Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Biofabrication ; 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39008993

RESUMO

Various anisotropic tissue structures exist in organisms, including muscle tissue, skin tissue, and nerve tissue. Replicating anisotropic tissue structures in vitro has posed a significant challenge. 3D printing technology is often used to fabricate biomimetic structures due to its advantages in manufacturing principle. However, direct 3D printing of freeform anisotropic bioactive structures has not been reported. To tackle this challenge, we developed a ternary F/G/P ink system that integrates the printability of Pluronic F127 (F), the robust bioactivity and photocrosslinking properties of GelMA (G), and the shear-induced alignment functionality of high-molecular-weight PEG (P). And through this strategic ternary system combination, freeform anisotropic tissue structures can be 3D printed directly. Moreover, these anisotropic structures exhibit excellent bioactivity, and promote orientational growth of different cells. This advancement holds promise for the repair and replacement of anisotropic tissues within the human body.

2.
Artigo em Inglês | MEDLINE | ID: mdl-38716540

RESUMO

Bone cancer pain (BCP), due to cancer bone metastasis and bone destruction, is a common symptom of tumors, including breast, prostate, and lung tumors. Patients often experience severe pain without effective treatment. Here, using a mouse model of bone cancer, we report that MOTS-c, a novel mitochondrial-derived peptide, confers remarkable protection against cancer pain and bone destruction. Briefly, we find that the plasma level of endogenous MOTS-c is significantly lower in the BCP group than in the sham group. Accordingly, intraperitoneal administration of MOTS-c robustly attenuates bone cancer-induced pain. These effects are blocked by compound C, an AMPK inhibitor. Furthermore, MOTS-c treatment significantly enhances AMPKα 1/2 phosphorylation. Interestingly, mechanical studies indicate that at the spinal cord level, MOTS-c relieves pain by restoring mitochondrial biogenesis, suppressing microglial activation, and decreasing the production of inflammatory factors, which directly contribute to neuronal modulation. However, in the periphery, MOTS-c protects against local bone destruction by modulating osteoclast and immune cell function in the tumor microenvironment, providing long-term relief from cancer pain. Additionally, we find that chronic administration of MOTS-c has little effect on liver, renal, lipid or cardiac function in mice. In conclusion, MOTS-c improves BCP through peripheral and central synergistic effects on nociceptors, immune cells, and osteoclasts, providing a pharmacological and biological rationale for the development of mitochondrial peptide-based therapeutic agents for cancer-induced pain.

3.
Mol Pain ; 20: 17448069241252654, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38658141

RESUMO

Painful Diabetic Neuropathy (PDN) is a common diabetes complication that frequently causes severe hyperalgesia and allodynia and presents treatment challenges. Mitochondrial-derived peptide (MOTS-c), a novel mitochondrial-derived peptide, has been shown to regulate glucose metabolism, insulin sensitivity, and inflammatory responses. This study aimed to evaluate the effects of MOTS-c in streptozocin (STZ)-induced PDN model and investigate the putative underlying mechanisms. We found that endogenous MOTS-c levels in plasma and spinal dorsal horn were significantly lower in STZ-treated mice than in control animals. Accordingly, MOTS-c treatment significantly improves STZ-induced weight loss, elevation of blood glucose, mechanical allodynia, and thermal hyperalgesia; however, these effects were blocked by dorsomorphin, an adenosine monophosphate-activated protein kinase (AMPK) inhibitor. In addition, MOTS-c treatment significantly enhanced AMPKα1/2 phosphorylation and PGC-1α expression in the lumbar spinal cord of PDN mice. Mechanistic studies indicated that MOTS-c significantly restored mitochondrial biogenesis, inhibited microglia activation, and decreased the production of pro-inflammatory factors, which contributed to the alleviation of pain. Moreover, MOTS-c decreased STZ-induced pain hypersensitivity in PDN mice by activating AMPK/PGC-1α signaling pathway. This provides the pharmacological and biological evidence for developing mitochondrial peptide-based therapeutic agents for PDN.


Assuntos
Neuropatias Diabéticas , Hiperalgesia , Mitocôndrias , Biogênese de Organelas , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Estreptozocina , Animais , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Neuropatias Diabéticas/tratamento farmacológico , Neuropatias Diabéticas/metabolismo , Neuropatias Diabéticas/patologia , Masculino , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por AMP/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Peptídeos/farmacologia , Camundongos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Microglia/efeitos dos fármacos , Microglia/metabolismo
4.
Sleep Med ; 115: 162-173, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38367358

RESUMO

The hippocampus (HPC) plays a pivotal role in fear learning and memory. Our two recent studies suggest that rapid eye movement (REM) sleep via the HPC downregulates fear memory consolidation and promotes fear extinction. However, it is not clear whether and how the dorsal and the ventral HPC regulates fear memory differently; and how the HPC in wake regulates fear memory. By chemogenetic stimulating in the HPC directly and its afferent entorhinal cortex that selectively activated the HPC in REM sleep for 3-6 h post-fear-acquisition, we found that HPC activation in REM sleep consolidated fear extinction memory. In particular, dorsal HPC (dHPC) stimulation in REM sleep virtually eliminated fear memory by enhancing fear extinction and reducing fear memory consolidation. By contrast, chemogenetic stimulating HPC afferent the supramammillary nucleus (SUM) induced 3-hr wake with HPC activation impaired fear extinction. Finally, desipramine (DMI) injection that selectively eliminated REM sleep for >6 h impaired fear extinction. Our results demonstrate that the HPC is critical for fear memory regulation; and wake HPC and REM sleep HPC have an opposite role in fear extinction of respective impairment and consolidation.


Assuntos
Medo , Consolidação da Memória , Humanos , Extinção Psicológica/fisiologia , Sono/fisiologia , Aprendizagem/fisiologia , Hipocampo , Consolidação da Memória/fisiologia
5.
Int J Biochem Cell Biol ; 169: 106541, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38309648

RESUMO

Microglial are major players in neuroinflammation that have recently emerged as potential therapeutic targets for neuropathic pain. Glucose metabolic programming has been linked to differential activation state and function in microglia. Tumor necrosis factor α-induced protein 8-like-2 (TNFAIP8L2) is an important component in regulating the anti-inflammatory response. However, the role of TNFAIP8L2 in microglia differential state during neuropathic pain and its interplay with glucose metabolic reprogramming in microglia has not yet been determined. Thus, we aimed to investigate the role of TNFAIP8L2 in the status of microglia in vitro and in vivo. BV2 microglial cells were treated with lipopolysaccharides plus interferon-gamma (LPS/IFNγ) or interleukin-4 (IL-4) to induce the two different phenotypes of microglia in vitro. In vivo experiments were conducted by chronic constriction injury of the sciatic nerve (CCI). We investigated whether TNFAIP8L2 regulates glucose metabolic programming in BV2 microglial cells. The data in vitro showed that TNFAIP8L2 lowers glycolysis and increases mitochondrial oxidative phosphorylation (OXPHOS) in inflammatory microglia. Blockade of glycolytic pathway abolished TNFAIP8L2-mediated differential activation of microglia. TNFAIP8L2 suppresses inflammatory microglial activation and promotes restorative microglial activation in BV2 microglial cells and in spinal cord microglia after neuropathic pain. Furthermore, TNFAIP8L2 controls differential activation of microglia and glucose metabolic reprogramming through the MAPK/mTOR/HIF-1α signaling axis. This study reveals that TNFAIP8L2 plays a critical role in neuropathic pain, providing important insights into glucose metabolic reprogramming and microglial phenotypic transition, which indicates that TNFAIP8L2 may be used as a potential drug target for the prevention of neuropathic pain.


Assuntos
Microglia , Neuralgia , Humanos , Microglia/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Reprogramação Metabólica , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Proteínas de Transporte/metabolismo , Fenótipo , Glucose/farmacologia , Glucose/metabolismo , Lipopolissacarídeos/farmacologia
6.
Nanomicro Lett ; 16(1): 74, 2024 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-38175408

RESUMO

Electrolytic aqueous zinc-manganese (Zn-Mn) batteries have the advantage of high discharge voltage and high capacity due to two-electron reactions. However, the pitfall of electrolytic Zn-Mn batteries is the sluggish deposition reaction kinetics of manganese oxide during the charge process and short cycle life. We show that, incorporating ZnO electrolyte additive can form a neutral and highly viscous gel-like electrolyte and render a new form of electrolytic Zn-Mn batteries with significantly improved charging capabilities. Specifically, the ZnO gel-like electrolyte activates the zinc sulfate hydroxide hydrate assisted Mn2+ deposition reaction and induces phase and structure change of the deposited manganese oxide (Zn2Mn3O8·H2O nanorods array), resulting in a significant enhancement of the charge capability and discharge efficiency. The charge capacity increases to 2.5 mAh cm-2 after 1 h constant-voltage charging at 2.0 V vs. Zn/Zn2+, and the capacity can retain for up to 2000 cycles with negligible attenuation. This research lays the foundation for the advancement of electrolytic Zn-Mn batteries with enhanced charging capability.

7.
Neurol Res ; 46(2): 165-177, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37899006

RESUMO

BACKGROUND: Inflammatory pain is caused by damaged tissue or noxious stimuli, accompanied by the release of inflammatory mediators that often leads to severe hyperalgesia and allodynia with limited therapy options. Recently, a novel mitochondrial-derived peptide (named MOTS-c) was reported to regulate obesity, metabolic homeostasis and inflammatory response. The aim of this study was to investigate the effects of MOTS-c and its related regulatory mechanisms involved in inflammatory pain. METHODS: Male Kunming mice (8-10 weeks-old) were intraplantar injected with formalin, capsaicin, λ-Carrageenan and complete Freund adjuvant (CFA) to establish acute and chronic inflammatory pain. The effects of MOTS-c on the above inflammatory pain mice and its underlying mechanisms were examined by behavioral tests, quantitative polymerase chain reaction (qPCR), western blotting, enzyme linked immunosorbent assay (ELISA), immunohistochemistry (IHC) and immunofluorescence (IF). RESULTS: Behavioral experiments investigated the potential beneficial effects of MOTS-c on multiple acute and chronic inflammatory pain in mice. The results showed that MOTS-c treatment produced potent anti-allodynic effects in formalin-induced acute inflammatory pain, capsaicin-induced nocifensive behaviors and λ-Carrageenan/CFA-induced chronic inflammatory pain model. Further mechanistic studies revealed that central MOTS-c treatment significantly ameliorated CFA-evoked the release of inflammatory factors and activation of glial cells and neurons in the spinal dorsal horn. Moreover, peripheral MOTS-c treatment reduced CFA-evoked inflammatory responses in the surface structure of hindpaw skin, accompanied by inhibiting excitation of peripheral calcitonin gene-related peptide (CGRP) and P2X3 nociceptive neurons. CONCLUSIONS: The present study indicates that MOTS-c may serve as a promising therapeutic target for inflammatory pain.


Assuntos
Capsaicina , Dor Crônica , Camundongos , Masculino , Animais , Carragenina/toxicidade , Carragenina/uso terapêutico , Capsaicina/farmacologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Hiperalgesia/metabolismo , Dor Crônica/complicações , Adjuvante de Freund/toxicidade , Formaldeído/toxicidade , Formaldeído/uso terapêutico
8.
Biomater Sci ; 12(3): 738-747, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38105707

RESUMO

The potential of 3D bioprinting in tissue engineering and regenerative medicine is enormous, but its implementation is hindered by the reliance on high-strength materials, which restricts the use of low-viscosity, biocompatible materials. Therefore, a major challenge for incorporating 3D bioprinting into tissue engineering is to develop a novel bioprinting platform that can reversibly provide high biological activity materials with a structural support. This study presents a room temperature printing system based on GelMA combined with carrageenan to address this challenge. By leveraging the wide temperature stability range and lubricating properties of carrageenan the room temperature stability of GelMA could be enhanced, as well as creating a solid ink to improve the performance of solid GelMA. Additionally, by utilizing the solubility of carrageenan at 37 °C, it becomes possible to prepare a porous GelMA structure while mimicking the unique extracellular matrix properties of osteocytes through residual carrageenan content and amplifying BMSCs' osteogenesis potential to some extent. Overall, this study provides an innovative technical platform for incorporating a low-viscosity ink into 3D bioprinting and resolves the long-standing contradiction between material printing performance and biocompatibility in bioprinting technology.


Assuntos
Bioimpressão , Alicerces Teciduais , Alicerces Teciduais/química , Carragenina , Impressão Tridimensional , Materiais Biocompatíveis/química , Engenharia Tecidual , Hidrogéis/química , Gelatina/química
9.
J Med Chem ; 66(24): 17138-17154, 2023 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-38095323

RESUMO

Our previous study reported the multifunctional agonist for opioid and neuropeptide FF receptors DN-9, along with its cyclic peptide analogues c[D-Cys2, Cys5]-DN-9 and c[D-Lys2, Asp5]-DN-9. These analogues demonstrated potent antinociceptive effects with reduced opioid-related side effects. To develop more stable and effective analgesics, we designed, synthesized, and evaluated seven hydrocarbon-stapled cyclic peptides based on DN-9. In vitro calcium mobilization assays revealed that most of the stapled peptides, except 3, displayed multifunctional agonistic activities at opioid and neuropeptide FF receptors. Subcutaneous administration of all stapled peptides resulted in effective and long-lasting antinociceptive activities lasting up to 360 min. Among these stapled peptides, 1a and 1b emerged as the optimized compounds, producing potent central antinociception following subcutaneous, intracerebroventricular, and oral administrations. Additionally, subcutaneous administration of 1a and 1b caused nontolerance antinociception, with limited occurrence of constipation and addiction. Furthermore, 1a was selected as the final optimized compound due to its wider safety window compared to 1b.


Assuntos
Analgésicos Opioides , Oligopeptídeos , Analgésicos Opioides/efeitos adversos , Oligopeptídeos/química , Analgésicos/química , Peptídeos/química , Receptores de Neuropeptídeos/agonistas , Encéfalo , Receptores Opioides mu/agonistas
10.
J Interferon Cytokine Res ; 43(10): 445-454, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37819621

RESUMO

Some progress has been made in immunotherapy with chimeric antigen receptor (CAR)-T cells targeting NKG2D-NKG2DL with the purpose of eradicating solid tumors. Non-small cell lung cancer (NSCLC) has been shown to express NKG2DL. This study hence evaluated the therapeutic effect of NKG2D CAR-T cells on NSCLC. Accordingly, NKG2D CAR-T cells were obtained from diverse human autologous T cell sources. T cells from peripheral blood T lymphocytes of healthy volunteers (without NKG2D CAR insertion) were used as NT-T cells. Coculture of effector cells (CAR-T cells or NT-T cells) with target cells (NSCLC cells such as PC-9 or NCL-H460 cells) was performed at different ratios. The cytotoxicity of CAR-T cells was examined using lactate dehydrogenase assay kits. Murine xenograft assay was conducted to investigate the in vivo antitumor effect of CAR-T cells. Cytokines secreted from CAR-T cells were assessed by enzyme-linked immunosorbent assay. CAR-T cell infiltration into xenografts was observed through immunochemical assay. Based on the results, NKG2DL was highly expressed in NSCLC cells. Compared with NT-T cells, NKG2D CAR-T cells from different sources of T cells delivered stronger toxicity, and secreted more effector and memory function-related cytokines to NSCLC cells, and those from the peripheral blood of healthy donors (H-T cells) exhibited the strongest effect. Furthermore, compared with NT-T cells, H-T cells and NKG2D CAR-T cells from NSCLC patients' peripheral blood diminished tumor, improved survival, increased body weight and tumor-infiltrating capacity, and upregulated serum IFN-γ level in NOG mice. Collectively speaking, NKG2D CAR-T cells exhibit a robust effect on eradicating NSCLC in a NKG2DL-dependent manner, thus making themselves a promising therapeutic candidate for NSCLC patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Camundongos , Animais , Linfócitos T , Carcinoma Pulmonar de Células não Pequenas/terapia , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Imunoterapia Adotiva , Neoplasias Pulmonares/terapia , Citocinas , Linhagem Celular Tumoral
11.
J Nanobiotechnology ; 21(1): 303, 2023 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-37641124

RESUMO

Skeletal muscle disease severity can often progress asymmetrically across muscle groups and heterogeneously within tissues. An example is Duchenne Muscular Dystrophy (DMD) in which lack of dystrophin results in devastating skeletal muscle wasting in some muscles whereas others are spared or undergo hypertrophy. An efficient, non-invasive approach to identify sites of asymmetry and degenerative lesions could enable better patient monitoring and therapeutic targeting of disease. In this study, we utilized a versatile intravenously injectable mesoporous silica nanoparticle (MSNP) based nanocarrier system to explore mechanisms of biodistribution in skeletal muscle of mdx mouse models of DMD including wildtype, dystrophic, and severely dystrophic mice. Moreover, MSNPs could be imaged in live mice and whole muscle tissues enabling investigation of how biodistribution is altered by different types of muscle pathology such as inflammation or fibrosis. We found MSNPs were tenfold more likely to aggregate within select mdx muscles relative to wild type, such as gastrocnemius and quadriceps. This was accompanied by decreased biodistribution in off-target organs. We found the greatest factor affecting preferential delivery was the regenerative state of the dystrophic skeletal muscle with the highest MSNP abundance coinciding with the regions showing the highest level of embryonic myosin staining and intramuscular macrophage uptake. To demonstrate, muscle regeneration regulated MSNP distribution, we experimentally induced regeneration using barium chloride which resulted in a threefold increase of intravenously injected MSNPs to sites of regeneration 7 days after injury. These discoveries provide the first evidence that nanoparticles have selective biodistribution to skeletal muscle in DMD to areas of active regeneration and that nanoparticles could enable diagnostic and selective drug delivery in DMD skeletal muscle.


Assuntos
Distrofina , Músculo Esquelético , Animais , Camundongos , Distribuição Tecidual , Camundongos Endogâmicos mdx , Regeneração
12.
Neuropharmacology ; 238: 109667, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37451333

RESUMO

Chronic morphine tolerance is a repulsive barrier to the clinical treatment of pain. Whereas the underlying molecular mechanisms of morphine tolerance remain unknown. Here, we proposed that tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) is an essential control point regarding the progression of chronic morphine antinociceptive tolerance. We found that TIPE2 levels in the lumbar spinal cord were significantly downregulated in the morphine tolerance mouse model. Specifically, decreased TIPE2 by morphine tolerance was primarily expressed in spinal neurons, while increased expression of spinal TIPE2 distinctly attenuated the chronic morphine antinociceptive tolerance and tolerance-associated hyperalgesia. We also observed that increased expression of spinal TIPE2 significantly reduced morphine tolerance-induced neuronal ROS production and apoptosis, along with the activation of MAPKs and NF-κB signaling pathways. Moreover, the increased TIPE2 expression inhibited neuronal activation and glial reactivity in the spinal dorsal horn after chronic morphine exposure. Additionally, TIPE2 overexpression in cultured SH-SY5Y cells significantly suppressed ROS production and apoptosis in response to morphine challenge. Therefore, we can conclude that the upregulation of spinal TIPE2 may attenuate the morphine antinociceptive tolerance via TIPE2-dependent downregulation of neuronal ROS, inhibition of neuronal apoptosis, suppression of MAPKs and NF-κB activation. TIPE2 may be a potential strategy for preventing morphine tolerance in the future studies and clinical settings.


Assuntos
Morfina , Neuroblastoma , Humanos , Camundongos , Animais , Morfina/farmacologia , Morfina/metabolismo , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neuroblastoma/patologia , Corno Dorsal da Medula Espinal/metabolismo , Medula Espinal/metabolismo , Transdução de Sinais , Analgésicos/farmacologia , Analgésicos/metabolismo , Apoptose , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo
13.
ACS Chem Neurosci ; 14(12): 2362-2374, 2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37285113

RESUMO

MOTS-c, a recently discovered mitochondrial-derived peptide, plays an important role in many physiological and pathological functions via adenosine monophosphate-activated protein kinase (AMPK) activation. Numerous studies have demonstrated that AMPK is an emerging target for the modulation of neuropathic pain. Meanwhile, microglia-activation-evoked neuroinflammation is known to contribute to the development and progression of neuropathic pain. MOTS-c is also known to inhibit microglia activation, chemokine and cytokine expression, and innate immune responses. Accordingly, in this study, we evaluated the effects of MOTS-c on neuropathic pain and investigated the putative underlying mechanisms. We found that MOTS-c levels in plasma and spinal dorsal horn were significantly lower in mice with spared nerve injury (SNI)-induced neuropathic pain than in control animals. Accordingly, MOTS-c treatment produced pronounced dose-dependent antinociceptive effects in SNI mice; however, these effects were blocked by dorsomorphin, an AMPK inhibitor, but not naloxone, a nonselective opioid receptor antagonist. Moreover, intrathecal (i.t.) injection of MOTS-c significantly enhanced AMPKα1/2 phosphorylation in the lumbar spinal cord of SNI mice. MOTS-c also significantly inhibited proinflammatory cytokine production and microglia activation in the spinal cord. The antinociceptive effects of MOTS-c were retained even when microglia activation in the spinal cord was inhibited by minocycline pretreatment, indicating that spinal cord microglia are dispensable for the antiallodynic effects of MOTS-c. In the spinal dorsal horn, MOTS-c treatment inhibited c-Fos expression and oxidative damage mainly in neurons rather than microglia. Finally, in contrast to morphine, i.t. administration of MOTS-c resulted in limited side effects relating to antinociceptive tolerance, gastrointestinal transit inhibition, locomotor function, and motor coordination. Collectively, the present study is the first to provide evidence that MOTS-c may be a promising therapeutic target for neuropathic pain.


Assuntos
Microglia , Neuralgia , Camundongos , Animais , Proteínas Quinases Ativadas por AMP/metabolismo , Medula Espinal , Neuralgia/metabolismo , Neurônios/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Citocinas/metabolismo , Analgésicos/farmacologia , Estresse Oxidativo , Hiperalgesia/metabolismo
14.
J Hazard Mater ; 457: 131740, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37269567

RESUMO

Efficient management of disguised toxic pollutants (DTPs), which can undergo microbial degradation and convert into more toxic substances, necessitates the collaboration of diverse microbial populations in wastewater treatment plants. However, the identification of key bacterial degraders capable of controlling the toxicity risks of DTPs through division of labor mechanisms in activated sludge microbiomes has received limited attention. In this study, we investigated the key degraders capable of controlling the risk of estrogenicity associated with nonylphenol ethoxylate (NPEO), a representative DTP, in textile activated sludge microbiomes. The results of our batch experiments revealed that the transformation of NPEO into NP and subsequent NP degradation were the rate-limiting processes for controlling the risk of estrogenicity, resulting in an inverted V-shaped curve of estrogenicity in water samples during the biodegradation of NPEO by textile activated sludge. By utilizing enrichment sludge microbiomes treated with NPEO or NP as the sole carbon and energy source, a total of 15 bacterial degraders, including Sphingbium, Pseudomonas, Dokdonella, Comamonas, and Hyphomicrobium, were identified as capable of participating in these processes, Among them, Sphingobium and Pseudomonas were the two key degraders that could cooperatively interact in the degradation of NPEO with division of labor mechanisms. Co-culturing Sphingobium and Pseudomonas isolates exhibited a synergistic effect in degrading NPEO and reducing estrogenicity. Our study underscores the potential of the identified functional bacteria for controlling estrogenicity associated with NPEO and provides a methodological framework for identifying key cooperators engaged in labor division, contributing to the management of risks associated with DTPs by leveraging intrinsic microbial metabolic interactions.


Assuntos
Biodegradação Ambiental , Poluentes Químicos da Água , Estrona , Etilenoglicóis , Esgotos/microbiologia , Sphingomonadaceae/metabolismo , Poluentes Químicos da Água/análise
15.
Front Immunol ; 14: 1146861, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37234150

RESUMO

Background/purpose: It has been demonstrated that gut microbes are closely associated with the pathogenesis of lymphoma, but the gut microbe landscape and its association with immune cells in diffuse large B-cell lymphoma (DLBCL) remain largely unknown. In this study, we explored the associations between gut microbiota, clinical features and peripheral blood immune cell subtypes in DLBCL. Method: A total of 87 newly diagnosed DLBCL adults were enrolled in this study. The peripheral blood samples were collected from all patients and then submitted to immune cell subtyping using full-spectral flow cytometry. Metagenomic sequencing was applied to assess the microbiota landscape of 69 of 87 newly diagnosed DLBCL patients. The microbiotas and peripheral blood immune cell subsets with significant differences between different National Comprehensive Center Network-International Prognostic Indexes (NCCN-IPIs) (low-risk, low-intermediate-risk, intermediate-high-risk, high-risk) groups were screened. Results: A total of 10 bacterial phyla, 31 orders and 455 bacteria species were identified in 69 patients with newly diagnosed DLBCL. The abundances of 6 bacteria, including Blautia sp.CAG 257, Actinomyces sp.S6 Spd3, Streptococcus parasanguinis, Bacteroides salyersiae, Enterococcus faecalls and Streptococcus salivarius were significantly different between the low-risk, low-intermediate-risk, intermediate-high-risk and high-risk groups, among which Streptococcus parasanguinis and Streptococcus salivarius were markedly accumulated in the high-risk group. The different bacteria species were mostly enriched in the Pyridoxal 5'-phosphate biosynthesis I pathway. In addition, we found that 2 of the 6 bacteria showed close associations with the different immune cell subtypes which were also identified from different NCCN-IPIs. In detail, the abundance of Bacteroides salyersiae was negatively correlated with Treg cells, CD38+ nonrescue exhausted T cells, nature killer 3 cells and CD38+CD8+ effector memory T cells, while the abundance of Streptococcus parasanguinis was negatively correlated with HLA-DR+ NK cells, CD4+ Treg cells, HLA-DR+ NKT cells and HLA-DR+CD94+CD159c+ NKT cells. Conclusion: This study first reveals the gut microbiota landscape of patients with newly diagnosed DLBCL and highlights the association between the gut microbiota and immunity, which may provide a new idea for the prognosis assessment and treatment of DLBCL.


Assuntos
Linfoma Difuso de Grandes Células B , Metagenoma , Adulto , Humanos , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Antígenos HLA-DR
16.
Nano Lett ; 23(6): 2137-2147, 2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36881967

RESUMO

We have developed tailor-designed mesoporous silica nanoparticles (MSNPs) specifically for delivering mRNA. Our unique assembly protocol involves premixing mRNA with a cationic polymer and then electrostatically binding it to the MSNP surface. Since the key physicochemical parameters of MSNPs could influence the biological outcome, we also investigated the roles of size, porosity, surface topology, and aspect ratio on the mRNA delivery. These efforts allow us to identify the best-performing carrier, which was able to achieve efficient cellular uptake and intracellular escape while delivering a luciferase mRNA in mice. The optimized carrier remained stable and active for at least 7 days after being stored at 4 °C and was able to enable tissue-specific mRNA expression, particularly in the pancreas and mesentery after intraperitoneal injection. The optimized carrier was further manufactured in a larger batch size and found to be equally efficient in delivering mRNA in mice and rats, without any obvious toxicity.


Assuntos
Nanopartículas , Dióxido de Silício , Animais , Camundongos , Ratos , Porosidade
17.
Colloids Surf B Biointerfaces ; 224: 113208, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36801524

RESUMO

A functional liquid-infused catheter surface strategy has recently attracted increasing attention for blood transport with the remarkable antibiofouling performance. Nevertheless, constructing porous structure inside a catheter with effective functional liquid-locking ability remains extremely challenging. Herein, the central cylinder mold and sodium chloride particle templates technique was used to create a PDMS sponge-based catheter that stores a stable functional liquid. Our multifunctional liquid-infused PDMS sponge-based catheter can not only exhibit bacterial resistant, less macrophages infiltration, a slighter inflammation response, but also capability to prevent platelet adhesion and activation, and impressively reduce thrombosis in vivo even at high shear. Therefore, these desirable properties will endow the prospective practical applications and serve as a watershed moment in the development of biomedical devices.


Assuntos
Catéteres , Estudos Prospectivos
18.
Eur J Pharmacol ; 939: 175469, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36528071

RESUMO

Inflammatory bowel disease (IBD) is a chronic and relapsing inflammatory disorder of the gastrointestinal tract (GI). Currently, the treatment options for IBD are limited. It has been reported that a novel bioactive mitochondrial-derived peptide (MOTS-c) encoded in the mitochondrial 12S rRNA, suppresses inflammatory response by enhancing the phagocytosis of macrophages. The aim of this study was to investigate the protective effects of MOTS-c against dextran sulfate sodium (DSS)-induced colitis. The results showed that intraperitoneal (i.p.) administration of MOTS-c significantly ameliorated the symptoms of DSS-induced experimental colitis, such as body weight loss, colon length shortening, diarrhea, and histological damage. MOTS-c down-regulated the expression of pro-inflammatory cytokines, decreased the plasma levels of myeloperoxidase, and inhibited the activation of macrophages and recruitment of neutrophils. Moreover, treatment with MOTS-c exhibited anti-apoptotic effects and significantly suppressed the phosphorylation of AMPKα1/2, ERK, and JNK. Notably, oral administration of MOTS-c did not result in any significant improvements. Screening of cell penetrating peptides was performed, (PRR)5 was linked to the C-terminus of MOTS-c through a linker to synthesize a new molecule (termed MP) with better penetration into the colon epithelium. In vitro experiments revealed the longer half-life of MP than MOTS-c, and in vivo experiments showed that oral administration of MP significantly ameliorated DSS-induced colitis. CONCLUSION: The present results demonstrate a protective role of MOTS-c in experimental IBD.


Assuntos
Colite , Inflamação , Doenças Inflamatórias Intestinais , Peptídeos , Animais , Camundongos , Apoptose/efeitos dos fármacos , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Colo , Citocinas/metabolismo , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/tratamento farmacológico , Camundongos Endogâmicos C57BL , Fatores de Transcrição/metabolismo , Peptídeos/uso terapêutico
19.
Cell Biol Int ; 46(9): 1510-1518, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35670241

RESUMO

The blood brain barrier (BBB) is a protective border that prevents noxious substances from gaining access to the central nervous system (CNS). CXCL13 is a chemokine from the CXC chemokine family, which has been shown to destroy the barrier function of umbilical vein endothelial cells with its receptor CXCR5. Here, we aimed to investigate the role of CXCL13/CXCR5 signaling axis in BBB. The invasive ability of bEnd.3 cells was determined by the Transwell invasion assay. The barrier integrity of bEnd.3 cells was assessed by detecting trans-endothelial electrical resistance, the permeability to fluorescein isothiocyanate-dextran, and the expression levels of the tight junction protein E-cadherin. Lipopolysaccharide (LPS)-activated microglia promoted invasion and barrier dysfunction, and upregulated CXCR5 and p-p38 expression levels in cocultured bEnd.3 cells. However, the effects of activated microglia were alleviated by knocking down CXCR5 in cocultured bEnd.3 cells. Furthermore, recombinant CXCL13 promoted invasion and barrier dysfunction, and upregulated the expression levels of p-p38 in bEnd.3 cells; however, its effects were abolished by treating bEnd.3 cells with the p38 inhibitor SB203580. Our data tentatively demonstrated that LPS-activated microglial cells may promote invasion and barrier dysfunction in bEnd.3 cells by regulating the CXCL13/CXCR5 axis and p38 signaling.


Assuntos
Barreira Hematoencefálica , Quimiocina CXCL13 , Células Endoteliais , Microglia , Receptores CXCR5 , Animais , Encéfalo/metabolismo , Quimiocina CXCL13/metabolismo , Células Endoteliais/metabolismo , Lipopolissacarídeos , Camundongos , Microglia/metabolismo , Receptores CXCR5/metabolismo
20.
Cancer Med ; 11(15): 2978-2989, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35621033

RESUMO

BACKGROUND: CARs are engineered receptors comprising an immunoglobulin single-chain variable fragment (scFv) that identifies and binds to the target antigen, a transmembrane domain, and an intracellular T-cell signaling domain. CD19 is a B lineage-specific transmembrane glycoprotein and is expressed in more than 95% of B-cell malignancies. Streptavidin (SA) is a homo-tetrameric protein derived from Streptomyces avidinii, which can bind four biotin molecules with an extremely high affinity at a Kd value of 10-15 M. AIMS: The aim of the study is to generate a novel soluble multimeric fusion protein, sCD19-streptavidin (sCD19-SA) for functional detection and selective expansion of CD19-targeted CAR-T cells. METHODS: The fusion proteins CD19-SA was expressed in CHO cells and purified by use of Ni-nitrilotriacetic acid agarose beads. RESULTS: A novel fusion protein (sCD19-SA) was generated, consisting of the extracellular domain of human CD19 and the core region of SA, and could be used to functionally detect CD19-targeted CAR-T cells. Furthermore, this protein was demonstrated to form multimers to activate CAR-T cells to induce their selective expansion. Importantly, sCD19-SA-stimulated CD19-targeted CAR-T cells could improve antitumor effects in vivo. CONCLUSIONS: Our study has highlighted the potential of utilizing antigen-SA fusion proteins such as sCD19-SA for CAR-T therapy for the functional detection of CAR expression and selective expansion of CAR-T cells.


Assuntos
Antígenos CD19 , Imunoterapia Adotiva , Animais , Cricetinae , Cricetulus , Humanos , Estreptavidina , Linfócitos T
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...