Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Int J Nanomedicine ; 19: 9459-9486, 2024.
Article in English | MEDLINE | ID: mdl-39371481

ABSTRACT

Given the global prevalence of prostate cancer in men, it is crucial to explore more effective treatment strategies. Recently, immunotherapy has emerged as a promising cancer treatment due to its unique mechanism of action and potential long-term effectiveness. However, its limited efficacy in prostate cancer has prompted renewed interest in developing strategies to improve immunotherapy outcomes. Nanomedicine offers a novel perspective on cancer treatment with its unique size effects and surface properties. By employing targeted delivery, controlled release, and enhanced immunogenicity, nanoparticles can be synergized with nanomedicine platforms to amplify the effectiveness of immunotherapy in treating prostate cancer. Simultaneously, nanotechnology can address the limitations of immunotherapy and the challenges of immune escape and tumor microenvironment regulation. Additionally, the synergistic effects of combining nanomedicine with other therapies offer promising clinical outcomes. Innovative applications of nanomedicine include smart nanocarriers, stimulus-responsive systems, and precision medicine approaches to overcome translational obstacles in prostate cancer immunotherapy. This review highlights the transformative potential of nanomedicine in enhancing prostate cancer immunotherapy and emphasizes the need for interdisciplinary collaboration to drive research and clinical applications forward.


Subject(s)
Immunotherapy , Nanomedicine , Prostatic Neoplasms , Tumor Microenvironment , Humans , Male , Prostatic Neoplasms/therapy , Prostatic Neoplasms/immunology , Immunotherapy/methods , Nanomedicine/methods , Tumor Microenvironment/drug effects , Nanoparticles/chemistry , Animals , Precision Medicine/methods , Drug Delivery Systems/methods
2.
Heliyon ; 10(17): e37031, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39286219

ABSTRACT

Diabetes represents a widely acknowledged global public health concern. Diabetic foot ulcer (DFU) stands as one of the most severe complications of diabetes, its occurrence imposing a substantial economic burden on patients, profoundly impacting their quality of life. Despite the deepening comprehension regarding the pathophysiology and cellular as well as molecular responses of DFU, the current therapeutic arsenal falls short of efficacy, failing to offer a comprehensive remedy for deep-seated chronic wounds and microvascular occlusions. Conventional treatments merely afford symptomatic alleviation or retard the disease's advancement, devoid of the capacity to effectuate further restitution of compromised vasculature and nerves. An escalating body of research underscores the prominence of mesenchymal stem cells (MSCs) owing to their paracrine attributes and anti-inflammatory prowess, rendering them a focal point in the realm of chronic wound healing. Presently, MSCs have been validated as a highly promising cellular therapeutic approach for DFU, capable of effectuating cellular repair, epithelialization, granulation tissue formation, and neovascularization by means of targeted differentiation, angiogenesis promotion, immunomodulation, and paracrine activities, thereby fostering wound healing. The secretome of MSCs comprises cytokines, growth factors, chemokines, alongside exosomes harboring mRNA, proteins, and microRNAs, possessing immunomodulatory and regenerative properties. The present study provides a systematic exposition on the etiology of DFU and elucidates the intricate molecular mechanisms and diverse functionalities of MSCs in the context of DFU treatment, thereby furnishing pioneering perspectives aimed at harnessing the therapeutic potential of MSCs for DFU management and advancing wound healing processes.

3.
Toxics ; 12(9)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39330591

ABSTRACT

Air pollution has become a serious public health problem and there is evidence that air pollution affects the incidence of allergic rhinitis. To further investigate the effect of ambient air pollutants on the severity of allergic rhinitis symptoms, a prospective follow-up study in patients with allergic rhinitis was conducted. A total of 167 allergic rhinitis patients with a mean age of 35.4 years, who were visiting the hospital, were enrolled. The daily symptom severity of allergic rhinitis and the concentrations of six air pollutants, including PM2.5, PM10, SO2, CO, O3 and NO2, were collected through follow-up investigations. The impact of ambient air pollutants on symptom severity was assessed via multi-pollutant models. Among several typical ambient air pollutants, we observed correlations of allergic rhinitis symptoms with PM2.5, PM10, CO, SO2 and NO2, whereas O3 showed no such correlation. Specifically, PM2.5 and PM10 were significantly associated with sneezing and nasal blockage. NO2 was significantly correlated with symptoms of rhinorrhea, itchy nose and itchy eyes. CO was significantly linked to sneezing and nasal blockage symptoms. These air pollutants not only had a direct impact on allergic rhinitis symptoms but also exhibited a lagging effect. This study indicates that short-term exposure to air pollutants is associated with exacerbation of nasal symptoms in patients with allergic rhinitis, leading to a decline in their quality of life.

4.
Int J Nanomedicine ; 19: 8641-8660, 2024.
Article in English | MEDLINE | ID: mdl-39188861

ABSTRACT

Osteosarcoma is the predominant primary malignant bone tumor that poses a significant global health challenge. MicroRNAs (miRNAs) that regulate gene expression are associated with osteosarcoma pathogenesis. Thus, miRNAs are potential therapeutic targets for osteosarcoma. Nanoparticles, widely used for targeted drug delivery, facilitate miRNA-based osteosarcoma treatment. Numerous studies have focused on miRNA delivery using nanoparticles to inhibit the progress of osteosarcoma. Polymer-based, lipid-based, inorganic-based nanoparticles and extracellular vesicles were used to deliver miRNAs for the treatment of osteosarcoma. They can be modified to enhance drug loading and delivery capabilities. Also, miRNA delivery was combined with traditional therapies, for example chemotherapy, to treat osteosarcoma. Consequently, miRNA delivery offers promising therapeutic avenues for osteosarcoma, providing renewed hope for patients. This review emphasizes the studies utilizing nanoparticles for miRNA delivery in osteosarcoma treatment, then introduced and summarized the nanoparticles in detail. And it also discusses the prospects for clinical applications.


Subject(s)
Bone Neoplasms , MicroRNAs , Nanoparticles , Osteosarcoma , Osteosarcoma/genetics , Osteosarcoma/drug therapy , Osteosarcoma/therapy , Humans , MicroRNAs/administration & dosage , MicroRNAs/genetics , Bone Neoplasms/genetics , Bone Neoplasms/therapy , Bone Neoplasms/drug therapy , Nanoparticles/chemistry , Animals , Drug Delivery Systems/methods
5.
Front Bioeng Biotechnol ; 12: 1426477, 2024.
Article in English | MEDLINE | ID: mdl-38915336

ABSTRACT

Background: Branched gold and silver nanoparticles coated with polydopamine (Au-Ag-PDA) demonstrate high photothermal conversion efficiency. Utilizing umbilical cord mesenchymal stem cell membranes (MSCM) as an effective drug delivery system, our preliminary studies investigated the suppression of sebum secretion in sebaceous glands using MSCM-coated Au-Ag-PDA nano-particles (Au-Ag-PDA@MSCM) combined with 808 nm laser irradiation, showing potential for dermatological applications in acne treatment. Methods: This study employs proteomic analysis, complemented by subsequent techniques such as Western blotting (WB), small interfering RNA (siRNA), and transmission electron microscopy, to further investigate the differential mechanisms by which Au-Ag-PDA and Au-Ag-PDA@MSCM-mediated photothermal therapy (PTT) suppress sebum secretion. Results: Our proteomic analysis indicated mitochondrial respiratory chain damage in sebaceous gland tissues post-PTT, with further validation revealing ferroptosis in sebaceous cells and tissues. Acyl-CoA Synthetase Long-Chain Family Member 4 (Acsl4) has been identified as a critical target, with Au-Ag-PDA@MSCM demonstrating enhanced ferroptotic effects. Conclusion: These findings significantly advance our understanding of how PTT mediated by Au-Ag-PDA@MSCM nanoparticles reduces sebum secretion and underscore the pivotal role of MSCM in inducing ferroptosis in sebaceous glands, thus providing a robust theoretical foundation for employing PTT via specific molecular pathways in acne treatment.

6.
Neurochem Res ; 49(8): 2005-2020, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38814357

ABSTRACT

Postoperative cognitive dysfunction (POCD) occurs after surgery and severely impairs patients' quality of life. Finding POCD-associated variables can aid in its diagnosis and prognostication. POCD is associated with noncoding RNAs, such as microRNAs (miRNAs), involved in metabolic function, immune response alteration, and cognitive ability impairment; however, the underlying mechanisms remain unclear. The aim of this study was to investigate hub miRNAs (i.e., miRNAs that have an important regulatory role in diseases) regulating postoperative cognitive function and the associated mechanisms. Hub miRNAs were identified by bioinformatics, and their expression in mouse hippocampus tissues was determined using real-time quantitative polymerase chain reaction. Hub miRNAs were overexpressed or knocked down in cell and animal models to test their effects on neuroinflammation and postoperative cognitive function. Six differentially expressed hub miRNAs were identified. miR-206-3p was the only broadly conserved miRNA, and it was used in follow-up studies and animal experiments. Its inhibitors reduced the release of proinflammatory cytokines in BV-2 microglia by regulating its target gene, brain-derived neurotrophic factor (BDNF), and the downstream signaling pathways. miR-206-3p inhibition suppressed microglial activation in the hippocampi of mice and improved learning and cognitive decline. Therefore, miR-206-3p significantly affects POCD, implying its potential as a therapeutic target.


Subject(s)
Brain-Derived Neurotrophic Factor , Cognition , Hippocampus , Mice, Inbred C57BL , MicroRNAs , Postoperative Cognitive Complications , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Mice , Postoperative Cognitive Complications/metabolism , Male , Hippocampus/metabolism , Cognition/physiology , Aging/metabolism , Aging/genetics , Microglia/metabolism , Cell Line
7.
Front Oncol ; 14: 1403719, 2024.
Article in English | MEDLINE | ID: mdl-38751816

ABSTRACT

Background: The primary treatment strategies for melanoma include surgical excision, chemotherapy, and radiotherapy. However, the efficacy of these treatments is often limited by drug resistance, recurrence, and severe side effects. Therefore, we aimed to develop a targeted drug delivery system capable of selectively locating tumor sites to minimize systemic toxicity and enhance therapeutic efficacy. This cell drug delivery system can also deliver chemotherapeutic drugs to the tumor microenvironment. Methods: We treated B16F10 cells with hyperosmotic cold shock (HCS) to obtain and characterize HCS cells. We then investigated the anti-tumor effects and immune activation capabilities of these cells and explored their potential as a targeted drug delivery system. Results: HCS cells not only maintained an intact cellular structure and tumor antigens but also exhibited high expression of the homologous melanoma-associated antigen glycoprotein 100. These cells demonstrated an exceptional capacity for loading and releasing doxorubicin, which has chemotherapeutic anti-tumor effects. HCS cells can precisely target the tumor microenvironment to minimize systemic toxicity, inducing an immune response by activating CD3+ and CD4+ T cells. Conclusion: HCS cells are non-carcinogenic, with both cellular and tumor antigens intact; thus, they are suitable drug delivery carriers. Our findings highlight the potential of HCS cells for carrying doxorubicin because of their high drug-loading efficiency, effective tumor-targeting and anti-tumor effects. Therefore, our results will facilitate the development of melanoma treatments that have higher efficacy than those in the literature.

8.
Biomed Pharmacother ; 174: 116515, 2024 May.
Article in English | MEDLINE | ID: mdl-38569276

ABSTRACT

Mesenchymal stem cell exosome (MSCs-exo) is a class of products secreted by mesenchymal stem cells (MSCs) that contain various biologically active substances. MSCs-exo is a promising alternative to MSCs due to their lower immunogenicity and lack of ethical constraints. Ginsenoside Rh2 (Rh2) is a hydrolyzed component of the primary active substance of ginsenosides. Rh2 has a variety of pharmacological functions, including anti-inflammatory, anti-tumor, and antioxidant. Studies have demonstrated that gut microbiota and metabolites are critical in developing rheumatoid arthritis (RA). In this study, we constructed a collagen-induced arthritis (CIA) model in rats. We used MSCs-exo combined with Rh2 to treat CIA rats. To observe the effect of MSCs-exo combined with Rh2 on joint inflammation, rat feces were collected for 16 rRNA amplicon sequencing and untargeted metabolomics analysis. The results showed that the arthritis index score and joint swelling of CIA rats treated with MSCs-exo in combination with Rh2 were significantly lower than those of the model and MSCs-exo alone groups. MSCs-exo and Rh2 significantly ameliorated the disturbed gut microbiota in CIA rats. The regulation of Candidatus_Saccharibacteria and Clostridium_XlVb regulation may be the most critical. Rh2 enhanced the therapeutic effect of MSCs-exo compared with the MSCs-exo -alone group. Furthermore, significant changes in gut metabolites were observed in the CIA rat group, and these differentially altered metabolites may act as messengers for host-microbiota interactions. These differential metabolites were enriched into relevant critical metabolic pathways, revealing possible pathways for host-microbiota interactions.


Subject(s)
Arthritis, Experimental , Gastrointestinal Microbiome , Ginsenosides , Mesenchymal Stem Cells , Animals , Humans , Male , Rats , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Experimental/microbiology , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/microbiology , Arthritis, Rheumatoid/therapy , Exosomes/metabolism , Gastrointestinal Microbiome/drug effects , Ginsenosides/pharmacology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Umbilical Cord , Collagen/metabolism , Collagen/pharmacology
12.
Stem Cell Res Ther ; 15(1): 87, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38520027

ABSTRACT

BACKGROUND: The clinical application of human bone-marrow derived mesenchymal stem cells (MSCs) for the treatment of refractory diseases has achieved remarkable results. However, there is a need for a systematic evaluation of the quality and safety of MSCs sourced from donors. In this study, we sought to assess one potential factor that might impact quality, namely the age of the donor. METHODS: We downloaded two data sets from each of two Gene Expression Omnibus (GEO), GSE39035 and GSE97311 databases, namely samples form young (< 65 years of age) and old (> 65) donor groups. Through, bioinformatics analysis and experimental validation to these retrieved data, we found that MSCs derived from aged donors can lead to differential expression of gene profiles compared with those from young donors, and potentially affect the function of MSCs, and may even induce malignant tumors. RESULTS: We identified a total of 337 differentially expressed genes (DEGs), including two upregulated and eight downregulated genes from the databases of both GSE39035 and GSE97311. We further identified 13 hub genes. Six of them, TBX15, IGF1, GATA2, PITX2, SNAI1 and VCAN, were highly expressed in many human malignancies in Human Protein Atlas database. In the MSCs in vitro senescent cell model, qPCR analysis validated that all six hub genes were highly expressed in senescent MSCs. Our findings confirm that aged donors of MSCs have a significant effect on gene expression profiles. The MSCs from old donors have the potential to cause a variety of malignancies. These TBX15, IGF1, GATA2, PITX2, SNAI1, VCAN genes could be used as potential biomarkers to diagnosis aging state of donor MSCs, and evaluate whether MSCs derived from an aged donor could be used for therapy in the clinic. Our findings provide a diagnostic basis for the clinical use of MSCs to treat a variety of diseases. CONCLUSIONS: Therefore, our findings not only provide guidance for the safe and standardized use of MSCs in the clinic for the treatment of various diseases, but also provide insights into the use of cell regeneration approaches to reverse aging and support rejuvenation.


Subject(s)
Mesenchymal Stem Cells , Neoplasms , Humans , Aged , Aging/metabolism , Tissue Donors , Biomarkers/metabolism , Mesenchymal Stem Cells/metabolism , Neoplasms/metabolism , T-Box Domain Proteins/metabolism
13.
Plants (Basel) ; 12(24)2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38140408

ABSTRACT

Actinidia chinensis Planch. is a fruit tree originating from China that is abundant in the wild. Actinidia eriantha Benth. is a type of A. chinensis that has emerged in recent years. The shape of A. eriantha is an elongated oval, and the skin is covered with dense, non-shedding milk-white hairs. The mature fruit has flesh that is bright green in colour, and the fruit has a strong flavour and a grass-like smell. It is appreciated for its rich nutrient content and unique flavour. Vitamin C, sugar, and organic acids are key factors in the quality and flavour composition of A. eriantha but have not yet been systematically analysed. Therefore, we sequenced the transcriptome of A. eriantha at three developmental stages and labelled them S1, S2, and S3, and comparisons of S1 vs. S2, S1 vs. S3, and S2 vs. S3 revealed 1218, 4019, and 3759 upregulated differentially expressed genes and 1823, 3415, and 2226 downregulated differentially expressed genes, respectively. Furthermore, the upregulated differentially expressed genes included 213 core genes, and Gene Ontology enrichment analysis showed that they were enriched in hormones, sugars, organic acids, and many organic metabolic pathways. The downregulated differentially expressed genes included 207 core genes, which were enriched in the light signalling pathway. We further constructed the metabolic pathways of sugars, organic acids, and vitamin C in A. eriantha and identified the genes involved in vitamin C, sugar, and organic acid synthesis in A. eriantha fruits at different stages. During fruit development, the vitamin C content decreased, the carbohydrate compound content increased, and the organic acid content decreased. The gene expression patterns were closely related to the accumulation patterns of vitamin C, sugars, and organic acids in A. eriantha. The above results lay the foundation for the accumulation of vitamin C, sugars, and organic acids in A. eriantha and for understanding flavour formation in A. eriantha.

14.
J Nanobiotechnology ; 21(1): 392, 2023 Oct 27.
Article in English | MEDLINE | ID: mdl-37891598

ABSTRACT

The development of nanotechnology has provided numerous possibilities for the diagnosis and treatment of cancer. Paradoxically, some in vivo experimental studies have also shown that nanoparticles (NPs) could promote tumor progression, but the specific mechanism is not yet clear. Primary tumors can release extracellular vesicles (EVs) which can promote the inoculation and growth of tumor cells that have metastasized to distant organs. So, whether nanomaterials can promote tumor progression through tumor-derived EVs deserves further research. Here, we showed that TiO2 NPs, widely used in nanomedicine, could trigger tumor-derived EVs with enhanced pro-metastatic capacity in vitro and in vivo. Mechanically, miR-301a-3p derived from NPs-elicited EVs could be delivered into vascular endothelial cells, which inhibited the expression of VEGFR2 and VE-cadherin by targeting S1PR1, leading to disrupt the tight junctions of vascular endothelial cells, thus to promote vascular permeability and angiogenesis, and induce the formation of pre-metastasis niches in vivo. This study emphasizes that it is urgent to consider the effect of NPs on EVs under long-term use conditions when designing nanodrugs for cancer treatment.


Subject(s)
Extracellular Vesicles , MicroRNAs , Nanoparticles , Neoplasms , Humans , Endothelial Cells , Neoplasms/metabolism , Extracellular Vesicles/metabolism , MicroRNAs/metabolism
15.
Sci Rep ; 13(1): 13213, 2023 08 14.
Article in English | MEDLINE | ID: mdl-37580391

ABSTRACT

The increase in the aging population has seriously affected our society. Neurodegenerative diseases caused by aging of the brain significantly impact the normal life of the elderly, and delaying brain aging is currently the focus of research. SIRT1 is a viable therapeutic target, and there is mounting evidence that it plays a significant role in the aging process. Mesenchymal stem cell-derived exosomes (MSC-Exos) have gained widespread interest as nanotherapeutic agents because of their ability to be injected at high doses to reduce the immune response. The present study focused on the ameliorative effect of MSC-Exos on aging mice and the potential mechanisms of this effect on cognitive impairment and brain aging. In this study, we first tested the neuroprotective effects of MSC-Exos in vitro on H2O2-induced oxidative damage in BV2 cells. An in vivo SAMP8 rapid senescence mouse model showed that MSC-Exos significantly increased SIRT1 gene expression in senescent mice. In addition, MSC-Exos also had an anti-apoptotic effect and reduced oxidative stress in the brains of SAMP8 senescent mice. In conclusion, MSC-Exos may exert neuroprotective effects and help prevent brain senescence in SAMP8 mice by activating the SIRT1 signaling pathway.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Neuroprotective Agents , Sirtuin 1 , Animals , Mice , Aging , Brain/metabolism , Exosomes/metabolism , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Mesenchymal Stem Cells/metabolism , Neuroprotective Agents/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
16.
Aging (Albany NY) ; 15(14): 6933-6949, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37470690

ABSTRACT

The extensive utilization of iron oxide nanoparticles in medical and life science domains has led to a substantial rise in both occupational and public exposure to these particles. The potential toxicity of nanoparticles to living organisms, their impact on the environment, and the associated risks to human health have garnered significant attention and come to be a prominent area in contemporary research. The comprehension of the potential toxicity of nanoparticles has emerged as a crucial concern to safeguard human health and facilitate the secure advancement of nanotechnology. As nanocarriers and targeting agents, the biocompatibility of them determines the use scope and application prospects, meanwhile surface modification becomes an important measure to improve the biocompatibility. Three different types of iron oxide nanoparticles (Fe3O4, Fe3O4@PDA and MSCM-Fe3O4@PDA) were injected into mice through the tail veins. The acute neurotoxicity of them in mice was evaluated by measuring the levels of autophagy and apoptosis in the brain tissues. Our data revealed that iron oxide nanoparticles could cause nervous system damage by regulating the ASK1/JNK signaling pathway. Apoptosis and autophagy may play potential roles in this process. Exposure to combined surface functionalization of mesenchymal stem cell membrane and polydopamine showed the neuroprotective effect and may alleviate brain nervous system disorders.


Subject(s)
MAP Kinase Signaling System , Nanoparticles , Mice , Humans , Animals , Autophagy , Apoptosis
17.
Life Sci ; 328: 121836, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37295713

ABSTRACT

PURPOSE: To investigate the effect of connexin 43 (Cx43) on corneal neovascularization and its regulation of VEGFR2 on vascular endothelial cells. METHODS: In vivo, we used mouse corneal suture model to induce corneal neovascularization and discovered the function of gap26 in corneal neovascularization. In vitro, the effect of gap26 on HUVEC was observed by cell proliferation, tube formation and scratch experiments. WB and PCR detected the changes in angiogenic protein and mRNA expression. Knockdown of key mRNA in neovascularization using siRNA confirmed that Cx43 regulates neovascularization through the ß-catenin-VE-cadherin-VEGFR2-Erk signaling pathway. RESULTS: In vivo, gap26 can reduce mouse corneal neovascularization. In vitro, we show that Cx43 expression is increased in the presence of VEGFA stimulation, and when we use gap26 to inhibit Cx43 can reduce vascular endothelial cell proliferation, tube formation and migration. We found that the expression of pVEGFR2 and pErk increased in response to VEGFA, while they decreased after using gap26. And the expression of ß-catenin and VE-cadherin decreased in response to VEGFA, while they increased after using gap26. Furthermore, we found that Cx43 regulates angiogenesis through the ß-catenin-VE-cadherin-VEGFR2-Erk pathway. CONCLUSIONS: Gap26 can downregulate VEGFR2 phosphorylation by stabilizing the expression of ß-catenin and VE-cadherin on the cell membrane, thereby inhibiting VEGFA-induced HUVECs proliferation, migration and tube formation and inhibiting corneal neovascularization.


Subject(s)
Connexin 43 , Corneal Neovascularization , Signal Transduction , Animals , Humans , Mice , beta Catenin/metabolism , Cadherins/metabolism , Cell Movement , Cell Proliferation , Connexin 43/genetics , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism
18.
Med Sci Monit ; 29: e940264, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37310931

ABSTRACT

Osteoradionecrosis (ORN) of the femoral head is an important issue for orthopedists and radiologists in clinical practice. With the rapid development of technological advances in radiation therapy and the improvement in cancer survival rates, the incidence of ORN is rising, and there is an unmet need for basic and clinical research. The pathogenesis of ORN is complex, and includes vascular injury, mesenchymal stem cell injury, bone loss, reactive oxygen species, radiation-induced fibrosis, and cell senescence. The diagnosis of ORN is challenging and requires multiple considerations, including exposure to ionizing radiation, clinical manifestations, and findings on physical examination and imaging. Differential diagnosis is essential, as clinical symptoms of ORN of the femoral head can resemble many other hip conditions. Hyperbaric oxygen therapy, total hip arthroplasty, and Girdlestone resection arthroplasty are effective treatments, each with their own advantages and disadvantages. The literature on ORN of the femoral head is incomplete and there is no criterion standard or clear consensus on management. Clinicians should gain a better and more comprehensive understanding on this disease to facilitate its early and better prevention, diagnosis, and treatment. This article aims to review the pathogenesis, diagnosis, and management of osteoradionecrosis of the femoral head.


Subject(s)
Arthroplasty, Replacement, Hip , Osteoradionecrosis , Humans , Osteoradionecrosis/diagnosis , Osteoradionecrosis/etiology , Osteoradionecrosis/therapy , Femur Head , Diagnosis, Differential , Radiation, Ionizing
19.
Front Immunol ; 14: 1125257, 2023.
Article in English | MEDLINE | ID: mdl-37251412

ABSTRACT

Systemic sclerosis (SSc) is an intricate systemic autoimmune disease with pathological features such as vascular injury, immune dysregulation, and extensive fibrosis of the skin and multiple organs. Treatment options are limited; however, recently, mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been acknowledged in preclinical and clinical trials as being useful in treating autoimmune diseases and are likely superior to MSCs alone. Recent research has also shown that MSC-EVs can ameliorate SSc and the pathological changes in vasculopathy, immune dysfunction, and fibrosis. This review summarizes the therapeutic effects of MSC-EVs on SSc and the mechanisms that have been discovered to provide a theoretical basis for future studies on the role of MSC-EVs in treating SSc.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Scleroderma, Systemic , Humans , Immunomodulation , Scleroderma, Systemic/therapy , Fibrosis
20.
Biomed Pharmacother ; 161: 114455, 2023 May.
Article in English | MEDLINE | ID: mdl-36905811

ABSTRACT

Mesenchymal stem cell-derived exosomes (MSCs-exo) can be used for treating Alzheimer's disease (AD) by promoting amyloid-ß (Aß) degradation, modulating immune responses, protecting neurology, promoting axonal growth, and improving cognitive impairment. Increasing evidence suggests that the alteration of gut microbiota is closely related to the occurrence and development of Alzheimer's disease. In this study, we hypothesized that dysbiosis of gut microbiota might limit the therapy of MSCs-exo, and the application of antibiotics would improve the therapy. METHODS: In this original research study, we used MSCs-exo to treat 5 ×FAD mice and fed them antibiotic cocktails for 1 week to detect cognitive ability and neuropathy. The mice's feces were collected to investigate alterations in the microbiota and metabolites. RESULTS: The results revealed that the AD gut microbiota eliminated the therapeutic effect of MSCs-exo, whereas antibiotic modulation of disordered gut microbiota and associated metabolites enhanced the therapeutic effect of MSCs-exo. CONCLUSIONS: These results encourage the research of novel therapeutics to enhance MSCs-exo treatment for AD, which could benefit a broader range of patients with AD.


Subject(s)
Alzheimer Disease , Exosomes , Gastrointestinal Microbiome , Mesenchymal Stem Cells , Mice , Animals , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL