Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Int J Mol Sci ; 25(12)2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38928156

RESUMO

Osteoarthritis (OA) is a degenerative joint disorder characterized by the progressive deterioration of articular cartilage driven and sustained by catabolic and inflammatory processes that lead to pain and functional impairment. Adipose-derived stem cells (ASCs) have emerged as a promising therapeutic strategy for OA due to their regenerative potential, which mainly relies on the adaptive release of paracrine molecules that are soluble or encapsulated in extracellular vesicles (EVs). The biological effects of EVs specifically depend on their cargo; in particular, microRNAs (miRNAs) can specifically modulate target cell function through gene expression regulation. This study aimed to investigate the impact of collection site (abdominal vs. peri-trochanteric adipose tissue) and collection method (surgical excision vs. lipoaspiration) on the miRNAs profile in ASC-derived EVs and their potential implications for OA therapy. EV-miRNA cargo profiles from ASCs of different origins were compared. An extensive bioinformatics search through experimentally validated and OA-related targets, pathways, and tissues was conducted. Several miRNAs involved in the restoration of cartilage homeostasis and in immunomodulation were identified in all ASC types. However, EV-miRNA expression profiles were affected by both the tissue-harvesting site and procedure, leading to peculiar characteristics for each type. Our results suggest that adipose-tissue-harvesting techniques and the anatomical site of origin influence the therapeutic efficacy of ASC-EVs for tissue-specific regenerative therapies in OA, which warrants further investigation.


Assuntos
Tecido Adiposo , Vesículas Extracelulares , Células-Tronco Mesenquimais , MicroRNAs , Humanos , Vesículas Extracelulares/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Mesenquimais/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/citologia , Osteoartrite/metabolismo , Osteoartrite/terapia , Osteoartrite/genética , Osteoartrite/patologia , Feminino , Masculino , Pessoa de Meia-Idade , Regulação da Expressão Gênica
2.
Artigo em Inglês | MEDLINE | ID: mdl-38842036

RESUMO

PURPOSE: Chondrocyte-based cell therapies are effective for the treatment of chondral lesions, but remain poorly indicated for diffuse lesions in the context of early osteoarthritis (OA). The aim of this study was to develop a protocol to obtain chondroprogenitor cells suitable for the treatment of diffuse chondral lesions within early OA. METHODS: Cartilage cells were expanded at low density in human platelet lysate (hPL). A test was performed to exclude senescence. The expression of surface cluster of differentiation 146, cluster of differentiation 166, major histocompatibility complex (MHC)-I and MHC-II and of genes of interest were evaluated, as well as the trophic potential of these cells, by the assessment of lubricin and matrix production. The immunomodulatory potential was assessed through their co-culture with macrophages. RESULTS: Cartilage cells expanded at low density in hPL showed higher proliferation rate than standard-density cells, no replicative senescence, low immunogenicity and expression of lubricin. Moreover, they presented an increased expression of chondrogenic and antihypertrophic markers, as well as a superior matrix deposition if compared to cells cultured at standard density. Cartilage cells induced on macrophages an upregulation of CD206, although a higher increase of CD163 expression was observed in the presence of low-density cells. CONCLUSIONS: These findings lay the grounds to explore the clinical usefulness of low-density cultured cartilage cells to treat diffuse lesions in early OA joints for both autologous and allogenic use. LEVEL OF EVIDENCE: Not applicable.

3.
BMC Musculoskelet Disord ; 24(1): 647, 2023 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-37573322

RESUMO

BACKGROUND: Knee osteoarthritis (OA) is a progressive and degenerative condition. Several pharmacological and non-pharmacological treatments are able to improve the OA symptoms and the structural characteristics of the affected joints. Among these, infiltrative therapy with hyaluronic acid (HA) is the most used and consolidated procedure for the pain management. The addition of skin conditioning peptides to HA promotes the cartilage remodeling processes and a better permeation of the HA-based gel containing a peptide mixture, CR500®. Furthermore, the topic route of administration is convenient over the routinely used intra-articular injective procedures. In this study, the effectiveness of CR500® was evaluated in terms of improvement of the algo-functional symptoms related to unilateral knee OA. METHODS: 38 mild and moderate OA patients were enrolled at a screening visit (V-1), treated at baseline visit (V1), and then continued the topical application of CR500® twice a week for 4 weeks, and followed-up for 3 visits (V2-V4) from week 2 to 4. Lequesne Knee Index (LKI) and Knee injury and Osteoarthritis Outcome Score (KOOS) were collected. Synovial fluid was collected and used for the quantification of neoepitope of type II collagen (C2C), C-terminal telopeptide of type II collagen (CTX-II), type II collagen propeptide (CPII), tumor necrosis factor alpha (TNFα) and HA. The expression of CD11c and CD206 was evaluated on cell pellets. RESULTS: Three patients were excluded, thus 35 patients were included in the analysis. The treatment with CR500® was safe and well tolerated, with 7.9% patients had mild adverse events, not related to the device. The LKI total score showed a significant decrease from V1 to V4. KOOS score also showed a significant improvement of patient condition at V2, V3 and V4 in comparison with V1 for all subscales, except for KOOS sport subscale which improved only from V3. At V1 a negative correlation among KOOS pain subscale values and C2C, CPII and TNFα levels was observed, as well as a positive correlation between KOOS pain subscale and CD11c/CD206 ratio. CONCLUSION: CR500® is safe and appear to be effective in improving pain and function in OA patients during the 4 weeks of treatment. TRIAL REGISTRATION: ClinicalTrials.gov ID: NCT05661162. This trial was registered on 22/12/2022.


Assuntos
Osteoartrite do Joelho , Humanos , Osteoartrite do Joelho/tratamento farmacológico , Osteoartrite do Joelho/induzido quimicamente , Colágeno Tipo II , Fator de Necrose Tumoral alfa , Resultado do Tratamento , Ácido Hialurônico , Dor/tratamento farmacológico , Injeções Intra-Articulares
4.
Front Surg ; 10: 1172313, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37425349

RESUMO

Introduction: A novel classification scheme for endplate lesions, based on T2-weighted images from magnetic resonance imaging (MRI) scan, has been recently introduced and validated. The scheme categorizes intervertebral spaces as "normal," "wavy/irregular," "notched," and "Schmorl's node." These lesions have been associated with spinal pathologies, including disc degeneration and low back pain. The exploitation of an automatic tool for the detection of the lesions would facilitate clinical practice by reducing the workload and the diagnosis time. The present work exploits a deep learning application based on convolutional neural networks to automatically classify the type of lesion. Methods: T2-weighted MRI scans of the sagittal lumbosacral spine of consecutive patients were retrospectively collected. The middle slice of each scan was manually processed to identify the intervertebral spaces from L1L2 to L5S1, and the corresponding lesion type was labeled. A total of 1,559 gradable discs were obtained, with the following types of distribution: "normal" (567 discs), "wavy/irregular" (485), "notched" (362), and "Schmorl's node" (145). The dataset was divided randomly into a training set and a validation set while preserving the original distribution of lesion types in each set. A pretrained network for image classification was utilized, and fine-tuning was performed using the training set. The retrained net was then applied to the validation set to evaluate the overall accuracy and accuracy for each specific lesion type. Results: The overall rate of accuracy was found equal to 88%. The accuracy for the specific lesion type was found as follows: 91% (normal), 82% (wavy/irregular), 93% (notched), and 83% (Schmorl's node). Discussion: The results indicate that the deep learning approach achieved high accuracy for both overall classification and individual lesion types. In clinical applications, this implementation could be employed as part of an automatic detection tool for pathological conditions characterized by the presence of endplate lesions, such as spinal osteochondrosis.

5.
Knee Surg Sports Traumatol Arthrosc ; 31(6): 2338-2348, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35716187

RESUMO

PURPOSE: To evaluate the mid- and long-term efficacy of autologous chondrocyte implantation (ACI) and matrix-assisted chondrocyte implantation (MACI) to treat patients with knee cartilage defects in the presence of osteoarthritis (OA). METHODS: PubMed and Cochrane databases were systematically searched for studies describing the treatment of knee OA with ACI or MACI (Kellgren-Lawrence (KL) ≥ 1, minimum follow-up 36 months). Results were reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines and included Lysholm, Western Ontario McMaster University and International Knee Documentation Committee scores. RESULTS: Of the 127 full-text articles assessed for eligibility, only five studies were selected based on inclusion/exclusion criteria (2 on ACI and 3 on MACI). In both groups, the defects were mainly located at femoral level, size 2.2-15.1 cm2 in the ACI and 2.0-7.6 cm2 in the MACI group. ACI was mostly used for patients affected by KL I, whereas MACI for patients with KL II-IV. The data obtained from 235 patients (161 ACI, 74 MACI) showed that ACI and MACI sustained stable clinical improvements up to 11 and 15 years, respectively, with a failure rate of about 10% up to 11 years. Scarce biological details regarding chondrocyte implantation were reported. CONCLUSIONS: ACI and MACI procedures for the treatment of knee cartilage lesions associated to OA showed long-term success and allowed delaying arthroplasty. Additional trials reporting homogenous data and precise patient characterization are needed to conduct an effective literature meta-analysis and identify the clinical relevance of these procedures. LEVEL OF EVIDENCE: IV.


Assuntos
Doenças das Cartilagens , Cartilagem Articular , Procedimentos Ortopédicos , Osteoartrite do Joelho , Humanos , Osteoartrite do Joelho/cirurgia , Condrócitos/transplante , Cartilagem Articular/cirurgia , Procedimentos Ortopédicos/métodos , Doenças das Cartilagens/cirurgia , Articulação do Joelho/cirurgia , Transplante Autólogo/métodos
6.
Knee Surg Sports Traumatol Arthrosc ; 31(8): 3079-3090, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36040510

RESUMO

PURPOSE: Current conservative treatments for knee OA provide limited benefits, with symptoms relief for a short amount of time. Regenerative medicine approaches such as the use of microfragmented adipose tissue (mFAT) showed promising results in terms of durable effects and the possibility to enhance tissue healing and counteract the progression of the pathology. Nevertheless, up to today, the large part of clinical data about mFAT use refers to uncontrolled studies, especially in the surgical setting. The purpose of this study was to evaluate the effectiveness of mFAT applied in association with arthroscopic debridement (AD) for the treatment of knee OA, in terms of symptoms relief and tissue healing. METHODS: This study is a prospective, randomized controlled clinical trial. 78 patients affected by knee OA grade 3-4 according to KL classification were randomly assigned to AD or AD + mFAT treatment groups. Clinical, radiological and serological assessments were performed at 6 months after treatment. Additional clinical evaluation was performed at the end of the study with an average follow-up of 26.1 ± 9.5 months. VAS, KOOS, WOMAC and SF-12 were also collected at both timepoints, KSS only at 6 months. RESULTS: Treatment with AD + mFAT improved functional scores at both 6 months (KOOS-PS: + 11.7 ± 20.2 vs + 24.4 ± 22.5, in AD and AD + mFAT, respectively, p = 0.024; KSS: + 14.9 ± 15.9 vs + 24.8 ± 23.5, in AD and AD + mFAT, respectively, p = 0.046) and 24-month follow-ups (KOOS-PS Functional subscale: - 2.0 ± 3.5 vs - 4.7 ± 4.2, in AD and AD + mFAT, respectively, p = 0.012). Lower T2-mapping scores were obtained in AD + mFAT-treated group in medial and lateral condyle compartments (p < 0.001). Slight increase was observed in the levels of a serum biomarker of cartilage deposition (PIIINP) in both groups at 6-month follow-up (p = 0.037). CONCLUSION: mFAT improves functional outcome and MRI appearance when used in association with AD, therefore supporting its use in the treatment of knee OA in an arthroscopic setting.


Assuntos
Osteoartrite do Joelho , Humanos , Desbridamento/métodos , Estudos Prospectivos , Articulação do Joelho/cirurgia , Tecido Adiposo , Resultado do Tratamento
7.
Brain Spine ; 2: 100858, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36248110

RESUMO

Introduction: A precise knowledge of the possible Adverse Events (AEs) related to spinal surgical procedures is crucial in clinical practice. Research Question: Purposes of this study are: to determine the prevalence and severity of perioperative AEs associated with pediatric and adult spine surgery in a high volume center; to estimate the impact of perioperative AEs on length of hospital stay (LOS). Material and Methods: This is a prospective, observational, monocenter study, including 346 consecutive patients (294 adults and 52 pediatrics). The SAVES-V2 questionnaire was used to record AEs. The form was updated by the medical staff every time an adverse event was recorded during hospitalization. Results: 21,2% of pediatric patients and 20,7% of adults had at least 1 perioperative AEs. In adults, dural tear (3.1%) and neuropathic pain (4,8%), were the most frequent intraoperative and postoperative AE, respectively. In pediatric patients, neurologic deterioration was the most frequent postoperative AE. A diagnosis of deformity (p=0.01), an ASA grade equal or superior to 3 (p=0.023) and the procedure 'Posterior Spinal Fusion' (p=0.001) were associated with a higher frequency of AEs. AEs required prolonged LOS in 40 cases, 7 (70%) pediatric patients and 33 (65%) adults. Discussion and Conclusion: The overall prevalence of AEs is 20.8%, and, although the distribution is almost equal between adult and pediatric patients, their severity is related to age, being higher in pediatric patients. Deformities, deformity correction, revision surgery and AP surgery are the most impactful factors. AEs seriously affect hospitalization, with prolonged LOS (mean 6 days).

8.
Front Med (Lausanne) ; 9: 992386, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36314003

RESUMO

The purpose of the present study is to predict by bioinformatics the activity of the extracellular vesicle (EV)-embedded micro RNA (miRNAs) secreted by cartilage cells (CCs), adipose tissue-derived- (ASCs), and bone marrow-derived stem cells (BMSCs) and verify their immunomodulatory potential supporting our bioinformatics findings to optimize the autologous cell-based therapeutic strategies for osteoarthritis (OA) management. Cells were isolated from surgical waste tissues of three patients who underwent total hip replacement, expanded and the EVs were collected. The expression of EV-embedded miRNA was evaluated with the QuantStudio 12 K Flex OpenArray® platform. Mientournet and ingenuity pathway analysis (IPA) were used for validated target prediction analysis and to identify miRNAs involved in OA and inflammation. Cells shared the expression of 325 miRNAs embedded in EVs and differed for the expression of a small number of them. Mienturnet revealed no results for miRNAs selectively expressed by ASCs, whereas miRNA expressed by CCs and BMSCs were putatively involved in the modulation of cell cycle, senescence, apoptosis, Wingless and Int-1 (Wnt), transforming growth factor beta (TGFß), vascular endothelial growth factor (VEGF), Notch, Hippo, tumor necrosis factor alpha (TNFα), interleukin 1 beta (IL-1ß), insulin like growth factor 1 (IGF-1), RUNX family transcription factor 2 (RUNX2), and endochondral ossification pathways. Cartilage homeostasis, macrophages and T cells activity and inflammatory mediators were identified by IPA as targets of the miRNAs found in all the cell populations. Co-culture tests on macrophages and T cells confirmed the immuno-modulatory ability of CCs, ASCs, and BMSCs. The study findings support the rationale behind the use of cell-based therapy for the treatment of OA.

9.
Int Orthop ; 46(2): 391-400, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34727209

RESUMO

PURPOSE: The aims of the present study were: (1) to characterize the bone-marrow aspirate (BMA) obtained with a centrifuge-free process, employing a dedicated aspiration device; (2) to test the in vitro efficacy of BMA in a model of cartilage inflammation; and (3) to report the preliminary clinical results in a small cohort of patients affected by knee OA. METHODS: Ten patients (4 M, 6 W; mean age: 51.9 ± 9.2 yy) affected by mild to moderate unicompartmental knee OA (KL grade 2-3) were treated by intra-articular and subchondral injections of BMA obtained by a centrifuge-free process. To evaluate the effectiveness of the device in harvesting mesenchymal stem cells (MSCs), samples of the obtained BMA were tested by flow cytometry before and after subculture; BMA ability to counteract inflammation was also tested in an in vitro model of cartilage cell inflammation, evaluating the expression of MMP1, MMP3, TGFß and TIMP-1 by real-time PCR. Patients were also evaluated up to two years' follow-up by using: VAS for pain, IKDC-subjective and KOOS scores. RESULTS: The laboratory analysis showed that BMSCs accounted for 0.011% of BMA cells, similar to what had been expected in native bone marrow. The paracrine activity of BMA was able to reduce in vitro the catabolic response of human chondrocyte, as shown by the decrease in metalloproteases concentration and increase in anti-inflammatory mediators. Moreover, the clinical evaluation showed significant improvements in all scores adopted, with stable results up to two years. CONCLUSION: The present data showed the effectiveness of the study device to harvest pure bone marrow with minimal peripheral blood contamination. The relevant content of MSCs resulted in the ability to counteract the catabolic cascade through a paracrine action. The clinical outcomes in patients affected by unicompartmental knee OA were encouraging in terms of pain reduction and functional improvement up to mid-term evaluation.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Osteoartrite do Joelho , Adulto , Medula Óssea , Células da Medula Óssea , Humanos , Injeções Intra-Articulares , Transplante de Células-Tronco Mesenquimais/métodos , Pessoa de Meia-Idade , Osteoartrite do Joelho/terapia , Projetos Piloto , Resultado do Tratamento
10.
Cells ; 10(5)2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-34066077

RESUMO

The study of the miRNA cargo embedded in extracellular vesicles (EVs) released from adipose-derived mesenchymal stromal cells (ASC) preconditioned with IL-1ß, an inflammatory stimulus driving osteoarthritis (OA), along with EVs-cartilage dynamic interaction represent poorly explored fields and are the purpose of the present research. ASCs were isolated from subcutaneous adipose tissue and EVs collected by ultracentrifugation. Shuttled miRNAs were scored by high-throughput screening and analyzed through bioinformatics approach that predicted the potentially modulated OA-related pathways. Fluorescently labeled EVs incorporation into OA cartilage explants was followed in vitro by time-lapse coherent anti-Stokes Raman scattering; second harmonic generation and two-photon excited fluorescence. After IL-1ß preconditioning, 7 miRNA were up-regulated, 4 down-regulated, 37 activated and 17 silenced. Bioinformatics allowed to identify miRNAs and target genes mainly involved in Wnt, Notch, TGFß and Indian hedgehog (IHH) pathways, cartilage homeostasis, immune/inflammatory responses, cell senescence and autophagy. As well, ASC-EVs steadily diffuse in cartilage cells and matrix, reaching a plateau 16 h after administration. Overall, ASCs preconditioned with IL-1ß allows secretion of EVs embedded with a chondro-protective miRNA cargo, able to fast penetrate in collagen-rich areas of cartilage with tissue saturation in a day. Further functional studies exploring the EVs dose-effects are needed to achieve clinical relevance.


Assuntos
Tecido Adiposo/metabolismo , Cartilagem Articular/metabolismo , Vesículas Extracelulares/metabolismo , Interleucina-1beta/metabolismo , Células-Tronco Mesenquimais/citologia , Adulto , Condrócitos/metabolismo , Colágeno/química , Biologia Computacional , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Inflamação , Cinética , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Osteoartrite/metabolismo , Espectrometria de Fluorescência , Análise Espectral Raman
11.
Cells ; 10(5)2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33946524

RESUMO

Intra-articular administration of adipose-derived mesenchymal stem cells (ASCs), either in vitro expanded or within adipose tissue-based products obtained at point-of-care, has gained popularity as innovative regenerative medicine approach for osteoarthritis (OA) treatment. ASCs can stimulate tissue repair and immunomodulation through paracrine factors, both soluble and extracellular vesicles (EV) embedded, collectively defining the secretome. Interaction with the degenerative/inflamed environment is a crucial factor in understanding the finely tuned molecular message but, to date, the majority of reports have described ASC-secretome features in resting conditions or under chemical stimuli far from the in vivo environment of degenerated OA joints. In this report, the secretory profile of ASCs treated with native synovial fluid from OA patients was evaluated, sifting 200 soluble factors and 754 EV-embedded miRNAs. Fifty-eight factors and 223 EV-miRNAs were identified, and discussed in the frame of cartilage and immune cell homeostasis. Bioinformatics gave a molecular basis for M2 macrophage polarization, T cell proliferation inhibition and T reg expansion enhancement, as well as cartilage protection, further confirmed in an in vitro model of OA chondrocytes. Moreover, a strong influence on immune cell chemotaxis emerged. In conclusion, obtained molecular data support the regenerative and immunomodulatory properties of ASCs when interacting with osteoarthritic joint environment.


Assuntos
Cartilagem/imunologia , Vesículas Extracelulares/metabolismo , Fatores Imunológicos/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Osteoartrite/terapia , Tecido Adiposo/citologia , Idoso , Idoso de 80 Anos ou mais , Cartilagem/fisiologia , Células Cultivadas , Citocinas/metabolismo , Vesículas Extracelulares/transplante , Humanos , Macrófagos/imunologia , Transplante de Células-Tronco Mesenquimais/métodos , Pessoa de Meia-Idade , Regeneração , Líquido Sinovial/metabolismo , Linfócitos T/imunologia
12.
Stem Cells Transl Med ; 10(7): 1044-1062, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33656805

RESUMO

Human amniotic membrane-derived mesenchymal stromal cells (hAMSCs) are easily obtained in large quantities and free from ethical concerns. Promising therapeutic results for both hAMSCs and their secreted factors (secretome) were described by several in vitro and preclinical studies, often for treatment of orthopedic disorders such as osteoarthritis (OA) and tendinopathy. For clinical translation of the hAMSC secretome as cell-free therapy, a detailed characterization of hAMSC-secreted factors is mandatory. Herein, we tested the presence of 200 secreted factors and 754 miRNAs in extracellular vesicles (EVs). Thirty-seven cytokines/chemokines were identified at varying abundance, some of which involved in both chemotaxis and homeostasis of inflammatory cells and in positive remodeling of extracellular matrix, often damaged in tendinopathy and OA. We also found 336 EV-miRNAs, 51 of which accounted for more than 95% of the genetic message. A focused analysis based on miRNAs related to OA and tendinopathy showed that most abundant EV-miRNAs are teno- and chondro-protective, able to induce M2 macrophage polarization, inhibit inflammatory T cells, and promote Treg. Functional analysis on IL-1ß treated tenocytes and chondrocytes resulted in downregulation of inflammation-associated genes. Overall, presence of key regulatory molecules and miRNAs explain the promising therapeutic results of hAMSCs and their secretome for treatment of musculoskeletal conditions and are a groundwork for similar studies in other pathologies. Furthermore, identified molecules will pave the way for future studies aimed at more sharply predicting disease-targeted clinical efficacy, as well as setting up potency and release assays to fingerprint clinical-grade batches of whole secretome or purified components.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , MicroRNAs , Osteoartrite , Secretoma , Tendinopatia , Âmnio/citologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Matriz Extracelular , Vesículas Extracelulares/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Sistema Musculoesquelético , Osteoartrite/terapia , Regeneração , Tendinopatia/terapia
13.
Cartilage ; 13(1_suppl): 1749S-1760S, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-32508140

RESUMO

OBJECTIVE: The purpose of this systematic review and meta-analysis was to evaluate the effect of electromagnetic field treatment on the symptoms of knee osteoarthritis (OA). In addition, the influence of the type of control group and other covariates have been investigated to identify the sources of heterogeneity in the results of the available clinical trials. METHODS: Randomized controlled trials reporting pulsed electromagnetic field-based therapies for the treatment of knee OA have been included. Main outcomes were self-reported pain and activity scores collected by Visual Analogue Scale (VAS) and/or Western Ontario McMaster Universities Osteoarthritis Index (WOMAC) at short term after treatment. RESULTS: Thirteen studies comprising 914 unique patients were included in the analysis. Overall reduction in pain score was observed after treatment (standardized mean difference -0.4059, P = 0.0091), while improvement in the activity score was not significant (standardized mean difference -0.4452, P = 0.0859). Type of control (i.e., placebo or alternative therapies) and time of follow-up resulted as the two major elements influencing the outcomes. Indeed, the restriction of the analysis to placebo-controlled trials demonstrated higher standardized mean differences between treatment and control groups, with lower P value for pain, while statistical significance became evident also for the activity score. On the contrary, no differences were observed pooling only studies comparing pulsed electromagnetic or magnetic fields to alternative treatments. In addition, longer follow-up correlated with lower differences between treated and control patients. CONCLUSIONS: Pulsed electromagnetic field therapy effectively relieves knee OA symptoms at short term, but it is not superior to other conservative therapies such as physiotherapy.


Assuntos
Osteoartrite do Joelho , Humanos , Campos Magnéticos , Osteoartrite do Joelho/tratamento farmacológico , Osteoartrite do Joelho/terapia , Dor , Medição da Dor , Modalidades de Fisioterapia
14.
Connect Tissue Res ; 62(5): 570-579, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-32921180

RESUMO

AIM OF THE STUDY: Tendons are exposed to mechanical stress constantly during movements and thus they are frequently subjected to injuries. Rotator cuff tears are common musculoskeletal disorders, mainly involving the supraspinatus tendon. The characterization of the tenocytes derived from this tendon and the comparison to cells isolated from the long head of the biceps tendon obtained from donors affected by rotator cuff disease may improve the knowledge of the cellular mechanisms involved in the initiation and progression of the pathology. Thus, the aim of the present study was to characterize and compare donor-matched human tendon cells (TCs) isolated from the long head of the biceps (LHB-TCs) and the supraspinatus tendons (SSP-TCs) of patients affected by rotator cuff tears. METHODS: donor-matched LHB-TCs and SSP-TCs were isolated and cultured up to passage 3. Phenotypic appearance, metabolic activity, DNA content, production of soluble mediators (IL-1Ra, IL-1ß, IL-6, and VEGF) and gene expression of tendon markers (SCX, COL1A1, COL3A1), inflammatory (PTGS2), and catabolic enzymes (MMP-1, MMP-3) were evaluated. RESULTS: LHB-TCs showed an elongated fibroblast-like shape, while SSP-TCs appeared irregular with jagged membrane. SSP-TCs gene expression revealed an augmented production of PTGS2, a marker of inflammation, whereas they produced a reduced amount of IL-6, in respect to LHB-TCs. CONCLUSION: SSP-TCs showed higher cellular stress and expression of inflammatory markers with respect to donor-matched LHB-TCs, suggesting that addressing the physio-pathological state of supraspinatus tendon cells during treatment of rotator cuff tears could favor tissue healing and possibly prevent relapses.


Assuntos
Lesões do Manguito Rotador , Manguito Rotador , Biomarcadores , Ciclo-Oxigenase 2 , Humanos , Interleucina-6 , Tendões
15.
J Clin Med ; 11(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35011909

RESUMO

BACKGROUND: The coronavirus disease 2019 (COVID-19) pandemic outbreak has posed new problems in the context of patients suffering from other diseases. In particular, musculoskeletal sequelae related to the state of debilitation associated with COVID-19 are important to consider in elderly patients undergoing surgery after lower limbs fracture, especially in the post-operative period. The objective of this study was to evaluate whether COVID-19 influenced biochemical parameter, recovery and mortality of surgically treated patients suffering from lower extremity fractures. METHODS: Laboratory and clinical data of 30 patients were extrapolated and analyzed in the pre-operative and post-operative periods. Among these patients, 13 had COVID-19 infection (COVID-19 +), whereas 17 had no signs of COVID-19 infections (COVID-19 -). Long-term clinical and functional outcomes were also analyzed. RESULTS: Lower calcium, slightly higher values of CRP and much higher values of CPK and AST were observed pre-operatively in COVID-19 + patients, who also showed higher prevalence of long-term sequelae than COVID-19 - patients. CONCLUSIONS: COVID-19 affects long-term outcome of elderly patients with lower limb fractures in a multifactorial way. First, the virus directly damages the muscle tissue. Secondly, the lung function impairment worsens the overall performance, making rehabilitation more challenging.

16.
PLoS One ; 15(9): e0239807, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32998161

RESUMO

Tendinopathy is a degenerative disease in which inflammatory mediators have been found to be sometimes present. The interaction between inflammation and matrix remodeling in human tendon cells (TCs) is supported by the secretion of cytokines such as IL-1ß, IL-6 and IL-33. In this context, it has been demonstrated that pulsed electromagnetic fields (PEMFs) were able to reduce inflammation and promote tendon marker synthesis. The aim of this study was to evaluate the anabolic and anti-inflammatory PEMF-mediated response on TCs in an in vitro model of inflammation. Moreover, since PEMFs enhance the anti-inflammatory efficacy of adenosine through the adenosine receptors (ARs), the study also focused on the role of A2AARs. Human TCs were exposed to PEMFs for 48 hours. After stimulation, A2AAR saturation binding experiments were performed. Along with 48 hours PEMF stimulation, TCs were treated with IL-1ß and A2AAR agonist CGS-21680. IL-1Ra, IL-6, IL-8, IL-10, IL-33, VEGF, TGF-ß1, PGE2 release and SCX, COL1A1, COL3A1, ADORA2A expression were quantified. PEMFs exerted A2AAR modulation on TCs and promoted COL3A1 upregulation and IL-33 secretion. In presence of IL-1ß, TCs showed an upregulation of ADORA2A, SCX and COL3A1 expression and an increase of IL-6, IL-8, PGE2 and VEGF secretion. After PEMF and IL-1ß exposure, IL-33 was upregulated, whereas IL-6, PGE2 and ADORA2A were downregulated. These findings demonstrated that A2AARs have a role in the promotion of the TC anabolic/reparative response to PEMFs and to IL-1ß.


Assuntos
Regulação para Baixo/efeitos da radiação , Campos Eletromagnéticos , Receptor A2A de Adenosina/metabolismo , Tendões/metabolismo , Regulação para Cima/efeitos da radiação , Agonistas do Receptor A2 de Adenosina/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Cultivadas , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Humanos , Interleucina-1beta/farmacologia , Interleucina-33/metabolismo , Interleucina-6/metabolismo , Receptor A2A de Adenosina/química , Receptor A2A de Adenosina/genética , Tendões/citologia , Tendões/efeitos da radiação , Regulação para Cima/efeitos dos fármacos
17.
Stem Cell Res Ther ; 11(1): 165, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32345351

RESUMO

BACKGROUND: Mesenchymal stromal cell (MSC)-enriched products showed positive clinical outcomes in regenerative medicine, where tissue restoration and inflammation control are needed. GMP-expanded MSCs displayed an even higher potential due to exclusive secretion of therapeutic factors, both free and conveyed within extracellular vesicles (EVs), collectively termed secretome. Moreover, priming with biochemical cues may influence the portfolio and biological activities of MSC-derived factors. For these reasons, the use of naive or primed secretome gained attention as a cell-free therapeutic option. Albeit, at present, a homogenous and comprehensive secretome fingerprint is still missing. Therefore, the aim of this work was to deeply characterize adipose-derived MSC (ASC)-secreted factors and EV-miRNAs, and their modulation after IFNγ preconditioning. The crucial influence of the target pathology or cell type was also scored in osteoarthritis to evaluate disease-driven potency. METHODS: ASCs were isolated from four donors and cultured with and without IFNγ. Two-hundred secreted factors were assayed by ELISA. ASC-EVs were isolated by ultracentrifugation and validated by flow cytometry, transmission electron microscopy, and nanoparticle tracking analysis. miRNome was deciphered by high-throughput screening. Bioinformatics was used to predict the modulatory effect of secreted molecules on pathologic cartilage and synovial macrophages based on public datasets. Models of inflammation for both macrophages and chondrocytes were used to test by flow cytometry the secretome anti-inflammatory potency. RESULTS: Data showed that more than 60 cytokines/chemokines could be identified at varying levels of intensity in all samples. The vast majority of factors are involved in extracellular matrix remodeling, and chemotaxis or motility of inflammatory cells. IFNγ is able to further increase the capacity of the secretome to stimulate cell migration signals. Moreover, more than 240 miRNAs were found in ASC-EVs. Sixty miRNAs accounted for > 95% of the genetic message that resulted to be chondro-protective and M2 macrophage polarizing. Inflammation tipped the balance towards a more pronounced tissue regenerative and anti-inflammatory phenotype. In silico data were confirmed on inflamed macrophages and chondrocytes, with secretome being able to increase M2 phenotype marker CD163 and reduce the chondrocyte inflammation marker VCAM1, respectively. IFNγ priming further enhanced secretome anti-inflammatory potency. CONCLUSIONS: Given the portfolio of soluble factors and EV-miRNAs, ASC secretome showed a marked capacity to stimulate cell motility and modulate inflammatory and degenerative processes. Preconditioning is able to increase this ability, suggesting inflammatory priming as an effective strategy to obtain a more potent clinical product which use should always be driven by the molecular mark of the target pathology.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , MicroRNAs , Osteoartrite , Humanos , Medicina Regenerativa
18.
Int J Mol Sci ; 21(5)2020 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-32111031

RESUMO

Mesenchymal stem cells (MSCs) derived from adipose tissue and used either as expanded cells or minimally manipulated cell preparations showed positive clinical outcomes in regenerative medicine approaches based on tissue restoration and inflammation control, like in osteoarthritis (OA). Recently, MSCs' healing capacity has been ascribed to the large array of soluble factors, including soluble cytokines/chemokines and miRNAs conveyed within extracellular vesicles (EVs). Therefore, in this study, 200 secreted cytokines, chemokines and growth factors via ELISA, together with EV-embedded miRNAs via high-throughput techniques, were scored in adipose-derived MSCs (ASCs) cultivated under inflammatory conditions, mimicking OA synovial fluid. Both factors (through most abundantly expressed TIMP1, TIMP2, PLG and CTSS) and miRNAs (miR-24-3p, miR-222-3p and miR-193b-3p) suggested a strong capacity for ASCs to reduce matrix degradation activities, as those activated in OA cartilage, and switch synovial macrophages, often characterized by an M1 inflammatory polarization, towards an M2 phenotype. Moreover, the crucial importance of selecting the target tissue is discussed, showing how a focused search may greatly improve potency prediction and explain clinical outcomes. In conclusion, herein presented data shed light about the way ASCs regulate cell homeostasis and regenerative pathways in an OA-resembling environment, therefore suggesting a rationale for the use of MSC-enriched clinical products, such as stromal vascular fraction and microfragmented adipose tissue, in joint pathologies.


Assuntos
Tecido Adiposo/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Osteoartrite do Joelho/terapia , Cicatrização/fisiologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Homeostase , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Macrófagos/metabolismo , MicroRNAs/genética , Líquido Sinovial/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Cicatrização/genética
19.
Cytotherapy ; 21(12): 1179-1197, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31784241

RESUMO

Osteoarthritis (OA) is a debilitating, degenerative joint disease characterized by progressive destruction of articular cartilage. Given the poor repair capacity of articular cartilage and the associated local destructive immune/inflammatory responses involving all joint structures, OA frequently ends up as a "whole joint failure" requiring prosthetic replacement. Current pharmacological efforts, belatedly started, mainly aim at symptomatic pain relief, underscoring the need for novel therapeutic schemes designed to modify the course of the disease. Mesenchymal stem cell (MSC)-based therapy has gained significant interest, sparking the design of multiple trials proving safety while providing promising preliminary efficacy results. MSCs possess 'medicinal signaling cell' properties related to their immunomodulatory and anti-inflammatory effects, which induce the establishment of a pro-regenerative microenvironment at the injured tissue. Those trophic effects are paralleled by the long-established chondroprogenitor capacity that can be harnessed to ex vivo fabricate engineered constructs to repair damaged articular cartilage. The present review focuses on these two aspects of the use of MSCs for articular cartilage damage, namely, cell therapy and tissue engineering, providing information on their use criteria, advancements, challenges and strategies to overcome them.


Assuntos
Cartilagem Articular/fisiologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Osteoartrite/terapia , Engenharia Tecidual/tendências , Animais , Regeneração Óssea/fisiologia , Cartilagem Articular/fisiopatologia , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Osteoartrite/fisiopatologia , Regeneração/fisiologia , Medicina Regenerativa/métodos , Medicina Regenerativa/tendências , Engenharia Tecidual/métodos
20.
Sci Rep ; 9(1): 10864, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31350444

RESUMO

The infrapatellar fat pad (IFP) serves as a reservoir of Mesenchymal Stem Cells (MSC), and with adjacent synovium plays key roles in joint disease including the production of Substance P (SP) affecting local inflammatory responses and transmitting nociceptive signals. Here, we interrogate human IFP-derived MSC (IFP-MSC) reaction to inflammatory and pro-fibrotic environments (cell priming by TNFα/IFNγ and TNFα/IFNγ/CTGF exposure respectively), compared with bone marrow-derived MSC (BM-MSC). Naïve IFP-MSC exhibit increased clonogenicity and chondrogenic potential compared with BM-MSC. Primed cells experienced dramatic phenotypic changes, including a sharp increase in CD10, upregulation of key immunomodulatory transcripts, and secreted growth factors/cytokines affecting key pathways (IL-10, TNF-α, MAPK, Ras and PI3K-Akt). Naïve, and more so primed MSC (both) induced SP degradation in vitro, reproduced with their supernatants and abrogated with thiorphan, a CD10 inhibitor. These findings were reproduced in vivo in a rat model of acute synovitis, where transiently engrafted human IFP-MSC induced local SP reduction. Functionally, primed IFP-MSC demonstrated sustained antagonism of activated human peripheral blood mononuclear cells (PBMC) proliferation, significantly outperforming a declining dose-dependent effect with naïve cohorts. Collectively, our in vitro and in vivo data supports cell priming as a way to enhance the immunoregulatory properties of IFP-MSC, which selectively engraft in areas of active synovitis/IFP fibrosis inducing SP degradation, resulting in a cell-based product alternative to BM-MSC to potentially treat degenerative/inflammatory joint diseases.


Assuntos
Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Neprilisina/metabolismo , Fenótipo , Proteólise/efeitos dos fármacos , Substância P/metabolismo , Sinovite/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/patologia , Adulto , Animais , Células da Medula Óssea/metabolismo , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Fibrose , Voluntários Saudáveis , Humanos , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interferon gama/farmacologia , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Sinovite/induzido quimicamente , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA