1
|
Kriukov K, Schneider D, Zeck S, Hahn L, Hofmann F, Altmann S, Luxenhofer R, Ebert R. Assessment of the viability and mechanoresponsiveness of hMSC-TERT printed with bioinert, thermoresponsive hydrogels. Sci Rep 2025; 15:12257. [PMID: 40210996 PMCID: PMC11986050 DOI: 10.1038/s41598-025-97196-9] [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: 11/27/2024] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
During three-dimensional (3D) bioprinting, the integration of living cells into hydrogel matrices results in complex biophysicochemical interactions between viscosity, shear stress, and temperature, critically influencing the structural and functional integrity of the resulting constructs. This study delves into the short-term biological ramifications of 3D extrusion printing of telomerase-immortalized human mesenchymal stromal cells (hMSC-TERT) embedded in bioinert hydrogels. Pluronic F127 and custom-synthesized poly(2-methyl-2-oxazoline)-block-poly(2-n-propyl-2-oxazine) (POx/POzi) are synthetic, block copolymers that create shear-thinning, physically crosslinked hydrogels that were used for this study. The rheological properties of the cell-free hydrogels and cell-laden bioinks were examined, revealing that they exhibited comparable behavior. Contrary to the original hypotheses, a key finding of this research is the reduction in cell viability (up to 50%) within 24 h post-printing, a trend consistently observed across varying initial conditions. The relative expression levels of the mechanoresponsive genes FOS and PTGS2 were increased, partly due to the suspension and incubation of the cells in both hydrogels. Only FOS was significantly upregulated in some cases because of the printing process after 2 and 4 h of incubation. These insights highlight the potential of using POx/POzi hydrogel as a matrix in 3D bioprinting, particularly for depositing hMSC-TERT into structures with vasculature-mimicking scaffolds or scaffolds designed for bone regeneration.
Collapse
Affiliation(s)
- Kirill Kriukov
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig Haus, University of Würzburg, Friedrich-Bergius-Ring 15, 97076, Würzburg, Germany
| | - Doris Schneider
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig Haus, University of Würzburg, Friedrich-Bergius-Ring 15, 97076, Würzburg, Germany
| | - Sabine Zeck
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig Haus, University of Würzburg, Friedrich-Bergius-Ring 15, 97076, Würzburg, Germany
| | - Lukas Hahn
- Institute for Functional Materials and Biofabrication, University of Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - Florian Hofmann
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Stephan Altmann
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig Haus, University of Würzburg, Friedrich-Bergius-Ring 15, 97076, Würzburg, Germany
| | - Robert Luxenhofer
- Department of Chemistry and Helsinki Institute of Sustainability Science, Faculty of Science, University of Helsinki, PB 55, Helsinki, 00014, Finland.
- Institute for Functional Materials and Biofabrication, University of Würzburg, Röntgenring 11, 97070, Würzburg, Germany.
| | - Regina Ebert
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig Haus, University of Würzburg, Friedrich-Bergius-Ring 15, 97076, Würzburg, Germany.
| |
Collapse
|
2
|
Qin X, Zhou K, Dong L, Yang L, Li W, Chen Z, Shen C, Han L, Li Y, Chan AK, Pokharel SP, Qing Y, Chen M, Wang K, Leung K, Sau L, Chen CW, Deng X, Su R, Chen J. CRISPR screening reveals ZNF217 as a vulnerability in high-risk B-cell acute lymphoblastic leukemia. Theranostics 2025; 15:3234-3256. [PMID: 40093906 PMCID: PMC11905140 DOI: 10.7150/thno.100295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 01/08/2025] [Indexed: 03/19/2025] Open
Abstract
Rationale: Despite substantial advancement in the treatment of B-cell acute lymphoblastic leukemia (B-ALL), it remains a leading cause of cancer mortality in children due to the high relapse rate. Moreover, the long-term survival rates for adult B-ALL patients are still less than 40%. The B-ALL patients carrying MLL rearrangements or BCR-ABL fusion represent high-risk B-ALL subtypes that face particularly dismal prognoses. This study aims to identify innovative therapeutic vulnerability for high-risk B-ALL. Methods: The CRISPR-Cas9 screen was conducted to pinpoint genes essential for high-risk B-ALL cell survival/growth. Both in vitro and in vivo models were then employed to investigate the pathological role of ZNF217 in high-risk B-ALL. To characterize the downstream functionally essential targets of ZNF217, we performed RNA-seq and CUT&RUN-seq, followed by integrative bioinformatics analysis and experimental validation. Results: Through the focused CRISPR-Cas9 screening, ZNF217 emerged as the most essential gene for the cell survival/growth of B-ALL driven by MLL rearrangement or BCR-ABL. Through in vitro gain- and loss-of-function assays, we demonstrated that ZNF217 is indeed required for B-ALL cell survival/growth. Moreover, we established the B-ALL xenograft model and patient-derived xenograft (PDX) model and demonstrated that ZNF217 depletion significantly suppressed B-ALL progression and substantially extended the survival of recipient mice. Through integrative multiple-omics analysis, we elucidated that ZNF217 exerts its oncogenic role in B-ALL through both CoREST-dependent and CoREST-independent mechanisms. Furthermore, we characterized FOS as a functionally essential downstream target of ZNF217, and ZNF217 inhibited FOS expression in a CoREST-independent manner. Conclusions: Our findings highlight ZNF217 as a promising therapeutic target for the treatment of high-risk B-ALL, such as those carrying MLL-rearrangements or BCR-ABL fusion.
Collapse
Affiliation(s)
- Xi Qin
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Keren Zhou
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Lei Dong
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Division of Epigenetic and Transcriptional Engineering, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Wei Li
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Zhenhua Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Chao Shen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Li Han
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Yangchan Li
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Anthony K.N. Chan
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Division of Epigenetic and Transcriptional Engineering, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Sheela Pangeni Pokharel
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Division of Epigenetic and Transcriptional Engineering, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Ying Qing
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Meiling Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Kitty Wang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Keith Leung
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Lillian Sau
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Chun-Wei Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA 91010, USA
- Division of Epigenetic and Transcriptional Engineering, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Xiaolan Deng
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA 91010, USA
| | - Rui Su
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA 91010, USA
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA 91010, USA
- Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
3
|
Running L, Cristobal JR, Karageorgiou C, Camdzic M, Aguilar JMN, Gokcumen O, Aga DS, Atilla-Gokcumen GE. Investigating the Mechanism of Neurotoxic Effects of PFAS in Differentiated Neuronal Cells through Transcriptomics and Lipidomics Analysis. ACS Chem Neurosci 2024; 15:4568-4579. [PMID: 39603830 DOI: 10.1021/acschemneuro.4c00652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Per- and polyfluorinated alkyl substances (PFAS) are pervasive environmental contaminants that bioaccumulate in tissues and pose risks to human health. Increasing evidence links PFAS to neurodegenerative and behavioral disorders, yet the underlying mechanisms of their effects on neuronal function remain largely unexplored. In this study, we utilized SH-SY5Y neuroblastoma cells, differentiated into neuronal-like cells, to investigate the impact of six PFAS compounds─perfluorooctanoic acid (PFOA), perfluorooctanesulfonic acid (PFOS), perfluorodecanoic acid (PFDA), perfluorodecanesulfonic acid (PFDS), 8:2 fluorotelomer sulfonate (8:2 FTS), and 8:2 fluorotelomer alcohol (8:2 FTOH)─on neuronal health. Following a 30 μM exposure for 24 h, PFAS accumulation ranged from 40-6500 ng/mg of protein. Transcriptomic analysis revealed 721 differentially expressed genes (DEGs) across treatments (padj < 0.05), with 11 DEGs shared among all PFAS exposures, indicating potential biomarkers for neuronal PFAS toxicity. PFOA-treated cells showed downregulation of genes involved in synaptic growth and neural function, while PFOS, PFDS, 8:2 FTS, and 8:2 FTOH exposures resulted in the upregulation of genes related to hypoxia response and amino acid metabolism. Lipidomic profiling further demonstrated significant increases in fatty acid levels with PFDA, PFDS, and 8:2 FTS and depletion of triacylglycerols with 8:2 FTOH treatments. These findings suggest that the neurotoxic effects of PFAS are structurally dependent, offering insights into the molecular processes that may drive PFAS-induced neuronal dysfunction.
Collapse
Affiliation(s)
- Logan Running
- Department of Chemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, New York 14260, United States
| | - Judith R Cristobal
- Department of Chemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, New York 14260, United States
- RENEW Institute, University at Buffalo, The State University of New York (SUNY), Buffalo, New York 14260, United States
| | - Charikleia Karageorgiou
- Department of Biological Sciences, University at Buffalo, The State University of New York (SUNY), Buffalo, New York 14260, United States
| | - Michelle Camdzic
- Department of Chemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, New York 14260, United States
| | - John Michael N Aguilar
- Department of Chemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, New York 14260, United States
| | - Omer Gokcumen
- Department of Biological Sciences, University at Buffalo, The State University of New York (SUNY), Buffalo, New York 14260, United States
| | - Diana S Aga
- Department of Chemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, New York 14260, United States
- RENEW Institute, University at Buffalo, The State University of New York (SUNY), Buffalo, New York 14260, United States
| | - G Ekin Atilla-Gokcumen
- Department of Chemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, New York 14260, United States
| |
Collapse
|
4
|
Duan X, Wan JMF, Yu ACH. The molecular impact of sonoporation: A transcriptomic analysis of gene regulation profile. ULTRASONICS SONOCHEMISTRY 2024; 111:107077. [PMID: 39368882 PMCID: PMC11600025 DOI: 10.1016/j.ultsonch.2024.107077] [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: 05/02/2024] [Revised: 08/17/2024] [Accepted: 09/17/2024] [Indexed: 10/07/2024]
Abstract
Sonoporation has long been known to disrupt intracellular signaling, yet the involved molecules and pathways have not been identified with clarity. In this study, we employed whole transcriptome shotgun sequencing (RNA-seq) to profile sonoporation-induced gene responses after membrane resealing has taken place. Sonoporation was achieved by microbubble-mediated ultrasound (MB-US) exposure in the form of 1 MHz ultrasound pulsing (0.50 MPa peak negative pressure, 10 % duty cycle, 30 s exposure period) in the presence of microbubbles (1:1 cell-to-bubble ratio). Using propidium iodide (PI) and calcein respectively as cell viability and cytoplasmic uptake labels, post-exposure flow cytometry was performed to identify three viable cell populations: 1) unsonoporated cells, 2) sonoporated cells with low uptake, and 3) sonoporated cells with high uptake. Fluorescence-activated cell sorting was then conducted to separate the different groups followed by RNA-seq analysis of the gene expressions in each group of cells. We found that sonoporated cells with low or high calcein uptake showed high similarity in the gene responses, including the activation of multiple heat shock protein (HSP) genes and immediate early response genes mediating apoptosis and transcriptional regulation. In contrast, unsonoporated cells exhibited a more extensive gene expression alteration that included the activation of more HSP genes and the upregulation of diverse apoptotic mediators. Four oxidative stress-related and three immune-related genes were also differentially expressed in unsonoporated cells. Our results provided new information for understanding the intracellular mobilization in response to sonoporation at the molecular level, including the identification of new molecules in the sonoporation-induced apoptosis regulatory network. Our data also shed light on the innovative therapeutic strategy which could potentially leverage the responses of viable unsonoporated cells as a synergistic effector in the microenvironment to favor tumor treatment.
Collapse
Affiliation(s)
- Xinxing Duan
- Schlegel Research Institute for Aging and Department of Electrical & Computer Engineering, University of Waterloo, Waterloo, ON N2L3G1, Canada; School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Jennifer M F Wan
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Alfred C H Yu
- Schlegel Research Institute for Aging and Department of Electrical & Computer Engineering, University of Waterloo, Waterloo, ON N2L3G1, Canada.
| |
Collapse
|
5
|
Zhang P, Li H, Gong H, Tian Y, Chen F, Li X, Xie C, Tu C, Qian S, Tan Y, Liu Q, Zhang B. c-Myc-XRCC2-FOS axis promotes the proliferation and the resistance to Doxorubicin of NSCLC. Biomed Pharmacother 2024; 179:117315. [PMID: 39153434 DOI: 10.1016/j.biopha.2024.117315] [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: 06/30/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024] Open
Abstract
Lung cancer represents one of the most prevalent malignant neoplasms, commanding an alarming incidence and mortality rate globally. Non-small cell lung cancer (NSCLC), constituting approximately 80 %-90 % of all lung cancer cases, is the predominant pathological manifestation of this disease, with a disconcerting 5-year survival rate scarcely reaching 10 %. Extensive prior investigations have elucidated that the aberrant expression of X-ray repair cross-complementing gene 2 (XRCC2), a critical meiotic gene intricately involved in the DNA damage repair process, is intimately associated with tumorigenesis. Nevertheless, the precise roles and underlying mechanistic pathways of XRCC2 in NSCLC remain largely elusive. In the present study, we discerned an overexpression of XRCC2 within NSCLC patient tissues, particularly in high-grade samples, when juxtaposed with normal tissues. Targeted knockdown of XRCC2 notably impeded the proliferation of NSCLC both in vitro and in vivo. Comprehensive RNA sequencing and flow rescue assays unveiled that XRCC2 augments the proliferation of NSCLC cells through the down-regulation of FOS expression. Moreover, the c-Myc gene was definitively identified as an XRCC2 transcriptional factor by means of chromatin immunoprecipitation (ChIP) and luciferase reporter assays, whereby pharmacological attenuation of c-Myc expression, in conjunction with Doxorubicin, synergistically curtailed NSCLC cell growth both in vitro and in vivo. Collectively, our findings proffer critical insights into the novel c-Myc-XRCC2-FOS axis in promoting both proliferation and resistance to Doxorubicin in NSCLC cells, thereby extending a promising avenue for potential new diagnostic strategies and therapeutic interventions in NSCLC.
Collapse
Affiliation(s)
- Peihe Zhang
- Department of Hepatobiliary and Intestinal Surgery of Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha 410013, China
| | - Hui Li
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Han Gong
- Department of Hepatobiliary and Intestinal Surgery of Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yuxuan Tian
- Department of Hepatobiliary and Intestinal Surgery of Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha 410013, China
| | - Fuxin Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha 410013, China
| | - Xiang Li
- Department of Pathology, Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Chunbo Xie
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China
| | - Chaofeng Tu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China; Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China
| | - Siyi Qian
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha 410013, China
| | - Yueqiu Tan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China; Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China; College of Life Sciences, Hunan Normal University, Changsha, China
| | - Qiang Liu
- Department of Hepatobiliary and Intestinal Surgery of Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.
| | - Bin Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha 410013, China.
| |
Collapse
|
6
|
Wang S, Yu H, Guo P, Feng L, Li Z. C-FOS inhibition promotes pancreatic cancer cell ferroptosis by transcriptionally regulating the expression of SLC7A11. Funct Integr Genomics 2024; 24:163. [PMID: 39292359 DOI: 10.1007/s10142-024-01429-5] [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: 06/05/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024]
Abstract
Cellular proto-oncogene C-Fos forms the AP-1 transcription factor by dimerizing with proto-oncogene c-Jun; this factor upregulates the transcription of genes associated with different malignancies. However, its functions in pancreatic adenocarcinoma (PAAD) remain poorly understood. In this study, the c-Fos was increased in PAAD cells and tissues through bioinformatic analysis, RT-PCR, and WB. In two PAAD cell lines, PANC-1 and BxPC-3, we performed c-Fos knockdown studies using short hairpin RNA (shRNA). Functional analysis indicated that c-Fos depletion in PAAD cells inhibits cell proliferation and promotes ferroptosis. Chromatin Immunoprecipitation (ChIP) and Dual-luciferase experiments showed that c-Fos coupled to the promoter region of SLC7A11 stimulated its transcription, providing mechanistic insight into the process. Moreover, SLC7A11 blocked the decline of proliferation and ferroptosis by c-Fos knockdown in PAAD cells. Furthermore, a xenograft nude mouse model was established to study the impact of c-Fos on tumorigenesis in vivo. Depletion of c-Fos could suppress PC tumor growth and the expressions of SLC7A11, ki-67, and 4HNE, but overexpression of SLC7A11 reversed this process. In summary, our investigation has shown that c-Fos acts as a transcriptional regulator of SLC7A11, which may enhance tumour growth in pancreatic cancer by inhibiting ferroptosis. These results indicate that c-Fos might be a promising target for treating ferroptosis in PAAD.
Collapse
Affiliation(s)
- Shuangjia Wang
- Department of Hepatobiliary Pancreatic Vascular Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No 55 Zhenhai Road, Xiamen, 361003, Fujian, China
| | - Hao Yu
- Department of Hepatobiliary Pancreatic Vascular Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No 55 Zhenhai Road, Xiamen, 361003, Fujian, China
| | - Ping Guo
- Department of Hepatobiliary Pancreatic Vascular Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No 55 Zhenhai Road, Xiamen, 361003, Fujian, China
| | - Liuxing Feng
- Department of Hepatobiliary Pancreatic Vascular Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No 55 Zhenhai Road, Xiamen, 361003, Fujian, China
| | - Zhimin Li
- Department of Hepatobiliary Pancreatic Vascular Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No 55 Zhenhai Road, Xiamen, 361003, Fujian, China.
| |
Collapse
|
7
|
Kew C, Prieto-Garcia C, Bhattacharya A, Tietgen M, MacNair CR, Carfrae LA, Mello-Vieira J, Klatt S, Cheng YL, Rathore R, Gradhand E, Fleming I, Tan MW, Göttig S, Kempf VAJ, Dikic I. The aryl hydrocarbon receptor and FOS mediate cytotoxicity induced by Acinetobacter baumannii. Nat Commun 2024; 15:7939. [PMID: 39261458 PMCID: PMC11390868 DOI: 10.1038/s41467-024-52118-7] [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: 03/07/2023] [Accepted: 08/27/2024] [Indexed: 09/13/2024] Open
Abstract
Acinetobacter baumannii is a pathogenic and multidrug-resistant Gram-negative bacterium that causes severe nosocomial infections. To better understand the mechanism of pathogenesis, we compare the proteomes of uninfected and infected human cells, revealing that transcription factor FOS is the host protein most strongly induced by A. baumannii infection. Pharmacological inhibition of FOS reduces the cytotoxicity of A. baumannii in cell-based models, and similar results are also observed in a mouse infection model. A. baumannii outer membrane vesicles (OMVs) are shown to activate the aryl hydrocarbon receptor (AHR) of host cells by inducing the host enzyme tryptophan-2,3-dioxygenase (TDO), producing the ligand kynurenine, which binds AHR. Following ligand binding, AHR is a direct transcriptional activator of the FOS gene. We propose that A. baumannii infection impacts the host tryptophan metabolism and promotes AHR- and FOS-mediated cytotoxicity of infected cells.
Collapse
Affiliation(s)
- Chun Kew
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
| | - Cristian Prieto-Garcia
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Anshu Bhattacharya
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
| | - Manuela Tietgen
- Institute for Medical Microbiology and Infection Control, Hospital of the Goethe University, Frankfurt, Germany
- University Center of Competence for Infection Control of the State of Hesse, Frankfurt, Germany
| | - Craig R MacNair
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Lindsey A Carfrae
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - João Mello-Vieira
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
| | - Stephan Klatt
- Institute for Vascular Signalling, Department of Molecular Medicine, CPI, Goethe University, Frankfurt, Germany
| | - Yi-Lin Cheng
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Rajeshwari Rathore
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Elise Gradhand
- Department of Pathology, Dr. Senckenberg Institute of Pathology, Goethe University, Frankfurt, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Department of Molecular Medicine, CPI, Goethe University, Frankfurt, Germany
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Stephan Göttig
- Institute for Medical Microbiology and Infection Control, Hospital of the Goethe University, Frankfurt, Germany
| | - Volkhard A J Kempf
- Institute for Medical Microbiology and Infection Control, Hospital of the Goethe University, Frankfurt, Germany
| | - Ivan Dikic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany.
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany.
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Branch Translational Medicine and Pharmacology, Frankfurt, Germany.
- Max Planck Institute of Biophysics, Frankfurt, Germany.
| |
Collapse
|
8
|
Wang Y, Yang S, Han B, Du X, Sun H, Du Y, Liu Y, Lu P, Di J, Luu LDW, Lv X, Hu S, Wang L, Jiang R. Single-cell landscape revealed immune characteristics associated with disease phases in brucellosis patients. IMETA 2024; 3:e226. [PMID: 39135683 PMCID: PMC11316929 DOI: 10.1002/imt2.226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 08/15/2024]
Abstract
A comprehensive immune landscape for Brucella infection is crucial for developing new treatments for brucellosis. Here, we utilized single-cell RNA sequencing (scRNA-seq) of 290,369 cells from 35 individuals, including 29 brucellosis patients from acute (n = 10), sub-acute (n = 9), and chronic (n = 10) phases as well as six healthy donors. Enzyme-linked immunosorbent assays were applied for validation within this cohort. Brucella infection caused a significant change in the composition of peripheral immune cells and inflammation was a key feature of brucellosis. Acute patients are characterized by potential cytokine storms resulting from systemic upregulation of S100A8/A9, primarily due to classical monocytes. Cytokine storm may be mediated by activating S100A8/A9-TLR4-MyD88 signaling pathway. Moreover, monocytic myeloid-derived suppressor cells were the probable contributors to immune paralysis in acute patients. Chronic patients are characterized by a dysregulated Th1 response, marked by reduced expression of IFN-γ and Th1 signatures as well as a high exhausted state. Additionally, Brucella infection can suppress apoptosis in myeloid cells (e.g., mDCs, classical monocytes), inhibit antigen presentation in professional antigen-presenting cells (APCs; e.g., mDC) and nonprofessional APCs (e.g., monocytes), and induce exhaustion in CD8+ T/NK cells, potentially resulting in the establishment of chronic infection. Overall, our study systemically deciphered the coordinated immune responses of Brucella at different phases of the infection, which facilitated a full understanding of the immunopathogenesis of brucellosis and may aid the development of new effective therapeutic strategies, especially for those with chronic infection.
Collapse
Affiliation(s)
- Yi Wang
- Experimental Research Center, Capital Institute of PediatricsBeijingChina
| | - Siyuan Yang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan HospitalCapital Medical UniversityBeijingChina
- Beijing Institute of Infectious DiseasesBeijingChina
- National Center for Infectious Diseases, Beijing Ditan HospitalCapital Medical UniversityBeijingChina
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious DiseasesBeijingChina
| | - Bing Han
- Clinical and Research Center of Infectious Diseases, Beijing Ditan HospitalCapital Medical UniversityBeijingChina
- Beijing Quality Control and Improvement Center of Infectious DiseaseBeijingChina
| | - Xiufang Du
- The Department of Infectious DiseasesThe Third People's Hospital of Linfen CityLinfenShanxiChina
| | - Huali Sun
- Department of Infectious DiseasesThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Yufeng Du
- The Department of Infectious DiseasesThe Third People's Hospital of Linfen CityLinfenShanxiChina
| | - Yinli Liu
- The Department of Infectious DiseasesThe Third People's Hospital of Linfen CityLinfenShanxiChina
| | - Panpan Lu
- The Department of Infectious DiseasesThe Third People's Hospital of Linfen CityLinfenShanxiChina
| | - Jinyu Di
- Department of Clinical LaboratoryThe Third People's Hospital of Lifen CityLinfenShanxiChina
| | | | - Xiao Lv
- Department of Clinical LaboratoryThe Third People's Hospital of Lifen CityLinfenShanxiChina
| | - Songnian Hu
- State Key Laboratory of Microbial Resources, Institute of MicrobiologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Linghang Wang
- National Center for Infectious Diseases, Beijing Ditan HospitalCapital Medical UniversityBeijingChina
| | - Rongmeng Jiang
- National Center for Infectious Diseases, Beijing Ditan HospitalCapital Medical UniversityBeijingChina
- Beijing Quality Control and Improvement Center of Infectious DiseaseBeijingChina
| |
Collapse
|
9
|
Xie N, Wang F, Chen D, Zhou J, Xu J, Qu F. Immune dysfunction mediated by the competitive endogenous RNA network in fetal side placental tissue of polycystic ovary syndrome. PLoS One 2024; 19:e0300461. [PMID: 38512862 PMCID: PMC10956758 DOI: 10.1371/journal.pone.0300461] [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: 11/29/2023] [Accepted: 02/27/2024] [Indexed: 03/23/2024] Open
Abstract
Polycystic ovary syndrome (PCOS), a common endocrine and metabolic disorder affecting women in their reproductive years. Emerging evidence suggests that the maternal-fetal immune system is crucial for proper pregnancy. However, whether immune function is altered at the end of pregnancy in PCOS women and the underlying molecular mechanisms is currently unexplored. Herein, the basic maternal immune system was investigated (n = 136 in the control group; n = 103 in the PCOS group), and whole-transcriptome sequencing was carried out to quantify the mRNAs, miRNAs, and lncRNAs expression levels in fetal side placental tissue of women with PCOS. GO, KEGG, and GSEA analysis were employed for functional enrichment analysis. The process of identifying hub genes was conducted utilizing the protein-protein interaction network. CIBERSORT and Connectivity Map were deployed to determine immune cell infiltration and predict potential drugs, respectively. A network of mRNA-miRNA-lncRNA was constructed and then validated by qRT-PCR. First, red blood cell count, neutrophil count, lymphocyte count, hypersensitive C-reactive protein, and procalcitonin were significantly elevated, while placental growth factor was hindered in PCOS women. We identified 308 DEmRNAs, 77 DEmiRNAs, and 332 DElncRNAs in PCOS samples. Functional enrichment analysis revealed that there were significant changes observed in terms of the immune system, especially the chemokine pathway. Eight genes, including FOS, JUN, EGR1, CXCL10, CXCR1, CXCR2, CXCL11, and CXCL8, were considered as hub genes. Furthermore, the degree of infiltration of neutrophils was dramatically decreased in PCOS tissues. In total, 57 ceRNA events were finally obtained, and immune-related ceRNA networks were validated. Some potential drug candidates, such as enalapril and RS-100329, could have a function in PCOS therapy. This study represents the inaugural attempt to evaluate the immune system at the end of pregnancy and placental ceRNA networks in PCOS, indicating alterations in the chemokine pathway, which may impact fetal and placental growth, and provides new therapy targets.
Collapse
Affiliation(s)
- Ningning Xie
- Department of Obstetrics and Gynecology, International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Fangfang Wang
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Danqing Chen
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jue Zhou
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, China
| | - Jian Xu
- Department of Obstetrics and Gynecology, International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Fan Qu
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
Wang Q, Deng J, Jiang Y, Wang Z, Xia B, Chen T. A medaka gonad-specific lncRNA may act as pri-miR-202 to regulate testicular endocrine homeostasis and spermatogenesis. Theriogenology 2024; 214:273-285. [PMID: 37948817 DOI: 10.1016/j.theriogenology.2023.10.020] [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: 05/09/2023] [Revised: 10/09/2023] [Accepted: 10/15/2023] [Indexed: 11/12/2023]
Abstract
A large number of long non-coding RNAs (lncRNAs) are expressed in animal gonads, but their functions are poorly understood. In this study, a gonad-specific lncRNA, termed lnc4, was identified and characterized in the model fish medaka (Oryzias latipes). The expression pattern and in vitro functional analyses indicated that lnc4 was likely to be a primary transcript of miR-202 (pri-miR-202). Results of single-molecule fluorescence in situ hybridization demonstrated that the precursor miR-202 (pre-miR-202) was highly expressed in the nuclei of testicular somatic cells, including Leydig and Sertoli cells, whereas only a small amount of lnc4 molecules could be detected co-expressed with pre-miR-202 in Sertoli cells due to its low expression level. Deletion of the lnc4 locus led to a significant reduction in testis size and a dramatic decrease in the number of male germ cells, as well as a reduction in sperm viability. Moreover, lnc4 knockout resulted in enhanced synthesis and secretion of testicular somatic cells and accelerated differentiation of immature male germ cells. Taken together, functional studies of lnc4 and its mature transcript miR-202 will contribute to the understanding of the important role of non-coding RNAs in animal or human reproductive disorders.
Collapse
Affiliation(s)
- Qian Wang
- College of Animal Science and Technology, Yangtze University, Jingzhou, Hubei, 434024, China; College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Jiajie Deng
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, Institute of Hydrobiology, Innovation Academy for Seed Design (INASEED), Chinese Academy of Sciences, Wuhan, Hubei, 430072, China
| | - Yuewen Jiang
- College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Zhi Wang
- College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Bilin Xia
- College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Tiansheng Chen
- Fisheries College of Jimei University, Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education, Xiamen, Fujian, 361021, China; College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| |
Collapse
|
11
|
Zhao D, He J, Zhao X, Sheng X, Feng Z, Wang X, Zhang C, Wang S, Geng B, Xia Y. A novel lncRNA GM15416 regulates osteoblast apoptosis and differentiation through the c-Fos/Fas axis and mitigates osteoporosis. Int J Biol Macromol 2024; 254:127824. [PMID: 37924900 DOI: 10.1016/j.ijbiomac.2023.127824] [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: 07/24/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023]
Abstract
Osteoporosis (OP) is a common systemic bone disorder, and the programmed cell death of osteoblasts is closely linked to the development of osteoporosis. Previous studies have shown that c-fos can cause osteoblast apoptosis. Furthermore, it has been demonstrated that long non-coding RNA (lncRNA) plays a pervasive role in regulating the biology of osteoblasts. Nevertheless, the precise role and mechanism of long non-coding RNA (lncRNA) in relation to c-Fos at the transcriptional level in osteoblast cell death remain uncertain. Compared with normal osteoblasts, serum deprivation resulted in significant upregulation of the transcription factor c-Fos and apoptosis-related Fas proteins in osteoblasts. In addition, the expression of lncRNA GM15416 related to c-Fos was significantly increased. The results showed that overexpression of c-Fos leads to an increase in downstream Fas protein, which subsequently leads to osteoblast apoptosis and hinders osteogenesis. On the contrary, a decrease in lncRNA GM15416 expression leads to a decrease in c-Fos/Fas expression, which hinders osteoblast apoptosis and promotes osteogenesis. Our results suggest that lncRNA GM15416 exerts inhibitory effects on osteoblast apoptosis and acts as a preventive factor against osteoporosis. As a result, GM15416 emerges as an important lncRNA associated with osteoporosis and holds potential as a future therapeutic target.
Collapse
Affiliation(s)
- Dacheng Zhao
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Pain Department of the Second Hospital of Lanzhou University, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, Pr China
| | - Jinwen He
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Xiaobing Zhao
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Xiaoyun Sheng
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Zhiwei Feng
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Xingwen Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Chengjun Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Shenghong Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Bin Geng
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China.
| | - Yayi Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China.
| |
Collapse
|
12
|
Zamanian Azodi M, Arjmand B, Hamzeloo-Moghadam M, Rezaei Tavirani M, Razzaghi Z, Ahmadzadeh A, Robati RM, Rezaei Tavirani M. Assessment of the Response of Human Umbilical Vein Endothelial Cells to Photodynamic Therapy: Highlighting the Role of Il-17 Signaling Pathway. J Lasers Med Sci 2023; 14:e59. [PMID: 38144940 PMCID: PMC10746882 DOI: 10.34172/jlms.2023.59] [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: 09/05/2023] [Accepted: 10/07/2023] [Indexed: 12/26/2023]
Abstract
Introduction: Photodynamic therapy (PDT) is a method based on the application of a photosensitive agent and the administration of light irradiation on the treated samples. PDT is applied as an effective tool with minimal side effects against tumor tissues. This study aimed to assess the targets of critical genes by PDT at the cellular level of cancer to provide a new perspective on its molecular mechanism. Methods: To assess the effect of PDT, we extracted the differentially expressed genes (DEGs) from the gene expression profiles of human umbilical vein endothelial cells (HUVECs) treated with PDT from Gene Expression Omnibus (GEO) databases. The queried DEGs were evaluated via a regulatory network and gene ontology enrichment to find the critical targets. Results: Among 76 queried significant DEGs, 27 individuals were interacted by activation, inhibition, and co-expression actions. Thirty DEGs were related to the five classes of biological terms. The IL-17 signaling pathway and PTGS2, CXCL8, FOS, JUN, CXCL1, ZFP36, and FOSB were identified as the crucial targets of PDT. Conclusion: PDT as a stimulator of gene expression and an activator of gene activity overexpressed and hyper-activated many genes. It seems that PDT introduces a number of genes and pathways that can be regulated by anticancer drugs to fight against cancers.
Collapse
Affiliation(s)
- Mona Zamanian Azodi
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Iranian Cancer Control Center (MACSA), Tehran, Iran
| | - Maryam Hamzeloo-Moghadam
- Traditional Medicine and Materia Medica Research Center, School of Traditional Medicine Shahid, Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Razzaghi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Ahmadzadeh
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza M Robati
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Rezaei Tavirani
- Department of surgery, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
He Y, Ling Y, Zhang Z, Mertens RT, Cao Q, Xu X, Guo K, Shi Q, Zhang X, Huo L, Wang K, Guo H, Shen W, Shen M, Feng W, Xiao P. Butyrate reverses ferroptosis resistance in colorectal cancer by inducing c-Fos-dependent xCT suppression. Redox Biol 2023; 65:102822. [PMID: 37494767 PMCID: PMC10388208 DOI: 10.1016/j.redox.2023.102822] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023] Open
Abstract
Ferroptosis has emerged to be a promising approach in cancer therapies; however, colorectal cancer (CRC) is relatively insensitive to ferroptosis. Exactly how the gut microenvironment impacts the ferroptotic sensitivity of CRC remains unknown. Herein, by performing metabolomics, we discovered that butyrate concentrations were significantly decreased in CRC patients. Butyrate supplementation sensitized CRC mice to ferroptosis induction, showing great in vivo translatability. Particularly, butyrate treatment reduced ferroptotic resistance of cancer stem cells. Mechanistically, butyrate inhibited xCT expression and xCT-dependent glutathione synthesis. Moreover, we identified c-Fos as a novel xCT suppressor, and further elucidated that butyrate induced c-Fos expression via disrupting class I HDAC activity. In CRC patients, butyrate negatively correlated with tumor xCT expression and positively correlated with c-Fos expression. Finally, butyrate was found to boost the pro-ferroptotic function of oxaliplatin (OXA). Immunohistochemistry data showed that OXA non-responders exhibited higher xCT expression compared to OXA responders. Hence, butyrate supplementation is a promising approach to break the ferroptosis resistance in CRC.
Collapse
Affiliation(s)
- Ying He
- Central Laboratory, The First Affiliated Hospital of Huzhou University, Huzhou, 313000, China; Huzhou Key Laboratory of Translational Medicine, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Yuhang Ling
- Central Laboratory, The First Affiliated Hospital of Huzhou University, Huzhou, 313000, China; Huzhou Key Laboratory of Translational Medicine, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Zhiyong Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | | | - Qian Cao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China; Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Xutao Xu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Ke Guo
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Qian Shi
- Central Laboratory, The First Affiliated Hospital of Huzhou University, Huzhou, 313000, China; Huzhou Key Laboratory of Translational Medicine, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Xilin Zhang
- Central Laboratory, The First Affiliated Hospital of Huzhou University, Huzhou, 313000, China; Huzhou Key Laboratory of Translational Medicine, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Lixia Huo
- Central Laboratory, The First Affiliated Hospital of Huzhou University, Huzhou, 313000, China; Huzhou Key Laboratory of Translational Medicine, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Kan Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Huihui Guo
- Central Laboratory, The First Affiliated Hospital of Huzhou University, Huzhou, 313000, China; Huzhou Key Laboratory of Translational Medicine, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Weiyun Shen
- Central Laboratory, The First Affiliated Hospital of Huzhou University, Huzhou, 313000, China; Huzhou Key Laboratory of Translational Medicine, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Manlu Shen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Wenming Feng
- Central Laboratory, The First Affiliated Hospital of Huzhou University, Huzhou, 313000, China; Huzhou Key Laboratory of Translational Medicine, The First People's Hospital of Huzhou, Huzhou, 313000, China.
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China; Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China; Institute of Immunology, Zhejiang University School of Medicine, 310058, Hangzhou, China; The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, 310058, China.
| |
Collapse
|
14
|
Bae S, Lim HK, Jeong Y, Kim SG, Park SM, Shon YM, Suh M. Deep brain stimulation of the anterior nuclei of the thalamus can alleviate seizure severity and induce hippocampal GABAergic neuronal changes in a pilocarpine-induced epileptic mouse brain. Cereb Cortex 2022; 32:5530-5543. [PMID: 35258078 DOI: 10.1093/cercor/bhac033] [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: 11/09/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 01/25/2023] Open
Abstract
Deep brain stimulation (DBS) of the anterior nucleus of the thalamus (ANT) has been widely used as an effective treatment for refractory temporal lobe epilepsy. Despite its promising clinical outcome, the exact mechanism of how ANT-DBS alleviates seizure severity has not been fully understood, especially at the cellular level. To assess effects of DBS, the present study examined electroencephalography (EEG) signals and locomotor behavior changes and conducted immunohistochemical analyses to examine changes in neuronal activity, number of neurons, and neurogenesis of inhibitory neurons in different hippocampal subregions. ANT-DBS alleviated seizure activity, abnormal locomotor behaviors, reduced theta-band, increased gamma-band EEG power in the interictal state, and increased the number of neurons in the dentate gyrus (DG). The number of parvalbumin- and somatostatin-expressing inhibitory neurons was recovered to the level in DG and CA1 of naïve mice. Notably, BrdU-positive inhibitory neurons were increased. In conclusion, ANT-DBS not only could reduce the number of seizures, but also could induce neuronal changes in the hippocampus, which is a key region involved in chronic epileptogenesis. Importantly, our results suggest that ANT-DBS may lead to hippocampal subregion-specific cellular recovery of GABAergic inhibitory neurons.
Collapse
Affiliation(s)
- Sungjun Bae
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, South Korea.,Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea.,IMNEWRUN Inc., N Center Bldg. A 5F, Sungkyunkwan University, Suwon 16419, South Korea
| | - Hyun-Kyoung Lim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, South Korea.,Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea
| | - Yoonyi Jeong
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, South Korea.,Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, South Korea.,Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea
| | - Sung-Min Park
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Young-Min Shon
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea.,Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Suwon 16419, South Korea
| | - Minah Suh
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, South Korea.,Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea.,IMNEWRUN Inc., N Center Bldg. A 5F, Sungkyunkwan University, Suwon 16419, South Korea.,Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea.,Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Suwon 16419, South Korea
| |
Collapse
|
15
|
Chattopadhyay P, Khare K, Kumar M, Mishra P, Anand A, Maurya R, Gupta R, Sahni S, Gupta A, Wadhwa S, Yadav A, Devi P, Tardalkar K, Joshi M, Sethi T, Pandey R. Single-cell multiomics revealed the dynamics of antigen presentation, immune response and T cell activation in the COVID-19 positive and recovered individuals. Front Immunol 2022; 13:1034159. [PMID: 36532041 PMCID: PMC9755500 DOI: 10.3389/fimmu.2022.1034159] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Despite numerous efforts to describe COVID-19's immunological landscape, there is still a gap in our understanding of the virus's infections after-effects, especially in the recovered patients. This would be important to understand as we now have huge number of global populations infected by the SARS-CoV-2 as well as variables inclusive of VOCs, reinfections, and vaccination breakthroughs. Furthermore, single-cell transcriptome alone is often insufficient to understand the complex human host immune landscape underlying differential disease severity and clinical outcome. Methods By combining single-cell multi-omics (Whole Transcriptome Analysis plus Antibody-seq) and machine learning-based analysis, we aim to better understand the functional aspects of cellular and immunological heterogeneity in the COVID-19 positive, recovered and the healthy individuals. Results Based on single-cell transcriptome and surface marker study of 163,197 cells (124,726 cells after data QC) from the 33 individuals (healthy=4, COVID-19 positive=16, and COVID-19 recovered=13), we observed a reduced MHC Class-I-mediated antigen presentation and dysregulated MHC Class-II-mediated antigen presentation in the COVID-19 patients, with restoration of the process in the recovered individuals. B-cell maturation process was also impaired in the positive and the recovered individuals. Importantly, we discovered that a subset of the naive T-cells from the healthy individuals were absent from the recovered individuals, suggesting a post-infection inflammatory stage. Both COVID-19 positive patients and the recovered individuals exhibited a CD40-CD40LG-mediated inflammatory response in the monocytes and T-cell subsets. T-cells, NK-cells, and monocyte-mediated elevation of immunological, stress and antiviral responses were also seen in the COVID-19 positive and the recovered individuals, along with an abnormal T-cell activation, inflammatory response, and faster cellular transition of T cell subtypes in the COVID-19 patients. Importantly, above immune findings were used for a Bayesian network model, which significantly revealed FOS, CXCL8, IL1β, CST3, PSAP, CD45 and CD74 as COVID-19 severity predictors. Discussion In conclusion, COVID-19 recovered individuals exhibited a hyper-activated inflammatory response with the loss of B cell maturation, suggesting an impeded post-infection stage, necessitating further research to delineate the dynamic immune response associated with the COVID-19. To our knowledge this is first multi-omic study trying to understand the differential and dynamic immune response underlying the sample subtypes.
Collapse
Affiliation(s)
- Partha Chattopadhyay
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kriti Khare
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manish Kumar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Pallavi Mishra
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Alok Anand
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Ranjeet Maurya
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rohit Gupta
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Shweta Sahni
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Ayushi Gupta
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Saruchi Wadhwa
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Aanchal Yadav
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Priti Devi
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kishore Tardalkar
- Department of Stem Cells and Regenerative Medicine, Dr. D. Y. Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra, India
| | - Meghnad Joshi
- Department of Stem Cells and Regenerative Medicine, Dr. D. Y. Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra, India
| | - Tavpritesh Sethi
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Rajesh Pandey
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
16
|
Buettner JM, Sowoidnich L, Gerstner F, Blanco-Redondo B, Hallermann S, Simon CM. p53-dependent c-Fos expression is a marker but not executor for motor neuron death in spinal muscular atrophy mouse models. Front Cell Neurosci 2022; 16:1038276. [DOI: 10.3389/fncel.2022.1038276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022] Open
Abstract
The activation of the p53 pathway has been associated with neuronal degeneration in different neurological disorders, including spinal muscular atrophy (SMA) where aberrant expression of p53 drives selective death of motor neurons destined to degenerate. Since direct p53 inhibition is an unsound therapeutic approach due carcinogenic effects, we investigated the expression of the cell death-associated p53 downstream targets c-fos, perp and fas in vulnerable motor neurons of SMA mice. Fluorescence in situ hybridization (FISH) of SMA motor neurons revealed c-fos RNA as a promising candidate. Accordingly, we identified p53-dependent nuclear upregulation of c-Fos protein in degenerating motor neurons from the severe SMNΔ7 and intermediate Smn2B/– SMA mouse models. Although motor neuron-specific c-fos genetic deletion in SMA mice did not improve motor neuron survival or motor behavior, p53-dependent c-Fos upregulation marks vulnerable motor neurons in different mouse models. Thus, nuclear c-Fos accumulation may serve as a readout for therapeutic approaches targeting neuronal death in SMA and possibly other p53-dependent neurodegenerative diseases.
Collapse
|
17
|
NT157 exerts antineoplastic activity by targeting JNK and AXL signaling in lung cancer cells. Sci Rep 2022; 12:17092. [PMID: 36224313 PMCID: PMC9556623 DOI: 10.1038/s41598-022-21419-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/27/2022] [Indexed: 01/04/2023] Open
Abstract
Combination therapies or multi-targeted drugs have been pointed out as an option to prevent the emergence of resistant clones, which could make long-term treatment more effective and translate into better clinical outcomes for cancer patients. The NT157 compound is a synthetic tyrphostin that leads to long-term inhibition of IGF1R/IRS1-2-, STAT3- and AXL-mediated signaling pathways. Given the importance of these signaling pathways for the development and progression of lung cancer, this disease becomes an interesting model for generating preclinical evidence on the cellular and molecular mechanisms underlying the antineoplastic activity of NT157. In lung cancer cells, exposure to NT157 decreased, in a dose-dependent manner, cell viability, clonogenicity, cell cycle progression and migration, and induced apoptosis (p < 0.05). In the molecular scenario, NT157 reduced expression of IRS1 and AXL and phosphorylation of p38 MAPK, AKT, and 4EBP1. Besides, NT157 decreased expression of oncogenes BCL2, CCND1, MYB, and MYC and increased genes related to cellular stress and apoptosis, JUN, BBC3, CDKN1A, CDKN1B, FOS, and EGR1 (p < 0.05), favoring a tumor-suppressive cell signaling network in the context of lung cancer. Of note, JNK was identified as a key kinase for NT157-induced IRS1 and IRS2 phosphorylation, revealing a novel axis involved in the mechanism of action of the drug. NT157 also presented potentiating effects on EGFR inhibitors in lung cancer cells. In conclusion, our preclinical findings highlight NT157 as a putative prototype of a multitarget drug that may contribute to the antineoplastic arsenal against lung cancer.
Collapse
|
18
|
Yang Z, Zhang H, Yin M, Cheng Z, Jiang P, Feng M, Liao B, Liu Z. Neurotrophin3 promotes hepatocellular carcinoma apoptosis through the JNK and P38 MAPK pathways. Int J Biol Sci 2022; 18:5963-5977. [PMID: 36263167 PMCID: PMC9576519 DOI: 10.7150/ijbs.72982] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022] Open
Abstract
Although liver cancer is a malignant tumor with the highest mortality across the world, its pathogenesis and therapeutic targets remain unclear. Apoptosis, a natural cell death mechanism, is an important target of anticancer therapy. The discovery of effective apoptotic regulators can lead to the identification of novel therapeutic targets for treating cancer. Neurotrophin 3 (NTF3) is a member of the nerve growth factor (NGF) family that is involved in the progression of various cancers, including medulloblastoma, primitive neuroectodermal brain tumors, and breast cancer. NTF3 is under-expressed in human hepatocellular carcinoma (HCC), albeit its specific effects and the action mechanism have not been elucidated. Here, we confirmed that NTF3 expression was significantly low in HCC with reference to the GSEA database. By collecting patient data from our center and performing qRT-PCR analysis, we found that NTF3 expression was significantly downregulated in 74 patients with HCC. Low NTF3 expression was associated with a shorter overall survival (OS), recurrence-free survival (RFS), progression-free survival (PFS), and disease-specific survival (DSS). Both in vivo and in vitro experiments revealed that NTF3 considerably inhibited the progression of HCC cells. We found that the ligand NTF3 is regulated by c-Jun and binds to the p75 neurotrophin receptor (p75NTR) and then activates the JNK and P38 MAPK pathways to induce apoptosis. Entinostat (the target of HDAC1/HDAC3) can activate the NTF3/p75NTR pathway. These results indicate that NTF3 is a tumor suppressor, and that its low expression can help in predict poor clinical outcomes in HCC. Therefore, NTF3 can be used as a potential treatment molecule for HCC.
Collapse
Affiliation(s)
- Zhangshuo Yang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| | - Hao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| | - Maohui Yin
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| | - Zhixiang Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| | - Ping Jiang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| | - Maohui Feng
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China.,✉ Corresponding authors: Zhisu Liu, E-mail: ; Bo Liao, E-mail: ; Maohui Feng, E-mail: ; Tel: +862767812588; Fax: +862767813493
| | - Bo Liao
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China.,✉ Corresponding authors: Zhisu Liu, E-mail: ; Bo Liao, E-mail: ; Maohui Feng, E-mail: ; Tel: +862767812588; Fax: +862767813493
| | - Zhisu Liu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, People's Republic of China.,✉ Corresponding authors: Zhisu Liu, E-mail: ; Bo Liao, E-mail: ; Maohui Feng, E-mail: ; Tel: +862767812588; Fax: +862767813493
| |
Collapse
|
19
|
Ziółkowska N, Lewczuk B. Profiles of Rho, Opn4, c-Fos, and Birc5 mRNA expression in Wistar rat retinas exposed to white or monochromatic light. Front Neuroanat 2022; 16:956000. [PMID: 36059433 PMCID: PMC9434339 DOI: 10.3389/fnana.2022.956000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/21/2022] [Indexed: 11/30/2022] Open
Abstract
Despite concern over potential retinal damage linked to exposure to light-emitting-diode (LED) light (particularly blue light), it remains unknown how exposure to low-intensity monochromatic LED light affects the expression of rhodopsin (Rho, a photopigment that mediates light-induced retinal degeneration), melanopsin (Opn4, a blue-light sensitive photopigment), c-Fos (associated with retinal damage/degeneration), and Birc5 (anti-apoptotic). This study investigated the mRNA expression profiles of these genes under exposure to white and monochromatic light (blue, red, green) in the retinas of albino rats under a cycle of 12 h of light and 12 h of darkness. In each group, 32 Wistar rats were exposed to one type of monochromatic-LED or white-fluorescent light for 7 day (150 lx). Retinal samples were taken for qPCR analysis and light and electron microscopy. Blue and green light exposure markedly decreased expression of Rho and Opn4 mRNA and increased expression of Birc5 and c-Fos mRNA (P < 0.05). In retinas from the blue-light group, loss and vesiculation of photoreceptor outer segments were visible, but not in retinas from the red-light and control group. Measurements of the photoreceptor inner and outer segments length revealed, that this length was significantly decreased in the blue- and green-light exposure groups (P < 0.02), but not in the red-light exposure group. Increased expression of Birc5 and decreased expression of Rho and Opn4 after exposure to blue and green light may be early responses that help to reduce light-induced retinal damage.
Collapse
Affiliation(s)
- Natalia Ziółkowska
- Department of Histology and Embryology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | | |
Collapse
|
20
|
Aljabban J, Rohr M, Syed S, Khorfan K, Borkowski V, Aljabban H, Segal M, Mukhtar M, Mohammed M, Panahiazar M, Hadley D, Spengler R, Spengler E. Transcriptome changes in stages of non-alcoholic fatty liver disease. World J Hepatol 2022; 14:1382-1397. [PMID: 36158924 PMCID: PMC9376779 DOI: 10.4254/wjh.v14.i7.1382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/29/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the United States and globally. The currently understood model of pathogenesis consists of a ‘multiple hit’ hypothesis in which environmental and genetic factors contribute to hepatic inflammation and injury.
AIM To examine the genetic expression of NAFLD and non-alcoholic steatohepatitis (NASH) tissue samples to identify common pathways that contribute to NAFLD and NASH pathogenesis.
METHODS We employed the Search Tag Analyze Resource for Gene Expression Omnibus platform to search the The National Center for Biotechnology Information Gene Expression Omnibus to elucidate NAFLD and NASH pathology. For NAFLD, we conducted meta-analysis of data from 58 NAFLD liver biopsies and 60 healthy liver biopsies; for NASH, we analyzed 187 NASH liver biopsies and 154 healthy liver biopsies.
RESULTS Our results from the NAFLD analysis reinforce the role of altered metabolism, inflammation, and cell survival in pathogenesis and support recently described contributors to disease activity, such as altered androgen and long non-coding RNA activity. The top upstream regulator was found to be sterol regulatory element binding transcription factor 1 (SREBF1), a transcription factor involved in lipid homeostasis. Downstream of SREBF1, we observed upregulation in CXCL10, HMGCR, HMGCS1, fatty acid binding protein 5, paternally expressed imprinted gene 10, and downregulation of sex hormone-binding globulin and insulin-like growth factor 1. These molecular changes reflect low-grade inflammation secondary to accumulation of fatty acids in the liver. Our results from the NASH analysis emphasized the role of cholesterol in pathogenesis. Top canonical pathways, disease networks, and disease functions were related to cholesterol synthesis, lipid metabolism, adipogenesis, and metabolic disease. Top upstream regulators included pro-inflammatory cytokines tumor necrosis factor and IL1B, PDGF BB, and beta-estradiol. Inhibition of beta-estradiol was shown to be related to derangement of several cellular downstream processes including metabolism, extracellular matrix deposition, and tumor suppression. Lastly, we found riciribine (an AKT inhibitor) and ZSTK-474 (a PI3K inhibitor) as potential drugs that targeted the differential gene expression in our dataset.
CONCLUSION In this study we describe several molecular processes that may correlate with NAFLD disease and progression. We also identified ricirbine and ZSTK-474 as potential therapy.
Collapse
Affiliation(s)
- Jihad Aljabban
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Michael Rohr
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, United States
| | - Saad Syed
- Department of Medicine, Northwestern Memorial Hospital, Chicago, IL 60611, United States
| | - Kamal Khorfan
- Department of Gastroenterology and Hepatology, University of California San Francisco-Fresno , Fresno, CA 93701, United States
| | - Vincent Borkowski
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Hisham Aljabban
- Department of Medicine, Barry University, Miami, FL 33161, United States
| | - Michael Segal
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Mohamed Mukhtar
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI 49503, United States
| | - Mohammed Mohammed
- Department of Medicine, Windsor University School of Medicine, Saint Kitts 1621, Cayon, Saint Kitts and Nevis
| | - Maryam Panahiazar
- Department of Surgery, University of California San Francisco, San Francisco, CA 94305, United States
| | - Dexter Hadley
- Department of Artificial Intelligence, Pathology, University of Central Florida College of Medicine , Orlando, FL 32827, United States
| | - Ryan Spengler
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Erin Spengler
- Department of Gastroenterology and Hepatology, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| |
Collapse
|
21
|
Puvvula PK, Buczkowski S, Moon AM. hnRNPK-derived cell-penetrating peptide inhibits cancer cell survival. Mol Ther Oncolytics 2021; 23:342-354. [PMID: 34820504 PMCID: PMC8586514 DOI: 10.1016/j.omto.2021.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/28/2021] [Accepted: 10/15/2021] [Indexed: 11/30/2022] Open
Abstract
hnRNPK is a multifunctional protein that plays an important role in cancer cell proliferation and metastasis via its RNA- and DNA-binding properties. Previously we showed that cell-penetrating peptides derived from the RGG RNA-binding domain of SAFA (hnRNPU) disrupt cancer cell proliferation and survival. Here we explore the efficacy of a peptide derived from the RGG domain of hnRNPK. This peptide acts in a dominant-negative manner on several hnRNPK functions to induce death of multiple types of cancer cells. The peptide phenocopies the effect of hnRNPK knockdown on its mRNA-stability targets such as KLF4 and EGR1 and alters the levels and locations of long non-coding RNAs (lncRNAs) and proteins required for nuclear and paraspeckle formation and function. The RGG-derived peptide also decreases euchromatin as evidenced by loss of active marks and polymerase II occupancy. Our findings reveal the potential therapeutic utility of the hnRNPK RGG-derived peptide in a range of cancers.
Collapse
Affiliation(s)
- Pavan Kumar Puvvula
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA
- Corresponding author: Pavan Kumar Puvvula, PhD, Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA.
| | - Stephanie Buczkowski
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA
| | - Anne M. Moon
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
- The Mindich Child Health and Development Institute, Hess Center for Science and Medicine at Mount Sinai, New York, NY, USA
- Corresponding author: Anne M. Moon, MD, PhD, Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA.
| |
Collapse
|
22
|
Parvin S, Williams CR, Jarrett SA, Garraway SM. Spinal Cord Injury Increases Pro-inflammatory Cytokine Expression in Kidney at Acute and Sub-chronic Stages. Inflammation 2021; 44:2346-2361. [PMID: 34417952 PMCID: PMC8616867 DOI: 10.1007/s10753-021-01507-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/21/2021] [Indexed: 11/26/2022]
Abstract
Accumulating evidence supports that spinal cord injury (SCI) produces robust inflammatory plasticity. We previously showed that the pro-inflammatory cytokine tumor necrosis factor (TNF)α is increased in the spinal cord after SCI. SCI also induces a systemic inflammatory response that can impact peripheral organ functions. The kidney plays an important role in maintaining cardiovascular health. However, SCI-induced inflammatory response in the kidney and the subsequent effect on renal function have not been well characterized. This study investigated the impact of high and low thoracic (T) SCI on C-fos, TNFα, interleukin (IL)-1β, and IL-6 expression in the kidney at acute and sub-chronic timepoints. Adult C57BL/6 mice received a moderate contusion SCI or sham procedures at T4 or T10. Uninjured mice served as naïve controls. mRNA levels of the proinflammatory cytokines IL-1β, IL-6, TNFα, and C-fos, and TNFα and C-fos protein expression were assessed in the kidney and spinal cord 1 day and 14 days post-injury. The mRNA levels of all targets were robustly increased in the kidney and spinal cord, 1 day after both injuries. Whereas IL-6 and TNFα remained elevated in the spinal cord at 14 days after SCI, C-fos, IL-6, and TNFα levels were sustained in the kidney only after T10 SCI. TNFα protein was significantly upregulated in the kidney 1 day after both T4 and T10 SCI. Overall, these results clearly demonstrate that SCI induces robust systemic inflammation that extends to the kidney. Hence, the presence of renal inflammation can substantially impact renal pathophysiology and function after SCI.
Collapse
Affiliation(s)
- Shangrila Parvin
- Department of Physiology, Emory University School of Medicine, 615 Michael Street, Suite 605G, Atlanta, GA 30322 USA
| | - Clintoria R. Williams
- Department of Physiology, Emory University School of Medicine, 615 Michael Street, Suite 605G, Atlanta, GA 30322 USA
- Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH USA
| | - Simone A. Jarrett
- Department of Physiology, Emory University School of Medicine, 615 Michael Street, Suite 605G, Atlanta, GA 30322 USA
| | - Sandra M. Garraway
- Department of Physiology, Emory University School of Medicine, 615 Michael Street, Suite 605G, Atlanta, GA 30322 USA
- Department of Physiology, Emory University School of Medicine, 615 Michael Street, Suite 605G, Atlanta, GA 30322 USA
| |
Collapse
|
23
|
Mu Q, Zhang Y, Gu L, Gerner ST, Qiu X, Tao Q, Pang J, Dipritu G, Zhang L, Yin S, Jiang Y, Peng J. Transcriptomic Profiling Reveals the Antiapoptosis and Antioxidant Stress Effects of Fos in Ischemic Stroke. Front Neurol 2021; 12:728984. [PMID: 34744970 PMCID: PMC8566985 DOI: 10.3389/fneur.2021.728984] [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: 06/22/2021] [Accepted: 09/09/2021] [Indexed: 12/02/2022] Open
Abstract
Arterial hypertension is considered the most prevalent risk factor for stroke. Both pathophysiologic and clinical data previously acquired suggest a strong correlation between the hemodynamic nature of arterial hypertension and an increase in the risk of ischemic insult to tissues. However, the knowledge of specific molecular interactions between hypertension and ischemic stroke (IS) is limited. In this study, we performed systematic bioinformatics analysis of stroke-prone spontaneous hypertensive brain tissue samples of rats (GSE41452), middle cerebral artery occlusion of brain tissue samples of rats (GSE97537), and peripheral blood array data of IS patients (GSE22255). We identified that Fos, an immediate-early gene (IEG) that responds to alterations in arterial blood pressure, has a strong correlation with the occurrence and prognosis of IS. To further evaluate the potential function of Fos, the oxygen–glucose deprivation model and RNA sequencing of HT22 neuronal cells were performed. Consistent with the sequencing results, real-time quantitative PCR and Western blot indicate that Fos was elevated at 3 h and returned to normal levels at 6 h after oxygen–glucose deprivation. Knock-down of Fos by lentivirus significantly increased the oxidative stress level, neuronal apoptosis, and inhibited the mitochondrial function. In conclusion, Fos acts as an important link between hypertension and IS. Furthermore, Fos can be used as a potential biomarker for target therapy in the prevention of stroke among hypertensive patients and also potential treatment targeting apoptosis and oxidative stress after its onset.
Collapse
Affiliation(s)
- Qiancheng Mu
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuxuan Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Long Gu
- Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Stefan T Gerner
- Department of Neurology, University Hospital Erlangen-Nuremberg, Erlangen, Germany
| | - Xiancheng Qiu
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qianke Tao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinwei Pang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China
| | - Ghosh Dipritu
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lifang Zhang
- Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shigang Yin
- Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Luzhou Key Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
24
|
Machuca A, Garcia-Calvo E, Anunciação DS, Luque-Garcia JL. Integration of Transcriptomics and Metabolomics to Reveal the Molecular Mechanisms Underlying Rhodium Nanoparticles-Based Photodynamic Cancer Therapy. Pharmaceutics 2021; 13:pharmaceutics13101629. [PMID: 34683922 PMCID: PMC8539937 DOI: 10.3390/pharmaceutics13101629] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Rhodium nanoparticles have recently been described as promising photosensitizers due to their low toxicity in the absence of near-infrared irradiation, but their high cytotoxicity when irradiated. Irradiation is usually carried out with a laser source, which allows the treatment to be localized in a specific area, thus avoiding undesirable side effects on healthy tissues. In this study, a multi-omics approach based on the combination of microarray-based transcriptomics and mass spectrometry-based untargeted and targeted metabolomics has provided a global picture of the molecular mechanisms underlying the anti-tumoral effect of rhodium nanoparticle-based photodynamic therapy. The results have shown the ability of these nanoparticles to promote apoptosis by suppressing or promoting anti- and pro-apoptotic factors, respectively, and by affecting the energy machinery of tumor cells, mainly blocking the β-oxidation, which is reflected in the accumulation of free fatty acids and in the decrease in ATP, ADP and NAD+ levels.
Collapse
Affiliation(s)
- Andres Machuca
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (E.G.-C.)
| | - Estefania Garcia-Calvo
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (E.G.-C.)
| | - Daniela S. Anunciação
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Campus A. C. Simões, 57072-900 Maceió, Brazil;
| | - Jose L. Luque-Garcia
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (E.G.-C.)
- Correspondence: ; Tel.: +34-913-944-212
| |
Collapse
|
25
|
Wang Y, Zheng Y, Dong J, Zhang X. Two-sided effects of prolonged hypoxia and sulfide exposure on juvenile ark shells (Anadara broughtonii). MARINE ENVIRONMENTAL RESEARCH 2021; 169:105326. [PMID: 33848850 DOI: 10.1016/j.marenvres.2021.105326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 06/12/2023]
Abstract
Oxygen deficit and sulfide have been restrictive factors in mariculture zones. However, the adaptive mechanism in aquatic lives is still unclear. The commercial ark shells Anadara broughtonii were selected to test the tolerance and adaptive responses to prolonged and intermittent hypoxia with or without exogenous sulfide (mild, moderate, high) by evaluating their behavior, mortality, oxidative level, antioxidant responses, and the MAPK-mediated apoptosis in gills. The results indicated that the clams were tolerant to hypoxia and sulfide exposure but vulnerable during reoxygenation from the challenges. Even so, sulfide had remarkable effect on attenuating the accumulation of reactive oxygen species (ROS) and lipid peroxides caused by reoxygenation from prolonged hypoxia. The increase of glutathione level was probably as an early and primary protective response to prevent the expected reperfusion injury from reoxygenation. The challenges suppressed the oxidative level with a dose-dependent effect of sulfide, with an exception when exposed to mild sulfide. Synchronously, biphasic effects of exogenous sulfide on apoptotic cascade, which was induced by mild sulfide while it was inhibited by higher sulfide, were also detected in gills. The induced or inhibited apoptosis by hypoxia and sulfide kept to a typical ROS-MAPK-CASPASE cascade, desiderating further investigation.
Collapse
Affiliation(s)
- Yihang Wang
- Fishery College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Yingqiu Zheng
- Fishery College, Ocean University of China, Qingdao 266003, China
| | - Jianyu Dong
- Fishery College, Ocean University of China, Qingdao 266003, China
| | - Xiumei Zhang
- Fishery College, Zhejiang Ocean University, Zhoushan 316022, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
26
|
LI Y, LI W, FENG Z, SONG J, ZHANG C, HUANG L, SONG Y. Effect of operative trauma and multiple propofol anesthesia on neurodevelopment and cognitive function in developmental rats. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:290-297. [PMID: 34402265 PMCID: PMC8710274 DOI: 10.3724/zdxbyxb-2021-0004] [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: 01/08/2021] [Accepted: 05/21/2021] [Indexed: 11/25/2022]
Abstract
To investigate the effect of multiple propofol anesthesia and operative trauma on neuroinflammation and cognitive function in development rats and its mechanism. A total of 104 13-day-old neonatal Sprague-Dawley rats were randomly divided into 4 groups with 26 rats in each group: control group was treated with saline q.d for propofol group was treated with propofol q.d for surgery group received abdominal surgery under local anesthesia and then treated with saline q.d for surgery with propofol group received propofol anesthesia plus abdominal surgery under local anesthesia with ropivacaine at d1, then treated with propofol q.d for At d2 of experiment, 13 rats from each group were sacrificed and brain tissue samples were taken, the concentration of TNF-α in hippocampus was detected with ELISA, the expression of caspase-3 and c-fos in hippocampal tissue was determined with immunohistochemical method, the number of apoptotic neurons in hippocampus was examined with TUNEL assay. Morris water maze test was used to examine the cognitive function of the rest rats at the age of 60 d, and the TNF-α concentration, caspase-3, c-fos expressions and the number of apoptotic neurons in hippocampus were also detected. Compared with control group, TNF-α concentration, caspase-3, c-fos expression and the neuroapoptosis in hippocampus increased significantly in other three groups (all <0.05). Compared with surgery group, propofol group and surgery with propofol group showed increased TNF-α level, caspase-3 and c-fos expressions and apoptotic cell numbers (all <0.05), but there was no significant difference between last two groups (all >0.05). Morris water maze test showed that there were no significant differences in swimming speed, escape latency, target quadrant residence time and crossing times among groups (all >0.05). TNF-α level, expressions of caspase-3 and c-fos and apoptotic cell numbers in hippocampus had no significant differences among the 4 adult rats groups (all >0.05). Abdominal surgery and multiple propofol treatment can induce neuroinflammation and neuroapoptosis in hippocampus of neonatal rats, however, which may not cause adverse effects on neurodevelopment and cognitive function when they grown up.
Collapse
Affiliation(s)
| | | | - Zeguo FENG
- 通信作者:冯泽国,主任医师,主要从事复杂肝脏手术、腹膜后巨大肿瘤手术、复杂脊柱矫形手术的麻醉及复杂老年患者的麻醉研究;E-mail:
;https://orcid.org/0000-0001-7922-5461
| | | | | | | | | |
Collapse
|
27
|
Duncan-Lewis C, Hartenian E, King V, Glaunsinger BA. Cytoplasmic mRNA decay represses RNA polymerase II transcription during early apoptosis. eLife 2021; 10:e58342. [PMID: 34085923 PMCID: PMC8192121 DOI: 10.7554/elife.58342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/03/2021] [Indexed: 12/22/2022] Open
Abstract
RNA abundance is generally sensitive to perturbations in decay and synthesis rates, but crosstalk between RNA polymerase II transcription and cytoplasmic mRNA degradation often leads to compensatory changes in gene expression. Here, we reveal that widespread mRNA decay during early apoptosis represses RNAPII transcription, indicative of positive (rather than compensatory) feedback. This repression requires active cytoplasmic mRNA degradation, which leads to impaired recruitment of components of the transcription preinitiation complex to promoter DNA. Importin α/β-mediated nuclear import is critical for this feedback signaling, suggesting that proteins translocating between the cytoplasm and nucleus connect mRNA decay to transcription. We also show that an analogous pathway activated by viral nucleases similarly depends on nuclear protein import. Collectively, these data demonstrate that accelerated mRNA decay leads to the repression of mRNA transcription, thereby amplifying the shutdown of gene expression. This highlights a conserved gene regulatory mechanism by which cells respond to threats.
Collapse
Affiliation(s)
- Christopher Duncan-Lewis
- Department of Molecular and Cell Biology; University of California, BerkeleyBerkeleyUnited States
| | - Ella Hartenian
- Department of Molecular and Cell Biology; University of California, BerkeleyBerkeleyUnited States
| | - Valeria King
- Department of Molecular and Cell Biology; University of California, BerkeleyBerkeleyUnited States
| | - Britt A Glaunsinger
- Department of Molecular and Cell Biology; University of California, BerkeleyBerkeleyUnited States
- Department of Plant and Microbial Biology; University of California, BerkeleyBerkeleyUnited States
- Howard Hughes Medical Institute, BerkeleyBerkeleyUnited States
| |
Collapse
|
28
|
Sood N, Verma DK, Paria A, Yadav SC, Yadav MK, Bedekar MK, Kumar S, Swaminathan TR, Mohan CV, Rajendran KV, Pradhan PK. Transcriptome analysis of liver elucidates key immune-related pathways in Nile tilapia Oreochromis niloticus following infection with tilapia lake virus. FISH & SHELLFISH IMMUNOLOGY 2021; 111:208-219. [PMID: 33577877 DOI: 10.1016/j.fsi.2021.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 06/12/2023]
Abstract
Nile tilapia (Oreochromis niloticus) is one of the most important aquaculture species farmed worldwide. However, the recent emergence of tilapia lake virus (TiLV) disease, also known as syncytial hepatitis of tilapia, has threatened the global tilapia industry. To gain more insight regarding the host response against the disease, the transcriptional profiles of liver in experimentally-infected and control tilapia were compared. Analysis of RNA-Seq data identified 4640 differentially expressed genes (DEGs), which were involved among others in antigen processing and presentation, MAPK, apoptosis, necroptosis, chemokine signaling, interferon, NF-kB, acute phase response and JAK-STAT pathways. Enhanced expression of most of the DEGs in the above pathways suggests an attempt by tilapia to resist TiLV infection. However, upregulation of some of the key genes such as BCL2L1 in apoptosis pathway; NFKBIA in NF-kB pathway; TRFC in acute phase response; and SOCS, EPOR, PI3K and AKT in JAK-STAT pathway and downregulation of the genes, namely MAP3K7 in MAPK pathway; IFIT1 in interferon; and TRIM25 in NF-kB pathway suggested that TiLV was able to subvert the host immune response to successfully establish the infection. The study offers novel insights into the cellular functions that are affected following TiLV infection and will serve as a valuable genomic resource towards our understanding of susceptibility of tilapia to TiLV infection.
Collapse
Affiliation(s)
- Neeraj Sood
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Dev Kumar Verma
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Anutosh Paria
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Shrish Chandra Yadav
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Manoj Kumar Yadav
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India
| | - Megha Kadam Bedekar
- ICAR-Central Institute of Fisheries Education, Versova, Andheri (W), Mumbai, 400 061, Maharashtra, India
| | - Saurav Kumar
- ICAR-Central Institute of Fisheries Education, Versova, Andheri (W), Mumbai, 400 061, Maharashtra, India
| | - Thangaraj Raja Swaminathan
- Peninsular and Marine Fish Genetic Resources Centre, ICAR-NBFGR, CMFRI Campus, Kochi, 682 018, Kerala, India
| | | | - K V Rajendran
- ICAR-Central Institute of Fisheries Education, Versova, Andheri (W), Mumbai, 400 061, Maharashtra, India
| | - Pravata Kumar Pradhan
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, Uttar Pradesh, India.
| |
Collapse
|
29
|
Tu Y, Wu W, Guo Y, Lu F, Li X, Xu D, Zou D, Tu Y, Chai Y, He L. Up-regulation of hsa-miR-221-3p induced by UVB affects proliferation and apoptosis of keratinocytes via Bcl-xL/Bax pathway. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2021; 37:269-277. [PMID: 33351232 DOI: 10.1111/phpp.12647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 12/19/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Chronic actinic dermatitis (CAD) is a photoallergic skin disease with abnormal hyperplasia. At present, the mechanism of abnormal proliferation is not clear. OBJECTIVE To explore possible mechanism of CAD proliferative lesions. METHODS Immunohistochemistry (IHC) assay and small RNA sequencing were carried out. Quantitative real-time PCR (qRT-PCR) analysis was performed to evaluate expression levels of hsa-miR-221-3p and FOS. The interaction between hsa-miR-221-3p and FOS was identified by dual-luciferase reporter assay. Expression of hsa-miR-221-3p also was detected by qRT-PCR after UVB irradiation. Influences of hsa-miR-221-3p and FOS on cell viability and apoptosis were assessed through a series of functional experiments and rescue experiments. Western blot analysis was used to detect protein expression of fos, Bax, Bcl-xL, and caspase-3. RESULTS Patients with CAD had marked epidermal hyperplasia. The expression of hsa-miR-221-3p was up-regulated in CAD while FOS was significantly down-regulated. Dual-luciferase reporter assay confirmed that hsa-miR-221-3p targeted FOS 3'UTR. Hsa-miR-221-3p induced by UVB ranged from 0 to 30 mJ. Moreover, hsa-miR-221-3p overexpression or FOS knockdown promoted cell proliferation and reduced cell apoptosis. Western blot showed that hsa-miR-221-3p negatively regulated fos, which regulated Bcl-xL/Bax. Cell proliferation caused by hsa-miR-221-3p overexpression or FOS knockdown could be reversed by Bcl-xL inhibitor. CONCLUSION Hsa-miR-221-3p induced by UVB targeted FOS 3'UTR, which played an important role in regulating proliferation and apoptosis of keratinocytes via Bcl-xL/Bax pathway; this may provide a new insight for CAD proliferative lesions.
Collapse
Affiliation(s)
- Yunhua Tu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Dermatology, The Second People's Hospital of Guiyang, Guizhou, China
| | - Wenjuan Wu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yanni Guo
- Department of Dermatology, The Second Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Fengyan Lu
- Department of Dermatology, The First People's Hospital of Qujing, Qujing, China
| | - Xing Li
- Department of Dermatology, People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, China
| | - Dan Xu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Dandan Zou
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ying Tu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yanjie Chai
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li He
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
30
|
Yang S, Zheng W, Yang C, Zu R, Ran S, Wu H, Mu M, Sun S, Zhang N, Thorne RF, Guan Y. Integrated Analysis of Hub Genes and MicroRNAs in Human Placental Tissues from In Vitro Fertilization-Embryo Transfer. Front Endocrinol (Lausanne) 2021; 12:774997. [PMID: 34867824 PMCID: PMC8632620 DOI: 10.3389/fendo.2021.774997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/22/2021] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE Supraphysiological hormone exposure, in vitro culture and embryo transfer throughout the in vitro fertilization-embryo transfer (IVF-ET) procedures may affect placental development. The present study aimed to identify differences in genomic expression profiles between IVF-ET and naturally conceived placentals and to use this as a basis for understanding the underlying effects of IVF-ET on placental function. METHODS Full-term human placental tissues were subjected to next-generation sequencing to determine differentially expressed miRNAs (DEmiRs) and genes (DEGs) between uncomplicated IVF-ET assisted and naturally conceived pregnancies. Gene ontology (GO) enrichment analysis and transcription factor enrichment analysis were used for DEmiRs. MiRNA-mRNA interaction and protein-protein interaction (PPI) networks were constructed. In addition, hub genes were obtained by using the STRING database and Cytoscape. DEGs were analyzed using GO and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Differentially expressed miRNAs were validated through qRT-PCR. RESULTS Compared against natural pregnancies, 12 DEmiRs and 258 DEGs were identified in IVF-ET placental tissues. In a validation cohort, it was confirmed that hsa-miR-204-5p, hsa-miR-1269a, and hsa-miR-941 were downregulation, while hsa-miR-4286, hsa-miR-31-5p and hsa-miR-125b-5p were upregulation in IVF-ET placentas. Functional analysis suggested that these differentially expressed genes were significantly enriched in angiogenesis, pregnancy, PI3K-Akt and Ras signaling pathways. The miRNA-mRNA regulatory network revealed the contribution of 10 miRNAs and 109 mRNAs while EGFR was the most highly connected gene among ten hub genes in the PPI network. CONCLUSION Even in uncomplicated IVF-ET pregnancies, differences exist in the placental transcriptome relative to natural pregnancies. Many of the differentially expressed genes in IVF-ET are involved in essential placental functions, and moreover, they provide a ready resource of molecular markers to assess the association between placental function and safety in IVF-ET offspring.
Collapse
Affiliation(s)
- Shuheng Yang
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Zheng
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen Yang
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruowen Zu
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shiyu Ran
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huan Wu
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingkun Mu
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Simin Sun
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nana Zhang
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yichun Guan
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
31
|
Martinez R, Huang W, Samadani R, Mackowiak B, Centola G, Chen L, Conlon IL, Hom K, Kane MA, Fletcher S, Shapiro P. Mechanistic Analysis of an Extracellular Signal-Regulated Kinase 2-Interacting Compound that Inhibits Mutant BRAF-Expressing Melanoma Cells by Inducing Oxidative Stress. J Pharmacol Exp Ther 2021; 376:84-97. [PMID: 33109619 PMCID: PMC7788356 DOI: 10.1124/jpet.120.000266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/06/2020] [Indexed: 11/22/2022] Open
Abstract
Constitutively active extracellular signal-regulated kinase (ERK) 1/2 signaling promotes cancer cell proliferation and survival. We previously described a class of compounds containing a 1,1-dioxido-2,5-dihydrothiophen-3-yl 4-benzenesulfonate scaffold that targeted ERK2 substrate docking sites and selectively inhibited ERK1/2-dependent functions, including activator protein-1-mediated transcription and growth of cancer cells containing active ERK1/2 due to mutations in Ras G-proteins or BRAF, Proto-oncogene B-RAF (Rapidly Acclerated Fibrosarcoma) kinase. The current study identified chemical features required for biologic activity and global effects on gene and protein levels in A375 melanoma cells containing mutant BRAF (V600E). Saturation transfer difference-NMR and mass spectrometry analyses revealed interactions between a lead compound (SF-3-030) and ERK2, including the formation of a covalent adduct on cysteine 252 that is located near the docking site for ERK/FXF (DEF) motif for substrate recruitment. Cells treated with SF-3-030 showed rapid changes in immediate early gene levels, including DEF motif-containing ERK1/2 substrates in the Fos family. Analysis of transcriptome and proteome changes showed that the SF-3-030 effects overlapped with ATP-competitive or catalytic site inhibitors of MAPK/ERK Kinase 1/2 (MEK1/2) or ERK1/2. Like other ERK1/2 pathway inhibitors, SF-3-030 induced reactive oxygen species (ROS) and genes associated with oxidative stress, including nuclear factor erythroid 2-related factor 2 (NRF2). Whereas the addition of the ROS inhibitor N-acetyl cysteine reversed SF-3-030-induced ROS and inhibition of A375 cell proliferation, the addition of NRF2 inhibitors has little effect on cell proliferation. These studies provide mechanistic information on a novel chemical scaffold that selectively regulates ERK1/2-targeted transcription factors and inhibits the proliferation of A375 melanoma cells through a ROS-dependent mechanism. SIGNIFICANCE STATEMENT: Constitutive activation of the extracellular signal-regulated kinase (ERK1/2) pathway drives the proliferation and survival of many cancer cell types. Given the diversity of cellular functions regulated by ERK1/2, the current studies have examined the mechanism of a novel chemical scaffold that targets ERK2 near a substrate binding site and inhibits select ERK functions. Using transcriptomic and proteomic analyses, we provide a mechanistic basis for how this class of compounds inhibits melanoma cells containing mutated BRAF and active ERK1/2.
Collapse
Affiliation(s)
- Ramon Martinez
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| | - Ramin Samadani
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| | - Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| | - Garrick Centola
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| | - Lijia Chen
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| | - Ivie L Conlon
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| | - Kellie Hom
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland
| |
Collapse
|
32
|
Wang F, Xie N, Zhou J, Dai M, Zhang Q, Hardiman PJ, Qu F. Molecular mechanisms underlying altered neurobehavioural development of female offspring of mothers with polycystic ovary syndrome: FOS-mediated regulation of neurotrophins in placenta. EBioMedicine 2020; 60:102993. [PMID: 32949999 PMCID: PMC7501055 DOI: 10.1016/j.ebiom.2020.102993] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This study explored the mechanisms underlying altered neurobehavioural development of female offspring born to mothers with polycystic ovary syndrome (PCOS). METHODS In total, 20 women with PCOS and 32 healthy women who underwent caesarean deliveries with a single female foetus were recruited. Infants were assessed with Dubowitz scoring. Swan71 cell line with stable FOS overexpression was used to verify the regulatory effects of FOS on brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) expression. Learning and memory in female first-generation (F1) and second-generation (F2) offspring in a rat model of PCOS was tested using the Morris water maze at puberty and adulthood. Transcriptome analysis of pubertal hippocampi and hypothalami of female F1 offspring was conducted. FINDINGS Total score and behaviour subscales of Dubowitz scoring were significantly lower in female infants of women with PCOS. FOS and NGF protein levels were downregulated in placental villi of the PCOS group. FOS played a key role in BDNF inhibition and enhancing NGF in Swan71 cells. PCOS female F1 rats exhibited lower target crossing times during puberty when compared to controls. Transcriptome analysis revealed significant changes in hippocampal and hypothalamic neuronal pathways in female F1 rats at puberty. INTERPRETATION FOS regulation of neurotrophins in the placenta negatively affects neurobehavioural development of female offspring of PCOS mothers. FUNDING This study was funded by the National Key R&D Program of China (2018YFC1004900 to F.Q. and F.W.) and the National Natural Science Foundation of China (81874480 to F.Q.; 81873837 to F.W.).
Collapse
Affiliation(s)
- Fangfang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Ningning Xie
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Jue Zhou
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Minchen Dai
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London NW3 2PF, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China; Institute for Women's Health, University College London, London NW3 2PF, United Kingdom.
| |
Collapse
|
33
|
Liao Y, Wang Z, Wang L, Lin Y, Ye Z, Zeng X, Wei F. MicroRNA-27a-3p directly targets FosB to regulate cell proliferation, apoptosis, and inflammation responses in immunoglobulin a nephropathy. Biochem Biophys Res Commun 2020; 529:1124-1130. [PMID: 32819575 DOI: 10.1016/j.bbrc.2020.06.115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 06/23/2020] [Indexed: 01/08/2023]
Abstract
Immunoglobulin A nephropathy (IgAN) constitutes the most common primary glomerulonephritis worldwide; however, the exact pathogenesis of IgAN is unknown. Previous genome-wide analysis of microRNA (miRNA) expression in the kidney has confirmed that miRNAs are closely related to the pathological changes of IgAN. Accordingly, in this study we found that miR-27a-3p is upregulated in IgAN kidney tissues in addition to human podocytes and tubule epithelial HK2 but not mesangial cells. Methylthiazolyldiphenyl-tetrazolium bromide (MTT), flow cytometry, real-time polymerase chain reaction, western blot, and enzyme-linked immunosorbent assays were used to verify the regulatory effects of miR-27a-3p and its inhibition on cell proliferation, apoptosis, and release of inflammatory factors in podocytes and HK2 cells. The target genes of miR-27a-3p were predicted using bioinformatics software; the identity of FosB as a target gene of miR-27a-3p was confirmed by luciferase report assay and western blot. Overall, our findings demonstrated that miR-27a-3p regulates cell apoptosis, cell proliferation, and the release of inflammatory cytokines of human podocytes and HK2 cells by directly targeting FosB. Our results therefore suggested that miR-27a-3p might be associated with the pathophysiology of IgAN and may represent a potential target for further studies related to IgAN mechanism or therapeutics.
Collapse
Affiliation(s)
- Yu Liao
- 2nd Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Ziyan Wang
- 2nd Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; General Hospital of Guangzhou Military Command of PLA, Guangzhou, 510062, China
| | - Lixin Wang
- Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China; 2nd Clinical Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510062, China
| | - Yanzhao Lin
- Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China; 2nd Clinical Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510062, China
| | - Ziyi Ye
- Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China; 2nd Clinical Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510062, China
| | - Xufang Zeng
- Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China; 2nd Clinical Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510062, China
| | - Fangning Wei
- Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China; 2nd Clinical Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510062, China.
| |
Collapse
|
34
|
Transcriptome Analysis of Deafness: Intracellular Signal Transduction Signaling Pathways Regulate Neuroplastic Changes in the Auditory Cortex. Otol Neurotol 2020; 41:986-996. [PMID: 32501934 DOI: 10.1097/mao.0000000000002691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
HYPOTHESIS AND BACKGROUND Hearing loss leads to synaptic changes in auditory neurons and their networks, and functions as a consequence of the interplay between genes and proteins. However, cellular and molecular mechanisms leading to deafness-induced plasticity in the auditory cortex (AC) remain unclear. Here, we examined the changes in gene expression and key signaling pathways that regulate differentially expressed genes (DEGs) in the AC following auditory deafferentation using RNA-sequencing (RNA-Seq) analysis. METHODS Cochlear ablation-induced bilaterally deafened Sprague-Dawley rats were maintained for 12 weeks and their ACs were harvested. RNA-seq analysis was performed on each sample to identify which genes were expressed. This information was then used for comparative analysis of DEGs between samples. The statistical significance of DEGs was determined by fold change (|FC| > 1.5) and independent t test (p < 0.05). RESULTS RNA-seq analysis identified 72 DEGs, of which 19 were upregulated and 53 were down-regulated after bilateral deafening in the ACs. Gene ontology (GO) analysis revealed the potential involvement of mitogen-activated protein kinase, tumor necrosis factor, and cyclic adenosine 3',5'-monophosphate (e.g., Bdnf, Gli1, and c-Fos) signaling pathways in regulating changes in the expression of the genes listed herein. The DEGs of interest-including c-Fos, Arc, Ntf3, and Gli1-from the RNA-seq analysis were consistent with result of quantitative reverse transcriptase polymerase chain reaction. CONCLUSION RNA-seq analysis revealed that auditory deprivation in adult rats elicited changes in gene expression, transcription factor levels, and their complex interaction at specific gene promoters in the AC. Particularly, activation of intracellular signal transduction signaling pathways may be key to neuronal plasticity in deafness.
Collapse
|
35
|
Zheng H, Ton H, Yang L, Liufu N, Dong Y, Zhang Y, Xie Z. Acute Fasting Does Not Induce Cognitive Impairment in Mice. Front Neurosci 2019; 13:896. [PMID: 31507368 PMCID: PMC6718131 DOI: 10.3389/fnins.2019.00896] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Preoperative baseline cognitive impairment is associated with postoperative neurocognitive disorder (PND). Fasting, and more generally, calorie restriction has been shown to exert controversial effects in clinical settings and various animal models of neurological disorders. Every patient needs acute fasting before anesthesia and surgery. However, the impact of acute fasting on cognitive function remain largely unknown. We, therefore, set out to determine whether acute fasting can induce neurotoxicity and neurobehavioral deficits in rodents. In the present system establishment study, a mouse model of acute fasting was established. The effects of the acute fasting on natural and learned behavior were evaluated in the buried food test, open field test and the Y maze test. The expression of c-Fos, the marker of neuronal activation, and caspase-3 activation, the marker of cellular apoptosis, were measured with immunohistochemistry. We found that the 9 h acute fasting increased the latency to eat food in the buried food test. The acute fasting also selectively increased the total distance and decreased the freezing time in open field test, and increased the duration in the novel arm in the Y maze test. Besides, the immunohistochemical study showed that the fasting significantly increased the c-Fos level in the hippocampus and various sub-cortical areas, including paraventricular thalamus (PVT), dorsomedial hypothalamus (DMH), lateral hypothalamus (LH), and basal amygdala (BMA). However, the acute fasting did not induce apoptosis, demonstrating by no appearance of caspase-3 activation in the corresponding brain areas. These data showed that acute fasting did not cause cellular apoptosis and cognitive impairment in the mice. Instead, the acute fasting increased the neuronal activity, enhanced the ambulatory activity and improved the spatial recognition memory in the mice. These findings will promote more research in the established system to further determine the effects of perioperative factors on the postoperative neurocognitive function and the underlying mechanisms.
Collapse
Affiliation(s)
- Hua Zheng
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Hoai Ton
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Lei Yang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States.,Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ning Liufu
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States.,Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
36
|
Holditch SJ, Brown CN, Lombardi AM, Nguyen KN, Edelstein CL. Recent Advances in Models, Mechanisms, Biomarkers, and Interventions in Cisplatin-Induced Acute Kidney Injury. Int J Mol Sci 2019; 20:ijms20123011. [PMID: 31226747 PMCID: PMC6627318 DOI: 10.3390/ijms20123011] [Citation(s) in RCA: 251] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/31/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022] Open
Abstract
Cisplatin is a widely used chemotherapeutic agent used to treat solid tumours, such as ovarian, head and neck, and testicular germ cell. A known complication of cisplatin administration is acute kidney injury (AKI). The development of effective tumour interventions with reduced nephrotoxicity relies heavily on understanding the molecular pathophysiology of cisplatin-induced AKI. Rodent models have provided mechanistic insight into the pathophysiology of cisplatin-induced AKI. In the subsequent review, we provide a detailed discussion of recent advances in the cisplatin-induced AKI phenotype, principal mechanistic findings of injury and therapy, and pre-clinical use of AKI rodent models. Cisplatin-induced AKI murine models faithfully develop gross manifestations of clinical AKI such as decreased kidney function, increased expression of tubular injury biomarkers, and tubular injury evident by histology. Pathways involved in AKI include apoptosis, necrosis, inflammation, and increased oxidative stress, ultimately providing a translational platform for testing the therapeutic efficacy of potential interventions. This review provides a discussion of the foundation laid by cisplatin-induced AKI rodent models for our current understanding of AKI molecular pathophysiology.
Collapse
Affiliation(s)
- Sara J Holditch
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Box C281, 12700 East, 19th Ave, Aurora, CO 80045, USA.
| | - Carolyn N Brown
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Box C281, 12700 East, 19th Ave, Aurora, CO 80045, USA.
| | - Andrew M Lombardi
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Box C281, 12700 East, 19th Ave, Aurora, CO 80045, USA.
| | - Khoa N Nguyen
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Box C281, 12700 East, 19th Ave, Aurora, CO 80045, USA.
| | - Charles L Edelstein
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Box C281, 12700 East, 19th Ave, Aurora, CO 80045, USA.
| |
Collapse
|
37
|
Kadokura H, Yamazaki T, Masuda Y, Kato Y, Hasegawa A, Sakagami H, Yokose S. Establishment of a Primary Culture System of Human Periodontal Ligament Cells that Differentiate into Cementum Protein 1-expressing Cementoblast-like Cells. In Vivo 2019; 33:349-352. [PMID: 30804111 DOI: 10.21873/invivo.11480] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM A better understanding of cementogenesis and cementoblast differentiation would be useful for periodontal therapy. The aim of this study was to establish a cell culture system that reflects cementum formation in periodontal tissue and determine whether or not isolated and cultured primary human periodontal ligament (PDL) cells could be used for the study of the differentiation of cementoblast. MATERIALS AND METHODS PDL cells were isolated from the outgrowths of tissue fragments of human PDL. PDL cells were incubated for up to 21 days in differentiation medium containing β-glycerophosphate and ascorbic acid. The changes in the cells were detected by alkaline phosphatase (ALP) and von Kossa staining. Real-time polymerase chain reaction was also performed for cementum protein 1 (CEMP1), which is a specific marker of cementoblasts and their progenitors. RESULTS On day 5, a small number of PDL cells, which were fibrous, were positive for ALP. On day 7, almost all cells were positive for ALP. On day 14, mineralization nodules appeared, as seen by positive von Kossa staining; the nodules increased in number and size by day 21. The expression of CEMP1 was detected on day 5, and its expression level increased gradually by day 7, reached a peak on day 14, and decreased by day 21. CONCLUSION Human PDL cells were used to establish a culture system that reflects cementum formation. Our results suggested that this culture method is convenient and useful for the study of cementogenesis and cementoblast differentiation.
Collapse
Affiliation(s)
- Hiroshi Kadokura
- Division of Endodontics and Operative Dentistry, Department of Restorative and Biomaterial Sciences, Meikai University School of Dentistry, Saitama, Japan
| | - Takahide Yamazaki
- Division of Endodontics and Operative Dentistry, Department of Restorative and Biomaterial Sciences, Meikai University School of Dentistry, Saitama, Japan
| | - Yoshiko Masuda
- Division of Endodontics and Operative Dentistry, Department of Restorative and Biomaterial Sciences, Meikai University School of Dentistry, Saitama, Japan
| | - Yuka Kato
- Division of Endodontics and Operative Dentistry, Department of Restorative and Biomaterial Sciences, Meikai University School of Dentistry, Saitama, Japan
| | - Akihiko Hasegawa
- Division of Internal Medicine, Department of Comprehensive Medical Sciences, Meikai University School of Dentistry, Saitama, Japan
| | - Hiroshi Sakagami
- Meikai University Research Institute of Odontology (M-RIO), Saitama, Japan
| | - Satoshi Yokose
- Division of Endodontics and Operative Dentistry, Department of Restorative and Biomaterial Sciences, Meikai University School of Dentistry, Saitama, Japan
| |
Collapse
|
38
|
Colmenárez-Raga AC, Díaz I, Pernia M, Pérez-González D, Delgado-García JM, Carro J, Plaza I, Merchán MA. Reversible Functional Changes Evoked by Anodal Epidural Direct Current Electrical Stimulation of the Rat Auditory Cortex. Front Neurosci 2019; 13:356. [PMID: 31031588 PMCID: PMC6473088 DOI: 10.3389/fnins.2019.00356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/28/2019] [Indexed: 12/26/2022] Open
Abstract
Rat auditory cortex was subjected to 0.1 mA anodal direct current in seven 10-min sessions on alternate days. Based on the well-known auditory cortex control of olivocochlear regulation through corticofugal projections, auditory brainstem responses (ABRs) were recorded as an indirect test of the effectiveness and reversibility of the multisession protocol of epidural stimulation. Increases of 20-30 dB ABR auditory thresholds shown after epidural stimulation reverted back to control levels 10 min after a single session. However, increases in thresholds revert 4 days after multisession stimulation. Less changes in wave amplitudes and threshold shifts were shown in ABR recorded contralaterally to the electrically stimulated side of the brain. To assess tissue effects of epidural electric stimulation on the brain cortex, well characterized functional anatomical markers of glial cells (GFAP/astrocytes and Iba1/microglial cells) and neurons (c-Fos) were analyzed in alternate serial sections by quantitative immunocytochemistry. Restricted astroglial and microglial reactivity was observed within the cytoarchitectural limits of the auditory cortex. However, interstitial GFAP overstaining was also observed in the ventricular surface and around blood vessels, thus supporting a potential global electrolytic stimulation of the brain. These results correlate with extensive changes in the distribution of c-Fos immunoreactive neurons among layers along sensory cortices after multisession stimulation. Quantitative immunocytochemical analysis supported this idea by showing a significant increase in the number of positive neurons in supragranular layers and a decrease in layer 6 with no quantitative changes detected in layer 5. Our data indicate that epidural stimulation of the auditory cortex induces a reversible decrease in hearing sensitivity due to local, restricted epidural stimulation. A global plastic response of the sensory cortices, also reported here, may be related to electrolytic effects of electric currents.
Collapse
Affiliation(s)
| | - Iván Díaz
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
| | - Marianny Pernia
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
| | - David Pérez-González
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
| | | | - Juan Carro
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
| | - Ignacio Plaza
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
| | - Miguel A. Merchán
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
| |
Collapse
|
39
|
The Possible Role of Complete Loss of Myostatin in Limiting Excessive Proliferation of Muscle Cells (C2C12) via Activation of MicroRNAs. Int J Mol Sci 2019; 20:ijms20030643. [PMID: 30717351 PMCID: PMC6386905 DOI: 10.3390/ijms20030643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Myostatin (MSTN) is a member of the TGF-β superfamily that negatively regulates skeletal muscle growth and differentiation. However, the mechanism by which complete MSTN deletion limits excessive proliferation of muscle cells remains unclear. In this study, we knocked out MSTN in mouse myoblast lines using a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR/Cas9) system and sequenced the mRNA and miRNA transcriptomes. The results show that complete loss of MSTN upregulates seven miRNAs targeting an interaction network composed of 28 downregulated genes, including TGFB1, FOS and RB1. These genes are closely associated with tumorigenesis and cell proliferation. Our study suggests that complete loss of MSTN may limit excessive cell proliferation via activation of miRNAs. These data will contribute to the treatment of rhabdomyosarcoma (RMS).
Collapse
|
40
|
Çomakli S, Köktürk M, Topal A, Özkaraca M, Ceyhun SB. Immunofluorescence/fluorescence assessment of brain-derived neurotrophic factor, c-Fos activation, and apoptosis in the brain of zebrafish (Danio rerio) larvae exposed to glufosinate. Neurotoxicology 2018; 69:60-67. [DOI: 10.1016/j.neuro.2018.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 09/13/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022]
|
41
|
Hop HT, Arayan LT, Huy TXN, Reyes AWB, Vu SH, Min W, Lee HJ, Rhee MH, Chang HH, Kim S. The Key Role of c-Fos for Immune Regulation and Bacterial Dissemination in Brucella Infected Macrophage. Front Cell Infect Microbiol 2018; 8:287. [PMID: 30186773 PMCID: PMC6110913 DOI: 10.3389/fcimb.2018.00287] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/27/2018] [Indexed: 12/25/2022] Open
Abstract
The cellular oncogene c-Fos (c-Fos) is a component of activator protein 1 (AP1), a master transcriptional regulator of cells. The suppression of c-Fos signaling by siRNA treatment resulted in significant induction of TLR4, which subsequently activates p38 and ERK1/2 mitogen-activated protein kinases (MAPKs) and enhances F-actin polymerization, leading to an increase in B. abortus phagocytosis. During B. abortus infection, c-Fos signaling is induced, which activates the downstream innate-immunity signaling cascade for bacterial clearance. The inhibition of c-Fos signaling led to increased production of interleukin 10 (IL-10), which partially suppressed lysosome-mediated killing, resulting in increased survival of B. abortus inside macrophages. We present evidence of the regulatory role played by the c-Fos pathway in proliferation during B. abortus infection; however, this was independent of the anti-Brucella effect of this pathway. Another finding is the essential contribution of c-Fos/TRAIL to infected-cell necrosis, which is a key event in bacterial dissemination. These data provide the mechanism via which c-Fos participates in host defense mechanisms against Brucella infection and in bacterial dissemination by macrophages.
Collapse
Affiliation(s)
- Huynh T Hop
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Lauren T Arayan
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Tran X N Huy
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Alisha W B Reyes
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Son H Vu
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - WonGi Min
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Hu J Lee
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Man H Rhee
- College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea
| | - Hong H Chang
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, South Korea
| | - Suk Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea.,Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
42
|
Hadipour M, Kaka G, Bahrami F, Meftahi GH, Pirzad Jahromi G, Mohammadi A, Sahraei H. Crocin improved amyloid beta induced long-term potentiation and memory deficits in the hippocampal CA1 neurons in freely moving rats. Synapse 2018; 72:e22026. [DOI: 10.1002/syn.22026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 01/15/2023]
Affiliation(s)
| | - Gholamreza Kaka
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences; Tehran Iran
| | - Farideh Bahrami
- Department of Physiology and Biophysics, Faculty of Medicine; Baqiyatallah University of Medical Sciences; Tehran Iran
| | | | - Gila Pirzad Jahromi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences; Tehran Iran
| | - Alireza Mohammadi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences; Tehran Iran
| | - Hedayat Sahraei
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences; Tehran Iran
| |
Collapse
|
43
|
Huang W, Chen C, Liu X. Hindlimb suspension-induced cell apoptosis in the posterior parietal cortex and lateral geniculate nucleus: corresponding changes in c-Fos protein and the PI3K/Akt signaling pathway. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
44
|
Nakajima H, Furukawa C, Chang YC, Ogata H, Magae J. Delayed Growth Suppression and Radioresistance Induced by Long-Term Continuous Gamma Irradiation. Radiat Res 2017; 188:181-190. [DOI: 10.1667/rr14666.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Hiroo Nakajima
- Department of Breast Surgery, Misugi-kai Sato Hospital, 65-1 Yabuhigashi-machi, Hirakata-shi, Osaka 573-1124, Japan
| | - Chiharu Furukawa
- Department of Biotechnology, Institute of Research and Innovation, 1201 Takada, Kashiwa 277-0861, Japan
| | - Young-Chae Chang
- Department of Cell Biology, Catholic University of Daegu, School of Medicine, 3056-6 Daemyung-4-Dong, Nam-gu, Daegu 705-718, Republic of Korea
| | - Hiromitsu Ogata
- Center for Public Health Informatics, National Institute of Public Health, 2-3-6 Minami, Wako, Saitama 351-0197, Japan
| | - Junji Magae
- Department of Biotechnology, Institute of Research and Innovation, 1201 Takada, Kashiwa 277-0861, Japan
- Center for Public Health Informatics, National Institute of Public Health, 2-3-6 Minami, Wako, Saitama 351-0197, Japan
- Magae Bioscience Institute, 49-4 Fujimidai, Tsukuba 300-1263, Japan
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry, 2-11-1 Iwado Kita, Komae, Tokyo 201-8511, Japan
| |
Collapse
|
45
|
Bail K, Notz Q, Rovituso DM, Schampel A, Wunsch M, Koeniger T, Schropp V, Bharti R, Scholz CJ, Foerstner KU, Kleinschnitz C, Kuerten S. Differential effects of FTY720 on the B cell compartment in a mouse model of multiple sclerosis. J Neuroinflammation 2017; 14:148. [PMID: 28738885 PMCID: PMC5525315 DOI: 10.1186/s12974-017-0924-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/14/2017] [Indexed: 12/11/2022] Open
Abstract
Background MP4-induced experimental autoimmune encephalomyelitis (EAE) is a mouse model of multiple sclerosis (MS), which enables targeted research on B cells, currently much discussed protagonists in MS pathogenesis. Here, we used this model to study the impact of the S1P1 receptor modulator FTY720 (fingolimod) on the autoreactive B cell and antibody response both in the periphery and the central nervous system (CNS). Methods MP4-immunized mice were treated orally with FTY720 for 30 days at the peak of disease or 50 days after EAE onset. The subsequent disease course was monitored and the MP4-specific B cell/antibody response was measured by ELISPOT and ELISA. RNA sequencing was performed to determine any effects on B cell-relevant gene expression. S1P1 receptor expression by peripheral T and B cells, B cell subset distribution in the spleen and B cell infiltration into the CNS were studied by flow cytometry. The formation of B cell aggregates and of tertiary lymphoid organs (TLOs) was evaluated by histology and immunohistochemistry. Potential direct effects of FTY720 on B cell aggregation were studied in vitro. Results FTY720 significantly attenuated clinical EAE when treatment was initiated at the peak of EAE. While there was a significant reduction in the number of T cells in the blood after FTY720 treatment, B cells were only slightly diminished. Yet, there was evidence for the modulation of B cell receptor-mediated signaling upon FTY720 treatment. In addition, we detected a significant increase in the percentage of B220+ B cells in the spleen both in acute and chronic EAE. Whereas acute treatment completely abrogated B cell aggregate formation in the CNS, the numbers of infiltrating B cells and plasma cells were comparable between vehicle- and FTY720-treated mice. In addition, there was no effect on already developed aggregates in chronic EAE. In vitro B cell aggregation assays suggested the absence of a direct effect of FTY720 on B cell aggregation. However, FTY720 impacted the evolution of B cell aggregates into TLOs. Conclusions The data suggest differential effects of FTY720 on the B cell compartment in MP4-induced EAE. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0924-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kathrin Bail
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Quirin Notz
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Damiano M Rovituso
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Andrea Schampel
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Marie Wunsch
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Tobias Koeniger
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Verena Schropp
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Richa Bharti
- Core Unit Systems Medicine, University Hospitals of Würzburg, Würzburg, Germany
| | - Claus-Juergen Scholz
- Core Unit Systems Medicine, University Hospitals of Würzburg, Würzburg, Germany.,LIMES Institute, University of Bonn, Bonn, Germany
| | - Konrad U Foerstner
- Core Unit Systems Medicine, University Hospitals of Würzburg, Würzburg, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany.,Department of Neurology, University Hospital Essen, Essen, Germany
| | - Stefanie Kuerten
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany. .,Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
46
|
Pernia M, Estevez S, Poveda C, Plaza I, Carro J, Juiz JM, Merchan MA. c-Fos and Arc/Arg3.1 expression in auditory and visual cortices after hearing loss: Evidence of sensory crossmodal reorganization in adult rats. J Comp Neurol 2017; 525:2677-2689. [PMID: 28472857 DOI: 10.1002/cne.24233] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 04/03/2017] [Accepted: 04/22/2017] [Indexed: 02/03/2023]
Abstract
Cross-modal reorganization in the auditory and visual cortices has been reported after hearing and visual deficits mostly during the developmental period, possibly underlying sensory compensation mechanisms. However, there are very few data on the existence or nature and timeline of such reorganization events during sensory deficits in adulthood. In this study, we assessed long-term changes in activity-dependent immediate early genes c-Fos and Arc/Arg3.1 in auditory and neighboring visual cortical areas after bilateral deafness in young adult rats. Specifically, we analyzed qualitatively and quantitatively c-Fos and Arc/Arg3.1 immunoreactivity at 15 and 90 days after cochlea removal. We report extensive, global loss of c-Fos and Arc/Arg3.1 immunoreactive neurons in the auditory cortex 15 days after permanent auditory deprivation in adult rats, which is partly reversed 90 days after deafness. Simultaneously, the number and labeling intensity of c-Fos- and Arc/Arg3.1-immunoreactive neurons progressively increase in neighboring visual cortical areas from 2 weeks after deafness and these changes stabilize three months after inducing the cochlear lesion. These findings support plastic, compensatory, long-term changes in activity in the auditory and visual cortices after auditory deprivation in the adult rats. Further studies may clarify whether those changes result in perceptual potentiation of visual drives on auditory regions of the adult cortex.
Collapse
Affiliation(s)
- M Pernia
- Laboratory of Neurobiology of Hearing, Institute of Neurosciences of Castilla y León (Instituto de Neurociencias de Castilla y León - INCYL), University of Salamanca (Universidad de Salamanca - US), Salamanca, Spain
| | - S Estevez
- Laboratory of Neurobiology of Hearing, Institute of Neurosciences of Castilla y León (Instituto de Neurociencias de Castilla y León - INCYL), University of Salamanca (Universidad de Salamanca - US), Salamanca, Spain
| | - C Poveda
- School of Medicine of Albacete, Institute for Research in Neurological Disabilities (Instituto de Investigación en Discapacidades Neurológicas - IDINE), University of Castilla-La Mancha (Universidad de Castilla La Mancha - UCLM), Albacete, Spain
| | - I Plaza
- Laboratory of Neurobiology of Hearing, Institute of Neurosciences of Castilla y León (Instituto de Neurociencias de Castilla y León - INCYL), University of Salamanca (Universidad de Salamanca - US), Salamanca, Spain
| | - J Carro
- Laboratory of Neurobiology of Hearing, Institute of Neurosciences of Castilla y León (Instituto de Neurociencias de Castilla y León - INCYL), University of Salamanca (Universidad de Salamanca - US), Salamanca, Spain
| | - J M Juiz
- School of Medicine of Albacete, Institute for Research in Neurological Disabilities (Instituto de Investigación en Discapacidades Neurológicas - IDINE), University of Castilla-La Mancha (Universidad de Castilla La Mancha - UCLM), Albacete, Spain
| | - M A Merchan
- Laboratory of Neurobiology of Hearing, Institute of Neurosciences of Castilla y León (Instituto de Neurociencias de Castilla y León - INCYL), University of Salamanca (Universidad de Salamanca - US), Salamanca, Spain
| |
Collapse
|
47
|
Novel miRNA-mRNA interactions conserved in essential cancer pathways. Sci Rep 2017; 7:46101. [PMID: 28387377 PMCID: PMC5384238 DOI: 10.1038/srep46101] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/08/2017] [Indexed: 12/23/2022] Open
Abstract
Cancer is a complex disease in which unrestrained cell proliferation results in tumour development. Extensive research into the molecular mechanisms underlying tumorigenesis has led to the characterization of oncogenes and tumour suppressors that are key elements in cancer growth and progression, as well as that of other important elements like microRNAs. These genes and miRNAs appear to be constitutively deregulated in cancer. To identify signatures of miRNA-mRNA interactions potentially conserved in essential cancer pathways, we have conducted an integrative analysis of transcriptomic data, also taking into account methylation and copy number alterations. We analysed 18,605 raw transcriptome samples from The Cancer Genome Atlas covering 15 of the most common types of human tumours. From this global transcriptome study, we recovered known cancer-associated miRNA-targets and importantly, we identified new potential targets from miRNA families, also analysing the phenotypic outcomes of these genes/mRNAs in terms of survival. Further analyses could lead to novel approaches in cancer therapy.
Collapse
|
48
|
Moretto JN, Duffy ÁM, Scharfman HE. Acute restraint stress decreases c-fos immunoreactivity in hilar mossy cells of the adult dentate gyrus. Brain Struct Funct 2017; 222:2405-2419. [PMID: 28190104 DOI: 10.1007/s00429-016-1349-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 12/14/2016] [Indexed: 11/29/2022]
Abstract
Although a great deal of information is available about the circuitry of the mossy cells (MCs) of the dentate gyrus (DG) hilus, their activity in vivo is not clear. The immediate early gene c-fos can be used to gain insight into the activity of MCs in vivo, because c-fos protein expression reflects increased neuronal activity. In prior work, it was identified that control rats that were perfusion-fixed after removal from their home cage exhibited c-fos immunoreactivity (ir) in the DG in a spatially stereotyped pattern: ventral MCs and dorsal granule cells (GCs) expressed c-fos protein (Duffy et al., Hippocampus 23:649-655, 2013). In this study, we hypothesized that restraint stress would alter c-fos-ir, because MCs express glucocorticoid type 2 receptors and the DG is considered to be involved in behaviors related to stress or anxiety. We show that acute restraint using a transparent nose cone for just 10 min led to reduced c-fos-ir in ventral MCs compared to control rats. In these comparisons, c-fos-ir was evaluated 30 min after the 10 min-long period of restraint, and if evaluation was later than 30 min c-fos-ir was no longer suppressed. Granule cells (GCs) also showed suppressed c-fos-ir after acute restraint, but it was different than MCs, because the suppression persisted for over 30 min after the restraint. We conclude that c-fos protein expression is rapidly and transiently reduced in ventral hilar MCs after a brief period of restraint, and suppressed longer in dorsal GCs.
Collapse
Affiliation(s)
- Jillian N Moretto
- The Nathan Kline Institute of Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Áine M Duffy
- The Nathan Kline Institute of Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Helen E Scharfman
- The Nathan Kline Institute of Psychiatric Research, Orangeburg, NY, 10962, USA. .,Departments of Child and Adolescent Psychiatry, Physiology and Neuroscience, and Psychiatry, New York University Langone Medical Center, New York, NY, 10016, USA.
| |
Collapse
|
49
|
Pozhitkov AE, Neme R, Domazet-Lošo T, Leroux BG, Soni S, Tautz D, Noble PA. Tracing the dynamics of gene transcripts after organismal death. Open Biol 2017; 7:160267. [PMID: 28123054 PMCID: PMC5303275 DOI: 10.1098/rsob.160267] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 12/12/2016] [Indexed: 12/13/2022] Open
Abstract
In life, genetic and epigenetic networks precisely coordinate the expression of genes-but in death, it is not known if gene expression diminishes gradually or abruptly stops or if specific genes and pathways are involved. We studied this by identifying mRNA transcripts that apparently increase in relative abundance after death, assessing their functions, and comparing their abundance profiles through postmortem time in two species, mouse and zebrafish. We found mRNA transcript profiles of 1063 genes became significantly more abundant after death of healthy adult animals in a time series spanning up to 96 h postmortem. Ordination plots revealed non-random patterns in the profiles by time. While most of these transcript levels increased within 0.5 h postmortem, some increased only at 24 and 48 h postmortem. Functional characterization of the most abundant transcripts revealed the following categories: stress, immunity, inflammation, apoptosis, transport, development, epigenetic regulation and cancer. The data suggest a step-wise shutdown occurs in organismal death that is manifested by the apparent increase of certain transcripts with various abundance maxima and durations.
Collapse
Affiliation(s)
- Alex E Pozhitkov
- Department of Oral Health Sciences, University of Washington, PO Box 357444, Seattle, WA 98195, USA
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, 24306 Ploen, Germany
| | - Rafik Neme
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, 24306 Ploen, Germany
| | - Tomislav Domazet-Lošo
- Laboratory of Evolutionary Genetics, Division of Molecular Biology, Ruđer Bošković Institute, 10002 Zagreb, Croatia
- Catholic University of Croatia, Ilica 242, Zagreb, Croatia
| | - Brian G Leroux
- Department of Oral Health Sciences, University of Washington, PO Box 357444, Seattle, WA 98195, USA
| | - Shivani Soni
- Department of Biological Sciences, Alabama State University, Montgomery, AL 36101-0271, USA
| | - Diethard Tautz
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, 24306 Ploen, Germany
| | - Peter A Noble
- Department of Periodontics, University of Washington, PO Box 357444, Seattle, WA 98195, USA
- Department of Biological Sciences, Alabama State University, Montgomery, AL 36101-0271, USA
- PhD Program in Microbiology, Alabama State University, Montgomery, AL 36101-0271, USA
| |
Collapse
|
50
|
Ronnekleiv-Kelly SM, Nukaya M, Díaz-Díaz CJ, Megna BW, Carney PR, Geiger PG, Kennedy GD. Aryl hydrocarbon receptor-dependent apoptotic cell death induced by the flavonoid chrysin in human colorectal cancer cells. Cancer Lett 2015; 370:91-9. [PMID: 26515162 DOI: 10.1016/j.canlet.2015.10.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 01/04/2023]
Abstract
The polyphenolic flavone chrysin has been evaluated as a natural chemopreventive agent due to its anti-cancer effects in a variety of cancer cell lines. However, the mechanism of the chemopreventive effect has been not well established, especially in human colorectal cancer cells. We evaluated the chemopreventive effect of chrysin in three different human colorectal cancer cell lines. We found that chrysin treatment consequently reduced cell viability via induction of apoptosis. We identified that the involvement of up-regulation of pro-apoptotic cytokines tumor necrosis factor (Tnf) α and β genes and consequent activation of the TNF-mediated transcriptional pathway in chrysin-induced apoptosis. Using our generated AHR siRNA expressing colorectal cancer cells, we demonstrated that the chrysin-induced up-regulation of Tnfα and β gene expression was dependent on the aryl hydrocarbon receptor (AHR), which is a ligand-receptor for chrysin. Subsequently, we found that the AHR siRNA expressing colorectal cancer cells were resistant to chrysin-induced apoptosis. Therefore, we concluded that AHR is required for the chrysin-induced apoptosis and the up-regulation of Tnfα and β gene expression in human colorectal cancer cells.
Collapse
Affiliation(s)
- Sean M Ronnekleiv-Kelly
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, G4/701A CSC, Madison, WI 53792, USA
| | - Manabu Nukaya
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, G4/701A CSC, Madison, WI 53792, USA
| | - Carol J Díaz-Díaz
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, G4/701A CSC, Madison, WI 53792, USA
| | - Bryant W Megna
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, G4/701A CSC, Madison, WI 53792, USA
| | - Patrick R Carney
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, G4/701A CSC, Madison, WI 53792, USA
| | - Peter G Geiger
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, G4/701A CSC, Madison, WI 53792, USA
| | - Gregory D Kennedy
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, G4/701A CSC, Madison, WI 53792, USA.
| |
Collapse
|