1
|
Sengupta A, Chakraborty S, Biswas S, Patra SK, Ghosh S. S-nitrosoglutathione (GSNO) induces necroptotic cell death in K562 cells: Involvement of p73, TSC2 and SIRT1. Cell Signal 2024; 124:111377. [PMID: 39222864 DOI: 10.1016/j.cellsig.2024.111377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 08/21/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Nitric oxide and Reactive Nitrogen Species are known to effect tumorigenicity. GSNO is one of the main NO carrying signalling moiety in cell. In the current study, we tried to delve into the effect of GSNO induced nitrosative stress in three different myelogenous leukemic K562, U937 and THP-1 cell lines. METHOD WST-8 assay was performed to investigate cell viability. RT-PCR and western-blot analysis were done to investigate mRNA and protein expression. Spectrophotometric and fluorimetric assays were done to investigate enzyme activities. RESULT We found that GSNO exposure led to reduced cell viability and the mode of cell death in K562 was non apoptotic in nature. GSNO promoted impaired autophagic flux and necroptosis. GSNO treatment heightened phosphorylation of AMPK and TSC2 and inhibited mTOR pathway. We observed increase in NAD+/ NADH ratio following GSNO treatment. Increase in both SIRT1 m-RNA and protein expression was observed. While total SIRT activity remained unaltered. GSNO increased tumor suppressor TAp73/ oncogenic ∆Np73 ratio in K562 cells which was correlated with cell mortality. Surprisingly, GSNO did not alter cellular redox status or redox associated protein expression. However, steep increase in total SNO and PSNO content was observed. Furthermore, inhibition of autophagy, AMPK phosphorylation or SIRT1 exacerbated the effect of GSNO. Altogether our work gives insights into GSNO mediated necroptotic event in K562 cells which can be excavated to develop NO based anticancer therapeutics. CONCLUSION Our data suggests that GSNO could induce necroptotic cell death in K562 through mitochondrial dysfunctionality and PTM of different cellular proteins.
Collapse
Affiliation(s)
- Ayantika Sengupta
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Subhamoy Chakraborty
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sanchita Biswas
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sourav Kumar Patra
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sanjay Ghosh
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| |
Collapse
|
2
|
Shechter S, Pal RK, Trovato F, Rozen O, Gage MJ, Avni D. p70S6K as a Potential Anti-COVID-19 Target: Insights from Wet Bench and In Silico Studies. Cells 2024; 13:1760. [PMID: 39513867 DOI: 10.3390/cells13211760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/27/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
The onset of SARS-CoV-2 infection in 2019 sparked a global COVID-19 pandemic. This infection is marked by a significant rise in both viral and host kinase activity. Our primary objective was to identify a pivotal host kinase essential for COVID-19 infection and the associated phenomenon of the cytokine storm, which may lead to long-term COVID-19 complications irrespective of viral genetic variations. To achieve this, our study tracked kinase phosphorylation dynamics in RAW264.7 macrophages following SPIKE transfection over time. Among the kinases surveyed, p70S6K (RPS6KB1) exhibited a 3.5-fold increase in phosphorylation at S418. This significant change prompted the selection of p70S6K for in silico investigation, utilizing its structure bound to M2698 (PDB: 7N93). M2698, an oral dual Akt/p70S6K inhibitor with an IC50 of 1.1 nM, exhibited psychosis side effects in phase I clinical trials, potentially linked to its interaction with Akt2. Our secondary objective was to discover a small-molecule analogue of M2698 that exhibits a distinct binding preference for p70S6K over Akt2 through computational modeling and analysis. The in silico part of our project began with validating the prediction accuracy of the docking algorithm, followed by an OCA analysis pinpointing specific atoms on M2698 that could be modified to enhance selectivity. Subsequently, our investigation led to the identification of an analog of M2698, designated as S34, that showed a superior docking score towards p70S6K compared to Akt2. To further assess the stability of S34 in its protein-ligand (PL) complexes with p70S6K and Akt2, MD simulations were conducted. These simulations suggest that S34, on average, forms two hydrogen bond interactions with p70S6K, whereas it only forms one hydrogen bond interaction with Akt2. This difference in hydrogen bond interactions likely contributed to the observed larger root mean square deviation (RMSD) of 0.3 nm in the S34-Akt2 complex, compared to 0.1 nm in the S34-p70S6K complex. Additionally, we calculated free binding energy to predict the strength of the binding interactions of S34 to p70S6K and Akt2, which showed ~2-fold favorable binding affinity of S34 in the p70S6K binding pocket compared to that in the Akt2 binding pocket. These observations may suggest that the S34-p70S6K complex is more stable than the S34-Akt2 complex. Our work focused on identifying a host kinase target and predicting the binding affinity of a novel small molecule to accelerate the development of effective treatments. The wet bench results specifically highlight p70S6K as a compelling anti-COVID-19 target. Meanwhile, our in silico investigations address the known off-target effects associated with M2698 by identifying a close analog called S34. In conclusion, this study presents novel and intriguing findings that could potentially lead to clinical applications with further investigations.
Collapse
Affiliation(s)
- Sharon Shechter
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | | | - Fabio Trovato
- Psivant Therapeutics, 451 D Street, Boston, MA 02210, USA
| | - Or Rozen
- Department of Natural Compound, Nutrition, and Health, MIGAL Galilee Research Institute, Kiryat Shmona 1101600, Israel
| | - Matthew J Gage
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Dorit Avni
- Department of Natural Compound, Nutrition, and Health, MIGAL Galilee Research Institute, Kiryat Shmona 1101600, Israel
| |
Collapse
|
3
|
Kondo S, Karasawa T, Koike A, Tsutsui M, Kunisawa J, Terada S. Decreased pancreatic amylase activity after acute high-intensity exercise and its effects on post-exercise muscle glycogen recovery. Appl Physiol Nutr Metab 2024; 49:1035-1046. [PMID: 38621297 DOI: 10.1139/apnm-2023-0265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Our prior results showed that an acute bout of endurance exercise for 6 h, but not 1 h, decreased pancreatic amylase activity, indicating that acute endurance exercise may affect carbohydrate digestive capacity in an exercise duration-dependent manner. Here, we investigated the effects of acute endurance exercise of different intensities on mouse pancreatic amylase activity. Male C57BL/6J mice performed low- or high-intensity running exercise for 60 min at either 10 (Ex-Low group) or 20 m/min (Ex-High group). The control group comprised sedentary mice. Immediately after acute exercise, pancreatic amylase activity was significantly decreased in the Ex-High group and not the Ex-Low group in comparison with the control group. To determine whether the decreased amylase activity induced by high-intensity exercise influenced muscle glycogen recovery after exercise, we investigated the rates of muscle glycogen resynthesis in Ex-High group mice administered either oral glucose or starch solution (2.0 mg/g body weight) immediately after exercise. The starch-fed mice exhibited significantly lower post-exercise glycogen accumulation rates in the 2-h recovery period compared with the glucose-fed mice. This difference in the glycogen accumulation rate was absent for starch- and glucose-fed mice in the sedentary (no exercise) control group. Furthermore, the plasma glucose AUC during early post-exercise recovery (0-60 min) was significantly lower in the starch-fed mice than in the glucose-fed mice. Thus, our findings suggest that acute endurance exercise diminishes the carbohydrate digestive capacity of the pancreas in a manner dependent on exercise intensity, with polysaccharides leading to delayed muscle glycogen recovery after exercise.
Collapse
Affiliation(s)
- Saki Kondo
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental Health, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Takuya Karasawa
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Atsuko Koike
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Momoko Tsutsui
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental Health, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Shin Terada
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Takahashi K, Kurokawa K, Hong L, Miyagawa K, Mochida-Saito A, Takeda H, Tsuji M. Hippocampal and gut AMPK activation attenuates enterocolitis-like symptoms and co-occurring depressive-like behavior in ulcerative colitis model mice: Involvement of brain-gut autophagy. Exp Neurol 2024; 373:114671. [PMID: 38160982 DOI: 10.1016/j.expneurol.2023.114671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/13/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
Patients with inflammatory bowel disease, including ulcerative colitis (UC) and Crohn's disease, have a high incidence of psychiatric disorders, including depression and anxiety. However, the underlying pathogenic mechanism remains unknown. Dextran sulfate sodium (DSS)-treated mice, a model of UC, exhibit depressive-like behavior and reduced adenosine monophosphate-activated protein kinase (AMPK) activity, which regulates various physiological functions in the brain and gut. However, comprehensive studies on UC pathophysiology with co-occurring depression focused on brain-gut AMPK activity are lacking. Therefore, we aimed to investigate whether resveratrol (RES), an AMPK activator, prevented DSS-induced UC-like symptoms and depressive-like behavior. DSS treatment induced UC-like pathology and depressive-like behavior, as assessed via the tail suspension test. Moreover, western blotting and immunohistochemical studies revealed that DSS increased p-p70S6 kinase (Thr389), p62, tumor necrosis factor-α, interleukin (IL)-1β, IL-18, NLR family pyrin domain containing 3 (NLRP3), cleaved caspase-1, cleaved Gasdermin-D (GSDMD), and cleaved caspase-3 expression levels in the rectum and hippocampus, and increased CD40, iNOS, and Kelch-like ECH-associated protein 1 expression levels, and the number of Iba1-positive cells in the hippocampus, and decreased p-AMPK and LC3II/I expression levels, and the number of NF-E2-related factor 2 (Nrf2)-positive cells, and reduced neurogenesis in the hippocampus. These changes were reversed by the RES administration. RES also enhanced PGC1α and SOD1 expression in the hippocampus of DSS-treated male mice. Moreover, NLRP3 staining was observed in the neurons and microglia, and cleaved GSDMD staining in neurons in the hippocampus of DSS-treated mice. Notably, RES prevented UC-like pathology and depressive-like behavior and enhancement of autophagy, decreased rectal and hippocampal inflammatory cytokines and inflammasome, and induced the Nrf2-PGC1α-SOD1 pathway in the hippocampus, resulting in neurogenesis in the hippocampal dentate gyrus. Our findings suggest that brain-gut AMPK activation may be an important therapeutic strategy in patients with UC and depression.
Collapse
Affiliation(s)
- Kohei Takahashi
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Kazuhiro Kurokawa
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Lihua Hong
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Kazuya Miyagawa
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Atsumi Mochida-Saito
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Hiroshi Takeda
- Department of Pharmacology, School of Pharmacy at Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa, Fukuoka 831-8501, Japan
| | - Minoru Tsuji
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan.
| |
Collapse
|
5
|
Yurube T, Buchser WJ, Zhang Z, Silwal P, Lotze MT, Kang JD, Sowa GA, Vo NV. Rapamycin mitigates inflammation-mediated disc matrix homeostatic imbalance by inhibiting mTORC1 and inducing autophagy through Akt activation. JOR Spine 2024; 7:e1303. [PMID: 38222800 PMCID: PMC10782056 DOI: 10.1002/jsp2.1303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/05/2023] [Accepted: 10/31/2023] [Indexed: 01/16/2024] Open
Abstract
Background Low back pain is a global health problem that originated mainly from intervertebral disc degeneration (IDD). Autophagy, negatively regulated by the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway, prevents metabolic and degenerative diseases by removing and recycling damaged cellular components. Despite growing evidence that autophagy occurs in the intervertebral disc, the regulation of disc cellular autophagy is still poorly understood. Methods Annulus fibrosus (rAF) cell cultures derived from healthy female rabbit discs were used to test the effect of autophagy inhibition or activation on disc cell fate and matrix homeostasis. Specifically, different chemical inhibitors including rapamycin, 3-methyladenine, MK-2206, and PP242 were used to modulate activities of different proteins in the PI3K/Akt/mTOR signaling pathway to assess IL-1β-induced cellular senescence, apoptosis, and matrix homeostasis in rAF cells grown under nutrient-poor culture condition. Results Rapamycin, an inhibitor of mTOR complex 1 (mTORC1), reduced the phosphorylation of mTOR and its effector p70/S6K in rAF cell cultures. Rapamycin also induced autophagic flux as measured by increased expression of key autophagy markers, including LC3 puncta number, LC3-II expression, and cytoplasmic HMGB1 intensity and decreased p62/SQSTM1 expression. As expected, IL-1β stimulation promoted rAF cellular senescence, apoptosis, and matrix homeostatic imbalance with enhanced aggrecanolysis and MMP-3 and MMP-13 expression. Rapamycin treatment effectively mitigated IL-1β-mediated inflammatory stress changes, but these alleviating effects of rapamycin were abrogated by chemical inhibition of Akt and mTOR complex 2 (mTORC2). Conclusions These findings suggest that rapamycin blunts adverse effects of inflammation on disc cells by inhibiting mTORC1 to induce autophagy through the PI3K/Akt/mTOR pathway that is dependent on Akt and mTORC2 activities. Hence, our findings identify autophagy, rapamycin, and PI3K/Akt/mTOR signaling as potential therapeutic targets for IDD treatment.
Collapse
Affiliation(s)
- Takashi Yurube
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic SurgeryUniversity of Pittsburgh Medical Cancer, University of PittsburghPittsburghPennsylvaniaUSA
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - William J. Buchser
- Damage Associated Molecular Pattern Molecule Laboratory, Department of Surgery, Hillman Cancer CenterUniversity of Pittsburgh Cancer Institute, University of PittsburghPittsburghPennsylvaniaUSA
| | - Zhongying Zhang
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic SurgeryUniversity of Pittsburgh Medical Cancer, University of PittsburghPittsburghPennsylvaniaUSA
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Prashanta Silwal
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic SurgeryUniversity of Pittsburgh Medical Cancer, University of PittsburghPittsburghPennsylvaniaUSA
| | - Michael T. Lotze
- Damage Associated Molecular Pattern Molecule Laboratory, Department of Surgery, Hillman Cancer CenterUniversity of Pittsburgh Cancer Institute, University of PittsburghPittsburghPennsylvaniaUSA
| | - James D. Kang
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic SurgeryUniversity of Pittsburgh Medical Cancer, University of PittsburghPittsburghPennsylvaniaUSA
- Department of Orthopedics, Brigham and Women's Hospital, School of MedicineHarvard UniversityBostonMassachusettsUSA
| | - Gwendolyn A. Sowa
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic SurgeryUniversity of Pittsburgh Medical Cancer, University of PittsburghPittsburghPennsylvaniaUSA
- Department of Physical Medicine and RehabilitationUniversity of Pittsburgh Medical Cancer, University of PittsburghPittsburghPennsylvaniaUSA
| | - Nam V. Vo
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic SurgeryUniversity of Pittsburgh Medical Cancer, University of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
6
|
Hu L, Sui X, Dong X, Li Z, Lun S, Wang S. Low beauvericin concentrations promote PC-12 cell survival under oxidative stress by regulating lipid metabolism and PI3K/AKT/mTOR signaling. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115786. [PMID: 38061083 DOI: 10.1016/j.ecoenv.2023.115786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 01/12/2024]
Abstract
Beauvericin (BEA), a naturally occurring cyclic peptide with good pharmacological activity, has been widely explored in anticancer research. Although BEA is toxic, studies have demonstrated its antioxidant activity. However, to date, the antioxidant mechanisms of BEA remain unclear. Herein, we conducted a comprehensive and detailed study of the antioxidant mechanism of BEA using an untargeted metabolomics approach, subsequently validating the results. BEA concentrations of 0.5 and 1 μM significantly inhibited H2O2-induced oxidative stress (OS), decreased reactive oxygen species levels in PC-12 cells, and restored the mitochondrial membrane potential. Untargeted metabolomics indicated that BEA was primarily involved in lipid-related metabolism, suggesting its role in resisting OS in PC-12 cells by participating in lipid metabolism. BEA combated OS damage by increasing phosphatidylcholine, phosphatidylethanolamine, and sphingolipid levels. In the current study, BEA upregulated proteins related to the PI3K/AKT/mTOR pathway, thereby promoting cell survival. These findings support the antioxidant activity of BEA at low concentrations, warranting further research into its pharmacological effects.
Collapse
Affiliation(s)
- Liming Hu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Xintong Sui
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Xin Dong
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Zhimeng Li
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Shiyi Lun
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Shumin Wang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| |
Collapse
|
7
|
Alattar A, Alshaman R, Althobaiti YS, Soliman GM, Ali HS, Khubrni WS, Koh PO, Rehman NU, Shah FA. Quercetin Alleviated Inflammasome-Mediated Pyroptosis and Modulated the mTOR/P70S6/P6/eIF4E/4EBP1 Pathway in Ischemic Stroke. Pharmaceuticals (Basel) 2023; 16:1182. [PMID: 37631097 PMCID: PMC10459024 DOI: 10.3390/ph16081182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Stroke ranks as the world's second most prevalent cause of mortality, and it represents a major public health concern with profound economic and social implications. In the present study, we elucidated the neuroprotective role of quercetin on NLRP3-associated pyroptosis, Nrf2-coupled anti-inflammatory, and mTOR-dependent downstream pathways. Male Sprague Dawley rats were subjected to 72 h of transient middle cerebral artery ischemia, followed by the administration of 10 mg/kg of quercetin. Our findings demonstrated that MCAO induced elevated ROS which were coupled to inflammasome-mediated pyroptosis and altered mTOR-related signaling proteins. We performed ELISA, immunohistochemistry, and Western blotting to unveil the underlying role of the Nrf2/HO-1 and PDK/AKT/mTOR pathways in the ischemic cortex and striatum. Our results showed that quercetin post-treatment activated the Nrf2/HO-1 cascade, reversed pyroptosis, and modulated the autophagy-related pathway PDK/AKT/mTOR/P70S6/P6/eIF4E/4EBP1. Further, quercetin enhances the sequestering effect of 14-3-3 and reversed the decrease in interaction between p-Bad and 14-3-3 and p-FKHR and 14-3-3. Our findings showed that quercetin exerts its protective benefits and rescues neuronal damage by several mechanisms, and it might be a viable neuroprotective drug for ischemic stroke therapy.
Collapse
Affiliation(s)
- Abdullah Alattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 47713, Saudi Arabia; (R.A.); (W.S.K.)
| | - Reem Alshaman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 47713, Saudi Arabia; (R.A.); (W.S.K.)
| | - Yusuf S. Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 21944, Taif 21944, Saudi Arabia;
- Addiction and Neuroscience Research Unit, Taif University, Taif 21944, Saudi Arabia
| | - Ghareb M. Soliman
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk 47713, Saudi Arabia;
| | - Howaida S. Ali
- Department of Pharmacology, Faculty of Medicine, Assuit University, Assuit 71515, Egypt;
- Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Waleed Salman Khubrni
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 47713, Saudi Arabia; (R.A.); (W.S.K.)
| | - Phil Ok Koh
- Department of Anatomy and Histology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Najeeb Ur Rehman
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sttam Bin Abdul Aziz University, Al-Kharj 11942, Saudi Arabia;
| | - Fawad Ali Shah
- Department of Anatomy and Histology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea;
| |
Collapse
|
8
|
Cox A, Tung M, Li H, Hallows KR, Chung EJ. In vitro delivery of mTOR inhibitors by kidney-targeted micelles for autosomal dominant polycystic kidney disease. SLAS Technol 2023; 28:223-229. [PMID: 36804177 PMCID: PMC10439257 DOI: 10.1016/j.slast.2023.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/22/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic kidney disease and is characterized by the formation of renal cysts and the eventual development of end-stage kidney disease. One approach to treating ADPKD is through inhibition of the mammalian target of rapamycin (mTOR) pathway, which has been implicated in cell overproliferation, contributing to renal cyst expansion. However, mTOR inhibitors, including rapamycin, everolimus, and RapaLink-1, have off-target side effects including immunosuppression. Thus, we hypothesized that the encapsulation of mTOR inhibitors in drug delivery carriers that target the kidneys would provide a strategy that would enable therapeutic efficacy while minimizing off-target accumulation and associated toxicity. Toward eventual in vivo application, we synthesized cortical collecting duct (CCD) targeted peptide amphiphile micelle (PAM) nanoparticles and show high drug encapsulation efficiency (>92.6%). In vitro analysis indicated that drug encapsulation into PAMs enhanced the anti-proliferative effect of all three drugs in human CCD cells. Analysis of in vitro biomarkers of the mTOR pathway via western blotting confirmed that PAM encapsulation of mTOR inhibitors did not reduce their efficacy. These results indicate that PAM encapsulation is a promising way to deliver mTOR inhibitors to CCD cells and potentially treat ADPKD. Future studies will evaluate the therapeutic effect of PAM-drug formulations and ability to prevent off-target side effects associated with mTOR inhibitors in mouse models of ADPKD.
Collapse
Affiliation(s)
- Alysia Cox
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Madelynn Tung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Hui Li
- Department of Medicine, Division of Nephrology and Hypertension, and USC/UKRO Kidney Research Center, University of Southern California, Los Angeles, CA, USA
| | - Kenneth R Hallows
- Department of Medicine, Division of Nephrology and Hypertension, and USC/UKRO Kidney Research Center, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Division of Nephrology and Hypertension, and USC/UKRO Kidney Research Center, University of Southern California, Los Angeles, CA, USA; Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Derwich A, Sykutera M, Bromińska B, Rubiś B, Ruchała M, Sawicka-Gutaj N. The Role of Activation of PI3K/AKT/mTOR and RAF/MEK/ERK Pathways in Aggressive Pituitary Adenomas-New Potential Therapeutic Approach-A Systematic Review. Int J Mol Sci 2023; 24:10952. [PMID: 37446128 PMCID: PMC10341524 DOI: 10.3390/ijms241310952] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/18/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Pituitary tumors (PT) are mostly benign, although occasionally they demonstrate aggressive behavior, invasion of surrounding tissues, rapid growth, resistance to conventional treatments, and multiple recurrences. The pathogenesis of PT is still not fully understood, and the factors responsible for its invasiveness, aggressiveness, and potential for metastasis are unknown. RAF/MEK/ERK and mTOR signaling are significant pathways in the regulation of cell growth, proliferation, and survival, its importance in tumorigenesis has been highlighted. The aim of our review is to determine the role of the activation of PI3K/AKT/mTOR and RAF/MEK/ERK pathways in the pathogenesis of pituitary tumors. Additionally, we evaluate their potential in a new therapeutic approach to provide alternative therapies and improved outcomes for patients with aggressive pituitary tumors that do not respond to standard treatment. We perform a systematic literature search using the PubMed, Embase, and Scopus databases (search date was 2012-2023). Out of the 529 screened studies, 13 met the inclusion criteria, 7 related to the PI3K/AKT/mTOR pathway, and 7 to the RAF/MEK/ERK pathway (one study was used in both analyses). Understanding the specific factors involved in PT tumorigenesis provides opportunities for targeted therapies. We also review the possible new targeted therapies and the use of mTOR inhibitors and TKI in PT management. Although the RAF/MEK/ERK and PI3K/AKT/mTOR pathways play a pivotal role in the complex signaling network along with many interactions, further research is urgently needed to clarify the exact functions and the underlying mechanisms of these signaling pathways in the pathogenesis of pituitary adenomas and their role in its invasiveness and aggressive clinical outcome.
Collapse
Affiliation(s)
- Aleksandra Derwich
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Monika Sykutera
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| | - Barbara Bromińska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| | - Błażej Rubiś
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| | - Nadia Sawicka-Gutaj
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| |
Collapse
|
10
|
J Tisdale E, R Artalejo C. Rab2 stimulates LC3 lipidation on secretory membranes by noncanonical autophagy. Exp Cell Res 2023; 429:113635. [PMID: 37201743 DOI: 10.1016/j.yexcr.2023.113635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/20/2023]
Abstract
The Golgi complex is a highly dynamic organelle that regulates various cellular activities and yet maintains a distinct structure. Multiple proteins participate in Golgi structure/organization including the small GTPase Rab2. Rab2 is found on the cis/medial Golgi compartments and the endoplasmic reticulum-Golgi intermediate compartment. Interestingly, Rab2 gene amplification occurs in a wide range of human cancers and Golgi morphological alterations are associated with cellular transformation. To learn how Rab2 'gain of function' influences the structure/activity of membrane compartments in the early secretory pathway that may contribute to oncogenesis, NRK cells were transfected with Rab2B cDNA. We found that Rab2B overexpression had a dramatic effect on the morphology of pre- and early Golgi compartments that resulted in a decreased transport rate of VSV-G in the early secretory pathway. We monitored the cells for the autophagic marker protein LC3 based on the findings that depressed membrane trafficking affects homeostasis. Morphological and biochemical studies confirmed that Rab2 ectopic expression stimulated LC3-lipidation on Rab2-containing membranes that was dependent on GAPDH and utilized a non-canonical LC3-conjugation mechanism that is nondegradative. Golgi structural alterations are associated with changes in Golgi-associated signalling pathways. Indeed, Rab2 overexpressing cells had elevated Src activity. We propose that increased Rab2 expression facilitates cis Golgi structural changes that are maintained and tolerated by the cell due to LC3 tagging, and subsequent membrane remodeling triggers Golgi associated signaling pathways that may contribute to oncogenesis.
Collapse
Affiliation(s)
- Ellen J Tisdale
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, 48202, USA.
| | - Cristina R Artalejo
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, 48202, USA
| |
Collapse
|
11
|
Clinical Implications of mTOR Expression in Papillary Thyroid Cancer—A Systematic Review. Cancers (Basel) 2023; 15:cancers15061665. [PMID: 36980552 PMCID: PMC10046096 DOI: 10.3390/cancers15061665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Papillary thyroid cancer (PTC) comprises approximately 80% of all thyroid malignancies. Although several etiological factors, such as age, gender, and irradiation, are already known to be involved in the development of PTC, the genetics of cancerogenesis remain undetermined. The mTOR pathway regulates several cellular processes that are critical for tumorigenesis. Activated mTOR is involved in the development and progression of PTC. Therefore, we performed a systematic review of papers studying the expression of the mTOR gene and protein and its relationship with PTC risk and clinical outcome. A systematic literature search was performed using PubMed, Embase, and Scopus databases (the search date was 2012–2022). Studies investigating the expression of mTOR in the peripheral blood or tissue of patients with PTC were deemed eligible for inclusion. Seven of the 286 screened studies met the inclusion criteria for mTOR gene expression and four for mTOR protein expression. We also analyzed the data on mTOR protein expression in PTC. We analyzed the association of mTOR expression with papillary thyroid cancer clinicopathological features, such as the TNM stage, BRAF V600E mutation, sex distribution, lymph node and distant metastases, and survival prognosis. Understanding specific factors involved in PTC tumorigenesis provides opportunities for targeted therapies. We also reviewed the possible new targeted therapies and the use of mTOR inhibitors in PTC. This topic requires further research with novel techniques to translate the achieved results to clinical application.
Collapse
|
12
|
Takahashi K, Tsuji M, Nakagawasai O, Katsuyama S, Hong L, Miyagawa K, Kurokawa K, Mochida-Saito A, Takeda H, Tadano T. Donepezil prevents olfactory dysfunction and α-synuclein aggregation in the olfactory bulb by enhancing autophagy in zinc sulfate-treated mice. Behav Brain Res 2023; 438:114175. [PMID: 36309244 DOI: 10.1016/j.bbr.2022.114175] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/18/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022]
Abstract
Alzheimer's disease is associated with marked olfactory dysfunction observed in the early stages. Clinical studies reported that acetylcholinesterase inhibitor donepezil (DNP) attenuated this deficit; however, the underlying mechanism remains unclear. Herein, we aimed to examine the effects and underlying mechanisms of DNP on olfactory deficits in zinc sulfate (ZnSO4) nasal-treated mice, which were used as a model of reversible olfactory impairment. We evaluated olfactory function using the buried food finding test and neurogenesis in the subventricular zone (SVZ) using immunohistochemistry. Finally, we measured the expression of doublecortin (DCX), neuronal nuclear antigen (NeuN), olfactory marker protein, tyrosine hydroxylase (TH), tryptophan hydroxylase 2, glutamic acid decarboxylase 67, p-α-synuclein (Ser129), α-synuclein, p-AMPK, p-p70S6 kinase (p70S6K) (Thr389), LC3 Ⅱ/Ⅰ, and p-p62 in the olfactory bulb (OB) by western blotting. On day 7 after treatment, ZnSO4-treated mice exhibited prolonged time to find the buried food, cell proliferation enhancement in the SVZ, increased NeuN, p-α-synuclein (Ser129), and α-synuclein levels, and decreased DCX and TH levels in the OB; except for TH, these changes normalized on day 14 after treatment. Repeated administration of DNP prevented the ZnSO4-induced changes on day 7 after treatment. Moreover, DNP increased p-AMPK and LC3 Ⅱ/Ⅰ, and decreased p-p70S6K and p-p62 (Ser351) levels in the OB, suggesting that DNP enhances autophagy in the OB. These findings indicate that DNP may help prevent olfactory dysfunction by autophagy that reduces α-synuclein aggregation via the AMPK/mTOC1 pathway.
Collapse
Affiliation(s)
- Kohei Takahashi
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Minoru Tsuji
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan.
| | - Osamu Nakagawasai
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan
| | - Soh Katsuyama
- Division of Clinical Pharmacology and Pharmaceutics, Nihon Pharmaceutical University, 10281 Komuro, Kitaadachigun Inamachi, Saitama 362-0806, Japan
| | - Lihua Hong
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Kazuya Miyagawa
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Kazuhiro Kurokawa
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Atsumi Mochida-Saito
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Hiroshi Takeda
- Department of Pharmacology, School of Pharmacy at Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa, Fukuoka 831-8501, Japan
| | - Takeshi Tadano
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan; Department of Environment and Preventive Medicine, Graduate School of Medicine Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan
| |
Collapse
|
13
|
Zhang G, Feng Y, Wang M, Liu X. ATL1 inhibits the proliferation and invasion of trophoblast cells via inhibition of the mTOR signaling pathway. J Biochem Mol Toxicol 2023; 37:e23237. [PMID: 36193555 DOI: 10.1002/jbt.23237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 08/01/2022] [Accepted: 09/22/2022] [Indexed: 01/18/2023]
Abstract
Pre-eclampsia (PE) is a major cause of hypertension in maternal and fetal. Atlastin-1 (ATL1), one regulator of endoplasmic reticulum (ER) morphology, participates in tubular ER formation and protein synthesis. The objective of this study is to investigate the role and molecular mechanism of ATL1 in PE. GEO databases showed that ATL1 was upregulated in PE patients. Our data also found that ATL1 was highly expressed in PE placental tissues. The cell viability, proliferation, migration, and invasion of HTR-8/SVneo cells increased/decreased after the downregulation/upregulation of ATL1. The mTOR pathway is the downstream pathway of ATL1. The levels of p-p70S6K and p-mTOR were increased/decreased after the downregulation/upregulation of ATL1. Moreover, rapamycin, an inhibitor of mTOR pathway, reversed the promotive effect of siATL1 on proliferation, migration, and invasion in HTR-8/SVneo cells. In conclusion, ATL1 inhibits the proliferation and invasion of trophoblast cells via the inhibition of the mTOR signaling pathway in HTR-8/SVneo cells.
Collapse
Affiliation(s)
- Guanli Zhang
- Department of Obstetrics, Yantaishan Hospital, Yantai, Shandong, China
| | - Yan Feng
- Department of Obstetrics, Liaocheng Dongchangfu Maternal and Child Health Hospital, Liaocheng, Shandong, China
| | - Min Wang
- Department of Obstetrics, Liaocheng Dongchangfu Maternal and Child Health Hospital, Liaocheng, Shandong, China
| | - Xin Liu
- Department of Obstetrics, Liaocheng Dongchangfu Maternal and Child Health Hospital, Liaocheng, Shandong, China
| |
Collapse
|
14
|
Yang CC, Masai H. Claspin is Required for Growth Recovery from Serum Starvation through Regulating the PI3K-PDK1-mTOR Pathway in Mammalian Cells. Mol Cell Biol 2023; 43:1-21. [PMID: 36720467 PMCID: PMC9936878 DOI: 10.1080/10985549.2022.2160598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Claspin plays multiple important roles in regulation of DNA replication as a mediator for the cellular response to replication stress, an integral replication fork factor that facilitates replication fork progression and a factor that promotes initiation by recruiting Cdc7 kinase. Here, we report a novel role of Claspin in growth recovery from serum starvation, which requires the activation of PI3 kinase (PI3K)-PDK1-Akt-mTOR pathways. In the absence of Claspin, cells do not proceed into S phase and eventually die partially in a ROS- and p53-dependent manner. Claspin directly interacts with PI3K and mTOR, and is required for activation of PI3K-PDK1-mTOR and for that of mTOR downstream factors, p70S6K and 4EBP1, but not for p38 MAPK cascade during the recovery from serum starvation. PDK1 physically interacts with Claspin, notably with CKBD, in a manner dependent on phosphorylation of the latter protein, and is required for interaction of mTOR with Claspin. Thus, Claspin plays a novel role as a key regulator for nutrition-induced proliferation/survival signaling by activating the mTOR pathway. The results also suggest a possibility that Claspin may serve as a common mediator that receives signals from different PI3K-related kinases and transmit them to specific downstream kinases.
Collapse
Affiliation(s)
- Chi-Chun Yang
- Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hisao Masai
- Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
15
|
Plangger A, Rath B, Stickler S, Hochmair M, Lang C, Weigl L, Funovics M, Hamilton G. Cytotoxicity of combinations of the pan-KRAS SOS1 inhibitor BAY-293 against pancreatic cancer cell lines. Discov Oncol 2022; 13:84. [PMID: 36048281 PMCID: PMC9437170 DOI: 10.1007/s12672-022-00550-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/26/2022] [Indexed: 11/27/2022] Open
Abstract
KRAS is mutated in approximately 25% of cancer patients and first KRAS G12C-specific inhibitors showed promising responses. Pancreatic cancer has the highest frequency of KRAS mutations but the prevailing KRAS G12D mutation is difficult to target. Inhibition of the GTP exchange factor (GEF) SOS1-KRAS interaction impairs oncogenic signaling independently of the specific KRAS mutations. In general, cell lines exhibiting KRAS mutations show specific alterations in respect to glucose utilization, signal transduction and stress survival. The aim of this investigation was to check the putative synergy of the SOS1 inhibitor BAY-293 with modulators targeting specific vulnerabilities of KRAS-mutated cell lines in vitro. The cytotoxicity of BAY-293 combinations was tested against MIA PaCa-2 (G12C), AsPC1 (G12D) and BxPC3 (KRAS wildtype) cell lines using MTT tests and calculation of the combination indices (CI) according to the Chou-Talalay method. The results show that BAY-293 synergizes with modulators of glucose utilization, inhibitors of the downstream MAPK pathway and several chemotherapeutics in dependence of the specific KRAS status of the cell lines. In particular, divergent responses for BAY-293 combinations between pancreatic and NSCLC cell lines were observed for linsitinib, superior inhibitory effects of trametinib and PD98059 in NSCLC, and lack of activity with doxorubicin in case of the pancreatic cell lines. Phosphoproteome analysis revealed inhibition of distinct signaling pathways by BAY-293 for MIA PaCa-2 on the one hand and for Aspc1 and BH1362 on the other hand. In conclusion, BAY-293 exhibits synergy with drugs in dependence of the tumor type and specific KRAS mutation.
Collapse
Affiliation(s)
- Adelina Plangger
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Sandra Stickler
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Maximilian Hochmair
- Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Klinik Floridsdorf, Vienna, Austria
| | - Clemens Lang
- Department of Trauma Surgery, Sozialmedizinisches Zentrum Ost, Donauspital, Vienna, Austria
| | - Lukas Weigl
- Division of Special Anesthesia and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Martin Funovics
- Department of Cardiovascular and Interventional Radiology, Medical University of Vienna, Vienna, Austria
| | - Gerhard Hamilton
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
16
|
Castro‐Guarda M, Arancibia Y, Chipón C, Matamala C, Oyarzo P, Vargas G, Reyes A, Salas M, Morera FJ, Zambrano A. Metabolic changes induced by DNA damage in Ramos cells: exploring the role of mTORC1 complex. FEBS Open Bio 2022; 12:1509-1522. [PMID: 35538662 PMCID: PMC9340868 DOI: 10.1002/2211-5463.13436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/26/2022] [Accepted: 05/09/2022] [Indexed: 11/09/2022] Open
Abstract
DNA damage induces the activation of many different signals associated with repair or cell death, but it is also connected with physiological events, such as adult neurogenesis and B-cell differentiation. DNA damage induces different signaling pathways, some of them linked to important metabolic changes. The mTORC1 pathway has a central role in the regulation of growth processes and cell division in response to environmental changes and also controls protein synthesis, lipid biogenesis, nucleotide synthesis, and expression of glycolytic genes. Here, we report that double-strand breaks induced with etoposide affect the expression of genes encoding different enzymes associated with specific metabolic pathways in Ramos cells. We also analyzed the role of mTOR signaling, demonstrating that double-strand breaks induce downregulation of mTOR signaling. Specific inhibition of mTORC1 using rapamycin also induced changes in the expression of metabolic genes. Finally, we demonstrated that DNA damage and rapamycin can regulate glucose uptake. In summary, our findings show that etoposide and rapamycin affect the expression of metabolic genes as well as apoptotic and proliferation markers in Ramos cells, increasing our understanding of cancer metabolism.
Collapse
Affiliation(s)
- Marcos Castro‐Guarda
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Yennyfer Arancibia
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Carina Chipón
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Christofer Matamala
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Paola Oyarzo
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Gabriela Vargas
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Alejandro Reyes
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
- Universidad Austral de ChileCoyhaiqueChile
| | - Mónica Salas
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Francisco J. Morera
- Facultad de Ciencias VeterinariasInstituto de Farmacología y MorfofisiologíaUniversidad Austral de ChileValdiviaChile
| | - Angara Zambrano
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
- Center for Interdisciplinary Studies on the Nervous System (CISNe)Universidad Austral de ChileValdiviaChile
| |
Collapse
|
17
|
Manetti AC, Maiese A, Baronti A, Mezzetti E, Frati P, Fineschi V, Turillazzi E. MiRNAs as New Tools in Lesion Vitality Evaluation: A Systematic Review and Their Forensic Applications. Biomedicines 2021; 9:1731. [PMID: 34829960 PMCID: PMC8615694 DOI: 10.3390/biomedicines9111731] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022] Open
Abstract
Wound vitality demonstration is one of the most challenging fields in forensic pathology. In recent years, researchers focused on the application of histological and immunohistochemical staining in this sphere of study. It is based on the detection of inflammation, red cell infiltration, and tissue alterations at the histological examination, all of which are supposedly present in antemortem rather than post-mortem wounds. Nevertheless, some doubts about the reliability of those markers have arisen. Furthermore, the lack of a standardized protocol and the operator dependency of this approach make the proper interpretation of its results difficult. Moreover, a differential miRNAs expression has been demonstrated in antemortem and post-mortem wounds. Herein, a systematic review concerning the current knowledge about the use of miRNAs in lesion vitality evaluation is carried out, to encourage researchers to deepen this peculiar study area. A compendium about the potential miRNAs that may be further investigated as vitality markers is also provided. The aim is to collect all available data about this topic to direct further studies on this field and highlight the future applications of miRNAs in forensic pathology. We found 20 articles and a total of 51 miRNAs that are involved in inflammation and wound healing. Further studies are certainly needed to deepen the role of miRNAs in inflammatory processes in lesioned skin and to evaluate their reliability in distinguishing between antemortem and post-mortem lesions.
Collapse
Affiliation(s)
- Alice Chiara Manetti
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Section of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (A.C.M.); (A.M.); (A.B.); (E.M.); (E.T.)
| | - Aniello Maiese
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Section of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (A.C.M.); (A.M.); (A.B.); (E.M.); (E.T.)
| | - Arianna Baronti
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Section of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (A.C.M.); (A.M.); (A.B.); (E.M.); (E.T.)
| | - Eleonora Mezzetti
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Section of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (A.C.M.); (A.M.); (A.B.); (E.M.); (E.T.)
| | - Paola Frati
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, 00186 Rome, Italy;
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, 00186 Rome, Italy;
| | - Emanuela Turillazzi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Section of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (A.C.M.); (A.M.); (A.B.); (E.M.); (E.T.)
| |
Collapse
|
18
|
O'Reilly CL, Uranga S, Fluckey JD. Culprits or consequences: Understanding the metabolic dysregulation of muscle in diabetes. World J Biol Chem 2021; 12:70-86. [PMID: 34630911 PMCID: PMC8473417 DOI: 10.4331/wjbc.v12.i5.70] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/21/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
The prevalence of type 2 diabetes (T2D) continues to rise despite the amount of research dedicated to finding the culprits of this debilitating disease. Skeletal muscle is arguably the most important contributor to glucose disposal making it a clear target in insulin resistance and T2D research. Within skeletal muscle there is a clear link to metabolic dysregulation during the progression of T2D but the determination of culprits vs consequences of the disease has been elusive. Emerging evidence in skeletal muscle implicates influential cross talk between a key anabolic regulatory protein, the mammalian target of rapamycin (mTOR) and its associated complexes (mTORC1 and mTORC2), and the well-described canonical signaling for insulin-stimulated glucose uptake. This new understanding of cellular signaling crosstalk has blurred the lines of what is a culprit and what is a consequence with regard to insulin resistance. Here, we briefly review the most recent understanding of insulin signaling in skeletal muscle, and how anabolic responses favoring anabolism directly impact cellular glucose disposal. This review highlights key cross-over interactions between protein and glucose regulatory pathways and the implications this may have for the design of new therapeutic targets for the control of glucoregulatory function in skeletal muscle.
Collapse
Affiliation(s)
| | - Selina Uranga
- Health and Kinesiology, Texas A&M University, TX 77843, United States
| | - James D Fluckey
- Health and Kinesiology, Texas A&M University, TX 77843, United States
| |
Collapse
|
19
|
Hashikawa-Hobara N, Otsuka A, Okujima C, Hashikawa N. CGRP overexpression does not alter depression-like behavior in mice. PeerJ 2021; 9:e11720. [PMID: 34249519 PMCID: PMC8256807 DOI: 10.7717/peerj.11720] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/13/2021] [Indexed: 12/11/2022] Open
Abstract
Background The calcitonin gene-related peptide (CGRP) is a neuropeptide that is released from capsaicin-sensitive nerves as a potent vasodilator involved in nociceptive transmission. While CGRP has been rigorously studied for its role in migraines owing to its vasodilation and inflammation activities, the effects of CGRP overexpression on depressive-like behaviors remain insufficiently understood. Methods In the present study, we performed a battery of behavioral tests, including the social interaction test, open field test, and sucrose preference test, to evaluate social defeat stress using male C57BL6J or CGRP overexpression in transgenic (Tg) mice (CGRP Tg). We performed mRNA and protein analyses on the brain-derived neurotrophic factor (BDNF), phosphorylated Akt, mTOR, and p70S6K in the hippocampi. Results CGRP Tg mice showed increased levels of Bdnf mRNAs, low locomotor activity, and no deficits in social interaction, which indicate that CGRP Tg mice exhibit stress resistance and not depression. However, the open field test significantly decreased after 15-day social defeat stress exposure. We also evaluated depressive-like behavior using the sucrose preference and social interaction tests. Our data indicate that defeated CGRP Tg mice exhibited a depressive-like phenotype, which was inferred from increased social avoidance and reduced sucrose preference compared with non-defeated controls. Although stress exposure did not change the BDNF levels in CGRP Tg mice, it significantly decreased the expression levels of p-Akt, p-mTOR and p-p70S6K in the mice hippocampi. We conclude that CGRP-overexpressing Tg mice have normal sensitivity to stress and down-regulated hippocampal Akt/mTOR/p70S6K pathways.
Collapse
Affiliation(s)
| | - Ami Otsuka
- Department of Life Science, Okayama University of Science, Okayama, Japan
| | - Chihiro Okujima
- Department of Life Science, Okayama University of Science, Okayama, Japan
| | - Naoya Hashikawa
- Department of Life Science, Okayama University of Science, Okayama, Japan
| |
Collapse
|
20
|
Allosteric Modulation of GSK-3β as a New Therapeutic Approach in Limb Girdle Muscular Dystrophy R1 Calpain 3-Related. Int J Mol Sci 2021; 22:ijms22147367. [PMID: 34298987 PMCID: PMC8308041 DOI: 10.3390/ijms22147367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022] Open
Abstract
Limb-girdle muscular dystrophy R1 calpain 3-related (LGMDR1) is an autosomal recessive muscular dystrophy produced by mutations in the CAPN3 gene. It is a rare disease and there is no cure or treatment for the disease while the pathophysiological mechanism by which the absence of calpain 3 provokes the dystrophy in muscles is not clear. However, key proteins implicated in Wnt and mTOR signaling pathways, which regulate muscle homeostasis, showed a considerable reduction in their expression and in their phosphorylation in LGMDR1 patients' muscles. Finally, the administration of tideglusib and VP0.7, ATP non-competitive inhibitors of glycogen synthase kinase 3β (GSK-3β), restore the expression and phosphorylation of these proteins in LGMDR1 cells, opening the possibility of their use as therapeutic options.
Collapse
|
21
|
Overexpression of Human Syndecan-1 Protects against the Diethylnitrosamine-Induced Hepatocarcinogenesis in Mice. Cancers (Basel) 2021; 13:cancers13071548. [PMID: 33801718 PMCID: PMC8037268 DOI: 10.3390/cancers13071548] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Syndecan-1 is a Janus-faced proteoglycan: depending on the type of cancer, it can promote or inhibit the development of tumors. Our previous in vitro experiments revealed that transfection of human syndecan-1 (hSDC1) into hepatoma cells, initiating hepatocyte-like differentiation. To further confirm the antitumor action of hSDC1 in the context of liver carcinogenesis, mice transgenic for albumin promoter-driven hSDC1 were created with exclusive expression of hSDC1 in the liver. Indeed, hSDC1 interfered with the development of liver cancer in diethylnitrosamine (DEN)-induced hepatocarcinogenesis experiments. The mechanism was found to be related to lipid metabolism that plays an important role in the induction of nonalcoholic liver cirrhosis. Nonalcoholic fatty liver disease is known to promote the development of cancer; therefore, the oncoprotective effect of hSDC1 may be mediated by a beneficial modulation of lipid metabolism. Abstract Although syndecan-1 (SDC1) is known to be dysregulated in various cancer types, its implication in tumorigenesis is poorly understood. Its effect may be detrimental or protective depending on the type of cancer. Our previous data suggest that SDC1 is protective against hepatocarcinogenesis. To further verify this notion, human SDC1 transgenic (hSDC1+/+) mice were generated that expressed hSDC1 specifically in the liver under the control of the albumin promoter. Hepatocarcinogenesis was induced by a single dose of diethylnitrosamine (DEN) at an age of 15 days after birth, which resulted in tumors without cirrhosis in wild-type and hSDC1+/+ mice. At the experimental endpoint, livers were examined macroscopically and histologically, as well as by immunohistochemistry, Western blot, receptor tyrosine kinase array, phosphoprotein array, and proteomic analysis. Liver-specific overexpression of hSDC1 resulted in an approximately six month delay in tumor formation via the promotion of SDC1 shedding, downregulation of lipid metabolism, inhibition of the mTOR and the β-catenin pathways, and activation of the Foxo1 and p53 transcription factors that lead to the upregulation of the cell cycle inhibitors p21 and p27. Furthermore, both of them are implicated in the regulation of intermediary metabolism. Proteomic analysis showed enhanced lipid metabolism, activation of motor proteins, and loss of mitochondrial electron transport proteins as promoters of cancer in wild-type tumors, inhibited in the hSDC1+/+ livers. These complex mechanisms mimic the characteristics of nonalcoholic steatohepatitis (NASH) induced human liver cancer successfully delayed by syndecan-1.
Collapse
|
22
|
Vanderplow AM, Eagle AL, Kermath BA, Bjornson KJ, Robison AJ, Cahill ME. Akt-mTOR hypoactivity in bipolar disorder gives rise to cognitive impairments associated with altered neuronal structure and function. Neuron 2021; 109:1479-1496.e6. [PMID: 33765445 PMCID: PMC8105282 DOI: 10.1016/j.neuron.2021.03.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/20/2021] [Accepted: 03/04/2021] [Indexed: 12/22/2022]
Abstract
The Akt family of kinases exerts many of its cellular effects via the activation of the mammalian target of rapamycin (mTOR) kinase through a series of intermediary proteins. Multiple lines of evidence have identified Akt-family kinases as candidate schizophrenia and bipolar disorder genes. Although dysfunction of the prefrontal cortex (PFC) is a key feature of both schizophrenia and bipolar disorder, no studies have comprehensively assessed potential alterations in Akt-mTOR pathway activity in the PFC of either disorder. Here, we examined the activity and expression profile of key proteins in the Akt-mTOR pathway in bipolar disorder and schizophrenia homogenates from two different PFC subregions. Our findings identify reduced Akt-mTOR PFC signaling in a subset of bipolar disorder subjects. Using a reverse-translational approach, we demonstrated that Akt hypofunction in the PFC is sufficient to give rise to key cognitive phenotypes that are paralleled by alterations in synaptic connectivity and function.
Collapse
Affiliation(s)
- Amanda M Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Andrew L Eagle
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Bailey A Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kathryn J Bjornson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Michael E Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
23
|
Sajan MP, Hansen BC, Acevedo‐Duncan M, Kindy MS, Cooper DR, Farese RV. Roles of hepatic atypical protein kinase C hyperactivity and hyperinsulinemia in insulin-resistant forms of obesity and type 2 diabetes mellitus. MedComm (Beijing) 2021; 2:3-16. [PMID: 34766133 PMCID: PMC8491214 DOI: 10.1002/mco2.54] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/21/2020] [Accepted: 12/28/2020] [Indexed: 11/10/2022] Open
Abstract
Diet-induced obesity, the metabolic syndrome, type 2 diabetes (DIO/MetS/T2DM), and their adverse sequelae have reached pandemic levels. In mice, DIO/MetS/T2DM initiation involves diet-dependent increases in lipids that activate hepatic atypical PKC (aPKC) and thereby increase lipogenic enzymes and proinflammatory cytokines. These or other hepatic aberrations, via adverse liver-to-muscle cross talk, rapidly impair postreceptor insulin signaling to glucose transport in muscle. The ensuing hyperinsulinemia further activates hepatic aPKC, which first blocks the ability of Akt to suppress gluconeogenic enzyme expression, and later impairs Akt activation, further increasing hepatic glucose production. Recent findings suggest that hepatic aPKC also increases a proteolytic enzyme that degrades insulin receptors. Fortunately, all hepatic aberrations and muscle impairments are prevented/reversed by inhibition or deficiency of hepatic aPKC. But, in the absence of treatment, hyperinsulinemia induces adverse events, some by using "spare receptors" to bypass receptor defects. Thus, in brain, hyperinsulinemia increases Aβ-plaque precursors and Alzheimer risk; in kidney, hyperinsulinemia activates the renin-angiotensin-adrenal axis, thus increasing vasoconstriction, sodium retention, and cardiovascular risk; and in liver, hyperinsulinemia increases lipogenesis, obesity, hepatosteatosis, hyperlipidemia, and cardiovascular risk. In summary, increases in hepatic aPKC are critically required for development of DIO/MetS/T2DM and its adverse sequelae, and therapeutic approaches that limit hepatic aPKC may be particularly effective.
Collapse
Affiliation(s)
- Mini P. Sajan
- Department of Internal MedicineUniversity of South Florida College of MedicineTampaFloridaUSA
- Research ServiceJames AHaley Veterans Administration Medical CenterTampaFloridaUSA
| | - Barbara C. Hansen
- Department of Internal MedicineUniversity of South Florida College of MedicineTampaFloridaUSA
| | - Mildred Acevedo‐Duncan
- Department of ChemistryCollege of Arts and SciencesUniversity of South FloridaTampaFloridaUSA
| | - Mark S. Kindy
- Research ServiceJames AHaley Veterans Administration Medical CenterTampaFloridaUSA
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of South FloridaTampaFloridaUSA
| | - Denise R. Cooper
- Research ServiceJames AHaley Veterans Administration Medical CenterTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Robert V. Farese
- Department of Internal MedicineUniversity of South Florida College of MedicineTampaFloridaUSA
- Research ServiceJames AHaley Veterans Administration Medical CenterTampaFloridaUSA
| |
Collapse
|
24
|
Chen TC, Kuo T, Dandan M, Lee RA, Chang M, Villivalam SD, Liao SC, Costello D, Shankaran M, Mohammed H, Kang S, Hellerstein MK, Wang JC. The role of striated muscle Pik3r1 in glucose and protein metabolism following chronic glucocorticoid exposure. J Biol Chem 2021; 296:100395. [PMID: 33567340 PMCID: PMC8010618 DOI: 10.1016/j.jbc.2021.100395] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 11/03/2022] Open
Abstract
Chronic glucocorticoid exposure causes insulin resistance and muscle atrophy in skeletal muscle. We previously identified phosphoinositide-3-kinase regulatory subunit 1 (Pik3r1) as a primary target gene of skeletal muscle glucocorticoid receptors involved in the glucocorticoid-mediated suppression of insulin action. However, the in vivo functions of Pik3r1 remain unclear. Here, we generated striated muscle-specific Pik3r1 knockout (MKO) mice and treated them with a dexamethasone (DEX), a synthetic glucocorticoid. Treating wildtype (WT) mice with DEX attenuated insulin activated Akt activity in liver, epididymal white adipose tissue, and gastrocnemius (GA) muscle. This DEX effect was diminished in GA muscle of MKO mice, therefore, resulting in improved glucose and insulin tolerance in DEX-treated MKO mice. Stable isotope labeling techniques revealed that in WT mice, DEX treatment decreased protein fractional synthesis rates in GA muscle. Furthermore, histology showed that in WT mice, DEX treatment reduced GA myotube diameters. In MKO mice, myotube diameters were smaller than in WT mice, and there were more fast oxidative fibers. Importantly, DEX failed to further reduce myotube diameters. Pik3r1 knockout also decreased basal protein synthesis rate (likely caused by lower 4E-BP1 phosphorylation at Thr37/Thr46) and curbed the ability of DEX to attenuate protein synthesis rate. Finally, the ability of DEX to inhibit eIF2α phosphorylation and insulin-induced 4E-BP1 phosphorylation was reduced in MKO mice. Taken together, these results demonstrate the role of Pik3r1 in glucocorticoid-mediated effects on glucose and protein metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Tzu-Chieh Chen
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Taiyi Kuo
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Mohamad Dandan
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Rebecca A Lee
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Maggie Chang
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Sneha D Villivalam
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Szu-Chi Liao
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Damian Costello
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Mahalakshmi Shankaran
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Hussein Mohammed
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Sona Kang
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Marc K Hellerstein
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Jen-Chywan Wang
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA.
| |
Collapse
|
25
|
Yin S, Liu L, Gan W. The Roles of Post-Translational Modifications on mTOR Signaling. Int J Mol Sci 2021; 22:ijms22041784. [PMID: 33670113 PMCID: PMC7916890 DOI: 10.3390/ijms22041784] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) is a master regulator of cell growth, proliferation, and metabolism by integrating various environmental inputs including growth factors, nutrients, and energy, among others. mTOR signaling has been demonstrated to control almost all fundamental cellular processes, such as nucleotide, protein and lipid synthesis, autophagy, and apoptosis. Over the past fifteen years, mapping the network of the mTOR pathway has dramatically advanced our understanding of its upstream and downstream signaling. Dysregulation of the mTOR pathway is frequently associated with a variety of human diseases, such as cancers, metabolic diseases, and cardiovascular and neurodegenerative disorders. Besides genetic alterations, aberrancies in post-translational modifications (PTMs) of the mTOR components are the major causes of the aberrant mTOR signaling in a number of pathologies. In this review, we summarize current understanding of PTMs-mediated regulation of mTOR signaling, and also update the progress on targeting the mTOR pathway and PTM-related enzymes for treatment of human diseases.
Collapse
|
26
|
Sawant OB, Meng C, Wu G, Washburn SE. Prenatal alcohol exposure and maternal glutamine supplementation alter the mTOR signaling pathway in ovine fetal cerebellum and skeletal muscle. Alcohol 2020; 89:93-102. [PMID: 32777475 DOI: 10.1016/j.alcohol.2020.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 01/25/2023]
Abstract
Prenatal alcohol exposure causes fetal neurodevelopmental damage and growth restriction. Among regions of the brain, the cerebellum is the most vulnerable to developmental alcohol exposure. Despite vast research in the field, there is still a need to identify specific mechanisms by which alcohol causes this damage in order to design effective therapeutic interventions. The mammalian target of rapamycin (mTOR) is known to be associated with axonal regeneration, dendritic arborization, synaptic plasticity, cellular growth, autophagy, and many other cellular processes. Glutamine and glutamine-related amino acids play a key role in fetal development and are known to alter the mTOR pathway; recent research has shown that disturbances in their bioavailability and signaling pathways may mediate adverse effects of prenatal alcohol exposure. This study investigated the role of the mTOR signaling pathway in the fetal cerebellum and skeletal muscle after third trimester-equivalent prenatal alcohol exposure and maternal l-glutamine (GLN) supplementation using a sheep model. Fetal cerebella and skeletal muscles were sampled for Western blot analysis of mTOR and its downstream targets S6 kinase and eukaryotic initiation factor 4E-bindin protein (4E-BP1). The expression of cerebellar phosphorylated mTOR relative to the total mTOR was elevated in the alcohol+GLN group compared to the saline and GLN groups. Alcohol exposure increased the ratio of phosphorylated S6K to total S6K in fetal cerebellum, and no significant effect of GLN supplementation was observed. On contrary, maternal GLN supplementation reduced the activation of mTOR and S6K in fetal skeletal muscle, possibly to make GLN and other amino acids available for use by other organs. These findings suggest prenatal alcohol exposure and maternal GLN supplementation during the third trimester-equivalent alter the mTOR signaling cascade, which plays a possible key role in alcohol-induced developmental damage.
Collapse
|
27
|
Hu R, Dong W, Liang Q, Wang F, Ou M, Li Z, Ren Y, Wu X, Liu Y, Wang W. A Litopenaeus vannamei p70S6K gene is involved in the antioxidative and apoptosis under low temperature. FISH & SHELLFISH IMMUNOLOGY 2020; 106:656-665. [PMID: 32858183 DOI: 10.1016/j.fsi.2020.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 06/11/2023]
Abstract
p70S6K is involved in cellular response, such as tumor metastases, the immune response and tissue repair in vertebrates. The role of p70S6K in these physiological processes in crustaceans remains, however, unknown. In this study, the Lvp70S6K was identified, containing a 5' UTR of 294 bp, an ORF of 1494 bp ad a 3' UTR of 211 bp, encoding 497 amino acids with a theoretical molecular weight of 70 kDa and an estimated isoelectric point of (pI) of 5.16. The multiple alignment found that Lvp70S6K was highly homologous with other invertebrates. Lvp70S6K mRNA was detected in all the tested tissues and the Lvp70S6K expression levels was significantly down-regulated and reached the lowest level (0.44-fold, p < 0.01) at 1.5 h after low temperature stress. The subcellular localization of Lvp70S6K could be detected in cytoplasm. ROS production was significantly up-regulation (1.19-fold, p < 0.01), total hemocyte count (THC) was significantly down-regulation (0.22-fold, p < 0.01), apoptosis rate was markedly increased (1.09-fold, p < 0.01), apoptosis-related genes of LvPDCD4 (1.61-fold, p < 0.01) and LvCyt.C (1.23-fold, p < 0.01) were up-regulated, and anti-apoptotic gene of LvBcl-2 (0.69-fold, p < 0.01), LvIAP1 (0.68-fold, p < 0.01) and LvIAP2 (0.45-fold, p < 0.01) were decreased after low temperature stress in hemolymph of Lvp70S6K-silenced shrimp at 1.5 h. Silencing of LvPTEN significantly increased Lvp70S6K, LvPI3K, LvAKT and LvmTOR expression. In summary, these results indicated that Lvp70S6K play a crucial role in oxidative and apoptosis, which was able to negatively regulate by PTEN.
Collapse
Affiliation(s)
- Rui Hu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - WenNa Dong
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - QingJian Liang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - FeiFei Wang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - MuFei Ou
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - ZhongHua Li
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - YingHao Ren
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - XuJian Wu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Yuan Liu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Weina Wang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, College of Life Science, South China Normal University, Guangzhou, 510631, PR China.
| |
Collapse
|
28
|
Mechanistic Target of Rapamycin Signaling Activation Antagonizes Autophagy To Facilitate Zika Virus Replication. J Virol 2020; 94:JVI.01575-20. [PMID: 32878890 DOI: 10.1128/jvi.01575-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Zika virus (ZIKV), a mosquito-transmitted flavivirus, is linked to microcephaly and other neurological defects in neonates and Guillain-Barré syndrome in adults. The molecular mechanisms regulating ZIKV infection and pathogenic outcomes are incompletely understood. Signaling by the mechanistic (mammalian) target of rapamycin (mTOR) kinase is important for cell survival and proliferation, and viruses are known to hijack this pathway for their replication. Here, we show that in human neuronal precursors and glial cells in culture, ZIKV infection activates both mTOR complex 1 (mTORC1) and mTORC2. Inhibition of mTOR kinase by Torin1 or rapamycin results in reduction in ZIKV protein expression and progeny production. Depletion of Raptor, the defining subunit of mTORC1, by small interfering RNA (siRNA) negatively affects ZIKV protein expression and viral replication. Although depletion of Rictor, the unique subunit of mTORC2, or the mTOR kinase itself also inhibits the viral processes, the extent of inhibition is less pronounced. Autophagy is transiently induced early by ZIKV infection, and impairment of autophagosome elongation by the class III phosphatidylinositol 3-kinase (PI3K) inhibitor 3-methyladenine (3-MA) enhances viral protein accumulation and progeny production. mTOR phosphorylates and inactivates ULK1 (S757) at later stages of ZIKV infection, suggesting a link between autophagy inhibition and mTOR activation by ZIKV. Accordingly, inhibition of ULK1 (by MRT68921) or autophagy (by 3-MA) reversed the effects of mTOR inhibition, leading to increased levels of ZIKV protein expression and progeny production. Our results demonstrate that ZIKV replication requires the activation of both mTORC1 and mTORC2, which negatively regulates autophagy to facilitate ZIKV replication.IMPORTANCE The re-emergence of Zika virus (ZIKV) and its association with neurological complications necessitates studies on the molecular mechanisms that regulate ZIKV pathogenesis. The mTOR signaling cascade is tightly regulated and central to normal neuronal development and survival. Disruption of mTOR signaling can result in neurological abnormalities. In the studies reported here, we demonstrate for the first time that ZIKV infection results in activation of both mTORC1 and mTORC2 to promote virus replication. Although autophagy is activated early in infection to counter virus replication, it is subsequently suppressed by mTOR. These results reveal critical roles of mTOR signaling and autophagy in ZIKV infection and point to a possible mechanism underlying ZIKV-induced pathogenesis. Elucidating the role of mTOR signaling in ZIKV infection will provide insights into the mechanisms of ZIKV-induced neurological complications and potential targets for therapeutic approaches.
Collapse
|
29
|
Chen YL, Yen IC, Lin KT, Lai FY, Lee SY. 4-Acetylantrocamol LT3, a New Ubiquinone from Antrodia cinnamomea, Inhibits Hepatocellular Carcinoma HepG2 Cell Growth by Targeting YAP/TAZ, mTOR, and WNT/β-Catenin Signaling. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:1243-1261. [PMID: 32668963 DOI: 10.1142/s0192415x20500615] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
4-acetylantrocamol LT3 (4AALT3), a new ubiquinone from the mycelium of Antrodia cinnamomea (Polyporaceae), has been recently shown to possess anticancer activity. However, the detailed mechanisms of such action remain unclear. In this study, the molecular mechanisms of 4AALT3 on hepatocellular carcinoma cells (HCC) were investigated. Human hepatocellular carcinoma cell line HepG2 cells were treated with concentrations of 4AALT3. Cell viability, colony formation, and the underlying mechanisms were then analyzed by CCK-8, colony formation, qPCR, and Western blotting assays. We found that 4AALT3 significantly decreased cell viability and colony formation in a dose-dependent manner. Accordingly, 4AALT3 significantly decreased protein levels of cyclin B, E1, D1, and D3, thereby facilitating cell cycle arrest. In addition, 4AALT3 significantly suppressed the nuclear localization of Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ), mammalian target of rapamycin (mTOR), and WNT/[Formula: see text]-catenin signaling pathways, all of which are well-known signaling pathways that contribute to the malignant properties of HCC. These effects are associated with activation of 5' AMP-activated protein kinase (AMPK) and autophagy. Our findings indicate that 4AALT3 exerts inhibitory effects on HepG2 cell growth via multiple signaling pathways and may be a potential agent for HCC therapy.
Collapse
Affiliation(s)
- Yen-Lin Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Radiology, Taoyuan Armed Forces General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - I-Chuan Yen
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Kuen-Tze Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Feng-Yi Lai
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Yu Lee
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
30
|
Estrogen Regulation of mTOR Signaling and Mitochondrial Function in Invasive Lobular Carcinoma Cell Lines Requires WNT4. Cancers (Basel) 2020; 12:cancers12102931. [PMID: 33053661 PMCID: PMC7650584 DOI: 10.3390/cancers12102931] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Invasive lobular carcinoma (ILC) is a common but understudied breast cancer subtype. ILC is presumed to be a low-risk disease in part because nearly all ILCs contain the estrogen receptor (ER). However, we previously showed that ER has unique functions in ILC cells, including driving expression of the Wnt ligand WNT4. WNT4 signaling is required for ILC cell proliferation and survival, but the mechanisms and targets of WNT4 signaling in ILC is unknown. We found that WNT4 regulates mTOR signaling via S6 kinase, and controls levels of MCL-1 protein, ultimately regulating mitochondrial function and cellular metabolism. These findings offer new insight into a novel Wnt signaling pathway and identify new targets to inhibit WNT4 signaling as potential treatments against ILC cells. Abstract Invasive lobular carcinoma of the breast (ILC) is strongly estrogen-driven and represents a unique context for estrogen receptor (ER) signaling. In ILC, ER controls the expression of the Wnt ligand WNT4, which is critical for endocrine response and anti-estrogen resistance. However, signaling mediated by WNT4 is cell type- and tissue-specific, and has not been explored in ILC. We utilized reverse phase protein array (RPPA) to characterize ER and WNT4-driven signaling in ILC cells and identified that WNT4 mediates downstream mTOR signaling via phosphorylation of S6 Kinase. Additionally, ER and WNT4 control levels of MCL-1, which is associated with regulation of mitochondrial function. In this context, WNT4 knockdown led to decreased ATP production and increased mitochondrial fragmentation. WNT4 regulation of both mTOR signaling and MCL-1 were also observed in anti-estrogen resistant models of ILC. We identified that high WNT4 expression is associated with similar mTOR pathway activation in ILC and serous ovarian cancer tumors, suggesting that WNT4 signaling is active in multiple tumor types. The identified downstream pathways offer insight into WNT4 signaling and represent potential targets to overcome anti-estrogen resistance for patients with ILC.
Collapse
|
31
|
Johnstone A, Mobley W. Local TrkB signaling: themes in development and neural plasticity. Cell Tissue Res 2020; 382:101-111. [DOI: 10.1007/s00441-020-03278-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/10/2020] [Indexed: 02/08/2023]
|
32
|
Molecular Interplay between Dormant Bone Marrow-Resident Cells (BMRCs) and CTCs in Breast Cancer. Cancers (Basel) 2020; 12:cancers12061626. [PMID: 32575420 PMCID: PMC7352937 DOI: 10.3390/cancers12061626] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/08/2020] [Accepted: 06/12/2020] [Indexed: 12/12/2022] Open
Abstract
Despite widespread knowledge that bone marrow-resident breast cancer cells (BMRCs) affect tumor progression, signaling mechanisms of BMRCs implicated in maintaining long-term dormancy have not been characterized. To overcome these hurdles, we developed a new experimental model of clinical dormancy employing patient-isolated Circulating Tumor Cells (de novo CTCs) and their injection in xenografts with subsequent tumor monitoring and CTC characterization (ex vivo CTCs). We hypothesized that significant distinctions exist between signaling pathways of bone marrow-homing vs metastasis-competent CTCs upon transplantation in xenografts. Comparative transcriptomic analyses of ex vivo vs de novo CTCs identified increased mTOR signaling—a critical pathway frequently dysregulated in breast cancer and implicated in cell survival and dormancy—with contrasting actions by its two complementary arms (mTORC2/mTORC1). Heightened mTORC2 downstream targets augmented quiescent CTCs (Ki67−/RBL2+ cells) in paired breast cancer tissues, along with high mTORC2 activity in solitary BMRCs and tissue-resident CTCs. Further, shRNA mediated the knockdown of RICTOR, an essential component of mTORC2, and augmented Ki67/PCNA biomarker expression and proliferation. Collectively, these findings suggest that the balance between mTORC1 vs mTORC2 signaling regulates CTC-associated mitotic and/or dormancy characteristics.
Collapse
|
33
|
Jamal MH, Nunes ACF, Vaziri ND, Ramchandran R, Bacallao RL, Nauli AM, Nauli SM. Rapamycin treatment correlates changes in primary cilia expression with cell cycle regulation in epithelial cells. Biochem Pharmacol 2020; 178:114056. [PMID: 32470549 DOI: 10.1016/j.bcp.2020.114056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023]
Abstract
Primary cilia are sensory organelles that regulate cell cycle and signaling pathways. In addition to its association with cancer, dysfunction of primary cilia is responsible for the pathogenesis of polycystic kidney disease (PKD) and other ciliopathies. Because the association between cilia formation or length and cell cycle or division is poorly understood, we here evaluated their correlation in this study. Using Spectral Karyotyping (SKY) technique, we showed that PKD and the cancer/tumorigenic epithelial cells PC3, DU145, and NL20-TA were associated with abnormal ploidy. We also showed that PKD and the cancer epithelia were highly proliferative. Importantly, the cancer epithelial cells had a reduction in the presence and/or length of primary cilia relative to the normal kidney (NK) cells. We then used rapamycin to restore the expression and length of primary cilia in these cells. Our subsequent analyses indicated that both the presence and length of primary cilia were inversely correlated with cell proliferation. Collectively, our data suggest that restoring the presence and/or length of primary cilia may serve as a novel approach to inhibit cancer cell proliferation.
Collapse
Affiliation(s)
- Maha H Jamal
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA, USA; Department of Pharmacology, School of Medicine, King Abdulaziz University, Jeddah, KSA, Saudi Arabia
| | - Ane C F Nunes
- Division of Nephrology and Hypertension, Department of Physiology and Biophysics Division of Nephrology and Hypertension, University of California, Irvine, USA
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, Department of Physiology and Biophysics Division of Nephrology and Hypertension, University of California, Irvine, USA
| | - Ramani Ramchandran
- Department of Pediatrics, Developmental Vascular Biology Program, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robert L Bacallao
- Division of Nephrology, Department of Cellular and Integrative Physiology Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andromeda M Nauli
- Department of Pharmaceutical Sciences, College of Pharmacy, Marshall B. Ketchum University, Fullerton, CA, USA
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA, USA; Department of Medicine, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
34
|
Iida K, Naiki T, Naiki-Ito A, Suzuki S, Kato H, Nozaki S, Nagai T, Etani T, Nagayasu Y, Ando R, Kawai N, Yasui T, Takahashi S. Luteolin suppresses bladder cancer growth via regulation of mechanistic target of rapamycin pathway. Cancer Sci 2020; 111:1165-1179. [PMID: 31994822 PMCID: PMC7156788 DOI: 10.1111/cas.14334] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/24/2022] Open
Abstract
Luteolin is a natural flavonoid with strong anti–oxidative properties that is reported to have an anti–cancer effect in several malignancies other than bladder cancer. In this study, we describe the effect of luteolin on a human bladder cancer cell line, T24, in the context of the regulation of p21, thioredoxin‐1 (TRX1) and the mechanistic target of rapamycin (mTOR) pathway. Luteolin inhibited cell survival and induced G2/M cell‐cycle arrest, p21 upregulation and downregulation of phospho(p)‐S6, which is downstream of mTOR signaling. Luteolin also upregulated TRX1 and reduced intracellular reactive oxygen species production. In a subcutaneous xenograft mouse model using the rat bladder cancer cell line, BC31, tumor volumes were significantly decreased in mice orally administered luteolin compared to control. Immunohistochemical analysis revealed that increased p21 and decreased p‐S6 expression were induced in the luteolin treatment group. Moreover, in another in vivo N‐butyl‐N‐(4‐hydroxybutyl) nitrosamine (BBN)‐induced rat bladder cancer model, the oral administration of luteolin led to a trend of decreased bladder tumor dimension and significantly decreased the Ki67‐labeling index and p‐S6 expression. Furthermore, the major findings on the metabolism of luteolin suggest that both plasma and urine luteolin‐3ʹ‐O‐glucuronide concentrations are strongly associated with the inhibition of cell proliferation and mTOR signaling. Moreover, a significant decrease in the squamous differentiation of bladder cancer is attributed to plasma luteolin‐3ʹ‐glucuronide concentration. In conclusion, luteolin, and in particular its metabolized product, may represent another natural product‐derived therapeutic agent that acts against bladder cancer by upregulating p21 and inhibiting mTOR signaling.
Collapse
Affiliation(s)
- Keitaro Iida
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Taku Naiki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Aya Naiki-Ito
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shugo Suzuki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroyuki Kato
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoshi Nozaki
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Nagai
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Toshiki Etani
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuko Nagayasu
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ryosuke Ando
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Noriyasu Kawai
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
35
|
Hankittichai P, Buacheen P, Pitchakarn P, Na Takuathung M, Wikan N, Smith DR, Potikanond S, Nimlamool W. Artocarpus lakoocha Extract Inhibits LPS-Induced Inflammatory Response in RAW 264.7 Macrophage Cells. Int J Mol Sci 2020; 21:ijms21041355. [PMID: 32079307 PMCID: PMC7072914 DOI: 10.3390/ijms21041355] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 02/07/2023] Open
Abstract
Artocarpus lakoocha Roxb. (AL) has been known for its high content of stilbenoids, especially oxyresveratrol. AL has been used in Thai traditional medicine for centuries. However, the role of AL in regulating inflammation has not been elucidated. Here we investigated the molecular mechanisms underlying the anti-inflammation of AL ethanolic extract in RAW 264.7 murine macrophage cell line. The HPLC results revealed that this plant was rich in oxyresveratrol, and AL ethanolic extract exhibited anti-inflammatory properties. In particular, AL extract decreased lipopolysaccharide (LPS)-mediated production and secretion of cytokines and chemokine, including IL-6, TNF-α, and MCP-1. Consistently, the extract inhibited the production of nitric oxide (NO) in the supernatants of LPS-stimulated cells. Data from the immunofluorescence study showed that AL extract suppressed nuclear translocation of nuclear factor-kappa B (NF-κB) upon LPS induction. Results from Western blot analysis further confirmed that AL extract strongly prevented the LPS-induced degradation of IκB which is normally required for the activation of NF-κB. The protein expression of iNOS and COX-2 in response to LPS stimulation was significantly decreased with the presence of AL extract. AL extract was found to play an anti-inflammatory role, in part through inhibiting LPS-induced activation of Akt. The extract had negligible impact on the activation of mitogen-activated protein kinase (MAPK) pathways. Specifically, incubation of cells with the extract for only 3 h demonstrated the rapid action of AL extract on inhibiting the phosphorylation of Akt, but not ERK1/2. Longer exposure (24 h) to AL extract was required to mildly reduce the phosphorylation of ERK1/2, p38, and JNK MAPKs. These results indicate that AL extract manipulates its anti-inflammatory effects mainly through blocking the PI3K/Akt and NF-κB signal transduction pathways. Collectively, we believe that AL could be a potential alternative agent for alleviating excessive inflammation in many inflammation-associated diseases.
Collapse
Affiliation(s)
- Phateep Hankittichai
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.H.); (M.N.T.); (S.P.)
- Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Pensiri Buacheen
- Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand;
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Pornsiri Pitchakarn
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Mingkwan Na Takuathung
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.H.); (M.N.T.); (S.P.)
- Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nitwara Wikan
- Institute of Molecular Biosciences, Mahidol University, Salaya, Nakorn Pathom 73170, Thailand; (N.W.); (D.R.S.)
| | - Duncan R. Smith
- Institute of Molecular Biosciences, Mahidol University, Salaya, Nakorn Pathom 73170, Thailand; (N.W.); (D.R.S.)
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.H.); (M.N.T.); (S.P.)
- Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.H.); (M.N.T.); (S.P.)
- Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: ; Tel./Fax: +66-53-934597
| |
Collapse
|
36
|
Valdez BC, Li Y, Murray D, Liu Y, Nieto Y, Bashir Q, Qazilbash MH, Andersson BS. Panobinostat and venetoclax enhance the cytotoxicity of gemcitabine, busulfan, and melphalan in multiple myeloma cells. Exp Hematol 2020; 81:32-41. [PMID: 31954171 DOI: 10.1016/j.exphem.2020.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/13/2022]
Abstract
Gemcitabine (Gem), busulfan (Bu), and melphalan (Mel) are used for hematopoietic stem cell transplantation. To further improve their efficacy, a preclinical study on their synergism with the histone deacetylase inhibitor panobinostat (Pano) and the BCL2 inhibitor venetoclax/ABT199 was performed. Multiple myeloma cell lines MM.1R and MC/CAR were exposed to ∼IC20 levels of the drugs. Synergistic cytotoxicity was observed in cells exposed to the five-drug combination as indicated by combination indexes <1, supported by ∼86% inhibition of proliferation and ∼84% annexin V positivity in MM.1R and ∼58% inhibition of proliferation and ∼46% annexin V positivity in MC/CAR cells. Activation of the DNA damage response and apoptosis were suggested by a modest increase in the phosphorylation of ATM and its substrates; significant cleavage of PARP1, caspase 3, and heat shock protein 90; DNA fragmentation; mitochondrial membrane depolarization; and reactive oxygen species production. The five-drug combination significantly decreased the levels of PI3K, AKT, mTOR, RAPTOR, P-P70S6K, and eIF2α, with concomitant increases in P-AMPK and its substrate Tuberin/TSC2, suggesting that the mTOR signaling pathway was compromised. Endoplasmic reticulum stress through activation of the unfolded protein response was also observed as suggested by increases in the levels of calnexin, BiP/GRP78, ERO1-Lα, and protein disulfide isomerase, which may relate to venetoclax-mediated inhibition of BCL2 in the endoplasmic reticulum. This is the first report on the effects of a venetoclax-containing regimen on the unfolded protein response. These results provide a rationale to propose a clinical trial on use of Gem + Bu + Mel + Pano + Venetoclax as part of a conditioning regimen for multiple myeloma patients undergoing autologous hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Benigno C Valdez
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Houston, TX.
| | - Yang Li
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - David Murray
- Department of Experimental Oncology, Cross Cancer Institute, Edmonton, AL T6G1Z2, Canada
| | - Yan Liu
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Yago Nieto
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Qaiser Bashir
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Muzaffar H Qazilbash
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Borje S Andersson
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
37
|
Cao R, Li L, Ying Z, Cao Z, Ma Y, Mao X, Li J, Qi X, Zhang Z, Wang X. A small molecule protects mitochondrial integrity by inhibiting mTOR activity. Proc Natl Acad Sci U S A 2019; 116:23332-23338. [PMID: 31653761 PMCID: PMC6859344 DOI: 10.1073/pnas.1911246116] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Apoptosis activation by cytochrome c release from mitochondria to cytosol is a normal cellular response to mitochondrial damage. Using cellular apoptosis assay, we have found small-molecule apoptosis inhibitors that protect cells from mitochondrial damage. Previously, we reported the discovery of a small molecule, Compound A, which blocks dopaminergic neuron death in a rat model of Parkinson's disease through targeting succinate dehydrogenase subunit B (SDHB) of complex II to protect the integrity of the mitochondrial respiratory chain. Here, we report a small molecule, Compound R6, which saves cells from apoptosis via mammalian target of rapamycin (mTOR)-mediated induction of autophagy. Additionally, we show that Compound R6 protects mitochondrial integrity and respiration after induction of the intrinsic apoptosis pathway. Encouragingly, and supporting the potential further application of Compound R6 as a tool for basic and medicinal research, a pharmacokinetics (PK) profiling study showed that Compound R6 is metabolically stable and can pass the blood-brain barrier. Moreover, Compound R6 accumulates in the brain of test animals via intravenous and intraperitoneal administration. Finally, we found that Compound R6 confers significant neuroprotective effects on a rat cerebral ischemia/reperfusion model, demonstrating its potential as a promising drug candidate for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ran Cao
- School of Life Sciences, Peking University, 100871 Beijing, China
- National Institute of Biological Sciences, 102206 Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 102206 Beijing, China
| | - Li Li
- National Institute of Biological Sciences, 102206 Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 102206 Beijing, China
| | - Zhengxin Ying
- National Institute of Biological Sciences, 102206 Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 102206 Beijing, China
| | - Zichen Cao
- National Institute of Biological Sciences, 102206 Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 102206 Beijing, China
| | - Yongfen Ma
- National Institute of Biological Sciences, 102206 Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 102206 Beijing, China
| | - Xiaoxu Mao
- National Institute of Biological Sciences, 102206 Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 102206 Beijing, China
| | - Jiaojiao Li
- National Institute of Biological Sciences, 102206 Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 102206 Beijing, China
| | - Xiangbing Qi
- National Institute of Biological Sciences, 102206 Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 102206 Beijing, China
| | - Zhiyuan Zhang
- National Institute of Biological Sciences, 102206 Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 102206 Beijing, China
| | - Xiaodong Wang
- National Institute of Biological Sciences, 102206 Beijing, China;
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 102206 Beijing, China
| |
Collapse
|
38
|
Maruska KP, Sohn YC, Fernald RD. Mechanistic target of rapamycin (mTOR) implicated in plasticity of the reproductive axis during social status transitions. Gen Comp Endocrinol 2019; 282:113209. [PMID: 31226256 PMCID: PMC6718321 DOI: 10.1016/j.ygcen.2019.113209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 01/04/2023]
Abstract
The highly conserved brain-pituitary-gonadal (BPG) axis controls reproduction in all vertebrates, so analyzing the regulation of this signaling cascade is important for understanding reproductive competence. The protein kinase mechanistic target of rapamycin (mTOR) functions as a conserved regulator of cellular growth and metabolism in all eukaryotes, and also regulates the reproductive axis in mammals. However, whether mTOR might also regulate the BPG axis in non-mammalian vertebrates remains unexplored. We used complementary experimental approaches in an African cichlid fish, Astatotilapia burtoni, to demonstrate that mTOR is involved in regulation of the brain, pituitary, and testes when males rise in rank to social dominance. mTOR or downstream components of its signaling pathway (p-p70S6K) were detected in gonadotropin-releasing hormone (GnRH1) neurons, the pituitary, and testes. Transcript levels of mtor in the pituitary and testes also varied when reproductively-suppressed subordinate males rose in social rank to become dominant reproductively-active males, a transition similar to puberty in mammals. Intracerebroventricular injection of the mTORC1 inhibitor, rapamycin, revealed a role for mTOR in the socially-induced hypertrophy of GnRH1 neurons. Rapamycin treatment also had effects at the pituitary and testes, suggesting involvement of the mTORC1 complex at multiple levels of the reproductive axis. Thus, we show that mTOR regulation of BPG function is conserved to fishes, likely playing important roles in regulating reproduction and fertility across all male vertebrates.
Collapse
Affiliation(s)
- Karen P Maruska
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, United States.
| | - Young Chang Sohn
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Russell D Fernald
- Department of Biology, Stanford University, Stanford, CA 94305, United States
| |
Collapse
|
39
|
Wang Q, Chen E, Cai Y, Zhou Y, Dong S, Zhang X, Wang O, Li Q. Serum deprivation response functions as a tumor suppressor gene in papillary thyroid cancer. Clin Genet 2019; 96:418-428. [PMID: 31334828 DOI: 10.1111/cge.13609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/29/2019] [Accepted: 07/14/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Qing‐Xuan Wang
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - En‐Dong Chen
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Ye‐Feng Cai
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Yi‐Li Zhou
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Si‐Yang Dong
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Xiao‐Hua Zhang
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Ou‐Chen Wang
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Quan Li
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| |
Collapse
|
40
|
TLR4 counteracts BVRA signaling in human leukocytes via differential regulation of AMPK, mTORC1 and mTORC2. Sci Rep 2019; 9:7020. [PMID: 31065010 PMCID: PMC6504875 DOI: 10.1038/s41598-019-43347-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/16/2019] [Indexed: 12/30/2022] Open
Abstract
TLR4 is implicated in diseases associated with chronic low-grade inflammation, yet homeostatic signaling mechanisms that prevent and/or are affected by chronic TLR4 activation are largely uncharacterized. We recently reported that LPS/TLR4 activates in human leukocytes signaling intermediates (SI), abbreviated TLR4-SI, which include mTORC1-specific effectors and targets, and that leukocytes of patients with T2D or after cardiopulmonary bypass (CPB) expressed similar SI. Extending these previous findings, here we show that TLR4-SI expression post-CPB was associated with low serum bilirubin and reduced preoperative expression of biliverdin reductase A (BVRA), the enzyme that converts biliverdin to bilirubin, in patient’s leukocytes. Biliverdin inhibited TLR4 signaling in leukocytes and triggered phosphorylation of mTORC2-specific targets, including Akt, PKCζ, AMPKα-LKB1-TSC1/2, and their association with BVRA. Torin, PP242, and a PKCζ inhibitory peptide, but not rapamycin, prevented these biliverdin-induced responses and TLR4 inhibition. In contrast, LPS/TLR4 triggered decreases in BVRA, AMPKα and PKCζ expression, and an increase in haptoglobin, a heme binding protein, in leukocytes in vivo and in vitro, indicating that activated TLR4 may suppress biliverdin/BVRA signaling. Significantly, compared to non-diabetics, BVRA and PKCζ expression was low and haptoglobin was high in T2D patients leukocytes. Sustained TLR4 activation may deregulate homeostatic anti-inflammatory BVRA/mTORC2 signaling and thereby contribute to chronic inflammatory diseases.
Collapse
|
41
|
Clomiphene citrate induces nuclear translocation of the TFEB transcription factor and triggers apoptosis by enhancing lysosomal membrane permeabilization. Biochem Pharmacol 2019; 162:191-201. [DOI: 10.1016/j.bcp.2018.11.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/20/2018] [Indexed: 01/13/2023]
|
42
|
Gao Q, Hou B, Yang H, Jiang X. Distinct role of 4E-BP1 and S6K1 in regulating autophagy and hepatitis B virus (HBV) replication. Life Sci 2019; 220:1-7. [DOI: 10.1016/j.lfs.2019.01.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/13/2019] [Accepted: 01/25/2019] [Indexed: 02/08/2023]
|
43
|
Kato Y, Yokoyama U, Fujita T, Umemura M, Kubota T, Ishikawa Y. Epac1 deficiency inhibits basic fibroblast growth factor-mediated vascular smooth muscle cell migration. J Physiol Sci 2019; 69:175-184. [PMID: 30084082 PMCID: PMC11117070 DOI: 10.1007/s12576-018-0631-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/24/2018] [Indexed: 01/12/2023]
Abstract
Vascular smooth muscle cell (VSMC) migration and the subsequent intimal thickening play roles in vascular restenosis. We previously reported that an exchange protein activated by cAMP 1 (Epac1) promotes platelet-derived growth factor (PDGF)-induced VSMC migration and intimal thickening. Because basic fibroblast growth factor (bFGF) also plays a pivotal role in restenosis, we examined whether Epac1 was involved in bFGF-mediated VSMC migration. bFGF-induced lamellipodia formation and migration were significantly decreased in VSMCs obtained from Epac1-/- mice compared to those in Epac1+/+-VSMCs. The bFGF-induced phosphorylation of Akt and glycogen synthase kinase 3β (GSK3β), which play a role in bFGF-induced cell migration, was attenuated in Epac1-/--VSMCs. Intimal thickening induced by the insertion of a large wire was attenuated in Epac1-/- mice, and was accompanied by the decreased phosphorylation of GSK3β. These data suggest that Epac1 deficiency attenuates bFGF-induced VSMC migration, possibly via Akt/GSK3β pathways.
Collapse
Affiliation(s)
- Yuko Kato
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
- Department of Immunopathology, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Utako Yokoyama
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan.
| | - Takayuki Fujita
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Masanari Umemura
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Tetsuo Kubota
- Department of Immunopathology, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan.
| |
Collapse
|
44
|
A novel PI3K/mTOR dual inhibitor, CMG002, overcomes the chemoresistance in ovarian cancer. Gynecol Oncol 2019; 153:135-148. [PMID: 30686552 DOI: 10.1016/j.ygyno.2019.01.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Ovarian cancer is the leading cause of gynecologic-related mortality worldwide. Despite successful initial treatment, overall survival rates are very low because tumors develop resistance to chemotherapeutic drugs. The PI3K/mTOR pathway is a key signaling pathway involved in drug resistance of ovarian cancer cells. The aim of this study was to examine the effect of a newly developed PI3K/mTOR dual inhibitor, CMG002, on chemoresistant ovarian cancer cells. METHODS We examined the effects of CMG002, and its synergistic effects when combined with paclitaxel or cisplatin, on cell viability, cell cycle arrest, and apoptosis of PTX-resistant SKpac17 or cisplatin-resistant A2780cis ovarian cancer cells in vitro. Western blot analysis was performed to assess expression of PI3K, p-mTOR, p-Akt, p-S6, Bim, and caspase-3. In vivo studies were carried out in a xenograft mouse model, followed by TUNEL and immunohistochemical staining of excised tumor tissue. RESULTS CMG002 showed marked toxicity against chemoresistant ovarian cancer cells and re-sensitized these cells to chemotherapeutic agents by suppressing cell proliferation and inducing G1 cell cycle arrest and apoptosis. In vivo xenograft studies revealed that treatment with CMG002, either alone or in combination with paclitaxel or cisplatin, led to a marked reduction in tumor growth. CMG002 caused marked suppression of mTOR (Ser2448), Akt (Ser473), Akt (Thr308), and S6 (Ser235/236) phosphorylation, both in vitro and in vivo. CONCLUSION Taken together, CMG002, a very potent PI3K/mTOR dual inhibitor, induced cytotoxicity in chemoresistant ovarian cancer cells, suggesting that this novel inhibitor might be a new therapeutic strategy for chemoresistant ovarian cancer.
Collapse
|
45
|
Pham DC, Chang YC, Lin SR, Fuh YM, Tsai MJ, Weng CF. FAK and S6K1 Inhibitor, Neferine, Dually Induces Autophagy and Apoptosis in Human Neuroblastoma Cells. Molecules 2018; 23:molecules23123110. [PMID: 30486505 PMCID: PMC6321370 DOI: 10.3390/molecules23123110] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 11/24/2018] [Accepted: 11/25/2018] [Indexed: 12/13/2022] Open
Abstract
Human neuroblastoma cancer is the most typical extracranial solid tumor. Yet, new remedial treatment therapies are demanded to overcome its sluggish survival rate. Neferine, isolated from the lotus embryos, inhibits the proliferation of various cancer cells. This study aimed to evaluate the anti-cancer activity of neferine in IMR32 human neuroblastoma cells and to expose the concealable molecular mechanisms. IMR32 cells were treated with different concentrations of neferine, followed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay to assess cell viability. In an effort to determine the molecular mechanisms in neferine-incubated IMR32 cells, cell cycle arrest, cell migration, and focal adhesion kinase (FAK), the 70-kDa ribosomal S6 kinase 1 (S6K1), poly (ADP-ribose) polymerase (PARP), caspase-3, Beclin-1, and microtubule-associated protein 1A/1B-light chain 3 (LC3) protein expressions were investigated. Neferine strongly disrupted the neuroblastoma cell growth via induction of G2/M phase arrest. Furthermore, neferine provoked autophagy and apoptosis in IMR32 cells, confirmed by p-FAK, and p-S6K1 reduction, LC3-II accumulation, Beclin-1 overexpression, and cleaved caspase-3/PARP improvement. Finally, neferine markedly retarded cell migration of neuroblastoma cancer cells. As a result, our findings for the first time showed an explicit anti-cancer effect of neferine in IMR32 cells, suggesting that neferine might be a potential candidate against human neuroblastoma cells to improve clinical outcomes with further in vivo investigation.
Collapse
Affiliation(s)
- Dinh-Chuong Pham
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Yu-Chuan Chang
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| | - Shian-Ren Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| | - Yuh-Ming Fuh
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| | - May-Jywan Tsai
- Neural Regeneration Laboratory, Taipei Veterans General Hospital, Taipei 11260, Taiwan.
| | - Ching-Feng Weng
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| |
Collapse
|
46
|
Natsume T, Yoshihara T, Naito H. Electromyostimulation with blood flow restriction enhances activation of mTOR and MAPK signaling pathways in rat gastrocnemius muscles. Appl Physiol Nutr Metab 2018; 44:637-644. [PMID: 30398900 DOI: 10.1139/apnm-2018-0384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuromuscular electrical stimulation (NMES) combined with blood flow restriction (BFR) induces muscle hypertrophy. However, cellular mechanisms underlying the muscle hypertrophy induced by NMES combined with BFR remain unclear. We tested the hypothesis that NMES combined with BFR would enhance the mechanistic target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK) signaling pathways. Age-matched male Wistar rats (6 months old, n = 7 per group) were assigned randomly to control, BFR alone (BFR), NMES alone (NMES), and NMES combined with BFR (NMES/BFR) groups. NMES induced 25 isometric contractions lasting 8 s with 4-s resting periods between contractions in the gastrocnemius muscle. Four sets in total were performed, with 1-min intervals between sets. A latex cuff was placed on the proximal portion of the hind limb and BFR at 200 mm Hg was conducted in 4 sets (each set 5 min) with 1-min rest intervals between sets. Venous blood was collected from the lateral tail vein to determine pH, H+ concentration, and lactate concentration before and immediately after the treatments. Expression levels of proteins related to muscle hypertrophy were determined by Western blot analysis. The application of NMES/BFR promoted muscle fatigue more than NMES alone. NMES/BFR induced greater changes in accumulation of metabolites and increase in gastrocnemius muscle weight. The phosphorylation of mTOR and MAPK signaling-related proteins was also enhanced following NMES/BFR, compared with other conditions. Thus, NMES enhanced the activation of mTOR and MAPK signaling pathways when combined with BFR.
Collapse
Affiliation(s)
- Toshiharu Natsume
- a Institute of Health and Sports Science & Medicine, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba 270-1695, Japan
| | - Toshinori Yoshihara
- b Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba 270-1695, Japan
| | - Hisashi Naito
- b Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba 270-1695, Japan
| |
Collapse
|
47
|
mTOR Modulates Methamphetamine-Induced Toxicity through Cell Clearing Systems. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6124745. [PMID: 30647813 PMCID: PMC6311854 DOI: 10.1155/2018/6124745] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/31/2018] [Indexed: 11/17/2022]
Abstract
Methamphetamine (METH) is abused worldwide, and it represents a threat for public health. METH exposure induces a variety of detrimental effects. In fact, METH produces a number of oxidative species, which lead to lipid peroxidation, protein misfolding, and nuclear damage. Cell clearing pathways such as ubiquitin-proteasome (UP) and autophagy (ATG) are involved in METH-induced oxidative damage. Although these pathways were traditionally considered to operate as separate metabolic systems, recent studies demonstrate their interconnection at the functional and biochemical level. Very recently, the convergence between UP and ATG was evidenced within a single organelle named autophagoproteasome (APP), which is suppressed by mTOR activation. In the present research study, the occurrence of APP during METH toxicity was analyzed. In fact, coimmunoprecipitation indicates a binding between LC3 and P20S particles, which also recruit p62 and alpha-synuclein. The amount of METH-induced toxicity correlates with APP levels. Specific markers for ATG and UP, such as LC3 and P20S in the cytosol, and within METH-induced vacuoles, were measured at different doses and time intervals following METH administration either alone or combined with mTOR modulators. Western blotting, coimmunoprecipitation, light microscopy, confocal microscopy, plain transmission electron microscopy, and immunogold staining were used to document the effects of mTOR modulation on METH toxicity and the merging of UP with ATG markers within APPs. METH-induced cell death is prevented by mTOR inhibition, while it is worsened by mTOR activation, which correlates with the amount of autophagoproteasomes. The present data, which apply to METH toxicity, are also relevant to provide a novel insight into cell clearing pathways to counteract several kinds of oxidative damage.
Collapse
|
48
|
Pandurangan AK, Ismail S, Esa NM, Munusamy MA. Inositol-6 phosphate inhibits the mTOR pathway and induces autophagy-mediated death in HT-29 colon cancer cells. Arch Med Sci 2018; 14:1281-1288. [PMID: 30393482 PMCID: PMC6209706 DOI: 10.5114/aoms.2018.76935] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 01/12/2017] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Colorectal cancer (CRC) is common, with a worldwide incidence estimated at more than 1 million cases annually. Therefore, the search for agents for CRC treatment is highly warranted. Inositol-6 phosphate (IP6) is enriched in rice bran and possesses many beneficial effects. In the present study the effect of IP6 on autophagy-mediated death by modulating the mTOR pathway in HT-29 colon cancer cells was studied. MATERIAL AND METHODS Autophagy was assessed by acridine orange (AO) staining, transmission electron microscopy, and western blotting to detect LC3-II and Beclin 1. Akt/mTOR signaling protein expression was also analyzed by western blotting. Apoptosis was analyzed by annexin V staining. RESULTS Incubation of cells with IP6 resulted in downregulation of the p-Akt at 3h. Along with that confocal microscopic analysis of p-AKT, IP6 administration resulted that a diminished expression of p-Akt. mTOR pathway regulates autophagy and incubation with IP6 to HT-29 cells showed decreased expression of p-70S6Kinase, 4-EBP-1 in a time-dependent manner. Inositol-6 phosphate (10 μg/ml, 24 and 48 h) induced autophagic vesicles, as confirmed by AO staining and transmission electron microscopy. We also found increased expression of LC3-II and Beclin 1 in a time-dependent manner after incubation with IP6. Furthermore, IP6 induced apoptosis, as revealed by annexin V staining. CONCLUSIONS Our results clearly indicate that IP6 induces autophagy by inhibiting the Akt/mTOR pathway.
Collapse
Affiliation(s)
- Ashok Kumar Pandurangan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Salmiah Ismail
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Norhaizan Mohd Esa
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Murugan A. Munusamy
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
49
|
Sabharwal SS, Rosen DB, Grein J, Tedesco D, Joyce-Shaikh B, Ueda R, Semana M, Bauer M, Bang K, Stevenson C, Cua DJ, Zúñiga LA. GITR Agonism Enhances Cellular Metabolism to Support CD8+ T-cell Proliferation and Effector Cytokine Production in a Mouse Tumor Model. Cancer Immunol Res 2018; 6:1199-1211. [DOI: 10.1158/2326-6066.cir-17-0632] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 05/11/2018] [Accepted: 08/23/2018] [Indexed: 11/16/2022]
|
50
|
Wang R, Bhattacharya R, Ye X, Fan F, Boulbes DR, Ellis LM. Endothelial Cells Promote Colorectal Cancer Cell Survival by Activating the HER3-AKT Pathway in a Paracrine Fashion. Mol Cancer Res 2018; 17:20-29. [PMID: 30131447 DOI: 10.1158/1541-7786.mcr-18-0341] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/11/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
The regulation of colorectal cancer cell survival pathways remains to be elucidated. Previously, it was demonstrated that endothelial cells (EC) from the liver (liver parenchymal ECs or LPEC), the most common site of colorectal cancer metastases, secrete soluble factors in the conditioned medium (CM) that, in turn, increase the cancer stem cell phenotype in colorectal cancer cells. However, the paracrine effects of LPECs on other colorectal cancer cellular functions have not been investigated. Here, results showed that CM from LPECs increased cell growth and chemoresistance by activating AKT in colorectal cancer cells in vitro. Using an unbiased receptor tyrosine kinase array, it was determined that human epidermal growth factor receptor 3 (ERBB3/HER3) was activated by CM from LPECs, and it mediated AKT activation, cell growth, and chemoresistance in colorectal cancer cells. Inhibition of HER3, either by an inhibitor AZD8931 or an antibody MM-121, blocked LPEC-induced HER3-AKT activation and cell survival in colorectal cancer cells. In addition, CM from LPECs increased in vivo tumor growth in a xenograft mouse model. Furthermore, inhibiting HER3 with AZD8931 significantly blocked tumor growth induced by EC CM. These results demonstrated a paracrine role of liver ECs in promoting cell growth and chemoresistance via activating HER3-AKT in colorectal cancer cells. IMPLICATIONS: This study suggested a potential of treating patients with metastatic colorectal cancer with HER3 antibodies/inhibitors that are currently being assessed in clinical trials for various cancer types.
Collapse
Affiliation(s)
- Rui Wang
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Rajat Bhattacharya
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Xiangcang Ye
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Fan Fan
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Delphine R Boulbes
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Lee M Ellis
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas. .,Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|