Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Biomaterials ; 312: 122755, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39151270

RESUMEN

Copper-catalyzed click chemistry offers creative strategies for activation of therapeutics without disrupting biological processes. Despite tremendous efforts, current copper catalysts face fundamental challenges in achieving high efficiency, atom economy, and tissue-specific selectivity. Herein, we develop a facile "mix-and-match synthetic strategy" to fabricate a biomimetic single-site copper-bipyridine-based cerium metal-organic framework (Cu/Ce-MOF@M) for efficient and tumor cell-specific bioorthogonal catalysis. This elegant methodology achieves isolated single-Cu-site within the MOF architecture, resulting in exceptionally high catalytic performance. Cu/Ce-MOF@M favors a 32.1-fold higher catalytic activity than the widely used MOF-supported copper nanoparticles at single-particle level, as first evidenced by single-molecule fluorescence microscopy. Furthermore, with cancer cell-membrane camouflage, Cu/Ce-MOF@M demonstrates preferential tropism for its parent cells. Simultaneously, the single-site CuII species within Cu/Ce-MOF@M are reduced by upregulated glutathione in cancerous cells to CuI for catalyzing the click reaction, enabling homotypic cancer cell-activated in situ drug synthesis. Additionally, Cu/Ce-MOF@M exhibits oxidase and peroxidase mimicking activities, further enhancing catalytic cancer therapy. This study guides the reasonable design of highly active heterogeneous transition-metal catalysts for targeted bioorthogonal reactions.

2.
Biomacromolecules ; 25(7): 4358-4373, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38924782

RESUMEN

Chitosan (CS)-based photo-cross-linkable hydrogels have gained increasing attention in biomedical applications. In this study, we grafted CS with gallic acid (GA) by carbodiimide chemistry to prepare the GA-CS conjugate, which was subsequently modified with methacrylic anhydride (MA) modification to obtain the methacrylated GA-CS conjugate (GA-CS-MA). Our results demonstrated that the GA-CS-MA hydrogel not only exhibited improved physicochemical properties but also showed antibacterial, antioxidative, and anti-inflammatory capacity. It showed moderate antibacterial activity and especially showed a more powerful inhibitory effect against Gram-positive bacteria. It modulated macrophage polarization, downregulated pro-inflammatory gene expression, upregulated anti-inflammatory gene expression, and significantly reduced reactive oxygen species (ROS) and nitric oxide (NO) production under lipopolysaccharide (LPS) stimulation. Subcutaneously implanted GA-CS-MA hydrogels induced significantly lower inflammatory responses, as evidenced by less inflammatory cell infiltration, thinner fibrous capsule, and predominately promoted M2 polarization. This study provides a feasible strategy to prepare CS-based photo-cross-linkable hydrogels with improved physicochemical properties for biomedical applications.


Asunto(s)
Antibacterianos , Antiinflamatorios , Antioxidantes , Quitosano , Ácido Gálico , Hidrogeles , Metacrilatos , Quitosano/química , Ácido Gálico/química , Ácido Gálico/farmacología , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/síntesis química , Animales , Hidrogeles/química , Hidrogeles/farmacología , Hidrogeles/síntesis química , Ratones , Antioxidantes/química , Antioxidantes/farmacología , Antioxidantes/síntesis química , Metacrilatos/química , Antiinflamatorios/farmacología , Antiinflamatorios/química , Células RAW 264.7 , Reactivos de Enlaces Cruzados/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Óxido Nítrico/metabolismo
3.
J Biomed Mater Res A ; 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38837524

RESUMEN

Methacrylated biopolymers are unique and attractive in preparing photocrosslinkable hydrogels in biomedical applications. Here we report a novel chitosan (CS) derivative-based injectable hydrogel with anti-inflammatory capacity via methacrylation modification. First, ibuprofen (IBU) was conjugated to the backbone of CS by carbodiimide chemistry to obtain IBU-CS conjugate, which converts water-insoluble unmodified CS into water-soluble IBU-CS conjugate. The IBU-CS conjugate did not precipitate at the pH of 7, which was beneficial to subsequent chemical modification with methacrylic anhydride to prepare IBU-CS methacrylate (IBU-CS-MA) with significantly higher methacrylation substitution. Photocrosslinkable in situ gel formation of injectable IBU-CS-MA hydrogel was verified using lithium phenyl-2,4,6-trimethylbenzoylphosphinate (LAP) initiator under visible light. The IBU-CS-MA hydrogel showed good cytocompatibility as revealed by encapsulating and in vitro culturing murine fibroblasts within hydrogels. It promoted macrophage polarization toward M2 phenotype, as well as downregulated pro-inflammatory gene expression and upregulated anti-inflammatory gene expression of macrophages. The hydrogel also significantly reduced the reactive oxygen specifies (ROS) and nitrogen oxide (NO) produced by lipopolysaccharides (LPS)-stimulated macrophages. Upon subcutaneous implantation in a rat model, it significantly mitigated inflammatory responses as shown by significantly lower inflammatory cell density, less cell infiltration, and much thinner fibrous capsule compared with CS methacrylate (CS-MA) hydrogel. This study suggests that IBU-CS conjugate represents a feasible strategy for preparing CS-based methacrylate hydrogels for biomedical applications.

4.
J Control Release ; 370: 528-542, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38705520

RESUMEN

Reversing the aggravated immunosuppression hence overgrowth of colorectal cancer (CRC) caused by the gut inflammation and microbiota dysbiosis is pivotal for effective CRC therapy and metastasis inhibition. However, the low delivery efficiency and severe dose-limiting off-target toxicities caused by unsatisfied drug delivery systems remain the major obstacles in precisely modulating gut inflammation and microbiota in CRC therapy. Herein, a multifunctional oral dextran-aspirin nanomedicine (P3C-Asp) was utilized for oral treatment of primary CRC, as it could release salicylic acid (SA) while scavenging reactive oxygen species (ROS) and held great potential in modulating gut microbiota with prebiotic (dextran). Oral P3C-Asp retained in CRC tissues for over 12 h and significantly increased SA accumulation in CRC tissues over free aspirin (10.8-fold at 24 h). The enhanced SA accumulation and ROS scavenging of P3C-Asp cooperatively induced more potent inflammation relief over free aspirin, characterized as lower level of cyclooxygenase-2 and immunosuppressive cytokines. Remarkably, P3C-Asp promoted the microbiota homeostasis and notably increased the relative abundance of strengthening systemic anti-cancer immune response associated microbiota, especially lactobacillus and Akkermansia to 6.66- and 103- fold over the control group. Additionally, a demonstrable reduction in pathogens associated microbiota (among 96% to 79%) including Bacteroides could be detected. In line with our findings, inflammation relief along with enhanced abundance of lactobacillus was positively correlated with CRC inhibition. In primary CRC model, P3C-Asp achieved 2.1-fold tumor suppression rate over free aspirin, with an overall tumor suppression rate of 85%. Moreover, P3C-Asp cooperated with αPD-L1 further reduced the tumor weight of each mouse and extended the median survival of mice by 29 days over αPD-L1 alone. This study unravels the synergistic effect of gut inflammation and microbiota modulation in primary CRC treatment, and unlocks an unconventional route for immune regulation in TME with oral nanomedicine.


