Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Mater ; 35(49): e2303979, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37515819

RESUMO

Conventional approaches to developing therapeutic cancer vaccines that primarily activate tumor-specific T cells via dendritic cells (DCs) often demonstrate limited efficacy due to the suboptimal activation of these T cells. To address this limitation, here a therapeutic cancer nanovaccine is developed that enhances T cell responses by interacting with both DCs and T cells. The nanovaccine is based on a cancer cell membrane nanoparticle (CCM-MPLA) that utilizes monophosphoryl lipid A (MPLA) as an adjuvant. To allow direct interaction between the nanovaccine and tumor-specific T cells, anti-CD28 antibodies (aCD28) are conjugated onto CCM-MPLA, resulting in CCM-MPLA-aCD28. This nanovaccine activates tumor-specific CD8+ T cells in both the presence and absence of DCs. Compared with nanovaccines that interact with either DCs (CCM-MPLA) or T cells (CCM-aCD28), CCM-MPLA-aCD28 induces more potent responses of tumor-specific CD8+ T cells and exhibits a higher antitumor efficacy in tumor-bearing mice. No differences in T cell activation efficiency and therapeutic efficacy are observed between CCM-MPLA and CCM-aCD28. This approach may lead to the development of effective personalized therapeutic cancer vaccines prepared from autologous cancer cells.


Assuntos
Vacinas Anticâncer , Neoplasias , Animais , Camundongos , Linfócitos T CD8-Positivos , Vacinas Anticâncer/uso terapêutico , Células Dendríticas , Neoplasias/patologia , Imunoterapia/métodos
2.
Adv Mater ; 35(36): e2303080, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37249019

RESUMO

To demonstrate potent efficacy, a cancer vaccine needs to activate both innate and adaptive immune cells. Personalized cancer vaccine strategies often require the identification of patient-specific neoantigens; however, the clonal and mutational heterogeneity of cancer cells presents inherent challenges. Here, extracellular nanovesicles derived from alpha-galactosylceramide-conjugated autologous acute myeloid leukemia (AML) cells (ECNV-αGC) are presented as a personalized therapeutic vaccine that activates both innate and adaptive immune responses, bypassing the need to identify patient-specific neoantigens. ECNV-αGC vaccination directly engages with and activates both invariant natural killer T (iNKT) cells and leukemia-specific CD8+ T cells in mice with AML, thereby promoting long-term anti-leukemic immune memory. ECNV-αGC sufficiently serves as an antigen-presenting platform that can directly activate antigen-specific CD8+ T cells even in the absence of dendritic cells, thereby demonstrating a multifaceted cellular mechanism of immune activation. Moreover, ECNV-αGC vaccination results in a significantly lower AML burden and higher percentage of leukemia-free survivors among cytarabine-treated hosts with AML. Human AML-derived ECNV-αGCs activate iNKT cells in both healthy individuals and patients with AML regardless of responsiveness to conventional therapies. Together, autologous AML-derived ECNV-αGCs may be a promising personalized therapeutic vaccine that efficiently establishes AML-specific long-term immunity without requiring the identification of neoantigens.


Assuntos
Vacinas Anticâncer , Leucemia Mieloide Aguda , Células T Matadoras Naturais , Humanos , Animais , Camundongos , Linfócitos T CD8-Positivos , Ativação Linfocitária , Leucemia Mieloide Aguda/terapia
3.
Tissue Eng Regen Med ; 20(3): 389-409, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36920675

RESUMO

Various immune cells participate in repair and regeneration following tissue injury or damage, orchestrating tissue inflammation and regeneration processes. A deeper understanding of the immune system's involvement in tissue repair and regeneration is critical for the development of successful reparatory and regenerative strategies. Here we review recent technologies that facilitate cell-based and biomaterial-based modulation of the immune systems for tissue repair and regeneration. First, we summarize the roles of various types of immune cells in tissue repair. Second, we review the principle, examples, and limitations of regulatory T (Treg) cell-based therapy, a representative cell-based immunotherapy. Finally, we discuss biomaterial-based immunotherapy strategies that aim to modulate immune cells using various biomaterials for tissue repair and regeneration.


Assuntos
Imunidade , Regeneração , Materiais Biocompatíveis , Imunomodulação
4.
Exp Mol Med ; 55(3): 541-554, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36854774

RESUMO

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.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Vacinas Anticâncer/uso terapêutico , Neoplasias/tratamento farmacológico , Antígenos de Neoplasias , Adjuvantes Imunológicos
5.
Nano Lett ; 23(2): 476-490, 2023 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-36638236

RESUMO

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.


Assuntos
Doença de Alzheimer , Células-Tronco Mesenquimais , Doenças Neurodegenerativas , Geleia de Wharton , Humanos , Camundongos , Animais , Idoso , Doença de Alzheimer/terapia , Doença de Alzheimer/metabolismo , Nanopartículas Magnéticas de Óxido de Ferro , Diferenciação Celular
6.
Adv Mater ; 35(3): e2207719, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36329674

RESUMO

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.


Assuntos
Doença de Alzheimer , Vacinas , Camundongos , Animais , Linfócitos T Reguladores , Doenças Neuroinflamatórias , Camundongos Transgênicos , Peptídeos beta-Amiloides , Anticorpos Monoclonais , Modelos Animais de Doenças
8.
ACS Biomater Sci Eng ; 8(5): 1921-1929, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35416659

RESUMO

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.


Assuntos
Reestenose Coronária , Stents Farmacológicos , Animais , Constrição Patológica , Reestenose Coronária/prevenção & controle , Stents Farmacológicos/efeitos adversos , Células Endoteliais , Everolimo/farmacologia , Peróxido de Hidrogênio/farmacologia , Coelhos , Senoterapia , Stents
9.
Metab Syndr Relat Disord ; 20(4): 210-218, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35100057

RESUMO

Background: The role of serum 25-hydroxyvitamin D [25(OH)D] levels in the development of sarcopenia in non-alcoholic fatty liver disease (NAFLD) remains controversial. We investigated the association between vitamin D levels, occurrence of sarcopenia, NAFLD, and sarcopenia in NAFLD in adults aged >50 years. Methods: This study used data pertaining to 5396 adults aged >50 years (1870 men and 3526 women) from the 2008-2011 Korea National Health and Nutrition Examination Survey. Appendicular skeletal muscle mass adjusted by weight (ASM/Wt) was used to diagnose sarcopenia, and NAFLD was diagnosed using the NAFLD fat score. Results: The lowest quintile of serum 25(OH)D level (4.85-15.26 ng/mL) was associated with an increased occurrence of sarcopenia [odds ratio (OR) 2.65; 95% confidence interval (CI) 1.64-4.27], NAFLD (OR 1.82; 95% CI 1.19-2.96), and sarcopenia in NAFLD (OR 2.25; 95% CI 1.26-4.03) in men. In women, sarcopenia (OR 1.80; 95% CI 1.29-2.51) was also significantly associated with serum vitamin D levels, whereas high levels of vitamin D were not significantly related to NAFLD. Conclusions: Serum vitamin D levels are associated with sarcopenia, NAFLD, and sarcopenia in NAFLD. Vitamin D level can be a useful marker of sarcopenia and NAFLD, especially in men.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Sarcopenia , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/diagnóstico , Hepatopatia Gordurosa não Alcoólica/epidemiologia , Inquéritos Nutricionais , Razão de Chances , República da Coreia/epidemiologia , Sarcopenia/complicações , Sarcopenia/diagnóstico , Sarcopenia/epidemiologia , Vitamina D
10.
Adv Mater ; 34(9): e2106516, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34962660

RESUMO

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.


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Animais , Antígenos de Neoplasias , Humanos , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico , Linfócitos T Citotóxicos
11.
Commun Biol ; 4(1): 1405, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34916605

RESUMO

Though various transgene expression switches have been adopted in a wide variety of organisms for basic and biomedical research, intrinsic obstacles of those existing systems, including toxicity and silencing, have been limiting their use in vertebrate transgenesis. Here we demonstrate a novel QF-based binary transgene switch (IQ-Switch) that is relatively free of driver toxicity and transgene silencing, and exhibits potent and highly tunable transgene activation by the chemical inducer tebufenozide, a non-toxic lipophilic molecule to developing zebrafish with negligible background. The interchangeable IQ-Switch makes it possible to elicit ubiquitous and tissue specific transgene expression in a spatiotemporal manner. We generated a RASopathy disease model using IQ-Switch and demonstrated that the RASopathy symptoms were ameliorated by the specific BRAF(V600E) inhibitor vemurafenib, validating the therapeutic use of the gene switch. The orthogonal IQ-Switch provides a state-of-the-art platform for flexible regulation of transgene expression in zebrafish, potentially applicable in cell-based systems and other model organisms.


Assuntos
Animais Geneticamente Modificados/genética , Técnicas de Transferência de Genes , Genes de Troca , Transgenes , Peixe-Zebra/genética , Animais
12.
Adv Mater ; 33(43): e2103258, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34510559

RESUMO

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.


Assuntos
Receptores de Antígenos Quiméricos
13.
Adv Mater ; 33(33): e2101110, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34235790

RESUMO

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.


Assuntos
Granzimas/química , Imunossupressores/química , Imunoterapia/métodos , Nanocápsulas/química , Neoplasias/terapia , Linfócitos T Citotóxicos/química , Animais , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Granzimas/metabolismo , Humanos , Imunossupressores/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Neoplasias Experimentais , Receptor de Morte Celular Programada 1/metabolismo , Transdução de Sinais , Microambiente Tumoral/efeitos dos fármacos
14.
Metab Syndr Relat Disord ; 19(8): 452-459, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34255575

RESUMO

Background: The association between sarcopenia and protein intake has been well studied. However, limited data are available on the association between sarcopenia and protein intake in people with nonalcoholic fatty liver disease (NAFLD). This study aimed to investigate the association between protein intake and sarcopenia among elderly participants with NAFLD using the Korea National Health and Nutrition Examination Survey (KNHANES). Methods: Data of 4179 participants (1576 men and 2603 women, age ≥60 years) who participated in the KNHANES during 2008-2011 were obtained. Sarcopenia was defined as appendicular skeletal muscle mass/wt (%) of 1 standard deviation below the gender-specific mean for healthy adults and NAFLD as liver fat score using the fatty liver prediction models. According to their daily protein intake, participants were grouped into the high protein intake group (>1.2 g/kg/day), middle protein intake group (0.8-1.2 g/kg/day), and low protein intake group (<0.8 g/kg/day). Generalized linear models and logistic regression models were used. Results: The overall prevalence of sarcopenia in participants with NAFLD was 16.53%. Compared with the highest protein intake group, the low protein intake group had a significantly higher risk of sarcopenia (adjustment odds ratio = 1.707; 95% confidence interval = 1.009-2.886). In the fully adjusted model, the highest protein intake group had significantly lower levels of insulin, total body fat, vitamin B, and fat intake, whereas muscle proportions, energy, carbohydrates, vitamin A, and vitamin C intake of high protein intake participants were significantly higher than other groups. Conclusion: The prevalence of sarcopenia and sarcopenia-related factors were significantly lower in NAFLD elderly participants with high protein intake. These results suggest that high protein intake can help prevent and manage sarcopenia in people with NAFLD.


Assuntos
Proteínas Alimentares , Hepatopatia Gordurosa não Alcoólica , Sarcopenia , Idoso , Comorbidade , Ingestão de Alimentos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/epidemiologia , Inquéritos Nutricionais/estatística & dados numéricos , Estado Nutricional , República da Coreia/epidemiologia , Sarcopenia/complicações , Sarcopenia/diagnóstico , Sarcopenia/epidemiologia
15.
Tissue Eng Regen Med ; 18(5): 807-818, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34251653

RESUMO

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.


Assuntos
Coração , Preparações Farmacêuticas , Células Cultivadas , Técnicas de Cocultura , Fibroblastos , Humanos
16.
Adv Mater ; 32(39): e2003368, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32812291

RESUMO

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.


Assuntos
Materiais Biomiméticos/química , Materiais Biomiméticos/uso terapêutico , Imunoterapia/métodos , Nanopartículas/uso terapêutico , Linfócitos T/imunologia , Linhagem Celular Tumoral , Humanos , Nanomedicina
17.
Adv Healthc Mater ; 9(5): e1901612, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31977158

RESUMO

Osteoarthritis (OA) is a painful intractable disease that significantly affects patients' quality of life. However, current therapies, such as pain killers and joint replacement surgery, do not lead to cartilage protection. Mesenchymal stem cells (MSCs) have been proposed as an alternative strategy for OA therapy because MSCs can secrete chondroprotective and anti-inflammatory factors. However, interleukin-4 (IL-4), a potent anti-inflammatory cytokine, is barely produced by MSCs, and MSC therapy suffers from rapid MSC death following intra-articular implantation. MSCs in spheroids survive better than naïve MSCs in vitro and in vivo. IL-4-transfected MSCs in spheroids (IL-4 MSC spheroid) show increased chondroprotective and anti-inflammatory effects in an OA chondrocyte model in vitro. Following intra-articular implantation in OA rats, IL-4 MSC spheroids show better cartilage protection and pain relief than naïve MSCs. Thus, IL-4 MSC spheroid may potentiate the therapeutic efficacy of MSCs for OA.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Osteoartrite , Animais , Humanos , Injeções Intra-Articulares , Interleucina-4 , Osteoartrite/terapia , Qualidade de Vida , Ratos , Transfecção
18.
Theranostics ; 9(23): 6734-6744, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31660065

RESUMO

Rationale: Cardiovascular diseases often cause substantial heart damage and even heart failure due to the limited regenerative capacity of adult cardiomyocytes. The direct cardiac reprogramming of fibroblasts could be a promising therapeutic option for these patients. Although exogenous transcriptional factors can induce direct cardiac reprogramming, the reprogramming efficiency is too low to be used clinically. Herein, we introduce a cardiac-mimetic cell-culture system that resembles the microenvironment in the heart and provides interactions with cardiomyocytes and electrical cues to the cultured fibroblasts for direct cardiac reprogramming. Methods: Nano-thin and nano-porous membranes and heart like electric stimulus were used in the cardiac-mimetic cell-culture system. The human neonatal dermal fibroblasts containing cardiac transcription factors were plated on the membrane and cultured with the murine cardiomyocyte in the presence of the electric stimulus. The reprogramming efficiency was evaluated by qRT-PCR and immunocytochemistry. Results: Nano-thin and nano-porous membranes in the culture system facilitated interactions between fibroblasts and cardiomyocytes in coculture. The cellular interactions and electric stimulation supplied by the culture system dramatically enhanced the cardiac reprogramming efficiency of cardiac-specific transcriptional factor-transfected fibroblasts. Conclusion: The cardiac-mimetic culture system may serve as an effective tool for producing a feasible number of reprogrammed cardiomyocytes from fibroblasts.


Assuntos
Biomimética/métodos , Técnicas de Reprogramação Celular/métodos , Miócitos Cardíacos/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Comunicação Celular , Transdiferenciação Celular , Células Cultivadas , Técnicas de Cocultura/métodos , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Humanos , Recém-Nascido , Masculino , Potenciais da Membrana , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
19.
Nano Lett ; 19(8): 5185-5193, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31298024

RESUMO

Liposomes are clinically used as drug carriers for cancer therapy; however, unwanted leakage of the encapsulated anticancer drug and poor tumor-targeting efficiency of liposomes may generate toxic side effects on healthy cells and lead to failure of tumor eradication. To overcome these limitations, we functionalized liposomes with a photosensitizer (KillerRed, KR)-embedded cancer cell membrane (CCM). A lipid adjuvant was also embedded in the lipocomplex to promote the anticancer immune response. KR proteins were expressed on CCM and did not leak from the lipocomplex. Owing to the homotypic affinity of the CCM for the source cancer cells, the lipocomplex exhibited a 3.3-fold higher cancer-targeting efficiency in vivo than a control liposome. The liposome functionalized with KR-embedded CCM and lipid adjuvant generated cytotoxic reactive oxygen species in photodynamic therapy and effectively induced anticancer immune responses, inhibiting primary tumor growth and lung metastasis in homotypic tumor-bearing mice. Taken together, the lipocomplex technology may improve liposome-based cancer therapy.


Assuntos
Fatores Imunológicos/uso terapêutico , Lipossomos/uso terapêutico , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Animais , Linhagem Celular Tumoral , Membrana Celular/patologia , Proteínas de Fluorescência Verde/uso terapêutico , Humanos , Camundongos , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/patologia , Neoplasias/patologia
20.
Adv Exp Med Biol ; 1064: 221-233, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30471036

RESUMO

Regulatory T-cells (Treg cells) are a specific group of T-cells that maintain immune homeostasis by counteracting the immune responses of conventional T-cells. So far, the therapeutic applications of Treg cells have focused on the treatment of autoimmune diseases, as depletion of Treg cells or Treg-related genes is known to cause autoimmune defects. However, Treg cells can be a potential solution for tissue repair as they can terminate the pro-inflammatory phase and initiate the anti-inflammatory or regenerative phase at the tissue injury site. This review summarizes the known characteristics of Treg cells and lists examples of their therapeutic applications. The use of Treg cells in the treatment of myocardial infarctions, skeletal muscle injuries, and ischemia injuries has revealed their potential as a promising tissue repair method. We have also discussed the limitations and scope of Treg cells in tissue repair.


Assuntos
Doenças Autoimunes/terapia , Terapia Baseada em Transplante de Células e Tecidos , Linfócitos T Reguladores/citologia , Humanos , Isquemia/terapia , Músculo Esquelético/lesões , Infarto do Miocárdio/terapia , Regeneração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...