Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
Adv Mater ; 36(23): e2312326, 2024 Jun.
Article En | MEDLINE | ID: mdl-38389502

Clinical treatment of cancer commonly incorporates X-ray radiation therapy (XRT), and developing spatially precise radiation-activatable drug delivery strategies may improve XRT efficacy while limiting off-target toxicities associated with systemically administered drugs. Nevertheless, achieving this has been challenging thus far because strategies typically rely on radical species with short lifespans, and the inherent nature of hypoxic and acidic tumor microenvironments may encourage spatially heterogeneous effects. It is hypothesized that the challenge could be bypassed by using scintillating nanoparticles that emit light upon X-ray absorption, locally forming therapeutic drug depots in tumor tissues. Thus a nanoparticle platform (Scintillating nanoparticle Drug Depot; SciDD) that enables the local release of cytotoxic payloads only after activation by XRT is developed, thereby limiting off-target toxicity. As a proof-of-principle, SciDD is used to deliver a microtubule-destabilizing payload MMAE (monomethyl auristatin E). With as little as a 2 Gy local irradiation to tumors, MMAE payloads are released effectively to kill tumor cells. XRT-mediated drug release is demonstrated in multiple mouse cancer models and showed efficacy over XRT alone (p < 0.0001). This work shows that SciDD can act as a local drug depot with spatiotemporally controlled release of cancer therapeutics.


Nanoparticles , Animals , Nanoparticles/chemistry , Mice , Humans , Cell Line, Tumor , Drug Liberation , Delayed-Action Preparations/chemistry , Oligopeptides/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Drug Carriers/chemistry
3.
Nat Nanotechnol ; 18(12): 1502-1514, 2023 Dec.
Article En | MEDLINE | ID: mdl-37884660

Commencing with the breakdown of immune tolerance, multiple pathogenic factors, including synovial inflammation and harmful cytokines, are conjointly involved in the progression of rheumatoid arthritis. Intervening to mitigate some of these factors can bring a short-term therapeutic effect, but other unresolved factors will continue to aggravate the disease. Here we developed a ceria nanoparticle-immobilized mesenchymal stem cell nanovesicle hybrid system to address multiple factors in rheumatoid arthritis. Each component of this nanohybrid works individually and also synergistically, resulting in comprehensive treatment. Alleviation of inflammation and modulation of the tissue environment into an immunotolerant-favourable state are combined to recover the immune system by bridging innate and adaptive immunity. The therapy is shown to successfully treat and prevent rheumatoid arthritis by relieving the main symptoms and also by restoring the immune system through the induction of regulatory T cells in a mouse model of collagen-induced arthritis.


Arthritis, Experimental , Arthritis, Rheumatoid , Mice , Animals , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Adaptive Immunity , Cytokines , Inflammation
4.
Clin Cancer Res ; 29(17): 3457-3470, 2023 09 01.
Article En | MEDLINE | ID: mdl-37289199

PURPOSE: Oncogene-driven macropinocytosis fuels nutrient scavenging in some cancer types, yet whether this occurs in thyroid cancers with prominent MAPK-ERK and PI3K pathway mutations remains unclear. We hypothesized that understanding links between thyroid cancer signaling and macropinocytosis might uncover new therapeutic strategies. EXPERIMENTAL DESIGN: Macropinocytosis was assessed across cells derived from papillary thyroid cancer (PTC), follicular thyroid cancer (FTC), non-malignant follicular thyroid, and aggressive anaplastic thyroid cancer (ATC), by imaging fluorescent dextran and serum albumin. The impacts of ectopic BRAFV600E and mutant RAS, genetic PTEN silencing, and inhibitors targeting RET, BRAF, and MEK kinases were quantified. BrafV600E p53-/- ATC tumors in immunocompetent mice were used to measure efficacy of an albumin-drug conjugate comprising microtubule-destabilizing monomethyl auristatin E (MMAE) linked to serum albumin via a cathepsin-cleavable peptide (Alb-vc-MMAE). RESULTS: FTC and ATC cells showed greater macropinocytosis than non-malignant and PTC cells. ATC tumors accumulated albumin at 8.8% injected dose per gram tissue. Alb-vc-MMAE, but not MMAE alone, reduced tumor size by >90% (P < 0.01). ATC macropinocytosis depended on MAPK/ERK activity and nutrient signaling, and increased by up to 230% with metformin, phenformin, or inhibition of IGF1Ri in monoculture but not in vivo. Macrophages also accumulated albumin and express the cognate IGF1R ligand, IGF1, which reduced ATC responsiveness to IGF1Ri. CONCLUSIONS: These findings identify regulated oncogene-driven macropinocytosis in thyroid cancers and demonstrate the potential of designing albumin-bound drugs to efficiently treat them.


Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms , Mice , Animals , Phosphatidylinositol 3-Kinases/genetics , Mutation , Proto-Oncogene Proteins B-raf , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Carcinoma, Anaplastic/genetics , Oncogenes , Thyroid Cancer, Papillary/genetics , Serum Albumin/genetics , Serum Albumin/therapeutic use
5.
Exp Mol Med ; 55(3): 541-554, 2023 03.
Article En | MEDLINE | ID: mdl-36854774

The development of therapeutic cancer vaccines (TCVs) that provide clinical benefits is challenging mainly due to difficulties in identifying immunogenic tumor antigens and effectively inducing antitumor immunity. Furthermore, there is an urgent need for personalized TCVs because only a limited number of tumor antigens are shared among cancer patients. Several autologous nanovaccines that do not require the identification of immunogenic tumor antigens have been proposed as personalized TCVs. However, these nanovaccines generally require exogenous adjuvants (e.g., Toll-like receptor agonists) to improve vaccine immunogenicity, which raises safety concerns. Here, we present senescent cancer cell-derived nanovesicle (SCCNV) as a personalized TCV that provides patient-specific tumor antigens and improved vaccine immunogenicity without the use of exogenous adjuvants. SCCNVs are prepared by inducing senescence in cancer cells ex vivo and subsequently extruding the senescent cancer cells through nanoporous membranes. In the clinical setting, SCCNVs can be prepared from autologous cancer cells from the blood of liquid tumor patients or from tumors surgically removed from solid cancer patients. SCCNVs also contain interferon-γ and tumor necrosis factor-α, which are expressed during senescence. These endogenous cytokines act as adjuvants and enhance vaccine immunogenicity, avoiding the need for exogenous adjuvants. Intradermally injected SCCNVs effectively activate dendritic cells and tumor-specific T cells and inhibit primary and metastatic tumor growth and tumor recurrence. SCCNV therapy showed an efficacy similar to that of immune checkpoint blockade (ICB) therapy and synergized with ICB. SCCNVs, which can be prepared using a simple and facile procedure, show potential as personalized TCVs.


Cancer Vaccines , Neoplasms , Humans , Cancer Vaccines/therapeutic use , Neoplasms/drug therapy , Antigens, Neoplasm , Adjuvants, Immunologic
6.
Nano Lett ; 23(2): 476-490, 2023 01 25.
Article En | MEDLINE | ID: mdl-36638236

Alzheimer's disease (AD) is a neurodegenerative disease with multifactorial pathogenesis. However, most current therapeutic approaches for AD target a single pathophysiological mechanism, generally resulting in unsatisfactory therapeutic outcomes. Recently, mesenchymal stem cell (MSC) therapy, which targets multiple pathological mechanisms of AD, has been explored as a novel treatment. However, the low brain retention efficiency of administered MSCs limits their therapeutic efficacy. In addition, autologous MSCs from AD patients may have poor therapeutic abilities. Here, we overcome these limitations by developing iron oxide nanoparticle (IONP)-incorporated human Wharton's jelly-derived MSCs (MSC-IONPs). IONPs promote therapeutic molecule expression in MSCs. Following intracerebroventricular injection, MSC-IONPs showed a higher brain retention efficiency under magnetic guidance. This potentiates the therapeutic efficacy of MSCs in murine models of AD. Furthermore, human Wharton's jelly-derived allogeneic MSCs may exhibit higher therapeutic abilities than those of autologous MSCs in aged AD patients. This strategy may pave the way for developing MSC therapies for AD.


Alzheimer Disease , Mesenchymal Stem Cells , Neurodegenerative Diseases , Wharton Jelly , Humans , Mice , Animals , Aged , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , Magnetic Iron Oxide Nanoparticles , Cell Differentiation
7.
Adv Mater ; 35(3): e2207719, 2023 Jan.
Article En | MEDLINE | ID: mdl-36329674

Alzheimer's disease (AD), the most common cause of dementia, is a complex condition characterized by multiple pathophysiological mechanisms including amyloid-ß (Aß) plaque accumulation and neuroinflammation in the brain. The current immunotherapy approaches, such as anti-Aß monoclonal antibody (mAb) therapy, Aß vaccines, and adoptive regulatory T (Treg) cell transfer, target a single pathophysiological mechanism, which may lead to unsatisfactory therapeutic efficacy. Furthermore, Aß vaccines often induce T helper 1 (Th1) cell-mediated inflammatory responses. Here, a nanovaccine composed of lipid nanoparticles loaded with Aß peptides and rapamycin is developed, which targets multiple pathophysiological mechanisms, exhibits the combined effects of anti-Aß antibody therapy and adoptive Aß-specific Treg cell transfer, and can overcome the limitations of current immunotherapy approaches for AD. The Nanovaccine effectively delivers rapamycin and Aß peptides to dendritic cells, produces both anti-Aß antibodies and Aß-specific Treg cells, removes Aß plaques in the brain, alleviates neuroinflammation, prevents Th1 cell-mediated excessive immune responses, and inhibits cognitive impairment in mice. The nanovaccine shows higher efficacy in cognitive recovery than an Aß vaccine. Unlike anti-Aß mAb therapy and adoptive Treg cell transfer, both of which require complicated and costly manufacturing processes, the nanovaccine is easy-to-prepare and cost-effective. The nanovaccines can represent a novel treatment option for AD.


Alzheimer Disease , Vaccines , Mice , Animals , T-Lymphocytes, Regulatory , Neuroinflammatory Diseases , Mice, Transgenic , Amyloid beta-Peptides , Antibodies, Monoclonal , Disease Models, Animal
8.
Adv Exp Med Biol ; 1395: 357-362, 2022.
Article En | MEDLINE | ID: mdl-36527662

Previously, anti-CD3 antibodies delivered intravenously have been known for their negative side effects. The experimental conditions for optimal liquid production are derived from the Fc-directed conjugation of anti-CD3 foralumab antibodies and magnetic nanoparticles (Ab-MNPs). The anti-CD3 antibodies are prepared for conjugation with MNPs using SiteClick antibody labelling kits. The successful conjugation of the Ab-MNPs is confirmed using a transmission electron microscopy (TEM) image and an energy dispersive spectroscopy (EDS) analysis. The average values ​​of the moving speed of MNPs and Ab-MNPs in phosphate buffer saline (PBS) were + 3.16 pix/frame and + 6.70 pix/frame in the x-axis, respectively. This implies that MNPs with CD3 antibodies attached to the surface through biocompatible ligand functional groups has better fluidity in PBS. Afterwards, a non-clinical animal testing for the flow characteristics of Ab-MNPs inside a blood vessel is carried out to observe the effects of Ab-MNP delivery through intravenous injection.


Magnetite Nanoparticles , Animals , Magnetite Nanoparticles/chemistry , Magnetics , Microscopy, Electron, Transmission , Physical Phenomena , Antibodies, Monoclonal
9.
ACS Biomater Sci Eng ; 8(5): 1921-1929, 2022 05 09.
Article En | MEDLINE | ID: mdl-35416659

The vast majority of drug-eluting stents (DES) elute either sirolimus or one of its analogues. While limus drugs stymie vascular smooth muscle cell (VSMC) proliferation to prevent in-stent restenosis, their antiproliferative nature is indiscriminate and limits healing of the endothelium in stented vessels, increasing the risk of late-stent thrombosis. Oxidative stress, which is associated with vascular injury from stent implantation, can induce VSMCs to undergo senescence, and senescent VSMCs can produce pro-inflammatory cytokines capable of inducing proliferation of neighboring nonsenescent VSMCs. We explored the potential of senolytic therapy, which involves the selective elimination of senescent cells, in the form of a senolytic-eluting stent (SES) for interventional cardiology. Oxidative stress was modeled in vitro by exposing VSMCs to H2O2, and H2O2-mediated senescence was evaluated by cytochemical staining of senescence-associated ß-galactosidase activity and qRT-PCR. Quiescent VSMCs were then treated with the conditioned medium (CM) of H2O2-treated VSMCs. Proliferative effects of CM were analyzed by staining for proliferating cell nuclear antigen. Senolytic effects of the first-generation senolytic ABT263 were observed in vitro, and the effects of ABT263 on endothelial cells were also investigated through an in vitro re-endothelialization assay. SESs were prepared by dip coating. Iliofemoral arteries of hypercholesteremic rabbits were implanted with SES, everolimus-eluting stents (EESs), or bare-metal stents (BMSs), and the area of stenosis was measured 4 weeks post-implantation using optical coherence tomography. We found that a portion of H2O2-treated VSMCs underwent senescence, and that CM of H2O2-treated senescent VSMCs triggered the proliferation of quiescent VSMCs. ABT263 reverted H2O2-mediated senescence and the proliferative capacity of senescent VSMC CM. Unlike everolimus, ABT263 did not affect endothelial cell migration and/or proliferation. SES, but not EES, significantly reduced stenosis area in vivo compared with bare-metal stents (BMSs). This study shows the potential of SES as an alternative to current forms of DES.


Coronary Restenosis , Drug-Eluting Stents , Animals , Constriction, Pathologic , Coronary Restenosis/prevention & control , Drug-Eluting Stents/adverse effects , Endothelial Cells , Everolimus/pharmacology , Hydrogen Peroxide/pharmacology , Rabbits , Senotherapeutics , Stents
10.
Adv Mater ; 34(9): e2106516, 2022 Mar.
Article En | MEDLINE | ID: mdl-34962660

Despite the clinically proven efficacies of immune checkpoint blockades, including anti-cytotoxic T lymphocyte-associated protein 4 antibody (αCTLA-4), the low response rate and immune-related adverse events (irAEs) in cancer patients represent major drawbacks of the therapy. These drawbacks of αCTLA-4 therapy are mainly due to the suboptimal activation of tumor-specific cytotoxic T lymphocytes (CTLs) and the systemic nonspecific activation of T cells. To overcome such drawbacks, αCTLA-4 is delivered by dendritic cell-derived nanovesicles presenting tumor antigens (DCNV-TAs) that exclusively interact with tumor-specific T cells, leading to selective activation of tumor-specific CTLs. Compared to conventional αCTLA-4 therapy, treatment with αCTLA-4-conjugated DCNV-TAs significantly inhibits tumor growth and reduces irAEs in syngeneic tumor-bearing mice. This study demonstrates that the spatiotemporal presentation of both αCTLA-4 and tumor antigens enables selective activation of tumor-specific T cells and potentiates the antitumor efficacy of αCTLA-4 without inducing systemic irAEs.


Immune Checkpoint Inhibitors , Neoplasms , Animals , Antigens, Neoplasm , Humans , Immunotherapy , Mice , Neoplasms/drug therapy , T-Lymphocytes, Cytotoxic
11.
Adv Healthc Mater ; 11(2): e2101483, 2022 01.
Article En | MEDLINE | ID: mdl-34699690

Intervertebral disc (IVD) degeneration (IVDD) is a leading cause of chronic low back pain. There is a strong clinical demand for more effective treatments for IVDD as conventional treatments provide only symptomatic relief rather than arresting IVDD progression. This study shows that senolytic therapy with local drug delivery can inhibit IVDD and restore IVD integrity. ABT263, a senolytic drug, is loaded in poly(lactic-co-glycolic acid) nanoparticles (PLGA-ABT) and intradiscally administered into injury-induced IVDD rat models. The single intradiscal injection of PLGA-ABT may enable local delivery of the drug to avascular IVD, prevention of potential systemic toxicity caused by systemic administration of senolytic drug, and morbidity caused by repetitive injections of free drug into the IVD. The strategy results in the selective elimination of senescent cells from the degenerative IVD, reduces expressions of pro-inflammatory cytokines and matrix proteases in the IVD, inhibits progression of IVDD, and even restores the IVD structure. This study demonstrates for the first time that local delivery of senolytic drug can effectively treat senescence-associated IVDD. This approach can be extended to treat other types of senescence-associated degenerative diseases.


Intervertebral Disc Degeneration , Intervertebral Disc , Animals , Drug Delivery Systems , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc Degeneration/metabolism , Pharmaceutical Preparations , Rats , Senotherapeutics
12.
Article En | MEDLINE | ID: mdl-36612770

This study investigated the effects of a multi-access, metaverse-based early onset schizophrenia nursing simulation program based on Raskin and Rogers' person-centered therapy. The program's effectiveness was tested using a nonequivalent control group pre-test/post-test design. A quasi-experimental simulation study with both pre- and post-test designs was adopted. The experimental group (n = 29) used the simulation program, whereas the control group (n = 29) received only an online lecture on schizophrenia nursing. Changes in scores among experimental and control groups were compared using independent t-tests and analyses of covariance with PASW SPSS-WIN 27.0. Post-intervention, the knowledge regarding patients with early onset schizophrenia, critical thinking ability, and the ability to facilitate communication increased significantly in the experimental group compared with the control group. The nursing simulation program for children with early onset schizophrenia using a metaverse improved nursing students' knowledge, critical thinking ability, and ability to facilitate communication. This training method should be adapted without spatiotemporal constraints by partially supplementing clinical and simulation-based practice. In clinical nursing training, metaverse technical limitations should be identified, and training topics should be selected. Employing EduTech in a metaverse environment can provide clinical education to nurses in psychiatric wards and improve therapeutic communication with their psychiatric patients.


Education, Nursing, Baccalaureate , Schizophrenia , Students, Nursing , Child , Humans , Education, Nursing, Baccalaureate/methods , Clinical Competence , Thinking , Students, Nursing/psychology
13.
Adv Mater ; 33(43): e2103258, 2021 Oct.
Article En | MEDLINE | ID: mdl-34510559

Chimeric antigen receptor-T (CAR-T) cell immunotherapy has shown impressive clinical outcomes for hematologic malignancies. However, its broader applications are challenged due to its complex ex vivo cell-manufacturing procedures and low therapeutic efficacy against solid tumors. The limited therapeutic effects are partially due to limited CAR-T cell infiltration to solid tumors and inactivation of CAR-T cells by the immunosuppressive tumor microenvironment. Here, a facile approach is presented to in vivo program macrophages, which can intrinsically penetrate solid tumors, into CAR-M1 macrophages displaying enhanced cancer-directed phagocytosis and anti-tumor activity. In vivo injected nanocomplexes of macrophage-targeting nanocarriers and CAR-interferon-γ-encoding plasmid DNA induce CAR-M1 macrophages that are capable of CAR-mediated cancer phagocytosis, anti-tumor immunomodulation, and inhibition of solid tumor growth. Together, this study describes an off-the-shelf CAR-macrophage therapy that is effective for solid tumors and avoids the complex and costly processes of ex vivo CAR-cell manufacturing.


Receptors, Chimeric Antigen
14.
Small ; 17(32): e2101207, 2021 08.
Article En | MEDLINE | ID: mdl-34216428

Severe cardiac damage following myocardial infarction (MI) causes excessive inflammation, which sustains tissue damage and often induces adverse cardiac remodeling toward cardiac function impairment and heart failure. Timely resolution of post-MI inflammation may prevent cardiac remodeling and development of heart failure. Cell therapy approaches for MI are time-consuming and costly, and have shown marginal efficacy in clinical trials. Here, nanoparticles targeting the immune system to attenuate excessive inflammation in infarcted myocardium are presented. Liposomal nanoparticles loaded with MI antigens and rapamycin (L-Ag/R) enable effective induction of tolerogenic dendritic cells presenting the antigens and subsequent induction of antigen-specific regulatory T cells (Tregs). Impressively, intradermal injection of L-Ag/R into acute MI mice attenuates inflammation in the myocardium by inducing Tregs and an inflammatory-to-reparative macrophage polarization, inhibits adverse cardiac remodeling, and improves cardiac function. Nanoparticle-mediated blocking of excessive inflammation in infarcted myocardium may be an effective intervention to prevent the development of post-MI heart failure.


Heart Failure , Myocardial Infarction , Nanoparticles , Animals , Disease Models, Animal , Heart Failure/prevention & control , Inflammation , Macrophages , Mice , Mice, Inbred C57BL , Myocardial Infarction/complications , Myocardium
15.
Adv Mater ; 33(33): e2101110, 2021 Aug.
Article En | MEDLINE | ID: mdl-34235790

Although T-cell therapy is a remarkable breakthrough in cancer immunotherapy, the therapeutic efficacy is limited for solid tumors. A major cause of the low efficacy is T-cell exhaustion by immunosuppressive mechanisms of solid tumors, which are mainly mediated by programmed death-ligand 1 (PD-L1) and transforming growth factor-beta (TGF-ß). Herein, T-cell-derived nanovesicles (TCNVs) produced by the serial extrusion of cytotoxic T cells through membranes with micro-/nanosized pores that inhibit T-cell exhaustion and exhibit antitumoral activity maintained in the immunosuppressive tumor microenvironment (TME) are presented. TCNVs, which have programmed cell death protein 1 and TGF-ß receptor on their surface, block PD-L1 on cancer cells and scavenge TGF-ß in the immunosuppressive TME, thereby preventing cytotoxic-T-cell exhaustion. In addition, TCNVs directly kill cancer cells via granzyme B delivery. TCNVs successfully suppress tumor growth in syngeneic-solid-tumor-bearing mice. Taken together, TCNV offers an effective cancer immunotherapy strategy to overcome the tumor's immunosuppressive mechanisms.


Granzymes/chemistry , Immunosuppressive Agents/chemistry , Immunotherapy/methods , Nanocapsules/chemistry , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/chemistry , Animals , B7-H1 Antigen/metabolism , Cell Line, Tumor , Granzymes/metabolism , Humans , Immunosuppressive Agents/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Neoplasms, Experimental , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction , Tumor Microenvironment/drug effects
16.
Tissue Eng Regen Med ; 18(5): 807-818, 2021 10.
Article En | MEDLINE | ID: mdl-34251653

BACKGROUND: Various cell-culture systems have been used to evaluate drug toxicity in vitro. However, factors that affect cytotoxicity outcomes in drug toxicity evaluation systems remain elusive. In this study, we used multilayered sheets of cardiac-mimetic cells, which were reprogrammed from human fibroblasts, to investigate the effects of the layer number on drug cytotoxicity outcomes. METHODS: Cell sheets of cardiac-mimetic cells were fabricated by reprogramming of human fibroblasts into cardiac-mimetic cells via coculture with cardiac cells and electric stimulation, as previously described. Double-layered cell sheets were prepared by stacking the cell sheets. The mono- and double-layered cell sheets were treated with 5-fluorouracil (5-FU), an anticancer drug, in vitro. Subsequently, apoptosis and lipid peroxidation were analyzed. Furthermore, effects of cardiac-mimetic cell density on cytotoxicity outcomes were evaluated by culturing cells in monolayer at various cell densities. RESULTS: The double-layered cell sheets exhibited lower cytotoxicity in terms of apoptosis and lipid peroxidation than the mono-layered sheets at the same 5-FU dose. In addition, the double-layered cell sheets showed better preservation of mitochondrial function and plasma membrane integrity than the monolayer sheets. The lower cytotoxicity outcomes in the double-layered cell sheets may be due to the higher intercellular interactions, as the cytotoxicity of 5-FU decreased with cell density in monolayer cultures of cardiac-mimetic cells. CONCLUSION: The layer number of cardiac-mimetic cell sheets affects drug cytotoxicity outcomes in drug toxicity tests. The in vitro cellular configuration that more closely mimics the in vivo configuration in the evaluation systems seems to exhibit lower cytotoxicity in response to drug.


Heart , Pharmaceutical Preparations , Cells, Cultured , Coculture Techniques , Fibroblasts , Humans
17.
Tissue Eng Regen Med ; 18(5): 841-850, 2021 10.
Article En | MEDLINE | ID: mdl-34003467

BACKGROUND: Ferroptosis is an iron-dependent, non-apoptotic programmed cell death. Cellular senescence contributes to aging and various age-related diseases through the expression of a senescence-associated secretory phenotype (SASP). Senescent cells are often resistant to ferroptosis via increased ferritin and impaired ferritinophagy. In this study, we investigated whether treatment with JQ1 could remove senescent cells by inducing ferroptosis. METHODS: Senescence of human dermal fibroblasts was induced in vitro by treating the cells with bleomycin. The senolytic effects of JQ1 were evaluated using a SA-ß gal assay, annexin V analysis, cell counting kit-8 assay, and qRT-PCR. Ferroptosis following JQ1 treatment was evaluated with qRT-PCR and BODIPY staining. RESULTS: At a certain range of JQ1 concentrations, JQ1 treatment reduced the viability of bleomycin-treated cells (senescent cells) but did not reduce that of untreated cells (non-senescent cells), indicating that JQ1 treatment can selectively eliminate senescent cells. JQ1 treatment also decreased SASP expression only in senescent cells. Subsequently, JQ1 treatment reduced the expression of ferroptosis-resistance genes in senescent cells. JQ1 treatment induced lipid peroxidation in senescent cells but not in non-senescent cells. CONCLUSION: The data indicate that JQ1 can eliminate senescent cells via ferroptosis. This study suggests ferroptosis as a new mechanism of senolytic therapy.


Ferroptosis , Pharmaceutical Preparations , Aging , Cellular Senescence , Fibroblasts , Humans
18.
Adv Mater ; 32(39): e2003368, 2020 Oct.
Article En | MEDLINE | ID: mdl-32812291

Cancer immunotherapies, including adoptive T cell transfer and immune checkpoint blockades, have recently shown considerable success in cancer treatment. Nevertheless, transferred T cells often become exhausted because of the immunosuppressive tumor microenvironment. Immune checkpoint blockades, in contrast, can reinvigorate the exhausted T cells; however, the therapeutic efficacy is modest in 70-80% of patients. To address some of the challenges faced by the current cancer treatments, here T-cell-membrane-coated nanoparticles (TCMNPs) are developed for cancer immunotherapy. Similar to cytotoxic T cells, TCMNPs can be targeted at tumors via T-cell-membrane-originated proteins and kill cancer cells by releasing anticancer molecules and inducing Fas-ligand-mediated apoptosis. Unlike cytotoxic T cells, TCMNPs are resistant to immunosuppressive molecules (e.g., transforming growth factor-ß1 (TGF-ß1)) and programmed death-ligand 1 (PD-L1) of cancer cells by scavenging TGF-ß1 and PD-L1. Indeed, TCMNPs exhibit higher therapeutic efficacy than an immune checkpoint blockade in melanoma treatment. Furthermore, the anti-tumoral actions of TCMNPs are also demonstrated in the treatment of lung cancer in an antigen-nonspecific manner. Taken together, TCMNPs have a potential to improve the current cancer immunotherapy.


Biomimetic Materials/chemistry , Biomimetic Materials/therapeutic use , Immunotherapy/methods , Nanoparticles/therapeutic use , T-Lymphocytes/immunology , Cell Line, Tumor , Humans , Nanomedicine
19.
Sci Adv ; 6(18): eaaz0952, 2020 05.
Article En | MEDLINE | ID: mdl-32494669

Because of poor engraftment and safety concerns regarding mesenchymal stem cell (MSC) therapy, MSC-derived exosomes have emerged as an alternative cell-free therapy for myocardial infarction (MI). However, the diffusion of exosomes out of the infarcted heart following injection and the low productivity limit the potential of clinical applications. Here, we developed exosome-mimetic extracellular nanovesicles (NVs) derived from iron oxide nanoparticles (IONPs)-incorporated MSCs (IONP-MSCs). The retention of injected IONP-MSC-derived NVs (IONP-NVs) within the infarcted heart was markedly augmented by magnetic guidance. Furthermore, IONPs significantly increased the levels of therapeutic molecules in IONP-MSCs and IONP-NVs, which can reduce the concern of low exosome productivity. The injection of IONP-NVs into the infarcted heart and magnetic guidance induced an early shift from the inflammation phase to the reparative phase, reduced apoptosis and fibrosis, and enhanced angiogenesis and cardiac function recovery. This approach can enhance the therapeutic potency of an MSC-derived NV therapy.


Exosomes , Mesenchymal Stem Cells , Myocardial Infarction/therapy , Apoptosis , Exosomes/metabolism , Humans , Magnetic Iron Oxide Nanoparticles
20.
Orthopedics ; 43(4): e251-e257, 2020 Jul 01.
Article En | MEDLINE | ID: mdl-32271933

A considerable number of patients who underwent a 2-stage exchange protocol for periprosthetic hip joint infection could not complete the second-stage reimplantation. The aim of this study was to evaluate the results of unintended retention of temporary articulating spacers for the treatment of periprosthetic hip joint infection. Ninety-four patients with infection after total hip arthroplasty were treated by using a 2-stage exchange protocol with temporary articulating spacers. Of the 94 patients, 35 did not complete the 2-stage exchange protocol and retained spacers for more than 12 months. The authors retrospectively investigated the clinical and radiographic results after a mean follow-up of 36.1 months. Thirty-one patients had well-healed wounds without recurrent infection and did not receive further surgery for any reason (success group). Spacers were revised in 2 patients, and the other 2 patients underwent incision and debridement because of recurrent infection (failure group). There were no statistical differences between the 2 groups in terms of demographics or presence of resistant organisms. After 3 years of follow-up, temporary articulating spacers functioned well in 89% of the patients who retained them. These results support that retention of temporary articulating spacers could be considered an alternative treatment option for select patients. [Orthopedics. 2020;43(4):e251-e257.].


Arthritis, Infectious/surgery , Foreign Bodies/surgery , Hip Joint/surgery , Prosthesis-Related Infections/surgery , Reoperation/methods , Adult , Aged , Aged, 80 and over , Arthroplasty, Replacement, Hip/methods , Bone Cements/adverse effects , Female , Follow-Up Studies , Hip Prosthesis/adverse effects , Humans , Male , Middle Aged , Replantation/adverse effects , Retrospective Studies
...