1
|
Schoepf AM, Gebhart M, Federspiel M, Heidegger I, Puhr M, Hotze M, Kwiatkowski M, Pircher A, Wolf D, Sopper S, Gust R, Salcher S. Eradication of Therapy-Resistant Cancer Stem Cells by Novel Telmisartan Derivatives. J Med Chem 2025; 68:287-306. [PMID: 39693499 PMCID: PMC11726677 DOI: 10.1021/acs.jmedchem.4c01865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
The present structure-activity relationship study investigates the development of novel chemosensitizers targeting therapy-resistant cancer stem cells (CSCs). We used 4'-((2-propyl-1H-benzo[d]imidazole-1-yl)methyl)-[1,1'-biphenyl]-2-carboxylic acid, derived from the angiotensin II type 1 receptor blocker telmisartan, as a lead structure, demonstrating that the biphenyl moiety is essential for chemosensitizing activity. Introducing a methyl carboxylate or carboxamide instead of the COOH-group significantly enhanced this effect, leading to the development of highly potent compounds. These novel, noncytotoxic chemosensitizers effectively target CSCs and overcome drug resistance by interfering with CSC persistence mechanisms─hyperactivated STAT5 signaling and increased drug transporter activity─with demonstrated efficacy in leukemia, ovarian, and prostate cancers. The carboxamide of telmisartan (telmi-amide, 7c) significantly reduced tumor growth in an imatinib-resistant leukemia xenograft model, both as monotherapy and combined with imatinib, showing promising oral bioavailability and tolerability. In summary, telmisartan derivatives act as effective chemosensitizers and offer an innovative strategy for targeting CSCs in various malignant diseases.
Collapse
Affiliation(s)
- Anna M. Schoepf
- CCB
- Centrum for Chemistry and Biomedicine, Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI
- Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Maximilian Gebhart
- CCB
- Centrum for Chemistry and Biomedicine, Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI
- Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Martin Federspiel
- CCB
- Centrum for Chemistry and Biomedicine, Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI
- Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Isabel Heidegger
- Department
of Urology, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Martin Puhr
- Department
of Urology, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Madlen Hotze
- Department
of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Marcel Kwiatkowski
- Department
of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Andreas Pircher
- Department
of Internal Medicine V, Hematology and Oncology, Tyrolean Cancer Research
Institute (TKFI), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Dominik Wolf
- Department
of Internal Medicine V, Hematology and Oncology, Tyrolean Cancer Research
Institute (TKFI), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Sieghart Sopper
- Department
of Internal Medicine V, Hematology and Oncology, Tyrolean Cancer Research
Institute (TKFI), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Ronald Gust
- CCB
- Centrum for Chemistry and Biomedicine, Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI
- Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Stefan Salcher
- Department
of Internal Medicine V, Hematology and Oncology, Tyrolean Cancer Research
Institute (TKFI), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck 6020, Austria
| |
Collapse
|
2
|
Motta JVDO, Gomes DS, Silva LLD, Oliveira MSD, Bastos DSS, Resende MTCS, Alvim JRL, Reis AB, Oliveira LLD, Afzal MBS, Serrão JE. Effects of sublethal concentration of thiamethoxam formulation on the wild stingless bee, Partamona helleri Friese (Hymenoptera: Apidae): Histopathology, oxidative stress and behavioral changes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177626. [PMID: 39566631 DOI: 10.1016/j.scitotenv.2024.177626] [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: 09/04/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Bees are pollinators of native and cultivated plants around the world. However, several factors are contributing to the decrease in their populations in recent years, with emphasis on the increasing use of insecticides in agriculture. Thiamethoxam is a neonicotinoid neurotoxicant, which binds to nicotinic acetylcholine receptors, causing hyperexcitation, paralysis and death of insects. Although thiamethoxam's target is the nervous system, it can affect other organs through ingestion, such as the midgut, affecting non-target insects such as bees. Partamona helleri Friese (Hymenoptera: Apidae) is a stingless bee, pollinator of several native and cultivated botanical families, and can be exposed to sublethal concentrations of thiamethoxam. This study evaluated the side effects of chronic oral exposure to thiamethoxam on the midgut, oxidative stress and behavior of P. helleri workers. The bees were exposed orally, for 7 days, to the approximate sublethal concentration of thiamethoxam found in pollen grains (0.09 ng/g). The results demonstrated changes in the midgut epithelium of workers treated with thiamethoxam, such as cytoplasmic vacuolization, cellular protrusions, increased apocrine transfer, mitochondrial damage, decreased proteins and neutral polysaccharides and the presence of cells undergoing autophagy and apoptosis. Sublethal concentration of thiamethoxam also induced oxidative stress, evidenced by changes in the activities of detoxification enzymes and antioxidant markers. Finally, thiamethoxam affects the bee's behavior, driving the distance covered and walking speed of this insect. The results indicate that the exposure of P. helleri workers to sublethal concentration of thiamethoxam have negative impacts upon midgut morphology and physiology and behavioral traits.
Collapse
Affiliation(s)
| | - Davy Soares Gomes
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | - Laryssa Lemos da Silva
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | - Mateus Soares de Oliveira
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | - Daniel Silva Sena Bastos
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais 36570-900, Brazil
| | | | | | - Aline Beatriz Reis
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | | | | | - José Eduardo Serrão
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| |
Collapse
|
3
|
Faber JG, Asensio JO, Caiment F, van den Beucken T. Knock-down of FOXO3, GATA2, NFE2L2 and AHR promotes doxorubicin-induced cardiotoxicity in human cardiomyocytes. Toxicology 2024; 509:153977. [PMID: 39427782 DOI: 10.1016/j.tox.2024.153977] [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: 09/27/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Recent advances in cancer therapy have substantially increased survival rates among patients, yet the prolonged effect of current treatment regimens with anthracyclines (ACs) often include severe long-term complications, notably in the form of anthracycline-induced cardiotoxicity (AIC). Despite known associations between AC treatment and AIC, a comprehensive understanding of the underlying molecular pathways remains elusive. This gap is highlighted by the scarcity of reliable therapeutic interventions, with dexrazoxane being the sole FDA-approved drug to mitigate AIC risks. This study aims at elucidating the transcriptional response of human cardiomyocytes (hCMs) to AC exposure by analyzing a previously generated RNA-sequencing dataset of cardiac spheroids subjected to clinically relevant doses of ACs. The analysis revealed a robust transcriptional response identified across various time points. We aimed at identifying important transcription factors (TFs) mediating AIC by employing predictive algorithms to highlight key TFs for further experimental validation. Using shRNA constructs, we further assessed the impact of these TFs on hCM response to doxorubicin (DOX) and revealed that these TFs had a notable impact on hCM survival upon DOX exposure. TFs FOXO3, GATA2, AHR and NFE2L2 were further investigated for their role in AIC including cell viability, DOX uptake, DNA damage repair and induction of apoptosis through Cleaved-Caspase 3. Our study demonstrated that eliminating FOXO3 and GATA2 made hCMs more vulnerable to DOX and the lack of GATA2, NFE2L2 and AHR led to significantly higher intracellular levels of DOX. Additionally, FOXO3 played a role in the repair of hCM DNA damage as we observed markedly enhanced levels of CDKN1A. We also noted significant increases in DNA damage through COMET-assays when FOXO3, GATA2, NFE2L2 and AHR were absent. Furthermore, we investigated the clinical relevance by comparing our results with those from a study based on hiPSC-CMs derived from patients with doxorubicin-induced cardiotoxicity, identifying overlapping TFs and their regulatory roles in critical cellular processes like the cell cycle and DNA repair. This approach not only advances the understanding of the molecular mechanisms behind AIC but also opens possible windows for new therapeutic approaches to mitigate the negative side-effects from patient AC treatment.
Collapse
Affiliation(s)
- J G Faber
- Maastricht University, Department of Translational Genomics, Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
| | - J Ochoteco Asensio
- Maastricht University, Department of Translational Genomics, Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
| | - F Caiment
- Maastricht University, Department of Translational Genomics, Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
| | - T van den Beucken
- Maastricht University, Department of Translational Genomics, Research Institute for Oncology and Reproduction, Maastricht, the Netherlands.
| |
Collapse
|
4
|
Morcos CA, Haiba NS, Bassily RW, Abu-Serie MM, El-Yazbi AF, Soliman OA, Khattab SN, Teleb M. Structure optimization and molecular dynamics studies of new tumor-selective s-triazines targeting DNA and MMP-10/13 for halting colorectal and secondary liver cancers. J Enzyme Inhib Med Chem 2024; 39:2423174. [PMID: 39513468 PMCID: PMC11552285 DOI: 10.1080/14756366.2024.2423174] [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: 10/27/2023] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
A series of triazole-tethered triazines bearing pharmacophoric features of DNA-targeting agents and non-hydroxamate MMPs inhibitors were synthesized and screened against HCT-116, Caco-2 cells, and normal colonocytes by MTT assay. 7a and 7g surpassed doxorubicin against HCT-116 cells regarding potency (IC50 = 0.87 and 1.41 nM) and safety (SI = 181.93 and 54.41). 7g was potent against liver cancer (HepG-2; IC50 = 65.08 nM), the main metastatic site of CRC with correlation to MMP-13 expression. Both derivatives induced DNA damage at 2.67 and 1.87 nM, disrupted HCT-116 cell cycle and triggered apoptosis by 33.17% compared to doxorubicin (DNA damage at 0.76 nM and 40.21% apoptosis induction). 7g surpassed NNGH against MMP-10 (IC50 = 0.205 μM) and MMP-13 (IC50 = 0.275 μM) and downregulated HCT-116 VEGF related to CRC progression by 38%. Docking and MDs simulated ligands-receptors binding modes and highlighted SAR. Their ADMET profiles, drug-likeness and possible off-targets were computationally predicted.
Collapse
Affiliation(s)
- Christine A. Morcos
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Nesreen S. Haiba
- Department of Physics and Chemistry, Faculty of Education, Alexandria University, Alexandria, Egypt
| | - Rafik W. Bassily
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Marwa M. Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, Egypt
| | - Amira F. El-Yazbi
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Omar A. Soliman
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Sherine N. Khattab
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Egypt
| |
Collapse
|
5
|
Wang R, Ji L, Yuan S, Liu X, Liang Z, Chen W, Wang B, Hu S, Liu Z, Zeng Z, Song Y, Wu T, Chen B. Microglial forkhead box O3a deficiency attenuates LPS-induced neuro-inflammation and depressive-like behaviour through regulating the expression of peroxisome proliferator-activated receptor-γ. Br J Pharmacol 2024; 181:3908-3925. [PMID: 38881194 DOI: 10.1111/bph.16474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND AND PURPOSE Depression is closely linked with microglial activation and neuro-inflammation. Peroxisome proliferator-activated receptor-γ (PPAR-γ) plays an important role in M2 activation of microglia. Forkhead box (FOX) O3a has been implicated in the regulation of mood-relevant behaviour. However, little is known about the inflammatory mechanisms of in the microglia of the brain. Here, we have investigated the role of microglial FOXO3a/PPAR-γ in the development of depression. EXPERIMENTAL APPROACH The effect of FOXO3a on microglia inflammation was analysed in vitro and in lipopolysaccharide (LPS)-induced depression-like behaviours in vivo. ChIP-seq and Dual-luciferase reporter assays were used to confirm the interaction between FOXO3a and PPAR-γ. Behavioural changes were measured, while inflammatory cytokines, microglial phenotype and morphological properties were determined by ELISA, qRT-PCR, western blotting and immunostaining. KEY RESULTS Overexpression of FOXO3a significantly attenuated expression of PPAR-γ and enhanced the microglial polarization towards the M1 phenotype, while knockdown of FOXO3a had the opposite effect. FOXO3a binds to the promoters of PPAR-γ and decreases its transcription activity. Importantly, deacetylation and activation of FOXO3a regulate LPS-induced neuro-inflammation by inhibiting the expression of PPAR-γ in microglia cells, supporting the antidepressant potential of histone deacetylase inhibitors. Microglial FOXO3a deficiency in mice alleviated LPS-induced neuro-inflammation and depression-like behaviours but failed to reduce anxiety behaviour, whereas pharmacological inhibition of PPAR-γ by GW9662 restored LPS-induced microglial activation and depressive-like behaviours in microglial FOXO3a-deficient mice. CONCLUSION AND IMPLICATIONS FOXO3a/PPAR-γ axis plays an important role in microglial activation and depression, identifying a new therapeutic avenue for the treatment of major depression.
Collapse
Affiliation(s)
- Rikang Wang
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lianru Ji
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Disease with Mental Disorders); Key Laboratory of Depression Animal Model Based on TCM syndrome, Jiangxi Administration of Traditional Chinese Medicine; Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive Dysfunction, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Shun Yuan
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Disease with Mental Disorders); Key Laboratory of Depression Animal Model Based on TCM syndrome, Jiangxi Administration of Traditional Chinese Medicine; Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive Dysfunction, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xiamin Liu
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Disease with Mental Disorders); Key Laboratory of Depression Animal Model Based on TCM syndrome, Jiangxi Administration of Traditional Chinese Medicine; Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive Dysfunction, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Zhi Liang
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wenjing Chen
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bocheng Wang
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Suifa Hu
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Disease with Mental Disorders); Key Laboratory of Depression Animal Model Based on TCM syndrome, Jiangxi Administration of Traditional Chinese Medicine; Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive Dysfunction, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhiwen Zeng
- Department for Bipolar Disorders, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, China
| | - Yonggui Song
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Disease with Mental Disorders); Key Laboratory of Depression Animal Model Based on TCM syndrome, Jiangxi Administration of Traditional Chinese Medicine; Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive Dysfunction, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Tao Wu
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Baodong Chen
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
6
|
Yu X, Yu Y, Huang X, Jiang Z, Wang Q, Yu X, Song C. Unraveling the causal links and novel molecular classification of Crohn's disease in breast Cancer: a two-sample mendelian randomization and transcriptome analysis with prognostic modeling. BMC Cancer 2024; 24:1134. [PMID: 39261800 PMCID: PMC11389480 DOI: 10.1186/s12885-024-12838-x] [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: 11/21/2023] [Accepted: 08/21/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Crohn's disease (CD), a prominent manifestation of chronic gastrointestinal inflammation, and breast cancer (BC), seemingly disparate in the medical domain, exhibit a shared characteristic. This convergence arises from their involvement in chronic inflammation and immune responses, an aspect that has progressively captivated the attention of investigators but remain controversial. METHODS We used two-sample Mendelian Randomization (MR) and transcriptomics to explore the relationship between CD and BC. MR assessed causality of CD on different BC subtypes and reverse causality of BC on CD. We identified CD-related differentially expressed genes and their prognostic impact on BC, and developed a new molecular BC classification based on these key genes. RESULTS MR revealed a causal link between CD and increased BC risk, especially in estrogen receptor-positive (ER+) patients, but not in ER-negative (ER-) cases. BC showed no causal effect on CD. Transcriptomics pinpointed genes like B4GALNT2 and FGF19 that affected BC prognosis in CD patients. A nomogram based on these genes predicted BC outcomes with high accuracy. Using these genes, a new molecular classification of BC patients was proposed. CONCLUSIONS CD is a risk factor for ER + BC but not for ER- BC. BC does not causally affect CD. Our prognostic model and new BC molecular classifications offer insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Xin Yu
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fu Ma Road, Jinan District, Fuzhou, Fujian Province, 350014, China
| | - Yushuai Yu
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fu Ma Road, Jinan District, Fuzhou, Fujian Province, 350014, China
| | - Xiewei Huang
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fu Ma Road, Jinan District, Fuzhou, Fujian Province, 350014, China
| | - Zirong Jiang
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fu Ma Road, Jinan District, Fuzhou, Fujian Province, 350014, China
| | - Qing Wang
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fu Ma Road, Jinan District, Fuzhou, Fujian Province, 350014, China
| | - Xiaoqin Yu
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fu Ma Road, Jinan District, Fuzhou, Fujian Province, 350014, China
| | - Chuangui Song
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fu Ma Road, Jinan District, Fuzhou, Fujian Province, 350014, China.
| |
Collapse
|
7
|
Wyant GA, Jiang Q, Singh M, Qayyum S, Levrero C, Maron BA, Kaelin WG. Induction of DEPP1 by HIF Mediates Multiple Hallmarks of Ischemic Cardiomyopathy. Circulation 2024; 150:770-786. [PMID: 38881449 PMCID: PMC11361356 DOI: 10.1161/circulationaha.123.066628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 05/22/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND HIF (hypoxia inducible factor) regulates many aspects of cardiac function. We and others previously showed that chronic HIF activation in the heart in mouse models phenocopies multiple features of ischemic cardiomyopathy in humans, including mitochondrial loss, lipid accumulation, and systolic cardiac dysfunction. In some settings, HIF also causes the loss of peroxisomes. How, mechanistically, HIF promotes cardiac dysfunction is an open question. METHODS We used mice lacking cardiac pVHL (von Hippel-Lindau protein) to investigate how chronic HIF activation causes multiple features of ischemic cardiomyopathy, such as autophagy induction and lipid accumulation. We performed immunoblot assays, RNA sequencing, mitochondrial and peroxisomal autophagy flux measurements, and live cell imaging on isolated cardiomyocytes. We used CRISPR-Cas9 gene editing in mice to validate a novel mediator of cardiac dysfunction in the setting of chronic HIF activation. RESULTS We identify a previously unknown pathway by which cardiac HIF activation promotes the loss of mitochondria and peroxisomes. We found that DEPP1 (decidual protein induced by progesterone 1) is induced under hypoxia in a HIF-dependent manner and localizes inside mitochondria. DEPP1 is both necessary and sufficient for hypoxia-induced autophagy and triglyceride accumulation in cardiomyocytes ex vivo. DEPP1 loss increases cardiomyocyte survival in the setting of chronic HIF activation ex vivo, and whole-body Depp1 loss decreases cardiac dysfunction in hearts with chronic HIF activation caused by VHL loss in vivo. CONCLUSIONS Our findings identify DEPP1 as a key component in the cardiac remodeling that occurs with chronic ischemia.
Collapse
Affiliation(s)
- Gregory A. Wyant
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.A.W., Q.J., C.L., W.G.K.)
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA (G.A.W., M.S., S.Q.)
| | - Qinqin Jiang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.A.W., Q.J., C.L., W.G.K.)
| | - Madhu Singh
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA (G.A.W., M.S., S.Q.)
| | - Shariq Qayyum
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA (G.A.W., M.S., S.Q.)
| | - Clara Levrero
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.A.W., Q.J., C.L., W.G.K.)
| | - Bradley A. Maron
- Department of Cardiovascular Medicine (B.A.M.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.A.W., Q.J., C.L., W.G.K.)
- Department of Medicine (W.G.K.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Howard Hughes Medical Institute, Chevy Chase, MD (W.G.K.)
| |
Collapse
|
8
|
Dyrkheeva NS, Zakharenko AL, Malakhova AA, Okorokova LS, Shtokalo DN, Medvedev SP, Tupikin AA, Kabilov MR, Lavrik OI. Transcriptomic analysis of HEK293A cells with a CRISPR/Cas9-mediated TDP1 knockout. Biochim Biophys Acta Gen Subj 2024; 1868:130616. [PMID: 38621596 DOI: 10.1016/j.bbagen.2024.130616] [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: 12/25/2023] [Revised: 03/21/2024] [Accepted: 04/01/2024] [Indexed: 04/17/2024]
Abstract
Tyrosyl-DNA phosphodiesterase 1 (TDP1) is a human DNA repair protein. It is a member of the phospholipase D family based on structural similarity. TDP1 is a key enzyme of the repair of stalled topoisomerase 1 (TOP1)-DNA complexes. Previously, with the CRISPR/Cas9 method, we obtained HEK293A cells with a homozygous knockout of the TDP1 gene and used the TDP1 knockout cells as a cellular model for studying mechanisms of action of an anticancer therapy. In the present work, we hypothesized that the TDP1 knockout would alter the expression of DNA repair-related genes. By transcriptomic analysis, we investigated for the first time the effect of the TDP1 gene knockout on genes' expression changes in the human HEK293A cell line. We obtained original data implying a role of TDP1 in other processes besides the repair of the DNA-TOP1 complex. Differentially expressed gene analysis revealed that TDP1 may participate in cell adhesion and communication, spermatogenesis, mitochondrial function, neurodegeneration, a cytokine response, and the MAPK signaling pathway.
Collapse
Affiliation(s)
- Nadezhda S Dyrkheeva
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of Russian Academy of Sciences (SB RAS), 8 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia.
| | - Alexandra L Zakharenko
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of Russian Academy of Sciences (SB RAS), 8 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia.
| | - Anastasia A Malakhova
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of Russian Academy of Sciences (SB RAS), 8 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia; Federal research center Institute of Cytology and Genetics, SB RAS, 10 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia.
| | | | - Dmitry N Shtokalo
- AcademGene LLC, 6 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia; A.P. Ershov Institute of Informatics Systems, SB RAS, 6 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia.
| | - Sergey P Medvedev
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of Russian Academy of Sciences (SB RAS), 8 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia; Federal research center Institute of Cytology and Genetics, SB RAS, 10 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia.
| | - Alexey A Tupikin
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of Russian Academy of Sciences (SB RAS), 8 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia.
| | - Marsel R Kabilov
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of Russian Academy of Sciences (SB RAS), 8 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia.
| | - Olga I Lavrik
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of Russian Academy of Sciences (SB RAS), 8 Akad. Lavrentyeva Ave., Novosibirsk 630090, Russia.
| |
Collapse
|
9
|
Bi CF, Liu J, Hu XD, Yang LS, Zhang JF. Novel insights into the regulatory role of N6-methyladenosine methylation modified autophagy in sepsis. Aging (Albany NY) 2023; 15:15676-15700. [PMID: 38112620 PMCID: PMC10781468 DOI: 10.18632/aging.205312] [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: 07/16/2023] [Accepted: 10/23/2023] [Indexed: 12/21/2023]
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. It is characterized by high morbidity and mortality and one of the major diseases that seriously hang over global human health. Autophagy is a crucial regulator in the complicated pathophysiological processes of sepsis. The activation of autophagy is known to be of great significance for protecting sepsis induced organ dysfunction. Recent research has demonstrated that N6-methyladenosine (m6A) methylation is a well-known post-transcriptional RNA modification that controls epigenetic and gene expression as well as a number of biological processes in sepsis. In addition, m6A affects the stability, export, splicing and translation of transcripts involved in the autophagic process. Although it has been suggested that m6A methylation regulates the biological metabolic processes of autophagy and is more frequently seen in the progression of sepsis pathogenesis, the underlying molecular mechanisms of m6A-modified autophagy in sepsis have not been thoroughly elucidated. The present article fills this gap by providing an epigenetic review of the processes of m6A-modified autophagy in sepsis and its potential role in the development of novel therapeutics.
Collapse
Affiliation(s)
- Cheng-Fei Bi
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Jia Liu
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Xiao-Dong Hu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Li-Shan Yang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| | - Jun-Fei Zhang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan 750000, Ningxia, China
| |
Collapse
|
10
|
Song J, Duivenvoorde LPM, Grefte S, Kuda O, Martínez-Ramírez F, van der Stelt I, Mastorakou D, van Schothorst EM, Keijer J. Normobaric hypoxia shows enhanced FOXO1 signaling in obese mouse gastrocnemius muscle linked to metabolism and muscle structure and neuromuscular innervation. Pflugers Arch 2023; 475:1265-1281. [PMID: 37656229 PMCID: PMC10567817 DOI: 10.1007/s00424-023-02854-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/02/2023]
Abstract
Skeletal muscle relies on mitochondria for sustainable ATP production, which may be impacted by reduced oxygen availability (hypoxia). Compared with long-term hypoxia, the mechanistic in vivo response to acute hypoxia remains elusive. Therefore, we aimed to provide an integrated description of the Musculus gastrocnemius response to acute hypoxia. Fasted male C57BL/6JOlaHsd mice, fed a 40en% fat diet for six weeks, were exposed to 12% O2 normobaric hypoxia or normoxia (20.9% O2) for six hours (n = 12 per group). Whole-body energy metabolism and the transcriptome response of the M. gastrocnemius were analyzed and confirmed by acylcarnitine determination and Q-PCR. At the whole-body level, six hours of hypoxia reduced energy expenditure, increased blood glucose and tended to decreased the respiratory exchange ratio (RER). Whole-genome transcriptome analysis revealed upregulation of forkhead box-O (FOXO) signalling, including an increased expression of tribbles pseudokinase 3 (Trib3). Trib3 positively correlated with blood glucose levels. Upregulated carnitine palmitoyltransferase 1A negatively correlated with the RER, but the significantly increased in tissue C14-1, C16-0 and C18-1 acylcarnitines supported that β-oxidation was not regulated. The hypoxia-induced FOXO activation could also be connected to altered gene expression related to fiber-type switching, extracellular matrix remodeling, muscle differentiation and neuromuscular junction denervation. Our results suggest that a six-hour exposure of obese mice to 12% O2 normobaric hypoxia impacts M. gastrocnemius via FOXO1, initiating alterations that may contribute to muscle remodeling of which denervation is novel and warrants further investigation. The findings support an early role of hypoxia in tissue alterations in hypoxia-associated conditions such as aging and obesity.
Collapse
Affiliation(s)
- Jingyi Song
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | | | - Sander Grefte
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Ondrej Kuda
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology, Czech Academy of Sciences, 14220, Prague 4, Czech Republic
| | - Felipe Martínez-Ramírez
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology, Czech Academy of Sciences, 14220, Prague 4, Czech Republic
| | - Inge van der Stelt
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Dimitra Mastorakou
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology, Czech Academy of Sciences, 14220, Prague 4, Czech Republic
| | | | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
11
|
Sun L, Liu J, Bao D, Hu C, Zhao Y, Chen S. Progress in the study of FOXO3a interacting with microRNA to regulate tumourigenesis development. Front Oncol 2023; 13:1293968. [PMID: 37965449 PMCID: PMC10641706 DOI: 10.3389/fonc.2023.1293968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
FOXO3a is a protein of the forkhead box family that inhibits tumour cell growth. One of the regulatory modes affecting the role of FOXO3a is microRNA targeting and degradation of its mRNA expression, and conversely, aberrant expression of FOXO3a as a transcription factor also influences microRNA levels. We summarized the results of the regulatory interactions of twenty-five microRNAs with FOXO3a in five types of malignant tumours and found that dual microRNAs synergize with FOXO3a to inhibit breast cancer cell growth including two groups; Three individual microRNAs collaborated with FOXO3a to restrain hepatocellular carcinoma progression; Twelve individual microRNAs antagonized FOXO3a to promote the development of a single tumour cell, respectively; and five microRNAs antagonized FOXO3a to contribute to the progression of more than two types of tumours. The above findings demonstrated the tumour suppressor effect of FOXO3a, but another result revealed that miR-485-5p and miR-498 inhibited the growth of hepatocellular carcinoma cells by antagonizing FOXO3a when acting in combination with other long-stranded non-coding RNAs, respectively, suggesting that FOXO3a at this moment plays the function of promoting the tumour progression. The PI3K/AKT, Snail, VEGF-NRP1, and Wnt/β-catenin signalling pathways perform crucial roles in the above process. It is anticipated that the above studies will assist in understanding the effects of FOXO3a-MicroRNA interactions in cancer genesis and development, and provide new perspectives in the treatment of malignant tumours.
Collapse
Affiliation(s)
- Liying Sun
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
- College of Medical Technology, Beihua University, Jilin, China
| | - Jiaqi Liu
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Dongbo Bao
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Cheng Hu
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Yundong Zhao
- College of Medical Technology, Beihua University, Jilin, China
| | - Shuang Chen
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| |
Collapse
|
12
|
Garrett TJ, Coatsworth H, Mahmud I, Hamerly T, Stephenson CJ, Ayers JB, Yazd HS, Miller MR, Lednicky JA, Dinglasan RR. Niclosamide as a chemical probe for analyzing SARS-CoV-2 modulation of host cell lipid metabolism. Front Microbiol 2023; 14:1251065. [PMID: 37901834 PMCID: PMC10603251 DOI: 10.3389/fmicb.2023.1251065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/15/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction SARS-CoV-2 subverts host cell processes to facilitate rapid replication and dissemination, and this leads to pathological inflammation. Methods We used niclosamide (NIC), a poorly soluble anti-helminth drug identified initially for repurposed treatment of COVID-19, which activates the cells' autophagic and lipophagic processes as a chemical probe to determine if it can modulate the host cell's total lipid profile that would otherwise be either amplified or reduced during SARS-CoV-2 infection. Results Through parallel lipidomic and transcriptomic analyses we observed massive reorganization of lipid profiles of SARS-CoV-2 infected Vero E6 cells, especially with triglycerides, which were elevated early during virus replication, but decreased thereafter, as well as plasmalogens, which were elevated at later timepoints during virus replication, but were also elevated under normal cell growth. These findings suggested a complex interplay of lipid profile reorganization involving plasmalogen metabolism. We also observed that NIC treatment of both low and high viral loads does not affect virus entry. Instead, NIC treatment reduced the abundance of plasmalogens, diacylglycerides, and ceramides, which we found elevated during virus infection in the absence of NIC, resulting in a significant reduction in the production of infectious virions. Unexpectedly, at higher viral loads, NIC treatment also resulted in elevated triglyceride levels, and induced significant changes in phospholipid metabolism. Discussion We posit that future screens of approved or new partner drugs should prioritize compounds that effectively counter SARS-CoV-2 subversion of lipid metabolism, thereby reducing virus replication, egress, and the subsequent regulation of key lipid mediators of pathological inflammation.
Collapse
Affiliation(s)
- Timothy J. Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
- Southeast Center for Integrated Metabolomics, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, United States
| | - Heather Coatsworth
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Iqbal Mahmud
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
- Southeast Center for Integrated Metabolomics, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, United States
| | - Timothy Hamerly
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Caroline J. Stephenson
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Jasmine B. Ayers
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Hoda S. Yazd
- Department of Chemistry, University of Florida, Gainesville, FL, United States
| | - Megan R. Miller
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - John A. Lednicky
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Rhoel R. Dinglasan
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
13
|
Pan Z, Huang J, Song H, Xiao Y, Liu T, Zeng Y, Zhu H, Yang K. PLCL1 suppresses tumour progression by regulating AMPK/mTOR-mediated autophagy in renal cell carcinoma. Aging (Albany NY) 2023; 15:10407-10427. [PMID: 37801481 PMCID: PMC10599749 DOI: 10.18632/aging.205085] [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: 06/08/2023] [Accepted: 09/09/2023] [Indexed: 10/08/2023]
Abstract
Autophagy has been increasingly recognized as a critical regulatory mechanism in the maintenance of cellular homeostasis. A previous study showed that phospholipase C-like protein 1 (PLCL1) is associated with lipid metabolism in renal cell carcinoma (RCC). However, it is unclear whether PLCL1 regulates autophagy, thereby influencing the progression of RCC. Bioinformatics analysis of five microarray datasets revealed that expression of PLCL1 is decreased in tumours and is positively correlated with prognosis in RCC patients. Three independent public datasets, clinical RCC tissues and RCC cell lines, were validated using real-time qPCR, western blotting and immunohistochemistry. Using wound healing and transwell assays, we observed that elevated PLCL1 levels decreased the migratory distance and the invasive number of 786-O and ACHN cells, but PLCL1 knockdown reversed these changes in 769P cell lines compared to those in controls. The results of flow cytometry analysis indicated that PLCL1 promotes apoptosis. Moreover, transcriptional analysis based on stable overexpression of PLCL1 in 786-O cells revealed that PLCL1 is related to autophagy, and western blotting and autophagic experimental results further verified these findings. Mechanistic investigations confirmed that PLCL1 activates the AMPK/mTOR pathway and interacts with decidual protein induced by progesterone (DEPP). Collectively, our data suggest that PLCL1 functions as a suppressor of RCC progression by activating the AMPK/mTOR pathway, interacting with DEPP, initiating autophagy and inducing apoptosis. PLCL1 may be a promising therapeutic target for the diagnosis and treatment of ccRCC patients.
Collapse
Affiliation(s)
- Zhou Pan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 443002, P.R. China
| | - Jing Huang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan 443002, P.R. China
| | - Huajie Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 443002, P.R. China
| | - Yusha Xiao
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 443002, P.R. China
| | - Ting Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 443002, P.R. China
| | - Yan Zeng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 443002, P.R. China
| | - Hengcheng Zhu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 443002, P.R. China
| | - Kang Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 443002, P.R. China
| |
Collapse
|
14
|
Lin JC, Liu TP, Chen YB, Yang PM. PF-429242 exhibits anticancer activity in hepatocellular carcinoma cells via FOXO1-dependent autophagic cell death and IGFBP1-dependent anti-survival signaling. Am J Cancer Res 2023; 13:4125-4144. [PMID: 37818050 PMCID: PMC10560959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/22/2023] [Indexed: 10/12/2023] Open
Abstract
Effective therapies for hepatocellular carcinoma (HCC) are urgently needed, as it is a type of cancer resistant to chemotherapy. Recent evidence showed that PF-429242, a membrane-bound transcription factor site-1 protease (MBTPS1) inhibitor, exhibited anticancer activities against glioblastomas, renal cell carcinoma, and pancreatic cancer. However, its anticancer activity against HCC has yet to be investigated. In this study, we found that PF-429242 induced autophagy-dependent cell death in HCC cells. RNA-sequencing analysis indicated that the primary effect of PF-429242 was inhibition of the sterol regulatory element-binding protein (SREBP) signaling pathway. However, overexpression of SREBP proteins did not efficiently rescue PF-429242-induced autophagy and cell death. Mechanistically, PF-429242 induced forkhead box protein O1 (FOXO1)-dependent autophagic cell death. Additionally, PF-429242 caused FOXO1-independent upregulation of insulin-like growth factor-binding protein 1 (IGFBP1), ultimately leading to autophagy-independent cell death. The in vivo anticancer activity of PF-429242 against HCC cells was demonstrated in a tumor xenograft mouse model. Therefore, PF-429242 is a potential anticancer agent to treat HCC by triggering FOXO1-dependent autophagic cell death and IGFBP1-mediated anti-survival signaling in parallel.
Collapse
Affiliation(s)
- Jiunn-Chang Lin
- Department of Surgery, MacKay Memorial HospitalTaipei 10449, Taiwan
- MacKay Junior College of Medicine, Nursing and ManagementNew Taipei 11260, Taiwan
- Department of Medicine, MacKay Medical CollegeNew Taipei 25245, Taiwan
- Liver Medical Center, MacKay Memorial HospitalTaipei 10449, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia SinicaTaipei 11031, Taiwan
| | - Tsang-Pai Liu
- Department of Surgery, MacKay Memorial HospitalTaipei 10449, Taiwan
- MacKay Junior College of Medicine, Nursing and ManagementNew Taipei 11260, Taiwan
- Department of Medicine, MacKay Medical CollegeNew Taipei 25245, Taiwan
- Liver Medical Center, MacKay Memorial HospitalTaipei 10449, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia SinicaTaipei 11031, Taiwan
| | - Yan-Bin Chen
- Department of Surgery, MacKay Memorial HospitalTaipei 10449, Taiwan
| | - Pei-Ming Yang
- Liver Medical Center, MacKay Memorial HospitalTaipei 10449, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia SinicaTaipei 11031, Taiwan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical UniversityTaipei 11031, Taiwan
- TMU Research Center of Cancer Translational MedicineTaipei 11031, Taiwan
- Cancer Center, Wan Fang Hospital, Taipei Medical UniversityTaipei 11696, Taiwan
- TMU and Affiliated Hospitals Pancreatic Cancer Groups, Taipei Medical UniversityTaipei 11031, Taiwan
| |
Collapse
|
15
|
Moghbeli M, Taghehchian N, Akhlaghipour I, Samsami Y, Maharati A. Role of forkhead box proteins in regulation of doxorubicin and paclitaxel responses in tumor cells: A comprehensive review. Int J Biol Macromol 2023; 248:125995. [PMID: 37499722 DOI: 10.1016/j.ijbiomac.2023.125995] [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: 06/20/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Chemotherapy is one of the common first-line therapeutic methods in cancer patients. Despite the significant effects in improving the quality of life and survival of patients, chemo resistance is observed in a significant part of cancer patients, which leads to tumor recurrence and metastasis. Doxorubicin (DOX) and paclitaxel (PTX) are used as the first-line drugs in a wide range of tumors; however, DOX/PTX resistance limits their use in cancer patients. Considering the DOX/PTX side effects in normal tissues, identification of DOX/PTX resistant cancer patients is required to choose the most efficient therapeutic strategy for these patients. Investigating the molecular mechanisms involved in DOX/PTX response can help to improve the prognosis in cancer patients. Several cellular processes such as drug efflux, autophagy, and DNA repair are associated with chemo resistance that can be regulated by transcription factors as the main effectors in signaling pathways. Forkhead box (FOX) family of transcription factor has a key role in regulating cellular processes such as cell differentiation, migration, apoptosis, and proliferation. FOX deregulations have been associated with resistance to chemotherapy in different cancers. Therefore, we discussed the role of FOX protein family in DOX/PTX response. It has been reported that FOX proteins are mainly involved in DOX/PTX response by regulation of drug efflux, autophagy, structural proteins, and signaling pathways such as PI3K/AKT, NF-kb, and JNK. This review is an effective step in introducing the FOX protein family as the reliable prognostic markers and therapeutic targets in cancer patients.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Negin Taghehchian
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yalda Samsami
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Zhu H, Gao X, Wang B, Niu B, Liu D, Zhang J, Jin Y, Yang X. A biodegradable hollow nanoagent enables a boosted chemodynamic therapy by simultaneous autophagy inhibition and macrophage reeducation. Int J Pharm 2023; 643:123248. [PMID: 37467817 DOI: 10.1016/j.ijpharm.2023.123248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/20/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023]
Abstract
Various therapeutic strategies, including chemotherapy, radiotherapy, photothermal therapy (PTT), and immunotherapy have been applied in cancer therapy. However, intrinsic or acquired therapeutic resistance is the main obstacle that attenuates the treatment effect of the therapeutic reagents used in these strategies. Studies have shown that autophagy and immunosuppressive tumor-associated macrophages (TAMs), as internal and external resistance mechanisms, would significantly compromise the effectiveness of cancer treatment. Therefore, selectively blocking the autophagy and repolarizing TAMs to anti-tumor phenotype (M1) will be effective for cancer treatments. Herein, an ambidextrous strategy that simultaneously inhibited autophagy and reeducated TAMs to promote anti-tumor therapy meditated by the iron-based nanocarriers was reported. The released Fe (II) ion reacted with the released artemisinin (ART) to produce ROS for chemodynamic therapy (CDT). The chloroquine (CQ) was used to inhibit autophagy in cancer cells and reset TAMs from the M2 phenotype to the M1 phenotype, eliminating the resistance of cancer cells and realizing an augmented therapeutic effect. This work provides a promising way for augmenting therapeutic efficiency by simultaneously interfering with two critical therapeutic resistance mechanisms.
Collapse
Affiliation(s)
- Han Zhu
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China
| | - Xin Gao
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China
| | - Bei Wang
- College of Basic Medical Science, Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases of Hebei Province, Hebei University, Baoding 071002, PR China
| | - Biao Niu
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China
| | - Dandan Liu
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China
| | - Jinchao Zhang
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China
| | - Yi Jin
- College of Basic Medical Science, Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases of Hebei Province, Hebei University, Baoding 071002, PR China.
| | - Xinjian Yang
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China.
| |
Collapse
|
17
|
Ji L, Zhang L, Liang Z, Zhong S, Liu X, Liu Z, Poon WS, Song Y, Chen B, Wang R. Role of omentin-1 in susceptibility to anxiety and depression like behaviors. Mol Cell Endocrinol 2023; 574:111990. [PMID: 37321286 DOI: 10.1016/j.mce.2023.111990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/17/2023]
Abstract
Neuro-inflammation and blood-brain barrier (BBB) dysfunction are associated with depression. Evidence shows that adipokines enter the brain from the circulation, which regulates depressive behaviors. Omentin-1 is a newly identified adipocytokine that has anti-inflammatory effects, but little is known about its role in neuro-inflammation and mood-relevant behavior. Our results showed omentin-1 knockout mice (Omentin-1-/-) increased susceptibility to anxiety and depressive-like behaviors, which are associated with abnormalities of cerebral blood flow (CBF) and impaired BBB permeability. Moreover, omentin-1 depletion significantly increased hippocampal pro-inflammatory cytokines (IL-1β, TNFα, IL-6), caused microglial activation, inhibited hippocampus neurogenesis, and resulted in autophagy impairment by dysregulating ATG genes. Omentin-1 deficiency also sensitized mice to the behavioral changes induced by lipopolysaccharide (LPS), suggesting that omentin-1 could rescue neuro-inflammation by acting as an anti-depressant. Our in vitro microglia cell culture data confirmed that recombinant omentin-1 suppresses microglial activation and pro-inflammatory cytokine expression induced by LPS. Our study suggests that omentin-1 can be used as a promising therapeutic agent for the prevention or treatment of depression by providing a barrier-promoting effect and an endogenous anti-inflammatory balance to downregulate the proinflammatory cytokines.
Collapse
Affiliation(s)
- Lianru Ji
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China; Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Disease with Mental Disorders), China; Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, China; Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive Dysfunction, Jiangxi University of Chinese Medicine, China
| | - Lang Zhang
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, China; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Zhi Liang
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China; Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Disease with Mental Disorders), China; Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, China; Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive Dysfunction, Jiangxi University of Chinese Medicine, China
| | - Sufang Zhong
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China; Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Disease with Mental Disorders), China; Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, China; Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive Dysfunction, Jiangxi University of Chinese Medicine, China
| | - Xiamin Liu
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China; Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Disease with Mental Disorders), China; Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, China; Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive Dysfunction, Jiangxi University of Chinese Medicine, China
| | - Zhiping Liu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Wai Sang Poon
- The University of Hong Kong, Hong Kong, China; Hong Kong University Shenzhen Hospital, Shen Zhen, China
| | - Yonggui Song
- Key Laboratory of Evaluation of Traditional Chinese Medicine Efficacy (Prevention and Treatment of Brain Disease with Mental Disorders), China; Key Laboratory of Depression Animal Model Based on TCM Syndrome, Jiangxi Administration of Traditional Chinese Medicine, China; Key Laboratory of TCM for Prevention and Treatment of Brain Diseases with Cognitive Dysfunction, Jiangxi University of Chinese Medicine, China.
| | - Baodong Chen
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Rikang Wang
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
18
|
Yan D, He Q, Pei L, Yang M, Huang L, Kong J, He W, Liu H, Xu S, Qin H, Lin T, Huang J. The APC/C E3 ligase subunit ANAPC11 mediates FOXO3 protein degradation to promote cell proliferation and lymph node metastasis in urothelial bladder cancer. Cell Death Dis 2023; 14:516. [PMID: 37573356 PMCID: PMC10423259 DOI: 10.1038/s41419-023-06000-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/18/2023] [Accepted: 07/14/2023] [Indexed: 08/14/2023]
Abstract
Urothelial bladder cancer (UBC) is one of the most prevalent malignancies worldwide, with striking tumor heterogeneity. Elucidating the molecular mechanisms that can be exploited for the treatment of aggressive UBC is a particularly relevant goal. Protein ubiquitination is a critical post-translational modification (PTM) that mediates the degradation of target protein via the proteasome. However, the roles of aberrant protein ubiquitination in UBC development and the underlying mechanisms by which it drives tumor progression remain unclear. In this study, taking advantage of clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein (Cas) 9 technology, we identified the ubiquitin E3 ligase ANAPC11, a critical subunit of the anaphase-promoting complex/cyclosome (APC/C), as a potential oncogenic molecule in UBC cells. Our clinical analysis showed that elevated expression of ANAPC11 was significantly correlated with high T stage, positive lymph node (LN) metastasis, and poor outcomes in UBC patients. By employing a series of in vitro experiments, we demonstrated that ANAPC11 enhanced the proliferation and invasiveness of UBC cells, while knockout of ANAPC11 inhibited the growth and LN metastasis of UBC cells in vivo. By conducting immunoprecipitation coupled with mass spectrometry, we confirmed that ANAPC11 increased the ubiquitination level of the Forkhead transcription factor FOXO3. The resulting decrease in FOXO3 protein stability led to the downregulation of the cell cycle regulator p21 and decreased expression of GULP1, a downstream effector of androgen receptor signaling. Taken together, these findings indicated that ANAPC11 plays an oncogenic role in UBC by modulating FOXO3 protein degradation. The ANAPC11-FOXO3 regulatory axis might serve as a novel therapeutic target for UBC.
Collapse
Affiliation(s)
- Dong Yan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingqing He
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lu Pei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Meihua Yang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lifang Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianqiu Kong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wang He
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Liu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shizhong Xu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haide Qin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
19
|
Li Q. scTour: a deep learning architecture for robust inference and accurate prediction of cellular dynamics. Genome Biol 2023; 24:149. [PMID: 37353848 PMCID: PMC10290357 DOI: 10.1186/s13059-023-02988-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 06/13/2023] [Indexed: 06/25/2023] Open
Abstract
Despite the continued efforts, a batch-insensitive tool that can both infer and predict the developmental dynamics using single-cell genomics is lacking. Here, I present scTour, a novel deep learning architecture to perform robust inference and accurate prediction of cellular dynamics with minimal influence from batch effects. For inference, scTour simultaneously estimates the developmental pseudotime, delineates the vector field, and maps the transcriptomic latent space under a single, integrated framework. For prediction, scTour precisely reconstructs the underlying dynamics of unseen cellular states or a new independent dataset. scTour's functionalities are demonstrated in a variety of biological processes from 19 datasets.
Collapse
Affiliation(s)
- Qian Li
- Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
Gao Y, Yuan Y, Wen S, Chen Y, Zhang Z, Feng Y, Jiang B, Ma S, Hu R, Fang C, Ruan X, Yuan Y, Fang X, Luo C, Meng Z, Wang X, Guo X. Dual role of ANGPTL8 in promoting tumor cell proliferation and immune escape during hepatocarcinogenesis. Oncogenesis 2023; 12:26. [PMID: 37188659 DOI: 10.1038/s41389-023-00473-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/17/2023] Open
Abstract
The interplay between hepatocellular carcinoma (HCC) cells and the tumor microenvironment is essential for hepatocarcinogenesis, but their contributions to HCC development are incompletely understood. We assessed the role of ANGPTL8, a protein secreted by HCC cells, in hepatocarcinogenesis and the mechanisms through which ANGPTL8 mediates crosstalk between HCC cells and tumor-associated macrophages. Immunohistochemical, Western blotting, RNA-Seq, and flow cytometry analyses of ANGPTL8 were performed. A series of in vitro and in vivo experiments were conducted to reveal the role of ANGPTL8 in the progression of HCC. ANGPTL8 expression was positively correlated with tumor malignancy in HCC, and high ANGPTL8 expression was associated with poor overall survival (OS) and disease-free survival (DFS). ANGPTL8 promoted HCC cell proliferation in vitro and in vivo, and ANGPTL8 KO inhibited the development of HCC in both DEN-induced and DEN-plus-CCL4-induced mouse HCC tumors. Mechanistically, the ANGPTL8-LILRB2/PIRB interaction promoted polarization of macrophages to the immunosuppressive M2 phenotype in macrophages and recruited immunosuppressive T cells. In hepatocytes, ANGPTL8-mediated stimulation of LILRB2/PIRB regulated the ROS/ERK pathway and upregulated autophagy, leading to the proliferation of HCC cells. Our data support the notion that ANGPTL8 has a dual role in promoting tumor cell proliferation and immune escape during hepatocarcinogenesis.
Collapse
Affiliation(s)
- Yujiu Gao
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
- Department of Nephrology, Taihe Hospital, 442000, Shiyan, China
- Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, 442000, Shiyan, China
| | - Yue Yuan
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
- College of Pharmacy, Hubei University of Medicine, 442000, Shiyan, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 430060, Wuhan, China
| | - Shu Wen
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Yanghui Chen
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Zongli Zhang
- Institute of Pediatric Disease, Taihe Hospital, 442000, Shiyan, China
| | - Ying Feng
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Bin Jiang
- Department of Hepatobiliary Pancreatic Surgery, Taihe Hospital, 442000, Shiyan, China
| | - Shinan Ma
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Rong Hu
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Chen Fang
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Xuzhi Ruan
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Yahong Yuan
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Xinggang Fang
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Chao Luo
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - Zhongji Meng
- Department of Infectious Diseases, Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, 442000, Shiyan, China.
| | - Xiaoli Wang
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China.
- Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, 442000, Shiyan, China.
| | - Xingrong Guo
- Department of Critical Care Medicine, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, 442000, Shiyan, China.
- Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, 442000, Shiyan, China.
| |
Collapse
|
21
|
Chen Y, Tang M, Li H, Huang J. Effects of C10orf10 on growth and prognosis of glioma under hypoxia. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:499-507. [PMID: 37385612 PMCID: PMC10930248 DOI: 10.11817/j.issn.1672-7347.2023.220396] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Indexed: 07/01/2023]
Abstract
OBJECTIVES Glioma is the most common malignant tumor in the central nervous system, and the hypoxic microenvironment is prevalent in solid tumors. This study aims to investigate the up-regulation of genes under the condition of hypoxia and their roles in glioma growth, as well as their impact on glioma prognosis. METHODS The hypoxia-related dataset with glioma was screened in the Gene Expression Omnibus database (GEO), and the differentially expressed genes were analyzed between hypoxia and normoxia through bioinformatics, and chromosome 10 open reading frame 10 (C10orf10) was verified and screened in hypoxia-treated cells through real-time PCR and Western blotting. The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) datasets were downloaded to analyze the mRNA expression of C10orf10 in different grades of glioma and its impact on prognosis. The glioma specimens and follow-up data of 68 gliomas who underwent surgical treatment in Xiangya Hospital of Central South University from March 2017 to January 2021 were collected, and real-time PCR was used to detect the mRNA expression of C10orf10 in different grades of glioma, and the Kaplan-Meier method was used to analyze the relationship between the expression C10orf10 and prognosis. The glioma cells, which could interfere the expression of C10orf10, were constructed, and the effect of C10orf10 on the proliferation of glioma cells was evaluated by cell counting kit-8 (CCK-8) and colony formation assays. RESULTS Compared with the condition of normoxia, the expression levels of C10orf10 mRNA and protein were significantly up-regulated in glioma cells under hypoxia (P<0.001), and the mRNA expression level of C10orf10 in glioma tissues was up-regulated with the increase of WHO grade in glioma (P<0.001). Based on Kaplan-Meier survival analysis, the higher the mRNA expression level of C10orf10 was, the shorter the survival time of the patient was (P<0.05). And the expression of C10orf10 mRNA was higher in recurrent gliomas than that in primary gliomas in the CGGA database (P<0.001). Knockdown of C10orf10 could significantly inhibit the growth of glioma cells both under hypoxia and normoxia (both P<0.001). CONCLUSIONS The expression level of C10orf10 can promote the proliferation and prognosis of glioma, which is expected to become a prognostic marker and therapeutic target for glioma.
Collapse
Affiliation(s)
- Yuanbing Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008.
| | - Miao Tang
- Department of Neurosurgery, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Hui Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008
| | - Jun Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008.
| |
Collapse
|
22
|
Yang DL, Li Y, Ma SQ, Zhang YJ, Huang JH, He LJ. Compound 275# Induces Mitochondria-Mediated Apoptosis and Autophagy Initiation in Colorectal Cancer Cells through an Accumulation of Intracellular ROS. Molecules 2023; 28:molecules28073211. [PMID: 37049976 PMCID: PMC10095895 DOI: 10.3390/molecules28073211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
Colorectal cancer (CRC) is the most common intestinal malignancy, and nearly 70% of patients with this cancer develop metastatic disease. In the present study, we synthesized a novel compound, termed N-(3-(5,7-dimethylbenzo [d]oxazol-2-yl)phenyl)-5-nitrofuran-2-carboxamide (compound 275#), and found that it exhibits antiproliferative capability in suppressing the proliferation and growth of CRC cell lines. Furthermore, compound 275# triggered caspase 3-mediated intrinsic apoptosis of mitochondria and autophagy initiation. An investigation of the molecular mechanisms demonstrated that compound 275# induced intrinsic apoptosis, and autophagy initiation was largely mediated by increasing the levels of the intracellular accumulation of reactive oxygen species (ROS) in CRC cells. Taken together, these data suggest that ROS accumulation after treatment with compound 275# leads to mitochondria-mediated apoptosis and autophagy activation, highlighting the potential of compound 275# as a novel therapeutic agent for the treatment of CRC.
Collapse
Affiliation(s)
- Dong-Lin Yang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing 402160, China
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Yong Li
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Shui-Qing Ma
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Ya-Jun Zhang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Jiu-Hong Huang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing 402160, China
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Liu-Jun He
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing 402160, China
| |
Collapse
|
23
|
Zanini F, Che X, Knutsen C, Liu M, Suresh NE, Domingo-Gonzalez R, Dou SH, Zhang D, Pryhuber GS, Jones RC, Quake SR, Cornfield DN, Alvira CM. Developmental diversity and unique sensitivity to injury of lung endothelial subtypes during postnatal growth. iScience 2023; 26:106097. [PMID: 36879800 PMCID: PMC9984561 DOI: 10.1016/j.isci.2023.106097] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/20/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
At birth, the lung is still immature, heightening susceptibility to injury but enhancing regenerative capacity. Angiogenesis drives postnatal lung development. Therefore, we profiled the transcriptional ontogeny and sensitivity to injury of pulmonary endothelial cells (EC) during early postnatal life. Although subtype speciation was evident at birth, immature lung EC exhibited transcriptomes distinct from mature counterparts, which progressed dynamically over time. Gradual, temporal changes in aerocyte capillary EC (CAP2) contrasted with more marked alterations in general capillary EC (CAP1) phenotype, including distinct CAP1 present only in the early alveolar lung expressing Peg3, a paternally imprinted transcription factor. Hyperoxia, an injury that impairs angiogenesis induced both common and unique endothelial gene signatures, dysregulated capillary EC crosstalk, and suppressed CAP1 proliferation while stimulating venous EC proliferation. These data highlight the diversity, transcriptomic evolution, and pleiotropic responses to injury of immature lung EC, possessing broad implications for lung development and injury across the lifespan.
Collapse
Affiliation(s)
- Fabio Zanini
- Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Kensington, NSW 2052, Australia
| | - Xibing Che
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Carsten Knutsen
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Min Liu
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nina E. Suresh
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Racquel Domingo-Gonzalez
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steve H. Dou
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daoqin Zhang
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gloria S. Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Robert C. Jones
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Stephen R. Quake
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - David N. Cornfield
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Cristina M. Alvira
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
24
|
Cha HS, Lee HK, Park SH, Nam MJ. Acetylshikonin induces apoptosis of human osteosarcoma U2OS cells by triggering ROS-dependent multiple signal pathways. Toxicol In Vitro 2022; 86:105521. [DOI: 10.1016/j.tiv.2022.105521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/18/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
|
25
|
Kohoutova K, Dočekal V, Ausserlechner MJ, Kaiser N, Tekel A, Mandal R, Horvath M, Obsilova V, Vesely J, Hagenbuchner J, Obsil T. Lengthening the Guanidine-Aryl Linker of Phenylpyrimidinylguanidines Increases Their Potency as Inhibitors of FOXO3-Induced Gene Transcription. ACS OMEGA 2022; 7:34632-34646. [PMID: 36188303 PMCID: PMC9521028 DOI: 10.1021/acsomega.2c04613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/07/2022] [Indexed: 06/16/2023]
Abstract
Increased FOXO3 nuclear localization is involved in neuroblastoma chemoresistance and tumor angiogenesis. Accordingly, FOXO3 inhibition is a promising strategy for boosting antitumor immune responses and suppressing FOXO3-mediated therapy resistance in cancer cells. However, no FOXO3 inhibitors are currently available for clinical use. Nevertheless, we have recently identified (4-propoxy)phenylpyrimidinylguanidine as a FOXO3 inhibitor in cancer cells in the low micromolar range. Here, we report the synthesis and structure-activity relationship study of a small library of its derivatives, some of which inhibit FOXO3-induced gene transcription in cancer cells in a submicromolar range and are thus 1 order of magnitude more potent than their parent compound. By NMR and molecular docking, we showed that these compounds differ in their interactions with the DNA-binding domain of FOXO3. These results may provide a foundation for further optimizing (4-propoxy)phenylpyrimidinylguanidine and developing therapeutics for inhibiting the activity of forkhead box (FOX) transcription factors and their interactions with other binding partners.
Collapse
Affiliation(s)
- Klara Kohoutova
- Department
of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
- Institute
of Physiology of the Czech Academy of Sciences, Laboratory of Structural
Biology of Signaling Proteins, Division
BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic
| | - Vojtěch Dočekal
- Department
of Organic Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
| | | | - Nora Kaiser
- Department
of Pediatrics I, Medical University Innsbruck, Innrain 66, Innsbruck 6020, Austria
| | - Andrej Tekel
- Department
of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
| | - Raju Mandal
- Department
of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
| | - Matej Horvath
- Department
of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
| | - Veronika Obsilova
- Institute
of Physiology of the Czech Academy of Sciences, Laboratory of Structural
Biology of Signaling Proteins, Division
BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic
| | - Jan Vesely
- Department
of Organic Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
| | - Judith Hagenbuchner
- Department
of Pediatrics II, Medical University Innsbruck, Innrain 66, Innsbruck 6020, Austria
| | - Tomas Obsil
- Department
of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
- Institute
of Physiology of the Czech Academy of Sciences, Laboratory of Structural
Biology of Signaling Proteins, Division
BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic
| |
Collapse
|
26
|
Kim YH, Jo DS, Park NY, Bae JE, Kim JB, Lee HJ, Kim SH, Kim SH, Lee S, Son M, Park K, Jeong K, Yeom E, Cho DH. Inhibition of BRD4 Promotes Pexophagy by Increasing ROS and ATM Activation. Cells 2022; 11:cells11182839. [PMID: 36139416 PMCID: PMC9497081 DOI: 10.3390/cells11182839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/09/2022] [Accepted: 09/10/2022] [Indexed: 01/18/2023] Open
Abstract
Although autophagy regulates the quality and quantity of cellular compartments, the regulatory mechanisms underlying peroxisomal autophagy (pexophagy) remain largely unknown. In this study, we identified several BRD4 inhibitors, including molibresib, a novel pexophagy inducer, via chemical library screening. Treatment with molibresib promotes loss of peroxisomes selectively, but not mitochondria, ER, or Golgi apparatus in HeLa cells. Consistently, depletion of BRD4 expression also induced pexophagy in RPE cells. In addition, the inhibition of BRD4 by molibresib increased autophagic degradation of peroxisome ATG7-dependency. We further found that molibresib produced reactive oxygen species (ROS), which potentiates ATM activation. Inhibition of ROS or ATM suppressed the loss of peroxisomes in molibresib-treated cells. Taken together, our data suggest that inhibition of BRD4 promotes pexophagy by increasing ROS and ATM activation.
Collapse
Affiliation(s)
- Yong Hwan Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Doo Sin Jo
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Na Yeon Park
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Ji-Eun Bae
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea
| | - Joon Bum Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Ha Jung Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - So Hyun Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Seong Hyun Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Sunwoo Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Mikyung Son
- Orgasis Corp., Suwon 16229, Gyeonggi-do, Korea
| | - Kyuhee Park
- Bio-Center, Gyeonggido Business & Science Accelerator, Suwon 16229, Gyeonggi-do, Korea
| | - Kwiwan Jeong
- Bio-Center, Gyeonggido Business & Science Accelerator, Suwon 16229, Gyeonggi-do, Korea
| | - Eunbyul Yeom
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Dong-Hyung Cho
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
- Orgasis Corp., Suwon 16229, Gyeonggi-do, Korea
- Correspondence: ; Tel.: +82-53-950-5382
| |
Collapse
|
27
|
Pro-oxidative priming but maintained cardiac function in a broad spectrum of murine models of chronic kidney disease. Redox Biol 2022; 56:102459. [PMID: 36099852 PMCID: PMC9482130 DOI: 10.1016/j.redox.2022.102459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Aims Patients with chronic kidney disease (CKD) have an increased risk of cardiovascular events and exhibit myocardial changes including left ventricular (LV) hypertrophy and fibrosis, overall referred to as ‘uremic cardiomyopathy’. Although different CKD animal models have been studied for cardiac effects, lack of consistent reporting on cardiac function and pathology complicates clear comparison of these models. Therefore, this study aimed at a systematic and comprehensive comparison of cardiac function and cardiac pathophysiological characteristics in eight different CKD models and mouse strains, with a main focus on adenine-induced CKD. Methods and results CKD of different severity and duration was induced by subtotal nephrectomy or adenine-rich diet in various strains (C57BL/6J, C57BL/6 N, hyperlipidemic C57BL/6J ApoE−/−, 129/Sv), followed by the analysis of kidney function and morphology, blood pressure, cardiac function, cardiac hypertrophy, fibrosis, myocardial calcification and inflammation using functional, histological and molecular techniques, including cardiac gene expression profiling supplemented by oxidative stress analysis. Intriguingly, despite uremia of variable degree, neither cardiac dysfunction, hypertrophy nor interstitial fibrosis were observed. However, already moderate CKD altered cardiac oxidative stress responses and enhanced oxidative stress markers in each mouse strain, with cardiac RNA sequencing revealing activation of oxidative stress signaling as well as anti-inflammatory feedback responses. Conclusion This study considerably expands the knowledge on strain- and protocol-specific differences in the field of cardiorenal research and reveals that several weeks of at least moderate experimental CKD increase oxidative stress responses in the heart in a broad spectrum of mouse models. However, this was insufficient to induce relevant systolic or diastolic dysfunction, suggesting that additional “hits” are required to induce uremic cardiomyopathy. Translational perspective Patients with chronic kidney disease (CKD) have an increased risk of cardiovascular adverse events and exhibit myocardial changes, overall referred to as ‘uremic cardiomyopathy’. We revealed that CKD increases cardiac oxidative stress responses in the heart. Nonetheless, several weeks of at least moderate experimental CKD do not necessarily trigger cardiac dysfunction and remodeling, suggesting that additional “hits” are required to induce uremic cardiomyopathy in the clinical setting. Whether the altered cardiac oxidative stress balance in CKD may increase the risk and extent of cardiovascular damage upon additional cardiovascular risk factors and/or events will be addressed in future studies. Development of a CKD mouse model with a clear cardiac functional or morphological phenotype is challenging. Cardiac oxidative stress response as well as oxidative stress markers are increased in a broad spectrum of CKD mouse models. Our findings suggest need of additional cardiovascular hits to clearly induce uremic cardiomyopathy as observed in patients.
Collapse
|
28
|
Gómez-Virgilio L, Silva-Lucero MDC, Flores-Morelos DS, Gallardo-Nieto J, Lopez-Toledo G, Abarca-Fernandez AM, Zacapala-Gómez AE, Luna-Muñoz J, Montiel-Sosa F, Soto-Rojas LO, Pacheco-Herrero M, Cardenas-Aguayo MDC. Autophagy: A Key Regulator of Homeostasis and Disease: An Overview of Molecular Mechanisms and Modulators. Cells 2022; 11:cells11152262. [PMID: 35892559 PMCID: PMC9329718 DOI: 10.3390/cells11152262] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy is a highly conserved lysosomal degradation pathway active at basal levels in all cells. However, under stress conditions, such as a lack of nutrients or trophic factors, it works as a survival mechanism that allows the generation of metabolic precursors for the proper functioning of the cells until the nutrients are available. Neurons, as post-mitotic cells, depend largely on autophagy to maintain cell homeostasis to get rid of damaged and/or old organelles and misfolded or aggregated proteins. Therefore, the dysfunction of this process contributes to the pathologies of many human diseases. Furthermore, autophagy is highly active during differentiation and development. In this review, we describe the current knowledge of the different pathways, molecular mechanisms, factors that induce it, and the regulation of mammalian autophagy. We also discuss its relevant role in development and disease. Finally, here we summarize several investigations demonstrating that autophagic abnormalities have been considered the underlying reasons for many human diseases, including liver disease, cardiovascular, cerebrovascular diseases, neurodegenerative diseases, neoplastic diseases, cancers, and, more recently, infectious diseases, such as SARS-CoV-2 caused COVID-19 disease.
Collapse
Affiliation(s)
- Laura Gómez-Virgilio
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
| | - Maria-del-Carmen Silva-Lucero
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
| | - Diego-Salvador Flores-Morelos
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Guerrero, Mexico;
| | - Jazmin Gallardo-Nieto
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Biotechnology Engeniering, Universidad Politécnica de Quintana Roo, Cancún 77500, Quintana Roo, Mexico
| | - Gustavo Lopez-Toledo
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
| | - Arminda-Mercedes Abarca-Fernandez
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Biotechnology Engeniering, Universidad Politécnica de Quintana Roo, Cancún 77500, Quintana Roo, Mexico
| | - Ana-Elvira Zacapala-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Guerrero, Mexico;
| | - José Luna-Muñoz
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlan Izcalli 53150, Estado de México, Mexico; (J.L.-M.); (F.M.-S.)
- Banco Nacional de Cerebros-UNPHU, Universidad Nacional Pedro Henríquez Ureña, Santo Domingo 11805, Dominican Republic
| | - Francisco Montiel-Sosa
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlan Izcalli 53150, Estado de México, Mexico; (J.L.-M.); (F.M.-S.)
| | - Luis O. Soto-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de México, Mexico;
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de México, Mexico
| | - Mar Pacheco-Herrero
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Santiago de los Caballeros 51000, Dominican Republic;
| | - Maria-del-Carmen Cardenas-Aguayo
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Correspondence: ; Tel.: +52-55-2907-0937
| |
Collapse
|
29
|
Potočnjak I, Šimić L, Vukelić I, Batičić L, Domitrović R. Oleanolic acid induces HCT116 colon cancer cell death through the p38/FOXO3a/Sirt6 pathway. Chem Biol Interact 2022; 363:110010. [PMID: 35690101 DOI: 10.1016/j.cbi.2022.110010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/14/2022] [Accepted: 06/07/2022] [Indexed: 12/08/2022]
Abstract
Oleanolic acid (OA) is a natural compound that possesses numerous beneficial health effects, including anticancer activity. The current study aimed to investigate the role of forkhead box O3a (FOXO3a) in autophagy/mitophagy by OA in HCT116 cell line. OA dose-dependently reduced viability of HCT116 cells, with IC50 = 29.8 μΜ. The expression of cleaved caspase-3 and poly (ADP-ribose) polymerase 1 increased after OA treatment, suggesting induction of apoptosis. Concurrently, OA induced autophagy, evidenced by increased expression of Beclin-1, autophagy-related protein 5 and microtubule-associated protein1A/1B-light chain 3 beta (LC3B), which played a prosurvival role. The induction of mitophagy was suggested by increased expression of p62 and PTEN-induced kinase 1 and reduced expression of translocase of outer mitochondrial membrane 20, which colocalized with LC3B. OA also induced nuclear accumulation of forkhead box O3a (FOXO3a). The cytotoxic activity of OA coincided with upregulation of p38. Inhibition of p38 led to increase in FOXO3a and NAD+-dependent deacetylase sirtuin 6 expression. In vivo, OA inhibited tumor growth in colon cancer xenograft mice. Our results suggest concomitant induction of apoptosis and prosurvival mitophagy by OA in colon cancer via p38/FOXO3a/Sirt6 signaling. Additionally, our data demonstrate that OA can chemosensitize colon cancer cells to 5-fluorouracil (5-FU).
Collapse
Affiliation(s)
- Iva Potočnjak
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Croatia
| | - Lidija Šimić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Croatia
| | - Iva Vukelić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Croatia
| | - Lara Batičić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Croatia
| | - Robert Domitrović
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Croatia.
| |
Collapse
|
30
|
Loh JS, Rahim NA, Tor YS, Foo JB. Simultaneous proteasome and autophagy inhibition synergistically enhances cytotoxicity of doxorubicin in breast cancer cells. Cell Biochem Funct 2022; 40:403-416. [PMID: 35485606 DOI: 10.1002/cbf.3704] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/22/2022] [Indexed: 01/18/2023]
Abstract
Ubiquitin-proteasome system (UPS) and autophagy are interconnected proteolysis pathways implicated in doxorubicin resistance of breast cancer cells. Following anticancer treatments, autophagy either plays a cytoprotective role or augments treatment-induced cytotoxicity. However, the role of autophagy in breast cancer cells cotreated with doxorubicin and ixazomib remains unclear. The expression of autophagy proteins (LC3A/B and Beclin-1) and UPS protein (ubiquitin) in MDA-MB-231 and MCF-7 cells following doxorubicin, ixazomib, and/or hydroxychloroquine were determined by western blot. The combinatorial effects and combination index (CI) of triple-combination were determined by cell viability assay and CompuSyn software, respectively. Doxorubicin and ixazomib cotreatment increased Beclin-1 (3.8- and 3.5-fold) and LC3-II expression (13.5- and 1.9-fold) in MDA-MB-231 and MCF-7 cells, respectively. Adding lysosomal inhibitor hydroxychloroquine to doxorubicin and ixazomib further increased LC3-II expression to 45.0- and 16.5-fold in MDA-MB-231 and MCF-7 cells, respectively, confirming autophagy induction. The triple-combination synergistically inhibited cell growth, achieving CI 0.672 and 0.157 in MDA-MB-231 and MCF-7 cells, respectively. The triple-combination also induced ubiquitinated proteins accumulation (2.5-fold and 3.0-fold) in MDA-MB-231 and MCF-7 cells, respectively. These results suggest that the autophagy induced by doxorubicin and ixazomib cotreatment serves cytoprotective role in breast cancer cells. Simultaneous UPS and autophagy inhibition synergistically enhanced doxorubicin-mediated cytotoxicity.
Collapse
Affiliation(s)
- Jian Sheng Loh
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Nusaibah Abdul Rahim
- Department of Clinical Pharmacy & Pharmacy Practice, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Yin Sim Tor
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia.,Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia.,Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
31
|
Oyabu M, Takigawa K, Mizutani S, Hatazawa Y, Fujita M, Ohira Y, Sugimoto T, Suzuki O, Tsuchiya K, Suganami T, Ogawa Y, Ishihara K, Miura S, Kamei Y. FOXO1 cooperates with C/EBPδ and ATF4 to regulate skeletal muscle atrophy transcriptional program during fasting. FASEB J 2022; 36:e22152. [DOI: 10.1096/fj.202101385rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Mamoru Oyabu
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Kaho Takigawa
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Sako Mizutani
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Yukino Hatazawa
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Mariko Fujita
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Yuto Ohira
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Takumi Sugimoto
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Osamu Suzuki
- Laboratory of Animal Models for Human Diseases National Institutes of Biomedical Innovation, Health and Nutrition Osaka Japan
| | - Kyoichiro Tsuchiya
- Third Department of Internal Medicine Interdisciplinary Graduate School of Medicine and Engineering University of Yamanashi Yamanashi Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism Research Institute of Environmental Medicine Nagoya University Nagoya Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Kengo Ishihara
- Department of Food Science and Human Nutrition Faculty of Agriculture Ryukoku University Shiga Japan
| | - Shinji Miura
- Graduate School of Nutritional and Environmental Sciences University of Shizuoka Shizuoka Japan
| | - Yasutomi Kamei
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| |
Collapse
|
32
|
Kuwahara M, Akasaki Y, Kurakazu I, Sueishi T, Toya M, Uchida T, Tsutsui T, Hirose R, Tsushima H, Teramura T, Nakashima Y. C10orf10/DEPP activates mitochondrial autophagy and maintains chondrocyte viability in the pathogenesis of osteoarthritis. FASEB J 2022; 36:e22145. [PMID: 34997944 DOI: 10.1096/fj.202100896r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/13/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA), the most prevalent joint disease, is characterized by the progressive loss of articular cartilage. Autophagy, a lysosomal degradation pathway, maintains cellular homeostasis, and autophagic dysfunction in chondrocytes is a hallmark of OA pathogenesis. However, the cause of autophagic dysfunction in OA chondrocytes remains incompletely understood. Recent studies have reported that decidual protein induced by progesterone (C10orf10/DEPP) positively regulates autophagic functions. In this study, we found that DEPP was involved in mitochondrial autophagic functions of chondrocytes, as well as in OA pathogenesis. DEPP expression decreased in human OA chondrocytes in the absence or presence of pro-inflammatory cytokines, and was induced by starvation, hydrogen peroxide (H2 O2 ), and hypoxia (cobalt chloride). For functional studies, DEPP knockdown decreased autophagic flux induced by H2 O2 , whereas DEPP overexpression increased autophagic flux and maintained cell viability following H2 O2 treatment. DEPP was downregulated by knockdown of forkhead box class O (FOXO) transcription factors and modulated the autophagic function regulated by FOXO3. In an OA mouse model by destabilization of the medial meniscus, DEPP-knockout mice exacerbated the progression of cartilage degradation with TUNEL-positive cells, and chondrocytes isolated from knockout mice were decreased autophagic flux and increased cell death following H2 O2 treatment. Subcellular fractionation analysis revealed that mitochondria-located DEPP activated mitochondrial autophagy via BCL2 interacting protein 3. Taken together, our data demonstrate that DEPP is a major stress-inducible gene involved in the activation of mitochondrial autophagy in chondrocytes, and maintains chondrocyte viability during OA pathogenesis. DEPP represents a potential therapeutic target for enhancing autophagy in patients with OA.
Collapse
Affiliation(s)
- Masanari Kuwahara
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Yukio Akasaki
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Ichiro Kurakazu
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Takuya Sueishi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Taisuke Uchida
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Tomoaki Tsutsui
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Ryota Hirose
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Hidetoshi Tsushima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka-sayama, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| |
Collapse
|
33
|
Nath LR, Swetha M, Vijayakurup V, Thangarasu AK, Haritha NH, Shabna A, Aiswarya SU, Rayginia TP, Keerthana CK, Kalimuthu K, Sundaram S, Lankalapalli RS, Pillai S, Towner R, Isakov N, Anto RJ. Blockade of Uttroside B-Induced Autophagic Pro-Survival Signals Augments Its Chemotherapeutic Efficacy Against Hepatocellular Carcinoma. Front Oncol 2022; 12:812598. [PMID: 35211405 PMCID: PMC8861526 DOI: 10.3389/fonc.2022.812598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/17/2022] [Indexed: 02/05/2023] Open
Abstract
Our previous study has demonstrated that Uttroside B (Utt-B), a saponin isolated from the leaves of Solanum nigrum Linn induces apoptosis in hepatic cancer cells and exhibits a remarkable growth inhibition of Hepatocellular Carcinoma (HCC). Our innovation has been granted a patent from the US (US 2019/0160088A1), Canada (3,026,426.), Japan (JP2019520425) and South Korea (KR1020190008323) and the technology have been transferred commercially to Q Biomed, a leading US-based Biotech company. Recently, the compound received approval as 'Orphan Drug' against HCC from US FDA, which reveals the clinical relevance of evaluating its antitumor efficacy against HCC. In the present study, we report that Utt-B promotes pro-survival autophagy in hepatic cancer cells as evidenced by the increased expression of autophagy-related proteins, including LC3-II, Beclin1, ATG 5, and ATG 7, as well as a rise in the autophagic flux. Hence, we investigated whether Utt-B-induced autophagic response is complementing or contradicting its apoptotic program in HCC. Inhibition of autophagy using the pharmacological inhibitors, Bafilomycin A1(Baf A1), and 3-methyl adenine (3-MA), and the biological inhibitor, Beclin1 siRNA, significantly enhances the apoptosis of hepatic cancer cells and hence the cytotoxicity induced by Utt-B. We also found increased expression of autophagy markers in Utt-B-treated xenografts derived from HCC. We further analyzed whether the antimalarial drug, Chloroquine (Cqn), a well-known autophagy inhibitor, can enhance the anticancer effect of Utt-B against HCC. We found that inhibition of autophagy using Cqn significantly enhances the antitumor efficacy of Utt-B in vitro and in vivo, in NOD SCID mice bearing HCC xenografts. Taken together, our results suggest that the antitumor effect of Utt-B against HCC can be further enhanced by blocking autophagy. Furthermore, Utt-B in combination with Cqn, a clinically approved drug, if repurposed and used in a combinatorial regimen with Utt-B, can further improve the therapeutic efficacy of Utt-B against HCC.
Collapse
Affiliation(s)
- Lekshmi R. Nath
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Mundanattu Swetha
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Vinod Vijayakurup
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Arun Kumar Thangarasu
- Chemical Sciences and Technology Division, Council for Scientific and Industrial Research (CSIR)-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Nair Hariprasad Haritha
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Anwar Shabna
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sreekumar U. Aiswarya
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Department of Biotechnology, University of Calicut, Malappuram, India
| | - Tennyson P. Rayginia
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - C. K. Keerthana
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Kalishwaralal Kalimuthu
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sankar Sundaram
- Department of Pathology, Government Medical College, Kottayam, India
| | - Ravi Shankar Lankalapalli
- Chemical Sciences and Technology Division, Council for Scientific and Industrial Research (CSIR)-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Sreekumar Pillai
- Department of Surgical Oncology, Jubilee Mission Medical College and Research Institute, Thrissur, India
| | - Rheal Towner
- Department of Pathology and Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma, United States
| | - Noah Isakov
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- *Correspondence: Ruby John Anto,
| |
Collapse
|
34
|
Autophagy-Related Chemoprotection against Sorafenib in Human Hepatocarcinoma: Role of FOXO3 Upregulation and Modulation by Regorafenib. Int J Mol Sci 2021; 22:ijms222111770. [PMID: 34769197 PMCID: PMC8583804 DOI: 10.3390/ijms222111770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
Early acquisition of sorafenib resistance is responsible for the dismal prognosis of advanced hepatocarcinoma (HCC). Autophagy, a catabolic process involved in liver homeostasis, has been associated with chemosensitivity modulation. Forkhead box O3 (FOXO3) is a transcription factor linked to HCC pathogenesis whose role on autophagy-related sorafenib resistance remains controversial. Here, we unraveled the linkage between autophagy and sorafenib resistance in HCC, focusing on the implication of FOXO3 and its potential modulation by regorafenib. We worked with two HepG2-derived sorafenib-resistant HCC in vitro models (HepG2S1 and HepG2S3) and checked HCC patient data from the UALCAN database. Resistant cells displayed an enhanced basal autophagic flux compared to HepG2, showing higher autophagolysosome content and autophagy markers levels. Pharmacological inhibition of autophagy boosted HepG2S1 and HepG2S3 apoptosis and subG1 cells, but reduced viability, indicating the cytoprotective role of autophagy. HCC samples displayed higher FOXO3 levels, being associated with shorter survival and autophagic genes expression. Consistently, chemoresistant in vitro models showed significant FOXO3 upregulation. FOXO3 knockdown suppressed autophagy and caused resistant cell death, demonstrating that overactivation of such pro-survival autophagy during sorafenib resistance is FOXO3-dependent; a cytoprotective mechanism that the second-line drug regorafenib successfully abolished. Therefore, targeting FOXO3-mediated autophagy could significantly improve the clinical efficacy of sorafenib.
Collapse
|
35
|
Nogueira LS, Vasconcelos CP, Plaça JR, Mitre GP, Bittencourt LO, Kataoka MSDS, de Oliveira EHC, Lima RR. Non-Lethal Concentration of MeHg Causes Marked Responses in the DNA Repair, Integrity, and Replication Pathways in the Exposed Human Salivary Gland Cell Line. Front Pharmacol 2021; 12:698671. [PMID: 34512333 PMCID: PMC8423918 DOI: 10.3389/fphar.2021.698671] [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] [Received: 04/21/2021] [Accepted: 07/07/2021] [Indexed: 12/02/2022] Open
Abstract
In Brazilian northern Amazon, communities are potentially exposed and vulnerable to methylmercury (MeHg) toxicity through the vast ingestion of fish. In vivo and in vitro studies demonstrated that the salivary glands as a susceptible organ to this potent environmental pollutant, reporting alterations on physiological, biochemical, and proteomic parameters. However, the alterations caused by MeHg on the gene expression of the exposed human salivary gland cells are still unknown. Therefore, the goal was to perform the transcriptome profile of the human salivary gland cell line after exposure to MeHg, using the microarray technique and posterior bioinformatics analysis. The cell exposure was performed using 2.5 µM MeHg. A previously published study demonstrated that this concentration belongs to a range of concentrations that caused biochemical and metabolic alterations in this linage. As a result, the MeHg exposure did not cause lethality in the human salivary gland cells line but was able to alter the expression of 155 genes. Downregulated genes (15) are entirety relating to the cell metabolism impairment, and according to KEGG analysis, they belong to the glycosphingolipid (GSL) biosynthesis pathway. On the other hand, most of the 140 upregulated genes were related to cell-cycle progression, DNA repair, and replication pathway, or cellular defenses through the GSH basal metabolism. These genomic changes revealed the effort to the cell to maintain physiological and genomic stability to avoid cell death, being in accordance with the nonlethality in the toxicity test. Last, the results support in-depth studies on nonlethal MeHg concentrations for biomarkers identification that interpret transcriptomics data in toxicological tests serving as an early alert of physiological changes in vitro biological models.
Collapse
Affiliation(s)
- Lygia Sega Nogueira
- Laboratory of Functional and Structural Biology, Federal University of Pará, Belém, Brazil
| | - Carolina P Vasconcelos
- Laboratory of Cell Culture and Cytogenetics, Environment Section, Evandro Chagas Institute, Ananindeua, Brazil
| | - Jessica Rodrigues Plaça
- Regional Blood Center at University Hospital of the Ribeirão Preto Medical School of University of São Paulo, Ribeirão Preto, Brazil
| | | | | | | | - Edivaldo H C de Oliveira
- Laboratory of Cell Culture and Cytogenetics, Environment Section, Evandro Chagas Institute, Ananindeua, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Federal University of Pará, Belém, Brazil
| |
Collapse
|
36
|
Xu Y, Ji Y, Li X, Ding J, Chen L, Huang Y, Wei W. URI1 suppresses irradiation-induced reactive oxygen species (ROS) by activating autophagy in hepatocellular carcinoma cells. Int J Biol Sci 2021; 17:3091-3103. [PMID: 34421352 PMCID: PMC8375238 DOI: 10.7150/ijbs.55689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 06/15/2021] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy has been extensively applied in cancer treatment. However, this treatment is ineffective in Hepatocellular carcinoma (HCC) due to lack of radiosensitivity. Unconventional prefoldin RPB5 interactor 1 (URI1) exhibits characteristics similar to those oncoproteins, which promotes survival of cancer cells. As a consequence of the irradiation, the levels of endogenous reactive oxygen species (ROS) rise. In the current study, we analyzed the role of URI1 in the control of ROS levels in HepG2 cells. Upon URI1 overexpression, HepG2 cells significantly suppressed irradiation-induced ROS, which may help cells escape from oxidative toxicity. And our data demonstrated that overexpression of URI1 not only resulted in an increase of autophagic flux, but also resulted in an further increased capacity of autophagy to eliminate ROS. It indicated that URI1 suppressed irradiation-induced ROS through activating autophagy. Moreover, URI1 activated autophagy by promoting the activities of AMP-activated protein kinase (AMPK). Results showed that overexpression of URI1 increased the phosphorylation of AMPKα at the Thr172 residue and the activated-AMPK promoted the phosphorylation of forkhead box O3 (FOXO3) at the Ser253 residue, which significantly induced autophagy. Taken together, our findings provide a mechanism that URI1 suppresses irradiation-induced ROS by activating autophagy through AMPK/FOXO3 signaling pathway. These new molecular insights will provide an important contribution to our better understanding about irradiation insensitivity of HCC.
Collapse
Affiliation(s)
- Yue Xu
- Department of Cell Biology, Institute of Bioengineering, School of Medicine, Soochow University, Suzhou 215123, China
| | - Yuan Ji
- Department of Cell Biology, Institute of Bioengineering, School of Medicine, Soochow University, Suzhou 215123, China
| | - Xiang Li
- Department of Endocrinology, Children's Hospital affiliated to Soochow University, Suzhou, 215000, China
| | - JiaZheng Ding
- Department of Cell Biology, Institute of Bioengineering, School of Medicine, Soochow University, Suzhou 215123, China
| | - LinQi Chen
- Department of Endocrinology, Children's Hospital affiliated to Soochow University, Suzhou, 215000, China
| | - YaFeng Huang
- Department of Cell Biology, Institute of Bioengineering, School of Medicine, Soochow University, Suzhou 215123, China
| | - Wenxiang Wei
- ✉ Corresponding author: Wenxiang Wei, Department of Cell Biology and Institute of Bioengineering, School of Medicine, Soochow University, Suzhou, 215123 China. 86-512-5188-0107;
| |
Collapse
|
37
|
Natural compounds modulate the autophagy with potential implication of stroke. Acta Pharm Sin B 2021; 11:1708-1720. [PMID: 34386317 PMCID: PMC8343111 DOI: 10.1016/j.apsb.2020.10.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/12/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Stroke is considered a leading cause of mortality and neurological disability, which puts a huge burden on individuals and the community. To date, effective therapy for stroke has been limited by its complex pathological mechanisms. Autophagy refers to an intracellular degrading process with the involvement of lysosomes. Autophagy plays a critical role in maintaining the homeostasis and survival of cells by eliminating damaged or non-essential cellular constituents. Increasing evidence support that autophagy protects neuronal cells from ischemic injury. However, under certain circumstances, autophagy activation induces cell death and aggravates ischemic brain injury. Diverse naturally derived compounds have been found to modulate autophagy and exert neuroprotection against stroke. In the present work, we have reviewed recent advances in naturally derived compounds that regulate autophagy and discussed their potential application in stroke treatment.
Collapse
Key Words
- AD, Alzheimer's disease
- ALS, amyotrophic lateral sclerosis
- AMPK, 5′-adenosine monophosphate-activated protein kinase
- ATF6, activating transcription factor 6
- ATG, autophagy related genes
- Autophagy
- BCL-2, B-cell lymphoma 2
- BNIP3L, BCL2/adenovirus
- COPII, coat protein complex II
- Cerebral ischemia
- ER, endoplasmic reticulum
- FOXO, forkhead box O
- FUNDC1, FUN14 domain containing 1
- GPCR, G-protein coupled receptor
- HD, Huntington's disease
- IPC, ischemic preconditioning
- IRE1, inositol-requiring enzyme 1
- JNK, c-Jun N-terminal kinase
- LAMP, lysosomal-associated membrane protein
- LC3, light chain 3
- LKB1, liver kinase B1
- Lysosomal activation
- Mitochondria
- Mitophagy
- Natural compounds
- Neurological disorders
- Neuroprotection
- OGD/R, oxygen and glucose deprivation-reperfusion
- PD, Parkinson's disease
- PERK, protein kinase R (PKR)-like endoplasmic reticulum kinase
- PI3K, phosphatidylinositol 3-kinase
- ROS, reactive oxygen species
- SQSTM1, sequestosome 1
- TFEB, transcription factor EB
- TIGAR, TP53-induced glycolysis and apoptosis regulator
- ULK, Unc-51- like kinase
- Uro-A, urolithin A
- eIF2a, eukaryotic translation-initiation factor 2
- mTOR, mechanistic target of rapamycin
- ΔΨm, mitochondrial membrane potential
Collapse
|
38
|
Mirra S, García-Arroyo R, B Domènech E, Gavaldà-Navarro A, Herrera-Úbeda C, Oliva C, Garcia-Fernàndez J, Artuch R, Villarroya F, Marfany G. CERKL, a retinal dystrophy gene, regulates mitochondrial function and dynamics in the mammalian retina. Neurobiol Dis 2021; 156:105405. [PMID: 34048907 DOI: 10.1016/j.nbd.2021.105405] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/06/2021] [Accepted: 05/21/2021] [Indexed: 11/30/2022] Open
Abstract
The retina is a highly active metabolic organ that displays a particular vulnerability to genetic and environmental factors causing stress and homeostatic imbalance. Mitochondria constitute a bioenergetic hub that coordinates stress response and cellular homeostasis, therefore structural and functional regulation of the mitochondrial dynamic network is essential for the mammalian retina. CERKL (ceramide kinase like) is a retinal degeneration gene whose mutations cause Retinitis Pigmentosa in humans, a visual disorder characterized by photoreceptors neurodegeneration and progressive vision loss. CERKL produces multiple isoforms with a dynamic subcellular localization. Here we show that a pool of CERKL isoforms localizes at mitochondria in mouse retinal ganglion cells. The depletion of CERKL levels in CerklKD/KO(knockdown/knockout) mouse retinas cause increase of autophagy, mitochondrial fragmentation, alteration of mitochondrial distribution, and dysfunction of mitochondrial-dependent bioenergetics and metabolism. Our results support CERKL as a regulator of autophagy and mitochondrial biology in the mammalian retina.
Collapse
Affiliation(s)
- Serena Mirra
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine (IBUB), Faculty of Biology, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona- Institut de Recerca Hospital Sant Joan de Déu, IBUB-IRSJD, Barcelona, Spain.
| | - Rocío García-Arroyo
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine (IBUB), Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Elena B Domènech
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine (IBUB), Faculty of Biology, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Aleix Gavaldà-Navarro
- Institut de Biomedicina de la Universitat de Barcelona- Institut de Recerca Hospital Sant Joan de Déu, IBUB-IRSJD, Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Barcelona, Spain; CIBEROBN, Instituto de Salud Carlos III, Spain
| | - Carlos Herrera-Úbeda
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine (IBUB), Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Clara Oliva
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Spain
| | - Jordi Garcia-Fernàndez
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine (IBUB), Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Rafael Artuch
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain; Clinical Biochemistry Department, Hospital Sant Joan de Déu, Spain
| | - Francesc Villarroya
- Institut de Biomedicina de la Universitat de Barcelona- Institut de Recerca Hospital Sant Joan de Déu, IBUB-IRSJD, Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Barcelona, Spain; CIBEROBN, Instituto de Salud Carlos III, Spain
| | - Gemma Marfany
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine (IBUB), Faculty of Biology, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona- Institut de Recerca Hospital Sant Joan de Déu, IBUB-IRSJD, Barcelona, Spain.
| |
Collapse
|
39
|
He M, Yan G, Wang Y, Gong R, Lei H, Yu S, He X, Li G, Du W, Ma T, Gao M, Yu M, Liu S, Xu Z, Idiiatullina E, Zagidullin N, Pavlov V, Cai B, Yuan Y, Yang L. Blue LED causes autophagic cell death in human osteosarcoma by increasing ROS generation and dephosphorylating EGFR. J Cell Mol Med 2021; 25:4962-4973. [PMID: 33960631 PMCID: PMC8178260 DOI: 10.1111/jcmm.16412] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/29/2021] [Accepted: 02/08/2021] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumour in adolescence. Lately, light‐emitting diodes (LED)‐based therapy has emerged as a new promising approach for several diseases. However, it remains unknown in human OS. Here, we found that the blue LED irradiation significantly suppressed the proliferation, migration and invasion of human OS cells, while we observed blue LED irradiation increased ROS production through increased NADPH oxidase enzymes NOX2 and NOX4, as well as decreased Catalase (CAT) expression levels. Furthermore, we revealed blue LED irradiation‐induced autophagy characterized by alterations in autophagy protein markers including Beclin‐1, LC3‐II/LC3‐I and P62. Moreover, we demonstrated an enhanced autophagic flux. The blockage of autophagy displayed a remarkable attenuation of anti‐tumour activities of blue LED irradiation. Next, ROS scavenger N‐acetyl‐L‐cysteine (NAC) and NOX inhibitor diphenyleneiodonium (DPI) blocked suppression of OS cell growth, indicating that ROS accumulation might play an essential role in blue LED‐induced autophagic OS cell death. Additionally, we observed blue LED irradiation decreased EGFR activation (phosphorylation), which in turn led to Beclin‐1 release and subsequent autophagy activation in OS cells. Analysis of EGFR colocalization with Beclin‐1 and EGFR‐immunoprecipitation (IP) assay further revealed the decreased interaction of EGFR and Beclin‐1 upon blue LED irradiation in OS cells. In addition, Beclin‐1 down‐regulation abolished the effects of blue LED irradiation on OS cells. Collectively, we concluded that blue LED irradiation exhibited anti‐tumour effects on OS by triggering ROS and EGFR/Beclin‐1‐mediated autophagy signalling pathway, representing a potential approach for human OS treatment.
Collapse
Affiliation(s)
- Mingyu He
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Gege Yan
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Yang Wang
- Department of Orthopedics, Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Rui Gong
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Hong Lei
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Shuting Yu
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Xiaoqi He
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Guanghui Li
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Weijie Du
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China.,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
| | - Tianshuai Ma
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Manqi Gao
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Meixi Yu
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Shenzhen Liu
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Zihang Xu
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Elina Idiiatullina
- Central Laboratory of Scientific Research, Bashkir State Medical University, Ufa, Russia
| | - Naufal Zagidullin
- Central Laboratory of Scientific Research, Bashkir State Medical University, Ufa, Russia
| | - Valentin Pavlov
- Central Laboratory of Scientific Research, Bashkir State Medical University, Ufa, Russia
| | - Benzhi Cai
- Department of Orthopedics, Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
| | - Ye Yuan
- Department of Orthopedics, Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
| | - Lei Yang
- Department of Orthopedics, Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
40
|
Felipe SMDS, de Freitas RM, Penha EDDS, Pacheco C, Martins DL, Alves JO, Soares PM, Loureiro ACC, Lima T, Silveira LR, Ferraz ASM, de Souza JES, Leal-Cardoso JH, Carvalho DP, Ceccatto VM. Transcriptional profile in rat muscle: down-regulation networks in acute strenuous exercise. PeerJ 2021; 9:e10500. [PMID: 33859869 PMCID: PMC8020866 DOI: 10.7717/peerj.10500] [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] [Received: 05/26/2020] [Accepted: 11/15/2020] [Indexed: 11/20/2022] Open
Abstract
Background Physical exercise is a health promotion factor regulating gene expression and causing changes in phenotype, varying according to exercise type and intensity. Acute strenuous exercise in sedentary individuals appears to induce different transcriptional networks in response to stress caused by exercise. The objective of this research was to investigate the transcriptional profile of strenuous experimental exercise. Methodology RNA-Seq was performed with Rattus norvegicus soleus muscle, submitted to strenuous physical exercise on a treadmill with an initial velocity of 0.5 km/h and increments of 0.2 km/h at every 3 min until animal exhaustion. Twenty four hours post-physical exercise, RNA-seq protocols were performed with coverage of 30 million reads per sample, 100 pb read length, paired-end, with a list of counts totaling 12816 genes. Results Eighty differentially expressed genes (61 down-regulated and 19 up-regulated) were obtained. Reactome and KEGG database searches revealed the most significant pathways, for down-regulated gene set, were: PI3K-Akt signaling pathway, RAF-MAP kinase, P2Y receptors and Signaling by Erbb2. Results suggest PI3K-AKT pathway inactivation by Hbegf, Fgf1 and Fgr3 receptor regulation, leading to inhibition of cell proliferation and increased apoptosis. Cell signaling transcription networks were found in transcriptome. Results suggest some metabolic pathways which indicate the conditioning situation of strenuous exercise induced genes encoding apoptotic and autophagy factors, indicating cellular stress. Conclusion Down-regulated networks showed cell transduction and signaling pathways, with possible inhibition of cellular proliferation and cell degeneration. These findings reveal transitory and dynamic process in cell signaling transcription networks in skeletal muscle after acute strenuous exercise.
Collapse
Affiliation(s)
| | | | | | - Christina Pacheco
- Superior Institute of Biomedic Sciences, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Danilo Lopes Martins
- Digital Metropolis Institute, Universidade Federal do Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Juliana Osório Alves
- Superior Institute of Biomedic Sciences, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Paula Matias Soares
- Superior Institute of Biomedic Sciences, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | | | - Tanes Lima
- Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Leonardo R Silveira
- Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | | | | | | | - Denise P Carvalho
- Carlos Chagas Filho Biophysics Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vania Marilande Ceccatto
- Superior Institute of Biomedic Sciences, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
41
|
Liu PW, Li CI, Huang KC, Liu CS, Chen HL, Lee CC, Chiou YY, Chen RJ. 3-MCPD and glycidol coexposure induces systemic toxicity and synergistic nephrotoxicity via NLRP3 inflammasome activation, necroptosis, and autophagic cell death. JOURNAL OF HAZARDOUS MATERIALS 2021; 405:124241. [PMID: 33187795 DOI: 10.1016/j.jhazmat.2020.124241] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/02/2020] [Accepted: 10/08/2020] [Indexed: 06/11/2023]
Abstract
3-Monochloropropane-1,2-diol (3-MCPD), 2,3-epoxy-1-propanol (glycidol), and their esters are well-known food contaminants mainly formed by the heat processing of certain refined oils and coexist in various kinds of foodstuffs. However, the combined health effect and the underlying mechanism of 3-MCPD and glycidol coexposure are not well-understood. In this study, we investigated the systemic toxicity effects and the nephrotoxicity mechanisms of 3-MCPD and glycidol coexposure with in vitro and in vivo models, and next-generation sequencing (NGS) analysis. It was found that 3-MCPD and glycidol coexposure for 28 days synergistically induced toxicity in the kidney, lung, testis, and heart in C57BL/6 mice. Kidney was the most sensitive organ to coexposure, and the coexposure had a synergistic effect on inflammation and cytotoxicity through activation of the NLRP3 inflammasome, and the induction of necroptosis, and autophagic cell death in NRK-52E cells. Moreover, the NGS results revealed the genes changes associated with nephrotoxicity, inflammation and with the broad toxicity effects induced by 3-MCPD or glycidol alone or in combination, which were consistent with the results of in vitro and in vivo models. In summary, we report for the first time of the comprehensive toxicity effects and the mechanisms caused by 3-MCPD and glycidol coexposure.
Collapse
Affiliation(s)
- Pei-Wen Liu
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chung-I Li
- Department of Statistics, College of Management, National Cheng Kung University, Tainan, Taiwan
| | - Kuo-Ching Huang
- Division of Nephrology, Department of Internal Medicine, Chi Mei Hospital, Liouying District, Tainan, Taiwan; Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chiang-Shin Liu
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Hsiu-Lin Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Chang Lee
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Research Center of Environmental Trace Toxic Substances, National Cheng Kung University, Tainan, Taiwan
| | - Yuan-Yow Chiou
- Department of Pediatrics, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Pediatric Nephrology, Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan; Institute of Clinical Medicine, Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Rong-Jane Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
42
|
Schoepf AM, Salcher S, Obexer P, Gust R. Tackling resistance in chronic myeloid leukemia: Novel cell death modulators with improved efficacy. Eur J Med Chem 2021; 216:113285. [PMID: 33662676 DOI: 10.1016/j.ejmech.2021.113285] [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: 10/10/2020] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 10/22/2022]
Abstract
The development of resistance poses a serious problem in the therapy of cancer due to the necessity of a multiple-drug and unlimited treatment of affected patients. In chronic myeloid leukemia (CML), the introduction of imatinib has revolutionized the therapy. The persistence of an untreatable cancer stem cell pool and other resistance-causing factors, however, also impede the cure of this malignancy. New therapeutic approaches are therefore essential to overcome current treatment drawbacks. In this regard, an intervention in the STAT5 signaling pathway can significantly improve drug response, as this central signaling node induces the formation of highly resistant CML cells. In the present study, we continued the design of efficient chemosensitizers derived from the partial PPARγ agonist telmisartan. The developed 2-carbonitriles or 2-carboxymethyl esters showed improved potency in sensitizing K562-resistant cells to imatinib treatment, even at concentrations, which are considered patient-relevant. At 5 μM, for instance, 2d sensitized the cells in such a manner that the resistance was fully overcome and the recovered efficacy of imatinib resulted in >76% cell death. Importantly, all compounds were non-cytotoxic per se. A transactivation experiment showed that only the carbonitriles are partial agonists of PPARγ, which does not seem to be involved in the mode of action. Yet, immunoassays revealed a suppression of the STAT5 phosphorylation status by co-application of the most active derivatives with imatinib. This mechanism consequently resulted in reduced cell proliferation and induction of cell death in resistant CML cells.
Collapse
Affiliation(s)
- Anna M Schoepf
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI - Center for Molecular Biosciences Innsbruck, University of Innsbruck, CCB - Centrum for Chemistry and Biomedicine, Innrain 80-82, 6020, Innsbruck, Austria; Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, United Kingdom
| | - Stefan Salcher
- Tyrolean Cancer Research Institute, Innrain 66, 6020, Innsbruck, Austria; Department of Internal Medicine V, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Petra Obexer
- Tyrolean Cancer Research Institute, Innrain 66, 6020, Innsbruck, Austria; Department of Pediatrics II, Medical University Innsbruck, Innrain 66, 6020, Innsbruck, Austria
| | - Ronald Gust
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI - Center for Molecular Biosciences Innsbruck, University of Innsbruck, CCB - Centrum for Chemistry and Biomedicine, Innrain 80-82, 6020, Innsbruck, Austria.
| |
Collapse
|
43
|
Farhan M, Silva M, Li S, Yan F, Fang J, Peng T, Hu J, Tsao M, Little P, Zheng W. The role of FOXOs and autophagy in cancer and metastasis-Implications in therapeutic development. Med Res Rev 2020; 40:2089-2113. [PMID: 32474970 PMCID: PMC7586888 DOI: 10.1002/med.21695] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 04/21/2020] [Accepted: 05/16/2020] [Indexed: 12/17/2022]
Abstract
Autophagy is a highly conserved intracellular degradation process that plays a crucial role in cell survival and stress reactions as well as in cancer development and metastasis. Autophagy process involves several steps including sequestration, fusion of autophagosomes with lysosomes and degradation. Forkhead box O (FOXO) transcription factors regulate the expression of genes involved in cellular metabolic activity and signaling pathways of cancer growth and metastasis. Recent evidence suggests that FOXO proteins are also involved in autophagy regulation. The relationship among FOXOs, autophagy, and cancer has been drawing attention of many who work in the field. This study summarizes the role of FOXO proteins and autophagy in cancer growth and metastasis and analyzes their potential roles in cancer disease management.
Collapse
Affiliation(s)
- Mohd Farhan
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Marta Silva
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Shuai Li
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Fengxia Yan
- Department of MedicineJinan UniversityGuangzhouChina
| | - Jiankang Fang
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Tangming Peng
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Jim Hu
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Ming‐Sound Tsao
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Peter Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of QueenslandWoolloongabbaQueenslandAustralia
| | - Wenhua Zheng
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| |
Collapse
|
44
|
Schoepf AM, Salcher S, Hohn V, Veider F, Obexer P, Gust R. Synthesis and Characterization of Telmisartan-Derived Cell Death Modulators to Circumvent Imatinib Resistance in Chronic Myeloid Leukemia. ChemMedChem 2020; 15:1067-1077. [PMID: 32298535 PMCID: PMC7318623 DOI: 10.1002/cmdc.202000092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/15/2020] [Indexed: 12/25/2022]
Abstract
New strategies to eradicate cancer stem cells in chronic myeloid leukemia (CML) include a combination of imatinib with peroxisome proliferator-activated receptor gamma (PPARγ) ligands. Recently, we identified the partial PPARγ agonist telmisartan as effective sensitizer of resistant K562 CML cells to imatinib treatment. Here, the importance of the heterocyclic core on the cell death-modulating effects of the telmisartan-derived lead 4'-((2-propyl-1H-benzo[d]imidazol-1-yl)methyl)-[1,1'-biphenyl]-2-carboxylic acid (3 b) was investigated. Inspired by the pharmacodynamics of HYL-6d and the selective PPARγ ligand VSP-51, the benzimidazole was replaced by a carbazole or an indole core. The results indicate no correlation between PPARγ activation and sensitization of resistant CML cells to imatinib. The 2-COOH derivatives of the carbazoles or indoles achieved low activity at PPARγ, while the benzimidazoles showed 60-100 % activation. Among the 2-CO2 CH3 derivatives, only the ester of the lead (2 b) slightly activated PPARγ. Sensitizing effects were further observed for this non-cytotoxic 2 b (80 % cell death), and to a lesser extent for the lead 3 b or the 5-Br-substituted ester of the benzimidazoles (5 b).
Collapse
Affiliation(s)
- Anna M. Schoepf
- Department of Pharmaceutical Chemistry Institute of Pharmacy CMBI – Center for Molecular Biosciences InnsbruckUniversity of Innsbruck, CCB – Centrum for Chemistry and BiomedicineInnrain 80/826020InnsbruckAustria
| | - Stefan Salcher
- Tyrolean Cancer Research InstituteInnrain 666020InnsbruckAustria
- Department of Internal Medicine VMedical University InnsbruckAnichstraße 356020InnsbruckAustria
| | - Verena Hohn
- Department of Pharmaceutical Chemistry Institute of Pharmacy CMBI – Center for Molecular Biosciences InnsbruckUniversity of Innsbruck, CCB – Centrum for Chemistry and BiomedicineInnrain 80/826020InnsbruckAustria
| | - Florina Veider
- Department of Pharmaceutical Chemistry Institute of Pharmacy CMBI – Center for Molecular Biosciences InnsbruckUniversity of Innsbruck, CCB – Centrum for Chemistry and BiomedicineInnrain 80/826020InnsbruckAustria
| | - Petra Obexer
- Tyrolean Cancer Research InstituteInnrain 666020InnsbruckAustria
- Department of Pediatrics IIMedical University InnsbruckInnrain 666020InnsbruckAustria
| | - Ronald Gust
- Department of Pharmaceutical Chemistry Institute of Pharmacy CMBI – Center for Molecular Biosciences InnsbruckUniversity of Innsbruck, CCB – Centrum for Chemistry and BiomedicineInnrain 80/826020InnsbruckAustria
| |
Collapse
|
45
|
Peng S, Sun Y, Luo Y, Ma S, Sun W, Tang G, Li S, Zhang N, Ren J, Xiao Y, Liu X, Zhang J, Gong Y, Xie C. MFP-FePt-GO Nanocomposites Promote Radiosensitivity of Non-Small Cell Lung Cancer Via Activating Mitochondrial-Mediated Apoptosis and Impairing DNA Damage Repair. Int J Biol Sci 2020; 16:2145-2158. [PMID: 32549761 PMCID: PMC7294941 DOI: 10.7150/ijbs.46194] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 04/25/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Recent advances in nanomedicine provided promising alternatives for tumor treatment to improve the survival and life quality of cancer patients. This study was designed to explore the insight mechanisms of the anti-tumor effects of the novel nanocomposites (NCs) MFP-FePt-GO with non-small cell lung cancer (NSCLC). Methods: A chemical co-reduction method was applied to the synthesis process of MFP-FePt-GO NCs. The chemical synthesis efficiency and morphology of the NCs were measured with spectroscope and transmission electron microscope. Colony formation assay and cell apoptosis were conducted to assess the radiosensitivity effect of NCs with radiation. Then, we detected cell mitochondrial membrane potential and reactive oxygen species (ROS) level by flow cytometry to further explore the cause of cell death. Immunofluorescence staining and Confocal were carried out to determine the DNA damage repair. A Lewis lung carcinoma animal model was used to measure safety and anti-tumor efficiency in vivo. Results: The novel NCs MFP-FePt-GO designed on a lamellar-structure magnetic graphene oxide and polyethylene glycol drug delivery system was synthesized and functionalized for co-delivery of metronidazole and 5-fluorouracil. While no severe allergies, liver and kidney damage, or drug-related deaths were observed, MFP-FePt-GO NCs promoted radiosensitivity of NSCLC cells both in vivo and in vitro. It improved the effects of radiation via activating intrinsic mitochondrial-mediated apoptosis and impairing DNA damage repair. This NCs also induced a ROS burst, which suppressed the antioxidant protein expression and induced cell apoptosis. Furthermore, MFP-FePt-GO NCs prevented NSCLC cell migration and invasion. Conclusion: MFP-FePt-GO NCs showed a synergistic anti-tumor effect with radiation to eliminate tumors. With good safety and efficacy, this novel NCs could be a potential radiosensitive agent for NSCLC patients.
Collapse
Affiliation(s)
- Shan Peng
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingming Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Luo
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shijing Ma
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenjie Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Guiliang Tang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shuying Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Nannan Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiangbo Ren
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu Xiao
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuefeng Liu
- Department of Pathology, Lombardi Comprehensive Cancer Center Georgetown University Medical School, Washington DC, USA
| | - Junhong Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
46
|
Zhang X, Wang LL, Wang B, Liu HL, Zhang J, Li YH, Wang LH. Effect of siRNA-induced Atg7 gene silencing on the sensitivity of ovarian cancer SKOV3 cells to cisplatin. Am J Transl Res 2020; 12:2052-2061. [PMID: 32509199 PMCID: PMC7269981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 04/22/2020] [Indexed: 06/11/2023]
Abstract
Ovarian cancer is one of the most common types of gynecological malignant tumors. A proclivity for, or the development of chemoresistance severely affects treatment efficacy for ovarian cancer. Herein we found that as concentrations of cisplatin (DDP) used in SKOV3 cells increased, expression of intracellular reactive oxygen species (ROS) increased as did amounts of proteins of Beclin-1 and Autophagy-Related Gene 7 (Atg7) whereas in contrast, expression of P62 protein decreased gradually. Expression of Atg7 protein in SKOV3 cells in the siRNA-Atg7 transfection treatment group was significantly reduced compared to the negative control group. Post-application of DDP treatments, the apoptotic ratio of SKOV3 cells in the siRNA-Atg7 transfection group increased, and the cell survival rate decreased to a level significantly lower than in the negative control group. Cellular morphological analyses revealed remarkably decreased measures of cell density, as well as shrunk, deformed, and rounded cells with unclear boundaries, and revealed a decreased measures of mitochondrial membrane potential. Taken together, autophagy may be involved in the dynamics and mechanistics underlying DDP resistance in ovarian cancer SKOV3 cells. Thus, inhibition of autophagy through down-regulation expression of Atg7 may have beneficially enhanced the sensitivity of SKOV3 cells to DDP-based chemotherapy which could help improve treatment outcomes for patients afflicted by ovarian cancer.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Oncological Gynecology, First Affiliated Hospital of Bengbu Medical College Bengbu 233000, China
| | - Liang-Liang Wang
- Department of Oncological Gynecology, First Affiliated Hospital of Bengbu Medical College Bengbu 233000, China
| | - Beibei Wang
- Department of Oncological Gynecology, First Affiliated Hospital of Bengbu Medical College Bengbu 233000, China
| | - Hong-Li Liu
- Department of Oncological Gynecology, First Affiliated Hospital of Bengbu Medical College Bengbu 233000, China
| | - Jing Zhang
- Department of Oncological Gynecology, First Affiliated Hospital of Bengbu Medical College Bengbu 233000, China
| | - Yan-Hua Li
- Department of Oncological Gynecology, First Affiliated Hospital of Bengbu Medical College Bengbu 233000, China
| | - Li-Hua Wang
- Department of Oncological Gynecology, First Affiliated Hospital of Bengbu Medical College Bengbu 233000, China
| |
Collapse
|
47
|
Identification and development of non-cytotoxic cell death modulators: Impact of sartans and derivatives on PPARγ activation and on growth of imatinib-resistant chronic myelogenous leukemia cells. Eur J Med Chem 2020; 195:112258. [PMID: 32272420 DOI: 10.1016/j.ejmech.2020.112258] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/12/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022]
Abstract
4'-((2-Propyl-1H-benzo[d]imidazol-1-yl)methyl)-[1,1'-biphenyl]-2-carboxylic acid derived from telmisartan was identified as lead for the design of cell death modulators. In this study, we evaluated the efficacy of telmisartan itself and other sartans in combination with imatinib against K562-resistant cells. The findings were directly used to further optimize the lead structure. Telmisartan and candesartan cilexetil represented the most effective sartans, thus the influence of carboxyl/methyl carboxylate groups at positions 7 (compounds 6, 7) or 4 (compounds 12-14) at the benzimidazole core was studied. Additionally, according to the results of a former structure-activity study, telmisartan was transformed to the related amide (1). Telmisartan amide 1, as well as the esters 6 and 12 markedly sensitized the resistant CML cells to imatinib treatment. Correlation with their potency to activate PPARγ is not given. Candesartan cilexetil, telmisartan and 1 showed the profile of partial agonists at PPARγ with EC50 values of 4.2, 4.3 and 9.1 μM, respectively, while 6 and 12 caused only marginal intrinsic activation at 10 μM (Amax = 22% and 13%). However, the repression of the STAT5 phosphorylation relates with the possibility to sensitize K562-resistant CML cells to imatinib treatment. It is worth mentioning that all compounds were per se non-cytotoxic at relevant concentrations.
Collapse
|
48
|
Liraglutide provides neuroprotection by regulating autophagy through the AMPK-FOXO3 signaling pathway in a spinal contusion injury rat model. Neurosci Lett 2020; 720:134747. [DOI: 10.1016/j.neulet.2020.134747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 11/23/2019] [Accepted: 01/06/2020] [Indexed: 12/25/2022]
|
49
|
The Expression of Decidual Protein Induced by Progesterone (DEPP) is Controlled by Three Distal Consensus Hypoxia Responsive Element (HRE) in Hypoxic Retinal Epithelial Cells. Genes (Basel) 2020; 11:genes11010111. [PMID: 31963726 PMCID: PMC7016973 DOI: 10.3390/genes11010111] [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] [Received: 11/19/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 11/16/2022] Open
Abstract
Hypoxia affects the development and/or progression of several retinopathies. Decidual protein induced by progesterone (DEPP) has been identified as a hypoxia-responsive gene that may be part of cellular pathways such as autophagy and connected to retinal diseases. To increase our understanding of DEPP regulation in the eye, we defined its expression pattern in mouse and human retina and retinal pigment epithelium (RPE). Interestingly, DEPP expression was increased in an age-dependent way in the central human RPE. We showed that DEPP was regulated by hypoxia in the mouse retina and eyecup and that this regulation was controlled by hypoxia-inducible transcription factors 1 and 2 (HIF1 and HIF2). Furthermore, we identified three hypoxia response elements (HREs) about 3.5 kb proximal to the transcriptional start site that were responsible for hypoxic induction of DEPP in a human RPE cell line. Comparative genomics analysis suggested that one of the three HREs resides in a highly conserved genomic region. Collectively, we defined the molecular elements controlling hypoxic induction of DEPP in an RPE cell line, and provided evidence for an enrichment of DEPP in the aged RPE of human donors. This makes DEPP an interesting gene to study with respect to aging and age-related retinal pathologies.
Collapse
|
50
|
Mármol-Sánchez E, Ramayo-Caldas Y, Quintanilla R, Cardoso TF, González-Prendes R, Tibau J, Amills M. Co-expression network analysis predicts a key role of microRNAs in the adaptation of the porcine skeletal muscle to nutrient supply. J Anim Sci Biotechnol 2020; 11:10. [PMID: 31969983 PMCID: PMC6966835 DOI: 10.1186/s40104-019-0412-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/04/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The role of non-coding RNAs in the porcine muscle metabolism is poorly understood, with few studies investigating their expression patterns in response to nutrient supply. Therefore, we aimed to investigate the changes in microRNAs (miRNAs), long intergenic non-coding RNAs (lincRNAs) and mRNAs muscle expression before and after food intake. RESULTS We measured the miRNA, lincRNA and mRNA expression levels in the gluteus medius muscle of 12 gilts in a fasting condition (AL-T0) and 24 gilts fed ad libitum during either 5 h. (AL-T1, N = 12) or 7 h. (AL-T2, N = 12) prior to slaughter. The small RNA fraction was extracted from muscle samples retrieved from the 36 gilts and sequenced, whereas lincRNA and mRNA expression data were already available. In terms of mean and variance, the expression profiles of miRNAs and lincRNAs in the porcine muscle were quite different than those of mRNAs. Food intake induced the differential expression of 149 (AL-T0/AL-T1) and 435 (AL-T0/AL-T2) mRNAs, 6 (AL-T0/AL-T1) and 28 (AL-T0/AL-T2) miRNAs and none lincRNAs, while the number of differentially dispersed genes was much lower. Among the set of differentially expressed miRNAs, we identified ssc-miR-148a-3p, ssc-miR-22-3p and ssc-miR-1, which play key roles in the regulation of glucose and lipid metabolism. Besides, co-expression network analyses revealed several miRNAs that putatively interact with mRNAs playing key metabolic roles and that also showed differential expression before and after feeding. One case example was represented by seven miRNAs (ssc-miR-148a-3p, ssc-miR-151-3p, ssc-miR-30a-3p, ssc-miR-30e-3p, ssc-miR-421-5p, ssc-miR-493-5p and ssc-miR-503) which putatively interact with the PDK4 mRNA, one of the master regulators of glucose utilization and fatty acid oxidation. CONCLUSIONS As a whole, our results evidence that microRNAs are likely to play an important role in the porcine skeletal muscle metabolic adaptation to nutrient availability.
Collapse
Affiliation(s)
- Emilio Mármol-Sánchez
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Yuliaxis Ramayo-Caldas
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - Raquel Quintanilla
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - Tainã Figueiredo Cardoso
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Present address: Embrapa Pecuária Sudeste, Empresa Brasileira de Pesquisa Agropecuária (EMBRAPA), São Carlos, SP 13560-970 Brazil
| | - Rayner González-Prendes
- Department of Animal Science, Universitat de Lleida - Agrotecnio Center, 25198 Lleida, Spain
| | - Joan Tibau
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - Marcel Amills
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| |
Collapse
|