistically, metformin-mediated downregulation of E2F1 in ovariectomized mice could downregulate BECN1 and BNIP3 levels, which subsequently perturbed the binding of BECN1 to BCL2. Furthermore, the disconnect between BECN1 and BCL2 was shown by BNIP3 overexpression. CONCLUSION: In summary, we demonstrated the effect and underlying mechanism of metformin on OVX-induced bone loss, which could be, at least in part, ascribed to its role in downregulating autophagy during osteoclastogenesis via E2F1-dependent BECN1 and BCL2 downregulation, suggesting that metformin or E2F1 inhibitor is a potential agent against postmenopausal bone loss. Video abstract.


Subject(s)
Bone Resorption , Metformin , Osteoporosis, Postmenopausal , Humans , Mice , Female , Animals , Osteoclasts , Osteoporosis, Postmenopausal/metabolism , Metformin/pharmacology , Bone Resorption/drug therapy , Autophagy , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Cell Differentiation , RANK Ligand/metabolism , E2F1 Transcription Factor/metabolism
3.
FASEB J ; 34(4): 5208-5222, 2020 04.
Article in English | MEDLINE | ID: mdl-32060985

ABSTRACT

Emerging evidence highlights the role of the long noncoding RNA (lncRNA) KCNQ1OT1 in fracture healing. Osteoblast proliferation, migration, and survival are pivotal during this process. In this study, we aimed to improve our understanding of the regulatory role of lncRNA KCNQ1OT1 during osteoblast proliferation, migration, and survival. We searched the gene expression omnibus databases and LncBase Experimental V.2 to identify key microRNAs (miRNAs) targets of KCNQ1OT1. MiR-701-3p was selected as a differentially expressed miRNA and RNA immunoprecipitation assays were performed to verify its interaction with KCNQ1OT1. Fibroblast growth factor receptor 3 (FGFR3) was also identified as a target of miR-701-3p. We further identified KCNQ1OT1 as a competing endogenous RNA of miR-701-3p that could influence osteoblast proliferation, migration, and apoptosis in vitro and in vivo. Taken together, our results indicate that the KCNQ1OT1/miR-701-3p/FGFR3 axis is an important regulator of osteoblast proliferation, migration, and apoptosis, and provide a new therapeutic avenue for fracture healing.


Subject(s)
Disease Models, Animal , Femoral Fractures/therapy , Fracture Healing/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Animals , Apoptosis , Cell Proliferation , Femoral Fractures/pathology , Male , Mice , Mice, Inbred C57BL , Receptor, Fibroblast Growth Factor, Type 3/genetics , Signal Transduction
4.
J Nanobiotechnology ; 19(1): 150, 2021 May 21.
Article in English | MEDLINE | ID: mdl-34020670

ABSTRACT

BACKGROUND: Enhanced angiogenesis can promote diabetic wound healing. Mesenchymal stem cells (MSCs)-derived exosomes, which are cell-free therapeutics, are promising candidates for the treatment of diabetic wound healing. The present study aimed to investigate the effect of exosomes derived from MSCs pretreated with pioglitazone (PGZ-Exos) on diabetic wound healing. RESULTS: We isolated PGZ-Exos from the supernatants of pioglitazone-treated BMSCs and found that PGZ-Exos significantly promote the cell viability and proliferation of Human Umbilical Vein Vascular Endothelial Cells (HUVECs) injured by high glucose (HG). PGZ-Exos enhanced the biological functions of HUVECs, including migration, tube formation, wound repair and VEGF expression in vitro. In addition, PGZ-Exos promoted the protein expression of p-AKT, p-PI3K and p-eNOS and suppressed that of PTEN. LY294002 inhibited the biological function of HUVECs through inhibition of the PI3K/AKT/eNOS pathway. In vivo modeling in diabetic rat wounds showed that pioglitazone pretreatment enhanced the therapeutic efficacy of MSCs-derived exosomes and accelerated diabetic wound healing via enhanced angiogenesis. In addition, PGZ-Exos promoted collagen deposition, ECM remodeling and VEGF and CD31 expression, indicating adequate angiogenesis in diabetic wound healing. CONCLUSIONS: PGZ-Exos accelerated diabetic wound healing by promoting the angiogenic function of HUVECs through activation of the PI3K/AKT/eNOS pathway. This offers a promising novel cell-free therapy for treating diabetic wound healing.


Subject(s)
Diabetes Mellitus/metabolism , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Pioglitazone/metabolism , Pioglitazone/pharmacology , Wound Healing/drug effects , Angiogenesis Inducing Agents/pharmacology , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Collagen/metabolism , Diabetes Mellitus, Experimental , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Male , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Sprague-Dawley , Skin/drug effects
5.
J Cell Mol Med ; 24(11): 6385-6396, 2020 06.
Article in English | MEDLINE | ID: mdl-32307908

ABSTRACT

N6-methyladenosine (m6A) modification has been reported in various diseases and implicated in increasing numbers of biological processes. However, previous studies have not focused on the role of m6A modification in fracture healing. Here, we demonstrated that m6A modifications are decreased during fracture healing and that methyltransferase-like 3 (METTL3) is the main factor involved in the abnormal changes in m6A modifications. Down-regulation of METTL3 promotes osteogenic processes both in vitro and in vivo, and this effect is recapitulated by the suppression of miR-7212-5p maturation. Further studies have shown that miR-7212-5p inhibits osteoblast differentiation in MC3T3-E1 cells by targeting FGFR3. The present study demonstrated an important role of the METTL3/miR-7212-5p/FGFR3 axis and provided new insights on m6A modification in fracture healing.


Subject(s)
Adenosine/analogs & derivatives , Cell Differentiation/genetics , Fracture Healing/genetics , Methyltransferases/metabolism , MicroRNAs/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Adenosine/metabolism , Animals , Cell Line , Gene Expression Regulation , Methylation , Methyltransferases/genetics , Mice, Inbred C57BL , MicroRNAs/genetics , RNA-Binding Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism
6.
J Cell Mol Med ; 24(1): 1076-1086, 2020 01.
Article in English | MEDLINE | ID: mdl-31755174

ABSTRACT

Interleukin-10 (IL-10) displays well-documented anti-inflammatory effects, but its effects on osteoblast differentiation have not been investigated. In this study, we found IL-10 negatively regulates microRNA-7025-5p (miR-7025-5p), the down-regulation of which enhances osteoblast differentiation. Furthermore, through luciferase reporter assays, we found evidence that insulin-like growth factor 1 receptor (IGF1R) is a miR-7025-5p target gene that positively regulates osteoblast differentiation. In vivo studies indicated that the pre-injection of IL-10 leads to increased bone formation, while agomiR-7025-5p injection delays fracture healing. Taken together, these results indicate that IL-10 induces osteoblast differentiation via regulation of the miR-7025-5p/IGF1R axis. IL-10 therefore represents a promising therapeutic strategy to promote fracture healing.


Subject(s)
Cell Differentiation , Fibroblasts/cytology , Fracture Healing/drug effects , Fractures, Bone/drug therapy , Interleukin-10/pharmacology , Osteoblasts/cytology , Osteogenesis , Animals , Fibroblasts/drug effects , Fibroblasts/metabolism , Fractures, Bone/metabolism , Fractures, Bone/pathology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Osteoblasts/drug effects , Osteoblasts/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism
7.
Small ; 16(3): e1904044, 2020 01.
Article in English | MEDLINE | ID: mdl-31867895

ABSTRACT

At present, developing therapeutic strategies to improve wound healing in individuals with diabetes remains challenging. Exosomes represent a promising nanomaterial from which microRNAs (miRNAs) can be isolated. These miRNAs have the potential to exert therapeutic effects, and thus, determining the potential therapeutic contributions of specific miRNAs circulating in exosomes is of great importance. In the present study, circulating exosomal miRNAs are identified in diabetic patients and assessed for their roles in the context of diabetic wound healing. A significant upregulation of miR-20b-5p is observed in exosomes isolated from patients with type 2 diabetes mellitus (T2DM), and this miRNA is able to suppress human umbilical vein endothelial cell angiogenesis via regulation of Wnt9b/ß-catenin signaling. It is found that the application of either miR-20b-5p or diabetic exosomes to wound sites is sufficient to slow wound healing and angiogenesis. In diabetic mice, it is found that knocking out miR-20b-5p significantly enhances wound healing and promotes wound angiogenesis. Together, these findings thus provide strong evidence that miR-20b-5p is highly enriched in exosomes from patients with T2DM and can be transferred to cells of the vascular endothelium, where it targets Wnt9b signaling to negatively regulate cell functionality and angiogenesis.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Exosomes/metabolism , MicroRNAs/antagonists & inhibitors , Wnt Proteins/metabolism , Wound Healing , Animals , Case-Control Studies , Diabetes Mellitus, Type 2/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , MicroRNAs/blood
8.
J Nanobiotechnology ; 18(1): 68, 2020 May 06.
Article in English | MEDLINE | ID: mdl-32375794

ABSTRACT

BACKGROUND: Enhancing angiogenesis is critical for accelerating wound healing. Application of different types of exosomes (Exos) to promote angiogenesis represents a novel strategy for enhanced wound repair. Saliva is known to accelerate wound healing, but the underlying mechanisms remain unclear. RESULTS: Our results have demonstrated that saliva-derived exosomes (saliva-Exos) induce human umbilical vein endothelial cells (HUVEC) proliferation, migration, and angiogenesis in vitro, and promote cutaneous wound healing in vivo. Further experiments documented that Ubiquitin-conjugating enzyme E2O (UBE2O) is one of the main mRNAs of saliva-Exos, and activation of UBE2O has effects similar to those of saliva-Exos, both in vitro and in vivo. Mechanistically, UBE2O decreases the level of SMAD family member 6 (SMAD6), thereby activating bone morphogenetic protein 2 (BMP2), which, in turn, induces angiogenesis. CONCLUSIONS: The present work suggests that administration of saliva-Exos and UBE2O represents a promising strategy for enhancing wound healing through promotion of angiogenesis.


Subject(s)
Exosomes/enzymology , Neovascularization, Physiologic/drug effects , Saliva/enzymology , Smad6 Protein/metabolism , Ubiquitin-Conjugating Enzymes , Animals , Cells, Cultured , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Saliva/cytology , Skin/injuries , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/pharmacology , Wound Healing/drug effects
9.
Arch Orthop Trauma Surg ; 140(1): 11-17, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31127408

ABSTRACT

OBJECTIVE: To compare the clinical effect of 3D-printed template technology with X-ray fluoroscopy in assisting surgery for sacroiliac screws placement. DESIGN: Institutional review board-approved retrospective analysis. PATIENTS: The clinical data of 31 cases of sacroiliac complex injury between January 2015 and December 2016 were analyzed. There were 16 patients, males 11 and females 5, who underwent surgery assisted by 3D-printed template in template group, and that of contemporaneous 15 patients, males 11 and females 4, who underwent traditional surgery were gathered as fluoroscopy group. All those patients were followed up for more than 6 months. MAIN OUTCOME MEASURES: The operation time and X-ray fluoroscopy times for each screw placement, and the Matta and Majeed score were analyzed and the difference between the two group was tested. RESULTS: All cases were followed up for 6-20 months, average 11.4 ± 0.6 months. In template group, 19 screws were implanted. Each screw spent 25-38 min, average 27.2 ± 5.3 min, and need 2-5 times fluoroscopy, average 2.7 ± 0.5. The fracture reduction quality was evaluated by Matta score scale: excellent 10, well 4, fair 2, good rate 87.5%; and pelvic function were evaluated by Majeed score scale: excellent 11, well 3, fair 2, and good rate 87.5%. In fluoroscopy group, 17 screws were implanted. Each screw spent 45-70 min, average 60.3 ± 5.8 min, and needs 11-23 times fluoroscopy, average 15.4 ± 3.5. The fracture reduction quality was evaluated by Matta score scale: excellent 7, well 6, fair 2, and good rate 86.7%; and pelvic function was evaluated by Majeed score scale: excellent 6, well 6, fair 3, and good rate 80.0%. The difference in operation time, X-ray fluoroscopy times between template group and fluoroscopy group had statistical significance. But the Matta and Majeed score had no difference between two groups. CONCLUSION: Compared with traditional surgery, 3D-printed template technology-assisted surgery for sacroiliac screws placement in sacroiliac complex injury patients possesses advantage such as shortened operation time and reduced X-ray exposure times. This technology improves the safety profile of this operation and should be further studied in future clinical applications.


Subject(s)
Bone Screws , Fluoroscopy/methods , Ilium , Printing, Three-Dimensional , Sacrum , Female , Fracture Fixation/instrumentation , Fracture Fixation/methods , Fractures, Bone/diagnostic imaging , Fractures, Bone/surgery , Humans , Ilium/diagnostic imaging , Ilium/injuries , Ilium/surgery , Male , Retrospective Studies , Sacrum/diagnostic imaging , Sacrum/injuries , Sacrum/surgery , Surgery, Computer-Assisted/methods
10.
J Cell Biochem ; 120(5): 7741-7750, 2019 May.
Article in English | MEDLINE | ID: mdl-30506715

ABSTRACT

The beneficial effects of icariin in the management of many diseases, such as chronic renal failure and heart failure, are well known. Icariin has also been shown to ameliorate osteoarthritis (OA) symptoms; however, the underlying mechanisms remain unclear. In this study, a bioinformatics analysis was performed to investigate the KEGG pathways of icariin-targeted genes involved in OA. Our study suggests that icariin plays a role in OA by regulating inflammatory cytokine production, insulin resistance, and cell survival through modulation of the NF-κB, MAPK, and Akt signaling pathways. Importantly, IKBKB, NFKBIA, MAPK8, MAPK9, and MAPK10 may be the hub genes affected by icariin when providing its beneficial effects on OA. In addition, we found that icariin decreases proinflammatory factors and inhibits chondrocyte apoptosis through suppression of the NF-κB pathway. Our study highlights a set of KEGG pathways that could explain the molecular mechanism of icariin's action on OA, suggesting that icariin could be considered as a promising therapeutic option for OA.

11.
Arch Orthop Trauma Surg ; 139(12): 1763-1770, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31435724

ABSTRACT

BACKGROUND: Shock-wave therapy (SWT) has been widely applied and proven to be an effective treatment in ameliorating symptoms of plantar fasciitis (PF). Ultrasound therapy (UT) is another common treatment of PF, and several researches have documented its advantages when compared to corticosteroid injection. Despite this, few studies have focused on comparing the use of SWT and UT in the treatment of PF. The purpose of our meta-analysis is to evaluate whether SWT is better than UT in managing PF, both in terms of ameliorating pain and improving functionality. METHODS: A systematic search of the literature was conducted to identify relevant articles that were published in Pubmed, Medline, Embase, the Cochrane Library, SpringerLink, Clinical Trials.gov and OVID from the databases' inception to October 2018. All studies comparing the efficacy of SWT and UT in terms of pain levels and functionality improvement were included. Data on the two primary outcomes were collected and analyzed using the Review Manager 5.3. RESULTS: Five studies were included in the current meta-analysis. A significant difference in VAS score (MD = - 13.14, Cl - 14.07 to - 12.75 P < 0.00001, I2 = 100%) was noted between the SWT group and the UT group. No significant difference was seen in the AOFAS (MD = 3.19, Cl - 1.72 to 8.10 P = 0.20, I2 = 100%); FFI or PFPS score was not found significant difference either (SMD = - 1.17, Cl - 4.45 to 2.10 P = 0.48, I2 = 96%). CONCLUSIONS: The results from this meta-analysis highlight the effectiveness of both SWT and UT in the treatment of PF. Although inter-group differences were not significant, the VAS score was better improved in the SWT group, suggesting that SWT may be a superior alternative for the treatment of PF.


Subject(s)
Extracorporeal Shockwave Therapy/methods , Fasciitis, Plantar/therapy , Pain Management/methods , Adrenal Cortex Hormones/therapeutic use , Fasciitis, Plantar/complications , Fasciitis, Plantar/physiopathology , Humans , Pain Measurement , Randomized Controlled Trials as Topic , Ultrasonic Therapy/methods
12.
Arch Orthop Trauma Surg ; 139(4): 529-536, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30426211

ABSTRACT

BACKGROUND: Corticosteroid (CS) injections have been proven to be effective in ameliorating symptoms of plantar fasciitis. Shock-wave (SW) therapy is another common treatment of plantar fasciitis, and several meta-analyses have documented its advantages when compared to placebo treatment. Despite this, few studies have focused on comparing the use of CS and SW in the treatment of plantar fasciitis. The purpose of this meta-analysis is to assess whether SW is superior to CS in managing plantar fasciitis, both in terms of ameliorating pain as well as improving functionality. METHODS: A systematic search of the literature was conducted to identify relevant articles that were published in Pubmed, Medline, Embase, the Cochrane Library, SpringerLink, Clinical Trials.gov and OVID from the databases' inception to July 2018. All studies comparing the efficacy of SW and CS in terms of pain levels and functionality improvement were included. Data on the two primary outcomes were collected and analyzed using the Review Manager 5.3. RESULTS: Six studies were included in the current meta-analysis. A significant difference in VAS score (MD = - 0.96, Cl - 1.28 to - 0.63, P < 0.00001, I2 = 96%) was noted between the SW group and the CS group. No significant difference was seen in the Mayo CSS or FFI or HFI or 100 Scoring System score at the 3 months follow-up (Chi2 = 0.62, I2 = 0%, P > 0.05). CONCLUSIONS: The clinical relevance of the present study is that both SW and CS were effective and successful in relieving pain and improving self-reported function in the treatment of plantar fasciitis at 3 months. Although inter-group differences were not significant, the VAS score was better improved in the SW group, highlighting that shock-wave therapy may be a better alternative for the management of chronic plantar fasciitis.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Extracorporeal Shockwave Therapy , Fasciitis, Plantar/therapy , Humans , Pain , Randomized Controlled Trials as Topic , Treatment Outcome
13.
Cell Physiol Biochem ; 51(2): 647-663, 2018.
Article in English | MEDLINE | ID: mdl-30463067

ABSTRACT

BACKGROUND/AIMS: Antimicrobial peptides are effective promoters of wound healing but are susceptible to degradation. In this study, we replaced the GIGDP unit on the N-terminal of the endogenous human antimicrobial peptide hBD-2 with APKAM to produce A-hBD-2 and analyzed the effect on wound healing both in vitro and in vivo. METHODS: The effects of A-hBD-2 and hBD-2 on cytotoxicity and proliferation in keratinocytes were assessed by Cell Counting Kit-8 assay. The structural stability and antimicrobial activity of hBD-2 and A-hBD-2 were evaluated against Staphylococcus aureus. RNA and proteins levels were evaluated by real-time PCR and western blotting, respectively. Cell migration was evaluated using a transwell assay. Cell cycle analysis was performed by flow cytometry. Wound healing was assessed in Sprague-Dawley rats. Epidermal thickness was evaluated by hematoxylin and eosin staining. RESULTS: We found that hBD-2 exhibited cytotoxicity at high concentrations and decreased the structural stability in the presence of high sodium chloride concentrations. A-hBD-2 exhibited increased structural stability and antimicrobial activity, and had lower cytotoxicity in keratinocytes. A-hBD-2 increased the migration and proliferation of keratinocytes via phosphorylation of EGFR and STAT3 and suppressed terminal differentiation of keratinocytes. We also found that A-hBD-2 elicited mobilization of intracellular Ca2+ and stimulated keratinocytes to produce pro- and anti-inflammatory cytokines and chemokines via phospholipase C activation. Furthermore, A-hBD-2 promoted wound healing in vivo. CONCLUSION: Our data suggest that A-hBD-2 may be a promising candidate therapy for wound healing.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Cell Proliferation/drug effects , Wound Healing/drug effects , beta-Defensins/pharmacology , Animals , Calcium/chemistry , Calcium/metabolism , Cell Cycle Checkpoints/drug effects , Cell Movement , Chemokines/metabolism , Cytokines/metabolism , ErbB Receptors/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratins/metabolism , Male , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Skin/pathology , beta-Defensins/chemistry
15.
Nat Commun ; 15(1): 5460, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937462

ABSTRACT

Developing superporous hemostatic sponges with simultaneously enhanced permeability and mechanical properties remains challenging but highly desirable to achieve rapid hemostasis for non-compressible hemorrhage. Typical approaches to improve the permeability of hemostatic sponges by increasing porosity sacrifice mechanical properties and yield limited pore interconnectivity, thereby undermining the hemostatic efficacy and subsequent tissue regeneration. Herein, we propose a temperature-assisted secondary network compaction strategy following the phase separation-induced primary compaction to fabricate the superporous chitosan sponge with highly-interconnected porous structure, enhanced blood absorption rate and capacity, and fatigue resistance. The superporous chitosan sponge exhibits rapid shape recovery after absorbing blood and maintains sufficient pressure on wounds to build a robust physical barrier to greatly improve hemostatic efficiency. Furthermore, the superporous chitosan sponge outperforms commercial gauze, gelatin sponges, and chitosan powder by enhancing hemostatic efficiency, cell infiltration, vascular regeneration, and in-situ tissue regeneration in non-compressible organ injury models, respectively. We believe the proposed secondary network compaction strategy provides a simple yet effective method to fabricate superporous hemostatic sponges for diverse clinical applications.


Subject(s)
Chitosan , Hemostasis , Hemostatics , Permeability , Animals , Porosity , Chitosan/chemistry , Hemostatics/chemistry , Hemostatics/pharmacology , Swine , Hemostasis/physiology , Hemorrhage/therapy , Male
16.
Signal Transduct Target Ther ; 9(1): 24, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38246920

ABSTRACT

The clinical role and underlying mechanisms of valproic acid (VPA) on bone homeostasis remain controversial. Herein, we confirmed that VPA treatment was associated with decreased bone mass and bone mineral density (BMD) in both patients and mice. This effect was attributed to VPA-induced elevation in osteoclast formation and activity. Through RNA-sequencing, we observed a significant rise in precursor miR-6359 expression in VPA-treated osteoclast precursors in vitro, and further, a marked upregulation of mature miR-6359 (miR-6359) in vivo was demonstrated using quantitative real-time PCR (qRT-PCR) and miR-6359 fluorescent in situ hybridization (miR-6359-FISH). Specifically, the miR-6359 was predominantly increased in osteoclast precursors and macrophages but not in neutrophils, T lymphocytes, monocytes and bone marrow-derived mesenchymal stem cells (BMSCs) following VPA stimulation, which influenced osteoclast differentiation and bone-resorptive activity. Additionally, VPA-induced miR-6359 enrichment in osteoclast precursors enhanced reactive oxygen species (ROS) production by silencing the SIRT3 protein expression, followed by activation of the MAPK signaling pathway, which enhanced osteoclast formation and activity, thereby accelerating bone loss. Currently, there are no medications that can effectively treat VPA-induced bone loss. Therefore, we constructed engineered small extracellular vesicles (E-sEVs) targeting osteoclast precursors in bone and naturally carrying anti-miR-6359 by introducing of EXOmotif (CGGGAGC) in the 3'-end of the anti-miR-6359 sequence. We confirmed that the E-sEVs exhibited decent bone/osteoclast precursor targeting and exerted protective therapeutic effects on VPA-induced bone loss, but not on ovariectomy (OVX) and glucocorticoid-induced osteoporotic models, deepening our understanding of the underlying mechanism and treatment strategies for VPA-induced bone loss.


