Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 269
Filter
1.
J Orthop Surg Res ; 18(1): 78, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36721157

ABSTRACT

BACKGROUND: Platelet-derived lyophilized growth factors (L-GFs) use a standardized number of allogenic pathogen-free platelets instead of autologous platelets used in PRP as a source of growth factors. This study aimed to evaluate the efficacy of L-GF injection versus placebo in subacromial impingement (SIS) treatment. METHODS: The current randomized double-blind placebo-controlled study included sixty patients (40 females and 20 males, aged between 24 and 75 years) diagnosed with SIS (both clinically and sonographically). Patients were randomly assigned to two equal groups. Under ultrasound guidance, group 1 received subacromial saline injection, and group 2 received L-GF injection. Clinical examination, pain visual analogue scale (VAS), shoulder pain and disability index (SPADI) and shoulder ultrasound were performed before and at the 8th week after injection. RESULTS: Follow-up assessment showed statistically significant improvement in the L-GF group regarding active flexion, active and passive internal rotation and extension, SPADI-disability scale, VAS and thickness of the supraspinatus tendon by US. Regression analysis showed that group 1 was approximately 30 times more likely than the L-GF group to experience painful arc at follow-up. Both groups showed statistically significant improvement in SPADI-pain scale and SPADI-total, flexion and abduction (still the mean value of abduction was significantly higher in the L-GF group). CONCLUSIONS: L-GF injection resulted in clinically significant reductions in pain and functional disability outcomes in patients with SIS. An objective significant reduction in the thickness of the supraspinatus tendon, measured by ultrasound, in the L-GF group hopefully encourages proper healing and functioning in SIS. TRIAL REGISTRATION: The identification number is NCT04330027, date of first registration (01/04/2020). Unique on 21/11/2019, Protocol ID: 0106178.


Subject(s)
Platelet-Derived Growth Factor , Shoulder Impingement Syndrome , Shoulder Pain , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Platelet-Derived Growth Factor/administration & dosage , Prospective Studies , Rotator Cuff , Shoulder Impingement Syndrome/diagnostic imaging , Shoulder Impingement Syndrome/drug therapy , Shoulder Pain/drug therapy , Injections, Intralesional
2.
Klin Monbl Augenheilkd ; 239(4): 378-381, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35472775

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF)-A, the most abundant subtype of the VEGF family in the eye, plays an important role in corneal homeostasis due to its ability to mediate corneal nerve repair. Repeated intravitreal anti-VEGF injections were shown to significantly reduce corneal nerve density, which might negatively affect corneal homeostasis and lead to a neuropathic dry eye disease. Currently, there are two effective modalities to treat dry eye while supplying VEGF to the ocular surface: serum eye drops (SED) and eye drops manufactured from plasma rich in growth factors (PRGF). The purpose of this study was to measure the VEGF-A concentration in SED and PRGF eye drops. MATERIAL AND METHODS: Ten healthy volunteers donated blood on two separate occasions, 2 - 8 days apart. Thus, a total of 20 blood samples were processed to obtain both SED and PRGF. Concentrations of VEGF-A were quantified by a Simple Plex platform run in triplicate. RESULTS: The VEGF-A concentration in SED and PRGF was very similar between the two blood samples drawn from one individual donor but showed substantial interindividual variability. However, in all 20 samples, VEGF concentrations were substantially higher in SED samples (mean 238.7 ± 146.6 pg/mL) compared to PRGF samples (mean 67.4 ± 46.3 pg/mL). Based upon the analysis of variance (ANOVA) model for the measured concentrations with fixed effects for specimen (SED vs. PRGF) and subject, the mean difference between the SED and PRGF concentration was 168.1 pg/mL (95% confidence interval: [142.4, 193.9], p < 0.001). CONCLUSION: Our study showed that the VEGF concentration was higher in SED than in PRGF. This is an important finding, particularly for potential treatment of dry eye disease in patients with neuropathic eye disease, especially in patients that received repeated anti-VEGF intravitreal injections, or in patients with Sjögren's disease, where the level of VEGF in tears might be pathologically decreased. Hypothetically, VEGF might be needed to restore ocular surface homeostasis. Although growing evidence has shown that VEGF-A plays an important role in corneal homeostasis, only a randomized prospective clinical trial will show whether supplying VEGF-A to the ocular surface might successfully restore the corneal homeostasis and overcome the problem of corneal neuropathy in these patients. For such a trial, based on our results, an undiluted SED should be preferred over a PRGF due to the higher content of VEGF-A.


Subject(s)
Dry Eye Syndromes , Platelet-Derived Growth Factor , Platelet-Rich Plasma , Vascular Endothelial Growth Factor A , Dry Eye Syndromes/drug therapy , Humans , Intercellular Signaling Peptides and Proteins/administration & dosage , Ophthalmic Solutions , Platelet-Derived Growth Factor/administration & dosage , Prospective Studies , Vascular Endothelial Growth Factor A/administration & dosage
3.
Int Orthop ; 46(3): 581-588, 2022 03.
Article in English | MEDLINE | ID: mdl-35020026

ABSTRACT

PURPOSE: The purpose of this study aimed to evaluate the efficacy of local injection of allogeneic platelet-derived growth factors in treatment of patients with tennis elbow. PATIENTS AND METHODS: This study included 120 tennis elbow patients randomly divided into two groups. The patients were locally injected with allogeneic growth factors (treatment group) or with normal saline (control group). The outcomes were assessed using Patient-Related Tennis Elbow Evaluation (PRTEE) and quick Disabilities of the Arm, Shoulder and Hand (qDASH) scales. The clinical outcomes were accordingly classified as excellent, good and poor. The patient's satisfaction and adverse effects were also recorded. RESULTS: There was no statistically significant difference between the two groups regarding the age, gender, dominant arm or the pre-injection scores. At three month follow-up, the reductions in the mean PRTEE and qDASH scores were 88.7% and 70.6% in the treatment group versus 21.8% and 14.9% in the control group, respectively. At the last follow-up, the outcomes in the treatment group were excellent in 85% of patients and good in 15%, versus 8% and 32% in the control group. Overall, 95% were satisfied in the treatment group compared to 25% in control group. Forty patients in the treatment group experienced mild transient post-injection pain. CONCLUSION: This study strongly suggests that local injection of allogeneic platelet-derived growth factors could be a promising safe treatment option for tennis elbow with significant pain relief, functional improvement and patient's satisfaction. Yet, additional larger studies are needed to assess the durability of these outcomes.


Subject(s)
Platelet-Derived Growth Factor , Tennis Elbow , Humans , Injections , Platelet-Derived Growth Factor/administration & dosage , Prospective Studies , Tennis Elbow/therapy , Treatment Outcome
4.
J Mater Chem B ; 9(31): 6176-6189, 2021 08 21.
Article in English | MEDLINE | ID: mdl-34297017

ABSTRACT

Wound healing is a well-orchestrated dynamic and interactive process, which needs a favorable microenvironment and suitable angiogenesis. Platelet derived growth factor-BB (PDGF-BB) plays a crucial role in wound healing. However, the short half-life of PDGF-BB limits its efficacy. In the present study, we successfully synthesized an injectable hydrogel with sodium alginate (SA) and dextran (Dex) as a delivery system to simultaneously deliver PDGF-BB and bone marrow-derived mesenchymal stem cells (BMSCs) in the wound. Our work demonstrates that the PDGF-BB protein enhanced the survival, migration and endothelial cell (EC) differentiation of BMSCs in vitro. The PDGF-BB/SA/Dex hydrogels could sustainably release PDGF-BB with excellent biocompatibility in vitro and in vivo. Besides, these composite hydrogels loaded with BMSCs could accelerate wound healing by improving epithelialization and collagen deposition. In addition, the PDGF-BB/SA/Dex hydrogels promoted the EC-differentiation of transplanted BMSCs and proliferation of hair follicle stem cells in the wound. Furthermore, the expressions of angiogenesis-specific markers, PDGFR-ß, p-PI3K, p-Akt, and p-eNOS, were obviously increased in the PDGF-BB/SA/Dex/BMSCs group. In conclusion, the PDGF-BB/SA/Dex injectable hydrogels could accelerate BMSC-mediated skin wound healing by promoting angiogenesis via the activation of the PDGF-BB/PDGFR-ß-mediated PI3K/Akt/eNOS pathway, which may provide a new therapeutic strategy for stem cell therapy in wound healing.


Subject(s)
Alginates/pharmacology , Biocompatible Materials/pharmacology , Dextrans/pharmacology , Hydrogels/pharmacology , Mesenchymal Stem Cells/drug effects , Platelet-Derived Growth Factor/pharmacology , Alginates/administration & dosage , Alginates/chemistry , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Dextrans/administration & dosage , Dextrans/chemistry , Humans , Hydrogels/administration & dosage , Hydrogels/chemistry , Materials Testing , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic/drug effects , Platelet-Derived Growth Factor/administration & dosage , Platelet-Derived Growth Factor/chemistry , Skin/drug effects , Skin/metabolism , Wound Healing/drug effects
5.
Int J Mol Sci ; 22(11)2021 May 28.
Article in English | MEDLINE | ID: mdl-34071329

ABSTRACT

Avascular transplantation of frozen-thawed testicular tissue fragments represents a potential future technique for fertility restoration in boys with cancer. A significant loss of spermatogonia was observed in xeno-transplants of human tissue most likely due to the hypoxic period before revascularization. To reduce the effect of hypoxia-reoxygenation injuries, several options have already been explored, like encapsulation in alginate hydrogel and supplementation with nanoparticles delivering a necrosis inhibitor (NECINH) or VEGF. While these approaches improved short-term (5 days) vascular surfaces in grafts, neovessels were not maintained up to 21 days; i.e., the time needed for achieving vessel stabilization. To better support tissue grafts, nanoparticles loaded with VEGF, PDGF and NECINH were developed. Testicular tissue fragments from 4-5-week-old mice were encapsulated in calcium-alginate hydrogels, either non-supplemented (control) or supplemented with drug-loaded nanoparticles (VEGF-nanoparticles; VEGF-nanoparticles + PDGF-nanoparticles; NECINH-nanoparticles; VEGF-nanoparticles + NECINH-nanoparticles; and VEGF-nanoparticles + PDGF-nanoparticles + NECINH-nanoparticles) before auto-transplantation. Grafts were recovered after 5 or 21 days for analyses of tissue integrity (hematoxylin-eosin staining), spermatogonial survival (immuno-histo-chemistry for promyelocytic leukemia zinc finger) and vascularization (immuno-histo-chemistry for α-smooth muscle actin and CD-31). Our results showed that a combination of VEGF and PDGF nanoparticles increased vascular maturity and induced a faster maturation of vascular structures in grafts.


Subject(s)
Hydrogels/chemistry , Nanoparticles/administration & dosage , Neovascularization, Physiologic/drug effects , Platelet-Derived Growth Factor/administration & dosage , Testis/transplantation , Vascular Endothelial Growth Factor A/administration & dosage , Alginates/chemistry , Animals , Drug Liberation , Fertility Preservation/methods , Humans , Male , Mice, Inbred Strains , Nanoparticles/chemistry , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/pharmacokinetics , Spermatogonia/drug effects , Testis/blood supply , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/pharmacokinetics
6.
Clin Podiatr Med Surg ; 38(2): 235-244, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33745654

ABSTRACT

The treatment of Achilles tendinitis from conservative to minimally invasive to surgery gives patients a wide range of treatment options for this common pathology. The use and role of biologics to augment this treatment is emerging. The use of biologics may enhance the healing potential of the Achilles tendon when conservative treatment fails. There are a handful of biologics being investigated to obtain if improved outcomes can be maximized.


Subject(s)
Achilles Tendon , Tendinopathy/therapy , Bone Marrow Transplantation , Hepatocytes/transplantation , Humans , Leukocytes, Mononuclear/transplantation , Platelet-Derived Growth Factor/administration & dosage , Platelet-Rich Plasma , Tissue Scaffolds , Transforming Growth Factor beta/administration & dosage , Ultrasonography, Interventional , Vascular Endothelial Growth Factor A/administration & dosage , Wound Healing
7.
J Orthop Surg Res ; 16(1): 43, 2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33430899

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) are used for the treatment of osteoarthritis (OA), and MSC genetic engineering is expected to enhance cartilage repair. Here, we aimed to investigate the effect of MSCs overexpressing platelet-derived growth factor (PDGF) or heme oxygenase-1 (HO-1) in chondrocytes and synovial cells with an OA phenotype and assess the in vivo efficacy of intra-articular injections of these MSCs in canine OA models. METHODS: Canine adipose-derived MSCs were transfected with canine PDGF (PDGF-MSCs) or HO-1 (HO-1-MSCs) using lentiviral vectors. Canine chondrocytes or synovial cells were stimulated with lipopolysaccharide (LPS) to mimic the inflammatory OA model and then co-cultured with MSCs, PDGF-MSCs, or HO-1-MSCs for 24 h and 72 h. The mRNA levels of pro-inflammatory, extracellular matrix-degradative/synthetic, or pain-related factors were measured after co-culture by real-time PCR. Furthermore, a surgery-induced canine OA model was established and the dogs were randomized into four groups: normal saline (n = 4), MSCs (n = 4), PDGF-MSCs (n = 4), and HO-1-MSCs (n = 4). The OA symptoms, radiographic OA severity, and serum matrix metallopeptidase (MMP)-13 levels were assessed before and 10 weeks after treatment, to evaluate the safety and efficacy of the modified MSCs. RESULTS: PDGF or HO-1 overexpression significantly reduced the expression of pro-inflammatory factors, MMP-13, and nerve growth factor elicited by LPS and increased that of aggrecan and collagen type 2 in chondrocytes (P < 0.05). In addition, the expression of aggrecanases was significantly downregulated in synovial cells, whereas that of tissue inhibitor of metalloproteinases was upregulated (P < 0.05). Furthermore, the co-cultured MSCs highly expressed genes that contributed to the maintenance of joint homeostasis (P < 0.05). In vivo studies showed that OA symptoms improved after administration of all MSCs. Also, PDGF-MSCs significantly improved limb function and reduced pain (P < 0.05). The results of the radiographic assessment and serum MMP-13 levels did not vary significantly compared to those of the control. CONCLUSIONS: Genetically modifying PDGF and HO-1 in MSCs is an effective strategy for treating OA, suggesting that PDGF-MSCs can be novel therapeutic agents for improving OA symptoms.


Subject(s)
Genetic Engineering/methods , Heme Oxygenase-1/administration & dosage , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Osteoarthritis/therapy , Platelet-Derived Growth Factor/administration & dosage , Animals , Biomarkers/blood , Cells, Cultured , Chondrocytes/metabolism , Disease Models, Animal , Dogs , Gene Expression , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Matrix Metalloproteinase 13/blood , Osteoarthritis/diagnosis , Osteoarthritis/genetics , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Radiography , Synovial Membrane/cytology , Synovial Membrane/metabolism
8.
Neurosci Lett ; 727: 134930, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32251708

ABSTRACT

Accumulated evidence indicates that platelet-derived growth factor (PDGF) contributes to various types of tissue regeneration. However, the effects and mechanisms of PDGF signaling for retina regeneration have not been sufficiently investigated. To clarify this, we investigated the role of PDGF signaling in retina regeneration process after needle puncture in zebrafish. Time-course analysis showed a spike peak of pdgf-a at 6 h after injury and a broad peak of pdgf-b during 6-96 h after injury. Inhibition of PDGF signaling with AG1295 suppressed BrdU-positive proliferative cell numbers at 4 days after injury. At the same time, retina regeneration-associated transcription factors, ascl1a and pax6b, were down-regulated by AG1295 treatment. Intravitreal injection of human recombinant PDGF-AA or -BB into intact zebrafish induced the cell proliferation. PDGF-BB injection induced the Müller glia-derived neurogenic cluster; PDGF-AA increased the 4C4-positive microglia. These findings indicate that PDGF signaling contributes to retina regeneration in zebrafish and causes different types of cell proliferation, depending on each subtype of PDGF. (160 words).


Subject(s)
Becaplermin/administration & dosage , Nerve Regeneration/physiology , Platelet-Derived Growth Factor/administration & dosage , Retina/physiology , Signal Transduction/drug effects , Animals , Animals, Genetically Modified , Becaplermin/metabolism , Humans , Intravitreal Injections/methods , Nerve Regeneration/drug effects , Platelet-Derived Growth Factor/metabolism , Retina/drug effects , Signal Transduction/physiology , Zebrafish , Zebrafish Proteins/metabolism
9.
J Cosmet Dermatol ; 19(3): 705-713, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31397540

ABSTRACT

BACKGROUND: As skin ages, a functional decrement occurs. To avoid future vulnerability to dermatologic diseases, an optimal cutaneous regeneration is mandatory. Biological therapies based on blood-derived autologous proteins are gaining attention of scientists and dermatologists. OBJECTIVES: A novel 100% autologous topical serum has been developed using plasma rich in growth factors technology. The physicochemical characterization and the biologic potential of the novel formulation have been studied. METHODS: Rheological and mechanical properties and the biological capacity of the formulation were characterized. Human dermal fibroblast culture and 3D organotypic skin explants were used as in vitro and ex vivo cutaneous models, respectively. RESULTS: The autologous topical serum presented an optimal spreadability index and appropriate shear thinning behavior that allowed an easy handling and rapid integration within the cutaneous tissue. The formulation has a high growth factor load with the ability to progressively penetrate into the dermal/epidermal layers of the skin. It is biocompatible and promotes cell proliferation and chemotactic activity. The autologous topical serum promotes the biosynthetic activity of cells by the stimulation of collagen and hyaluronic acid expression. CONCLUSIONS: These findings present an in situ and easy to prepare autologous topical serum based on the patient's own blood with physicochemical and bioactive properties that may be used for skin regeneration purposes.


Subject(s)
Biological Factors/administration & dosage , Blood Transfusion, Autologous/methods , Platelet-Derived Growth Factor/administration & dosage , Regeneration/drug effects , Skin Aging/drug effects , Administration, Cutaneous , Biological Factors/pharmacokinetics , Cells, Cultured , Collagen/metabolism , Drug Evaluation, Preclinical , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hyaluronic Acid/metabolism , Platelet-Derived Growth Factor/pharmacokinetics , Primary Cell Culture , Skin/cytology , Skin/drug effects , Skin Absorption , Tissue Culture Techniques
10.
J Orthop Res ; 37(11): 2307-2315, 2019 11.
Article in English | MEDLINE | ID: mdl-31318103

ABSTRACT

The objective of this study was to improve cartilage repair and integration using self-assembling KLD hydrogel functionalized with platelet-derived growth factor-BB and heparin-binding insulin-like growth factor-1 with associated enzymatic trypsin pre-treatment of the native cartilage. Bilateral osteochondral defects were created at the central portion of the femoral trochlear groove of 48 skeletally mature, white New Zealand rabbits. One limb received a randomly assigned treatment and the contralateral limb served as the control. Treated defects were exposed to trypsin for 2 min and filled with self-assembling KLD hydrogel only, or associated to growth factors. All control limbs received KLD hydrogel alone or received only trypsin but not hydrogel. Ninety days post-defect creation, the rabbits were euthanized and magnetic resonance imaging, radiography, macroscopic evaluation, histology, and immunohistochemistry of the joint and repaired tissue were performed. Mixed model analyses of variance were utilized to assess the outcome parameters and individual comparisons were performed using Least Square Means procedure and differences with p-value < 0.05 were considered significant. Trypsin enzymatic pre-treatment improved cellular morphology, cluster formation and subchondral bone reconstitution. Platelet-derived growth factor-BB improved subchondral bone healing and basal integration. Heparin-binding insulin-like growth factor-1 associated with platelet-derived growth factor improved tissue and cell morphology. The authors conclude that self-assembling KLD hydrogel functionalized with platelet-derived growth factor and heparin-binding insulin-like growth factor-1 with associated enzymatic pre-treatment of the native cartilage with trypsin resulted in an improvement on the cartilage repair process. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2307-2315, 2019.


Subject(s)
Fractures, Cartilage/therapy , Insulin-Like Growth Factor I/administration & dosage , Platelet-Derived Growth Factor/administration & dosage , Trypsin/administration & dosage , Animals , Cartilage, Articular/pathology , Drug Carriers , Drug Evaluation, Preclinical , Fractures, Cartilage/diagnostic imaging , Fractures, Cartilage/pathology , Hydrogels , Rabbits
11.
Curr Drug Deliv ; 16(5): 472-483, 2019.
Article in English | MEDLINE | ID: mdl-30894109

ABSTRACT

Chronic wounds are the result of alterations in the complex series of events of physiological wound healing. In particular, the prolonged inflammation results in increased protease activity, in the degradation of extracellular matrix (ECM) and of growth factors (GFs). The relevance of platelet GFs in maintaining and restoring the complex equilibrium of different moments in wound healing is well recognized. Moreover, the observed decrease of their levels in chronic wounds suggested a possible therapeutic role of the external application to the wounds. It has been also pointed out that tissue regeneration can be more efficiently obtained by the synergic use of different GFs. Platelet derivatives such as platelet- rich plasma (PRP) and platelet lysate (PL) are able to release GFs in a balanced pool. Their therapeutic use in regenerative medicine and wound healing has been therefore more and more frequently proposed in clinical trials and in the literature. The development of a suitable formulation able to control the GFs release rate, to protect the GFs, and to assure their prolonged contact with the wound site, is of paramount importance for the therapeutic success. The present review considers some formulation approaches for PRP and PL application to wounds.


Subject(s)
Platelet-Derived Growth Factor/administration & dosage , Platelet-Rich Plasma , Wound Healing , Animals , Humans
12.
Biotechnol Bioeng ; 116(1): 155-167, 2019 01.
Article in English | MEDLINE | ID: mdl-30229864

ABSTRACT

Spinal cord injury (SCI) results in paralysis below the injury and strategies are being developed that support axonal regrowth, yet recovery lags, in part, because many axons are not remyelinated. Herein, we investigated strategies to increase myelination of regenerating axons by overexpression of platelet-derived growth factor (PDGF)-AA and noggin either alone or in combination in a mouse SCI model. Noggin and PDGF-AA have been identified as factors that enhance recruitment and differentiation of endogenous progenitors to promote myelination. Lentivirus encoding for these factors was delivered from a multichannel bridge, which we have previously shown creates a permissive environment and supports robust axonal growth through channels. The combination of noggin+PDGF enhanced total myelination of regenerating axons relative to either factor alone, and importantly, enhanced functional recovery relative to the control condition. The increase in myelination was consistent with an increase in oligodendrocyte-derived myelin, which was also associated with a greater density of cells of an oligodendroglial lineage relative to each factor individually and control conditions. These results suggest enhanced myelination of regenerating axons by noggin+PDGF that act on oligodendrocyte-lineage cells post-SCI, which ultimately led to improved functional outcomes.


Subject(s)
Carrier Proteins/administration & dosage , Genetic Therapy/methods , Myelin Sheath/drug effects , Nerve Regeneration , Platelet-Derived Growth Factor/administration & dosage , Regenerative Medicine/methods , Spinal Cord Injuries/therapy , Animals , Carrier Proteins/genetics , Disease Models, Animal , Drug Carriers/administration & dosage , Genetic Vectors , Lentivirus/genetics , Mice , Platelet-Derived Growth Factor/genetics , Treatment Outcome
13.
Eur Rev Med Pharmacol Sci ; 22(20): 6912-6921, 2018 10.
Article in English | MEDLINE | ID: mdl-30402857

ABSTRACT

OBJECTIVE: This study aims to explore whether homeobox A11 antisense RNA (HOXA11-AS) could regulate inflammation induced by diabetic arteriosclerosis (DAA) via PI3K/AKT pathway. PATIENTS AND METHODS: Quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) was used to detect expressions of HOXA11-AS and proinflammatory genes in carotid endarterectomy samples of symptomatic and asymptomatic atherosclerosis (AS) patients, diabetes mellitus (DM), and non-DM patients. The above-mentioned genes in DM animal model and non-DM animal model were also detected. We detected the expression of HOXA11-AS in vascular smooth muscle cells (VSMCs) treated with platelet-derived growth factor (PDGF) or PDGF inhibitor imatinib, respectively. Subsequently, we applied cell transfection technology to interfere with the expression of HOXA11-AS in VSMCs. In vascular endothelial cells (VECs) and VSMCs, we detected the effect of HOXA11-AS on the expressions of genes related to the proliferation, migration, and cell cycle. Then, VSMCs were treated with tumor necrosis factor-α (TNF-α), and the expression of HOXA11-AS was examined in VSMCs. The effect of HOXA11-AS on TNF-α-induced inflammation in VSMCs was detected as well. Finally, we analyzed the effect of HOXA11-AS on PDGF-induced activation of PI3K/AKT pathway in VSMCs and VECs. RESULTS: HOXA11-AS expression was markedly increased in carotid endarterectomy specimens of symptomatic AS patients compared to that of asymptomatic AS patients. Expression levels of HOXA11-AS and pro-inflammatory genes were significantly elevated in carotid endarterectomy specimens of DM patients. Similarly, HOXA11-AS expression was also significantly increased in carotid arteries of DM mice compared with that of non-DM mice. PDGF could upregulate HOXA11-AS expression in VSMCs, which was reversed by PDGF inhibitor imatinib. HOXA11-AS knockdown could reduce the expressions of the proliferation-associated gene (PCNA) and the cycle-related genes (p21, p53), and also inhibited the proliferation and migration of VSMCs induced by PDGF. HOXA11-AS was upregulated by TNF-α. HOXA11-AS knockdown remarkably downregulated expressions of inflammation-related genes in VSMCs induced by TNF-α. In VECs, low expression of HOXA11-AS can inhibit the expression of TNF-α-induced pro-inflammatory genes and PDGF-induced vascular inflammation-related genes. Low expression of HOXA11-AS inhibited PDGF-induced activation of PI3K/AKT pathway in VSMCs and VECs. CONCLUSIONS: HOXA11-AS may participate in DAA by activating the PI3K/AKT pathway to regulate inflammation in VSMCs and VECs.


Subject(s)
Arteriosclerosis/genetics , Diabetic Angiopathies/etiology , Homeodomain Proteins/genetics , Animals , Cell Cycle/genetics , Cell Proliferation/genetics , Cells, Cultured , Down-Regulation , Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Mice , Myocytes, Smooth Muscle/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Up-Regulation
14.
Injury ; 49(4): 775-783, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29503013

ABSTRACT

BACKGROUND: It has been previously suggested that the use of regenerative promoters, which include bone marrow-derived mesenchymal stem cells (MSCs) or natural growth factors supplement such as platelet-rich concentrate (PRC) could promote cartilage regeneration. However, the notion that the concurrent use of both promoters may provide a synergistic effect that improves the repair outcome of focal cartilage injury has not been previously demonstrated. This study was thus conducted to determine whether the concomitant use of PRC could further enhance the reparative potential of MSCs encapsulated in alginate transplanted into focal cartilage injury in rabbits. METHODS: Artifically created full thickness cartilage defects were made on the weight-bearing region of medial femoral condyles in bilateral knees of New Zealand White rabbits (N = 30). After one month, the right knee was treated with either i) PRC (n = 10), ii) MSCs (n = 10), or, iii) a combination of PRC and MSCs (PRC + MSC) (n = 10), all encapsulated in alginate. The left knee remained untreated (control). Rabbits were sacrificed at 3 and 6 months after treatment. Cartilage tissue regeneration was accessed using ICRS morphologic scoring, histologic grading by O'Driscoll scoring, immunohistochemical staining and quantitative analysis of glycosaminoglycans (GAG) per total protein content. RESULTS: At 3 months, transplantation using PRC alone was equally effective as MSCs in inducing the repair of cartilage defects. However, PRC + MSC resulted in significantly higher ICRS and O'Driscoll scores (p < 0.05) as compared to other groups. The regenerated tissues from the PRC + MSC group also had stronger staining for Safranin-O and collagen type II. By 6 months, in addition to superior ICRS and O'Driscoll scores as well as stronger staining, glycosaminoglycan per total protein content was also significantly higher (p < 0.05) in the PRC + MSC group (3.4 ±â€¯0.3 µg/mg) as compared to the MSC (2.6 ±â€¯0.2 µg/mg) or PRC (2.1 ±â€¯0.2 µg/mg) groups. CONCLUSION: PRC enhances the reparative effects of MSC in treating focal articular cartilage injuries.


Subject(s)
Biological Products/pharmacology , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Chondrocytes/metabolism , Knee Injuries/pathology , Knee Injuries/therapy , Platelet-Rich Plasma/cytology , Alginates/pharmacology , Animals , Biological Products/administration & dosage , Cartilage, Articular/injuries , Cells, Cultured , Disease Models, Animal , Intra-Articular Fractures/pathology , Intra-Articular Fractures/therapy , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Platelet-Derived Growth Factor/administration & dosage , Platelet-Derived Growth Factor/pharmacology , Rabbits
15.
Clin Rehabil ; 32(3): 330-339, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28783969

ABSTRACT

OBJECTIVE: Aim of this trial was to compare efficacy of activated platelet-rich plasma against hyaluronic acid as intra-articular injections to people with osteoarthritis of the knee. DESIGN: Phase-2 randomized controlled trial, with blind patients and outcome assessors. SETTING: Outpatient rehabilitation service; years 2011-2013. SUBJECTS: Patients with knee osteoarthritis grades 2-3 at magnetic resonance imaging (MRI) were included after consent and randomized. Target sample size was 25 patients per group. INTERVENTIONS: Patients received three activated platelet-rich plasma (intervention group) or hyaluronic acid (controls) intra-articular injections at 4-week intervals. MAIN MEASURES: Main outcome measure was proportion of patients with >1 grade improvement at six months from last injection, as assessed by a radiologist blind to study group. Patients were evaluated over time clinically and with functional scales (Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), Lysholm, Tegner, American Knee Society Score (AKSS), Lequesne, visual analogue scale (VAS) for pain). RESULTS: Overall, 30 patients were randomized to intervention and 28 to control group. For primary outcome, 28 patients (29 knees) in the intervention and 22 (25 knees) in the control group were available. Patients with at least 1 grade improvement at repeat MRI were 14 (48.3%) in the intervention and 2 (8%) in the control group ( P < 0.003). Improvement in symptoms and functional scales was consistently higher in the intervention group. No side-effects were observed in either group. CONCLUSION: Activated platelet-rich plasma reduces articular damage as evident at MRI, as soon as six months after treatment; it reduces pain and improves patient's function and overall quality of life.


Subject(s)
Hyaluronic Acid/administration & dosage , Osteoarthritis, Knee/drug therapy , Platelet-Derived Growth Factor/administration & dosage , Range of Motion, Articular/drug effects , Adult , Aged , Confidence Intervals , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Intra-Articular , Magnetic Resonance Imaging/methods , Male , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Pain Measurement , Proportional Hazards Models , Range of Motion, Articular/physiology , Recovery of Function , Severity of Illness Index , Single-Blind Method , Treatment Outcome
16.
Cell Immunol ; 323: 19-32, 2018 01.
Article in English | MEDLINE | ID: mdl-29111157

ABSTRACT

Angiogenesis is an organized series of events, beginning with vessel destabilization, followed by endothelial cell re-organization, and ending with vessel maturation. The formation of a mature vascular network requires precise spatial and temporal regulation of a large number of angiogenic factors, including vascular endothelial growth factor (VEGF), basic fibroblast growth factor-2 (FGF-2) and platelet-derived growth factor (PDGF). VEGF aids in vascular permeability and endothelial cell recruitment, FGF-2 activates endothelial cell proliferation and migration while PDGF stimulates vascular stability. Accordingly, VEGF may inhibit vessel stabilization while PDGF may inhibit endothelial cell recruitment. Therefore, a new polymeric system was prepared by the supercritical carbon dioxide foaming technology, which realized sequential delivery of two or more growth factors with the controlled dose and rate. Increased release of VEGF (71.10%) and FGF-2 (69.76%) compared to PDGF (43.17%) was observed for the first 7 days. Thereafter, up till 21 days, an increased rate of release of BMP-2 compared to VEGF 165 was observed. The effects of PDGF-PLAms/VEGF-FGF-2-PLGA scaffolds on angiogenesis were investigated by human umbilical vein endothelial cells (HUVECs) angiogenic differentiation in vitro and chorioallantoic membrane (CAM) angiogenesis in vivo. Sequential delivery of VEGF, FGF-2 and PDGF from structural polymer scaffolds with distinct kinetics resulted in significant angiogenic differentiation of HUVECs and rapid formation of mature vascular networks in chorioallantoic membrane. This study reported a composite scaffold with distinct release kinetics, and these results clearly indicated the importance of sequential delivery of multiple growth factors in tissue regeneration and engineering.


Subject(s)
Drug Delivery Systems/methods , Fibroblast Growth Factor 2/administration & dosage , Neovascularization, Physiologic/drug effects , Platelet-Derived Growth Factor/administration & dosage , Vascular Endothelial Growth Factor A/administration & dosage , Angiogenesis Inducing Agents/administration & dosage , Angiogenesis Inducing Agents/metabolism , Bone Morphogenetic Protein 2/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chorioallantoic Membrane , Delayed-Action Preparations , Fibroblast Growth Factor 2/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Platelet-Derived Growth Factor/metabolism , Polyesters/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Vascular Endothelial Growth Factor A/metabolism
17.
Acta Biomater ; 65: 237-247, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29111372

ABSTRACT

Spontaneous endothelialization of synthetic vascular grafts may occur via three independent or concurrent modalities: transanastomotic (TA) outgrowth, transmural (TM) ingrowth or fallout (FO) from the blood. The limited TA and FO endothelialization, which occurs in humans, results in poor long-term patency in the small diameter position, where TM ingrowth may offer a clinically relevant alternative. To achieve sequential analysis of each mode of healing, loop grafts comprising anastomotically isolated angiopermissive polyurethane control grafts were abluminally sealed using either ePTFE wraps or solid polyurethane skins and implanted in the rat infrarenal aortic loop model for twelve weeks. Positive control grafts showed improved endothelialization and patency compared to the abluminally isolated mid-grafts. Furthermore, the mid-graft healing was accelerated with surface heparin and heparin-growth factor (VEGF, PDGF) modification in a three-week sub-study. We are thus able to distinguish between the three vascular graft endothelialization modes, and conclude that fallout plays a secondary role to TM healing. The increased endothelialisation for growth factor presenting grafts indicates the promise of this simple approach but further optimization is required. STATEMENT OF SIGNIFICANCE: In addition to the full elucidation of, and differentiation between, the three healing/endothelialisation modes of vascular grafts, the significance of the work relates to the near-complete lack of endothelialisation of small diameter vascular grafts in humans (1-2 cm transanastomotic outgrowth on a graft that may be 60 cm long) even after decades of implantation. The concomitant retained midgraft thrombogenicity leads, together with anastomotic hyperplastic responses, to poor long-term outcomes. The large impact of successful translation of the current research to the achievement of full endothelialisation of long peripheral grafts in humans via transmural ingrowth (half a millimetre distance; thickness of the graft wall), is evident, and supported by the large improvements in clinical patencies achievable in by pre-seeding of ePTFE grafts with confluent endothelia.


Subject(s)
Blood Vessel Prosthesis , Capillaries/growth & development , Endothelium, Vascular/growth & development , Endothelium, Vascular/pathology , Neovascularization, Pathologic , Wound Healing , Animals , Aorta/surgery , Heparin/administration & dosage , Humans , Male , Platelet-Derived Growth Factor/administration & dosage , Polyurethanes , Rats, Wistar , Vascular Endothelial Growth Factor A/administration & dosage
18.
Br J Clin Pharmacol ; 84(3): 434-444, 2018 03.
Article in English | MEDLINE | ID: mdl-29148070

ABSTRACT

AIMS: Topical growth factors accelerate wound healing in patients with diabetic foot ulcers (DFU). Due to the absence of head-to-head comparisons, we carried out Bayesian network meta-analysis to compare the efficacy and safety of growth factors. METHODS: Using an appropriate search strategy, randomized controlled trials on topical growth factors compared with standard of care in patients with DFU, were included. Proportion of patients with complete healing was the primary outcome. Odds ratio (95% confidence interval) was used as the effect estimate and random effects model was used for both direct and indirect comparisons. Markov Chain Monte Carlo simulation was used to obtain pooled estimates. Rankogram was generated based on surface under the cumulative ranking curve (SUCRA). RESULTS: A total of 26 studies with 2088 participants and 1018 events were included. The pooled estimates for recombinant epidermal growth factor (rhEGF), autologous platelet rich plasma (PRP), recombinant human platelet-derived growth factor (rhPDGF) were 5.72 [3.34, 10.37], 2.65 [1.60, 4.54] and 1.97 [1.54, 2.55] respectively. SUCRA for rhEGF was 0.95. Sensitivity analyses did not reveal significant changes from the pooled estimates and rankogram. No differences were observed in the overall risk of adverse events between the growth factors. However, the growth factors were observed to lower the risk of lower limb amputation compared to standard of care. CONCLUSION: To conclude, rhEGF, rhPDGF and autologous PRP significantly improved the healing rate when used as adjuvants to standard of care, of which rhEGF may perform better than other growth factors. The strength of most of the outcomes assessed was low and the findings may not be applicable for DFU with infection or osteomyelitis. The findings of this study needs to be considered with caution as the results might change with findings from head-to-head studies.


Subject(s)
Diabetic Foot/therapy , Epidermal Growth Factor/administration & dosage , Platelet-Derived Growth Factor/administration & dosage , Platelet-Rich Plasma , Amputation, Surgical/statistics & numerical data , Animals , Bayes Theorem , Diabetic Foot/pathology , Humans , Markov Chains , Monte Carlo Method , Randomized Controlled Trials as Topic , Recombinant Proteins , Wound Healing/drug effects
19.
Exp Cell Res ; 362(2): 252-259, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29174980

ABSTRACT

Erectile dysfunction (ED) is the most common sexual disorder that men report to healthcare providers. Gap junctions (GJs) are thought to be responsible for synchronous shrinkage of corpus cavernosum smooth muscle cells (CCSMCs), and play thus an important role in the maintenance of an erection. Hypoxia has been suggested as a pathological mechanism underlying ED. Here we demonstrate that hypoxia increased the expression of platelet-derived growth factor (PDGF) and the main GJ component connexin (Cx)43 in CCSMCs. Inhibiting PDGF receptor (PDGFR) activity decreased Cx43 expression. Treatment with different concentrations of PDGF increased the levels of phosphorylated protein kinase B (AKT), ß-catenin, and Cx43, whereas inhibition of PDGFR or activation of phosphatidylinositol 3 kinase (PI3K)/AKT signaling altered ß-catenin and Cx43 expression. Meanwhile, silencing ß-catenin resulted in the downregulation of Cx43. These results demonstrate that PDGF secretion by CCSMCs and vascular endothelial cells is enhanced under hypoxic conditions, leading to increased Cx43 expression through PI3K/AKT/ß-catenin signaling and ultimately affecting GJ function in ED. Thus, targeting this pathway is a potential therapeutic strategy for the treatment of ED.


Subject(s)
Connexin 43/genetics , Erectile Dysfunction/genetics , Receptors, Platelet-Derived Growth Factor/genetics , beta Catenin/genetics , Animals , Cell Hypoxia/genetics , Erectile Dysfunction/pathology , Gap Junctions/genetics , Gap Junctions/pathology , Gene Expression Regulation/drug effects , Humans , Male , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Penis/metabolism , Penis/physiopathology , Phosphatidylinositol 3-Kinases/genetics , Platelet-Derived Growth Factor/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction/drug effects
20.
Invest Ophthalmol Vis Sci ; 58(14): 6299-6307, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29242904

ABSTRACT

Purpose: Platelet-derived growth factor (PDGF)-BB is known to have neuroprotective effects against various neurodegenerative disorders. The purpose of this study was to determine whether PDGF-BB can be neuroprotective against light-induced photoreceptor damage in mice. Methods: Mice were exposed to 8000-lux luminance for 3 hours to induce phototoxicity. Two hours before light exposure, the experimental mice were injected with PDGF-BB intravitreally, and the control mice were injected with phosphate-buffered saline. The light-exposed PDGF-BB-injected mice and saline-injected mice were evaluated electroretinographically and histologically. The site and expression levels of PDGFR-ß and PDGF-BB were determined by immunostaining and Western blotting, respectively. The effect of PDGF-BB on light-induced cone and rod photoreceptor damage was also evaluated in vitro in 661W cells, a murine cone photoreceptor cell line, and in primary retinal cell cultures. Results: An intravitreal injection of PDGF-BB significantly reduced the decrease in the amplitudes of the electroretinograms (ERGs) and the thinning of the outer nuclear layer (ONL) induced by the light exposure. It also reduced the number of TUNEL-positive cells in the ONL. PDGFR-ß was expressed in the rod outer segments (OSs) but not the cone OSs. The levels of PDGF-BB and PDGFR-ß were decreased after light irradiation. In addition, PDGF-BB had protective effects against light-induced damage to cells of rod photoreceptors but had no effect on the 661W cells in vitro. Conclusions: These findings indicate that PDGF-BB reduces the degree of light-induced retinal damage by activating PDGFR-ß in rod photoreceptors. These findings suggest that PDGF-BB could play a role in the prevention of degeneration in eyes susceptible to phototoxicity.


Subject(s)
Light/adverse effects , Photoreceptor Cells, Vertebrate/drug effects , Pregnancy, Animal , Proto-Oncogene Proteins c-sis/administration & dosage , Radiation Injuries, Experimental/prevention & control , Retinal Diseases/prevention & control , Angiogenesis Inducing Agents/administration & dosage , Animals , Animals, Newborn , Becaplermin , Blotting, Western , Cell Death/drug effects , Cell Death/radiation effects , Cell Line , Electroretinography , Female , Immunohistochemistry , In Situ Nick-End Labeling , Intravitreal Injections , Male , Mice , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/radiation effects , Platelet-Derived Growth Factor/administration & dosage , Pregnancy , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/physiopathology , Recombinant Proteins , Retinal Diseases/etiology , Retinal Diseases/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...