For Authors|For Editors|For Reviewers

20 July 2024, Volume 36 Issue 186
    

  • Select all
    |
    Review
  • Yanping Xing, Xianli Lan, Xiyu Liu, Yong Huang
    Discovery Medicine. 2024, 36(186): 1316-1333. https://doi.org/10.24976/Discov.Med.202436186.122
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    The detection of tumor markers is crucial for assessing the progression of specific cancers. Numerous research studies have shown that immunosensors can convert immune-specific response biosignals into visual signals, enabling the highly sensitive tracking and detection of tumor markers. This offers a promising solution for early cancer diagnosis. However, most tumor markers are inert molecules that are challenging to detect at low concentrations in the early stages of cancer. Therefore, there is a need to develop immunosensor analysis platforms with a higher sensitivity. Nanomaterials, with their advantages of high stability, low cost, and versatility in design, have emerged as ideal candidates for enhancing the performance of immunosensor analysis. In this paper, we review the design ideas of nanomaterials in antibody-based electrochemical, electrochemiluminescent, and photoelectrochemical immunosensors, including electrode interface modification, signaling probes for stimulating sensing signals, and design strategies of modified materials in signaling mechanisms. In addition, we have thoroughly analyzed the performance, advantages and disadvantages of different immunosensors. Therefore, the aim of this paper is to review the recent advances in advanced nanomaterial strategies for different immunosensors and their biomedical applications, and to point out the challenges and prospects of immunosensors in future clinical applications.

  • Teng Cai, Yanyan Lin, Wenbo Meng
    Discovery Medicine. 2024, 36(186): 1334-1344. https://doi.org/10.24976/Discov.Med.202436186.123
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    N-acetyltransferase 10 (NAT10) is an important acetyltransferase that regulates telomerase activity and participates in DNA damage reactions, ribosomal RNA (rRNA) transcriptional activation, cell division, microtubule acetylation, and other important cellular processes. Abnormalities in the expression or distribution of NAT10 result in diseases such as Hutchinson-Gilford progeria syndrome (HGPS) and various tumors, with serious consequences. Remodelin, an inhibitor of NAT10, delays HGPS progression; many studies have been conducted on its role in tumor therapy. A major breakthrough in the study of NAT10 was the discovery of mRNA N4-acetylcytidine (ac4C) modification, which can increase mRNA stability and translation efficiency significantly. In addition, NAT10 modifies the mRNA of ac4C, which is associated with tumor development. Here, we present a review of pertinent studies focusing on NAT10, particularly its role in cancer, to provide researchers with a concise and informative summary of the current state of knowledge about this topic. The conclusions drawn from this review could provide a new direction for tumor treatment.

  • Guang Liu, Hong Zhang
    Discovery Medicine. 2024, 36(186): 1345-1353. https://doi.org/10.24976/Discov.Med.202436186.124
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Lactylation plays an important regulatory role in neural development, neural stem cell fate determination, and the physiological function of the nervous system. Abnormal lactylation is closely related to the occurrence and development of hippocampal microglial inflammation in post-traumatic stress disorder (PTSD), so lactylation may serve as a potential therapeutic target for PTSD. This article reviewed the latest research progress on the involvement of lactylation in hippocampal microglial inflammation and its molecular mechanisms in PTSD.

  • Jixin Lin, Yan Chen, Ting Li, Cheng Zhu, Yaju Qiu, Enyan Yu
    Discovery Medicine. 2024, 36(186): 1354-1362. https://doi.org/10.24976/Discov.Med.202436186.125
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Major depressive disorder (MDD) is a clinical condition that significantly impacts patients' physical and mental well-being, quality of life, and social functioning. The pathogenesis of MDD remains unclear, but accumulating evidence suggests a close relationship between gut microbiota and the occurrence and progression of MDD. Gut microbiota refers to the microbial community in the human intestine, which engages in bidirectional communication with the host via the “gut-brain axis” and plays a pivotal role in influencing the host's metabolism, immune system, endocrine system, and nervous system. Modulating gut microbiota entails restoring the balance and function of the intestinal flora through methods such as probiotic intake, fecal transplantation, and dietary intervention. Such modulation has been shown to effectively alleviate depressive symptoms in the host. This review synthesizes recent advancements in research on gut microbiota modulation for ameliorating depressive symptoms and can serve as a foundation for further exploration of the gut microbiota's role in MDD and its potential therapeutic benefits.

  • Article
  • Franziska Haumaier, Theresa Dregelies, William Sterlacci, Raja Atreya, Michael Vieth
    Discovery Medicine. 2024, 36(186): 1363-1369. https://doi.org/10.24976/Discov.Med.202436186.126
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Ulcerative colitis is a well-known inflammatory bowel disease. Patients have an increased risk of developing colitis associated carcinoma (CAC). It is important for patient management to be able to distinguish between ulcerative colitis associated carcinoma and sporadic carcinoma (sCRC). However, this distinction is frequently very challenging. It is not readily possible to differentiate this histologically. However, the diagnosis is crucial for the patient's further treatment and follow-up. An attempt was therefore made to develop a diagnostic regime that would enable a reliable distinction between sCRC and CAC.

    Methods: We screened 96 patients analyzing more than 850,000 methylation hotspots, to detect distinct epigenetic patterns between both types of carcinomas. Patients with sporadic carcinoma and colitis-associated carcinoma as well as patients with normal colon and patients with confirmed ulcerative colitis without neoplasia were used for the analysis. By extensively filtering the results, methylation sites relevant to distinguish between CAC and sCRC were identified.

    Results: After the results were filtered, three methylation sites relevant to distinguish between CAC and sCRC were identified. For this purpose, methylation limit values were defined, which favor the samples as CAC or sCRC up to a certain methylation value of the methylation sites. The combination of three methylation sites allows a correct assignment to CAC or sCRC in 94.5% of the cases.

    Conclusion: The results show that these three methylation sites are promising markers in the diagnosis of CAC vs sCRC. Nevertheless, the diagnosis should always be made in conjunction with histomorphological analyses.

  • Xin-ying Li, Chun-rong Zhong, Jin-chan Wu, Cai-hong Yuan, Jian-min Ran
    Discovery Medicine. 2024, 36(186): 1370-1377. https://doi.org/10.24976/Discov.Med.202436186.127
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Glucose and fatty acid overload-induced glucolipid toxicity of pancreatic β-cells is associated with the development of diabetes. Endoplasmic reticulum stress (ERS) plays an essential role in this process. Ghrelin, a peptide secreted by the pancreas, negatively correlates with oxidative stress. The study aimed to investigate ghrelin's role in glycolipid-induced β-cell dysfunction and its possible mechanism.

    Methods: Mouse insulinoma β-cell, NIT-1 cells, were stimulated with high fat and high glucose to induce glucolipid toxicity. High fat and high glucose-induced NIT-1 cells were treated with acylated ghrelin (AG) or [d-Lys3]-growth hormone releasing peptide (GHRP)-6. Flow cytometry and Cell Counting Kit-8 (CCK-8) assay were performed to assess apoptosis and cell viability. The protein expression related to apoptosis, inositol-requiring kinase 1 (IRE1)/c-Jun N-terminal kinase (JNK) signaling, and ERS were investigated using western blot. Enzyme-linked immunosorbent assay (ELISA) was adopted to examine insulin's synthesis and secretion levels.

    Results: Ghrelin treatment improved cell viability while inhibiting cell glucolipotoxicity-induced NIT-1 cell apoptosis. Ghrelin can promote the synthesis and secretion of insulin in NIT-1 cells. Mechanistically, ghrelin attenuates ERS and inhibits the IRE1/JNK signaling pathway in NIT-1 cells induced by glucolipotoxicity.

    Conclusion: Ghrelin improves β-cellular dysfunction induced by glucolipotoxicity by inhibiting the IRE1/JNK pathway induced by ERS. It could be an effective treatment for β-cellular dysfunction.

  • Tian Dai, Yunfang Xiao, Hong Zhang, Yingying Shi, Fangyuan Wu
    Discovery Medicine. 2024, 36(186): 1378-1385. https://doi.org/10.24976/Discov.Med.202436186.128
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Objective: To investigate the alleviating effect of chlorogenic acid (CGA) on oxidative damage in high glucose (HG)-induced HK-2 cells and to explore its potential mechanisms.

    Methods: We cultured the human proximal tubular cell line HK-2 and divided them into the control group and different concentrations of CGA groups (0, 5, 10, 25, 50, 100, 200 μM). The trypan blue dye test was used to detect CGA's potential cytotoxicity on HK-2 cells. Then, we treated HK-2 with HG and CGA; the Cell Counting Kit-8 (CCK-8) method was used to detect the cell viability of HK-2 cells in each group. Flow cytometry was employed to measure the apoptosis rate of cells. Western blot was performed to detect the expression of apoptosis proteins B-cell lymphoma-2 (BCL-2), BCL-2-associated X protein (BAX), cysteinyl aspartate specific proteinase (CASPASE)-9, and CASPASE-3. In addition, enzymatic activities, including superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), and lipid peroxide (LPO), were measured with the corresponding detection kits. 2′,7′-Dichlorodihydrofluorescein diacetate (DCFH-DA) assay and flow cytometry were performed to detect reactive oxygen species (ROS) production. Western blot analysis and Reverse Transcription-Polymerase Chain Reaction (RT-PCR) were conducted to evaluate protein and mRNA expressions of the Kelch-like ECH-associated protein-1 (KEAP1)/Nuclear factor erythroid 2-related factor 2 (NRF2)/Antioxidant Response Elements (ARE) signaling pathway.

    Results: The outcomes showed that, in a dose-dependent way, CGA dramatically increased the vitality of HK-2 induced by HG. Furthermore, CGA significantly reduced the HG-stimulated HK-2 cell apoptosis, which may be linked to the promotion of BCL-2 and the suppression of BAX, cleaved-CASPASE-3, and cleaved-CASPASE-9 expression. In HK-2 cells, CGA reduced the formation of ROS generated by HG levels and markedly boosted the activity of the antioxidant enzymes SOD, GSH-Px, and CAT. Furthermore, compared with the HG group, CGA significantly raised NRF2 nuclear expression and downregulated NRF2 cytosolic expression and increased the mRNA expression of NRF2 and its target genes, heme oxygenase-1 (HO-1), KEAP1, and NAD(P)H dehydrogenase quinone 1 (NQO1).

    Conclusion: These results show that CGA might be useful in managing oxidative damage in HG-induced HK-2 cells.

  • Aiping Hu, Yuqing She, Yang Wang, Xue Cao, Ran Li, Jing Wang, Lizhi Yu, Haifeng Jiang, Yang Zhao
    Discovery Medicine. 2024, 36(186): 1386-1397. https://doi.org/10.24976/Discov.Med.202436186.129
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: More and more evidence has shown the process of Parkinson's disease (PD). Probably, inflammation exerts a crucial role between them. Therefore, the aim of this study was to analyze the impact of interleukin-6 receptor (IL-6R) expression on the IL-6/signal transducer and activator of transcription 3 (STAT3)/hypoxia-inducible factor-1α (HIF-1α) inflammatory signaling pathway within a mouse model of PD with type 2 diabetes mellitus (T2DM) as co-morbidity.

    Methods: We chose healthy wild-type C57BL/6J male mice at the age of 10 weeks to prepare a mouse model of PD with T2DM co-morbidity. Adeno-associated virus (AAV) overexpressing IL-6R or AAV IL-6R-shRNA genes were injected into the substantia nigra (SN) of the mice. The behavioral indices of the pole test were used for examining the motor function of the mice. Using immunofluorescence analysis, the impacts of IL-6R on the level of tyrosine hydroxylase (TH) and anti-ionized calcium-binding adaptor molecule 1 (IBA-1) on dopaminergic neurons and microglia were examined. Additionally, enzyme-linked immunosorbent assay (ELISA) was adopted for determining the expressions of HIF-1α and inflammatory cytokines like tumor necrosis factor-α (TNF-α), IL-1β, IL-6, and IL-4 in the serum. In this study, the protein expression levels of TH, α-Synuclein (α-Syn), IBA-1, IL-6, IL-6R, phosphorylated and total signal transducer and activator of transcription 3 (p-STAT3 (Tyr705) and STAT3) and HIF-1α in the SN were tested via western blotting. To ascertain the mRNA expressions of TNF-α, IL-1β, IL-6, IL-4, and HIF-1α, we used quantitative Real-Time Polymerase Chain Reaction (RT-qPCR).

    Results: IL-6R-shRNA treatment could markedly shorten the total time of PD in the T2DM co-morbidity mouse model based on the pole test results, reverse the decrease in TH-positive neurons stimulated by 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP), and lower the activation of microglia (all p < 0.05). Further, IL-6R-shRNA treatment hindered the expression of IL-6, p-STAT3 (Tyr705), and HIF-1α in the SN, lowered the levels of TNF-α, IL-1β, IL-6, IL-4, and HIF-1α in the serum, and mRNA expressions of TNF-α, IL-1β, IL-6, and HIF-1α in the SN (all p < 0.05). In contrast, IL-6R overexpression reduced TH levels, upregulated the level of IBA-1, IL-6, p-STAT3 (Tyr705), and HIF-1α, increased the level of IL-1β, TNF-α, IL-6, IL-4, and HIF-1α (all p < 0.05) in the serum and SN in the PD mouse model with T2DM as a co-morbidity.

    Conclusions: PD progression with T2DM as a co-morbidity can be boosted by AAV IL-6R-overexpression through upregulation of the IL-6/STAT3/HIF-1α axis. Conversely, AAV IL-6R-shRNA treatment suppressed the IL-6/STAT3/HIF-1α pathway and alleviated neuroinflammation, thus weakening the development of PD with T2DM as a co-morbidity.

  • Xiaosheng Lv, Yuwen Song, Yanyan Shi, Yinghong Xue, Shuncheng Ge, Chao Liu
    Discovery Medicine. 2024, 36(186): 1398-1407. https://doi.org/10.24976/Discov.Med.202436186.130
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Subarachnoid hemorrhage (SAH) is a severe cerebrovascular disease, often leading to neuroinflammation and neuronal damage. Activation of the Nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome is closely associated with post-SAH neuroinflammation, while activation of Nicotinamide Adenine Dinucleotide (NAD)-dependent deacetylase sirtuin-1 (SIRT1) has neuroprotective effects. This study aimed to investigate the impact of injectable Collagen Binding Domain-Brain Derived Neurotrophic Factor (CBD-BDNF) on neuroinflammation and neuronal damage following SAH.

    Methods: After establishing the SAH model, experimental animals were divided into three groups: sham surgery group (Sham), SAH group, and SAH+neuroregenerative scaffold (CBD-BDNF treatment) group. Behavioral performance was evaluated using neurofunctional deficit, beam balance, and Y-maze tests. Expression of inflammatory factors and essential proteins was quantitatively analyzed using Enzyme-Linked Immunosorbent Assay (ELISA) kits and immunoblotting. Terminal deoxynucleotidyl transferase dUTP Nick End Labeling (TUNEL) staining was used to assess cell apoptosis. To further investigate the mechanism of action of CBD-BDNF on SIRT1, the model animals were treated with EX527 (SIRT1 inhibitor) for comparative studies.

    Results: Neurological deficit tests, CBD-BDNF improves functional outcomes after SAH. Compared to the SAH group, the SAH+neuroregenerative scaffold group showed significantly increased expression of SIRT1 protein and significantly decreased expression of NLRP3, Apoptosis-associated speck-like protein containing a CARD (ASC), and c-caspase-1. The inflammatory cytokines Interleukin-1 beta (IL-1β), IL-6, and IL-18 levels also significantly decreased in the SAH+neuroregenerative scaffold group. Additionally, animals in the SAH+neuroregenerative scaffold group showed better neurofunctional recovery in neurofunctional deficit and beam balance tests. The number of apoptotic cells significantly decreased in the SAH+neuroregenerative scaffold group compared to the SAH group. However, when SIRT1 was inhibited with EX527, the aforementioned neuroprotective effects were reversed, indicating the involvement of CBD-BDNF through SIRT1 activation.

    Conclusion: This study demonstrates that injectable CBD-BDNF can significantly alleviate neuroinflammation and neuronal damage resulting from SAH by blocking NLRP3 inflammasome activation and promoting SIRT1 expression. These findings provide a new therapeutic strategy for neuroprotection after SAH and reveal the mechanism of action of CBD-BDNF as a potential therapeutic agent. Future research will further explore the long-term efficacy and safety of CBD-BDNF.

  • Daoxu Huang, Zengpai Zheng, Yiling Huang, Xiang Chen
    Discovery Medicine. 2024, 36(186): 1408-1419. https://doi.org/10.24976/Discov.Med.202436186.131
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Obesity is linked to impaired intestinal barrier function and inflammation. Saikosaponin A (SSA), a triterpene saponin from Bupleurum chinense, has shown beneficial effects on intestinal colitis in mice. However, the mechanisms underlying SSA's protective effects against obesity are not fully understood.

    Objective: To investigate the effects of SSA on body weight, metabolic disturbances, and intestinal health in diet-induced obese (DIO) mice, and to elucidate the potential mechanisms involved.

    Methods: In the in vivo study, DIO mice were supplemented with SSA. Body weight, fasting blood glucose, and metabolic parameters were measured. Intestinal barrier function and inflammation were assessed. In the in vitro study, intestinal epithelial cells were treated with palmitic acid and lipopolysaccharide to induce inflammation. SSA was then administered to evaluate its effects on cell barrier integrity and inflammatory responses. The role of the nuclear factor-E2-related factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathway was investigated by silencing Nrf2.

    Results: SSA supplementation significantly (p < 0.05) decreased body weight and fasting blood glucose levels in DIO mice, and markedly improved metabolic disturbances. This treatment also enhanced intestinal barrier function and reduced metabolic inflammation, likely through increased antioxidant capacity of intestinal epithelial cells via activation of the Nrf2/ARE signaling pathway. In vitro, SSA maintained cell barrier integrity and reduced inflammatory responses by activating the Nrf2/ARE signaling pathway, decreasing intracellular reactive oxygen species content, and increasing transepithelial electrical resistance. However, silencing Nrf2 abolished SSA's protective effects.

    Conclusion: SSA enhances the antioxidant capacity of intestinal epithelial cells, maintains intestinal barrier integrity, and reduces intestinal inflammation in DIO mice through the activation of the Nrf2/ARE signaling pathway. These findings offer new insights into the protective role of SSA in obesity and metabolic diseases.

  • Shimaa Saad El-Din, Basma Emad Aboulhoda, Amira Hassouna, Marwa M. Shakweer, Mansour A. Alghamdi, Doha Essam, Mohamed Essam, Nawaf A. AlRakaf, Hanaa M. Elzahed, Asmaa Selmy, Dina Sabry, Dina M. Mekawy
    Discovery Medicine. 2024, 36(186): 1420-1429. https://doi.org/10.24976/Discov.Med.202436186.132
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Bone marrow-derived mesenchymal stem cells (BM-MSCs) have recently attracted great attention due to their crucial anti-inflammatory and regenerative properties. This work aims to examine the curative impact of intra-articular injection of BM-MSCs-derived exosomes in ameliorating osteoarthritis (OA) progression in rats and to explore the interaction between circular RNA of Yes-associated protein 1 (circYAP1) and microRNA-21 (miRNA-21) in the rat knee joints.

    Methodology: Gene expression circYAP1, miRNA-21, toll-like receptor-7 (TLR7), aggrecan, and collagen type II were evaluated by quantitative reverse transcription polymerase chain reaction (qRT-PCR) in the rat articular tissues. In addition, the Enzyme-linked immunosorbent assay (ELISA) technique was used to estimate the level of the inflammatory markers interleukin 4 (IL-4), interleukin 10 (IL-10), interleukin 1β (IL-1β), and tumor necrosis factor-alpha (TNF-α); and the oxidative markers glutathione (GSH), malondialdehyde (MDA) and total reactive oxygen species (ROS). Histopathological examination using Hematoxylin and Eosin (H&E) staining of the rat articular tissue was also performed along with an estimation of the articular cartilage thickness.

    Results: Our results showed that BM-MSCs-derived exosomes significantly elevated circYAP1 gene expression level (p < 0.05) along with subsequent downregulation of miRNA-21 and TLR7 (p < 0.05). These effects impacted the inflammatory milieu of rat articular surfaces, where there was a significant reduction (p < 0.05) of the pro-inflammatory and oxidative markers with significantly increased production of the anti-inflammatory and antioxidative markers (p < 0.05). Marked elevation in aggrecan and collagen type II gene expression was also found in the treated groups (p < 0.05).

    Conclusion: Those data suggest that BM-MSCs-derived exosomes have a crucial role in mitigating OA symptoms and pathology progression and might be regarded as an effective as well as acceptable treatment option for OA.

  • Tao Zhou, Yajie Chen, Chao Wang, Zhiyong Huang, Ziming Tan, Yan Ma
    Discovery Medicine. 2024, 36(186): 1430-1440. https://doi.org/10.24976/Discov.Med.202436186.133
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Keloid, a fibroproliferative disorder, significantly impacts patients' quality of life, yet effective therapies remain elusive. This study explored the role of silent information regulator 6 (SIRT6) in modulating the proliferation, invasion, and collagen synthesis of keloid fibroblasts.

    Methods: Keloid and normal skin specimens were collected, and fibroblasts were isolated from the keloid tissue. SIRT6 recombinant adenovirus (Ad) was constructed to infect keloid fibroblasts to overexpress SIRT6. This study entails three groups: Control group, adenovirus-Negative Control (Ad-NC) group, and Ad-SIRT6 group. SIRT6 protein and mRNA levels were measured via Western blotting and Quantitative reverse transcription polymerase chain reaction (qRT-PCR), respectively. Cell viability was determined using 5-ethynyl-2′-deoxyuridine (EdU) assay. Flow cytometry was exploited to measure cell apoptosis. To investigate cell migration, wound healing assay and Transwell assay were employed. Western blotting was also utilized to study the expression levels of apoptotic proteins, collagen deposition-related proteins, and Mitogen-Activated Protein Kinases (MAPK)/extracellular regulated protein kinases (ERK) pathway-related proteins.

    Results: Compared to the control and Ad-NC groups, the Ad-SIRT6 group exhibited significantly elevated SIRT6 level; diminished cell proliferation, migration and invasion; reduced protein levels of α-smooth muscle actin (α-SMA), collagen I, collagen III, phospho SMAD Family Member 3 (p-Smad3), transforming growth factor-β 1 (TGF-β1), and MAPK/ERK pathway proteins (phospho extracellular signal-regulated protein kinase 1/2 (p-ERK1/2), phospho MAP kinase-ERK kinase (p-MEK) and phospho-c-Raf (p-c-Raf)). Treatment with epidermal growth factor (EGF), an MAPK/ERK pathway agonists, reversed the inhibitory effect of SIRT6 on cell activity and inhibited apoptosis in keloid fibroblasts.

    Conclusion: SIRT6 overexpression in keloid fibroblasts attenuates proliferation, invasion, and collagen synthesis, while fostering apoptosis, likely through the suppression of MAPK/ERK pathway activity. This suggests a potential therapeutic target for keloid treatment.

  • Yong Hu, Ming Li, Xiao-yan Chen, Chun-hang Huang, Hui-yuan Cao, Guang-ji Wang, Xiu-fan Du
    Discovery Medicine. 2024, 36(186): 1441-1452. https://doi.org/10.24976/Discov.Med.202436186.134
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Synovial inflammation plays a crucial role in osteoarthritis (OA). Gastrodin (GAS), an active ingredient derived from the Gastrodia elata Blume rhizome, possesses antioxidant and anti-inflammatory pharmacological effects. This research aimed to evaluate the function and molecular mechanism of GAS on human fibroblast-like synoviocytes of osteoarthritis (HFLS-OA) induced by interleukin (IL)-1β.

    Methods: The impact of GAS on the viability of IL-1β-treated HFLS-OA cells was assessed using the cell counting kit-8 (CCK-8). Quantitative real-time reverse transcription PCR (qRT-PCR) was employed to detect changes in IL-8, IL-6, monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor (TNF)-α, and Gremlin-1 mRNA expression in each group. Corresponding kits were utilized to measure the catalase (CAT) and superoxide dismutase (SOD) activities, as well as the nitric oxide (NO) level. Western blot analysis was conducted to examine the expression of extracellular matrix degradation-associated proteins and nuclear factor kappa-B (NF-κB) pathway-correlated proteins in each group.

    Results: GAS significantly promoted the proliferation of IL-1β-induced HFLS-OA cells and concurrently down-regulated Gremlin-1 mRNA expression (p < 0.05). Through the down-regulation of Gremlin-1 expression, GAS exhibited the following effects: decreased IL-8, IL-6, and TNF-α mRNA expression, as well as NO levels (p < 0.05); increased SOD and CAT activities (p < 0.05); down-regulated matrix metallopeptidase 13 (MMP-13) and MMP-1 protein expression levels (p < 0.01); and up-regulated collagen II protein expression level (p < 0.01) in IL-1β-treated HFLS-OA cells. Additionally, GAS decreased phospho-inhibitory kappa B (p-IκB)/IκB, phospho-inhibitory kappa B kinase (p-IKK)/IKK, and p-p65/p65 ratios in IL-1β-induced HFLS-OA cells by inhibiting Gremlin-1 expression (p < 0.01).

    Conclusion: GAS demonstrates a positive impact on inflammation, oxidative stress, and extracellular matrix degradation in IL-1β-mediated HFLS-OA cells. This effect is achieved by suppressing Gremlin-1 expression and reducing NF-κB pathway activity.

  • Wei Meng, Chang Shu, Hong Gao
    Discovery Medicine. 2024, 36(186): 1453-1463. https://doi.org/10.24976/Discov.Med.202436186.135
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Levosimendan (Levo) is a drug commonly used to treat heart failure. Recent studies have suggested that Levo may have neuroprotective effects, but it is still unknown how exactly it contributes to hypoxia-induced brain damage. Thus, the aim of this study was to investigate how Levo affects hypoxia-induced brain damage and to clarify any possible underlying mechanisms.

    Methods: One group of rats (Levo group) was pretreated with Levo via oral force-feeding for four weeks. Another group (Ferrostatin-1 (Fer-1) group) was pretreated with intraperitoneal injections of Fer-1 for four weeks. A rat model of chronic hypoxia was created by treating rats with 13% O2 for 14 days in a closed hypoxia chamber. For each group (Control, Model, Levo, Fer-1), we evaluated learning and memory capacity and the morphology and structure of neurons in the rats' brain tissue. Other measurements included tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6); malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px); Fe2+; apoptosis; cleaved caspase-3, caspase-3; phosphatase and tensin homolog (PTEN), protein kinase B (Akt), phosphorylated Akt (p-Akt); and ferroptosis-related proteins Nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11).

    Results: The Model group rats had considerably fewer neurons than the Control group, with loosely arranged cells, and markedly impaired learning and memory abilities (p < 0.05). Oxidative damage and inflammation in brain tissues of the Model group were significantly intensified, accompanied by a substantial increase in neuronal apoptosis (p < 0.05). PTEN protein, Fe2+ concentration, and cleaved caspase-3 expression were all significantly upregulated, whereas p-Akt, Nrf2, GPX4, and SLC7A11 proteins were dramatically downregulated (p < 0.05). Both the Levo and Fer-1 groups demonstrated significantly more neurons and closely arranged cells than the Model group, along with a notable improvement in learning and memory abilities (p < 0.05). Oxidative damage and inflammation in brain tissues of the Levo and Fer-1 groups were markedly alleviated, and neuronal apoptosis was suppressed (p < 0.05). p-Akt, Nrf2, GPX4, and SLC7A11 proteins were dramatically upregulated, whereas the expression of cleaved caspase-3, PTEN protein, and Fe2+ content was considerably downregulated (p < 0.05).

    Conclusions: Levo effectively mitigates brain injury in rats with chronic hypoxia, likely by regulating ferroptosis via the PTEN/Akt signaling pathway.

  • Qingcong Li, Yan Li, Jiayu Liu, Xin Huang, Zikang Li
    Discovery Medicine. 2024, 36(186): 1464-1476. https://doi.org/10.24976/Discov.Med.202436186.136
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Monotherapy consisting of radiotherapy or chemotherapy has limited efficacy in pancreatic tumors. This study aims to investigate whether the combination of 125I brachytherapy and gemcitabine (GEM) chemotherapy has a synergistic effect on pancreatic cancer (PC).

    Methods: In vitro, PANC-1 cells in the exponential phase were treated with 125I radioactive seeds (6 Gy) and GEM (30 nM). Cell proliferation, apoptosis, and mitochondrial membrane potential were measured using the Cell Counting Kit-8 (CCK-8) assay, Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, and flow cytometry, respectively. In vivo, we examined the inhibitory effect of three different treatment regimens on tumor growth in mice when combined with 125I brachytherapy and GEM. Next, we investigated the effects of the optimal scheme among the three on the tumor microenvironment, tumor tissue morphology, tumor cell apoptosis, systemic inflammatory response, and levels of apoptosis-related proteins in the tumor. Changes in the tumor microenvironment and levels of apoptosis-related proteins were measured by Western blot. The extent of damage to tumor tissue morphology was assessed by Hematoxylin and Eosin (HE) staining. Tumor cell apoptosis was measured by TUNEL staining. Changes in inflammation-related factors were determined by Enzyme-Linked Immunosorbent Assay (ELISA).

    Results: The results of in vitro cell experiments demonstrated that the combination of 125I radioactive seeds (6 Gy) and GEM (30 nM) had a stronger inhibitory effect on PANC-1 cells than either alone (p < 0.05). In vivo, data showed that the GEM (after 3 d) + 125I treatment group had the strongest tumor inhibition effect on PC (p < 0.05). Western blot analysis showed that the combined treatment of 125I brachytherapy and GEM caused changes in the expression of collagen and connexin in the tumor microenvironment, promoted tumor cell apoptosis, upregulated the expression of pro-apoptotic proteins, and helped to restore pancreatic function (p < 0.01).

    Conclusion: Our research results suggest that the strategy of 125I seed implantation surgery in mice after 3 days of GEM treatment has a more pronounced synergistic effect on the treatment of PC.

  • Xu-hui Zhang, Jie-qiong Zhou, Qing Wei, Jian Li, Ting Xu, Chun-mei Bai, Jian-feng Zhou, Xi-cheng Wang
    Discovery Medicine. 2024, 36(186): 1477-1485. https://doi.org/10.24976/Discov.Med.202436186.137
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Metastatic colorectal cancer (mCRC) is increasingly characterized by myriad genomic alterations beyond the well-known factors such as RAS, BRAF, and microsatellite instability (MSI). Novel genomic changes, including ERBB2 amplifications, mutations, and gene fusions, are now recognized as potential targets for precision therapy. This study aims to explore the genomic landscape of a Chinese cohort with mCRC to identify potentially targetable genetic alterations for personalized treatment strategies.

    Methods: A total of 500 mCRC patients in China were enrolled, based on which genomic profiling was performed using capture-based targeted sequencing across a panel of 520 genes on tumor tissues to identify prevalent genomic alterations. The mutations were analyzed by optimized proprietary algorithms. MSI and mismatch repair deficiency status were analyzed using the read-count-distribution approach. Besides, the overall survival (OS) related to these molecular changes was estimated.

    Results: The cohort's genomic profiling revealed TP53 mutations in 78%, APC in 60%, and KRAS in 47% of the patients. MSI-High status was confirmed in 5.8% of cases via a next-generation sequencing (NGS)-based algorithm. ERBB2/HER2 amplifications were found in 12% (60/500) of patients, with potential therapeutic implications for those without concurrent KRAS mutations. A subset of patients (1.2%; 6/500) showed fusions and DNA damage response (DDR) gene mutations (except TP53) that could be targeted therapeutically. The KRAS (G12C) variant was detected in 14 patients (2.8%), and 61 (12.2%) had a BRAF V600E mutation. Notably, survival analysis showed no significant differences in OS between KRAS mutant loci and NRAS mutations (p = 0.436). However, BRAF V600E mutations were associated with a poorer prognosis than BRAF wild-type and non-V600E mutations (16.3 months vs. 29.5 and 31.1 months, respectively; p < 0.001).

    Conclusions: This study validates the feasibility of using NGS to detect prognostic and therapeutically actionable genetic variants in Chinese mCRC patients, contributing to understanding the genomic variation within this population and highlighting the potential for personalized medicine in managing mCRC.

  • Dandan Qiu, Lingling Gao, Xingwei Yu
    Discovery Medicine. 2024, 36(186): 1486-1498. https://doi.org/10.24976/Discov.Med.202436186.138
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: The previous study has proved the oncogenic role of Yes-associated protein 1 (YAP1) in bladder cancer (BLCA), thus this study focused on its impact on bladder cancer stem cells (BCSCs) and underlying mechanism.

    Method: BCSCs were obtained by treating human BLCA cells UMUC3 with cisplatin and identified by measuring CD133+ in UMUC3/BCSCs via flow cytometry. YAP1 interaction proteins and mothers against decapentaplegic homolog 7 (SMAD7) N6-methyladenosine (m6A) site were analyzed by bioinformatics. BCSCs were transfected. SMAD7 m6A level, YTH domain-containing family proteins 3 (YTHDF3)-SMAD7 interaction, YAP1/YTHDF3 expression in BCSCs were assessed by methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP) or quantitative reverse transcription PCR (qRT-PCR), respectively. BCSC proliferation was detected by 5-Bromo-2-deoxyuridine (BrdU) staining. UMUC3/BCSC migration/invasion and tumour sphere formation were determined by Transwell or tumour sphere formation assays. YAP1/YTHDF3/SMAD7/transforming growth factor (TGF)-β1/stemness marker expressions in UMUC3/BCSCs were measured by Western blot assay.

    Result: BCSCs showed higher CD133+ ratio, expressions of stemness marker/YAP1/YTHDF3/TGF-β1, lower SMAD7 expression and greater invasion/migration/tumour sphere formation capabilities than UMUC3 cells. YAP1 knockdown decreased SMAD7 m6A level and impaired YTHDF3-SMAD7 interaction in BCSCs. YAP1 silencing inhibited cell growth/invasiveness/migration/tumour sphere formation and stemness-associated protein/YTHDF3/TGF-β1 expressions while upregulating SMAD7 expression in BCSCs, which was offset by YTHDF3 overexpression.

    Conclusion: The silencing of YAP1 in BCSCs impedes the YTHDF3-mediated degradation of m6A-modified SMAD7, culminating in diminished cell stemness.

  • Enjian Guan, Junping Zhang, Juntao Zhou, Yanna Liu, Ning Xu
    Discovery Medicine. 2024, 36(186): 1499-1512. https://doi.org/10.24976/Discov.Med.202436186.139
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Melanoma is a highly invasive skin cancer with limited treatment strategies. Bupivacaine, a commonly used local anesthetic recognized for its safety, has shown promise in combating tumors. 3′-phosphoadenosine 5′-phosphosulfate synthase 2 (PAPSS2) is a key enzyme in the sulfation process and is associated with the development and metastasis of various tumors. This study aimed to explore the mechanism by which bupivacaine inhibits melanoma proliferation and metastasis by targeting PAPSS2.

    Methods: The effects of bupivacaine on the proliferation of A375 and A2058 melanoma cells were evaluated using Cell Counting Kit-8 (CCK-8), 5-Ethynyl-2'-deoxyuridine (EdU) labeling, and clonogenic assays. Cell migration, invasion, and PAPSS2 expression were evaluated using Transwell experiments and Quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR) analysis. Additionally, an in vivo melanoma tumor model in nude mice was constructed to evaluate the impact of bupivacaine on melanoma growth and metastasis. Immunohistochemistry was used to assess tumor metastasis and PAPSS2 expression levels in the nude mouse model.

    Results: Experimental results demonstrated that bupivacaine significantly inhibited melanoma proliferation and invasion compared to the control group. Notably, this inhibitory effect was partially reversed by PAPSS2 overexpression. In vivo experiments demonstrated that bupivacaine-treated nude mice exhibited reduced tumor volumes, weights, and fewer lung metastatic foci. Molecular analysis via qRT-PCR and immunohistochemistry analysis further indicated that bupivacaine significantly reduced PAPSS2 in tumor tissues.

    Conclusion: This study confirms that bupivacaine, a local anesthetic, can inhibit melanoma proliferation and metastasis by targeting the PAPSS2 signaling pathway. These findings suggest its potential as an anti-tumor medication and present new treatment strategies for melanoma.

  • Rashid Mir, Imadeldin Elfaki, Mohammad A Alanazi, Naseh A. Algehainy, Faisal H Altemani, Badr A Alsayed, Elsiddig Idriss Mohamed, Syed Khalid Mustafa, Mamdoh S Moawadh, Faris J Tayeb, Jaber Alfaifi, Sael M Alatawi, Mohammad Muzaffar Mir, Mohammad Fahad Ullah
    Discovery Medicine. 2024, 36(186): 1513-1526. https://doi.org/10.24976/Discov.Med.202436186.140
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: In recent years, various coronaviruses have caused severe respiratory illnesses worldwide. For example the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections of COVID-19 outbreak in 2019 in Wuhan, China. Genome-wide association studies (GWAS) have significantly expanded our comprehension of how specific genetic variations are linked to diseases. Research has demonstrated the existence of genetic factors influencing susceptibility to coronaviruses. The objective of this study was to examine the association of certain loci with the COVID-19 in Saudi population.

    Methods: In the present study we have examined the link between the COVID-19 disease and certain genetic variants in hospitalized COVID-19 patients (n = 16) in Tabuk and Bisha, Kingdom Saudi Arabia. We used the genome Analysis Toolkit (GATK) and Comprehensive variant annotation was performed different databases and tools such as Search Tool for the Retrieval of Interacting Genes (STRING), PanelApp and PolyPhen-2.

    Results: The study showed that the genetic variants associated with genes such as Homeostatic Iron Regulator (HFE) (found in 7 patients, representing 44%), complement factor H (CFH) (6 patients, 38%), cadherin 23 (CDH23) (4 patients, 25%), cytotoxic T-lymphocyte associated protein 4 (CTLA-4) (3 patients, 19%), Transforming Growth Factor Beta 1 (TGFB1) (3 patients, 19%), CREB-binding protein (CREBBP) (2 patients, 13%), E1A Binding Protein P300 (EP300) (2 patients, 13%), hemoglobin subunit beta (HBB) (2 patients, 13%), interferon regulatory factor 7 (IRF7) (2 patients, 13%), and unc-119 lipid binding chaperone (UNC119) (2 patients, 13%) might be associated with susceptibility to coronavirus. We also identified mutations in the COVID-19 patient that are pathogenic or likely pathogenic.

    Conclusion: A recurrent pathogenic mutation, HFE p.His63Asp (H63D), was identified in 7 patients, suggesting its potential contribution to disease severity. Additionally, a likely pathogenic variant, HBB p.Glu7Val (E7V), was present in 2 patients, highlighting its potential role in disease susceptibility. Our results shed light on the key genetic mechanisms of COVID-19 pathogenesis and help to identify and stratify the individuals or populations that are at risk to corona virus infection. The identification of susceptible individuals or populations assist in prevention and/or in treatment programs.

  • Systematic Review
  • Sileshi Mulatu, Gebrehiwot Berie Mekonnen, Yeshimebet Tamir Tsehay, Sosina Tamre Mamo, Mengistu Abebe Messelu, Alamirew Enyew Belay, Asnake Gashaw Belayneh, Ousman Adal, Chernet Tafere
    Discovery Medicine. 2024, 36(186): 1527-1543. https://doi.org/10.24976/Discov.Med.202436186.141
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Feeding diversified food for children is the major indicator of nutritional quality and adequacy that is crucial during the complementary feeding period for infants and young children aged 6–23 months. Ensuring diversified food is highly essential for the normal growth and development of the infant and young children. In Ethiopia, malnutrition and food insecurity remain prevalent, underscoring the need to understand and improve dietary diversity among children. The primary objective of this review was to determine the pooled prevalence of dietary diversity and its associated factors among children aged 6–23 months in Ethiopia.

    Methods: We thoroughly searched some electronic databases, including Pub Med, Africa Index Medicus, Science Direct, Hinari, and Google Scholar, to perform a meta-analysis. Excel was used to extract and combine the data, while Stata 17 was used for statistical analysis. To estimate pooled prevalence rates and related associated factors, we used a random-effect model and the Der Simonian-Laird technique. The I2-test was utilized to examine heterogeneity, and funnel plots, in conjunction with Egger's and Begg's tests, were employed to investigate publication bias.

    Result: This review analyzed 42 full-text studies, finding a pooled prevalence of 26.78% (95% confidence interval (CI): 23.35–30.21) with significant heterogeneity (I2 = 98.95%). Maternal education levels—college & above Adjusted Odds Ratio (AOR: 5.377, 95% CI: 3.116–9.279), secondary and above (AOR: 3.324, 95% CI: 1.939–5.700), primary (AOR: 3.065, 95% CI: 2.275–3.129), and formal education (AOR: 2.484, 95% CI: 1.722–3.583)—showed higher odds than counterparts. Similarly, fathers' education—secondary and above (AOR: 2.837, 95% CI: 1.981–4.065) and primary (AOR: 2.082, 95% CI: 1.016–4.266)—and father's occupation as merchant (AOR: 2.739, 95% CI: 1.355–5.539), and mother's occupation as housewife (AOR: 3.636, 95% CI: 2.457–5.381) showed higher odds. Additionally, male child sex (AOR: 1.877, 95% CI: 1.185–2.972), child age 18–23 months (AOR: 2.470, 95% CI: 1.568–3.987), and 12–17 months (AOR: 2.460, 95% CI: 1.914–3.163) indicated higher odds than counterparts. Having Postnatal Care (PNC) follow-up, counseling on infant and young child feeding (IYCF) practices, and no history of child illness were associated with higher odds (AOR: 3.155, 95% CI: 2.104–4.732), (AOR: 2.960, 95% CI: 2.288–3.829), and (AOR: 2.420, 95% CI: 1.765–3.318), respectively. Maternal knowledge of dietary diversity, urban residency, Antenatal Care (ANC) follow-up, child growth monitoring, and media exposure also showed higher odds. Similarly, maternal age groups 25–34 years and 35–44 years had higher odds compared to those aged 15–24 years. Other factors associated with higher odds included home grading, food security, institutional delivery, availability of cow milk, and household wealth index.

    Conclusion: Among Ethiopian children aged 6–23 months, the prevalence of recommended dietary diversity feeding practices was remarkably low, with only about 25% meeting the minimum recommended diversified food. The scientific predictors factor affecting dietary diversity included maternal media exposure, place of delivery, food security, urban residency, availability of cow milk, child growth monitoring, age, and knowledge of IYCF practices; paternal factors like education and occupation; child-related variables like age, sex, and history of illness; and history of ANC and PNC.