For Authors|For Editors|For Reviewers

20 March 2024, Volume 36 Issue 182
    

  • Select all
    |
    Review
  • John Dawi, Scarlet Affa, Abraham Chorbajian, Yura Misakyan, Aishvaryaa Shree Mohan, Brandon Norris, Jacob Ochsner, Sonyeol Yoon, Vishwanath Venketaraman
    Discovery Medicine. 2024, 36(182): 437-447. https://doi.org/10.24976/Discov.Med.202436182.41
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    This research project delves into the multifaceted dynamics of Mycobacterium tuberculosis (M.tb) endocarditis, a significant yet uncommon manifestation of tuberculosis (TB). Beginning with an overview of M.tb and the global challenges posed by TB, we navigate through the bacterium's evolution, transmission modes, and the intricate host immune response. The pathology and pathophysiology of M.tb endocarditis are explored, emphasizing its complexities and the host's efforts to contain the pathogen. The study extends to atypical mycobacterial endocarditis, highlighting the emergence of species like M.chimaera, M.fortuitum, and M.chelonae, with a focus on their association with life-threatening mycobacterial endocarditis. Clinical presentations and complications of M.tb endocarditis are detailed, addressing challenges in diagnosis, drug-resistant, co-infections with Human Immunodeficiency Virus (HIV), and potential sepsis. The research underscores the need for a deeper understanding of M.tb endocarditis to enhance prevention, diagnosis, and treatment strategies. Examining the genetic and environmental factors influencing M.tb endocarditis, the study discusses the interplay of immune-related genes, environmental conditions, and predispositions contributing to infection susceptibility. Despite challenges in treatment due to its rarity, the research highlights current protocols, surgical interventions, and promising pharmaceutical developments. Lastly, unraveling these intricate factors is crucial for refining strategies and conducting large-scale trials to address this global health threat effectively.

  • Qixin Leng, Aishwarya Anand, Archibald James Mixson
    Discovery Medicine. 2024, 36(182): 448-456. https://doi.org/10.24976/Discov.Med.202436182.42
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Although Warburg discovered pH discrepancies between tumor and normal tissues nearly 100 years ago, developing therapies to take advantage of this concept was relatively slow for the first 70 years. During the last 30 years, there has been an exponential increase in the use of pH-dependent strategies for both low molecular weight drugs and nanoparticles. Two frequently discussed approaches are the chemotherapy's release from pH-sensitive covalent linkages of macromolecules or from pH-dependent disruption of charged polymeric nanoparticles. In contrast, pH-dependent non-covalent bonds between the chemotherapy agent and macromolecules have rarely been discussed, yet this underappreciated strategy has great potential. These non-covalent interactions are primarily ionic or hydrogen bonds with supporting roles from hydrophobic bonds. In addition to the facile coupling of the drug with the carrier, these non-covalent interactions may show marked pH dependence. Consistent with pH dependence, many of these drug-loaded carriers showed significant in vitro and, in some cases, striking in vivo activity. In this review, we will focus on pH-sensitive non-covalent bonds, highlighting the release of drugs from diverse carriers such as tetrahedron DNA structures, cyclodextrin, polymeric carriers, and carbon-based quantum particles.

  • Moeka Nakashima, Naoko Suga, Yuka Ikeda, Sayuri Yoshikawa, Satoru Matsuda
    Discovery Medicine. 2024, 36(182): 457-466. https://doi.org/10.24976/Discov.Med.202436182.43
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Chitosan seems to be an innovative biological material potentially utilized as a nanoparticle carrier for drug delivery, which could be low toxic, biocompatible, and easy to prepare. Chitosan nanoparticles have been employed in gene delivery. As a type of multifunctional adjuvant, chitosan nanoparticles could activate the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway to induce cell protection and/or proliferation via the modulation of autophagy within dendritic cells. In general, adjuvants may improve the innate and/or adaptive immune responses to a vaccine antigen by facilitating the antigen presentation of antigen presenting cells such as dendritic cells. The choice of a suitable adjuvant has become vital for improved safety and/or expanded application of vaccines. Fortunately, chitosan nanoparticles could be designed to target the dendritic cells to be enhanced by its adjuvant effect and for stimulating robust immune responses. Therefore, chitosan nanoparticles may be a good immune stimulant with encouraging properties for the development of superior vaccine delivery. Indeed, vaccines could play a key role in human health. In this review, we summarize the concept and/or recent progress in the field of chitosan nanoparticles, providing a valuable resource for investigating the molecular mechanisms of chitosan for the development of a greater vaccine.

  • Karl Livori, Jean Calleja-Agius
    Discovery Medicine. 2024, 36(182): 467-481. https://doi.org/10.24976/Discov.Med.202436182.44
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Endometriosis is a medical condition affecting at least up to 10% of women of reproductive age. This condition occurs when ectopic endometrial glands and stroma implant outside the uterus and there are several theories regarding the underlying origins of the disease. Endometriosis is one of the major causes of severe dysmenorrhoea, chronic pelvic pain and infertility. While endometriosis is generally a non-malignant condition, it rarely may transform into an invasive cancer, and increase the risk for epithelial ovarian cancer, notably endometrioid or clear cell ovarian cancer. Despite the increased risk, the mechanisms behind the development of endometriosis-associated ovarian cancer (EAOC) are not yet well understood. Recent investigations have delved into the intricate interplay between endometriosis and EAOC, exploring pathways involving oxidative stress, inflammation, hyperestrogenism, and the discovery of genetic mutations within endometriotic lesions that hint at a transition towards invasive carcinoma. Efforts have been made to identify intermediary lesions between endometriosis and EAOC, which may enable earlier detection of endometriosis at risk of malignant transformation or even prevention of the transformation altogether. However, given the rarity of this malignancy, there is still the risk of late or missed diagnosis, with the risk of inappropriate management being offered to the patient, and the higher risk of poor prognosis and increased morbidity and mortality. This scoping review aims to summarize existing data on EAOC, with a focus on endometrioid and clear cell histologic subtypes. It also provides insights into its identification, prognosis, and delineating management strategies, seeking to provide a holistic understanding of the complexities surrounding EAOC, facilitating further research and the development of more effective prevention and treatment approaches.

  • Ruoyan Zhang, Haiwen Chen, Yanning Ma, Zuolin Jin
    Discovery Medicine. 2024, 36(182): 482-493. https://doi.org/10.24976/Discov.Med.202436182.45
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    With the rapid development of dental artificial intelligence systems (DAIS), a new field known as “Data Dentistry”, proposed by Schwendicke in 2021, has successfully bridged the gap between medicine and engineering. This literature review introduces advanced techniques in data collection, outlines the current state of DAIS in data processing, and anticipates the future of DAIS by emphasizing the importance of more extensive and enhanced datasets. The key findings include: Versatility of imaging data: Various types of imaging data, such as X-ray, cone beam computed tomography (CBCT), facial photos, and face and oral scans, can be transformed into datasets used by artificial intelligence systems. Uniform rules in electronic dental record (EDR) systems: EDR systems require standardized rules for general use in DAIS, ensuring compatibility and seamless integration. Potential of wearable device data: Data from wearable devices, including bioelectric signals (such as electromyography), stress sensors, AR glasses, etc., show great potential for enhancing DAIS capabilities. Current DAIS performance focus: Presently, DAIS demonstrate superior performance in object location and disease diagnosis compared to information integration and clinical decision-making. Need for data quality and quantity improvement: Further improvements are needed in both the quality and quantity of data for DAIS.

  • Article
  • Yiwen Ru, Hong Gu, Lian Sun, WeiBing Zhang, Lin Wang
    Discovery Medicine. 2024, 36(182): 494-508. https://doi.org/10.24976/Discov.Med.202436182.46
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Mandibular distraction osteogenesis (MDO) is a highly effective method for bone regeneration, commonly employed in treating craniofacial defects and deformities. Osteocytes sense mechanical forces in the pericellular space, relay external stimuli to biochemical changes, and send signals to other effector cells, including bone marrow mesenchymal stem cells (BM-MSCs), to regulate bone resorption and formation. Piezo1 potentially affects the secretion signal molecules of bone cells under mechanical stretch. The primary aim of this study was to enhance our comprehension of the molecular biology underlying this therapeutic approach and to identify specific signaling molecules that facilitate bone formation in response to stretch forces.

    Methods: Mechanical stretching was applied to negative controls and Piezo1 knockdown osteocyte-like MLO-Y4 cells. Alkaline phosphatase and Alizarin Red S staining were used to survey the osteogenic potential of BM-MSCs. The production and secretion content of adenosine triphosphate (ATP) was measured using ATP content determination analysis. Pathway-related and osteo-specific genes and proteins were evaluated using real-time polymerase chain reaction (RT-PCR), Western blots, and immunofluorescence. Mitochondrial organization was examined with a transmission electron microscope.

    Results: The conditioned medium of stretch-exposed MLO-Y4s significantly upregulated osteogenesis-related indicators of BM-MSCs (p < 0.001). The upregulation of BM-MSC osteogenesis was associated with ATP release from osteocytes. Mechanically induced calcium transfer and transcriptional coactivator with PDZ-binding motif (TAZ) nuclear translocation mediated by Piezo1 could promote mitochondrial fission and ATP release. Osteocytes detected stretch forces through Piezo1, triggering calcium influx, TAZ nuclear translocation, and ATP production.

    Conclusions: The stretch stimulation of Piezo1 induces calcium influx, which in turn promotes calcium-related TAZ nuclear translocation, changes in mitochondrial dynamics, and the release of ATP in osteocytes. This signaling cascade leads to an up-regulation in the osteogenic capacity of BM-MSCs. Mitochondrial energy metabolism of mechanosensitive protein Piezo1-dependent and ATP release may provide a new effective intervention method for mechanically related bone remodeling.

  • Wei Gu, Mimi Wu, Shihe Cui, Jinhua Bo, Hao Wu
    Discovery Medicine. 2024, 36(182): 509-517. https://doi.org/10.24976/Discov.Med.202436182.47
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Currently, the role of melatonin (MT) in neuronal damage remains unclear and this study aimed to explore the protective effects of MT on neurons in an in vitro cell injury model.

    Methods: The Sprague Dawley (SD) rat traumatic brain injury (TBI) model was prepared, and brain tissue extract (BTE) from the injured area were generated. To establish a cell injury model in vitro, the BTE was added to the culture medium during the neuron culture process. MT was introduced into the culture medium of the cell injury model to observe its protective effects on neurons. Relevant molecular biology experiments were conducted to observe cellular oxidative stress status, inflammation, endoplasmic reticulum (ER) stress, mitochondrial damage, and neuronal apoptosis.

    Results: When compared to the control group, the BTE group exhibited a significant increase in cellular oxidative stress, inflammation, neurofilament light polypeptide (NEFL) expression, and ER stress. Additionally, the mitochondrial DNA (mtDNA) copy number significantly decreased, and there was a higher count of apoptotic cells (p < 0.05). Upon the addition of MT to the culture medium of the in vitro cell injury model, there was a significant reduction in cellular oxidative stress, inflammation, and NEFL levels. This addition also mitigated ER stress, increased mtDNA copy numbers, and decreased the ratio of cell apoptosis (p < 0.05).

    Conclusions: In the in vitro cell injury model, MT demonstrates the capacity to inhibit cellular oxidative stress, inflammation, and ER stress levels. Additionally, it diminishes mtDNA damage, fosters cell viability, and serves as a protective agent against both apoptosis and necrosis in neurons.

  • Wei Guo, Shuyun Yan, Ge Zhao
    Discovery Medicine. 2024, 36(182): 518-526. https://doi.org/10.24976/Discov.Med.202436182.48
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Periodontitis is a chronic inflammatory disease resulting from bacterial plaque infection. While the involvement of activating transcription factor 1 (ATF1) has been extensively explored in various human diseases, its specific role in periodontitis remains unclear. This study aims to elucidate the expression and biological function of ATF1 in the context of periodontitis.

    Methods: Primary human periodontal ligament cells (hPDLCs) were procured from clinical samples and subsequently characterized. Following treatment with P. gingivalis lipopolysaccharide (LPS, 10 μg/mL), hPDLCs underwent transfection with either ATF1 vector or siRNA. The expression levels of ATF1 in LPS-treated hPDLCs or transfected cells were evaluated through real-time quantitative polymerase chain reaction (RT-qPCR) and western blot assay. Inflammatory factors, including interleukin-6 (IL-6), interleukin-8 (IL-8), tumor necrosis factor-alpha (TNF-α), and interleukin-1beta (IL-1β), were quantified using Enzyme-linked Immunosorbent Assay (ELISA). The assessment of osteogenic proteins, such as runt-related transcription factor 2 (Runx2), osteopontin (OPN), and osteoprotegerin (OPG), as well as noncanonical nuclear factor-kappaB (NF-κB) pathway-related proteins (p65, p-p65, IkBα, p-IkBα), was conducted using western blot assay. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay and flow cytometry assays were employed to detect cell viability.

    Results: LPS induced an inflammatory response and hindered the osteogenic differentiation of hPDLCs (p < 0.05, p < 0.01). Furthermore, ATF1 silencing enhanced cell proliferation and suppressed apoptosis in LPS-stimulated hPDLCs (p < 0.05, p < 0.01). ATF1 silencing not only restrained the inflammatory response but also promoted the osteogenic differentiation of LPS-stimulated hPDLCs (p < 0.05, p < 0.01). Importantly, ATF1 silencing effectively blocked the LPS-induced activation of the NF-κB signaling pathway (p < 0.05, p < 0.01, p < 0.001).

    Conclusions: ATF1 emerges as a promising treatment option, inhibiting the osteogenic differentiation of hPDLCs and mitigating the inflammatory response by preventing the phosphorylation of the NF-κB signaling pathway.

  • Rong Wang, Qiao-Yan Zhu, Wei-Wu Ye, Yuan Huang, Zhan-Hong Chen, Ya-Bing Zheng, Xiao Zou, Jian Wang, Dan-Lu Jiang, Xiao-Jia Wang, Zheng-Yang Xu, Wen-Ming Cao
    Discovery Medicine. 2024, 36(182): 527-537. https://doi.org/10.24976/Discov.Med.202436182.49
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: The clinical outcomes of chemotherapy (CT) for the treatment of metastatic triple-negative (TN) and hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (mBC) have proven to be disappointing. The phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway, a tumor-promoting signaling cascade frequently mutated in breast cancer (BC), has been implicated in chemoresistance. In this study, our objective is to investigate the efficacy and safety of combining everolimus with chemotherapy in mBC patients exhibiting mutations in the PI3K/AKT/mTOR pathway.

    Methods: We conducted a retrospective analysis to characterize the efficacy, safety, and their association with clinical and molecular characteristics of metastatic lesions in 14 patients with HER2- mBC. These patients harbored at least one altered member of the PI3K/AKT/mTOR signaling pathway and were treated with a combination of a chemotherapy agent and the mTOR inhibitor everolimus (CT+EVE).

    Results: The majority of patients belonged to the triple-negative (TN) subtype (9/14, 64.3%), having already undergone 2 lines of chemotherapy (CT) in the metastatic setting (11, 78.6%). These patients carried altered phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) and were administered a vinorelbine-containing regimen (10, 71.4%). The objective response rate (ORR) was 42.9%, with a disease control rate of 92.9%. The median progression-free survival (PFS) and overall survival (OS) were 5.9 (95% confidence interval (CI): 4.9–13.6) months and 14.3 (95% CI: 8.5–not reached (NR)) months, respectively. Patients with fewer prior treatment lines tended to exhibit longer PFS. OS, PFS, and ORR were comparable between hormone receptor-positive (HR+) and triple-negative breast cancer (TNBC) patients, but numerical improvements were noted in patients with a single PI3K pathway alteration compared to those with more than one alteration. Genomic alterations that surfaced upon progression on CT+EVE included cyclin dependent kinase 4 (CDK4) and epidermal growth factor receptor (EGFR) amplification, as well as neurofibromin 1 (NF1) mutation, suggesting potential mechanisms of acquired resistance. An analysis of adverse events indicated manageable toxicities.

    Conclusions: The findings of this study suggest both activity and safety for the combination of chemotherapy and the mTOR inhibitor everolimus (CT+EVE) in patients with HER2- mBC who have alterations in the PI3K pathway, particularly those who have received fewer prior chemotherapy. However, it is crucial to note that large-scale, randomized control studies are warranted to more comprehensively characterize the efficacy and safety of this combination therapy.

  • Fan Zhang, Guangping Li, Xueyin Li, Yi Zheng, Wanting Du, Bo Zhao, Da Sun
    Discovery Medicine. 2024, 36(182): 538-545. https://doi.org/10.24976/Discov.Med.202436182.50
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Atherosclerosis (AS) is a chronic vascular inflammatory disease resulting from vascular endothelial injury and lipid deposition, closely linked to abnormal lipid metabolism within the body. The critical processes involved in atherosclerosis encompass lipid deposition, oxidation, metabolic disruptions, and inflammatory stimulation within the inner vessel wall. Lipid deposition emerges as a pivotal factor triggering these pathological changes, with vascular smooth muscle cells (VSMCs) playing a significant role in the development of AS. Therefore, the goal was to employ lipids, specifically palmitic acid (PA) and oleic acid (OA) solutions, to stimulate VSMCs and create an in vitro atherosclerosis model. This approach allows for the establishment of a rapid and efficient cell model for simulating atherosclerosis in vitro.

    Methods: Primary vascular smooth muscle cells (VSMCs) were isolated and cultured from the thoracic aorta of healthy rats using the tissue-block method. VSMCs were identified through cell climbing slices and immunofluorescence. The growth of VSMCs was observed using light microscopy. The logarithmic growth phase of VSMCs was induced and stimulated by various concentrations of palmitic acid (PA) and oleic acid (OA) ranging from 0 to 650 μmol/L, with a gradient dilution of 50 μmol/L. VSMC activity was assessed using the 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Intracellular lipid deposition was visualized through Oil Red O staining. The levels of total cholesterol (TC), triglyceride (TG), high-density lipoprotein-cholesterol (HDL-C), and low-density lipoprotein-cholesterol (LDL-C) within VSMCs were quantified using commercially available kits.

    Results: The optimal conditions for VSMC proliferation were determined to be an OA concentration of 500 μmol/L, a PA concentration of 300 μmol/L, and a culture duration of 48 hours. In comparison to the control group, the presence of lipid droplets within VSMCs became significantly evident following treatment with OA or PA. Furthermore, the levels of TC, TG, and LDL-C increased, while the HDL-C content decreased after treatment with OA or PA.

    Conclusions: A research model for atherosclerosis (AS) and the early stages of cardiovascular events, specifically lipid deposition, was successfully established through the use of OA and PA solutions. This model has the potential to open up new research avenues for gaining a deeper understanding of the pathogenesis and progression of AS.

  • Fan-li Liu, Zhan-cheng Zhang, Sheng-li Zhou, Xu-liang Liu, Wei Xu
    Discovery Medicine. 2024, 36(182): 546-558. https://doi.org/10.24976/Discov.Med.202436182.51
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Identifying the key molecular targets in hypopharynx squamous cell carcinoma (HSCC) is crucial for understanding this prevalent and highly fatal type of head and neck tumor. The study aims to enhance comprehension of the HSCC process by accurately identifying these key molecular targets.

    Materials and Methods: In this study, we examined 47 clinical tissue samples from individuals diagnosed with HSCC using RNA-seq high-throughput assay. Quantitative real-time PCR (RT-PCR) was used to compare long non-coding RNA (lncRNA) bladder cancer-associated transcript 1 (BLACAT1) expression in HSCC tissues versus adjacent non-tumor tissues. The influence of highly expressed lncRNA BLACAT1 on prognostic survival was assessed. Subsequently, we cultured human pharynx squamous cell carcinoma FaDu cells. After reducing lncRNA BLACAT1 expression, we assessed FaDu cell proliferation, invasion, and migration using Cell Counting kit-8 (CCK-8) assay, colony formation assay, EUD assay, Transwell assay, and scratch assay. Additionally, liquid chromatography-tandem mass spectrometry/mass spectrometry (LC-MS/MS) and western blotting analysis were used to analyze proteins that bind to lncRNA BLACAT1. During in vivo experiments, mice received subcutaneous injections of FaDu cells transfected with lncRNA BLACAT1 shRNA or Scr plasmid (Control) in the dorsal region to observe and compare tumor growth. Lastly, tumor tissues underwent hematoxylin-eosin (HE) and immunohistochemical (IHC) staining.

    Results: lncRNA BLACAT1 was screened as one of the most significant genes among the group of differentially expressed lncRNAs. RT-PCR exhibited elevated lncRNA BLACAT1 expression in HSCC tissues when compared to non-tumor tissues (p < 0.001). Furthermore, increased lncRNA BLACAT1 expression correlated with advanced clinical stages, heightened lymphatic invasion, and a poor prognosis. Subsequent in vitro experiments solidified our observations, demonstrating lncRNA BLACAT1's promotion of HSCC cell proliferation (p < 0.05), migration (p < 0.01), and invasion (p < 0.01) compared with the control group. Moreover, LC-MS/MS identified signal transducer and activator of transcription 3 (STAT3) and Prohibitin 2 (PHB2) as lncRNA BLACAT1-binding proteins and sh-lncRNA BLACAT1 inhibits STAT3/AKT phosphorylation (p < 0.01) and alters the subcellular distribution of PHB2 and P21 compared with the control group (p < 0.01). Moreover, in vivo experiments showed that lncRNA BLACAT1 inhibition suppresses tumorigenicity in an HSCC xenograft model compared to the control group (p < 0.01).

    Conclusions: lncRNA BLACAT1 is highly expressed in HSCC tumor tissues and plays a crucial role in the development of HSCC in vitro and in vivo. This increased expression may be caused by STAT3/AKT pathway activation, consequently inhibiting P21 expression through PHB2.

  • Xiaofeng Xie, Sijia Tang, Zhengyang Yu
    Discovery Medicine. 2024, 36(182): 559-570. https://doi.org/10.24976/Discov.Med.202436182.52
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: The epidermal growth factor receptor 2 (HER2) is overexpressed in 30% of breast cancers, and this overexpression is strongly correlated with a poor prognosis. Herceptin is a common treatment for HER2-positive breast cancer; however, cancer cells tend to adapt gradually to the drug, rendering it ineffective. The study revealed an association between the methylation status of the Homeobox C8 (HOXC8) gene and tumor development. Therefore, it is of paramount importance to delve into the interaction between HOXC8 and HER2-positive breast cancer, along with its molecular mechanisms. This exploration holds significant implications for a deeper understanding of the pathophysiological processes underlying HER2-positive breast cancer.

    Method: Tumor tissue and pathological data from patients with HER2-positive breast cancer were systematically collected. Additionally, the human HER2-positive breast cancer cell line, SKBR3, was cultured in vitro to assess both the expression level of HOXC8 and the degree of DNA methylation. The study aimed to explore the relationship between the relative expression of HOXC8 and the clinical characteristics of breast cancer patients. The expression level of HOXC8 and the promoter methylation of HOXC8 were verified by methylation treatment of SKBR3 breast cancer cells. The regulation of HOXC8 was meticulously carried out, leading to the division of the cells into distinct groups. The study further analyzed the expression levels and biological capabilities within each group. Finally, the in vitro and in vivo sensitivity of the cells to Herceptin, a common treatment for HER2-positive breast cancer, was measured to assess the efficacy of the drug.

    Result: In HER2-positive breast cancer cases characterized by poor methylation, there was an up-regulation of HOXC8. Its expression was found to be correlated with key clinical factors such as tumor size, lymph node status, clinical tumor, node, metastasis (cTNM) staging, and Herceptin resistance (p < 0.05). Upon methylation of breast cancer cells, there was a significant decrease in HOXC8 expression (p < 0.05). The study revealed that overexpression of HOXC8 resulted in increased proliferation, cloning, and metastasis of HER2-positive breast cancer cells, along with a reduced apoptosis rate (p < 0.05). Conversely, interference with HOXC8 expression reversed this scenario (p < 0.05). A Herceptin-resistant substrain, POOL2, was established using SKBR3 cells. Animal studies demonstrated that overexpressing HOXC8 accelerated tumor development and enhanced POOL2 cells' resistance to Herceptin (p < 0.05). However, following interference with HOXC8, POOL2 cells exhibited increased responsiveness to Herceptin, leading to a gradual reduction in tumor size (p < 0.05).

    Conclusions: In HER2-positive breast cancer, the expression of HOXC8 is elevated in a manner dependent on DNA methylation, and this elevated expression is closely linked to the pathology of the patient. Interfering with HOXC8 expression demonstrates the potential to partially inhibit the development and spread of breast cancer, as well as to alleviate resistance to Herceptin.

  • Guozhen Ni, Jian Xu
    Discovery Medicine. 2024, 36(182): 571-580. https://doi.org/10.24976/Discov.Med.202436182.53
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: The apoptosis of vascular smooth muscle cells (VSMCs) contributes to the progression of atherosclerosis (AS). Long intergenic non-protein coding RNA 1128 (LINC01128) has been implicated in AS, and this study aims to uncover the role and mechanism of LINC01128 in regulating oxidized low-density lipoprotein (oxLDL)-induced VSMCs.

    Methods: The position of LINC01128 in cells and its target genes were predicted using bioinformatics. The localization of LINC01128 in human VSMCs was determined through fluorescence in situ hybridization. VSMCs were transfected, and the interaction between LINC01128 and fucosyltransferase 8 (FUT8) was validated by chromatin immunoprecipitation assay. The apoptotic VSMC model was established using oxLDL. LINC01128 expression in VSMCs was analyzed by quantitative real-time polymerase chain reaction (qRT-PCR), and FUT8 expression was detected by qRT-PCR and western blot. VSMC viability, migration, invasion abilities, and apoptosis were assessed using cell counting kit-8, transwell assay, and flow cytometry, respectively.

    Results: OxLDL (200 μg/mL) upregulated the expression of LINC01128 and FUT8 mRNA, as well as FUT8 protein, in VSMCs. LINC01128 was expressed in the nucleus of VSMCs and bound to FUT8. Knockdown of LINC01128 alleviated the inhibitory effects of oxLDL (200 μg/mL) on viability, migration, and invasion, and mitigated the promotion of apoptosis and FUT8 expression in VSMCs. On the other hand, FUT8 overexpression enhanced the suppressive effects of oxLDL (200 μg/mL) on viability, migration, and invasion activities, and amplified the facilitating effect of oxLDL on apoptosis in VSMCs. Moreover, FUT8 overexpression reversed the impact of LINC01128 silencing on viability, migration, invasion, and apoptosis in oxLDL-stimulated VSMCs.

    Conclusion: The knockdown of LINC01128 downregulates FUT8, inhibiting the progression of VSMCs in AS.

  • Qian Liu, Shengwei Wu, Zhenlin Chen, Jin Cai
    Discovery Medicine. 2024, 36(182): 581-590. https://doi.org/10.24976/Discov.Med.202436182.54
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Phentolamine is an α-adrenergic receptor blocker that can be used to treat neonatal pneumonia, but its underlying mechanism is unclear. The purpose of this study is to probe the function of phentolamine on lipopolysaccharide (LPS)-induced inflammation and cell death in an in vitro model of neonatal pneumonia.

    Methods: Human MRC-5 cells were incubated with varying doses of phentolamine in vitro to evaluate cell viability. Subsequently, LPS was introduced to further investigate the combined effects of phentolamine and LPS on cell viability and apoptosis in MRC-5 cells. The effect of phentolamine/LPS treatment on the Neurotrophic Tyrosine Kinase Receptor A (TrkA)/Protein Kinase B (Akt) signaling pathway and the phosphorylation of pathway proteins in MRC-5 cells was further analyzed via western blot. Additionally, knockout of TrkA and Akt genes in MRC-5 cells was performed to explore the effects of phentolamine/LPS treatment on cell viability, apoptosis levels, and inflammatory factor levels in MRC-5 cells.

    Results: Preincubation of MRC-5 cells with a low concentration of phentolamine (≤6 μg/mL) protected against LPS-induced cell inflammatory injury. Phentolamine promoted both TrkA and Akt phosphorylation and Akt activation induced by LPS in MRC-5 cells. The protective effect of phentolamine against LPS-induced apoptosis and inflammation was significantly reduced in response to TrkA or Akt gene knockdown in MRC-5 cells.

    Conclusions: Phentolamine may protect LPS-induced apoptosis and inflammation by activating the TrkA and Akt signaling pathways.

  • Chengfang Wang, Yijia Zhou, Ruhui Ruan
    Discovery Medicine. 2024, 36(182): 591-597. https://doi.org/10.24976/Discov.Med.202436182.55
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: The continuous advancement in ultrasound technology has given rise to Acoustic Radiation Force Impulse (ARFI) elastography, which boasts non-invasiveness, ease of operation, rapid inspection, and high accuracy. It has been successfully employed in detecting tissue hardness across various diseases. This study aims to investigate the application of acoustic radiation force pulse imaging technology in evaluating the efficacy of calf intermuscular vein thrombosis.

    Methods: This study is retrospective in nature, involving a total of 120 patients diagnosed with calf intermuscular venous thrombosis (MCVT) who were admitted to our hospital. These patients were selected retrospectively as the subjects for our research. They were subsequently divided into two groups: the control group and the observation group. The control group received standard nursing care and simple pressure therapy, while the observation group underwent anticoagulant drug treatment. The shear wave elastic hardness of both groups was measured, with the recording of ultrasonic elasticity scores and the average elastic modulus value (E-mean, in kPa). Furthermore, a comparison was made between the two groups regarding thrombus disappearance time, blood flow patency, and the clinical treatment effect.

    Results: At the 1, 3, and 6-month marks of the treatment period, the ultrasonic elasticity scores in the observation group were consistently higher compared to those in the control group. Additionally, the shear wave elastic hardness in the observation group was consistently lower than that in the control group, and these differences were found to be statistically significant (p < 0.05). The total effective rates for the control and observation groups were 83.33% and 95.00%, respectively. Notably, the clinical total effective rate in the observation group was significantly higher than that in the control group, and this difference was statistically significant (p < 0.05). The thrombus disappearance time in the observation group was significantly shorter than that in the control group, and the blood flow rate was significantly higher than in the control group, with both differences being statistically significant (p < 0.05).

    Conclusion: ARFI plays a crucial role in assessing the efficacy of MCVT by effectively revealing the hardness and location of the patient's thrombus tissue. This technology aids doctors in gaining a more precise understanding of the deep vein thrombosis condition. Notably, ARFI is characterized by high safety levels and exhibits positive effects due to its painless and non-invasive nature.

  • Qinxia Yu, Shuyan Yu
    Discovery Medicine. 2024, 36(182): 598-603. https://doi.org/10.24976/Discov.Med.202436182.56
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Acute myocardial infarction (AMI) is a prevalent cardiovascular disease resulting from myocardial ischemia and necrosis due to coronary artery occlusion. AMI is characterized by a sudden onset and high mortality, underscoring the significance of early diagnosis and treatment for improving patient prognosis. This study endeavors to assess the utility of a combined assessment involving serum brain natriuretic peptide (BNP), cardiac troponin-I (cTnI), and dynamic electrocardiogram (ECG) in the early clinical diagnosis and prognosis prediction of AMI.

    Methods: This paper constitutes a retrospective study. All enrolled patients underwent dynamic ECG examination. The study compared the serum levels of BNP and cTnI, along with pertinent dynamic ECG parameters [turbulence slope (TS) and standard deviation (SDNN) of the 24-hour interval between normal atrial depolarization and ventricular depolarization (R-R)], between the observation group (AMI patients) and the control group (patients with unstable angina (UA)). To evaluate the early diagnostic potential of AMI, we utilized receiver operating characteristic (ROC) curves to analyze serum BNP, cTnI, dynamic ECG, and their combined utility. Furthermore, a follow-up period of 6 months was conducted for AMI patients to record major adverse cardiovascular events (MACE).

    Results: In the observation group, the serum levels of BNP and cTnI were significantly higher than those in the control group (p < 0.001), while dynamic ECG parameters, specifically TS and SDNN, were significantly lower in the observation group compared to the control group (p < 0.001). The results obtained from the ROC curve analysis revealed that the area under the curve (AUC) for BNP, cTnI, dynamic ECG, and their combination in early AMI diagnosis were 0.838, 0.887, 0.874, and 0.974, respectively. The 95% confidence intervals (CI) were 0.781~0.884, 0.836~0.926, 0.822~0.915, and 0.942~0.991, respectively. Sensitivity values were 64.29%, 82.14%, 91.07%, and 88.39%, and specificity values were 91.00%, 88.00%, 70.00%, and 98.00%, respectively. Significantly, the combination of all three markers demonstrated superior efficacy in early AMI diagnosis compared to any single index (p < 0.05). During the 6-month follow-up of 112 AMI patients, 22 experienced MACE. The MACE group exhibited notably higher serum BNP and cTnI levels compared to the non-MACE group. Additionally, dynamic electrocardiogram parameters TS and SDNN demonstrated a significant decrease (p < 0.05) in the MACE group.

    Conclusions: The combined assessment of serum BNP, cTnI, and dynamic electrocardiogram enhances the early clinical diagnostic potential for AMI and holds value in assessing the prognosis of AMI patients.

  • Pei Dong, Sheng Zhang, Da-jun Hu, Rui Hou, Li Lei
    Discovery Medicine. 2024, 36(182): 604-612. https://doi.org/10.24976/Discov.Med.202436182.57
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: The hedgehog signaling pathway exerts vital functions in regulating epithelial-to-mesenchymal transition (EMT) in renal interstitial fibrosis (RIF). It was reported that lncRNA-maternally expressed gene 3 (lncRNA Meg3) can regulate hepatic fibrosis by regulating the expression of smoothened (Smo) in the hedgehog signaling pathway. However, the specific role of lncRNA Meg3 in renal fibrosis resulting from unilateral ureteral obstruction (UUO) by regulating the hedgehog signaling pathway has not been reported. Hence, this research aimed to expound the effects of lncRNA Meg3 on renal fibrosis induced by UUO in rats via the hedgehog pathway.

    Methods: Peripheral blood was collected from patients with chronic kidney disease (CKD, CKD group) and healthy volunteers (Normal group) at the same period. In addition, 6-week-old male Sprague-Dawley (SD) rats were divided to Sham, UUO, UUO+shRNA Negative control (shNC), and UUO+sh-Meg3 groups, and their kidney tissues and serum were gathered. Next, quantitative real-time polymerase chain reaction (qRT-PCR) was employed for detecting the lncRNA Meg3 expression level in the serum of patients and renal tissue of rats; kits for testing levels of blood urea nitrogen (BUN), creatinine (Cr), hydroxyproline (HYP), and 24-hour urine protein (24-up) in rats of each group; hematoxylin and eosin (HE) staining and Masson staining for observing kidney tissue and renal fibrosis level in rats; western blot for measuring levels of collagen type III (Col III), α-Smooth muscle actin (α-SMA), fibronectin, E-cadherin, sonic hedgehog (Shh), patched (Ptch) protein, smoothened (Smo) protein and glioma-associated oncogene homolog 1 (Gli1) protein expression.

    Results: LncRNA Meg3 was highly expressed in CKD patients and UUO rats (p < 0.01). In contrast to the UUO+shNC group, knocking down lncRNA Meg3 improved renal injury, relieved pathological renal lesions, and reduced kidney fibrosis and related protein levels. It inhibited the hedgehog pathway in kidney tissues of UUO rats (p < 0.05 and p < 0.01).

    Conclusions: LncRNA Meg3 can aggravate UUO-induced rat renal fibrosis by activating the hedgehog pathway.

  • Hongmei Liu, Zhipeng Wang, Zhigang Liu
    Discovery Medicine. 2024, 36(182): 613-620. https://doi.org/10.24976/Discov.Med.202436182.58
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Breast cancer (BC), a common tumor in women, has high morbidity and mortality. Formononetin, an active ingredient in red clover and Astragalus membranaceus, has a wide range of pharmacological applications, including as an anticancer agent. Since immunotherapy is a hot topic in the treatment strategy of BC, it was dedicated to appraising the specific mechanism of formononetin in BC immunotherapy in this research.

    Methods: Different formononetin concentrations (0, 20, 40, 60, 80, 100 μM) were used to treat BC cells transfected with pcDNA3.1-Programmed death ligand 1 (PD-L1) or Short-hairpin RNA (sh)-PD-L1. Cells were separated into four subgroups: CTRL, pcDNA3.1-PD-L1, sh-CTRL, and sh-PD-L1. Cell viability and cell cycle were assessed through Methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay and flow cytometry. Programmed death ligand 1 (PD-L1) mRNA concentration was validated via quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). Cell metastasis was evaluated via cloning assay and transwell assay. The p-STING/stimulator of interferon genes (STING), p-p65/p65, and PD-L1 concentrations were determined by western blot.

    Results: Formononetin restrained the proliferation of MCF-7 and MDA-MB-468 cells, and reduced PD-L1 mRNA, p-STING/STING, and p-p65/p65 protein concentrations. Whereas PD-L1 inhibition restrained the viability of BC cells, pcDNA3.1-PD-L1 intervention had the opposite result. STING pathway inhibitor C-176 combined with formononetin treatment further restrained cell proliferation, colony formation, and cell invasion, in contrast to cells treated with formononetin alone.

    Conclusion: Formononetin can restrain the proliferation of BC cells, which may be mediated through the interference of PD-L1 and suppression of the activation of the STING-NF-κB pathway.

  • Yujing Zhang, Yao Chen, Qiang Guo, Ying Zhang, Aichun Liu
    Discovery Medicine. 2024, 36(182): 621-631. https://doi.org/10.24976/Discov.Med.202436182.59
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Diffuse large B-cell lymphoma (DLBCL) poses a significant threat to the quality of life for people worldwide. Regrettably, effective treatment strategies for this disease remain elusive in clinical practice due to the unclear understanding of its molecular mechanisms. Therefore, this study was devised to address these issues and identify novel diagnostic, therapeutic, and prognostic biomarkers for DLBCL.

    Methods: Gene expression and clinical data for DLBCL patients were retrieved from The Cancer Genome Atlas (TCGA) database, and relevant clinical data, tumor mutational burden (TMB), and gene expression levels were extracted. Bioinformatics analysis was conducted to screen for differentially expressed genes (DEGs). The prognostic significance of flotillin-2 (FLOT2) was assessed using Kaplan-Meier survival analysis. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses were employed to evaluate mRNA and protein levels of the genes. Cell proliferation, apoptosis, and invasion were assessed using cell counting kit-8 (CCK-8) assay, flow cytometry analysis, and Transwell assay, respectively.

    Results: Our bioinformatics analysis revealed that FLOT2 was significantly overexpressed in DLBCL tissues compared to normal tissues, a finding corroborated by subsequent immunohistochemistry staining, qRT-PCR, and Western blot analyses. To elucidate its biological functions, shRNAs targeting FLOT2 were transfected into DLBCL cell lines (LY-3 and U2932), resulting in suppressed cell proliferation and invasion, while promoting apoptosis. Furthermore, a positive correlation between TMB and FLOT2 expression in DLBCL was observed. Subsequently, quanTIseq was utilized to calculate the immune score and assess FLOT2 gene expression. In DLBCL, FLOT2 gene expression was found to be associated with T cell CD4+ (non-regulatory) (p < 0.01), monocytes (p < 0.05), and uncharacterized cells (p < 0.05). Regarding immune checkpoint markers, including the cluster of differentiation 274 (CD274), cytotoxic T lymphocyte-associated antigen-4 (CTLA4), hepatitis A virus cellular receptor 2 (HAVCR2), lymphocyte activation gene-3 (LAG3), programmed cell death protein 1 (PDCD1), programmed cell death 1 ligand 2 (PDCD1LG2), Siglec-15 (SIGLEC15), and T cell immunoreceptor with Ig and ITIM domains (TIGIT), our analysis indicated that in DLBCL, FLOT2 exhibited a relationship only with TIGIT (p < 0.05).

    Conclusions: In summary, FLOT2 functions as an oncogene and is linked to DLBCL prognosis and the tumor microenvironment. Targeting FLOT2 deletion emerges as a novel strategy to impede DLBCL aggressiveness by inhibiting cell proliferation and invasion, ultimately inducing apoptotic cell death.

  • Mohamed A.A.A Hegazi, Fabio Pasqualini, Gianluigi Taverna, Robert S. Bresalier, Maurizio Chiriva-Internati, Fabio Grizzi
    Discovery Medicine. 2024, 36(182): 632-645. https://doi.org/10.24976/Discov.Med.202436182.60
    Abstract ( ) Download PDF ( ) HTML ( ) Knowledge map Save

    Background: Ovarian cancer (OC) accounts for about 4% of female cancers globally. While Ki67-immunopositive (Ki67+) cell density is commonly used to assess proliferation in OC, the two-dimensional (2D) distribution pattern of these cells is poorly understood. This study explores the 2D distribution pattern of Ki67+ cells in primary OC tissues and models the proliferation process to improve our understanding of this hallmark of cancer.

    Methods: A total of 100 tissue cores, included in a tissue microarray (TMA) representing 5 clear cell carcinomas, 62 serous carcinomas, 10 mucinous adenocarcinomas, 3 endometrioid adenocarcinomas, 10 lymph node metastases from OC, and 10 samples of adjacent normal ovary tissue, were stained using a standardized immunohistochemical protocol. The computer-aided image analysis system assessed the 2D distribution pattern of Ki67+ proliferating cells, providing the cell number and density, patterns of randomness, and cell-to-cell closeness. Three computer models were created to simulate behavior and responses, aiming to gain insights into the variations in the proliferation process.

    Results: Significant differences in Ki67+ cell density were found between low- and high-grade serous carcinoma/mucinous adenocarcinomas (p = 0.003 and p = 0.01, respectively). The Nearest Neighbor Index of Ki67+ cells differed significantly between high-grade serous carcinomas and endometrioid adenocarcinomas (p = 0.01), indicating distinct 2D Ki67+ distribution patterns. Proxemics analysis revealed significant differences in Ki67+ cell-to-cell closeness between low- and high-grade serous carcinomas (p = 0.002). Computer models showed varied effects on the overall organization of Ki67+ cells and the ability to preserve the original 2D distribution pattern when altering the location and/or density of Ki67+ cells.

    Conclusions: Cell proliferation is a hallmark of OCs. This study provides new evidence that investigating the Ki67+ cell density and 2D distribution pattern can assist in understanding the proliferation status of OCs. Moreover, our computer models suggest that changes in Ki67+ cell density and their location are critical for maintaining the 2D distribution pattern.