Subject(s)
Extracellular Vesicles , MicroRNAs , Female , Humans , Animals , Mice , Valproic Acid/pharmacology , Antagomirs , In Situ Hybridization, Fluorescence , Extracellular Vesicles/genetics , MicroRNAs/genetics
17.
Theranostics ; 13(6): 2015-2039, 2023.
Article in English | MEDLINE | ID: mdl-37064871

ABSTRACT

Increasing data reveals that gelatin that has been methacrylated is involved in a variety of physiologic processes that are important for therapeutic interventions. Gelatin methacryloyl (GelMA) hydrogel is a highly attractive hydrogels-based bioink because of its good biocompatibility, low cost, and photo-cross-linking structure that is useful for cell survivability and cell monitoring. Methacrylated gelatin (GelMA) has established itself as a typical hydrogel composition with extensive biomedical applications. Recent advances in GelMA have focused on integrating them with bioactive and functional nanomaterials, with the goal of improving GelMA's physical, chemical, and biological properties. GelMA's ability to modify characteristics due to the synthesis technique also makes it a good choice for soft and hard tissues. GelMA has been established to become an independent or supplementary technology for musculoskeletal problems. Here, we systematically review mechanism-of-action, therapeutic uses, and challenges and future direction of GelMA in musculoskeletal disorders. We give an overview of GelMA nanocomposite for different applications in musculoskeletal disorders, such as osteoarthritis, intervertebral disc degeneration, bone regeneration, tendon disorders and so on.


Subject(s)
Intervertebral Disc Degeneration , Nanocomposites , Humans , Gelatin/chemistry , Hydrogels/chemistry , Tissue Engineering/methods
18.
ACS Appl Mater Interfaces ; 14(30): 34427-34442, 2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35866896

ABSTRACT

Fracture nonunion can result in considerable physical harm and limitation of quality of life in patients, exerting an extensive economic burden to the society. Nonunion largely results from unresolved inflammation and impaired osteogenesis. Despite advancements in surgical techniques, the indispensable treatment for nonunion is robust anti-inflammation therapy and the promotion of osteogenic differentiation. Herein, we report that plasma exosomes derived from infected fracture nonunion patients (Non-Exos) delayed fracture repair in mice by inhibiting the osteogenic differentiation of bone marrow stromal cells in vivo and in vitro. Unique molecular identifier microRNA-sequencing (UID miRNA-seq) suggested that microRNA-708-5p (miR-708-5p) was overexpressed in Non-Exos. Mechanistically, miR-708-5p targeted structure-specific recognition protein 1, thereby suppressing the Wnt/ß-catenin signaling pathway, which, in turn, impaired osteogenic differentiation. AntagomicroRNA-708-5p (antagomiR-708-5p) could partly reverse the above process. A bacteria-sensitive natural polymer hyaluronic-acid-based hydrogel (HA hydrogel) loaded with antagomiR-708-5p exhibited promising effects in an in vivo study through antibacterial and pro-osteogenic differentiation functions in infected fractures. Overall, the effectiveness and reliability of an injectable bacteria-sensitive hydrogel with sustained release of agents represent a promising approach for infected fractures.


Subject(s)
Fractures, Bone , MicroRNAs , Animals , Antagomirs , Bacteria/metabolism , Cell Differentiation/genetics , Delayed-Action Preparations/pharmacology , Fractures, Bone/drug therapy , Humans , Hydrogels/pharmacology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Osteogenesis/genetics , Quality of Life , Reproducibility of Results
19.
Front Surg ; 9: 984431, 2022.
Article in English | MEDLINE | ID: mdl-36157415

ABSTRACT

Background: A coronal comminuted femoral intertrochanteric fracture is a special type of fracture that easily leads to internal fixation failure, and the current internal fixation techniques remain controversial. This study aims to evaluate the effect of traction-bed-assisted reduction and double-plate internal fixation in the treatment of comminuted and coronally split intertrochanteric femoral fracture. Method: Retrospective analyses of the clinical data of 83 patients diagnosed with, and treated for, comminuted and coronally split intertrochanteric femoral fracture from December 2017 to November 2019 were conducted. Among the total number of 83 patients, 40 patients received traction-bed-assisted reduction and PFNA fixation (the control group), whereas 43 patients received traction-bed-assisted reduction and double-plate internal fixation (the experimental group). The major indicators for the research analysis such as the general information of patients, perioperative data, and follow-up data of both groups were collected, sorted out, and meticulously analyzed. Results: The time taken for traction-bed-assisted reduction and double-plate intern fixation in the experimental group was significantly shorter than that in the control group (P < .05). The post-operative Harris Hip Score (HHS) at 3 months and at the final follow-up after the surgery was significantly better in the experimental group compared with that in the control group, both of which were statistically significant (P < .05). However, there were statistically no significant differences between the two groups in terms of preoperative hemoglobin (Hb) level, amount of intraoperative total blood loss, immediate post-operative Hb level, incidence of wound infection within 14 days post-operatively, time taken to step up on the ground after surgery, HHS 2 weeks after surgery, time taken for fracture healing, and the incidence of complications (P > .05). Conclusion: The use of a traction bed to achieve adequate reduction, followed by internal fixation using double plates, comparatively takes less time for both reduction and operation in the treatment of comminuted and coronally split intertrochanteric femoral fractures, which also restores proper hip joint movements relatively early and hence provides better hip joint functions in the long run.

20.
Front Cell Dev Biol ; 10: 826023, 2022.
Article in English | MEDLINE | ID: mdl-35445030

ABSTRACT

With the worldwide aging population, the prevalence of osteoporosis is on the rise, particularly the number of postmenopausal women with the condition. However, the various adverse side effects associated with the currently available treatment options underscore the need to develop novel therapies. In this study, we investigated the use of AQX-1125, a novel clinical-stage activator of inositol phosphatase-1 (SHIP1), in ovariectomized (OVX) mice, identifying a protective role. We then found that the effect was likely due to increased osteogenesis and mineralization and decreased osteoclastogenesis caused by AQX-1125 in a time- and dose-dependent manner. The effect against OVX-induced bone loss was identified to be SHIP1-dependent as pretreatment of BMSCs and BMMs with SHIP1 RNAi could greatly diminish the osteoprotective effects. Furthermore, SHIP1 RNAi administration in vivo induced significant bone loss and decreased bone mass. Mechanistically, AQX-1125 upregulated the expression level and activity of SHIP1, followed upregulating the phosphorylation levels of PI3K and Akt to promote osteoblast-related gene expressions, including Alp, cbfa1, Col1a1, and osteocalcin (OCN). NF-κB signaling was also inhibited through suppression of the phosphorylation of IκBα and P65 induced by RANKL, resulting in diminished osteoclastogenesis. Taken together, our results demonstrate that AQX-1125 may be a promising candidate for preventing and treating bone loss.

SELECTION OF CITATIONS
SEARCH DETAIL