Current Issue

  • Select all
    |
    Contents
  • Contents
    2025, 39(4): 0.
    Abstract ( ) Download PDF ( )   Knowledge map   Save
  • ORIGINAL ARTICLES
  • ORIGINAL ARTICLES
    ZHU Yunfeng, BIAN Xiangyu, CHANG Yanyun, YAO Zhanxin, LI Aiping, GAO Weina
    Abstract ( ) Download PDF ( )   Knowledge map   Save
    OBJECTIVE  To study the anti-fatigue effects of differently-cultivated Astragalus extract in a hypoxic environment of the plateau and explore the related mechanisms. METHODS  Fifty-six male KM mice were randomly divided into the hypoxic swimming control (HSC) group, imitation wild Astragalus extract (IWA) 430, 860 and 1 720 mg·kg-1 groups, and cultivated Astragalus extract (CA) 463, 925 and 1 850 mg·kg-1 groups. The drug was administered by gavage once daily for 15 days, while body mass was monitored every three days. After 15 days of gavage, the mice were subjected to load swimming (5% body weight) in a hypobaric chamber (simulating a 4 000 m altitude), with exhaustive swimming time measured to identify the optimal dosage. Following randomization, fifty male KM mice were assigned to five groups: normoxic control (NC), hypoxic control (HC), HSC, IWA 860 and CA 925 mg·kg-1. All groups underwent daily gavage for 15 d before 90 min non-weight-bearing swimming was conducted in the HSC, IWA 860 and CA 925 mg·kg-1 groups within a hypobaric chamber, followed by immediate measurement of muscle strength. Hematoxylin-eosin (HE) staining was used to observe the histopathological changes in liver and gastrocnemius muscle tissues. Blood urea nitrogen (BUN), blood glucose (BG) and serum lactic acid (LA), glutathione (GSH), glutathione peroxidase (GSH-Px), malondialdehyde (MDA), superoxide dismutase (SOD), liver glycogen (LG) and muscle glycogen (MG) in livers and muscles, and total antioxidant capacity (T-AOC) and reactive oxygen species (ROS) in muscles were measured by commercial kits. Taurine and hypotaurine were measured by HPLC. Enzyme-linked immunosorbent assay (ELISA) was used for cysteine sulfenic acid decarboxylase (CSAD) measurement. Western blotting was used to detect protein expressions of phosphatidylinositol 3-kinase (PI3K), protein kinase B (Akt), nuclear factor E2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) in skeletal muscles. RESULTS  Compared with the HSC group, the swimming time was prolonged in IWA 463, IWA 860, CA 925 and CA 1 850 mg·kg-1 groups. Compared with the HSC group, the muscle strength of mice in the IWA 860 mg·kg-1 group and the CA 925 mg·kg-1 group was significantly increased, histopathological damage in the liver and gastrocnemius muscle was reduced, serum levels of LA and BUN were significantly decreased, levels of BG, LG and MG were significantly increased, levels of GSH, GSH-Px and SOD were significantly increased,  contents of MDA were significantly decreased, expressions of CSAD were significantly increased in liver tissue, contents of GSH, T-AOC, taurine and hypotaurine were significantly increased, levels of ROS were significantly decreased, and protein expressions of PI3K, Akt, Nrf2, HO-1 were significantly upregulated in muscle tissues. CONCLUSION  Under simulated high-altitude hypoxic conditions, extracts of Astragalus membranaceus cultivated by two methods consistently exhibit anti-fatigue effects. Its mechanisms may be mitigating oxidative stress, augmenting taurine and hypotaurine metabolic regulation, and activating  PI3K/Akt and Nrf2/HO-1 signaling pathways. IWA has a better anti-fatigue effect than CA.
  • ORIGINAL ARTICLES
    LI Jing, ZHANG Kaidi, XIAO Zhiyong, ZHOU Wenxia
    Abstract ( ) Download PDF ( )   Knowledge map   Save
    OBJECTIVE  To investigate the regulatory effect of Rasfonin on SOS1 (Son of Sevenless, one of the major guanylate exchange factors) expressions and the underlying mechanism. METHODS  ① Human cancer cells MCF-7 (breast cancer cells, KRASWT wild-type), Calu-1 (non-small cell lung cancer, KRASG12C mutation), and UM-UC-3 (bladder metastatic cell carcinoma, KRASG12C mutation) were divided into the control group and Rasfonin (1, 5, 10 and 15 μmol·L-1) treated groups. CCK-8 assay was used to observe the effects of Rasfonin on the proliferation of MCF-7, Calu-1, and UM-UC-3 cells after 24 h of Rasfonin treatment. In addition, these cells were divided into the control group, EGF stimulation group (EGF 50 μg·L-1, stimulated for 5 min), and Rasfonin treated groups (pretreated with 5 and 10 μmol·L-1 Rasfonin before 5 min EGF stimulation). Quantitative real-time PCR (real-time fluorescence PCR) and Western blotting were employed to identify the expression levels of SOS1 mRNA and protein in MCF-7, Calu-1 and UM-UC-3 cells. ② The co-expression systems of KRAS and SOS1 were established by transfecting plasmids (KRAS-NC, KRASWT, KRASG12C and SOS1) into 293T cells that were divided into the control group and Rasfonin (1, 5 and 10 μmol·L-1) treated group. The dual luciferase reporter gene assay was used to evaluate the effects of Rasfonin on activities of the SOS1 promoter. Moreover, 293T cells were divided into the EGF stimulation group (EGF 50 μg·L-1, stimulated for 5 min) and Rasfonin treated groups (12 h of treatment with 10 μmol·L-1 Rasfonin before 5 min EGF stimulation). Western blotting was performed to determine the role of KRASG12C protein in the inhibition of Rasfonin on SOS1 expression. RESULTS  ① Compared with the control group, Rasfonin inhibited the proliferation of Calu-1 and UM-UC-3 cells at concentrations of 5, 10 and 15 μmol·L-1 (IC50 was 8.22 and 4.94 μmol·L-1). But for MCF-7 cells, only 15 μmol·L-1 Rasfonin could decrease their viability (IC50 was 45.15 μmol·L-1). Compared with the EGF stimulation group, mRNA expressions of SOS1 were increased after Rasfonin treatment of 1 h. mRNA expressions of SOS1 were decreased in Calu-1 cells after 3 h of Rasfonin treatment. These changes also occurred after Rasfonin treatment of 3 h and 6 h in UM-UC-3 cells. Furthermore, Rasfonin treatment did not influence SOS1 protein expressions in MCF-7 cells, but can significantly inhibit SOS1 expression of in UM-UC-3 and Calu-1 cells. ② Rasfonin had no significant effects on the activity of SOS1 promoter and its protein level in 293T cells when only SOS1 was expressed, but significantly inhibited its activity and its protein level when SOS1 was co-expressed with KRAS protein. CONCLUSION  One of the anti-tumor mechanisms of Rasfonin is to inhibit the activity of SOS1 promoter to decrease mRNA and protein expressions of SOS1 through KRASG12C protein.
  • ORIGINAL ARTICLES
    WU Chao, YANG Xiaomin, XU Xiaojia, SHI Xiaowan, LIU Yu, ZHANG Mingsheng
    Abstract ( ) Download PDF ( )   Knowledge map   Save
    OBJECTIVE  To investigate the vasodilatory effect of dimethyl sulfoxide (DMSO) on isolated aortic arteries and intrarenal arteries of rats and the mechanisms. METHODS  ① Rat aortas and intrarenal arteries were pre-contracted with KCl 60 mmol·L-1, U46619 0.3 μmol·L-1 or PE 3 μmol·L-1. Following equilibration, dimethyl sulfoxide (DMSO) was cumulatively administered at concentrations of 0.1%, 0.3%, 1.0%, 3.0% and 10.0%. Changes in the vascular tension of aortic and intrarenal arterial rings were recorded using an isolated vessel tension measurement system. Rat intrarenal arteries were pre-contracted with KCl 60 mmol·L-1, U46619 0.3 μmol·L-1 or PE 3 μmol·L-1. Cumulative additions of DMSO (3.0% and 10.0%) were administered as the control group. Following pre-contraction with each of the three stimulants, the voltage-gated potassium channel (Kv) inhibitor 4-AP (0.5 mmol·L-1) was incubated for 15 min. Cumulative additions of DMSO (3.0% and 10.0%) were then administered, and the vascular relaxation percentages induced by DMSO before and after treatment were calculated. Whole-cell patch-clamp recordings were performed on acutely isolated intrarenal arterial smooth muscle cells of rat to assess Kv currents during cumulative DMSO applications (0.1%, 0.3%, 1.0% and 3.0%). ② Rat aortic smooth muscle cells (A7r5) were exposed to DMSO concentration of 0.0% (control), 0.1%, 0.3%, 1.0% and 3.0% for 24 h. Intracellular reactive oxygen species (ROS) levels were detected using the DCFH-DA fluorescent probe. Malondialdehyde (MDA) content, catalase (CAT) activity, and superoxide dismutase (SOD) activity in A7r5 cells were measured by chemical colorimetry. Mitochondrial membrane potential was evaluated using the JC-1 fluorescent probe. RESULTS  ① DMSO (0.1%-10.0%) dose-dependently relaxed rats aortic and intrarenal arteries pre-contracted with either KCl 
    60 mmol·L-1, U46619 0.3 μmol·L-1 or PE 3 μmol·L-1. The values of maximum relaxation were (42.3±9.7)%, (73.2±8.4)%, (99.2±4.7)% and (84.0±1.9)%, (80.5±6.1)% and (81.2±4.4)%, respectively. Compared with the control group, vasorelaxation of DMSO on IRAs precontracted with U46619 was significantly attenuated by Kv inhibitor 4-AP. ② Low concentrations (0.1% and 0.3%) of DMSO significantly increased Kv currents, while high concentrations (1.0% and 3.0%) of DMSO significantly decreased Kv currents. High concentrations (1.0% and 3.0%) of DMSO remarkablely increased ROS and MDA levels, but significantly decreased CAT activity, SOD activity and mitochondrial membrane potential in A7r5 cells. CONCLUSION DMSO (0.1%-10.0%) can relax rats aortal and intrarenal arteries in a concentration-dependent manner, and the mechanism may be related to Kv, oxidative stress and mitochondrial damage.
  • ORIGINAL ARTICLES
    XIAO Zhenyu, SUN Yangyang, YANG Yi, MA Yingkai, WANG Yongan, LUO Yuan
    Abstract ( ) Download PDF ( )   Knowledge map   Save
    OBJCTIVE  To evaluate the therapeutic efficacy of intranasal administration of pirfenidone in treating paraquat-induced pulmonary fibrosis in mice across treatment durations. METHODS  Eight-week-old male C57BL/6 mice were randomly divided into six groups (n=8 per group): the normal control group (saline), pirfenidone control group, paraquat group, and three treatment groups receiving a combination of paraquat and pirfenidone for 15, 10 and 5 d, respectively. Except the normal and pirfenidone control groups, all the mice received intraperitoneal injection of paraquat (35 mg·kg-1) to induce pulmonary fibrosis. In the treatment groups, pirfenidone (20 mg·kg-1) was delivered intranasally once daily, beginning on days 1, 6, and 11 post-paraquat exposure, until day 15. Fifteen days after paraquat exposure, pulmonary function tests, micro-CT imaging, and arterial blood gas analysis were performed. Histopathological changes and collagen fiber deposition in lung tissues were examined using HE and Masson staining respectively.The protein expression levels of fibrosis markers, including fibronectin (FN), collagen typeⅠ(CollⅠ), E-cadherin (E-cad), vimentin (Vim), and α-smooth muscle actin (α-SMA), were detected by Western blotting.  Additionally, inflammatory and pro-fibrotic cytokines, such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), IL-8, IL-1β, and connective tissue growth factor (CTGF), were quantified using ELISA. RESULTS  Compared with the normal control group, mice treated with paraquat exhibited significant respiratory alterations, including prolonged expiratory time (TE), increased enhanced pause (PENH), and reduced tidal volume (TV). CT imaging revealed reticular high-density shadows and ground-glass opacities in paraquat-treated mice. Blood gas analysis showed reduced partial pressure of oxygen (PaO2), oxygen saturation of blood (SaO2), fractional oxygen saturation of hemoglobin (FO2Hb), and central venous oxygen saturation (ScvO2), along with increased partial pressure of carbon dioxide (PaCO2) and fractional deoxyhemoglobin saturation (FHHb), indicating that mice exposed to paraquat exhibited severe hypoxemia and hypercapnia. Histological evaluation highlighted pronounced lung interstitial thickening, alveolar collapse, inflammatory infiltration, and extensive fibrotic changes marked by collagen accumulation. Furthermore, exposure to paraquat significantly increased the protein levels of FN, CollⅠ, vimentin, and α-SMA, markedly reduced E-cadherin levels and elevated the levels of inflammatory cytokines (TNF-α, IL-6, IL-8 and IL-1β) as well as the pro-fibrotic cytokine CTGF. Pirfenidone treatment demonstrated time-dependent efficacy, with the 15 d group showing the most significant improvements in pulmonary function as evidenced by reduced PENH levels and increased TV, EV and MV. CT imaging revealed a decrease in high-density opacities and improved lung transparency after pirfenidone treatment. In addition, arterial blood gas measurements indicated markedly elevated levels of PaO2, SaO2 and FO2Hb. Histological analysis showed that pirfenidone alleviated lung interstitial thickening, reduced inflammatory cell infiltration, and decreased collagen deposition. At the molecular level, pirfenidone significantly reduced the protein expressions of FN, CollⅠ, Vim and α-SMA, while increasing E-cad levels. Furthermore, inflammatory cytokines, particularly IL-6 and IL-1β, were notably suppressed following pirfenidone intervention. CONCLUSION  Intranasal administration of pirfenidone can exhibit potent time-dependent anti-fibrotic efficacy in paraquat-induced pulmonary fibrosis, with early interventions delivering the most substantial therapeutic benefits.
  • ORIGINAL ARTICLES
    GUO Shuai, XING Huanchun, CAO Wenbin, WANG Lin, CHEN Aibing, WANG Yongan, YANG Jun
    Abstract ( ) Download PDF ( )   Knowledge map   Save
    OBJECTIVE  To establish skin contamination models with sulfur mustard analogue 1-chloro-2-ethylsulfanyl ethane (CEES), and evaluate the therapeutic efficacy of reactive skin decontamination lotion kit (RSDL). METHODS  ① Kunming, BALB/c, BALB/c-nu, and C57BL/6N were contaminted with CEES 75 μL·kg-1 using exposed method and covered method for 10 min on the dorsal skin. Wound healing times and areas were assessed to determine the stock of mice and exposure method. ② BALB/c mice were exposed to a gradient of CEES at the doses of 75, 150, 250, 350 and 500 μL·kg-1 (10 min) using the covered method. Wound healing times, areas and Kaplan-Meier survival curves were used to determine the optimal dose. ③ BALB/c mice were exposed to CEES 150 μL·kg-1 for varying durations (5, 10 and 20 min) using the covered method. Wound healing times, areas and Kaplan-Meier survival curves were analyzed to determine the optimal exposure time. ④ BALB/c mice were divided into four groups: control (exposed with the covered method at 150 μL·kg-1, 10 min), treatment (20 μL RSDL treatment after exposure, as the control group), 5 min treatment (20 μL RSDL treatment after 5 min of exposure, followed by 5 min of coverage) and immediate-treatment (exposed with the exposed method at 150 μL·kg-1, immediately treated with 20 μL RSDL, followed by 10 min of coverage). The therapeutic efficacy was evaluated based on the wound area, subcutaneous microvesicle count, epidermal thickness, and inflammatory cytokine (IL-6 and TNF-α) expressions. RESULTS  ① The covered method caused more severe and prolonged wounds than the exposed method. BALB/c mice exhibited a high sensitivity to CEES with delayed wound healing and were therefore selected as the model animal. 
    ② Survival rates in BALB/c mice dropped below 50% at doses of 250, 350 and 500 μL·kg-1, whereas an 83.3% survival rate was observed at 150 μL·kg-1. ③ The mice exposed to CEES (150 μL·kg-1, 20 min) died within 3 days. The wound area was consistently smaller in the 5 min covered group than in the 10 min covered group. ④ CEES-induced skin injury led to epidermal nuclear pyknosis, follicular disruption, inflammatory infiltration, and microvesicle formation in both treatment and poisoned control groups. As more immediate treatment, the wound area significantly decreased. While the IL-6 expression showed no significant intergroup difference, the TNF-α expression was significantly higher in the treatment group. CONCLUSION  A CEES-induced skin contamination model has been established in BALB/c mice using the covered method (150 μL·kg-1, 10 min covered). However, RSDL should be administered in 10 min post-contamination. 
  • ORIGINAL ARTICLES
    FU Dawei, FU Yujin, YAN Lailai, CHEN Jie, LIU Zhiyu, FU Juanling, YAO Biyun, HAO Weidong, ZHAO Peng
    Abstract ( ) Download PDF ( )   Knowledge map   Save
    OBJECTIVE  To establish an in vitro simulated one compartment extravascular administration model with lanthanum nitrate as the test substance, and explore the differences between this model and the classic in vitro administration model in lanthanum nitrate induced HepG2 cell death. METHODS  An in vitro administration device was designed based on compartment model theories which consisted of four functional chambers: the liquid storage chamber, mixing chamber, toxicant exposure chamber, and waste liquid receiving chamber. The four chambers were connected by peristaltic pump hoses. The peristaltic pumps were employed to ensure unidirectional and constant speed transmission of liquid between these chambers. According to the preset toxicokinetic parameters such as T1/2a and T1/2, an in vitro simulated one compartment extravascular administration model of lanthanum nitrate was constructed using the device. The content of lanthanum nitrate in the toxicant exposure chamber at different time points was measured using inductively coupled plasma mass spectrometry. The concentration-time curves of lanthanum nitrate were analyzed using PKsolver and GraphPad Prism 8.0 software. The constructed in vitro simulated one compartment extravascular administration model was evaluated by comparing the measured and theoretical values of toxicokinetic parameters. HepG2 cells were treated with lanthanum nitrate in the in vitro simulated one compartment extravascular administration model and classic in vitro administration model, respectively, and cell death was measured using the Hoechst 33342/propidium iodide staining method. RESULTS  Within the Cmax range of 3.91-1 000.00 μmol·L-1, the measured concentration-time curves of lanthanum nitrate in the toxicant exposure chamber almost conformed with the corresponding calculated theoretical curves (the correlation coefficients were all>0.998 0). The measured values of toxicokinetic parameters, including Ke, T1/2, Ka, T1/2a, Tmax, Cmax, CL and AUC0-∞, were close to the corresponding theoretical values. The fitting coefficients (R2) of the concentration-time curves for each experimental group were all>0.990 0, which was consistent with one compartment model for extravascular administration. In the simulated one compartment extravascular administration model, no significant death of HepG2 cells was observed in any lanthanum nitrate dose group. In the classic in vitro administration model, the cell death rate of the 0.500 mmol·L-1 lanthanum nitrate group was higher than that of the solvent control group, but no significant cell death was observed in the 0.119 mmol·L-1 group or 0.243 mmol·L-1 group. When Cmax or Cadministration was 0.500 mmol·L-1, classic in vitro administration induced a higher cell death rate than simulated one compartment extravascular administration. However, there was no statistically significant difference in lanthanum nitrate induced HepG2 cell death between the two administration models when the AUC was equal. CONCLUSION  The device designed in this study can be used to in vitro simulate one compartment extravascular administration, making in vitro toxicity testing more similar to in vivo scenarios, and providing data for optimizing administration methods of in vitro toxicity testing. There are differences in lanthanum nitrate induced HepG2 cell death between simulated one compartment extravascular administration and classic in vitro administration, indicating that different in vitro exposure modes can affect toxicity.
  • REVIEWS
  • REVIEWS
    HAN Dongmei, ZHOU Di, ZHANG Yi, WEI Kaikun
    Abstract ( ) Download PDF ( )   Knowledge map   Save
    Mesenchymal stem cells (MSCs) have become a key focus in the development of cell-based therapeutic products due to their wide availability, lack of ethical constraints, and potential for immune privileges. However, the transition from basic MSC transplantation techniques to fully developed therapeutic drugs presents numerous challenges given the current regulatory framework in China and from the perspective of drug development and review. This article summarizes the problems faced in this transition, particularly the challenges posed by theheterogeneity of MSC sources, the complexity of unique manufacturing processes, and the complexities of quality characterization. The article also offers suggestions and countermeasures in the hopes of advancing the research, development, and registration of MSC-based therapeutic products.
  • REVIEWS
    PENG Yunhui, ZHANG Shikun, JI Shouping
    Abstract ( ) Download PDF ( )   Knowledge map   Save
    Extracellular vesicles (EVs) are subcellular components released by cells, which can carry proteins, lipids, mRNA and miRNA, and other biomolecules. They play a significant role in maintaining the intracellular environment and mediating intercellular communication. In recent years, EVs have attracted much attention in the field of drug delivery. Platelet-derived extracellular vesicles (P-EVs) not only possess the advantages of extracellular vesicles but also integrate the unique properties of platelets. P-EVs have unique application value in in vivo drug delivery. This article reviews the extraction methods of P-EVs, their targeting mechanisms and drug loading methods as drug delivery carriers, as well as the research progress in the treatment of various diseases, providing new solutions and ideas for drug application and disease treatment.
  • REVIEWS
    GAN Xinyue, SONG Juxingsi, ZOU Shuaijun, WANG Qianqian, ZHANG Liming
    Abstract ( ) Download PDF ( )   Knowledge map   Save
    Biotoxins are natural chemical substances produced by animals, plants or microorganisms that exhibit toxicity toward other species. These biotoxins exhibit a wide range of structural complexities. Some of them are highly toxic, posing significant threats to human health and public safety. In recent years, researchers have employed the cell membranes of biotoxin target cells to coat nanomaterials, thereby constructing biomimetic nanosystems with high biocompatibility, low immunogenicity, 
    prolonged circulation time, and strong targeting capabilities. By leveraging the interactions between biotoxins and their target cell membranes, these systems effectively trap toxins, leading to notable advancements in this field. This article reviews the toxic mechanisms of biotoxins, the historical development of using biomimetic nanosystems for toxin detoxification, common preparation methods and characterization techniques, as well as the composition and detoxification efficacy of these nanosystems, thus providing references for their future design, development, and application.