Asunto(s)
Aspirina , Neoplasias Colorrectales , Dextranos , Microbioma Gastrointestinal , Homeostasis , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Aspirina/administración & dosificación , Aspirina/uso terapéutico , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Homeostasis/efectos de los fármacos , Administración Oral , Dextranos/administración & dosificación , Dextranos/química , Nanomedicina , Ratones Endogámicos BALB C , Inflamación/tratamiento farmacológico , Masculino , Ratones , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/uso terapéutico , Nanopartículas/administración & dosificación , Línea Celular Tumoral , Femenino
5.
Nano Lett ; 24(9): 2921-2930, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38411094

RESUMEN

Immune checkpoint inhibitor (ICI) therapy is effectively employed in treating various malignancies. However, the response rate is constrained to 5-30%, which is attributed to differences in immune responses across different tumors. Overcoming all obstacles of multistep immune activation with monotherapy is difficult. Here, maleimide-modified resiquimod (R848) prodrug nanoparticles (MAL-NPs) are reported and combined with radiotherapy (RT) and anti-PD1 to enhance ICI therapy. MAL-NPs can promote antigen endocytosis by dendritic cells and are radio-reduced to produce R848. When combined with RT, MAL-NPs can augment the concentration of nanoparticles at tumor sites and be selectively radio-reduced within the tumor, thereby triggering a potent antitumor immune response. The systemic immune response and long-term memory efficacy induced by MAL-NPs + RT + anti-PD1 significantly inhibit the abscopal tumor growth and prevent tumor recurrence. This strategy can achieve systemic therapy through selective training of the tumor immune microenvironment, offering a new approach to overcome the obstacles of ICI therapy.


Asunto(s)
Nanoestructuras , Neoplasias , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Imidazoles/farmacología , Imidazoles/uso terapéutico , Microambiente Tumoral , Línea Celular Tumoral , Inmunoterapia
6.
ACS Appl Mater Interfaces ; 16(7): 8484-8498, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38334265

RESUMEN

Malignant tumors are still one of the most deadly diseases that threaten human life and health. However, developing new drugs is challenging due to lengthy trials, funding constraints, and regulatory approval procedures. Consequently, researchers have devoted themselves to transforming some clinically approved old drugs into antitumor drugs with certain active ingredients, which have become an attractive alternative. Disulfiram (DSF), an antialcohol medication, can rapidly metabolize in the physiological environment into diethyldithiocarbamate (DTC) which can readily react with Cu2+ ions in situ to form the highly toxic bis(N,N-diethyldithiocarbamate)-copper(II) (CuET) complex. In this study, DSF is loaded into mesoporous dopamine nanocarriers and surface-chelated with tannin and Cu2+ to construct M-MDTC nanoprodrugs under the camouflage of K7 tumor cell membranes. After intravenous injection, M-MDTC nanoprodrugs successfully reach the tumor sites with the help of mediated cell membranes. Under slightly acidic pH and photothermal stimulation conditions, DSF and Cu2+ are simultaneously released, forming a highly toxic CuET to kill tumor cells in situ. The generated CuET can also induce immunogenic cell death of tumor cells, increase the proportion of CD86+ CD80+ cells, and promote dendritic cell maturation. In vitro and in vivo studies of M-MDTC nanoprodrugs have shown excellent tumor-cell-killing ability and solid tumor suppression. This approach enables in situ amplification of chemotherapy in the tumor microenvironment, achieving an effective antitumor treatment.


Asunto(s)
Cadaverina/análogos & derivados , Cobre , Neoplasias , Humanos , Línea Celular Tumoral , Cobre/farmacología , Cobre/uso terapéutico , Microambiente Tumoral , Biomimética , Disulfiram/farmacología , Ditiocarba/farmacología , Ditiocarba/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología
7.
Biomater Sci ; 12(9): 2203-2228, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38293828

RESUMEN

Unmethylated cytosine-phosphate-guanine (CpG) oligodeoxynucleotides (ODNs), which were therapeutic DNA with high immunostimulatory activity, have been applied in widespread applications from basic research to clinics as therapeutic agents for cancer immunotherapy, viral infection, allergic diseases and asthma since their discovery in 1995. The major factors to consider for clinical translation using CpG motifs are the protection of CpG ODNs from DNase degradation and the delivery of CpG ODNs to the Toll-like receptor-9 expressed human B-cells and plasmacytoid dendritic cells. Therefore, great efforts have been devoted to the advances of efficient delivery systems for CpG ODNs. In this review, we outline new horizons and recent developments in this field, providing a comprehensive summary of the nanoparticle-based CpG delivery systems developed to improve the efficacy of CpG-mediated immune responses, including DNA nanostructures, inorganic nanoparticles, polymer nanoparticles, metal-organic-frameworks, lipid-based nanosystems, proteins and peptides, as well as exosomes and cell membrane nanoparticles. Moreover, future challenges in the establishment of CpG delivery systems for immunotherapeutic applications are discussed. We expect that the continuously growing interest in the development of CpG-based immunotherapy will certainly fuel the excitement and stimulation in medicine research.


Asunto(s)
Nanopartículas , Oligodesoxirribonucleótidos , Humanos , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/administración & dosificación , Nanopartículas/química , Animales , Inmunoterapia/métodos , Receptor Toll-Like 9/metabolismo , Sistemas de Liberación de Medicamentos
8.
Int J Biol Macromol ; 259(Pt 2): 129283, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38199538

RESUMEN

Diabetic wounds, resulting from skin atrophy due to localized ischemia and hypoxia in diabetic patients, lead to chronic pathological inflammation and delayed healing. Using electrospinning technology, we developed magnesium ion-chelated nanofiber membranes to explore their efficacy in antibacterial, anti-inflammatory, and angiogenic applications for wound healing. These membranes are flexible and elastic, resembling native skin tissue, and possess good hydrophilicity for comfortable wound bed contact. The mechanical properties of nanofiber membranes are enhanced by the chelation of magnesium ions (Mg2+), which also facilitates a long-term slow release of Mg2+. The cytocompatibility of the nanofibrous membranes is influenced by their Mg2+ content: lower levels encourage the proliferation of fibroblasts, endothelial cells, and macrophages, while higher levels are inhibitory. In a diabetic rat model, magnesium ion-chelated nanofibrous membranes effectively reduced early wound inflammation and notably accelerated wound healing. This study highlights the potential of magnesium ion-chelated nanofiber membranes in treating diabetic wounds.


Asunto(s)
Diabetes Mellitus , Nanofibras , Humanos , Ratas , Animales , Magnesio/farmacología , Células Endoteliales/patología , Cicatrización de Heridas , Diabetes Mellitus/patología , Inflamación
9.
Nano Lett ; 23(10): 4191-4200, 2023 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-37186944

RESUMEN

Multispecific antibodies (MsAbs) maintain the specificity of versatile antibodies while simultaneously addressing different epitopes for a cumulative, collaborative effect. They could be an alternative treatment to chimeric antigen receptor-T cell therapy by helping to redirect T cells to tumors in vivo. However, one major limitation of their development is their relatively complex production process, which involves performance of a massive screen with low yield, inconsistent quality, and nonnegligible impurities. Here, a poly(l-glutamic acid)-conjugated multiple Fc binding peptide-based synthesis nanoplatform was proposed, in which MsAbs were constructed by mixing the desired monoclonal antibodies (mAbs) with polymeric Fc binding peptides in aqueous solution without purification. To determine its efficacy, a dual immune checkpoint-based PD1/OX40 bispecific antibody and PDL1/CD3e/4-1BB trispecific antibody-based T cell engager were generated to trigger antitumor CD8+ T responses in mice, showing superior tumor suppression over free mixed mAbs. In this study, a facile, versatile build platform for MsAbs was established.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Animales , Ratones , Neoplasias/terapia , Anticuerpos Monoclonales , Linfocitos T , Inmunoterapia Adoptiva
10.
Adv Mater ; 35(29): e2207733, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37086177

RESUMEN

Resiquimod (R848) is an immunomodulator that causes a severe systemic inflammatory reaction due to low tumor selectivity, thus hindering its use in cancer therapy. Therefore, an azide-masked prodrug (R848-N3 ) of R848 is developed, which is selectively activated to R848 in hypoxic tumors. R848-N3 significantly reduces pro-inflammatory cytokines in treated mice to 1/12 compared to R848 at the same dose. In addition, combretastatin A4 nanoparticles (CA4-NPs) are used to enhance the tumor selectivity of R848-N3 by elevating the level of tumor hypoxia. R848-N3 +CA4-NPs has higher tumor selectivity than the intratumoral injection of R848 at 1 h after administration. The concentration of the active R848 in the tumor is 21.45-fold that in the heart. Benefiting from the high tumor selectivity of R848-N3 , R848-N3 +CA4-NPs+anti-PD1 exerted 94.1% tumor suppression and 40.0% tumor cure. Therefore, this work highlights the potential of azide-masking strategy in the development of tumor-selective prodrugs with reduced toxicity.


Asunto(s)
Nanopartículas , Neoplasias , Profármacos , Ratones , Animales , Azidas , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Imidazoles/farmacología , Imidazoles/uso terapéutico , Profármacos/farmacología , Profármacos/uso terapéutico , Hipoxia
11.
Nano Lett ; 22(8): 3328-3339, 2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35404605

RESUMEN

Nanomedicines are highly promising for cancer therapy due to their minimal side effects. However, little is known regarding their host immune response, which may limit their clinical efficacy and applications. Here, we find that cisplatin (CDDP)-loaded poly(l-glutamic acid)-graft-methoxy poly(ethylene glycol) complex nanoparticles (CDDP-NPs) elicit a strong antitumor CD8+ T cell-mediated immune response in a tumor-bearing mouse model compared to free CDDP. Mechanistically, the sustained retention of CDDP-NPs results in persistent tumor MHC-I overexpression, which promotes the formation of MHC-I-antigen peptide complex (pMHC-I), enhances the interaction between pMHC-I and T cell receptor (TCR), and leads to the activation of TCR signaling pathway and CD8+ T cell-mediated immune response. Furthermore, CDDP-NPs upregulate the costimulatory OX40 on intratumoral CD8+ T cells, and synergize with the agonistic OX40 antibody (aOX40) to suppress tumor growth by 89.2%. Our study provides a basis for the efficacy advantage of CDDP-based nanomedicines and immunotherapy.


Asunto(s)
Cisplatino , Nanopartículas , Animales , Presentación de Antígeno , Linfocitos T CD8-positivos , Línea Celular Tumoral , Cisplatino/farmacología , Cisplatino/uso terapéutico , Ratones , Receptores de Antígenos de Linfocitos T
12.
Adv Mater ; 34(10): e2109254, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34984753

RESUMEN

In recent years, significant evolutions have been made in applying nanotechnologies for prophylactic and therapeutic cancer vaccine design. However, the clinical translation of nanovaccines is still limited owing to their complicated compositions and difficulties in the spatiotemporal coordination of antigen-presenting cell activation and antigen cross-presentation. Herein, a minimalist binary nanovaccine (BiVax) is designed that integrates innate stimulating activity into the carrier to elicit robust antitumor immunity. The authors started by making a series of azole molecules end-capped polyethylenimine (PEI-M), and were surprised to find that over 60% of the PEI-M polymers have innate stimulating activity via activation of the stimulator of interferon genes pathway. PEI-4BImi, a PEI-M obtained from a series of polymers, elicits robust antitumor immune responses when used as a subcutaneously injected nanovaccine by simply mixing with ovalbumin antigens, and this BiVax system performs much better than the traditional ternary vaccine system, as well as, commercialized aluminum-containing adjuvants. This system also enables the fast preparation of personalized BiVax by compositing PEI-4BImi with autologous tumor cell membrane protein antigens, and a 60% postoperative cure rate is observed when combined with immune checkpoint inhibitors.


Asunto(s)
Vacunas contra el Cáncer , Nanopartículas , Neoplasias , Animales , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia
13.
Biomater Sci ; 9(20): 6879-6888, 2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34505857

RESUMEN

Cancer vaccines artificially stimulate the immune system against cancer and are considered the most promising treatment of cancer. However, the current progress in vaccine research against cancer is still limited and slow, partially due to the difficulties in identifying and obtaining tumor-specific antigens. Considering surgery as the first choice for tumor treatment in most cases, the authors evaluated whether the resected tumor can be directly used as a source of tumor antigens for designing personalized cancer vaccines. Based on this idea, herein, the authors report a dynamic covalent hydrogel-based vaccine (DCHVax) for personalized postsurgical management of tumors. The study uses proteins extracted from the resected tumor as antigens, CpG as the adjuvant, and a multi-armed poly(ethylene glycol) (8-arm PEG)/oxidized dextran (ODEX) dynamically cross-linked hydrogel as the matrix. Subcutaneous injection of DCHVax recruits dendritic cells to the matrix in situ and elicits robust tumor-specific immune responses. Thus, it effectively inhibits the postoperative growth of the residual tumor in several murine tumor models. This simple and personalized method to develop cancer vaccines may be promising in developing clinically relevant strategies for postoperative cancer treatment.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Adyuvantes Inmunológicos , Animales , Antígenos de Neoplasias , Hidrogeles , Ratones , Neoplasias/tratamiento farmacológico
14.
Front Oncol ; 11: 632976, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33816273

RESUMEN

MicroRNAs (miRNAs) participated in the regulation of tumorigenesis, progression, metastasis, recurrence and chemo-resistance of cancers. However, the potential function of miRNAs in cancer stem cells (CSCs) or tumor-initiating cells (T-ICs) was not clearly elucidated. In the present study, we found that miR-186 expression was reduced in liver CSCs. Functional studies showed that miR-186 knockdown facilitated liver CSCs self-renewal and tumorigenesis. Conversely, forced miR-186 expression suppressed liver CSCs self-renewal and tumorigenesis. Mechanically, miR-186 downregulated PTPN11 via binding to its 3'-UTR in liver CSCs. The correlation of miR-186 and PTPN11 was confirmed in Hepatocellular carcinoma (HCC) patients' tissues. Further study showed that interference of PTPN11 can abolished the discrepancy between miR-186 mimic and control HCC cells in self-renewal and the proportion of CSCs. Additionally, we found that miR-186 overexpression HCC cells were more sensitive to cisplatin treatment. Clinical cohort analysis showed that HCC patients with high miR-186 were benefited more from transcatheter arterial chemoembolization (TACE) treatment. In conclusion, our study demonstrates a new regulation mechanism of liver CSCs, a new target for HCC, and a biomarker for postoperative TACE.

15.
Urologiia ; (1): 112-119, 2021 03.
Artículo en Ruso | MEDLINE | ID: mdl-33818946

RESUMEN

Hemorrhagic fever with renal syndrome (HFRS) is an acute natural focal viral disease caused by viruses of the genus hantavirus, characterized by damage to small blood vessels, kidneys, lungs and other organs of a person. MicroRNAs (miRNAs) are 18-22 nucleotide endogenously expressed RNA molecules that inhibit gene expression at the post-transcriptional level by binding to the 3-untranslated region of the target mRNA. It has been proven that miRNAs play a significant role in various biological processes, including the cell cycle, apoptosis, cell proliferation and differentiation. It has been proven that miRNAs may be involved in the pathogenesis of infectious diseases, including HFRS. Hantavirus infection predominantly affects endothelial cells and causes dysfunction of the endothelium of capillaries and small vessels. It is known that the immune response induced by Hantavirus infection plays an important role in disrupting the endothelial barrier. In a few studies, both in vitro and in vivo, it has been shown that endothelial dysfunction and the immune response after infection with Hantavirus can be partially regulated by miRNAs by acting on certain genes. Most of the miRNAs is expressed within the cells themselves. However, in some biological fluids of the human body, for example, plasma or blood serum, numerous miRNAs, called circulating miRNAs, have been found. Circulating miRNAs can be secreted by cells into human biological fluids as part of extracellular vesicles as exosomes or be part of an RNA-bound protein complex as miRNA-Argonaute 2 (Ago2). These miRNAs are resistant to nucleases, which makes them attractive as potential biomarkers in various human diseases. There is no specific antiviral therapy for HFRS, and the determination of laboratory parameters that are used to diagnose, assess the severity, and predict the course of the disease remains a challenge due to the peculiarities of the pathophysiology and clinical course of the disease. Studying the role of miRNAs in HFRS seems to be expedient for the development of specific and effective therapy, as well as for use as diagnostic and prognostic biomarkers (in relation to circulating miRNAs).


Asunto(s)
Fiebre Hemorrágica con Síndrome Renal , MicroARNs , Orthohantavirus , Células Endoteliales , Orthohantavirus/genética , Fiebre Hemorrágica con Síndrome Renal/genética , Humanos , Riñón , MicroARNs/genética
16.
Adv Mater ; 33(3): e2004559, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33296110

RESUMEN

Surgical resection is the first-line therapy for colorectal cancer (CRC). However, for advanced CRC, the curative effect of surgical resection is limited due to either local recurrence or distal metastasis. Postoperative in situ immunotherapy, presents a promising option for preventing tumor recurrence and metastasis, owing to the fact that surgeons have unique opportunities and direct access to the surgical site. Herein, a designed biopolymer immune implant for CRC post-surgical therapy, characterized with tissue adhesion, sustained drug release, and sequential elicitation of innate immunity, adaptive immunity, and immune memory effects, is reported. With gradual release of the loaded resiquimod (R848) and anti-OX40 antibody (aOX40), the immune implant can eradicate residual tumors post-surgery (with no tumor recurrence in 150 days), inhibit the growth of distal tumors and elicit immune memory effects to resist tumor re-challenge. Immunological analysis reveal that the biopolymer immune plant treatment leads to a two-stage action, with enhanced natural killer cells (NK cells) infiltration and activation of dendritic cells (DCs) in the first several days, then a greatly increased population of infiltrating T cells, and finally immune memory effects are established. The reported biopolymer immune implants provide a valuable and clinically-relevant option for post-surgical CRC management.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Biopolímeros/farmacología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Inmunidad Innata/efectos de los fármacos , Prótesis e Implantes , Biopolímeros/uso terapéutico , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Células Asesinas Naturales/inmunología , Periodo Posoperatorio
17.
Biomaterials ; 268: 120542, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33249316

RESUMEN

STING (stimulator of interferon genes) signaling pathway has attracted considerable attention in cancer immunotherapy due to its capacity to boost vigorous antitumor immunity. However, the shortage of effective STING agonists limits the promotion of STING pathway in cancer treatment. Herein, we present an approach for in situ activation of STING pathway with nanoparticles delivered DNA-targeting chemo agents, based on the understanding that cytosol DNA is a pre-requisite for STING pathway activation. Through in vitro screening among several DNA-targeting chemo agents, we identified 7-ethyl-10-hydroxycamptothecin (SN38) as the most potent drug for stimulating interferon (IFN)-ß secretion and proved that this process is mediated by the passage of DNA-containing exosomes from treated tumor cells to bone marrow-derived dendritic cells (BMDCs) and subsequent activation of the STING pathway. Furthermore, we designed a polymeric-SN38 conjugate that could self-assemble into nanoparticles (SN38-NPs) for in vivo application. The SN38-NPs formulation reduced toxicity of free SN38, effectively stimulated the activation of STING pathway in E0771 tumors, and resulted in a tumor suppression rate (TSR%) of 82.6%. Our results revealed a new mechanism of SN38 in cancer treatment and should inspire using more DNA-targeting agents, especially in nanoformulation, for activating STING pathway and cancer chemoimmunotherapy.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Inmunoterapia , Irinotecán , Neoplasias/tratamiento farmacológico , Polímeros
18.
Acta Pharm Sin B ; 10(11): 2171-2182, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33304784

RESUMEN

Primary bile acids were reported to augment secretion of chemokine (C‒X‒C motif) ligand 16 (CXCL16) from liver sinusoidal endothelial cells (LSECs) and trigger natural killer T (NKT) cell-based immunotherapy for liver cancer. However, abundant expression of receptors for primary bile acids across the gastrointestinal tract overwhelms the possibility of using agonists against these receptors for liver cancer control. Taking advantage of the intrinsic property of LSECs in capturing circulating nanoparticles in the circulation, we proposed a strategy using nanoemulsion-loaded obeticholic acid (OCA), a clinically approved selective farnesoid X receptor (FXR) agonist, for precisely manipulating LSECs for triggering NKT cell-mediated liver cancer immunotherapy. The OCA-nanoemulsion (OCA-NE) was prepared via ultrasonic emulsification method, with a diameter of 184 nm and good stability. In vivo biodistribution studies confirmed that the injected OCA-NE mainly accumulated in the liver and especially in LSECs and Kupffer cells. As a result, OCA-NE treatment significantly suppressed hepatic tumor growth in a murine orthotopic H22 tumor model, which performed much better than oral medication of free OCA. Immunologic analysis revealed that the OCA-NE resulted in augmented secretion of CXCL16 and IFN-γ, as well as increased NKT cell populations inside the tumor. Overall, our research provides a new evidence for the antitumor effect of receptors for primary bile acids, and should inspire using nanotechnology for precisely manipulating LSECs for liver cancer therapy.

19.
Biosaf Health ; 2(3): 164-168, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32838284

RESUMEN

This study described the epidemiologic and clinical characteristics of patients who died from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and pointed out the potential risk factors associated with fatal outcomes. Retrospective data from 42 death cases due to SARS-CoV-2 infection at Tongji Hospital Affiliated to Huazhong University of Science and Technology, Wuhan, China was analyzed. Demographics, clinical detection, laboratory findings, and treatments of the deceased were collected and analyzed. The average time between onset of symptoms and admission to the hospitals was 11 ± 5 days of hospitalization. Among the deceased, 60% were with co-morbidities. All of them were having fever and bilateral pneumonia on computed tomography, abnormal infection-related biomarkers, and renal impairment. Abnormal blood coagulation parameters that appeared in more than half of them, were consistent with disseminated intravascular coagulation. All of the patients were treated in the ICU. Based on the fact that SARS-CoV-2 infection carries a risk of mortality, we may infer a few older male patients with underlying comorbidities are likely to have the increased risk. Impaired consciousness level, markers of renal impairment and coagulation abnormalities may be poor prognostic factors.

20.
Front Cell Dev Biol ; 8: 683, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32850809

RESUMEN

The recent outbreak of the coronavirus disease-2019 (COVID-19) caused serious challenges to the human society in China and across the world. COVID-19 induced pneumonia in human hosts and carried a highly inter-person contagiousness. The COVID-19 patients may carry severe symptoms, and some of them may even die of major organ failures. This study utilized the machine learning algorithms to build the COVID-19 severeness detection model. Support vector machine (SVM) demonstrated a promising detection accuracy after 32 features were detected to be significantly associated with the COVID-19 severeness. These 32 features were further screened for inter-feature redundancies. The final SVM model was trained using 28 features and achieved the overall accuracy 0.8148. This work may facilitate the risk estimation of whether the COVID-19 patients would develop the severe symptoms. The 28 COVID-19 severeness associated biomarkers may also be investigated for their underlining mechanisms how they were involved in the COVID-19 infections.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA