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
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (123)

Search Parameters:
Keywords = thapsigargin

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 2091 KiB  
Article
Opposing Calcium-Dependent Effects of GsMTx4 in Acute Lymphoblastic Leukemia: In Vitro Proliferation vs. In Vivo Survival Advantage
by Souleymane Abdoul-Azize, Rachid Zoubairi and Olivier Boyer
Int. J. Mol. Sci. 2025, 26(10), 4822; https://doi.org/10.3390/ijms26104822 - 18 May 2025
Viewed by 445
Abstract
Mechanogated (MG) ion channels play a crucial role in mechano-transduction and immune cell regulation, yet their impact on blood cancers, particularly acute lymphoblastic leukemia (ALL), remains poorly understood. This study investigates the pharmacological effects of GsMTx4, an MG channel inhibitor, in human ALL [...] Read more.
Mechanogated (MG) ion channels play a crucial role in mechano-transduction and immune cell regulation, yet their impact on blood cancers, particularly acute lymphoblastic leukemia (ALL), remains poorly understood. This study investigates the pharmacological effects of GsMTx4, an MG channel inhibitor, in human ALL cells both in vitro and in vivo. Unexpectedly, we found that GsMTx4 remarkably increased basal calcium (Ca2+) levels in ALL cells through constitutive Ca2+ entry and enhanced store-operated Ca2⁺ influx upon thapsigargin stimulation. This increase in basal Ca2+ signaling promoted ALL cell viability and proliferation in vitro. Notably, chelating intracellular Ca2+ with BAPTA-AM reduces GsMTx4-mediated leukemia cell viability and proliferation. However, in vivo, GsMTx4 decreases cytosolic Ca2+ levels in Nalm-6 GFP⁺ cells isolated from mouse blood, effectively countering leukemia progression and significantly extending survival in NSG mice transplanted with leukemia cells (median survival: GsMTx4 vs. control, 37.5 days vs. 29 days, p = 0.0414). Our results highlight the different properties of GsMTx4 activity in in vitro and in vivo models. They also emphasize that Ca2+ signaling is a key vulnerability in leukemia, where its precise modulation dictates disease progression. Thus, targeting Ca2+ channels could offer a novel therapeutic strategy for leukemia by exploiting Ca2+ homeostasis. Full article
Show Figures

Figure 1

15 pages, 6628 KiB  
Article
Ginsenoside Rh2 Mitigates Endoplasmic Reticulum Stress-Induced Apoptosis and Inflammation and Through Inhibition of Hepatocyte–Macrophage Inflammatory Crosstalk
by Shinjung Park, Inae Jeong and Ok-Kyung Kim
Nutrients 2025, 17(10), 1682; https://doi.org/10.3390/nu17101682 - 15 May 2025
Viewed by 752
Abstract
Background/Objectives: Endoplasmic reticulum stress (ERS) contributes to hepatocyte inflammation, triggered by prolonged exposure to lipotoxicity, and promotes non-alcoholic fatty liver disease (NAFLD) progression by recruiting and activating hepatic macrophages, which accelerate fibrosis and exacerbate disease progression. Here, we aimed to evaluate the therapeutic [...] Read more.
Background/Objectives: Endoplasmic reticulum stress (ERS) contributes to hepatocyte inflammation, triggered by prolonged exposure to lipotoxicity, and promotes non-alcoholic fatty liver disease (NAFLD) progression by recruiting and activating hepatic macrophages, which accelerate fibrosis and exacerbate disease progression. Here, we aimed to evaluate the therapeutic potential of ginsenoside Rh2 (Rh2) in a cell model of NAFLD induced by the ERS inducer thapsigargin (THA). Methods: HepG2 cells were treated with THA to induce ERS and mimic NAFLD conditions. The effects of Rh2 on ERS, lipid accumulation, and apoptosis were assessed in HepG2 cells. Additionally, THP-1 cells were used to investigate macrophage activation upon exposure to conditioned medium (CM) from THA- and Rh2-treated HepG2 cells. Gene and protein expression of inflammatory and lipid synthesis markers were analyzed, as well as M1/M2 macrophage polarization markers. Results: Rh2 inhibited THA-induced apoptosis, ERS, and lipid accumulation in HepG2 cells. It also reduced the expression of lipid synthesis genes (SREBF1, FAS) and inflammatory markers (IL-6, IL-1β, TNF-α, MCP-1). CM from Rh2-treated HepG2 cells suppressed macrophage activation in THP-1 cells, decreased M1 polarization markers (CD80, CD86), and increased M2 markers (CD163, Arg1, MRC-1). Conclusions: These results suggest that Rh2 effectively suppresses inflammation and lipid storage in ERS-induced HepG2 cells while modulating the crosstalk between hepatocytes and macrophages. These findings underscore the potential of Rh2 as a promising therapeutic agent for the prevention and early intervention of NAFLD progression. Full article
Show Figures

Figure 1

17 pages, 2739 KiB  
Article
TP53 Mutation-Specific Dysregulation of Store-Operated Calcium Entry and Apoptotic Sensitivity in Triple-Negative Breast Cancer
by Kaneez E. Rabab, Paul J. Buchanan, Grace Colley, Anita White, Aisling Murphy, Chloe McCormack and Alex J. Eustace
Cancers 2025, 17(10), 1614; https://doi.org/10.3390/cancers17101614 - 10 May 2025
Cited by 1 | Viewed by 1041
Abstract
Background: Triple-negative breast cancer (TNBC) is an aggressive subtype lacking estrogen, progesterone, and HER2 receptors, and is associated with poor prognosis and limited targeted therapeutic options. TP53 mutations occur in the majority of TNBC cases, disrupting p53’s role in DNA repair and apoptosis. [...] Read more.
Background: Triple-negative breast cancer (TNBC) is an aggressive subtype lacking estrogen, progesterone, and HER2 receptors, and is associated with poor prognosis and limited targeted therapeutic options. TP53 mutations occur in the majority of TNBC cases, disrupting p53’s role in DNA repair and apoptosis. Beyond gene regulation, p53 also influences calcium signalling through store-operated calcium entry (SOCE), a critical pathway for cell survival and death. However, the impact of different TP53 mutation types on calcium signalling remains unclear. Methods: Calcium channel gene expression was analysed using publicly available TNBC datasets. Calcium channel expression and SOCE activity were assessed in TNBC cell lines with different TP53 mutations using quantitative PCR and calcium imaging (Fura-2AM). Cell proliferation was measured using acid phosphatase assays, while apoptosis was evaluated through caspase 3/7 activation using the Incucyte live-cell fluorescent imager. The p53 reactivator COTI-2 was tested for its ability to restore TP53 function and modulate calcium signalling. Results: Analysis revealed significant downregulation of CACNA1D in TP53-mutant TNBCs. TNBC cell lines harbouring frameshift and stop TP53 mutations exhibited reduced SOCE, lower CACNA1D expression, and resistance to thapsigargin-induced apoptosis compared to wild-type cells. In contrast, cells with the TP53 R273H missense mutation demonstrated similar calcium signalling and proliferation to TP53 wild-type cels. COTI-2 treatment restored CACNA1D expression and SOCE in frameshift and stop mutant cells, enhancing apoptotic sensitivity. Combined treatment with COTI-2 and thapsigargin resulted in a synergistic increase in apoptosis. Conclusions: This study identifies a novel link between TP53 mutation type and calcium signalling in TNBC. Reactivating mutant p53 with COTI-2 restores calcium-mediated apoptosis, supporting combination strategies targeting both TP53 dysfunction and calcium signalling. Full article
(This article belongs to the Special Issue Calcium Signaling in Cancer Cell Progression)
Show Figures

Figure 1

19 pages, 5913 KiB  
Article
Putative Endoplasmic Reticulum Stress Inducers Enhance Triacylglycerol Accumulation in Chlorella sorokiniana
by Yoomi Roh, Sujeong Je, Naeun Sheen, Chang Hun Shin and Yasuyo Yamaoka
Bioengineering 2025, 12(5), 452; https://doi.org/10.3390/bioengineering12050452 - 25 Apr 2025
Cited by 1 | Viewed by 548
Abstract
Chlorella, recognized for its high lipid and protein content, is increasingly studied for its potential in the food and bio industries. To enhance its production and understand the underlying mechanisms of lipid accumulation, this study investigated the role of endoplasmic reticulum (ER) [...] Read more.
Chlorella, recognized for its high lipid and protein content, is increasingly studied for its potential in the food and bio industries. To enhance its production and understand the underlying mechanisms of lipid accumulation, this study investigated the role of endoplasmic reticulum (ER) stress in modulating lipid metabolism in Chlorella sorokiniana UTEX 2714, using six putative ER stress inducers: 2-deoxy-D-glucose (2-DG), dithiothreitol (DTT), tunicamycin (TM), thapsigargin (TG), brefeldin A (BFA), and monensin (Mon). The results showed that 2-DG, DTT, TM, BFA, and Mon significantly inhibited cell growth in C. sorokiniana. Treatment with 2-DG, DTT, TM, BFA, or Mon resulted in substantial increases in the triacylglycerol (TAG) to total fatty acid (tFA) ratio, with fold changes of 14.8, 7.9, 6.2, 10.1, and 8.9, respectively. Among the tFAs, cells treated with these compounds exhibited higher levels of saturated fatty acids and lower levels of polyunsaturated fatty acids (PUFAs). In contrast, the fatty acid composition of TAGs showed the opposite trend, with relative enrichment in PUFAs. This study enhances our understanding of Chlorella lipid metabolism, providing valuable insights for optimizing lipid production, particularly TAGs enriched with PUFA content, for applications in functional foods, nutraceuticals, and sustainable bioresources. Full article
(This article belongs to the Special Issue Microalgae Biotechnology and Microbiology: Prospects and Applications)
Show Figures

Graphical abstract

20 pages, 10727 KiB  
Article
α-Latrotoxin Actions in the Absence of Extracellular Ca2+ Require Release of Stored Ca2+
by Jennifer K. Blackburn, Quazi Sufia Islam, Ouafa Benlaouer, Svetlana A. Tonevitskaya, Evelina Petitto and Yuri A. Ushkaryov
Toxins 2025, 17(2), 73; https://doi.org/10.3390/toxins17020073 - 6 Feb 2025
Viewed by 1104
Abstract
α-Latrotoxin (αLTX) causes exhaustive release of neurotransmitters from nerve terminals in the absence of extracellular Ca2+ (Ca2+e). To investigate the mechanisms underlying this effect, we loaded mouse neuromuscular junctions with BAPTA-AM. This membrane-permeable Ca2+-chelator demonstrates that Ca [...] Read more.
α-Latrotoxin (αLTX) causes exhaustive release of neurotransmitters from nerve terminals in the absence of extracellular Ca2+ (Ca2+e). To investigate the mechanisms underlying this effect, we loaded mouse neuromuscular junctions with BAPTA-AM. This membrane-permeable Ca2+-chelator demonstrates that Ca2+e-independent effects of αLTX require an increase in cytosolic Ca2+ (Ca2+cyt). We also show that thapsigargin, which depletes Ca2+ stores, induces neurotransmitter release, but inhibits the effect of αLTX. We then studied αLTX’s effects on Ca2+cyt using neuroblastoma cells expressing signaling-capable or signaling-incapable variants of latrophilin-1, a G protein-coupled receptor of αLTX. Our results demonstrate that αLTX acts as a cation ionophore and a latrophilin agonist. In model cells at 0 Ca2+e, αLTX forms membrane pores and allows the influx of Na+; this reverses the Na+-Ca2+ exchanger, leading to the release of stored Ca2+ and inhibition of its extrusion. Concurrently, αLTX stimulates latrophilin signaling, which depletes a Ca2+ store and induces transient opening of Ca2+ channels in the plasmalemma that are sensitive to inhibitors of store-operated Ca2+ entry. These results indicate that Ca2+ release from intracellular stores and that Ca2+ influx through latrophilin-activated store-operated Ca2+ channels contributes to αLTX actions and may be involved in physiological control of neurotransmitter release at nerve terminals. Full article
(This article belongs to the Special Issue Animal Venoms: Unraveling the Molecular Complexity (2nd Edition))
Show Figures

Figure 1

18 pages, 5275 KiB  
Article
Effect of Media Composition and Oxygen Tension on Cellular Stress Response and Nrf2 Activation in HepG2ARE Cells
by Rutt Taba, Marie Põlluaed, Karin Tein, Marju Puurand, Tuuli Käämbre and Anton Terasmaa
Antioxidants 2025, 14(2), 137; https://doi.org/10.3390/antiox14020137 - 24 Jan 2025
Viewed by 996
Abstract
Cell models play a central role in preclinical research aimed at the mechanism of disease and drug discovery. The outside environment of the cells, including levels of nutrients and oxygen tension, regulates cellular stress response pathways. Routinely used in vitro disease models often [...] Read more.
Cell models play a central role in preclinical research aimed at the mechanism of disease and drug discovery. The outside environment of the cells, including levels of nutrients and oxygen tension, regulates cellular stress response pathways. Routinely used in vitro disease models often overlook cell growth conditions. This study aimed to evaluate the effect of substituting classic cell media (DMEM) with media matching the nutrient composition of human plasma (Plasmax) on cell viability, the activation of nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione (GSH), and malondialdehyde (MDA) levels by different pharmacological inducers of cell stress. The cells were grown at ambient (~19%) and reduced (5%) oxygen levels. The activation of Nrf2 by ferroptosis activators (erastin and RSL3) was dependent on cell media and oxygen tension. The induction of Nrf2 by an inducer of endoplasmic reticulum stress, thapsigargin, was observable only in cells grown in DMEM and at low oxygen tension. GSH and MDA levels were elevated in Plasmax media. Results indicate that stress tolerance and the activation of Nrf2 in the HepG2ARE cell line depend on the growth conditions, including cell media and oxygen. Cell culture conditions should be critically considered when designing in vitro models of diseases involving oxidative stress. Full article
(This article belongs to the Section Aberrant Oxidation of Biomolecules)
Show Figures

Figure 1

15 pages, 2990 KiB  
Article
A Novel Approach for In Vitro Testing and Hazard Evaluation of Nanoformulated RyR2-Targeting siRNA Drugs Using Human PBMCs
by Valeria Bettinsoli, Gloria Melzi, Angelica Crea, Lorenzo Degli Esposti, Michele Iafisco, Daniele Catalucci, Paolo Ciana and Emanuela Corsini
Life 2025, 15(1), 95; https://doi.org/10.3390/life15010095 - 14 Jan 2025
Viewed by 1642
Abstract
Nucleic acid (NA)-based drugs are promising therapeutics agents. Beyond efficacy, addressing safety concerns—particularly those specific to this class of drugs—is crucial. Here, we propose an in vitro approach to screen for potential adverse off-target effects of NA-based drugs. Human peripheral blood mononuclear cells [...] Read more.
Nucleic acid (NA)-based drugs are promising therapeutics agents. Beyond efficacy, addressing safety concerns—particularly those specific to this class of drugs—is crucial. Here, we propose an in vitro approach to screen for potential adverse off-target effects of NA-based drugs. Human peripheral blood mononuclear cells (PBMCs), purified from buffy coats of healthy donors, were used to investigate the ability of NA-drugs to trigger toxicity pathways and inappropriate immune stimulation. PBMCs were selected for their ability to represent potential human responses, given their likelihood of interacting with administered drugs. As proof of concept, a small interfering RNA (siRNA) targeting Ryanodine Receptor mRNA (RyR2) identified by the Italian National Center for Gene Therapy and Drugs based on RNA Technology as a potential therapeutic target for dominant catecholaminergic polymorphic ventricular tachycardia, was selected. This compound and its scramble were formulated within a calcium phosphate nanoparticle-based delivery system. Positive controls for four toxicity pathways were identified through literature review, each associated with a specific type of cellular stress: oxidative stress (tert-butyl hydroperoxide), mitochondrial stress (rotenone), endoplasmic reticulum stress (thapsigargin), and autophagy (rapamycin). These controls were used to define specific mRNA signatures triggered in PBMCs, which were subsequently used as indicators of off-target effects. To assess immune activation, the release of pro-inflammatory cytokines (interleukin-6, interleukin-8, tumor necrosis factor-α, and interferon-γ) was measured 24 h after exposure. The proposed approach provides a rapid and effective screening method for identifying potential unintended effects in a relevant human model, which also allows to address gender effects and variability in responses. Full article
(This article belongs to the Section Pharmaceutical Science)
Show Figures

Figure 1

15 pages, 1664 KiB  
Article
Anti-Inflammatory 8-Shogaol Mediates Apoptosis by Inducing Oxidative Stress and Sensitizes Radioresistance in Gastric Cancer
by Tae Woo Kim and Hee Gu Lee
Int. J. Mol. Sci. 2025, 26(1), 173; https://doi.org/10.3390/ijms26010173 - 28 Dec 2024
Cited by 2 | Viewed by 1236
Abstract
Radiotherapy is a powerful tumor therapeutic strategy for gastric cancer patients. However, radioresistance is a major obstacle to kill cancer cells. Ginger (Zingiber officinale Roscoe) exerts a potential function in various cancers and is a noble combined therapy to overcome radioresistance in [...] Read more.
Radiotherapy is a powerful tumor therapeutic strategy for gastric cancer patients. However, radioresistance is a major obstacle to kill cancer cells. Ginger (Zingiber officinale Roscoe) exerts a potential function in various cancers and is a noble combined therapy to overcome radioresistance in gastric cancer radiotherapy. In this study, we suggested that 8-shogaol, a monomethoxybenzene compound extracted from Zingiber officinale Roscoe, has an anti-cancer and anti-inflammatory activity. In lipopolysaccharide (LPS)-induced inflammatory murine models in vivo and in vitro, 8-shogaol suppressed LPS-mediated cytokine production, including COX-2, TNFα, IL-6, and IL-1β. In xenograft mouse models of AGS gastric cancer cell lines, 8-shogaol reduced tumor volume. In gastric cancer cell lines AGS and NCI-N87, 8-shogaol reduced cell viability and increased caspase-3 activity and cytotoxicity LDH. However, combined with Z-VAD-FMK, 8-shogaol blocked caspase-dependent apoptotic cell death. 8-Shogaol induced intracellular reactive oxygen species (ROS) production, intracellular calcium (Ca2+) release, and endoplasmic reticulum (ER) stress response via the PERK-CHOP signaling pathway. Thapsigargin (TG), an ER stressor, mediated synergistic apoptosis and cell death in 8-shogaol-treated AGS and NCI-N87 cell lines. Nevertheless, loss of PERK or CHOP function suppressed ER-stress-induced apoptosis and cell death in 8-shogaol-treated AGS and NCI-N87 cell lines. 8-Shogaol-induced NADPH oxidase 4 (NOX4) activation is related to ROS generation. However, NOX4 knockdown and ROS inhibitors DPI or NAC blocked ER-stress-induced apoptosis by suppressing the inhibition of cell viability and the enhance of caspase-3 activity, intracellular ROS activity, and cytotoxicity LDH in 8-shogaol-treated AGS and NCI-N87 cell lines. Radioresistant gastric cancer models (AGSR and NCI-N87R) were developed and combined with 8-shogaol and radiation (2 Gy) to overcome radioresistance via the upregulation of N-cadherin and vimentin and the downregulation of E-cadherin. Therefore, these results indicated that 8-shogaol is a novel combined therapeutic strategy in gastric cancer radiotherapy. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

18 pages, 1859 KiB  
Article
A Novel PPARγ Modulator Falcarindiol Mediates ER Stress-Mediated Apoptosis by Regulating NOX4 and Overcomes Radioresistance in Breast Cancer
by Tae Woo Kim and Seong-Gyu Ko
Antioxidants 2024, 13(12), 1533; https://doi.org/10.3390/antiox13121533 - 14 Dec 2024
Cited by 1 | Viewed by 1262
Abstract
The extract of the rhizome of Cnidium officinale Makino has potential anti-cancer and anti-inflammatory effects in many diseases, such as cancer. However, the biological functions of falcarindiol (FAD) in breast cancer are not fully understood. This study proved the anti-inflammatory and anti-cancer effects [...] Read more.
The extract of the rhizome of Cnidium officinale Makino has potential anti-cancer and anti-inflammatory effects in many diseases, such as cancer. However, the biological functions of falcarindiol (FAD) in breast cancer are not fully understood. This study proved the anti-inflammatory and anti-cancer effects of FAD in breast cancer. Breast cancer models confirmed that FAD reduces cell viability and decreases the tumor volume of xenograft mouse models in a dose-dependent manner. FAD mediated caspase-3-dependent apoptosis in MDA-MB-231 and MCF-7 cells, whereas Z-VAD-FMK in combination with FAD inhibited caspase-3-induced apoptosis. FAD mediates apoptosis through cytosolic reactive oxygen species (ROS) and calcium (Ca2+) production and ER stress signaling pathways. In addition, FAD combined with thapsigargin (TG) exerts a synergistic apoptotic cell death effect. In the loss-of-function experiments, PERK or CHOP ablation suppressed intracellular ROS and Ca2+ release and ER stress-induced apoptosis in FAD-treated breast cancer models. Since there is a relationship between ROS and NADPH Oxidase 4 (NOX4), Nox4 ablation blocked ER stress-mediated apoptotic cell death by inhibiting ROS release in FAD-induced breast cancer models. Radioresistant models, such as MCF-7R and MDA-MB-231R, were developed to address the cellular radioresistance in clinical radiotherapy. FAD combined with radiation (2 Gy) overcame radioresistance via the inhibition of the epithelial–mesenchymal transition (EMT) phenomenon, such as the upregulation of PPARγ, VIM, and CDH2 and the downregulation of CDH1. Consequently, these results show that FAD may be a novel treatment as a breast cancer therapy. Full article
Show Figures

Figure 1

17 pages, 1966 KiB  
Article
Anti-Inflammatory and Anticancer Effects of Kaurenoic Acid in Overcoming Radioresistance in Breast Cancer Radiotherapy
by Tae Woo Kim and Seong-Gyu Ko
Nutrients 2024, 16(24), 4320; https://doi.org/10.3390/nu16244320 - 14 Dec 2024
Cited by 2 | Viewed by 1638
Abstract
Background/Objectives: Peroxisome proliferator–activated receptor γ (PPARγ) plays a key role in mediating anti-inflammatory and anticancer effects in the tumor microenvironment. Kaurenoic acid (KA), a diterpene compound isolated from Sphagneticola trilobata (L.) Pruski, has been demonstrated to exert anti-inflammatory, anticancer, and antihuman immunodeficiency [...] Read more.
Background/Objectives: Peroxisome proliferator–activated receptor γ (PPARγ) plays a key role in mediating anti-inflammatory and anticancer effects in the tumor microenvironment. Kaurenoic acid (KA), a diterpene compound isolated from Sphagneticola trilobata (L.) Pruski, has been demonstrated to exert anti-inflammatory, anticancer, and antihuman immunodeficiency virus effects. Methods: In this study, we identified KA as a novel activator of PPARγ with potent anti-inflammatory and antitumor effects both in vitro and in vivo. Given the potential of PPARγ regulators in overcoming radioresistance and chemoresistance in cancer therapies, we hypothesized that KA may enhance the efficacy of breast cancer radiotherapy. Results: In a lipopolysaccharide (LPS)-induced mouse inflammation model, KA treatment reduced the levels of pro-inflammatory cytokines, including COX-2, IL-6, IL-1β, and TNFα. In a xenograft mouse mode of breast cancer, KA treatment inhibited tumor growth. Specifically, KA treatment enhanced caspase-3 activity and cytotoxicity against MDA-MB-231 and MCF-7 breast cancer cells. When KA was co-treated with a caspase inhibitor, Z-VAD-FMK, caspase-dependent apoptosis was suppressed in these cells. KA was found to induce the generation of cytosolic calcium ions (Ca2+) and reactive oxygen species (ROS), triggering endoplasmic reticulum (ER) stress via the PERK-ATF4-CHOP axis. Hence, the ER stressor thapsigargin (TG) synergized with KA treatment to enhance apoptosis in these cells, while the loss of the PERK or CHOP function inhibited this phenomenon. KA treatment was shown to induce oxidative stress via the NADPH oxidase 4 (NOX4) and stimulate ROS production. Specifically, NOX4 knockdown (KD) and antioxidant treatment (N-acetyl cysteine or diphenyleneiodonium) suppressed such ER stress–mediated apoptosis by inhibiting KA-enhanced caspase-3 activity, cytotoxicity, and intracellular ROS production in the treated cells. In radioresistant MDA-MB-231R and MCF-7R cells, KA combined with 2 Gy radiation overcame radioresistance by upregulating PPARγ and modulating epithelial–mesenchymal transition (EMT) markers, such as E-cadherin, N-cadherin, and vimentin. In PPARγ KD MDA-MB-231R and MCF-7R cells, this phenomenon was inhibited due to reduced PPARγ and NOX4 expression. Conclusions: In conclusion, these findings demonstrated KA as a novel PPARγ regulator with promising potential to enhance the efficacy of breast cancer radiotherapy. Full article
(This article belongs to the Section Nutritional Immunology)
Show Figures

Figure 1

25 pages, 2250 KiB  
Article
SERCA Modulators Reveal Distinct Signaling and Functional Roles of T Lymphocyte Ca2+ Stores
by Md Nasim Uddin and David W. Thomas
Int. J. Mol. Sci. 2024, 25(22), 12095; https://doi.org/10.3390/ijms252212095 - 11 Nov 2024
Cited by 5 | Viewed by 1318
Abstract
The allosteric SERCA (Sarcoplasmic/Endoplasmic Reticulum Ca2+-ATPase) activator CDN1163 has been recently added to the group of pharmacological tools for probing SERCA function. We chose to investigate the effects of the compound on T lymphocyte Ca2+ stores, using the well-described Jurkat [...] Read more.
The allosteric SERCA (Sarcoplasmic/Endoplasmic Reticulum Ca2+-ATPase) activator CDN1163 has been recently added to the group of pharmacological tools for probing SERCA function. We chose to investigate the effects of the compound on T lymphocyte Ca2+ stores, using the well-described Jurkat T lymphocyte as a reliable cell system for Ca2+ signaling pathways. Our study identified the lowest concentrations of the SERCA inhibitors thapsigargin (TG) and 2,5-di-(tert butyl)-1,4-benzohydroquinone (tBHQ) capable of releasing Ca2+, permitting the differentiation of the TG-sensitive SERCA 2b Ca2+ store from the tBHQ-sensitive SERCA 3 Ca2+ store. We proceeded to test the effects of CDN1163 on Ca2+ stores, examining specific actions on the SERCA 2b and SERCA 3 Ca2+ pools using our low-dose SERCA blocker regimen. In contrast to previous work, we find CDN1163 exerts complex time-sensitive and SERCA isoform-specific actions on Ca2+ stores. Surprisingly, short-term exposure (0–30 min) to CDN1163 perturbs T cell Ca2+ stores by suppressing Ca2+ uptake with diminished Ca2+ release from the SERCA 2b-controlled store. Concomitantly, we find evidence for a SERCA-activating effect of CDN1163 on the SERCA-3 regulated store, given the observation of increased Ca2+ release inducible by low-dose tBHQ. Intriguingly, longer-term (>12 h) CDN1163 exposure reversed this pattern, with increased Ca2+ release from SERCA 2b-regulated pools yet decreased Ca2+ release responses from the tBHQ-sensitive SERCA 3 pool. Indeed, this remodeling of SERCA 2b Ca2+ stores with longer-term CDN1163 exposure also translated into the compound’s ability to protect Jurkat T lymphocytes from TG but not tBHQ-induced growth suppression. Full article
(This article belongs to the Special Issue Calcium Signaling in Health and Diseases)
Show Figures

Figure 1

13 pages, 2066 KiB  
Article
Inhibition of K+ Channels Affects the Target Cell Killing Potential of CAR T Cells
by Ghofrane Medyouni, Orsolya Vörös, Vivien Jusztus, György Panyi, György Vereb, Árpád Szöőr and Péter Hajdu
Cancers 2024, 16(22), 3750; https://doi.org/10.3390/cancers16223750 - 6 Nov 2024
Cited by 1 | Viewed by 1298
Abstract
Ion channels of T cells (Kv1.3, KCa3.1, and CRAC) participate in the regulation of activation and effector functions via modulation of the Ca2+-dependent pathway. T cells expressing chimeric antigen receptors (CAR T cells) showed a remarkable role in anti-tumor therapy, especially [...] Read more.
Ion channels of T cells (Kv1.3, KCa3.1, and CRAC) participate in the regulation of activation and effector functions via modulation of the Ca2+-dependent pathway. T cells expressing chimeric antigen receptors (CAR T cells) showed a remarkable role in anti-tumor therapy, especially in the treatment of chemotherapy-resistant liquid cancers. Nevertheless, many challenges remain to be overcome to improve the treatment for solid tumors. In this study, we assessed the expression and role of ion channels in CAR T cells. We found that HER2-specific CAR T cells had higher KCa3.1 conductance compared to the non-transduced (NT, control) cells, which was more prominent in the CD8+ population (CD4+ cell also showed elevation). Conversely, the Kv1.3 expression level was the same for all cell types (CD4+, CD8+, CAR, and NT). Single-cell Ca2+ imaging revealed that thapsigargin-induced SOCE via CRAC is suppressed in CD8+ CAR T cells, unlike for CD4+ and CD8+ NT cells. To dissect the functional role of Kv1.3 and KCa3.1, we used specific antagonists (Kv1.3: Vm24; KCa3.1: TRAM-34): the target cell elimination capacity of the CD8+ CAR T cells was improved either by blocking KCa3.1 or Kv1.3. These results imply that ion channels could be a target in CAR T cell immunotherapy elaboration. Full article
(This article belongs to the Special Issue The Emerging Role of Ion Channels in Cancer Treatment)
Show Figures

Figure 1

19 pages, 1704 KiB  
Article
Readdressing the Localization of Apolipoprotein E (APOE) in Mitochondria-Associated Endoplasmic Reticulum (ER) Membranes (MAMs): An Investigation of the Hepatic Protein–Protein Interactions of APOE with the Mitochondrial Proteins Lon Protease (LONP1), Mitochondrial Import Receptor Subunit TOM40 (TOMM40) and Voltage-Dependent Anion-Selective Channel 1 (VDAC1)
by Johanna Rueter, Gerald Rimbach, Stephanie Bilke, Andreas Tholey and Patricia Huebbe
Int. J. Mol. Sci. 2024, 25(19), 10597; https://doi.org/10.3390/ijms251910597 - 1 Oct 2024
Cited by 1 | Viewed by 2396
Abstract
As a component of circulating lipoproteins, APOE binds to cell surface receptors mediating lipoprotein metabolism and cholesterol transport. A growing body of evidence, including the identification of a broad variety of cellular proteins interacting with APOE, suggests additional independent functions. Investigating cellular localization [...] Read more.
As a component of circulating lipoproteins, APOE binds to cell surface receptors mediating lipoprotein metabolism and cholesterol transport. A growing body of evidence, including the identification of a broad variety of cellular proteins interacting with APOE, suggests additional independent functions. Investigating cellular localization and protein–protein interactions in cultured human hepatocytes, we aimed to contribute to the elucidation of hitherto unnoted cellular functions of APOE. We observed a strong accumulation of APOE in MAMs, equally evident for the two major isoforms APOE3 and APOE4. Using mass spectrometry proteome analyses, novel and previously noted APOE interactors were identified, including the mitochondrial proteins TOMM40, LONP1 and VDAC1. All three interactors were present in MAM fractions, which we think initially facilitates interactions with APOE. LONP1 is a protease with chaperone activity, which migrated to MAMs in response to ER stress, displaying a reinforced interaction with APOE. We therefore hypothesize that APOE may help in the unfolded protein response (UPR) by acting as a co-chaperone in cooperation with LONP1 at the interface of mitochondria and ER membranes. The interaction of APOE with the integral proteins TOMM40 and VDAC1 may point to the formation of bridging complexes connecting mitochondria with other organelles. Full article
(This article belongs to the Special Issue Apolipoproteins and Lipoproteins in Health and Disease, 3rd Edition)
Show Figures

Figure 1

14 pages, 6818 KiB  
Communication
UTRs and Ago-2/miR-335 Complex Restricts Amylin Translation in Insulinoma and Human Pancreatic β-Cells
by Zhanar Kudaibergenova, Satyabrata Pany, Elizabeth Placheril and Aleksandar M. Jeremic
Int. J. Mol. Sci. 2024, 25(17), 9614; https://doi.org/10.3390/ijms25179614 - 5 Sep 2024
Cited by 1 | Viewed by 1231
Abstract
Amylin promoter and transcriptional factors are well-established, inducible factors in the production of the main amyloidogenic pancreatic hormone, human islet amyloid peptide (hIAPP) or amylin. However, posttranscriptional mechanisms driving hIAPP expression in pancreas remain enigmatic, and hence were explored here. The translational assay [...] Read more.
Amylin promoter and transcriptional factors are well-established, inducible factors in the production of the main amyloidogenic pancreatic hormone, human islet amyloid peptide (hIAPP) or amylin. However, posttranscriptional mechanisms driving hIAPP expression in pancreas remain enigmatic, and hence were explored here. The translational assay revealed that both 5′ and 3′ untranslated regions (UTRs) of hIAPP restricted expression of the luciferase constructs only in constructs driven by the hIAPP promoter. Bioinformatics analysis revealed several putative seed sequences for a dozen micro RNAs (miRNAs) in hIAPP’s 3′ UTR. miR-182, miR-335, and miR-495 were the most downregulated miRNAs in stressed human islets exposed to endoplasmic reticulum (ER) or metabolic stressors, thapsigargin (TG) or high glucose (HG). Correspondingly, miR-335 mimics alone or in combination with miR-495 and miR-182 mimics significantly and potently (>3-fold) reduced hIAPP protein expression in HG-treated cultured human islets. siRNA-mediated silencing of Ago2 but not Ago1 significantly stimulated hIAPP expression and secretion from transfected, HG-treated human islets. Conversely, ectopic expression of Ago2 in hIAPP-expressing RIN-m5F cell line driven by CMV promoter reduced hIAPP intracellular protein levels. Collectively, the results point to a novel and synergistic role for hIAPP promoter, 5/3′ UTRs and Ago-2/miR-335 complex in post-transcriptional regulation of hIAPP gene expression in normal and metabolically active β-cells. Full article
(This article belongs to the Special Issue Molecular Research on Diabetes)
Show Figures

Figure 1

17 pages, 2660 KiB  
Article
The ER Stress Induced in Human Neuroblastoma Cells Can Be Reverted by Lumacaftor, a CFTR Corrector
by Michela Pecoraro, Adele Serra, Maria Pascale and Silvia Franceschelli
Curr. Issues Mol. Biol. 2024, 46(9), 9342-9358; https://doi.org/10.3390/cimb46090553 - 24 Aug 2024
Cited by 2 | Viewed by 1641
Abstract
Most neurodegenerative diseases share a common etiopathogenesis, the accumulation of protein aggregates. An imbalance in homeostasis brought on by the buildup of misfolded proteins within the endoplasmic reticulum (ER) results in ER stress in the cell. Three distinct proteins found in the ER [...] Read more.
Most neurodegenerative diseases share a common etiopathogenesis, the accumulation of protein aggregates. An imbalance in homeostasis brought on by the buildup of misfolded proteins within the endoplasmic reticulum (ER) results in ER stress in the cell. Three distinct proteins found in the ER membrane—IRE1α, PERK, and ATF6—control the unfolded protein response (UPR), a signal transduction pathway that is triggered to restore normal physiological conditions. Buildup of misfolded proteins in ER lumen leads to a shunting of GRP78/BiP, thus triggering the UPR. PERK autophosphorylation leads to activation of ATF4, the transcription factor; finally, ATF6 activates the UPR’s target genes, including GRP78/Bip. Accordingly, the UPR is a cellular reaction to an ER stress state that, if left unchecked for an extended period, results in apoptosis and irreversible damage. The identification of caspase 4, which is in the ER and is selectively activated by apoptotic stimuli caused by reticular stress, further demonstrated the connection between reticular stress and programed cell death. Moreover, oxidative stress and ER stress are linked. Oxidative stress is brought on by elevated quantities of radical oxygen species, both mitochondrial and cytosolic, that are not under the enzymatic regulation of superoxide dismutases, whose levels fall with increasing stress. Here, we evaluated the activity of Vx-809 (Lumacaftor), a drug used in cystic fibrosis, in SH-SY5Y neuronal cells, in which an ER stress condition was induced by Thapsigargin, to verify whether the drug could improve protein folding, suggesting its possible therapeutic use in proteinopathies, such as neurodegenerative diseases (NDs). Our data show that Vx-809 is involved in the significant reduction in protein produced under ER stress, particularly in the levels of Bip, ATF4, and ATF6 by Western blotting analysis, the reduction in ROS in the cytosol and mitochondria, and the reduction in the activation of the apoptotic pathway, measured by flow cytofluorimetry analysis and in restoring calcium homeostasis. Full article
(This article belongs to the Special Issue Molecules at Play in Neurological Diseases)
Show Figures

Figure 1

Back to TopTop