Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Control Release ; 372: 251-264, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38908755

ABSTRACT

Drug-loaded porous membranes have been deemed to be effective physicochemical barriers to separate postoperative adhesion-prone tissues in tendon healing. However, cell viability and subsequent tissue regeneration might be severely interfered with the unrestricted release and the locally excessive concentration of anti-inflammatory drugs. Herein, we report a double-layered membrane with sustained and uni-directional drug delivery features to prevent peritendinous adhesion without hampering the healing outcome. A vortex-assisted electrospinning system in combination with ibuprofen (IBU)-in-water emulsion was utilized to fabricate IBU-loaded poly-ʟ-lactic-acid (PLLA) fiber bundle membrane (PFB-IBU) as the anti-adhesion layer. The resultant highly porous structure, oleophilic and hydrophobic nature of PLLA fibers enabled in situ loading of IBU with a concentration gradient across the membrane thickness. Aligned collagen nanofibers were further deposited at the low IBU concentration side of the membrane for regulating cell growth and achieving uni-directional release of IBU. Drug release kinetics showed that the release amount of IBU from the high concentration side reached 79.32% at 14 d, while it was only 0.35% at the collagen side. Therefore, fibroblast proliferation at the high concentration side was successfully inhibited without affecting the oriented growth of tendon-derived stem cells at the other side. In vivo evaluation of the rat Achilles adhesion model confirmed the successful peritendinous anti-adhesion of our double-layered membrane, in that the macrophage recruitment, the inflammatory factor secretion and the deposition of pathological adhesion markers such as α-SMA and COL-III were all inhibited, which greatly improved the peritendinous fibrosis and restored the motor function of tendon.

2.
Heliyon ; 9(11): e21411, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37954299

ABSTRACT

Background: Inadequate repair capacity and disturbed immune compartments are the main pathological causes of tendinopathy. Transplantation of mesenchymal stem cells (MSCs) become an effective clinic option to alleviate tendinopathy. Interleukin-1ß (IL-1ß) could confer on MSCs enhanced immunoregulatory capability to remodel the repair microenvironment favoring tissue repair. Therefore, IL-1ß activated UC-MSCs (1ßUC-MSCs) may exert favorable efficacy in promoting tendon repair in a preclinical tendinopathy rat model. Methods: Tendon-derived stem cells (TDSCs) were isolated and characterized. In vitro, the levels of immunoregulatory-related cytokines such as IL-1ß, IL-6, IL-10, and TGF-ß secreted by 1ßUC-MSCs and unprimed UC-MSCs was measured. And tendon-specific markers expressed by TDSCs cultured with primed cultured medium (CM) or unprimed CM were detected. In vivo, Achilles tendinopathy was induced by 30 µL collagenase I injection in Sprague Dawley rats. One week later, the rats were randomly injected with UC-MSCs primed with IL-1ß (106 cells per tendon), UC-MSCs, or PBS. After rats were sacrificed, histological evaluation, electron microscopy, biomechanical tests, gait performance were conducted to evaluate the structural and functional recovery of Achilles tendons. The inflammation and metabolic state of the extracellular matrix, and the potential mechanism were assessed by immunohistochemical staining and Western blot. Results: UC-MSCs were activated by IL-1ß to secrete higher levels of IL-10 and TGF-ß while the secretion levels of IL-6 and IL-1ß were not changed significantly, promoting a higher expression level of COL I and TNMD in TDSCs under proinflammatory environment. In vivo, the transplanted 1ßUC-MSCs could survive up to 5 weeks after injection with tenogenic differentiation and improved tendon healing histologically semi-quantified by modified Bonar scores. This structural regeneration was further confirmed by observation of ultrastructural morphology, and led to good functional recovery including improved biomechanical properties and gait performance. During this process, the inflammatory response and metabolism of the extracellular matrix was improved through TGF-ß/IL-10 pathway. Conclusion: This study demonstrated that the transplantation of UC-MSCs activated by IL-1ß exhibited satisfactory ability for promoting tendon functional repair in a tendinopathy rat model. During this process, the balance of inflammatory response and extracellular matrix metabolism was remodeled, and the TGF-ß/Smad2/3 and IL-10 signaling pathways were activated simultaneously. We cautiously conclude that the IL-1ß primed UC-MSCs could be a promising strategy for enhancing the ability of MSCs to treat tendinopathy.

3.
IUBMB Life ; 75(12): 1003-1016, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37503658

ABSTRACT

Tendinopathy is a condition characterized by chronic, complex, and multidimensional pathological changes in the tendons. The etiology of tendinopathy is the combination of several factors, and diabetes mellitus (DM) is a risk factor. Increasing evidence has shown that the diabetic microenvironment plays an important role in tendinopathy. However, the mechanism causing tendinopathy in patients with DM remains unclear. Our study found that ferroptosis played an important role in tendinopathy in patients with DM. In vitro, high glucose and high fat treatment was used to simulate the DM microenvironment. Results showed that such a mechanism significantly increased ferroptosis, which was characterized by mass cell death, lipid peroxide accumulation, mitochondrial morphological changes, mitochondrial membrane potential decline, iron overload, and the activation of ferroptosis-related genes, in tendon-derived stem cells cultured in vitro. In the animal studies, db/db mice were used in the DM model, and the db mice had severe tendon injury and high ACSL4 and TfR1 expressions. These phenomena could be alleviated by the ferroptosis inhibitor ferrostatin-1. In conclusion, ferroptosis is associated with tendinopathy in patients with DM, and ferroptosis targeting may be a novel approach for treating diabetic tendinopathy. Our results can provide a new strategy for managing tendinopathy clinically in patients with DM.


Subject(s)
Diabetes Mellitus , Ferroptosis , Hypercholesterolemia , Tendinopathy , Humans , Mice , Animals , Ferroptosis/genetics , Tendons/metabolism , Diabetes Mellitus/pathology , Hypercholesterolemia/metabolism , Tendinopathy/pathology , Stem Cells/metabolism
4.
Adv Sci (Weinh) ; 10(19): e2207383, 2023 07.
Article in English | MEDLINE | ID: mdl-37204068

ABSTRACT

Heterotopic ossification (HO) represents an unwanted ossific wound healing response to the soft tissue injury which caused catastrophic limb dysfunction. Recent studies established the involvement of inflammation and cellular senescence in the tissue healing process, though their role in HO still remained to be clarified. Here, a novel crosstalk where the pyroptotic macrophages aroused tendon-derived stem cells (TDSCs) senescence is revealed to encourage osteogenic healing during trauma-induced HO formation. Macrophage pyroptosis blockade reduces the senescent cell burden and HO formation in NLRP3 knockout mice. Pyroptosis-driven IL-1ß and extracellular vesicles (EVs) secretion from macrophages are determined to motivate TDSCs senescence and resultant osteogenesis. Mechanistically, pyroptosis in macrophages enhances the exosomal release of high mobility group protein 1 (HMGB1), which directly bounds TLR9 in TDSCs to trigger morbid signaling. NF-κB signaling is confirmed to be the converging downstream pathway of TDSCs in response to HMGB1-containing EVs and IL-1ß. This study adds insights into aberrant regeneration-based theory for HO formation and boosts therapeutic strategy development.


Subject(s)
HMGB1 Protein , Ossification, Heterotopic , Animals , Mice , Cellular Senescence , HMGB1 Protein/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Ossification, Heterotopic/etiology , Ossification, Heterotopic/metabolism , Wound Healing
5.
Adv Sci (Weinh) ; 9(31): e2202542, 2022 11.
Article in English | MEDLINE | ID: mdl-36000796

ABSTRACT

Tendon injury is a tricky and prevalent motor system disease, leading to compromised daily activity and disability. Insufficient regenerative capability and dysregulation of immune microenvironment are the leading causes of functional loss. First, this work identifies persistent oxidative stress and mitochondrial impairment in the regional tendon tissues postinjury. Therefore, a smart scaffold incorporating the enzyme mimicry nanoparticle-ceria nanozyme (CeNPs) into the nanofiber bundle scaffold (NBS@CeO) with porous, anisotropic, and enhanced mechanical properties is designed to innovatively explore a targeted energy-supporting repair strategy by rescuing mitochondrial function and remodeling the microenvironment favoring endogenous regeneration. The integrated CeNPs scavenge excessive reactive oxygen species (ROS), stabilize the mitochondria membrane potential (ΔΨm), and ATP synthesis of tendon-derived stem cells (TDSCs) under oxidative stress. In a rat Achilles tendon defect model, NBS@CeO reduces oxidative damage and accelerates structural regeneration of collagen fibers, manifesting as recovering mechanical properties and motor function. Furthermore, NBS@CeO mediates the rebalance of endogenous regenerative signaling and dysregulated immune microenvironment by alleviating senescence and apoptosis of TDSCs, downregulating the secretion of senescence-associated secretory phenotype (SASP), and inducing macrophage M2 polarization. This innovative strategy highlights the role of NBS@CeO in tendon repair and thus provides a potential therapeutic approach for promoting tendon regeneration.


Subject(s)
Achilles Tendon , Rats , Animals , Rats, Sprague-Dawley , Stem Cells , Regeneration , Mitochondria
6.
Carbohydr Polym ; 277: 118865, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34893270

ABSTRACT

Tendon injury is one of the most common musculoskeletal diseases in the world, severely challenging the public health care system. Electrospinning technique using polymer materials (i.e. polycaprolactone (PCL)) and hydrogels (i.e. sodium alginate (ALG)) contribute to the development and application of smart composite scaffolds in the tendon tissue engineering by advantageously integrating mechanical properties and biocompatibility. As a potential natural antioxidant, melatonin (MLT) represents the potential to promote tendon repair. Here, we develop an MLT-loaded PCL/ALG composite scaffold that effectively promotes tendon injury repair in vivo and in vitro via a controlled release of MLT, possibly mechanically relying on an antioxidant stress pathway. This biomimetic composite scaffold will be of great significance in the tendon tissue engineering.


Subject(s)
Achilles Tendon/drug effects , Alginates/pharmacology , Biomimetic Materials/pharmacology , Hydrogels/pharmacology , Melatonin/pharmacology , Polyesters/pharmacology , Achilles Tendon/injuries , Achilles Tendon/pathology , Alginates/chemistry , Animals , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , Cells, Cultured , Hydrogels/chemistry , Male , Melatonin/chemistry , Polyesters/chemistry , Rats , Rats, Sprague-Dawley , Tissue Engineering , Tissue Scaffolds/chemistry
7.
J Nanobiotechnology ; 19(1): 169, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-34090456

ABSTRACT

BACKGROUND: Exosomes are extracellular vesicles of nano-structures and represent an emerging nano-scale acellular therapy in recent years. Tendon regeneration is a sophisticated process in the field of microsurgery due to its poor natural healing ability. To date, no successful long-term solution has been provided for the healing of tendon injuries. Functional recovery requires advanced treatment strategies. Human umbilical cord mesenchymal stem cell-derived exosomes (HUMSC-Exos) are considered as promising cell-free therapeutic agents. However, few studies reported their potential in the tendon repair previously. In this study, we explored the roles and underlying mechanisms of HUMSC-Exos in the tendon regeneration. RESULTS: Expression of tendon-specific markers in, and collagen deposition by, tendon-derived stem cells (TDSCs) treated with HUMSC-Exos increased in vitro. In a rat Achilles tendon injury model, treatment with HUMSC-Exos improved the histological structure, enhanced tendon-specific matrix components, and optimized biomechanical properties of the Achilles tendon. Findings in miRNA sequencing indicated a significant increase in miR-29a-3p in HUMSC-Exo-treated Achilles tendons. Next, luciferase assay in combination with western blot identified phosphatase and tensin homolog (PTEN) as the specific target of miR-29a-3p. Furthermore, we applied a miR-29a-3p-specific agonist to engineer HUMSC-Exos. These HUMSC-Exos overexpressing miR-29a-3p amplified the gain effects of HUMSC-Exos on tendon healing in vivo. To explore the underlying mechanisms, a transforming growth factor-ß1 (TGF-ß1) inhibitor (SB-431542), mTOR inhibitor (rapamycin), and engineered HUMSC-Exos were employed. The results showed that TGF-ß1 and mTOR signaling were involved in the beneficial effects of HUMSC-Exos on tendon regeneration. CONCLUSION: The findings in our study suggest that PTEN/mTOR/TGF-ß1 signaling cascades may be a potential pathway for HUMSC-Exos to deliver miR-29a-3p for tendon healing and implicate a novel therapeutic strategy for tendon regeneration via engineered stem cell-derived exosomes.


Subject(s)
Exosomes/metabolism , MicroRNAs/metabolism , Signal Transduction , Stem Cells , TOR Serine-Threonine Kinases/metabolism , Tendons/metabolism , Umbilical Cord/metabolism , Animals , Humans , Male , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Rats , Regeneration , Tendons/pathology , Umbilical Cord/cytology
8.
Front Immunol ; 12: 649285, 2021.
Article in English | MEDLINE | ID: mdl-34093537

ABSTRACT

Heterotopic ossification (HO) is one of the most intractable disorders following musculoskeletal injury and is characterized by the ectopic presence of bone tissue in the soft tissue leading to severe loss of function in the extremities. Recent studies have indicated that immune cell infiltration and inflammation are involved in aberrant bone formation. In this study, we found increased monocyte/macrophage and mast cell accumulation during early HO progression. Macrophage depletion by clodronate liposomes and mast cell stabilization by cromolyn sodium significantly impeded HO formation. Therefore, we proposed that the dietary phytochemical quercetin could also suppress immune cell recruitment and related inflammatory responses to prevent HO. As expected, quercetin inhibited the monocyte-to-macrophage transition, macrophage polarization, and mast cell activation in vitro in a dose-dependent manner. Using a murine burn/tenotomy model, we also demonstrated that quercetin attenuated inflammatory responses and HO in vivo. Furthermore, elevated SIRT1 and decreased acetylated NFκB p65 expression were responsible for the mechanism of quercetin, and the beneficial effects of quercetin were reversed by the SIRT1 antagonist EX527 and mimicked by the SIRT agonist SRT1720. The findings in this study suggest that targeting monocyte/macrophage and mast cell activities may represent an attractive approach for therapeutic intervention of HO and that quercetin may serve as a promising therapeutic candidate for the treatment of trauma-induced HO by modulating SIRT1/NFκB signaling.


Subject(s)
Burns/complications , Ossification, Heterotopic/drug therapy , Quercetin/administration & dosage , Tendon Injuries/complications , Animals , Burns/immunology , Carbazoles/administration & dosage , Cells, Cultured , Disease Models, Animal , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Humans , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Mice , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Ossification, Heterotopic/diagnosis , Ossification, Heterotopic/immunology , Ossification, Heterotopic/pathology , Primary Cell Culture , Signal Transduction/drug effects , Signal Transduction/immunology , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , THP-1 Cells , Tendon Injuries/immunology , Tendons/pathology , Tenotomy/adverse effects , Transcription Factor RelA/metabolism , X-Ray Microtomography
9.
Neural Plast ; 2021: 6684176, 2021.
Article in English | MEDLINE | ID: mdl-33679970

ABSTRACT

To date, failed back surgery syndrome (FBSS) remains a therapy-refractory clinical condition after spinal surgery. The antiadhesion membrane is applied to prevent FBSS by isolating fibrosis; however, the inflammation stimulated by the foreign body and surgical trauma needs to be further resolved simultaneously. Therefore, we developed new electrospun polycaprolactone (PCL) fibrous membranes loaded with celecoxib (CEL) to prevent fibrosis and inflammation associated with FBSS. The CEL-loaded PCL fibers were randomly distributed, and the drug was released over two weeks. Fluorescence micrographs revealed that the fibroblasts proliferated less on the PCL-CEL fibrous membranes than in the PCL group and the blank control. In the rat laminectomy model after 4 weeks, magnetic resonance imaging of epidural fibrosis was least in the PCL-CEL group. Expression of COX-2 and PGE2 was lower in the PCL-CEL group. It concluded that the CEL-loaded PCL membrane could reduce fibrosis and inflammation in a rat model of FBSS via COX-2/PGE2 signaling pathways.


Subject(s)
Celecoxib/pharmacology , Failed Back Surgery Syndrome/drug therapy , Inflammation/metabolism , Polyesters/pharmacology , Animals , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Epidural Space/pathology , Failed Back Surgery Syndrome/pathology , Inflammation/drug therapy , Male , Rats, Sprague-Dawley
10.
Mater Sci Eng C Mater Biol Appl ; 119: 111446, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33321586

ABSTRACT

Skin wounds are associated with huge economic and emotional burdens for millions of people annually and are a challenge for health workers worldwide. At present, for skin defects after traumatic accidents, especially large-area skin defects, newly developed strategies such as the use of emerging biomaterials and cell therapy could be considered as options besides classic skin grafts. However, the new strategies have to deal with problems such as immune rejection and high costs for patients. An insufficient understanding of the mechanisms of skin wound healing further hinders the development of innovative treatment approaches. In this study, we developed a parathyroid hormone (PTH)-loaded phase-transition microneedle (PTMN) patch to deliver PTH subcutaneously in an efficient manner and change microneedle patch daily to achieve intermittent and systematic drug administration. By evaluating wound closure, re-epithelialization, collagen deposition, and extracellular matrix (ECM) expression in a Sprague-Dawley rat model of traumatic skin wounds, we demonstrated that intermittent systemic administration of PTH using our PTMN patches accelerated skin wound healing. Further, we demonstrated that the use of the patch may accelerate skin wound healing depending on the activation of the transforming growth factor (TGF)-ß/Smad3/mammalian target of rapamycin (mTOR) cascade pathway. Our results suggest that the PTH-loaded PTMN patch may be a novel therapeutic strategy for treating skin wounds.


Subject(s)
Collagen , Parathyroid Hormone , Animals , Collagen/metabolism , Humans , Rats , Rats, Sprague-Dawley , Skin/metabolism , Smad3 Protein , TOR Serine-Threonine Kinases , Wound Healing
11.
Stem Cell Res Ther ; 11(1): 500, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33239069

ABSTRACT

BACKGROUND: Peritendinous fibrosis represents a fibrotic healing process that usually occurs after tendon injury or surgery. This worldwide challenge hampers the functional rehabilitation and the mobility of extremities. However, effective treatment is still lacking at present. The aim of our study was to explore the effect of extracellular vesicles derived from hydroxycamptothecin primed human umbilical cord stem cells (HCPT-EVs) on post-traumatic tendon adhesion. METHODS: Extracellular vesicles derived from unprimed human umbilical cord mesenchymal stem cells (Unprimed EVs) or HCPT-EVs were isolated and characterized. A rat model of Achilles tendon injury was used to confirm the anti-adhesion effect of HCPT-EVs and compared with that of Unprimed EVs in vivo. In vitro, the inhibitory effects of HCPT-EVs on fibroblast proliferation, viability, and myofibroblast differentiation upon TGF-ß1 stimulation were compared with the effects of Unprimed EVs. For mechanistic analysis, the expression of endoplasmic reticulum stress (ERS)-associated proteins was examined among the effector cargos of HCPT-EVs and Unprimed EVs. The ERS antagonist salubrinal was used to determine the ERS dependence of the anti-adhesion effects of HCPT-EVs. RESULTS: There were no obvious differences between Unprimed EVs and HCPT-EVs in terms of morphology, particle size, characteristic protein expression, and cellular uptake. HCPT-EVs exhibited a fortified anti-adhesion effect after Achilles tendon injury compared with Unprimed EVs. Fibroblast proliferation and viability and myofibroblast differentiation were all inhibited by HCPT-EVs. These properties were superior for HCPT-EVs relative to Unprimed EVs. Mechanistically, HCPT-EVs contained more ERS-associated protein than Unprimed EVs and activated the ERS pathway in fibroblast to counteract myofibroblast differentiation. CONCLUSION: This study demonstrates that HCPT-EVs show high anti-adhesion potential for the treatment of tendon injury by provoking ERS in fibroblasts. HCPT-EVs represent a promising strategy for clinical use in treating adhesion-related diseases.


Subject(s)
Camptothecin/analogs & derivatives , Extracellular Vesicles , Stem Cells , Tendon Injuries , Animals , Rats , Tendon Injuries/therapy , Umbilical Cord
12.
Ther Adv Chronic Dis ; 11: 2040622320944779, 2020.
Article in English | MEDLINE | ID: mdl-32821363

ABSTRACT

INTRODUCTION: Synthetic fibrous membranes unveil a promising field in anti-adhesion of tendons. Meanwhile, oriented nanofiber structures have been widely studied and used in the application of biomedical engineering, particularly in repairing and strengthening effects. METHODS: In this study, a bi-layer poly(L-lactic acid) (PLLA) electrospun membrane was fabricated, in which the inner oriented fibrous layer was designed to promote tendon healing while outer random aligned layer was designed to prevent peritendinous adhesion. RESULTS: It was found that fibroblasts were aligned along the oriented fiber of membranes in vitro and in a Leghorn chicken model. In biomechanical tests of repaired tendons, no significant difference was found between oriented fibrous membrane and blank control in maximum tensile strength; both oriented fibrous membranes and random fibrous membranes showed lower work of flexion than blank control, which was consistent with gross assessment. CONCLUSION: It was practicable to promote tendon healing while preventing adhesion via bi-layer PLLA membranes with an inner-oriented-fiber fabricated structure.

13.
Mater Sci Eng C Mater Biol Appl ; 116: 111166, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32806293

ABSTRACT

Posttraumatic peritendinous adhesion leads to limb disability. Physical barrier was widely used and thus focus was paid to fabricate the hydrophobic surfaces of electrospun membrane for anti-adhesion. However, current methods are limited and complicated. In this study, beeswax (Wax)/poly-L-lactic acid (PLA) anti-adhesion membranes were fabricated by blending electrospinning of Wax and PLA. The water contact angle was tested to investigate the hydrophobicity of the surfaces. Incorporation of Wax into PLA did not destroy the micro-pores between Wax/PLA fibers. After 7-day culture, proliferation of fibroblasts on Wax/PLA anti-adhesion membranes were significantly less than that on culture dish and PLA membranes. In rat Achilles adhesion model, least histological peritendinous adhesion formation was detected on the repaired sites in the group treated with Wax/PLA membranes than PLA membranes. Consequently, blending electrospinning of Wax into PLA is an easy method to fabricate hydrophobic surface of electrospun membrane with advanced peritendinous anti-adhesion outcome.


Subject(s)
Membranes, Artificial , Polyesters , Tissue Adhesions , Animals , Hydrophobic and Hydrophilic Interactions , Rats , Waxes
14.
J Inflamm Res ; 13: 303-316, 2020.
Article in English | MEDLINE | ID: mdl-32753931

ABSTRACT

PURPOSE: As a common complication of tendon injury, tendon adhesion is an unresolved problem in clinical work. The aim of this study was to investigate whether human umbilical cord mesenchymal stem cell-derived exosomes (HUMSC-Exos), one of the most promising new-generation cell-free therapeutic agents, can improve tendon adhesion and explore potential-related mechanisms. METHODS: The rat Achilles tendon injury adhesion model was constructed in vivo, and the localization of HUMSC-Exos was used to evaluate the tendon adhesion. Rat fibroblast cell lines were treated with transforming growth factor ß1 (TGF-ß1) and/or HUMSC-Exos in vitro, and cell proliferation, apoptosis and gene expression were measured. MicroRNA (miRNA) sequencing and quantitative PCR (qPCR) analysis confirmed differential miRNAs. A specific miRNA antagonist (antagomir-21a-5p) was used to transform HUMSC-Exos and obtain modified exosomes to verify its efficacy and related mechanism of action. RESULTS: In this study, we found HUMSC-Exos reduced rat fibroblast proliferation and inhibited the expression of fibrosis genes: collagen III (COL III) and α-smooth muscle actin (α-SMA) in vitro. In the rat tendon adhesion model, topical application of HUMSC-Exos contributed to relief of tendon adhesion. Specifically, the fibrosis and inflammation-related genes were simultaneously inhibited by HUMSC-Exos. Further, miRNA sequencing of HUMSCs and HUMSC-Exos showed that miR-21a-3p was expressed at low abundance in HUMSC-Exos. The antagonist targeting miR-21a-3p was recruited for treatment of HUMSCs, and harvested HUMSC-Exos, which expressed low levels of miR-21a-3p, and expanded the inhibition of tendon adhesion in subsequent in vitro experiments. CONCLUSION: Our results indicate that HUMSC-Exos may manipulate p65 activity by delivering low-abundance miR-21a-3p, ultimately inhibiting tendon adhesion. The findings may be promising for dealing with tendon adhesion.

15.
Article in English | MEDLINE | ID: mdl-32478044

ABSTRACT

Anti-adhesion membranes are prospective scaffolds for preventing peritendinous adhesion after injury. However, currently available scaffolds have some limitations, such as low efficacy for anti-adhesion, low quality of tendon healing, and unknown drug interactions. Thus, in this study, we designed an innovative structure involving an integrated dual-layer poly(L-lactic acid) (PLLA) electrospun membrane for preventing peritendonous adhesion by promoting tendon gliding. We investigated the surface morphology and wettability of the fiber scaffold. The adhesion and proliferation of fibroblasts were low on the PLLA fibrous membrane. Compared with single-layer membranes, the dual-layer PLLA fiber scaffold reduced adhesion to the tissues. The gliding space persisted until recovery in chicken extensor flexor tendons in vivo. Thus, this innovative PLLA membrane scaffold could prevent adhesion and promote gliding to facilitate tendon healing.

16.
Cell Prolif ; 53(1): e12730, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31746040

ABSTRACT

OBJECTIVES: In peripheral neuropathy, the underlying mechanisms of nerve and muscle degeneration include chronic inflammation and oxidative stress in fibrotic tissues. (-)-Epigallocatechin gallate (EGCG) is a major, active component in green tea and may scavenge free radical oxygen and attenuate inflammation. Conservative treatments such as steroid injection only deal with early, asymptomatic, peripheral neuropathy. In contrast, neurolysis and nerve conduit implantation work effectively for treating advanced stages. MATERIALS AND METHODS: An EGCG-loaded polycaprolactone (PCL) porous scaffold was fabricated using an integrated moulding method. We evaluated proliferative, oxidative and inflammatory activity of rat Schwann cells (RSCs) and rat skeletal muscle cells (RSMCs) cultured on different scaffolds in vitro. In a rat radiation injury model, we assessed the morphological, electrophysiological and functional performance of regenerated sciatic nerves and gastrocnemius muscles, as well as oxidative stress and inflammation state. RESULTS: RSCs and RSMCs exhibited higher proliferative, anti-oxidant and anti-inflammatory states in an EGCG/PCL scaffold. In vivo studies showed improved nerve and muscle recovery in the EGCG/PCL group, with increased nerve myelination and muscle fibre proliferation and reduced macrophage infiltration, lipid peroxidation, inflammation and oxidative stress indicators. CONCLUSIONS: The EGCG-modified PCL porous nerve scaffold alleviates cellular oxidative stress and repairs peripheral nerve and muscle structure in rats. It attenuates oxidative stress and inflammation in vivo and may provide further insights into peripheral nerve repair in the future.


Subject(s)
Catechin/analogs & derivatives , Nerve Regeneration/drug effects , Neurogenesis/drug effects , Oxidative Stress , Peripheral Nervous System Diseases/drug therapy , Polyesters , Radiation Injuries, Experimental/drug therapy , Schwann Cells/metabolism , Sciatic Nerve/physiology , Tissue Scaffolds/chemistry , Animals , Catechin/chemistry , Catechin/pharmacology , Cell Line , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Polyesters/chemistry , Polyesters/pharmacology , Porosity , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Rats , Schwann Cells/pathology , Sciatic Nerve/injuries , Sciatic Nerve/pathology
17.
Front Pharmacol ; 10: 967, 2019.
Article in English | MEDLINE | ID: mdl-31551777

ABSTRACT

Traumatic peritendinous fibrosis is a worldwide clinical problem resulting in severe limb disability. Hydroxycamptothecin (HCPT) is an anti-neoplastic drug widely exploited in clinical practice. It has shown potential of anti-fibrosis in recent years. We previously demonstrated that HCPT inhibited the characterization of fibrosis in vitro. However, it is still unclear whether it ameliorates peritendinous adhesion in an in vivo animal tendon injury model. The underlying mechanism is also worth investigating. The present study aims to determine whether HCPT inhibits tendon adhesion and to explore the underlying mechanisms. In a rat tendon injury model, we observed that topical application of HCPT significantly attenuated peritendinous adhesion as revealed by the results of macroscopic observation, biomechanical, histological, immunohistochemical evaluation, western blot, and quantitative PCR (q-PCR) analyses. Furthermore, western blot and q-PCR analyses revealed that this phenomenon is correlated with HCPT activation of endoplasmic reticulum (ER) stress. In addition, in vitro studies show that HCPT significantly inhibits fibroblast proliferation and induces apoptosis by reducing the expression of extracellular matrix (ECM) proteins COL3A1 and α-smooth muscle actin (α-SMA). Finally, we employed small interfering RNA (siRNA) to target inositol requiring kinase 1 (IRE1) and activated transcription factor 6 (ATF-6) to verify that the effect of inhibitory fibrosis of HCPT disappears after knockdown of ATF-6 and IRE1, thereby suggesting that an anti-fibrotic effect of HCPT is mediated by the ER-dependent apoptotic pathway. In conclusion, our results indicate that HCPT inhibits peritendinous fibrosis through the ER-dependent apoptotic pathway and might serve as a potential solution to prevent traumatic peritendinous adhesion.

18.
IUBMB Life ; 71(5): 653-662, 2019 05.
Article in English | MEDLINE | ID: mdl-30690843

ABSTRACT

Peritendinous fibrosis, which leads to impaired tendon function, is a clinical problem worldwide, and it is urgent to explore potential ways to reduce the formation of peritendinous adhesion. Several studies have demonstrated the biological roles of hydroxycamptothecin (HCPT) in inhibiting fibrosis in different tissues. In this study, we investigated whether HCPT could inhibit tendon fibrosis in vitro. Our results revealed that HCPT inhibited transforming growth factor (TGF)-ß1-induced cell viability of human fibroblasts, decreased excessive cell hyperproliferation and promoted fibroblasts apoptosis. In addition, HCPT treatment also inhibited expression of fibrosis genes COL3A1 and α-smooth muscle actin (α-SMA). In terms of mechanism, we pretreated fibroblasts with the endoplasmic reticulum stress (ER) inhibitor salubrinal and RNA-dependent protein kinase-like ER kinase (PERK) short hairpin RNA, these treatments abolished the inhibitory effects of HCPT on fibrosis, thereby suggesting that HCPT's inhibition of TGF-ß1-induced tendon fibrosis might be mediated by the PERK signaling pathway in vitro. In conclusion, our results suggested that HCPT had protective effects on peritendinous tissue fibrosis and might be promising in future clinical applications. © 2019 IUBMB Life, 71(5):653-662, 2019.


Subject(s)
Apoptosis/drug effects , Camptothecin/analogs & derivatives , Cell Proliferation/drug effects , Fibroblasts/drug effects , Fibrosis/prevention & control , Signal Transduction/drug effects , Tendons/drug effects , Camptothecin/pharmacology , Cell Adhesion , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Humans , In Vitro Techniques , Tendons/cytology , Tendons/metabolism , eIF-2 Kinase/metabolism
19.
J Mol Histol ; 48(2): 63-72, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27913976

ABSTRACT

Follistain-like protein 1 (FSTL1), has been recently demonstrated to be involved in the embryo development of nervous system and glioblastoma. However, the role of FSTL1 in neuroinflammation remains unexplored. In this study, the expression of FSTL1 in astrocytes was verified and its role was studied in neuroinflammation induced by in vivo intracerebroventricular (ICV) injection of lipopolysaccharide (LPS) or LPS treatment to astrocytes in vitro. FSTL1 was significantly induced after ICV LPS injection or LPS treatment. FSTL1 suppressed upregulation of pro-inflammatory cytokines in astrocytes after LPS treatment. Moreover, FSTL1 downregulated expression of pro-inflammatory cytokines through suppressing MAPK/p-ERK1/2 pathway in astrocytes. Our results suggest that FSTL1 may play an anti-inflammatory role in neuroinflammation mediated by astrocytes.


Subject(s)
Astrocytes/pathology , Cytokines/metabolism , Follistatin-Related Proteins/physiology , Inflammation/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Astrocytes/metabolism , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/pharmacology , Gene Expression Regulation , Humans , Inflammation/chemically induced , Lipopolysaccharides , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism
20.
Inflammation ; 39(6): 1997-2007, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27590238

ABSTRACT

Matrix metalloproteinase 13 (MMP-13) plays an important role in the process of pro-inflammatory cytokine-induced intervertebral disc degeneration (IDD). This study examined the effect of IL-17 on the regulation of MMP-13 and the extracellular matrix (ECM) in the intervertebral disc (IVD). We then examined whether salubrinal, a known inhibitor of eIF2α dephosphorylation, inhibited the IL-17-induced changes mentioned above. Furthermore, we demonstrated a potential therapeutic role for salubrinal in alleviating the chronic inflammatory-dependent degenerative state commonly observed in IDD. After inflammatory distress with IL-17, RT-PCR and western blot were employed to investigate the expression of MMP-13, collagen type II (COL2A1), collagen type I (COL1A1), and aggrecan (ACAN) in nucleus pulpous (NP) tissue. Activation of the NF-kB pathway was measured by western blot and immunocytochemistry following IL-17 treatment. We also examine the level of eIF2α phosphorylation after IL-17 treatment with or without salubrinal. Then, we investigated interactions of the NF-kB pathway to eIF2α phosphorylation. Moreover, we employed salubrinal and a specific inhibitor of NF-kB (BAY11-7082) to evaluate their effects on IL-17-driven regulation of MMP-13 and the ECM, as well as on the activation of NF-kB. The results showed that IL-17 increased the production of MMP-13 and decreased expression of COL2A1 and ACAN via the NF-kB pathway. Either IL-17 or salubrinal increased the level of eIF2α phosphorylation, but the effects of BAY11-7082 on the level of p-eIF2α were not detectable. BAY11-7082 and salubrinal significantly suppressed IL-17-driven intervertebral disc degeneration. Furthermore, salubrinal produced stronger effects than BAY11-7082. These results imply the potential involvement of IL-17 in IDD through activation of NF-kB signaling, which successively upregulated the expression of MMP-13 and led to the degradation of the ECM. Furthermore, salubrinal can inhibit this process through inhibition of NF-kB activation that is not directly linked to eIF2α phosphorylation, suggesting a potential therapeutic role in IDD.


Subject(s)
Cinnamates/pharmacology , Extracellular Matrix/metabolism , Interleukin-17/pharmacology , Matrix Metalloproteinase 13/metabolism , NF-kappa B/metabolism , Nucleus Pulposus/cytology , Signal Transduction/drug effects , Thiourea/analogs & derivatives , Up-Regulation/drug effects , Aggrecans/analysis , Cinnamates/therapeutic use , Collagen Type I/analysis , Collagen Type II/analysis , Eukaryotic Initiation Factor-2/metabolism , Humans , Intervertebral Disc Degeneration/drug therapy , Matrix Metalloproteinase 13/drug effects , Nitriles/pharmacology , Nucleus Pulposus/drug effects , Nucleus Pulposus/metabolism , Sulfones/pharmacology , Thiourea/pharmacology , Thiourea/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...