Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (128)

Search Parameters:
Keywords = lung resistance-related protein

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 3781 KiB  
Article
Ginsenoside Rg3 Adjunctively Increases the Efficacy of Gefitinib Against NSCLC by Regulating EGFR Copy Number
by Xinyi Lv, Yuehan Song, Tianhua Liu, Dingdan Zhang, Xinpeng Ye, Qingqing Wang, Rongrong Li, Jiayi Chen, Shujing Zhang, Xue Yu and Chunying Hou
Pharmaceuticals 2025, 18(7), 1077; https://doi.org/10.3390/ph18071077 - 21 Jul 2025
Viewed by 331
Abstract
Background: Lung cancer has the highest morbidity and mortality of all tumors, and the development of TKI drugs targeting EGFR activating mutations has brought lung cancer treatment into the targeted era. In view of their low efficacy and susceptibility to drug resistance, [...] Read more.
Background: Lung cancer has the highest morbidity and mortality of all tumors, and the development of TKI drugs targeting EGFR activating mutations has brought lung cancer treatment into the targeted era. In view of their low efficacy and susceptibility to drug resistance, there is an urgent need to find strategies to increase their efficacy and reduce the incidence of drug resistance. Methods: In this study, we examined the distribution and probability of EGFR mutations in non-small cell lung cancer patients in the cBioPortal database and compared the survival prognosis of patients with normal and abnormal EGFR, NSCLC patients treated with and without TKI, and NSCLC patients with different EGFR gene copy numbers. We established a mouse lung cancer model and examined the histomorphological characteristics of lung tissues via hematoxylin and eosin staining. Additionally, changes in the copy number of the EGFR gene and its protein expression levels were detected using RT-qPCR and Western blotting. Furthermore, we quantified the concentration of the EGFR protein using ELISA. Results: We found no significant advantage of EGFR-TKI therapy over first-line chemotherapeutic agents in patients with EGFR-abnormal NSCLC. The reason for this may be related to the abnormal EGFR gene copy number; the higher the copy number increases, the worse the survival prognosis of the patients. In molecular biology experiments, we demonstrated that ginsenoside Rg3 down-regulated the copy number of 18, 19, 20, and 21 exons and protein expression of EGFR in lung adenocarcinoma cells. The results of in vivo pharmacodynamic assays confirmed that sequential administration of ginsenoside Rg3 with TKI drugs could achieve a gainful complementary effect. Conclusions: Ginsenoside Rg3 down-regulates the copy number of EGFR important exons in EGFR-mutant cells of lung adenocarcinoma and reduces EGFR protein expression, thus providing a high gainful complementary effect in combination with EGFR-TKI. Full article
(This article belongs to the Topic Advances in Anti-Cancer Drugs: 2nd Edition)
Show Figures

Figure 1

23 pages, 2440 KiB  
Article
LRP1 Shedding in Ricin-Induced Lung Injury: A Cell-Specific Response to Toxin Exposure
by Anita Sapoznikov, Yentl Evgy, Moshe Aftalion and Reut Falach
Int. J. Mol. Sci. 2025, 26(12), 5448; https://doi.org/10.3390/ijms26125448 - 6 Jun 2025
Viewed by 470
Abstract
Ricin is a highly potent toxin that causes severe lung injury upon inhalation by initiating a complex cascade of cellular responses that ultimately leads to cell death. The low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional receptor involved in various physiological processes, [...] Read more.
Ricin is a highly potent toxin that causes severe lung injury upon inhalation by initiating a complex cascade of cellular responses that ultimately leads to cell death. The low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional receptor involved in various physiological processes, including ricin-mediated toxicity. This study explores the role of LRP1 shedding in the development of ricin-induced lung injury. Analysis of bronchoalveolar lavage fluid (BALF) from ricin-intoxicated mice and swine showed a significant increase in soluble LRP1 (sLRP1) levels, whereas serum LRP1 levels remained largely unchanged, suggesting the lungs are the primary source of sLRP1 release. In vitro assays demonstrated the formation of ricin-sLRP1 complexes, indicating that sLRP1 in BALF retained ricin-binding capability. Flow cytometric analysis of lung cells revealed a reduction in both the percentage and total number of LRP1-expressing cells following ricin exposure. Further investigation of specific lung cell populations showed that alveolar epithelial type II (AT-II) cells, despite experiencing significant injury, exhibited minimal LRP1 shedding. No shedding of LRP1 occurred in neutrophils. In contrast, fibroblasts, which were resistant to ricin-induced cell death, exhibited increased shedding of LRP1 and a corresponding decrease in membrane-bound LRP1 expression. This shedding of the LRP1 ectodomain was mediated by metalloproteinases. Immunohistochemical staining further confirmed decreased LRP1 expression in fibroblasts from ricin-exposed mice. Macrophages also showed substantial LRP1 shedding, despite undergoing significant depletion. These findings highlight the complex cell-specific nature of LRP1 shedding in response to ricin intoxication and suggests the potential role of LRP1 in modulation of cellular susceptibility and resistance to ricin-induced lung injury. Full article
Show Figures

Figure 1

37 pages, 2106 KiB  
Review
Decoding the NRF2–NOTCH Crosstalk in Lung Cancer—An Update
by Angelo Sparaneo, Filippo Torrisi, Floriana D’Angeli, Giovanni Giurdanella, Sara Bravaccini, Lucia Anna Muscarella and Federico Pio Fabrizio
Antioxidants 2025, 14(6), 657; https://doi.org/10.3390/antiox14060657 - 29 May 2025
Viewed by 978
Abstract
The Nuclear factor erythroid 2-related factor 2 (NRF2) Neurogenic locus NOTCH homolog protein (NOTCH) crosstalk has emerged as a critical regulatory axis in the progression of solid cancers, especially lung, affecting tumor growth and resistance to therapy. NRF2 is a master transcription factor [...] Read more.
The Nuclear factor erythroid 2-related factor 2 (NRF2) Neurogenic locus NOTCH homolog protein (NOTCH) crosstalk has emerged as a critical regulatory axis in the progression of solid cancers, especially lung, affecting tumor growth and resistance to therapy. NRF2 is a master transcription factor that orchestrates the cellular antioxidant response, while NOTCH signaling is involved in the cell–cell communication processes by influencing the patterns of gene expression and differentiation. Although frequently altered independently, genetic and epigenetic dysregulation of both NRF2 and NOTCH pathways often converge to deregulate oxidative stress responses and promote tumor cell survival. Recent findings reveal that the NRF2/NOTCH interplay extends beyond canonical signaling, contributing to metabolic reprogramming and reshaping the tumor microenvironment (TME) to promote cancer malignancy. Emerging scientific evidences highlight the key role of biochemical and metabolomic changes within NRF2–NOTCH crosstalk, in contributing to cancer progression and metabolic reprogramming, beyond facilitating the adaptation of cancer cells to the TME. Actually, the effects of the NRF2–NOTCH bidirectional interaction in either supporting or suppressing lung tumor phenotypes are still unclear. This review explores the molecular mechanisms underlying NRF2–NOTCH crosstalk in lung cancer, highlighting the impact of genetic and epigenetic deregulation mechanisms on neoplastic processes, modulating the TME and driving the metabolic reprogramming. Furthermore, we discuss therapeutic opportunities for targeting this regulatory network, which may open new avenues for overcoming drug resistance and improving clinical outcomes in lung cancer. Full article
(This article belongs to the Special Issue Novel Antioxidant Mechanisms for Health and Diseases)
Show Figures

Graphical abstract

17 pages, 2794 KiB  
Review
Long Non-Coding TP73-AS1: A Potential Biomarker and Therapeutic Target in Cancer
by Kejing Li, Dapeng Zhao, Xuena Liu, Qiyou Cao, Longzhu Ruan, Huiwen Lei, Xiaohua Chen, Xiaodong Jin, Qiang Li, Xiaodong Xie and Cuixia Di
Int. J. Mol. Sci. 2025, 26(8), 3886; https://doi.org/10.3390/ijms26083886 - 20 Apr 2025
Viewed by 716
Abstract
Tumor protein 73 antisense RNA 1 (TP73-AS1), a newly discovered long non-coding RNA (lncRNA), the dysregulated expression of which is closely related to the occurrence, drug resistance, and prognosis of various cancers. Exploring the regulatory mechanism of TP73-AS1 provides a new research direction [...] Read more.
Tumor protein 73 antisense RNA 1 (TP73-AS1), a newly discovered long non-coding RNA (lncRNA), the dysregulated expression of which is closely related to the occurrence, drug resistance, and prognosis of various cancers. Exploring the regulatory mechanism of TP73-AS1 provides a new research direction for cancer diagnosis and treatment. On this basis, we briefly review the molecular structural and dual regulatory roles of TP73-AS1 in cancer. In addition, we outline its three molecular mechanisms in cancer: binding to proteins, regulating signaling pathways, and serving as molecular sponges. Subsequently, we introduce the role of TP73-AS1 in common malignant tumors such as gastric cancer (GC), lung cancer, colorectal cancer (CRC), etc. Last, emphasis is given to the potential clinical value of TP73-AS1, especially as single nucleotide polymorphisms of this lncRNA are associated with the risk of GC and CRC. Therefore, this review highlights the significance of TP73-AS1 as a novel biomarker and therapeutic target. Full article
(This article belongs to the Special Issue Roles and Mechanisms of Non-Coding RNAs in Human Health and Disease)
Show Figures

Figure 1

22 pages, 8508 KiB  
Article
Combining AdipoRon with Paclitaxel Unveils Synergistic Potential in Non-Small Cell Lung Cancer Cells via AMPK-ERK1/2 Signaling
by Sanober Kafeel, Giuseppina Palmiero, Alessia Salzillo, Angela Ragone, Silvio Naviglio and Luigi Sapio
Cells 2025, 14(8), 602; https://doi.org/10.3390/cells14080602 - 16 Apr 2025
Cited by 2 | Viewed by 833
Abstract
As part of chemotherapy regimens, Paclitaxel improves the overall survival of many non-small cell lung cancer (NSCLC) patients. However, the development of drug resistance and adverse events limits its clinical usage, reinforcing the need for further advancements in NSCLC therapeutics. We recently recognized [...] Read more.
As part of chemotherapy regimens, Paclitaxel improves the overall survival of many non-small cell lung cancer (NSCLC) patients. However, the development of drug resistance and adverse events limits its clinical usage, reinforcing the need for further advancements in NSCLC therapeutics. We recently recognized the adiponectin receptor agonist AdipoRon as a promising anticancer compound in NSCLC. Consequently, this study aimed to evaluate the therapeutic potential of combining AdipoRon with Paclitaxel (Combo) in NSCLC cells. With respect to individual treatments, Combo triggered a stronger inhibition of both cell growth and clonogenic potential, as well as a greater induction of cell death. The Combo-mediated cytotoxicity was also corroborated by cleavage of poly-ADP ribose polymerase (PARP) and caspase-3 apoptotic markers. Notably, AMP-activated protein kinase (AMPK) emerged as a critical sensor in Combo efficacy, as its inhibition by Compound-C unveiled a significant rescue in cell growth. Although Combo caused a gradual downregulation of extracellular signal-regulated kinase 1/2 (ERK1/2), the hindrance in the upstream cascade by PD98059 partially counteracted the Combo outcomes. In conclusion, our findings designate AdipoRon as an effective candidate in Paclitaxel-based therapy. Nevertheless, future studies aimed at exploring the Combo aptitude in overcoming the Paclitaxel-related restraints need to be investigated in NSCLC. Full article
Show Figures

Figure 1

25 pages, 4721 KiB  
Article
Cytotoxic Effect of Escitalopram/Etoposide Combination on Etoposide-Resistant Lung Cancer
by Serap Özkaya Gül, Beyzanur Şimşek, Fidan Yıldız and Esra Aydemir
Pharmaceuticals 2025, 18(4), 531; https://doi.org/10.3390/ph18040531 - 5 Apr 2025
Cited by 1 | Viewed by 1011
Abstract
Background: Antidepressants are a class of pharmaceuticals utilized for the management of many psychiatric disorders, including depression. A considerable number of antidepressants, particularly selective serotonin reuptake inhibitors (SSRIs), have been documented to demonstrate significant anticancer properties in various cancer cell lines. Objectives: The [...] Read more.
Background: Antidepressants are a class of pharmaceuticals utilized for the management of many psychiatric disorders, including depression. A considerable number of antidepressants, particularly selective serotonin reuptake inhibitors (SSRIs), have been documented to demonstrate significant anticancer properties in various cancer cell lines. Objectives: The aim of this study was to evaluate the selective cytotoxic and apoptotic effects of escitalopram oxalate (ES) alone and in combination with etoposide (ET) on ET-resistant A549 (A549/90E) lung cancer cells. Methods: The cytotoxic effects of the drugs were determined by CCK-8, trypan blue, and neutral red assays. Apoptosis was observed by Annexin V fluorescein isothiocyanate (FITC)/PI and mitochondrial membrane potential (ΔΨm) assays. Moreover, the effects of the drugs, alone and in combination, on apoptosis-related proteins, caspase-3, PTEN, and resistance-related P-gP were determined by ELISA. The relationship between drugs and lung cancer was determined with protein–protein interaction (PPI) network analysis. Results: Our results revealed that ES significantly exerted cytotoxic effects on both wild-type and A549/90E cells compared with BEAS-2B cells. The IC50 values of 48.67 and 51.6 μg/mL obtained for ET and ES, respectively, at the end of 24 h of incubation for A549 cells were applied reciprocally for each cell by including BEAS-2B together with the 2xIC50 and ½ IC50 values. The results of each combination were statistically evaluated with combination indices (CIs) obtained using the Compusyn synergistic effect analysis program. Combination doses with a synergistic effect in A549 and A549/90E cells and an antagonistic effect in BEAS-2B cells have been determined as ½ IC50 for ET and ½ IC50 for ES. ET ½ IC50, ES ½ IC50, and an ET ½ IC50 + ES ½ IC50 combination caused 18.37%, 55.19%, and 57.55% death in A549 cells, whereas they caused 44.9%, 22.4%, and 51.94% death in A549/90E cells, respectively. In A549 cells, the combination of ES ½ IC50 and ET ½ IC50 caused increased levels of caspase-3 (p < 0.01) and P-gP (p < 0.001), while PTEN levels remained unchanged. The combination resulted in an increase in caspase-3 (p < 0.001) and PTEN (p < 0.001) amounts, alongside a decrease in P-gP (p < 0.01) levels in A549/90E cells. The death mechanism induced by the combination was found to be apoptotic by Annexin V-FITC and ΔΨm assays. Conclusions: Based on our findings, ES was observed to induce cytotoxic and apoptotic activities in A549/90E cells in vitro. ES in combination therapy is considered to be effective to overcome ET resistance by reducing the amount of P-gP in A549/90E cells. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

17 pages, 3166 KiB  
Article
Synergistic Anti-Cancer Activity of Melittin and Erlotinib in Non-Small Cell Lung Cancer
by Hairulislam M. Ibrahim, Jihad Alessa, Hala Badr Khalil, Gamal A. Bekhet and Ashraf Khalifa
Int. J. Mol. Sci. 2025, 26(7), 2903; https://doi.org/10.3390/ijms26072903 - 22 Mar 2025
Cited by 1 | Viewed by 1077
Abstract
Lung cancer remains a leading cause of cancer-related mortality worldwide. Despite advancements in current therapies, the development of drug resistance and the need for improved treatment outcomes necessitate the exploration of novel therapeutic approaches. This study aimed to investigate the synergistic anti-cancer effects [...] Read more.
Lung cancer remains a leading cause of cancer-related mortality worldwide. Despite advancements in current therapies, the development of drug resistance and the need for improved treatment outcomes necessitate the exploration of novel therapeutic approaches. This study aimed to investigate the synergistic anti-cancer effects of Melittin, a bee venom peptide, in combination with Erlotinib, an EGFR inhibitor, in non-small cell lung cancer (NSCLC). The study evaluated the combined effects of Melittin and Erlotinib on A549 NSCLC cells. Cell viability, proliferation, migration, and apoptosis were assessed using standard in vitro assays. Mechanistic studies investigated the impact of the combination treatment on key signaling pathways, including those involving JAK2 and JAK3. Molecular docking simulations were performed to predict the binding interactions between Melittin and these kinases. The combination of Melittin and Erlotinib significantly inhibited A549 cell proliferation and migration, with a marked reduction in cell viability and enhanced apoptosis compared to either agent alone. Mechanistically, Melittin demonstrated interactions with JAK2 and JAK3, key proteins involved in apoptotic signaling. Molecular docking simulations further supported these findings, predicting strong binding affinities between Melittin and both kinases. These findings demonstrate a synergistic anti-cancer effect of Melittin and Erlotinib in A549 NSCLC cells. The observed interactions with JAK2 and JAK3 suggest a potential mechanism for Melittin’s activity. These results highlight the potential of Melittin as a promising adjuvant to Erlotinib for the treatment of NSCLC. Full article
(This article belongs to the Special Issue Innovative Biological Molecules for Cancer Therapy)
Show Figures

Figure 1

22 pages, 10922 KiB  
Article
TMEM160 Promotes Tumor Growth in Lung Adenocarcinoma and Cervical Adenocarcinoma Cell Lines
by Gloria Angelina Herrera-Quiterio, Heriberto Abraham Valencia-González, Karen Griselda de la Cruz-López, Diana Lashidua Fernández-Coto, Jeovanis Gil, György Marko-Varga, Josué Morales-Gálvez, Nilda C. Sánchez, Rubén Rodríguez-Bautista, Alejandro Avilés-Salas, Oscar Arrieta, Alejandro García-Carrancá and Sergio Encarnación-Guevara
Int. J. Mol. Sci. 2025, 26(3), 1097; https://doi.org/10.3390/ijms26031097 - 27 Jan 2025
Viewed by 1558
Abstract
The Chromosome-Centric Human Proteome Project (C-HPP) is an international initiative. It aims to create a protein list expressed in human cells by each chromosomal and mitochondrial DNA to enhance our understanding of disease mechanisms, akin to the gene list generated by the Human [...] Read more.
The Chromosome-Centric Human Proteome Project (C-HPP) is an international initiative. It aims to create a protein list expressed in human cells by each chromosomal and mitochondrial DNA to enhance our understanding of disease mechanisms, akin to the gene list generated by the Human Genome Project. Transmembrane protein 160 (TMEM160) is a member of the transmembrane proteins (TMEM) family. TMEM proteins have been implicated in cancer-related processes, including cell proliferation, migration, epithelial-mesenchymal transition, metastasis, and resistance to chemotherapy and radiotherapy. This study aimed to investigate the role of TMEM160 in non-small cell lung cancer and cervical cancer using cell lines, clinical samples, and xenograft studies. Our findings demonstrated that TMEM160 knockdown decreased the proliferation of lung and cervical cancer cell lines. We observed that TMEM160 is localized in the nucleus and cytoplasm and dynamic localization during mitosis of cancer cells and discovered a novel interaction between TMEM160 and nuclear proteins such as NUP50. Furthermore, the TMEM160 interactome was enriched in processes associated with apical junctions, xenobiotic metabolism, glycolysis, epithelial-mesenchymal transition, reactive oxygen species, UV response DNA, the P53 pathway, and the mitotic spindle. This study provides an initial understanding of the function of TMEM160 in lung and cervical cancer progression and clarifies the need to continue investigating the participation of TMEM160 in these cancers. Full article
(This article belongs to the Special Issue Genomics and Proteomics of Cancer)
Show Figures

Figure 1

25 pages, 5054 KiB  
Review
Anthrax: Transmission, Pathogenesis, Prevention and Treatment
by Nitika Sangwan, Aakriti Gangwal, Preksha Jain, Chokey Langtso, Shruti Srivastava, Uma Dhawan, Renu Baweja and Yogendra Singh
Toxins 2025, 17(2), 56; https://doi.org/10.3390/toxins17020056 - 24 Jan 2025
Cited by 1 | Viewed by 5438
Abstract
Bacillus anthracis is a deadly pathogen that under unfavourable conditions forms highly resistant spores which enable them to survive for a long period of time. Spores of B. anthracis are transmitted through the contaminated soil or animal products and enter to the host [...] Read more.
Bacillus anthracis is a deadly pathogen that under unfavourable conditions forms highly resistant spores which enable them to survive for a long period of time. Spores of B. anthracis are transmitted through the contaminated soil or animal products and enter to the host through the skin, lungs or oral route and can cause cutaneous, injection, inhalation and gastrointestinal anthrax, respectively. The disease is caused by the toxin which is produced by them once they germinate within the host cell. Anthrax toxin is the major virulence factor which has the ability to kill the host cell. The role of protein kinases and phosphatases of B. anthracis in toxin production and other virulence related properties have also been reported. There are two vaccines, BioThrax and CYFENDUSTM, which are approved by the FDA-USA to prevent anthrax disease. Recently, anthrax toxin has also been shown to be a potential candidate for cancer therapeutics. Through present review, we aim to provide insights into sporulation, transmission and pathogenesis of B. anthracis as well as the current state of its prevention, treatment, vaccines and possible therapeutic uses in cancer. Full article
Show Figures

Figure 1

15 pages, 923 KiB  
Review
Resistance to Radiotherapy in Cancer
by Almaz A. Akhunzianov, Elvira V. Rozhina, Yuliya V. Filina, Albert A. Rizvanov and Regina R. Miftakhova
Diseases 2025, 13(1), 22; https://doi.org/10.3390/diseases13010022 - 17 Jan 2025
Cited by 3 | Viewed by 2350
Abstract
Radiation therapy or radiotherapy is a medical treatment that uses high doses of ionizing radiation to eliminate cancer cells and shrink tumors. It works by targeting the DNA within the tumor cells restricting their proliferation. Radiotherapy has been used for treating cancer for [...] Read more.
Radiation therapy or radiotherapy is a medical treatment that uses high doses of ionizing radiation to eliminate cancer cells and shrink tumors. It works by targeting the DNA within the tumor cells restricting their proliferation. Radiotherapy has been used for treating cancer for more than 100 years. Along with surgery and chemotherapy, it is one of the three main and most common approaches used in cancer therapy. Nowadays, radiotherapy has become a standard treatment option for a wide range of cancers around the world, including lung, breast, cervical, and colorectal cancers. Around 50% of all patients will require radiotherapy, 60% of whom are treated with curative intent. Moreover, it is commonly used for palliative treatment. Radiotherapy provides 5-year local control and overall survival benefit in 10.4% and 2.4% of all cancer patients, respectively. The highest local control benefit is reported for cervical (33%), head and neck (32%), and prostate (26%) cancers. But no benefit is observed in pancreas, ovary, liver, kidney, and colon cancers. Such relatively low efficiency is related to the development of radiation resistance, which results in cancer recurrence, metastatic dissemination, and poor prognosis. The identification of radioresistance biomarkers allows for improving the treatment outcome. These biomarkers mainly include proteins involved in metabolism and cell signaling pathways. Full article
Show Figures

Figure 1

15 pages, 4307 KiB  
Article
Expression of ABCB1, ABCC1, and LRP in Mesenchymal Stem Cells from Human Amniotic Fluid and Bone Marrow in Culture—Effects of In Vitro Osteogenic and Adipogenic Differentiation
by Carolina Martinez Romão, Felipe de Lara Janz, Jorge Luis Maria Ruiz, Marco Antônio Borges Lopes, Alexandre Fogaça Cristante, Tarcísio Eloy Pessoa de Barros Filho, Débora Levy and Sérgio Paulo Bydlowski
Int. J. Mol. Sci. 2025, 26(2), 510; https://doi.org/10.3390/ijms26020510 - 9 Jan 2025
Viewed by 1198
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells with the potential to differentiate into various lineages. They have also the potential to protect themselves against harmful stimuli to maintain their functional integrity. Drug resistance-related transporters such as ABCB1 (P-glycoprotein; P-gp), ABCC1 (MRP1; multidrug resistance-related [...] Read more.
Mesenchymal stem cells (MSCs) are multipotent cells with the potential to differentiate into various lineages. They have also the potential to protect themselves against harmful stimuli to maintain their functional integrity. Drug resistance-related transporters such as ABCB1 (P-glycoprotein; P-gp), ABCC1 (MRP1; multidrug resistance-related Protein 1), and LRP (lung resistance protein) may protect MSCs against toxic substances such as chemotherapeutic agents. This study evaluated ABCB1, ABCC1, and LRP before and after the differentiation of MSCs derived from human amniotic fluid (AF) and bone marrow (BM). P-gp expression in both AFMSCs and BMMSCs was analyzed by immunocytochemistry, and pump function was analyzed by cell viability assay with doxorubicin (DOX) and Rhodamine 123 (Rh 123) dye exclusion. ABCB1, ABCC1, and LRP gene expression was determined by RT-PCR both before and after osteogenic and adipogenic differentiation. The MES-SA/DX5 cell line was used as a model of resistance to DOX and the overexpression of P-gp. Both AFMSCs and BMMSCs displayed a high P-gp expression, although lower than MES-SA/DX5 control cells. It was shown that both, undifferentiated AFMSCs and BMMSCs, have high cell viability in response to DOX, similar to the MES-SA/DX5 lineage. ABCB1 was less expressed in BM than in AFMSCs in undifferentiated samples, while no differences were observed in the expression of ABCC1 and LRP. AFMSCs showed an increase in ABCB1 after osteogenic differentiation, whereas BMMSCs exhibited lower ABCB1 and ABCC1 expression after osteogenic and adipogenic differentiation. The findings suggest that ABCB1, ABCC1, and LRP gene expression in AFMSCs and BMMSCs is influenced by differentiation processes and further support the concept that these transporters modulate MSC differentiation in a cell source-dependent way. Full article
Show Figures

Figure 1

14 pages, 3105 KiB  
Article
Investigating the Pulmonary Host Response of Acinetobacter baumannii Infection-Associated Pneumonia by Metagenomic Next-Generation Sequencing
by Mu-Jung Chou, Chih-Hung Cheng, Hui-Ching Wang, Ming-Ju Tsai, Chau-Chyun Sheu and Wei-An Chang
Biomedicines 2025, 13(1), 142; https://doi.org/10.3390/biomedicines13010142 - 9 Jan 2025
Cited by 1 | Viewed by 1266
Abstract
Background: For investigating the host response in Acinetobacter baumannii associated pneumonia, we analyzed the host genetic sequences obtained from metagenomic next-generation sequencing (mNGS). Methods: The samples for mNGS were bronchoalveolar lavage fluid (BALF) collected from the lungs of patients infected with A. baumannii [...] Read more.
Background: For investigating the host response in Acinetobacter baumannii associated pneumonia, we analyzed the host genetic sequences obtained from metagenomic next-generation sequencing (mNGS). Methods: The samples for mNGS were bronchoalveolar lavage fluid (BALF) collected from the lungs of patients infected with A. baumannii and from patients without bacterial infections. BALF samples from patients with pneumonia were collected from the lungs of patients infected with A. baumannii with New Delhi metallo-β-lactamase (NDM, before treatment), A. baumannii with NDM (post-treatment), A. baumannii without resistant genes, and those without bacterial infection. Partek was used for investigating enriched functions and pathways related to the pulmonary host response to pneumonia caused by A. baumannii with NDM infection and A. baumannii without antimicrobial-resistant genes. The STRING was employed for identifying protein interaction pathways related to the pulmonary host response to pneumonia caused by A. baumannii without antimicrobial-resistant genes. Results: In pulmonary host response to pneumonia caused by A. baumannii with NDM, five immune system-related pathways and five pathways related to signal transduction were identified. No significant differences were observed in the immune system and signal transduction pathways in the pulmonary host response to pneumonia caused by A. baumannii without antimicrobial-resistant genes. However, significant differences were noted in the phagosome, ferroptosis, and regulation of the actin cytoskeleton in cellular processes. Conclusions: mNGS provides information not only on pathogen gene expression but also on host gene expression. In this study, we found that pneumonia with A. baumannii carrying the NDM resistance gene triggers stronger immune responses in the lung, while pneumonia with A. baumannii lacking antimicrobial resistance genes is more linked to iron-related pathways. Full article
(This article belongs to the Section Microbiology in Human Health and Disease)
Show Figures

Figure 1

17 pages, 2539 KiB  
Article
C/EBPβ Regulates HIF-1α-Driven Invasion of Non-Small-Cell Lung Cancer Cells
by Seung Hee Seo, Ji Hae Lee, Eun Kyung Choi, Seung Bae Rho and Kyungsil Yoon
Biomolecules 2025, 15(1), 36; https://doi.org/10.3390/biom15010036 - 30 Dec 2024
Viewed by 1154
Abstract
Metastatic cancer accounts for most cancer-related deaths, and identifying specific molecular targets that contribute to metastatic progression is crucial for the development of effective treatments. Hypoxia, a feature of solid tumors, plays a role in cancer progression by inducing resistance to therapy and [...] Read more.
Metastatic cancer accounts for most cancer-related deaths, and identifying specific molecular targets that contribute to metastatic progression is crucial for the development of effective treatments. Hypoxia, a feature of solid tumors, plays a role in cancer progression by inducing resistance to therapy and accelerating metastasis. Here, we report that CCAAT/enhancer-binding protein beta (C/EBPβ) transcriptionally regulates hypoxia-inducible factor 1 subunit alpha (HIF1A) and thus promotes migration and invasion of non-small-cell lung cancer (NSCLC) cells under hypoxic conditions. Our results show that knockdown or forced expression of C/EBPβ was correlated with HIF-1α expression and that C/EBPβ directly bound to the promoter region of HIF1A. Silencing HIF1A inhibited the enhanced migration and invasion induced by C/EBPβ overexpression in NSCLC cells under hypoxia. Expression of the HIF-1α target gene, SLC2A1, was also altered in a C/EBPβ-dependent manner, and knockdown of SLC2A1 reduced migration and invasion enhanced by C/EBPβ overexpression. These results indicate that C/EBPβ is a critical regulator for the invasion of NSCLC cells in the hypoxic tumor microenvironment. Collectively, the C/EBPβ-HIF-1α-GLUT1 axis represents a potential therapeutic target for preventing metastatic progression of NSCLC and improving patient outcomes. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

15 pages, 4400 KiB  
Article
GPR68 Mediates Lung Endothelial Dysfunction Caused by Bacterial Inflammation and Tissue Acidification
by Pratap Karki, Yunbo Ke, Chenou Zhang, Kamoltip Promnares, Yue Li, Charles H. Williams, Charles C. Hong, Konstantin G. Birukov and Anna A. Birukova
Cells 2024, 13(24), 2125; https://doi.org/10.3390/cells13242125 - 22 Dec 2024
Viewed by 1512
Abstract
Tissue acidification resulting from dysregulated cellular bioenergetics accompanies various inflammatory states. GPR68, along with other members of proton-sensing G protein-coupled receptors, responds to extracellular acidification and has been implicated in chronic inflammation-related diseases such as ischemia, cancer, and colitis. The present study examined [...] Read more.
Tissue acidification resulting from dysregulated cellular bioenergetics accompanies various inflammatory states. GPR68, along with other members of proton-sensing G protein-coupled receptors, responds to extracellular acidification and has been implicated in chronic inflammation-related diseases such as ischemia, cancer, and colitis. The present study examined the role of extracellular acidification on human pulmonary endothelial cell (EC) permeability and inflammatory status per se and investigated potential synergistic effects of acidosis on endothelial dysfunction caused by bacterial lipopolysaccharide (LPS, Klebsiella pneumoniae). Results showed that medium acidification to pH 6.5 caused a delayed increase in EC permeability illustrated by a decrease in transendothelial electrical resistance and loss of continuous VE-cadherin immunostaining at cell junctions. Likewise, acidic pH induced endothelial inflammation reflected by increased mRNA and protein expression of EC adhesion molecules VCAM-1 and ICAM-1, upregulated mRNA transcripts of tumor necrosis factor-α, IL-6, IL-8, IL-1β, and CXCL5, and increased secretion of ICAM-1, IL-6, and IL-8 in culture medium monitored by ELISA. Among the GPCRs tested, acidic pH selectively increased mRNA and protein expression of GPR68, and only the GPR68-specific small molecule inhibitor OGM-8345 rescued acidosis-induced endothelial permeability and inflammation. Furthermore, acidic pH exacerbated LPS-induced endothelial permeability and inflammatory response in cultured lung macrovascular as well as microvascular endothelial cells. These effects were suppressed by OGM-8345 in both EC types. Altogether, these results suggest that GPR68 is a critical mediator of acidic pH-induced dysfunction of human pulmonary vascular endothelial cells and mediates the augmenting effect of tissue acidification on LPS-induced endothelial cell injury. Full article
(This article belongs to the Section Cell Microenvironment)
Show Figures

Figure 1

11 pages, 1221 KiB  
Article
Association Between Visceral Fat and Lung Function Impairment in Overweight and Grade I Obese Women: A Cross-Sectional Study
by Anamei Silva-Reis, Boris Brill, Maysa Alves Rodrigues Brandao-Rangel, Renilson Moraes-Ferreira, Dobroslav Melamed, Helida Cristina Aquino-Santos, Claudio Ricardo Frison, Regiane Albertini, Rodrigo Álvaro Brandao Lopes-Martins, Luís Vicente Franco de Oliveira, Gustavo Paixao-Santos, Carlos Rocha Oliveira, Asghar Abbasi and Rodolfo P. Vieira
Adv. Respir. Med. 2024, 92(6), 548-558; https://doi.org/10.3390/arm92060048 - 18 Dec 2024
Cited by 1 | Viewed by 2215
Abstract
Beyond the common comorbidities related to obesity, such as type 2 diabetes and cardiovascular diseases, impaired lung function is already known, but whether the fat distribution (sub-cutaneous, visceral) affects the lung function and pulmonary immune response are poorly known. Few evidence has shown [...] Read more.
Beyond the common comorbidities related to obesity, such as type 2 diabetes and cardiovascular diseases, impaired lung function is already known, but whether the fat distribution (sub-cutaneous, visceral) affects the lung function and pulmonary immune response are poorly known. Few evidence has shown that visceral fat is associated with insulin resistance, low-grade inflammation, and reduced lung function. In the present study, the body composition and fat distribution were evaluated by multi-frequency octopolar bioimpedance. This study demonstrated a possible association of increased visceral fat with impaired lung function in obesity grade I (n = 28; 45.46 ± 10.38 years old) women that was not observed in normal weight (n = 20; 43.20 ± 10.78 years old) and in overweight women (n = 30; 47.27 ± 10.25 years old). We also identified a negative correlation in FVC% (R2 = 0.9129; p < 0.0236), FEV1% (R2 = 0.1079; p < 0.0134), PEF% (R2 = 0.1673; p < 0.0018), and VC IN% (R2 = 0.1330; p < 0.0057) in the obesity grade I group, clearly demonstrating that higher levels of visceral fat correlate with reduced lung function, but not with sub-cutaneous fat. In addition, for the first time, a negative correlation among anti-fibrotic protein klotho (R2 = 0.09298; p < 0.0897) and anti-inflammatory IL-10 (R2 = 0.1653; p < 0.0487) in plasma was observed, in contrast to increased visceral fat. On the contrary, in breath condensate, a positive correlation for adiponectin (R2 = 0.5665; p < 0.0120), IL1-Ra (R2 = 0.2121; p < 0.0544), and IL1-Beta (R2 = 0.3270; p < 0.0084) was found. Thus, increased visceral fat directly influences the impairment of lung function and the systemic and pulmonary immune response of women with obesity grade I. Full article
Show Figures

Figure 1

Back to TopTop