Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Ann Med ; 56(1): 2403721, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39291917

RESUMO

OBJECTIVES: The relationship between serum calcium and occurrence of MHO (metabolically healthy obesity) and MUNO (metabolically unhealthy non-obesity) remains unclear, and distinguishing these two phenotypes is difficult within primary healthcare units. This study explores that relationship. METHODS: This survey included 28590 adults from the National Health and Nutrition Examination Survey (NHANES) 2001-2018. Obesity phenotypes were categorized based on BMI and presence or absence of metabolic syndrome components. Weighted multivariate logistic regression analyses were used to assess the association between serum calcium levels and the obesity phenotype. Restricted cubic spline analysis characterized dose-response relationships, and stratified analyses explored these relationships across sociodemographic and lifestyle factors. RESULTS: The overall prevalence of MHO and MUNO were 2.6% and 46.6%, respectively. After adjusting for covariates, serum calcium exhibited a negative association with MHO [OR (95%): 0.49 (0.36,0.67), p < 0.001], while exhibiting a positive association with MUNO [OR (95%): 1.48 (1.26,1.84), p < 0.001]. Additionally, we found a non-linear association between serum calcium levels and the incidences of MHO and MUNO. Stratified analyses demonstrated a strong negative correlation between serum calcium levels and MHO occurrence across various subgroups. There was no significant interaction between calcium and stratified variables except sex; the association between calcium and the occurrence of MHO was remarkable in female patients. Meanwhile, the predictive ability of serum calcium level for the occurrence of MUNO among all patients was consistent across various subgroups. There was a significant interaction between calcium level and stratified variables based on age, sex, race, and smoking status; the association was remarkable in older (≥ 40 years old), white, none or less smoking, and female patients. CONCLUSIONS: A significant correlation was identified between serum calcium levels and MHO or MUNO. The findings suggest that serum calcium levels may serve as an indicator for more accurate assessment and diagnosis of MUNO and MHO, especially among individuals with abdominal obesity.


Serum calcium levels exhibited an inverse relationship with metabolically healthy obesity (MHO) and a positive relationship with metabolically unhealthy non-obese (MUNO).A nonlinear association exists between serum calcium levels and the incidence of both MHO and MUNO.Serum calcium has the potential to enhance evaluation and screening for MUNO or MHO in the general US adult population.


Assuntos
Cálcio , Inquéritos Nutricionais , Obesidade Metabolicamente Benigna , Humanos , Feminino , Masculino , Estudos Transversais , Adulto , Pessoa de Meia-Idade , Inquéritos Nutricionais/estatística & dados numéricos , Cálcio/sangue , Estados Unidos/epidemiologia , Obesidade Metabolicamente Benigna/sangue , Obesidade Metabolicamente Benigna/epidemiologia , Síndrome Metabólica/sangue , Síndrome Metabólica/epidemiologia , Prevalência , Índice de Massa Corporal , Idoso , Obesidade/sangue , Obesidade/epidemiologia
2.
Front Endocrinol (Lausanne) ; 15: 1417485, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39345882

RESUMO

Objective: The exact relationship between the serum uric acid-to-HDL cholesterol ratio (UHR) and mortality rates remains enigmatic among American adults. This study aims to clarify the association between UHR and both all-cause and cardiovascular disease (CVD) mortality in US adults. Methods: This study enrolled 48054 patients from the National Health and Nutrition Examination Survey (NHANES). Mortality outcomes were determined by linking to National Death Index (NDI) records up to December 31,2019. Multivariate Cox proportional hazards models were constructed to analyze explore the associations between UHR and mortality. Dose-response relationships were explored using restricted cubic splines, and stratified analyses were conducted based on gender, age, race, education, PIR, smoking status, alcohol intake, physical activity, BMI, diabetes and hypertension. Results: During the follow-up period, the overall mortality for all-cause and CVD was 10.9% and 2.7%, respectively. The adjusted HRs in the highest quintile were 1.16 (95% CI: 1.05, 1.29) for all-cause mortality and 1.2 (95% CI: 1, 1.45) for CVD mortality. In diabetes, obese, and CVD subgroups, significantly elevated adjusted HRs were observed for both all-cause and CVD mortality. Specifically, diabetes patients had adjusted HRs of 1.32 (95% CI: 1.11, 1.57) and 1.38 (95% CI: 1.01, 1.90), obese individuals had HRs of 1.32 (95% CI: 1.10, 1.58) and 1.55 (95% CI: 1.06, 2.28), and CVD patients had HRs of 1.29 (95% CI: 1.10, 1.50) and 1.38 (95% CI: 1.06, 1.79), respectively. A non-linear relationship between UHR and mortality was identified, with critical thresholds of 12.4 for all-cause mortality and 10.7 for CVD mortality in the general population. Significant interactions were observed between UHR and stratified variables, including gender, BMI, education, smoking, alcohol use, and hypertension for all-cause mortality, while significant interactions were observed based on gender, smoking, and alcohol intake for CVD mortality. Comparable trends were also observed in patient with diabetes, obese and CVD. Conclusions: In this cohort study, we provide novel insights into the association between serum UHR concentrations and mortality in the general population. UHR is a strong predictor of all-cause and cardiovascular mortality in the general population.


Assuntos
Doenças Cardiovasculares , HDL-Colesterol , Inquéritos Nutricionais , Ácido Úrico , Humanos , Masculino , Doenças Cardiovasculares/mortalidade , Doenças Cardiovasculares/sangue , Feminino , Ácido Úrico/sangue , Pessoa de Meia-Idade , Estudos Transversais , HDL-Colesterol/sangue , Adulto , Idoso , Causas de Morte , Fatores de Risco , Biomarcadores/sangue , Estados Unidos/epidemiologia , Prognóstico
4.
Front Immunol ; 15: 1360237, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38576617

RESUMO

Comprising only 1-10% of the circulating T cell population, γδT cells play a pivotal role in cancer immunotherapy due to their unique amalgamation of innate and adaptive immune features. These cells can secrete cytokines, including interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), and can directly eliminate tumor cells through mechanisms like Fas/FasL and antibody-dependent cell-mediated cytotoxicity (ADCC). Unlike conventional αßT cells, γδT cells can target a wide variety of cancer cells independently of major histocompatibility complex (MHC) presentation and function as antigen-presenting cells (APCs). Their ability of recognizing antigens in a non-MHC restricted manner makes them an ideal candidate for allogeneic immunotherapy. Additionally, γδT cells exhibit specific tissue tropism, and rapid responsiveness upon reaching cellular targets, indicating a high level of cellular precision and adaptability. Despite these capabilities, the therapeutic potential of γδT cells has been hindered by some limitations, including their restricted abundance, unsatisfactory expansion, limited persistence, and complex biology and plasticity. To address these issues, gene-engineering strategies like the use of chimeric antigen receptor (CAR) T therapy, T cell receptor (TCR) gene transfer, and the combination with γδT cell engagers are being explored. This review will outline the progress in various engineering strategies, discuss their implications and challenges that lie ahead, and the future directions for engineered γδT cells in both monotherapy and combination immunotherapy.


Assuntos
Neoplasias , Receptores de Antígenos de Linfócitos T gama-delta , Receptores de Antígenos de Linfócitos T gama-delta/genética , Linfócitos T , Imunoterapia , Imunoterapia Adotiva , Engenharia Celular , Neoplasias/terapia
5.
Cancer Metab ; 11(1): 24, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38057929

RESUMO

BACKGROUND: The relationship between obesity and non-Hodgkin's lymphoma (NHL) was controversial, which may be due to the crudeness definition of obesity based on body mass index (BMI). As obesity and metabolic abnormalities often coexist, we aimed to explore whether the classification of obesity based on metabolic status can help to evaluate the real impact of obesity on the readmission of NHL. METHODS: In this retrospective cohort study, utilizing the 2018 Nationwide Readmissions Database, we identified NHL-related index hospitalizations and followed them for non-elective readmission. The patients with NHL were classified as metabolically healthy non-obese (MHNO) and obese (MHO) and metabolically unhealthy non-obese (MUNO) and obese (MUO). Readmission rates for each phenotype were calculated at 30-day intervals. Multiple COX regression was used to analyze the association of metabolic-defined obesity with 30-day, 90-day, and 180-day readmission rates in patients with NHL. RESULTS: There were 22,086 index hospitalizations with NHL included. In the multivariate COX regression, MUNO was associated with increased 30-day (HR = 1.113, 95% CI 1.036-1.195), 90-day (HR = 1.148, 95% CI 1.087-1.213), and 180-day readmission rates (HR = 1.132, 95% CI 1.077-1.189), and MUO was associated with increased 30-day (HR=1.219, 95% CI: 1.081-1.374), 90-day (HR = 1.228, 95% CI 1.118-1.348), and 180-day readmission rates (HR = 1.223, 95% CI 1.124-1.33), while MHO had no associations with readmission rates. CONCLUSIONS: The presence of metabolic abnormalities with or without obesity increased the risk of non-selective readmission in patients with NHL. However, obesity alone had no associations with the risk of non-selective readmission, suggesting that interventions for metabolic abnormalities may be more important in reducing readmissions of NHL patients.

6.
Front Genet ; 14: 1004481, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37007970

RESUMO

Hepatocellular carcinoma (HCC) is the most common type of liver cancer with a high morbidity and fatality rate. Traditional diagnostic methods for HCC are primarily based on clinical presentation, imaging features, and histopathology. With the rapid development of artificial intelligence (AI), which is increasingly used in the diagnosis, treatment, and prognosis prediction of HCC, an automated approach to HCC status classification is promising. AI integrates labeled clinical data, trains on new data of the same type, and performs interpretation tasks. Several studies have shown that AI techniques can help clinicians and radiologists be more efficient and reduce the misdiagnosis rate. However, the coverage of AI technologies leads to difficulty in which the type of AI technology is preferred to choose for a given problem and situation. Solving this concern, it can significantly reduce the time required to determine the required healthcare approach and provide more precise and personalized solutions for different problems. In our review of research work, we summarize existing research works, compare and classify the main results of these according to the specified data, information, knowledge, wisdom (DIKW) framework.

7.
FEBS Open Bio ; 13(1): 89-101, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36333974

RESUMO

Type 2 diabetes (T2DM) is characterized by insulin secretion deficiencies and systemic insulin resistance (IR) in adipose tissue, skeletal muscle, and the liver. Although the mechanism of T2DM is not yet fully known, inflammation and insulin resistance play a central role in the pathogenesis of T2DM. G protein-coupled receptors (GPCRs) are involved in endocrine and metabolic processes as well as many other physiological processes. GPR50 (G protein-coupled receptor 50) is an orphan GPCR that shares the highest sequence homology with melatonin receptors. The aim of this study was to investigate the effect of GPR50 on inflammation and insulin resistance in 3T3-L1 preadipocytes. GPR50 expression was observed to be significantly increased in the adipose tissue of obese T2DM mice, while GPR50 deficiency increased inflammation in 3T3-L1 cells and induced the phosphorylation of AKT and insulin receptor substrate (IRS) 1. Furthermore, GPR50 knockout in the 3T3-L1 cell line suppressed PPAR-γ expression. These data suggest that GPR50 can attenuate inflammatory levels and regulate insulin signaling in adipocytes. Furthermore, the effects are mediated through the regulation of the IRS1/AKT signaling pathway and PPAR-γ expression.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Animais , Camundongos , Células 3T3-L1 , Proteínas de Transporte/metabolismo , Inflamação/metabolismo , Insulina/metabolismo , PPAR gama/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
8.
J Orthop Translat ; 36: 64-74, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35979174

RESUMO

Background: A critical size bone defect is a clinical scenario in which bone is lost or excised due to trauma, infection, tumor, or other causes, and cannot completely heal spontaneously. The most common treatment for this condition is autologous bone grafting to the defect site. However, autologous bone graft is often insufficient in quantity or quality for transplantation to these large defects. Recently, tissue engineering methods using mesenchymal stem cells (MSCs) have been proposed as an alternative treatment. However, the underlying biological principles and optimal techniques for tissue regeneration of bone using stem cell therapy have not been completely elucidated. Methods: In this study, we compare the early cellular dynamics of healing between bone graft transplantation and MSC therapy in a murine chronic femoral critical-size bone defect. We employ high-dimensional mass cytometry to provide a comprehensive view of the differences in cell composition, stem cell functionality, and immunomodulatory activity between these two treatment methods one week after transplantation. Results: We reveal distinct cell compositions among tissues from bone defect sites compared with original bone graft, show active recruitment of MSCs to the bone defect sites, and demonstrate the phenotypic diversity of macrophages and T cells in each group that may affect the clinical outcome. Conclusion: Our results provide critical data and future directions on the use of MSCs for treating critical size defects to regenerate bone.Translational Potential of this article: This study showed systematic comparisons of the cellular and immunomodulatory profiles among different interventions to improve the healing of the critical-size bone defect. The results provided potential strategies for designing robust therapeutic interventions for the unmet clinical need of treating critical-size bone defects.

9.
Immun Ageing ; 19(1): 14, 2022 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-35279175

RESUMO

BACKGROUND: Despite the high incidence of fractures and pseudoarthrosis in the aged population, a potential role for the use of mesenchymal stem cells (MSCs) in the treatment of bone defects in elderly patients has not been elucidated. Inflammation and the innate immune system, including macrophages, play crucial roles in the differentiation and activation of MSCs. We have developed lentivirus-transduced interleukin 4 (IL4) over-expressing MSCs (IL4-MSCs) to polarize macrophages to an M2 phenotype to promote bone healing in an established young murine critical size bone defect model. In the current study, we explore the potential of IL4-MSCs in aged mice. METHODS: A 2 mm femoral diaphyseal bone defect was created and fixed with an external fixation device in 15- to 17-month-old male and female BALB/c mice. Microribbon (µRB) scaffolds (Sc) with or without encapsulation of MSCs were implanted in the defect sites. Accordingly, the mice were divided into three treatment groups: Sc-only, Sc + MSCs, and Sc + IL4-MSCs. Mice were euthanized six weeks after the surgery; subsequently, MicroCT (µCT), histochemical and immunohistochemical analyses were performed. RESULTS: µCT analysis revealed that bone formation was markedly enhanced in the IL4-MSC group. Compared with the Sc-only, the amount of new bone increased in the Sc + MSCs and Sc + IL4-MSC groups. However, no bridging of bone was observed in all groups. H&E staining showed fibrous tissue within the defect in all groups. Alkaline phosphatase (ALP) staining was increased in the Sc + IL4-MSC group. The Sc + IL4-MSCs group showed a decrease in the number of M1 macrophages and an increase in the number of M2 macrophages, with a significant increase in the M2/M1 ratio. DISCUSSION: IL4 promotes macrophage polarization to an M2 phenotype, facilitating osteogenesis and vasculogenesis. The addition of IL4-MSCs in the µRB scaffold polarized macrophages to an M2 phenotype and increased bone formation; however, complete bone bridging was not observed in any specimens. These results suggest that IL4-MSCs are insufficient to heal a critical size bone defect in aged mice, as opposed to younger animals. Additional therapeutic strategies are needed in this challenging clinical scenario.

10.
ACS Nano ; 16(1): 1421-1435, 2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-34962119

RESUMO

Combinatorial cancer therapies based on nanomedicine have emerged as a promising strategy to achieve potentiated treatment efficiency. Herein, cisplatin (CDDP) prodrug (Pt-CD) and a mitochondria-targeted near-infrared (NIR) photosensitizer IR780 were combined to construct a multifunctional nanomedicine IR780@Pt NPs through a supramolecular self-assembly strategy. Targeted mitochondrial dysfunction of cancer cells was sufficiently induced under NIR laser irradiation through both photothermal and photodynamic effects, inhibiting the overactive mitochondrial energy pathways of cancer cells. The mitochondrial dysfunction significantly attenuated the crosstalk between mitochondria and nucleus via the cellular ATP energy chain, leading to obvious down-regulation of the key proteins of the nucleotide excision repair (NER) pathway. Thereby, the chemotherapeutic effect of CDDP could be significantly potentiated because of reduced DNA lesion repair capacity by ERCC1-XPF nuclease system. Moreover, IR780@Pt NPs exhibited excellent NIR fluorescence and photoacoustic (PA) imaging capacity for in vivo imaging-guided NIR laser treatment. Ultimately, the IR780@Pt NPs mediated combinatorial chemophototherapy achieved potentiated anticancer efficacy against cancer cells in vitro and tumor inhibition performance in vivo. Overall, this study highlighted the significance of nanomedicine mediated targeted induction of mitochondrial dysfunction to potentiate chemotherapy for efficient combinatorial cancer therapy.


Assuntos
Nanopartículas , Fotoquimioterapia , Cisplatino/farmacologia , Fotoquimioterapia/métodos , Nanomedicina , Raios Infravermelhos , Nanopartículas/uso terapêutico , Nanomedicina Teranóstica/métodos , Mitocôndrias , Fototerapia/métodos , Linhagem Celular Tumoral
11.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-34884641

RESUMO

Fibroblasts in the synovial membrane secrete molecules essential to forming the extracellular matrix (ECM) and supporting joint homeostasis. While evidence suggests that fibroblasts contribute to the response to joint injury, the outcomes appear to be patient-specific and dependent on interactions between resident immune cells, particularly macrophages (Mφs). On the other hand, the response of Mφs to injury depends on their functional phenotype. The goal of these studies was to further explore these issues in an in vitro 3D microtissue model that simulates a pathophysiological disease-specific microenvironment. Two sources of fibroblasts were used to assess patient-specific influences: mesenchymal stem cell (MSC)- and induced pluripotent stem cell (iPSC)-derived fibroblasts. These were co-cultured with either M1 or M2 Mφs, and the cultures were challenged with polyethylene particles coated with lipopolysaccharide (cPE) to model wear debris generated from total joint arthroplasties. Our results indicated that the fibroblast response to cPE was dependent on the source of the fibroblasts and the presence of M1 or M2 Mφs: the fibroblast response as measured by gene expression changes was amplified by the presence of M2 Mφs. These results demonstrate that the immune system modulates the function of fibroblasts; furthermore, different sources of differentiated fibroblasts may lead to divergent results. Overall, our research suggests that M2 Mφs may be a critical target for the clinical treatment of cPE induced fibrosis.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Polietileno/farmacologia , Artroplastia/métodos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Matriz Extracelular , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibrose/tratamento farmacológico , Fibrose/imunologia , Fibrose/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Células-Tronco Mesenquimais/imunologia
12.
Front Bioeng Biotechnol ; 9: 755964, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34738008

RESUMO

Mesenchymal stem cell (MSC)-based therapy and novel biomaterials are promising strategies for healing of long bone critical size defects. Interleukin-4 (IL-4) over-expressing MSCs within a gelatin microribbon (µRB) scaffold was previously shown to enhance the bridging of bone within a critical size femoral bone defect in male Balb/c mice. Whether sex differences affect the healing of this bone defect in conjunction with different treatments is unknown. In this study, we generated 2-mm critical-sized femoral diaphyseal bone defects in 10-12-week-old female and male Balb/c mice. Scaffolds without cells and with unmodified MSCs were implanted immediately after the primary surgery that created the bone defect; scaffolds with IL-4 over-expressing MSCs were implanted 3 days after the primary surgery, to avoid the adverse effects of IL-4 on the initial inflammatory phase of fracture healing. Mice were euthanized 6 weeks after the primary surgery and femurs were collected. MicroCT (µCT), histochemical and immunohistochemical analyses were subsequently performed of the defect site. µRB scaffolds with IL-4 over-expressing MSCs enhanced bone healing in both female and male mice. Male mice showed higher measures of bone bridging and increased alkaline phosphatase (ALP) positive areas, total macrophages and M2 macrophages compared with female mice after receiving scaffolds with IL-4 over-expressing MSCs. Female mice showed higher Tartrate-Resistant Acid Phosphatase (TRAP) positive osteoclast numbers compared with male mice. These results demonstrated that sex differences should be considered during the application of MSC-based studies of bone healing.

13.
Front Cell Dev Biol ; 9: 757830, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34722543

RESUMO

Wear particles from total joint arthroplasties (TJAs) induce chronic inflammation, macrophage infiltration and lead to bone loss by promoting bone destruction and inhibiting bone formation. Inhibition of particle-associated chronic inflammation and the associated bone loss is critical to the success and survivorship of TJAs. The purpose of this study is to test the hypothesis that polyethylene particle induced chronic inflammatory bone loss could be suppressed by local injection of NF-κB sensing Interleukin-4 (IL-4) over-expressing MSCs using the murine continuous polyethylene particle infusion model. The animal model was generated with continuous infusion of polyethylene particles into the intramedullary space of the femur for 6 weeks. Cells were locally injected into the intramedullary space 3 weeks after the primary surgery. Femurs were collected 6 weeks after the primary surgery. Micro-computational tomography (µCT), histochemical and immunohistochemical analyses were performed. Particle-infusion resulted in a prolonged pro-inflammatory M1 macrophage dominated phenotype and a decrease of the anti-inflammatory M2 macrophage phenotype, an increase in TRAP positive osteoclasts, and lower alkaline phosphatase staining area and bone mineral density, indicating chronic particle-associated inflammatory bone loss. Local injection of MSCs or NF-κB sensing IL-4 over-expressing MSCs reversed the particle-associated chronic inflammatory bone loss and facilitated bone healing. These results demonstrated that local inflammatory bone loss can be effectively modulated via MSC-based treatments, which could be an efficacious therapeutic strategy for periprosthetic osteolysis.

14.
Stem Cell Res Ther ; 12(1): 503, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34526115

RESUMO

BACKGROUND: Approximately one third of patients undergoing core decompression (CD) for early-stage osteonecrosis of the femoral head (ONFH) experience progression of the disease, and subsequently require total hip arthroplasty (THA). Thus, identifying adjunctive treatments to optimize bone regeneration during CD is an unmet clinical need. Platelet-derived growth factor (PDGF)-BB plays a central role in cell growth and differentiation. The aim of this study was to characterize mesenchymal stromal cells (MSCs) that were genetically modified to overexpress PDGF-BB (PDGF-BB-MSCs) in vitro and evaluate their therapeutic effect when injected into the bone tunnel at the time of CD in an in vivo rabbit model of steroid-associated ONFH. METHODS: In vitro studies: Rabbit MSCs were transduced with a lentivirus vector carrying the human PDGF-BB gene under the control of either the cytomegalovirus (CMV) or phosphoglycerate (PGK) promoter. The proliferative rate, PDGF-BB expression level, and osteogenic differentiation capacity of unmodified MSCs, CMV-PDGF-BB-MSCs, and PGK-PDGF-BB-MSCs were assessed. In vivo studies: Twenty-four male New Zealand white rabbits received an intramuscular (IM) injection of methylprednisolone 20 mg/kg. Four weeks later, the rabbits were divided into four groups: the CD group, the hydrogel [HG, (a collagen-alginate mixture)] group, the MSC group, and the PGK-PDGF-BB-MSC group. Eight weeks later, the rabbits were sacrificed, their femurs were harvested, and microCT, mechanical testing, and histological analyses were performed. RESULTS: In vitro studies: PGK-PDGF-BB-MSCs proliferated more rapidly than unmodified MSCs (P < 0.001) and CMV-PDGF-BB-MSCs (P < 0.05) at days 3 and 7. CMV-PDGF-BB-MSCs demonstrated greater PDGF-BB expression than PGK-PDGF-BB-MSCs (P < 0.01). However, PGK-PDGF-BB-MSCs exhibited greater alkaline phosphatase staining at 14 days (P < 0.01), and osteogenic differentiation at 28 days (P = 0.07) than CMV-PDGF-BB-MSCs. In vivo: The PGK-PDGF-BB-MSC group had a trend towards greater bone mineral density (BMD) than the CD group (P = 0.074). The PGK-PDGF-BB-MSC group demonstrated significantly lower numbers of empty lacunae (P < 0.001), greater osteoclast density (P < 0.01), and greater angiogenesis (P < 0.01) than the other treatment groups. CONCLUSION: The use of PGK-PDGF-BB-MSCs as an adjunctive treatment with CD may reduce progression of osteonecrosis and enhance bone regeneration and angiogenesis in the treatment of early-stage ONFH.


Assuntos
Necrose da Cabeça do Fêmur , Células-Tronco Mesenquimais , Osteonecrose , Animais , Becaplermina , Descompressão , Cabeça do Fêmur , Necrose da Cabeça do Fêmur/induzido quimicamente , Necrose da Cabeça do Fêmur/genética , Necrose da Cabeça do Fêmur/terapia , Humanos , Masculino , Osteogênese , Coelhos , Esteroides
15.
Biomaterials ; 275: 120950, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34119886

RESUMO

Clinical treatment of Osteosarcoma (OS) encounters great challenges of postsurgical tumor recurrence and extensive bone defect. To address these issues, innovative multifunctional PLGA/Mg porous scaffolds were designed for comprehensive postsurgical management of OS. The PLGA/Mg composite scaffolds exhibited several unique features: (1) The multiple functions of Mg particles were explored for the first time to fulfill the requirement for postsurgical management of OS. The intact Mg particles exhibits excellent photothermal effect for tumor eradication, and the released Mg ions could subsequently promote bone regeneration, thus endowing the PLGA/Mg scaffolds dual functions of suppressing OS recurrence and repairing bone defect in a sequential way; (2) A low temperature rapid prototyping (LT-RP) 3D-printing technology was used to fabricate the scaffolds with biomimetic hierarchical porous structures, which could structurally promote bone regeneration; (3) The PLGA/Mg scaffolds have excellent biodegradability and biocompatibility, exhibiting great promise for clinical translation. Finally, the PLGA/Mg scaffolds achieved complete suppression of tumor recurrence in the presence of near-infrared laser irradiation, as well as efficient bone defect repair in vivo. Activation of the AKT and ß-catenin pathways of osteoblast cells by PLGA/Mg scaffolds was identified, which might be the modulators to accelerate the ossification. The innovative PLGA/Mg scaffolds demonstrated excellent capabilities in postsurgical OS recurrence suppression and bone regeneration, providing a promising clinical strategy for comprehensive postsurgical management of OS.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Neoplasias Ósseas/cirurgia , Regeneração Óssea , Humanos , Magnésio , Recidiva Local de Neoplasia , Osteogênese , Osteossarcoma/cirurgia , Impressão Tridimensional , Alicerces Teciduais
16.
Biomaterials ; 275: 120972, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34186237

RESUMO

Cell-based therapy for augmentation of core decompression (CD) using mesenchymal stromal cells (MSCs) is a promising treatment for early stage osteonecrosis of the femoral head (ONFH). Recently, the therapeutic potential for immunomodulation of osteogenesis using preconditioned (with pro-inflammatory cytokines) MSCs (pMSCs), or by the timely resolution of inflammation using MSCs that over-express anti-inflammatory cytokines has been described. Here, pMSCs exposed to tumor necrosis factor-alpha and lipopolysaccharide for 3 days accelerated osteogenic differentiation in vitro. Furthermore, injection of pMSCs encapsulated with injectable hydrogels into the bone tunnel facilitated angiogenesis and osteogenesis in the femoral head in vivo, using rabbit bone marrow-derived MSCs and a model of corticosteroid-associated ONFH in rabbits. In contrast, in vitro and in vivo studies demonstrated that genetically-modified MSCs that over-express IL4 (IL4-MSCs), established by using a lentiviral vector carrying the rabbit IL4 gene under the cytomegalovirus promoter, accelerated proliferation of MSCs and decreased the percentage of empty lacunae in the femoral head. Therefore, adjunctive cell-based therapy of CD using pMSCs and IL4-MSCs may hold promise to heal osteonecrotic lesions in the early stage ONFH. These interventions must be applied in a temporally sensitive fashion, without interfering with the mandatory acute inflammatory phase of bone healing.


Assuntos
Corticosteroides/efeitos adversos , Necrose da Cabeça do Fêmur , Células-Tronco Mesenquimais , Animais , Medula Óssea , Cabeça do Fêmur , Necrose da Cabeça do Fêmur/induzido quimicamente , Necrose da Cabeça do Fêmur/terapia , Interleucina-4 , Osteogênese , Coelhos
17.
Front Cell Dev Biol ; 9: 631063, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33816480

RESUMO

Chronic inflammation is a common feature in many diseases of different organ systems, including bone. However, there are few interventions to mitigate chronic inflammation and preserve host tissue. Previous in vitro studies demonstrated that preconditioning of mesenchymal stem cells (pMSCs) using lipopolysaccharide and tumor necrosis factor-α polarized macrophages from a pro-inflammatory to an anti-inflammatory phenotype and increased osteogenesis compared to unaltered MSCs. In the current study, we investigated the local injection of MSCs or pMSCs during the acute versus chronic inflammatory phase in a murine model of inflammation of bone: the continuous femoral intramedullary polyethylene particle infusion model. Chronic inflammation due to contaminated polyethylene particles decreased bone mineral density and increased osteoclast-like cells positively stained with leukocyte tartrate resistant acid phosphatase (TRAP) staining, and resulted in a sustained M1 pro-inflammatory macrophage phenotype and a decreased M2 anti-inflammatory phenotype. Local injection of MSCs or pMSCs during the chronic inflammatory phase reversed these findings. Conversely, immediate local injection of pMSCs during the acute inflammatory phase impaired bone healing, probably by mitigating the mandatory acute inflammatory reaction. These results suggest that the timing of interventions to facilitate bone healing by modulating inflammation is critical to the outcome. Interventions to facilitate bone healing by modulating acute inflammation should be prudently applied, as this phase of bone healing is temporally sensitive. Alternatively, local injection of MSCs or pMSCs during the chronic inflammatory phase may be a potential intervention to mitigate the adverse effects of contaminated particles on bone.

18.
J Biomed Mater Res A ; 109(10): 1828-1839, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33779115

RESUMO

Wear particle-associated bone loss (periprosthetic osteolysis) constrains the longevity of total joint arthroplasty (TJA). Wear particles induce a prolonged upregulation of nuclear factor kappa B (NF-κB) signaling in macrophages and osteoclasts. Synthetic double-stranded oligodeoxynucleotides (ODNs) can prevent the binding of NF-κB to the promoter regions of targeted genes and inhibit genetic activation. We tested the hypothesis that polyethylene-particle induced chronic inflammatory bone loss could be suppressed by local delivery of NF-κB decoy ODNs in murine in vivo model. Polyethylene particles were continuously infused into the medullary cavity of the distal femur for 6 weeks to induce chronic inflammation, and micro-computational tomography and immunohistochemical analysis were performed. Particle-induced chronic inflammation resulted in lower BMD values, an increase in osteoclastogenesis and nuclear translocation of p65, a prolonged M1 pro-inflammatory macrophage phenotype, and a decrease of M2 anti-inflammatory macrophage phenotype. Delayed timing of local infusion of NF-κB decoy ODN for the last 3 weeks reversed polyethylene-particle associated chronic inflammatory bone loss and facilitated bone healing. This study demonstrated that polyethylene-particle associated chronic inflammatory osteolysis can be effectively modulated via interference with the NF-κB pathway; this minimally invasive intervention could potentially be an efficacious therapeutic strategy for periprosthetic osteolysis after TJA.


Assuntos
Inflamação/patologia , NF-kappa B/metabolismo , Osteólise/patologia , Polietileno/toxicidade , Fosfatase Alcalina/metabolismo , Animais , Núcleo Celular/metabolismo , Doença Crônica , Modelos Animais de Doenças , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Oligodesoxirribonucleotídeos/farmacologia , Osteogênese/efeitos dos fármacos , Fenótipo , Fator de Transcrição RelA/metabolismo
19.
Stem Cell Res Ther ; 12(1): 40, 2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33413614

RESUMO

BACKGROUND: Mesenchymal stem cell (MSC)-based therapy has the potential for immunomodulation and enhancement of tissue regeneration. Genetically modified MSCs that over-express specific cytokines, growth factors, or chemokines have shown great promise in pre-clinical studies. In this regard, the anti-inflammatory cytokine interleukin (IL)-4 converts pro-inflammatory M1 macrophages into an anti-inflammatory M2 phenotype; M2 macrophages mitigate chronic inflammation and enhance osteogenesis by MSC lineage cells. However, exposure to IL-4 prematurely inhibits osteogenesis of MSCs in vitro; furthermore, IL-4 overexpressing MSCs inhibit osteogenesis in vivo during the acute inflammatory period. Platelet-derived growth factor (PDGF)-BB has been shown to enhance osteogenesis of MSCs with a dose-dependent effect. METHODS: In this study, we generated a lentiviral vector that produces PDGF-BB under a weak promoter (phosphoglycerate kinase, PGK) and lentiviral vector producing IL-4 under a strong promoter (cytomegalovirus, CMV). We infected MSCs with PDGF-BB and IL-4-producing lentiviral vectors separately or in combination to investigate cell proliferation and viability, protein expression, and the capability for osteogenesis. RESULTS: PDGF-BB and IL-4 co-overexpression was observed in the co-infected MSCs and shown to enhance cell proliferation and viability, and osteogenesis compared to IL-4 overexpressing MSCs alone. CONCLUSIONS: Overexpression of PDGF-BB together with IL-4 mitigates the inhibitory effect of IL-4 on osteogenesis by IL-4 overexpressing MSCS. PDGF-BB and IL-4 overexpressing MSCs may be a potential strategy to facilitate osteogenesis in scenarios of both acute and chronic inflammation.


Assuntos
Células-Tronco Mesenquimais , Becaplermina , Regeneração Óssea , Interleucina-4/genética , Osteogênese
20.
J Biomed Mater Res A ; 109(8): 1512-1520, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33340244

RESUMO

Periprosthetic osteolysis remains as a major complication of total joint replacement surgery. Modulation of macrophage polarization with interleukin-4 (IL-4) has emerged as an effective means to limit wear particle-induced osteolysis. The aim of this study was to evaluate the efficacy of local IL-4 delivery in treating preexisting particle-induced osteolysis. To this end, recently established 8 week modification of murine continuous femoral intramedullary particle infusion model was utilized. Subcutaneous infusion pumps were used to deliver polyethylene (PE) particles into mouse distal femur for 4 weeks to induce osteolysis. IL-4 was then added to the particle infusion for another 4 weeks. This delayed IL-4 treatment (IL-4 Del) was compared to IL-4 delivered continuously (IL-4 Cont) with PE particles from the beginning and to the infusion of particles alone for 8 weeks. Both IL-4 treatments were highly effective in preventing and repairing preexisting particle-induced bone loss as assessed by µCT. Immunofluorescence indicated a significant reduction in the number of F4/80 + iNOS + M1 macrophages and increase in the number of F4/80 + CD206 + M2 macrophages with both IL-4 treatments. Reduction in the number of tartrate resistant acid phosphatase + osteoclasts and increase in the amount of alkaline phosphatase (ALP) + osteoblasts was also observed with both IL-4 treatments likely explaining the regeneration of bone in these samples. Interesting, slightly more bone formation and ALP + osteoblasts were seen in the IL-4 Del group than in the IL-4 Cont group although these differences were not statistically significant. The study is a proof of principle that osteolytic lesions can be repaired via modulation of macrophage polarization.


Assuntos
Remodelação Óssea/efeitos dos fármacos , Interleucina-4/uso terapêutico , Prótese Articular/efeitos adversos , Osteólise/tratamento farmacológico , Osteólise/etiologia , Animais , Artroplastia de Substituição/efeitos adversos , Interleucina-4/administração & dosagem , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos BALB C
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA