Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Adv Mater ; : e2314294, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38572797

ABSTRACT

Current synthetic grafts for ligament rupture repair often fail to integrate well with the surrounding biological tissue, leading to complications such as graft wear, fatigue, and subsequent re-rupture. To address this medical challenge, this study aims at advancing the development of a biological ligament through the integration of physiologically-inspired principles and tissue engineering strategies. In this study, interfacial polyelectrolyte complexation (IPC) spinning technique, along with a custom-designed collection system, to fabricate a hierarchical scaffold mimicking native ligament structure, is utilized. To emulate the bone-ligament interface and alleviate stress concentration, a hydroxyapatite (HAp) mineral gradient is strategically introduced near both ends of the scaffold to enhance interface integration and diminish the risk of avulsion rupture. Biomimetic viscoelasticity is successfully displayed to provide similar mechanical support to native ligamentous tissue under physiological conditions. By introducing the connective tissue growth factor (CTGF) and conducting mesenchymal stem cells transplantation, the regenerative potential of the synthetic ligament is significantly amplified. This pioneering study offers a multifaceted solution combining biomimetic materials, regenerative therapies, and advanced techniques to potentially transform ligament rupture treatment.

2.
Am J Sports Med ; 52(2): 406-422, 2024 02.
Article in English | MEDLINE | ID: mdl-38193194

ABSTRACT

BACKGROUND: Tendons have limited regenerative potential, so healing of ruptured tendon tissue requires a prolonged period, and the prognosis is suboptimal. Although stem cell transplantation-based approaches show promise for accelerating tendon repair, the resultant therapeutic efficacy remains unsatisfactory. HYPOTHESIS: The transplantation of stem cells preassembled as 3-dimensional spheroids achieves a superior therapeutic outcome compared with the transplantation of single-cell suspensions. STUDY DESIGN: Controlled laboratory study. METHODS: Adipose-derived stem cells (ADSCs) were assembled as spheroids using a methylcellulose hydrogel system. The secretome of ADSC suspensions or spheroids was collected and utilized to treat tenocytes and macrophages to evaluate their therapeutic potential and investigate the mechanisms underlying their effects. RNA sequencing was performed to investigate the global difference in gene expression between ADSC suspensions and spheroids in an in vitro inflammatory microenvironment. For the in vivo experiment, rabbits that underwent Achilles tendon transection, followed by stump suturing, were randomly assigned to 1 of 3 groups: intratendinous injection of saline, rabbit ADSCs as conventional single-cell suspensions, or preassembled ADSC spheroids. The tendons were harvested for biomechanical testing and histological analysis at 4 weeks postoperatively. RESULTS: Our in vitro results demonstrated that the secretome of ADSCs assembled as spheroids exhibited enhanced modulatory activity in (1) tenocyte proliferation (P = .015) and migration (P = .001) by activating extracellular signal-regulated kinase (ERK) signaling and (2) the suppression of the secretion of interleukin-6 (P = .005) and interleukin-1α (P = .042) by M1 macrophages via the COX-2/PGE2/EP4 signaling axis. Gene expression profiling of cells exposed to an inflammatory milieu revealed significantly enriched terms that were associated with the immune response, cytokines, and tissue remodeling in preassembled ADSC spheroids. Ex vivo fluorescence imaging revealed that the engraftment efficiency of ADSCs in the form of spheroids was higher than that of ADSCs in single-cell suspensions (P = .003). Furthermore, the transplantation of ADSC spheroids showed superior therapeutic effects in promoting the healing of sutured stumps, as evidenced by improvements in the tensile strength (P = .019) and fiber alignment (P < .001) of the repaired tendons. CONCLUSION: The assembly of ADSCs as spheroids significantly advanced their potential to harness tenocytes and macrophages. As a proof of concept, this study clearly demonstrates the effectiveness of using ADSC spheroids to promote tendon regeneration. CLINICAL RELEVANCE: The present study lays a foundation for future clinical applications of stem cell spheroid-based therapy for the management of tendon injuries.


Subject(s)
Achilles Tendon , Tendon Injuries , Animals , Rabbits , Achilles Tendon/pathology , Tenocytes , Adipose Tissue/pathology , Tendon Injuries/surgery , Macrophages/pathology , Stem Cells/physiology , Cell Proliferation
3.
Biomedicines ; 11(7)2023 Jun 24.
Article in English | MEDLINE | ID: mdl-37509451

ABSTRACT

Osteoarthritis (OA) is a major public and animal health challenge with significant economic consequences. Cartilage degradation plays a critical role in the initiation and progression of degenerative joint diseases, such as OA. Mesenchymal stem cells (MSCs) have become increasingly popular in the field of cartilage regeneration due to their promising results. The objective of this preclinical study was to evaluate the regenerative effects of mesenchymal stem cells (MSCs) in the repair of knee cartilage defects using a porcine model. Seven healthy LYD breed white pigs, aged 9-10 weeks and weighing approximately 20 ± 3 kg, were used in the experimental protocol. Full-thickness defects measuring 8 mm in diameter and 5 mm in depth were induced in the lateral femoral condyle of the posterior limbs in both knee joints using a sterile puncture technique while the knee was maximally flexed. Following a 1-week induction phase, the pig treatment groups received a 0.3 million/kg MSC transplant into the damaged knee region, while the placebo group received a control solution as a treatment. Magnetic resonance imaging (MRI), computerized tomography (CT), visual macroscopic examination, histological analysis, and cytokine concentration analysis were used to assess cartilage regeneration. The findings revealed that human adipose-derived mesenchymal stem cells (hADSCs) were more effective in repairing cartilage than pig umbilical cord-derived mesenchymal stem cells (pUCMSCs). These results suggest that MSC-based treatments hold promise as a treatment option for cartilage repair, which aid in the treatment of OA. However, further studies with larger sample sizes and longer follow-up periods are required to fully demonstrate the safety and efficacy of these therapies in both animals and humans.

4.
Int J Biol Macromol ; 241: 124636, 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37119896

ABSTRACT

Peripheral nerve injuries are commonly encountered in extremity traumas. Their motor and sensory recovery following microsurgical repair is limited by slow regeneration speed (<1 mm/d) and subsequent muscle atrophy, which are consequently correlated with the activity of local Schwann cells and efficacy of axon outgrowth. To promote post-surgical nerve regeneration, we synthesized a nerve wrap consisting of an aligned polycaprolactone (PCL) fiber shell with a Bletilla striata polysaccharide (BSP) core (APB). Cell experiments demonstrated that the APB nerve wrap markedly promoted neurite outgrowth and Schwann cell migration and proliferation. Animal experiments applying a rat sciatic nerve repair model indicated that the APB nerve wrap restored conduction efficacy of the repaired nerve and the compound action potential as well as contraction force of the related leg muscles. Histology of the downstream nerves disclosed significantly higher fascicle diameter and myelin thickness with the APB nerve wrap compared to those without BSP. Thus, the BSP-loaded nerve wrap is potentially beneficial for the functional recovery after peripheral nerve repair and offers sustained targeted release of a natural polysaccharide with good bioactivity.


Subject(s)
Myelin Sheath , Peripheral Nerve Injuries , Rats , Animals , Schwann Cells , Sciatic Nerve , Peripheral Nerve Injuries/drug therapy , Polysaccharides/pharmacology , Nerve Regeneration/physiology
5.
Int J Biol Macromol ; 231: 123312, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36669628

ABSTRACT

An anti-adhesive barrier membrane incorporating hyaluronic acid (HA) can reduce fibroblasts attachment and impart lubrication effect for smooth tendon gliding during management of post-surgical tendon adhesion. On the other hand, as numerous growth factors are required during tendon recovery, growth factors released by platelets in platelet-rich plasma (PRP) can provide beneficial therapeutic effects to facilitate tendon recovery post tendon injury. Furthermore, PRP is reported to be associated with anti-inflammatory properties for suppressing postoperative adhesion. Toward this end, we fabricate core-shell nanofiber membranes (NFM) with HA/PRP-infused core and polycaprolactone shell in this study. Different NFM with 100 % (H-P), 75 % (HP31-P), 50 % (HP11-P) and 25 % (H31-P) HA in the core was fabricated through coaxial electrospinning and analyzed through microscopic, pore size, mechanical, as well as HA and growth factor release studies. In vitro study with fibroblasts indicates the NFM can act as a barrier to prevent cell penetration and reduce cell attachment/focal adhesion, in addition to promoting tenocyte migration in tendon healing. In vivo studies in a rabbit flexor tendon rupture model indicates the HP11-P NFM shows improved efficacy over H-P NFM and control in reducing tendon adhesion formation and inflammation, while promoting tendon healing, from functional assays and histological analysis.


Subject(s)
Nanofibers , Platelet-Rich Plasma , Animals , Rabbits , Hyaluronic Acid , Nanofibers/therapeutic use , Tendons , Wound Healing , Tissue Adhesions/prevention & control
6.
Asian J Surg ; 46(1): 180-186, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35305874

ABSTRACT

BACKGROUND: Ulnar tunnel syndrome (UTS) is relatively uncommon compared to the carpal tunnel or cubital tunnel syndromes. Few reports dedicated to the functional outcomes after surgical intervention of the UTS exist. Herein we compare the outcomes of patients with UTS of different etiologies. METHODS: Patients diagnosed with UTS between 2016 and 2020 were recruited. Ulnar tunnel release was performed in all patients, along with other necessary osteosynthesis or reconstructive procedures in the traumatic group. Patients were followed-up every six months post-operatively. Outcomes measured include: objective evaluations, subjective questionnaires, records of clinical signs, and grading of the British Medical Research Council scale for intrinsic muscle strength. RESULTS: 21 patients were recruited, and favorable results were noted in all of them after surgery. Traumatic UTS patients had a worse initial presentation than the non-traumatic cases, but had a greater improvement after surgery and yielded outcomes comparable with those of the patients without trauma. Patients with aberrant muscles in their wrists had better outcomes in some objective measurements than those without aberrant muscles. CONCLUSIONS: Ulnar tunnel release improves the outcome of patients regardless of the etiology, especially in patients with trauma-induced UTS. Thus, a proper diagnosis of the UTS should be alerted in all patients encountering paresthesia in the ulnar digits, ulnar-sided pain, weakness of grip strength, or intrinsic weakness to ensure good outcomes.


Subject(s)
Carpal Tunnel Syndrome , Cubital Tunnel Syndrome , Ulnar Nerve Compression Syndromes , Humans , Ulnar Nerve Compression Syndromes/etiology , Ulnar Nerve Compression Syndromes/surgery , Prospective Studies , Cubital Tunnel Syndrome/diagnosis , Cubital Tunnel Syndrome/etiology , Cubital Tunnel Syndrome/surgery , Carpal Tunnel Syndrome/surgery , Carpal Tunnel Syndrome/complications , Wrist
7.
APL Bioeng ; 6(4): 046103, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36345317

ABSTRACT

Peripheral nerve injuries are commonly occurring traumas of the extremities; functional recovery is hindered by slow nerve regeneration (<1 mm/day) following microsurgical repair and subsequent muscle atrophy. Functional recovery after peripheral nerve repair is highly dependent on local Schwann cell activity and axon regeneration speed. Herein, to promote nerve regeneration, paracrine signals of adipose-derived stem cells were applied in the form of extracellular vesicles (EVs) loaded in a thermosensitive hydrogel (PALDE) that could solidify rapidly and sustain high EV concentration around a repaired nerve during surgery. Cell experiments revealed that PALDE hydrogel markedly promotes Schwann-cell migration and proliferation and axon outgrowth. In a rat sciatic nerve repair model, the PALDE hydrogel increased repaired-nerve conduction efficacy; contraction force of leg muscles innervated by the repaired nerve also recovered. Electromicroscopic examination of downstream nerves indicated that fascicle diameter and myeline thickness in the PALDE group (1.91 ± 0.61 and 1.06 ± 0.40 µm, respectively) were significantly higher than those in PALD and control groups. Thus, this EV-loaded thermosensitive hydrogel is a potential cell-free therapeutic modality to improve peripheral-nerve regeneration, offering sustained and focused EV release around the nerve-injury site to overcome rapid clearance and maintain EV bioactivity in vivo.

9.
Int J Mol Sci ; 22(22)2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34830254

ABSTRACT

Healing of ruptured tendons remains a clinical challenge because of its slow progress and relatively weak mechanical force at an early stage. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have therapeutic potential for tissue regeneration. In this study, we isolated EVs from adipose-derived stem cells (ADSCs) and evaluated their ability to promote tendon regeneration. Our results indicated that ADSC-EVs significantly enhanced the proliferation and migration of tenocytes in vitro. To further study the roles of ADSC-EVs in tendon regeneration, ADSC-EVs were used in Achilles tendon repair in rabbits. The mechanical strength, histology, and protein expression in the injured tendon tissues significantly improved 4 weeks after ADSC-EV treatment. Decorin and biglycan were significantly upregulated in comparison to the untreated controls. In summary, ADSC-EVs stimulated the proliferation and migration of tenocytes and improved the mechanical strength of repaired tendons, suggesting that ADSC-EV treatment is a potential highly potent therapeutic strategy for tendon injuries.


Subject(s)
Achilles Tendon/injuries , Adipocytes/metabolism , Extracellular Vesicles/transplantation , Stem Cells/chemistry , Tendon Injuries/therapy , Wound Healing/physiology , Adipocytes/cytology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Biglycan/genetics , Biglycan/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Decorin/genetics , Decorin/metabolism , Extracellular Vesicles/chemistry , Female , Gene Expression , Rabbits , Stem Cells/cytology , Stem Cells/metabolism , Tendon Injuries/metabolism , Tendon Injuries/pathology , Tenocytes/cytology , Tenocytes/metabolism , Treatment Outcome
10.
Int J Mol Sci ; 19(5)2018 May 04.
Article in English | MEDLINE | ID: mdl-29734717

ABSTRACT

To improve intraperitoneal chemotherapy and to prevent postsurgical peritoneal adhesion, we aimed to develop a drug delivery strategy for controlled release of a chemotherapeutic drug from the intraperitoneally injected thermosensitive poly(N-isopropylacrylamide)-based hydrogel (HACPN), which is also endowed with peritoneal anti-adhesion properties. Anticancer drug doxorubicin (DOX) was loaded into the hydrogel (HACPN-DOX) to investigate the chemotherapeutic and adhesion barrier effects in vivo. A burst release followed by sustained release of DOX from HACPN-DOX was found due to gradual degradation of the hydrogel. Cell culture studies demonstrated the cytotoxicity of released DOX toward CT-26 mouse colon carcinoma cells in vitro. Using peritoneal carcinomatosis animal model in BALB/c mice with intraperitoneally injected CT-26 cells, animals treated with HACPN-DOX revealed the best antitumor efficacy judging from tumor weight and volume, survival rate, and bioluminescence signal intensity when compared with treatment with free DOX at the same drug dosage. HACPN (or HACPN-DOX) also significantly reduced the risk of postoperative peritoneal adhesion, which was generated by sidewall defect-cecum abrasion in tumor-bearing BALB/c mice, from gross and histology analyses. This study could create a paradigm to combine controlled drug release with barrier function in a single drug-loaded injectable hydrogel to enhance the intraperitoneal chemotherapeutic efficacy while simultaneously preventing postsurgical adhesion.


Subject(s)
Doxorubicin/administration & dosage , Drug Delivery Systems , Peritoneal Neoplasms/drug therapy , Peritoneum/drug effects , Acrylamides/administration & dosage , Acrylamides/chemistry , Animals , Carcinoma/complications , Carcinoma/surgery , Cell Line, Tumor , Colonic Neoplasms/complications , Colonic Neoplasms/surgery , Doxorubicin/chemistry , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Hydrogels/administration & dosage , Hydrogels/chemistry , Mice , Mice, Inbred BALB C , Peritoneal Neoplasms/pathology , Peritoneum/pathology , Peritoneum/surgery , Tissue Adhesions/drug therapy , Tissue Adhesions/pathology , Tissue Adhesions/prevention & control
11.
Acta Biomater ; 63: 85-95, 2017 11.
Article in English | MEDLINE | ID: mdl-28919215

ABSTRACT

In this study, we aimed to assess whether thermo-responsive in-situ forming hydrogels based on poly(N-isopropylacrylamide) (PNIPAM) could prevent post-operative peritendinous adhesion. The clinical advantages of the thermo-responsive hydrogels are acting as barrier material to block penetration of fibroblasts, providing mobility and flexibility during application and enabling injection through a small opening to fill spaces of any shape after surgery. The thermo-responsiveness of hydrogels was determined to ensure their clinic uses. By grafting hydrophilic biopolymers chitosan (CS) and hyaluronic acid (HA) to PNIPAM, the copolymer hydrogels show enhanced water retention and lubrication, while reduced volume shrinkage during phase transition. In cell culture experiments, the thermo-responsive hydrogel has good biocompatibility and reduces fibroblast penetration. In animal experiments, the effectiveness of preventing post-operative peritendinous adhesion was studied in a rabbit deep flexor tendon model. From gross examination, histology, bending angles of joints, tendon gliding excursion and pull-out force, HA-CS-PNIPAM (HACPN) was confirmed to be the best barrier material to prevent post-operative peritendinous adhesion compared to PNIPAM and CS-PNIPAM (CPN) hydrogels and a commercial barrier film Seprafilm®. There was no significant difference in the breaking strength of HACPN-treated tendons and spontaneously healed ones, indicating HACPN hydrogel application did not interfere with normal tendon healing. We conclude that HACPN hydrogel can provide the best functional outcomes to significantly prevent post-operative tendon adhesion in vivo. STATEMENT OF SIGNIFICANCE: We prepared thermo-responsive in-situ forming hydrogels based on poly(N-isopropylacrylamide) (PNIPAM) to prevent post-operative peritendinous adhesion. The injectable barrier hydrogel could have better anti-adhesive properties than current commercial products by acting as barrier material to block penetration of fibroblasts, providing mobility and flexibility during application and enabling injection through a small opening to fill spaces of any shape after surgery. The effectiveness of preventing post-operative peritendinous adhesion was studied in a rabbit deep flexor tendon model. From gross examination, histology, bending angles of joints, tendon gliding excursion and pull-out force, HA-CS-PNIPAM (HACPN) was confirmed to be the best barrier material to prevent post-operative peritendinous adhesion compared to PNIPAM and CS-PNIPAM (CPN) hydrogels and a commercial barrier film Seprafilm®.


Subject(s)
Hydrogels/pharmacology , Temperature , Tendons/pathology , Tissue Adhesions/prevention & control , Animals , Biomechanical Phenomena , Cell Death/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Mice , NIH 3T3 Cells , Phase Transition , Rabbits , Range of Motion, Articular/drug effects , Tendons/drug effects , Tendons/physiopathology , Tissue Adhesions/pathology , Tissue Adhesions/physiopathology , Viscosity , Water/chemistry
12.
Nanomaterials (Basel) ; 7(8)2017 Aug 11.
Article in English | MEDLINE | ID: mdl-28800110

ABSTRACT

Silk fibroin (SF) and fiber alignment were introduced into polycaprolactone (PCL)-based electrospun nanofibers as chemical and physical cues for tendon tissue engineering applications. The physicochemical properties of random PCL (RP) nanofibers, random PCL/SF (RPSF) nanofibers and aligned PCL/SF (APSF) nanofibers were characterized for fiber orientation and SF blending effects. An in vitro cell culture with rabbit dermal fibroblasts (RDFBs) on nanofibers indicated that SF promotes cell proliferation to a higher extent than fiber alignment. Cells aligned in the direction of fiber axes could be confirmed through scanning electron microscopy (SEM) observation and cytoskeleton staining. The quantitative real-time polymerase chain reaction (qRT-PCR) experiments indicated up-regulated gene expression of tendon marker proteins (type I collagen (Col I), fibronectin and biglycan) on APSF nanofibers and tendon reconstruction was confirmed from Col III gene expression. Animal experiments with Achilles tendon defect repairs in rabbits were carried out with RPSF and APSF scaffolds. The beneficial effects of fiber alignment were verified from histological and immunohistochemical staining, where cell migration and extracellular matrix protein deposition tend to stretch in a parallel direction along the axial direction of APSF nanofibers with enhanced Col I and tenascin C production. Biomechanical testing indicated the tensile stiffness and maximum load of cell-seeded APSF scaffolds were 60.2 and 81.3% of normal tendon values, respectively, which are significantly higher than cell-seeded RPSF or acellular APSF and RPSF scaffolds. These results suggest that APSF nanofiber scaffolds combined with RDFBs have the potential to repair the gap defects of Achilles tendons in vivo and to effectively restore the function and structure of tendons.

13.
Carbohydr Polym ; 173: 721-731, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28732919

ABSTRACT

Peritoneal adhesion is one of the common complications after abdominal surgery. Injectable thermosensitive hydrogel could serve as an ideal barrier to prevent this postoperative tissue adhesion. In this study, poly(N-isopropylacrylamide) (PNIPAm) was grafted to chitosan (CS) and the polymer was further conjugated with hyaluronic acid (HA) to form thermosensitive HA-CS-PNIPAm hydrogel. Aqueous solutions of PNIPAm and HA-CS-PNIPAm at 10%(w/v) are both free-flowing and injectable at room temperature and exhibit sol-gel phase transition around 31°C; however, HA-CS-PNIPAm shows less volume shrinkage after gelation and higher complex modulus than PNIPAm. Cell culture studies indicate both injectable hydrogel show barrier effects to reduce fibroblasts penetration while induce little cytotoxicity in vitro. From a sidewall defect-bowel abrasion model in rats, significant reduction of postoperative peritoneal adhesion was found for peritoneal defects treated with HA-CS-PNIPAm compared with those treated with PNIPAm and untreated controls from gross and histological evaluation. Furthermore, HA-CS-PNIPAm did not interfere with normal peritoneal tissue healing and did not elicit acute toxicity from blood analysis and tissue biopsy examination. By taking advantage of the easy handling and placement properties of HA-CS-PNIPAm during application, this copolymer hydrogel would be a potentially ideal injectable anti-adhesion barrier after abdominal surgeries.


Subject(s)
Chitosan/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Tissue Adhesions/drug therapy , Animals , Male , Mice , NIH 3T3 Cells , Peritoneum/drug effects , Postoperative Complications/prevention & control , Rats , Rats, Sprague-Dawley , Temperature , Tissue Adhesions/prevention & control
14.
Acta Biomater ; 26: 225-35, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26234491

ABSTRACT

Peritendinous adhesions, one of the common complications after tendon injury and subsequent surgery, could be minimized by directly placing a physical barrier between the injured site and the surrounding tissue. We used silver (Ag) nanoparticles embedded in electrospun hyaluronic acid (HA)/polycaprolactone (PCL) nanofibrous membranes (NFMs) (HA/PCL+Ag NFMs) to prevent peritendinous adhesions and bacterial infection after tendon surgery. HA was used for effective lubrication, and Ag provided antibacterial activity. A dual functional anti-adhesion barrier with core-sheath nanofibrous architecture was made from an HA core solution and a photo-reduced silver nitrate/PCL sheath solution. Polycaprolactone NFMs (PCL NFMs), hyaluronic acid/polycaprolactone core-sheath NFMs (HA/PCL NFMs) and HA/PCL+Ag NFMs with comparable fiber diameters and pore sizes were prepared and analyzed. The microporous structure of NFMs is expected to effectively block the penetration of adhesion-forming fibroblasts during tendon healing. The release of Ag from HA/PCL+Ag NFMs plateaued after 4 days, which confirmed the short-term anti-bacterial effect, and this result was verified with agar diffusion tests. In contrast, the release of HA was extended up to 21 days to simulate the lubrication effect offered by HA in the synovial fluid of the tendon sheath. In vitro cell culture experiments revealed that HA/PCL+Ag NFMs exhibited the highest inhibition of fibroblast attachment and proliferation without significant cytotoxicity due to the synergistic effect of Ag and HA. In vivo studies with a rabbit flexor tendon model further confirmed the efficacy of HA/PCL+Ag NFMs in reducing peritendinous adhesion as determined by gross observation, histology, joint range-of-motion, tendon gliding and biomechanical tests.


Subject(s)
Bacterial Infections/prevention & control , Bandages , Hyaluronic Acid/chemistry , Polyesters/chemistry , Tendinopathy/prevention & control , Tissue Adhesions/prevention & control , Animals , Anti-Bacterial Agents/administration & dosage , Bacterial Infections/pathology , Electroplating/methods , Equipment Design , Equipment Failure Analysis , Materials Testing , Membranes, Artificial , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Nanofibers/chemistry , Nanofibers/therapeutic use , Rabbits , Silver/administration & dosage , Silver/chemistry , Tendinopathy/pathology , Tissue Adhesions/pathology , Treatment Outcome
15.
Colloids Surf B Biointerfaces ; 133: 221-30, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26115533

ABSTRACT

Postoperative adhesion formation is the major complication that could occur after acute tendon surgery. The application of an anti-adhesive membrane at the post-surgical site is deemed as a potential way to solve this problem by preventing adhesive fibrotic tissue development. In this study, we fabricated electrospun composite poly(ethylene glycol) (PEG)/poly(caprolactone) (PCL) nanofibrous membrane (NFM) to prevent peritendinous adhesions, which could act as a barrier between the tendon and surrounding tissues, without interrupting mass transfer and normal tendon gliding. PCL/PEG NFMs of 0% PEG (PCL), 25% PEG (25PECL), 50% PEG (50PECL) and 75% PEG (75PECL) were prepared and characterized for physico-chemical properties. The PCL NFM shows the lowest protein permeability while 25PECL NFM exhibited the largest fiber diameter, smallest pore size and the largest ultimate stress and strain. The 75PECL NFM had the lowest water contact angle and the highest Young's modulus. In vitro cell adhesion and migration experiments with fibroblasts indicate that all NFMs could prevent cell penetration, with 75PECL NFM having the least cell attachment. In vivo application of 75PECL NFM on the repaired site of rabbit flexor tendon rupture model demonstrated improved efficacy compared with the PCL NFM and a commercial anti-adhesion barrier (Seprafilm™), from gross observation, histological analysis and functional assays. We concluded that 75PECL NFM could function as an effective anti-adhesion membrane after tendon surgery in a clinical setting.


Subject(s)
Membranes, Artificial , Nanofibers , Polyesters/pharmacology , Polyethylene Glycols/pharmacology , Tendons/pathology , Tissue Adhesions/prevention & control , Animals , Cells, Cultured , Humans , Polyesters/chemistry , Polyethylene Glycols/chemistry , Rabbits
16.
Acta Biomater ; 10(12): 4971-4982, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25192729

ABSTRACT

As one of the common complications after tendon injury and subsequent surgery, peritendinous adhesions could be minimized by directly placing a physical barrier between the injured site and the surrounding tissue. With the aim of solving the shortcomings of current biodegradable anti-adhesion barrier membranes, we propose the use of an electrospun chitosan-grafted polycaprolactone (PCL-g-CS) nanofibrous membrane (NFM) to prevent peritendinous adhesions. After introducing carboxyl groups on the surface by oxygen plasma treatment, the polycaprolactone (PCL) NFM was covalently grafted with chitosan (CS) molecules, with carbodiimide as the coupling agent. Compared with PCL NFM, PCL-g-CS NFM showed a similar fiber diameter, permeation coefficient for bovine serum albumin, ultimate tensile strain, reduced pore diameter, lower water contact angle, increased water sorption and tensile strength. With its submicrometer pore diameter (0.6-0.9µm), both NFMs could allow the diffusion of nutrients and waste while blocking fibroblast penetration to prevent adhesion formation after tendon surgery. Cell culture experiments verified that PCL-g-CS NFM can reduce fibroblast attachment while maintaining the biocompatibility of PCL NFM, implicating a synergistic anti-adhesion effect to raise the anti-adhesion efficacy. In vivo studies with a rabbit flexor digitorum profundus tendon surgery model confirmed that PCL-g-CS NFM effectively reduced peritendinous adhesion from gross observation, histology, joint flexion angle, gliding excursion and biomechanical evaluation. An injured tendon wrapped with PCL-g-CS NFM showed the same tensile strength as the naturally healed tendon, indicating that the anti-adhesion NFM will not compromise tendon healing.


Subject(s)
Chitosan/chemistry , Membranes, Artificial , Nanofibers/chemistry , Tendons/physiopathology , Tissue Adhesions/prevention & control , Tissue Adhesions/physiopathology , Tissue Scaffolds , Animals , Electroplating/methods , Equipment Design , Equipment Failure Analysis , Guided Tissue Regeneration/instrumentation , Materials Testing , Nanofibers/ultrastructure , Particle Size , Rabbits , Rotation , Tendinopathy/pathology , Tendinopathy/physiopathology , Tendinopathy/prevention & control , Tendons/pathology , Tendons/surgery , Tensile Strength , Tissue Adhesions/pathology , Treatment Outcome
17.
Int J Nanomedicine ; 9: 4079-92, 2014.
Article in English | MEDLINE | ID: mdl-25187711

ABSTRACT

Peritendinous adhesion is one of the common complications encountered after tendon injury and subsequent surgery, and it can be minimized by introducing a physical barrier between the injured site and the surrounding tissue. An electrospun hyaluronic acid-grafted poly(caprolactone) (PCL-g-HA) nanofibrous membrane (NFM) is proposed as an alternative to current antiadhesion barrier films. HA is covalently grafted to surface-aminolyzed PCL nanofibers, using carbodiimide as the coupling agent. Pristine PCL and PCL-g-HA NFMs were characterized by scanning electron microscopy, thermogravimetric analysis, X-ray photoelectron spectroscopy, Fourier-transform infrared spectroscopy, and mechanical testing. In vitro cell culture with fibroblasts showed that PCL-g-HA NFMs reduced cellular adhesion on the membrane surface while maintaining cell proliferation. Animal experiments using a rabbit flexor digitorum profundus tendon model confirmed the efficacy of PCL-g-HA in reducing peritendinous adhesion, based on gross observation, histology, joint flexion-angle measurements, gliding tests, and biomechanical evaluation.


Subject(s)
Hyaluronic Acid/pharmacology , Membranes, Artificial , Nanofibers/chemistry , Polyesters/pharmacology , Tissue Adhesions/prevention & control , Animals , Cell Adhesion/drug effects , Cell Line , Cell Survival/drug effects , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/therapeutic use , Male , Nanofibers/therapeutic use , Particle Size , Polyesters/chemistry , Polyesters/therapeutic use , Rabbits , Tendons/drug effects , Tendons/surgery , Tissue Adhesions/drug therapy
18.
Carbohydr Polym ; 111: 288-97, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25037354

ABSTRACT

Nanofibrous membrane scaffolds of chitosan (CS), silk fibroin (SF) and CS/SF blend were prepared by electrospinning and studied for growth and osteogenic differentiation of human bone marrow mesenchymal stem cells (hMSCs). The morphology and physico-chemical properties of all membrane scaffolds were compared. The influence of CS and SF on cell proliferation was assessed by the MTS assay, whereas osteogenic differentiation was determined from the Alizarin Red staining, alkaline phosphatase activity and expression of osteogenic marker genes. The osteogenic differentiation and proliferation of hMSCs were enhanced by CS and SF nanofibers, respectively. Blending CS with SF retained the osteogenesis nature of CS without negatively influencing the cell proliferative effect of SF. By taking advantage of the differentiation/proliferation cues from individual components, the electrospun CS/SF composite nanofibrous membrane scaffold is suitable for bone tissue engineering.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chitosan/pharmacology , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Silk/pharmacology , Alkaline Phosphatase/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Calcification, Physiologic , Chitosan/chemistry , Gene Expression , Humans , Mesenchymal Stem Cells/enzymology , Nanofibers/chemistry , Silk/chemistry
19.
Kaohsiung J Med Sci ; 29(5): 246-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23639510

ABSTRACT

We compared the enhancement effect between a newly synthesized tissue-specific contrast agent, [Gd-DOTA-FPßG], and a commercially available agent, [Gd(DOTA)](-), in a murine model of liver tumor using a clinical magnetic resonance imaging scanner. The colon cancer cell lines with and without ß-glucuronidase (ßG) expression were implanted into the liver of mice. Self-synthesized gadolinium-based magnetic resonance contrast agent, [Gd(DOTA-FPßG)], was administered to measure enhancement on magnetic resonance images using a commercially available agent, [Gd(DOTA)](-), as control in a clinical 3.0 tesla (T) magnetic resonance scanner. In vivo fluorescence imaging and histopathology of the liver were also performed to compare and correlate with the magnetic resonance studies. The in vivo fluorescence imaging failed to depict a sufficiently intense signal for liver or liver tumor of mice without exposure of the liver following an incision on the abdominal wall. The tissue-specific magnetic resonance agent, [Gd(DOTA-FPßG)], caused significantly stronger enhancement in tumors expressing ßG (CT26/mßG-eB7) than in tumors not expressing ßG (CT26) (p < 0.05). In the magnetic resonance imaging studies using control agent [Gd(DOTA)](-), the tumors with and without ßG expression depicted no significant difference in enhancement on the T1-weighted images. The [Gd(DOTA-FPßG)] also provided significantly more contrast uptake in the CT26/mßG-eB7 tumor than in the normal liver parenchyma, whereas the [Gd(DOTA)](-) did not. This study confirms that better contrast enhancement can be readily detected in vivo by the use of a tissue-specific magnetic resonance contrast agent to target tumor cells with specific biomarkers in a clinical magnetic resonance imaging scanner.


Subject(s)
Contrast Media , Coordination Complexes , Glucuronates , Heterocyclic Compounds , Liver Neoplasms, Experimental/pathology , Organometallic Compounds , Animals , Cell Line, Tumor , Contrast Media/pharmacokinetics , Coordination Complexes/pharmacokinetics , Glucuronates/pharmacokinetics , Heterocyclic Compounds/pharmacokinetics , Liver/metabolism , Liver/pathology , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Optical Imaging , Organometallic Compounds/pharmacokinetics
20.
Inorg Chem ; 51(22): 12426-35, 2012 Nov 19.
Article in English | MEDLINE | ID: mdl-23116118

ABSTRACT

ß-Glucuronidase is a key lysosomal enzyme and is often overexpressed in necrotic tumor masses. We report here the synthesis of a pro receptor-induced magnetization enhancement (pro-RIME) magnetic resonance imaging (MRI) contrast agent ([Gd(DOTA-FPßGu)]) for molecular imaging of ß-glucuronidase activity in tumor tissues. The contrast agent consists of two parts, a gadolinium complex and a ß-glucuronidase substrate (ß-d-glucopyranuronic acid). The binding association constant (KA) of [Gd(DOTA-FPßGu)] is 7.42 × 10(2), which is significantly lower than that of a commercially available MS-325 (KA = 3.0 × 10(4)) RIME contrast agent. The low KA value of [Gd(DOTA-FPßGu)] is due to the pendant ß-d-glucopyranuronic acid moiety. Therefore, [Gd(DOTA-FPßGu)] can be used for detection of ß-glucuronidase through RIME modulation. The detail mechanism of enzymatic activation of [Gd(DOTA-FPßGu)] was elucidated by LC-MS. The kinetics of ß-glucuronidase catalyzed hydrolysis of [Eu(DOTA-FPßGu)] at pH 7.4 best fit the Miechalis-Menten kinetic mode with Km = 1.38 mM, kcat = 3.76 × 10(3), and kcat/Km = 2.72 × 10(3) M(-1) s(-1). The low Km value indicates high affinity of ß-glucuronidase for [Gd(DOTA-FPßGu)] at physiological pH. Relaxometric studies revealed that T1 relaxivity of [Gd(DOTA-FPßGu)] changes in response to the concentration of ß-glucuronidase. Consistent with the relaxometric studies, [Gd(DOTA-FPßGu)] showed significant change in MR image signal in the presence of ß-glucuronidase and HSA. In vitro and in vivo MR images demonstrated appreciable differences in signal enhancement in the cell lines and tumor xenografts in accordance to their expression levels of ß-glucuronidase.


Subject(s)
Antineoplastic Agents/pharmacology , Contrast Media/pharmacology , Gadolinium , Glucuronidase/metabolism , Neoplasms, Experimental/drug therapy , Organometallic Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Contrast Media/chemical synthesis , Contrast Media/chemistry , Dose-Response Relationship, Drug , Enzyme Activation , Gadolinium/chemistry , Ligands , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Structure-Activity Relationship , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...