1
|
Jiang N, Hu Z, Wang Q, Hao J, Yang R, Jiang J, Wang H. Fibroblast growth factor 2 enhances BMSC stemness through ITGA2-dependent PI3K/AKT pathway activation. J Cell Physiol 2024:e31423. [PMID: 39188080 DOI: 10.1002/jcp.31423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSC) are promising cellular reservoirs for treating degenerative diseases, tissue injuries, and immune system disorders. However, the stemness of BMSCs tends to decrease during in vitro cultivation, thereby restricting their efficacy in clinical applications. Consequently, investigating strategies that bolster the preservation of BMSC stemness and maximize therapeutic potential is necessary. Transcriptomic and single-cell sequencing methodologies were used to perform a comprehensive examination of BMSCs with the objective of substantiating the pivotal involvement of fibroblast growth factor 2 (FGF2) and integrin alpha 2 (ITGA2) in stemness regulation. To investigate the impact of these genes on the BMSC stemness in vitro, experimental approaches involving loss and gain of function were implemented. These approaches encompassed the modulation of FGF2 and ITGA2 expression levels via small interfering RNA and overexpression plasmids. Furthermore, we examined their influence on the proliferation and differentiation capacities of BMSCs, along with the expression of stemness markers, including octamer-binding transcription factor 4, Nanog homeobox, and sex determining region Y-box 2. Transcriptomic analyzes successfully identified FGF2 and ITGA2 as pivotal genes responsible for regulating the stemness of BMSCs. Subsequent single-cell sequencing revealed that elevated FGF2 and ITGA2 expression levels within specific stem cell subpopulations are closely associated with stemness maintenance. Moreover, additional in vitro experiments have convincingly demonstrated that FGF2 effectively enhances the BMSC stemness by upregulating ITGA2 expression, a process mediated by the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway. This conclusion was supported by the observed upregulation of stemness markers following the induction of FGF2 and ITGA2. Moreover, administration of the BEZ235 pathway inhibitor resulted in the repression of stemness transcription factors, suggesting the substantial involvement of the PI3K/AKT pathway in stemness preservation facilitated by FGF2 and ITGA2. This study elucidates the involvement of FGF2 in augmenting BMSC stemness by modulating ITGA2 and activating the PI3K/AKT pathway. These findings offer valuable contributions to stem cell biology and emphasize the potential of manipulating FGF2 and ITGA2 to optimize BMSCs for therapeutic purposes.
Collapse
Affiliation(s)
- Nizhou Jiang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
- Department of Spine Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhenxin Hu
- Department of Spine Surgery, Peking University Fourth School of Clinical Medicine, Beijing Jishuitan Hospital, Beijing, China
| | - Quanxiang Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Jiayu Hao
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Rui Yang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Jian Jiang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Hong Wang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| |
Collapse
|
2
|
Lee H, Park W, An G, Park J, Lim W, Song G. Hexaconazole induces developmental toxicities via apoptosis, inflammation, and alterations of Akt and MAPK signaling cascades. Comp Biochem Physiol C Toxicol Pharmacol 2024; 279:109872. [PMID: 38423198 DOI: 10.1016/j.cbpc.2024.109872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/16/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Hexaconazole is a highly effective triazole fungicide that is frequently applied in various countries to elevate crop productivity. Given its long half-life and high water solubility, this fungicide is frequently detected in the environment, including water sources. Moreover, hexaconazole exerts hazardous effects on nontarget organisms. However, little is known about the toxic effects of hexaconazole on animal development. Thus, this study aimed to investigate the developmental toxicity of hexaconazole to zebrafish, a valuable animal model for toxicological studies, and elucidate the underlying mechanisms. Results showed that hexaconazole affected the viability and hatching rate of zebrafish at 96 h postfertilization. Hexaconazole-treated zebrafish showed phenotypic defects, such as reduced size of head and eyes and enlarged pericardiac edema. Moreover, hexaconazole induced apoptosis, DNA fragmentation, and inflammation in developing zebrafish. Various organ defects, including neurotoxicity, cardiovascular toxicity, and hepatotoxicity, were observed in transgenic zebrafish models olig2:dsRed, fli1:eGFP, and l-fabp:dsRed. Furthermore, hexaconazole treatment altered the Akt and MAPK signaling pathways, which possibly triggered the organ defects and other toxic mechanisms. This study demonstrated the developmental toxicity of hexaconazole to zebrafish and elucidated the underlying mechanisms.
Collapse
Affiliation(s)
- Hojun Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Wonhyoung Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Garam An
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Junho Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
3
|
Xie X, Zhang X, Li S, Du W. Involvement of Fgf2-mediated tau protein phosphorylation in cognitive deficits induced by sevoflurane in aged rats. Mol Med 2024; 30:39. [PMID: 38493090 PMCID: PMC10943822 DOI: 10.1186/s10020-024-00784-0] [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: 10/30/2023] [Accepted: 01/11/2024] [Indexed: 03/18/2024] Open
Abstract
OBJECTIVE Anesthetics have been linked to cognitive alterations, particularly in the elderly. The current research delineates how Fibroblast Growth Factor 2 (Fgf2) modulates tau protein phosphorylation, contributing to cognitive impairments in aged rats upon sevoflurane administration. METHODS Rats aged 3, 12, and 18 months were subjected to a 2.5% sevoflurane exposure to form a neurotoxicity model. Cognitive performance was gauged, and the GEO database was employed to identify differentially expressed genes (DEGs) in the 18-month-old cohort post sevoflurane exposure. Bioinformatics tools, inclusive of STRING and GeneCards, facilitated detailed analysis. Experimental validations, both in vivo and in vitro, examined Fgf2's effect on tau phosphorylation. RESULTS Sevoflurane notably altered cognitive behavior in older rats. Out of 128 DEGs discerned, Fgf2 stood out as instrumental in regulating tau protein phosphorylation. Sevoflurane exposure spiked Fgf2 expression in cortical neurons, intensifying tau phosphorylation via the PI3K/AKT/Gsk3b trajectory. Diminishing Fgf2 expression correspondingly curtailed tau phosphorylation, neurofibrillary tangles, and enhanced cognitive capacities in aged rats. CONCLUSION Sevoflurane elicits a surge in Fgf2 expression in aging rats, directing tau protein phosphorylation through the PI3K/AKT/Gsk3b route, instigating cognitive aberrations.
Collapse
Affiliation(s)
- Xin Xie
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dandong District, Liaoning Province, Shenyang, 110042, P. R. China
| | - Xiaomin Zhang
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dandong District, Liaoning Province, Shenyang, 110042, P. R. China
| | - Songze Li
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dandong District, Liaoning Province, Shenyang, 110042, P. R. China
| | - Wei Du
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dandong District, Liaoning Province, Shenyang, 110042, P. R. China.
| |
Collapse
|
4
|
Lee H, An G, Park J, You J, Song G, Lim W. Mevinphos induces developmental defects via inflammation, apoptosis, and altered MAPK and Akt signaling pathways in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2024; 275:109768. [PMID: 37858660 DOI: 10.1016/j.cbpc.2023.109768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
Mevinphos, an organophosphate insecticide, is widely used to control pests and enhance crop yield. Because of its high solubility, it can easily flow into water and threaten the aquatic environment, and it is known to be hazardous to non-target organisms. However, little is known about its developmental toxicity and the underlying toxic mechanisms. In this study, we utilized zebrafish, which is frequently used for toxicological research to estimate the toxicity in other aquatic organisms or vertebrates including humans, to elucidate the developmental defects induced by mevinphos. Here, we observed that mevinphos induced various phenotypical abnormalities, such as diminished eyes and head sizes, shortened body length, loss of swim bladder, and increased pericardiac edema. Also, exposure to mevinphos triggered inflammation, apoptosis, and DNA fragmentation in zebrafish larvae. In addition, MAPK and Akt signaling pathways, which control apoptosis, inflammation, and proper development of various organs, were also altered by the treatment of mevinphos. Furthermore, these factors induced various organ defects which were confirmed by various transgenic models. We identified neuronal toxicity through transgenic olig2:dsRed zebrafish, cardiovascular toxicity through transgenic fli1:eGFP zebrafish, and hepatotoxicity and pancreatic toxicity through transgenic lfabp:dsRed;elastase:GFP zebrafish. Overall, our results elucidated the developmental toxicities of mevinphos in zebrafish and provided the parameters for the assessment of toxicities in aquatic environments.
Collapse
Affiliation(s)
- Hojun Lee
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Garam An
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Junho Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Jeankyoung You
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
5
|
Sills ES, Harrity C, Wood SH, Tan SL. mTOR Inhibition via Low-Dose, Pulsed Rapamycin with Intraovarian Condensed Platelet Cytokines: An Individualized Protocol to Recover Diminished Reserve? J Pers Med 2023; 13:1147. [PMID: 37511761 PMCID: PMC10381109 DOI: 10.3390/jpm13071147] [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: 06/15/2023] [Revised: 07/03/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
No major breakthroughs have entered mainstream clinical fertility practice since egg donation and intracytoplasmic sperm injection decades ago, and oocyte deficits secondary to advanced age continue as the main manifestation of diminished ovarian reserve. In the meantime, several unproven IVF 'accessories' have emerged including so-called ovarian rejuvenation which entails placing fresh autologous platelet-rich plasma (PRP) directly into ovarian tissue. Among cellular responses attributed to this intervention are reduced oxidative stress, slowed apoptosis and improved metabolism. Besides having an impact on the existing follicle pool, platelet growth factors might also facilitate de novo oocyte recruitment by specified gene upregulation targeting uncommitted ovarian stem cells. Given that disordered activity at the mechanistic target of rapamycin (mTOR) has been shown to exacerbate or accelerate ovarian aging, PRP-discharged plasma cytokines combined with mTOR suppression by pulsed/cyclic rapamycin represents a novel fusion technique to enhance ovarian function. While beneficial effects have already been observed experimentally in oocytes and embryos with mTOR inhibition alone, this proposal is the first to discuss intraovarian platelet cytokines followed by low-dose, phased rapamycin. For refractory cases, this investigational, tailored approach could amplify or sustain ovarian capacity sufficient to permit retrieval of competent oocytes via distinct but complementary pathways-thus reducing dependency on oocyte donation.
Collapse
Affiliation(s)
- E Scott Sills
- Plasma Research Section, Regenerative Biology Group/CAG, San Clemente, CA 92673, USA
- Department of Obstetrics & Gynecology, Palomar Medical Center, Escondido, CA 92029, USA
| | - Conor Harrity
- Department of Obstetrics & Gynaecology, Royal College of Surgeons in Ireland, D02 HC66 Dublin, Ireland
| | - Samuel H Wood
- Department of Obstetrics & Gynecology, Palomar Medical Center, Escondido, CA 92029, USA
- Gen 5 Fertility Center, San Diego, CA 92121, USA
| | - Seang Lin Tan
- OriginElle Fertility Clinic, Montreal, QC H4A 3J3, Canada
- Department of Obstetrics & Gynecology, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
6
|
Bicalutamide, an androgen receptor antagonist, effectively alleviate allergic rhinitis via suppression of PI3K-PKB activity. Eur Arch Otorhinolaryngol 2023; 280:703-711. [PMID: 35831673 PMCID: PMC9849187 DOI: 10.1007/s00405-022-07538-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/01/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVES To investigate the therapeutic effect of Bicalutamide, an androgen receptor antagonist on the onset and development of allergic rhinitis in an animal model. METHODS 40 male BALB/c mice were randomly divided into five groups (eight mice per group). Aluminum hydroxide powder was used as an adjuvant, combined with Ovalbumin (OVA) to establish the mouse model of allergic rhinitis via ultrasonic nebulization of OVA to stimulate the nasal cavity. Mice in Bica#1 group were intraperitoneally injected with 0.02 mg Bicalutamide/0.5 ml of normal saline daily for 7 consecutive days; mice in Bica#2 group were administered 0.02 mg Bicalutamide/0.5 ml of normal saline via intraperitoneal injection for 5 consecutive days, and then the same amount of normal saline was injected intraperitoneally for 2 consecutive days. Enzyme-linked immunosorbent assay was adopted to detect the serological levels of IgE, IL-4, and IL-6 production. Eosinophil infiltration was observed under microscope after hematoxylin and eosin staining of nasal mucosa. Quantitative PCR and Western blot were employed for determination of histamine receptors mRNA expression and PI3K/PKB associated protein levels, respectively. RESULTS Histological analysis shown that allergic lesion was induced after OVA sensitization. Intraperitoneal injection with 0.02 mg Bicalutamide daily for 7 consecutive days significantly reduced the allergic lesion; however, mice injected with the same amount of normal saline at the same time demonstrated no allergic rhinitis symptoms. In addition, there was a significant reduction in eosinophils number in Bicalutamide treated mice (n = 8) compared to the OVA group (n = 8) (OVA: 19.6 ± 5.3 vs. Bica#1: 7.7 ± 0.8 vs. Bica#2: 9.4 ± 1.2, both p < 0.01). Furthermore, ELISA results revealed that the serological levels of IgE (OVA: 17.3 ± 1.7 µg/ml vs. Bica#1: 9.2 ± 0.6 vs. Bica#2: 10.4 ± 2.3, both p < 0.05), IL-4 (OVA: 164.3 ± 5.1 pg/ml vs. Bica#1: 110.2 ± 3.1 vs. Bica#2: 115.3 ± 4.1, both p < 0.05) and IL-6 (OVA: 167.3 ± 3.7 pg/ml vs. Bica#1: 117.5 ± 6.5 vs. Bica#2: 114.8 ± 2.4, both p < 0.05) were significantly decreased after two different dosage of Bicalutamide treatment. Similarly, histamine receptors in mast cells were significantly reduced after two different dosage of Bicalutamide treatment. More importantly, p-PKB protein was notably reduced after two different dosage of Bicalutamide treatment compared to the OVA group, mTOR protein levels were also down regulated after two different dosage of Bicalutamide treatment. CONCLUSIONS Our data demonstrated that androgen receptor antagonist Bicalutamide can significantly alleviate allergic rhinitis lesion in the animal model. PI3K/PKB activity in mast cells was suppressed after Bicalutamide injection. Our results provide important implication in allergic rhinitis prevention and treatment.
Collapse
|
7
|
Eupafolin regulates non-small-cell lung cancer cell proliferation, migration, and invasion by suppressing MMP9 and RhoA via FAK/PI3K/AKT signaling pathway. J Biosci 2023. [DOI: 10.1007/s12038-022-00323-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
8
|
Bustamante-Barrientos FA, Méndez-Ruette M, Molina L, Koning T, Ehrenfeld P, González CB, Wyneken U, Henzi R, Bátiz LF. Alpha-SNAP (M105I) mutation promotes neuronal differentiation of neural stem/progenitor cells through overactivation of AMPK. Front Cell Dev Biol 2023; 11:1061777. [PMID: 37113766 PMCID: PMC10127105 DOI: 10.3389/fcell.2023.1061777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 03/13/2023] [Indexed: 04/29/2023] Open
Abstract
Background: The M105I point mutation in α-SNAP (Soluble N-ethylmaleimide-sensitive factor attachment protein-alpha) leads in mice to a complex phenotype known as hyh (hydrocephalus with hop gait), characterized by cortical malformation and hydrocephalus, among other neuropathological features. Studies performed by our laboratory and others support that the hyh phenotype is triggered by a primary alteration in embryonic neural stem/progenitor cells (NSPCs) that leads to a disruption of the ventricular and subventricular zones (VZ/SVZ) during the neurogenic period. Besides the canonical role of α-SNAP in SNARE-mediated intracellular membrane fusion dynamics, it also negatively modulates AMP-activated protein kinase (AMPK) activity. AMPK is a conserved metabolic sensor associated with the proliferation/differentiation balance in NSPCs. Methods: Brain samples from hyh mutant mice (hydrocephalus with hop gait) (B6C3Fe-a/a-Napahyh/J) were analyzed by light microscopy, immunofluorescence, and Western blot at different developmental stages. In addition, NSPCs derived from WT and hyh mutant mice were cultured as neurospheres for in vitro characterization and pharmacological assays. BrdU labeling was used to assess proliferative activity in situ and in vitro. Pharmacological modulation of AMPK was performed using Compound C (AMPK inhibitor) and AICAR (AMPK activator). Results: α-SNAP was preferentially expressed in the brain, showing variations in the levels of α-SNAP protein in different brain regions and developmental stages. NSPCs from hyh mice (hyh-NSPCs) displayed reduced levels of α-SNAP and increased levels of phosphorylated AMPKα (pAMPKαThr172), which were associated with a reduction in their proliferative activity and a preferential commitment with the neuronal lineage. Interestingly, pharmacological inhibition of AMPK in hyh-NSPCs increased proliferative activity and completely abolished the increased generation of neurons. Conversely, AICAR-mediated activation of AMPK in WT-NSPCs reduced proliferation and boosted neuronal differentiation. Discussion: Our findings support that α-SNAP regulates AMPK signaling in NSPCs, further modulating their neurogenic capacity. The naturally occurring M105I mutation of α-SNAP provokes an AMPK overactivation in NSPCs, thus connecting the α-SNAP/AMPK axis with the etiopathogenesis and neuropathology of the hyh phenotype.
Collapse
Affiliation(s)
| | - Maxs Méndez-Ruette
- Neuroscience Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- PhD Program in Biomedicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Luis Molina
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| | - Tania Koning
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Carlos B. González
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Ursula Wyneken
- Neuroscience Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- School of Medicine, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Roberto Henzi
- Neuroscience Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Laboratorio de Reproducción Animal, Escuela de Medicina Veterinaria, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
- *Correspondence: Luis Federico Bátiz, ; Roberto Henzi,
| | - Luis Federico Bátiz
- Neuroscience Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- School of Medicine, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- *Correspondence: Luis Federico Bátiz, ; Roberto Henzi,
| |
Collapse
|
9
|
Arthur R, Navik U, Kumar P. Repurposing artemisinins as neuroprotective agents: a focus on the PI3k/Akt signalling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 396:593-605. [PMID: 36464748 DOI: 10.1007/s00210-022-02350-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/22/2022] [Indexed: 12/09/2022]
Abstract
Artemisinin and its derivatives, since their discovery by professor Tu Youyou in the early 1970s, have been the bedrock for the management of malaria globally. Recent works have implied that they could be used to manage other diseases including neurodegenerative disorders. Neurodegenerative disorders mainly occur in the adult population resulting from a progressive deterioration of neuronal structures. These include Parkinson's disease (PD), Alzheimer's disease (AD), Huntington's disease (HD), and Multiple sclerosis (MS), among others. The PI3K/Akt signaling pathway plays a significant role in the central nervous system. It has been investigated extensively for its role in central nervous system physiological processes such as cell survival, autophagy, neuronal proliferation, and synaptic plasticity. Therefore, the modulation of this pathway will be crucial in the management of neurodegenerative disorders. This review seeks to compile most of the research findings on the possible neuroprotective role of artemisinins with special emphasis on their modulatory role on the PI3k/Akt pathway. A literature survey was conducted on PubMed, EBSCO, Web of Science, and EMBASE using the keyword artemisinins, and a total of 10,281 articles were retrieved from 1956 to 2022. Among these, 120 articles were examined using Mesh words like PI3k/Akt, neurodegeneration, and neuroinflammation coupled with boolean operators. Most research revealed that artemisinins could help neurodegenerative disorders by modulating the PI3k/Akt with subsequent inhibition of oxidative stress, neuroinflammation, and apoptosis. This paper illustrates that artemisinins could be repurposed as a neuroprotective agent.
Collapse
|
10
|
Schmidt V, Hogan AE, Fallon PG, Schwartz C. Obesity-Mediated Immune Modulation: One Step Forward, (Th)2 Steps Back. Front Immunol 2022; 13:932893. [PMID: 35844529 PMCID: PMC9279727 DOI: 10.3389/fimmu.2022.932893] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 05/27/2022] [Indexed: 11/15/2022] Open
Abstract
Over the past decades, the relationship between the immune system and metabolism has become a major research focus. In this arena of immunometabolism the capacity of adipose tissue to secrete immunomodulatory molecules, including adipokines, within the underlying low-grade inflammation during obesity brought attention to the impact obesity has on the immune system. Adipokines, such as leptin and adiponectin, influence T cell differentiation into different T helper subsets and their activation during immune responses. Furthermore, within the cellular milieu of adipose tissue nutrient availability regulates differentiation and activation of T cells and changes in cellular metabolic pathways. Upon activation, T cells shift from oxidative phosphorylation to oxidative glycolysis, while the differential signaling of the kinase mammalian target of rapamycin (mTOR) and the nuclear receptor PPARγ, amongst others, drive the subsequent T cell differentiation. While the mechanisms leading to a shift from the typical type 2-dominated milieu in lean people to a Th1-biased pro-inflammatory environment during obesity are the subject of extensive research, insights on its impact on peripheral Th2-dominated immune responses become more evident. In this review, we will summarize recent findings of how Th2 cells are metabolically regulated during obesity and malnutrition, and how these states affect local and systemic Th2-biased immune responses.
Collapse
Affiliation(s)
- Viviane Schmidt
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Andrew E. Hogan
- Kathleen Lonsdale Human Health Institute, Maynooth University, Maynooth, Ireland
- Obesity Immunology Research, St. Vincent’s University Hospital and University College Dublin, Dublin, Ireland
| | - Padraic G. Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Christian Schwartz
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- *Correspondence: Christian Schwartz,
| |
Collapse
|
11
|
Tan H, Li M, Han L, Zhao Y, Zhang X. Gypensapogenin I Suppresses Cell Proliferation in Triple-Negative Breast Cancer Via Triggering the Closure of AKT/GSK3β/β-Catenin and Notch-1 Signaling Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:5438-5449. [PMID: 35465659 DOI: 10.1021/acs.jafc.2c02512] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Jiaogulan (Gynostemma pentaphyllum) tea is a functional food that is commercially available worldwide. Gypensapogenin I (Gyp I), which is a natural damarane-type saponin, was obtained from the hydrolysates of total gypenosides. The present research was performed to investigate the potential antiproliferation effect of Gyp I in MDA-MB-231 cells and the underlying mechanisms. Here, we found that Gyp I attenuated survival, inhibited proliferation, and induced apoptosis in MDA-MB-231 cells. Target prediction by binding molecule docking and western blot assays confirmed the mechanism by which Gyp I inhibited the proliferation of breast cancer cells via the AKT/GSK3β/β-catenin signaling pathway. We also showed that Gyp I exhibited superior in vivo efficacy that was dose dependent. Tumor tissue transcriptome analysis indicated that Gyp I could decrease the expression levels of NOTCH1 and HES1, which was in contrast to the effect on MAML and NUMBL, indicating that our compound hindered the activation of the Notch-1 signaling pathway. In summary, we report for the first time that Gyp I shows excellent anti-breast cancer activity in vivo and in vitro and that its pathway of action is related to the AKT/GSK3β/β-catenin and Notch-1 signaling pathways. Therefore, Jiaogulan tea can not only be used as a health food but also possesses the possibility to treat triple-negative breast cancer (TNBC).
Collapse
Affiliation(s)
- Hongyan Tan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Minjie Li
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linlin Han
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuqing Zhao
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaoshu Zhang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
12
|
Computational identification of host genomic biomarkers highlighting their functions, pathways and regulators that influence SARS-CoV-2 infections and drug repurposing. Sci Rep 2022; 12:4279. [PMID: 35277538 PMCID: PMC8915158 DOI: 10.1038/s41598-022-08073-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 02/15/2022] [Indexed: 12/13/2022] Open
Abstract
The pandemic threat of COVID-19 has severely destroyed human life as well as the economy around the world. Although, the vaccination has reduced the outspread, but people are still suffering due to the unstable RNA sequence patterns of SARS-CoV-2 which demands supplementary drugs. To explore novel drug target proteins, in this study, a transcriptomics RNA-Seq data generated from SARS-CoV-2 infection and control samples were analyzed. We identified 109 differentially expressed genes (DEGs) that were utilized to identify 10 hub-genes/proteins (TLR2, USP53, GUCY1A2, SNRPD2, NEDD9, IGF2, CXCL2, KLF6, PAG1 and ZFP36) by the protein–protein interaction (PPI) network analysis. The GO functional and KEGG pathway enrichment analyses of hub-DEGs revealed some important functions and signaling pathways that are significantly associated with SARS-CoV-2 infections. The interaction network analysis identified 5 TFs proteins and 6 miRNAs as the key regulators of hub-DEGs. Considering 10 hub-proteins and 5 key TFs-proteins as drug target receptors, we performed their docking analysis with the SARS-CoV-2 3CL protease-guided top listed 90 FDA approved drugs. We found Torin-2, Rapamycin, Radotinib, Ivermectin, Thiostrepton, Tacrolimus and Daclatasvir as the top ranked seven candidate drugs. We investigated their resistance performance against the already published COVID-19 causing top-ranked 11 independent and 8 protonated receptor proteins by molecular docking analysis and found their strong binding affinities, which indicates that the proposed drugs are effective against the state-of-the-arts alternatives independent receptor proteins also. Finally, we investigated the stability of top three drugs (Torin-2, Rapamycin and Radotinib) by using 100 ns MD-based MM-PBSA simulations with the two top-ranked proposed receptors (TLR2, USP53) and independent receptors (IRF7, STAT1), and observed their stable performance. Therefore, the proposed drugs might play a vital role for the treatment against different variants of SARS-CoV-2 infections.
Collapse
|
13
|
Maharati A, Zanguei AS, Khalili-Tanha G, Moghbeli M. MicroRNAs as the critical regulators of tyrosine kinase inhibitors resistance in lung tumor cells. Cell Commun Signal 2022; 20:27. [PMID: 35264191 PMCID: PMC8905758 DOI: 10.1186/s12964-022-00840-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/05/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the second most common and the leading cause of cancer related deaths globally. Tyrosine Kinase Inhibitors (TKIs) are among the common therapeutic strategies in lung cancer patients, however the treatment process fails in a wide range of patients due to TKIs resistance. Given that the use of anti-cancer drugs can always have side effects on normal tissues, predicting the TKI responses can provide an efficient therapeutic strategy. Therefore, it is required to clarify the molecular mechanisms of TKIs resistance in lung cancer patients. MicroRNAs (miRNAs) are involved in regulation of various pathophysiological cellular processes. In the present review, we discussed the miRNAs that have been associated with TKIs responses in lung cancer. MiRNAs mainly exert their role on TKIs response through regulation of Tyrosine Kinase Receptors (TKRs) and down-stream signaling pathways. This review paves the way for introducing a panel of miRNAs for the prediction of TKIs responses in lung cancer patients. Video Abstract
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zanguei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Rosa-Fernandes L, Bedrat A, dos Santos MLB, Pinto A, Lucena E, Silva TP, Melo RC, Palmisano G, Cardoso CA, Barbosa RH. Global RNAseq of ocular cells reveals gene dysregulation in both asymptomatic and with Congenital Zika Syndrome infants exposed prenatally to Zika virus. Exp Cell Res 2022; 414:113086. [DOI: 10.1016/j.yexcr.2022.113086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/24/2022]
|
15
|
Lee J, Hong EM, Kim JH, Kim JH, Jung JH, Park SW, Koh DH, Jang HJ. Ursodeoxycholic acid shows antineoplastic effects in bile duct cancer cells via apoptosis induction; p53 activation; and EGFR-ERK, COX-2, and PI3K-AKT pathway inhibition. Mol Biol Rep 2021; 48:6231-6240. [PMID: 34392440 DOI: 10.1007/s11033-021-06331-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/31/2021] [Indexed: 11/24/2022]
Abstract
Unlike in normal cells, ursodeoxycholic acid (UDCA) causes apoptosis rather than protection in cancer cells. Aim of this study was to demonstrate whether UDCA actually inhibits proliferation and induces apoptosis in bile duct cancer cells; the effect of UDCA on the expression of COX-2, PI3K/AKT, ERK, and EGFR; how UDCA affects cancer cell invasiveness and metastasis, since these effects are not established in bile duct cancer cells. SNU-245 cells (human extrahepatic bile duct cancer cells) were cultured. MTT assays were performed to evaluate the effect of UDCA on the cell proliferation. A cell death detection enzyme-linked immunosorbent assay and a caspase-3 activity assay were used to determine apoptosis. Western blot analysis measured expression levels of various proteins. The invasiveness of the cancer cells was evaluated by invasion assay. In cultured bile duct cancer cells, UDCA suppressed cell proliferation in bile duct cancer cells by inducing apoptosis and p53 activation, blocking deoxycholic acid (DCA)-induced activated EGFR-ERK signaling and COX-2, inhibiting DCA-induced activated PI3K-AKT signaling, and suppressing the invasiveness of bile duct cancer cells. In addition, a MEK inhibitor impaired UDCA-induced apoptosis in bile duct cancer cells. UDCA has antineoplastic and apoptotic effects in bile duct cancer cells. Thus, UDCA could be a chemopreventive agent in patients with a high risk of cancer, and/or a therapeutic option that enhances other chemotherapeutics.
Collapse
Affiliation(s)
- Jin Lee
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Gangwon-Do, South Korea. .,Division of Gastroenterology, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-Gil, Hwasung, Gyeonggi-Do, 18450, South Korea.
| | - Eun Mi Hong
- Division of Gastroenterology, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-Gil, Hwasung, Gyeonggi-Do, 18450, South Korea
| | - Jung Han Kim
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Gangwon-Do, South Korea
| | - Jung Hee Kim
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Gangwon-Do, South Korea.,Division of Gastroenterology, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-Gil, Hwasung, Gyeonggi-Do, 18450, South Korea
| | - Jang Han Jung
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Gangwon-Do, South Korea.,Division of Gastroenterology, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-Gil, Hwasung, Gyeonggi-Do, 18450, South Korea
| | - Se Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Gangwon-Do, South Korea.,Division of Gastroenterology, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-Gil, Hwasung, Gyeonggi-Do, 18450, South Korea
| | - Dong Hee Koh
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Gangwon-Do, South Korea.,Division of Gastroenterology, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-Gil, Hwasung, Gyeonggi-Do, 18450, South Korea
| | - Hyun Joo Jang
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Gangwon-Do, South Korea.,Division of Gastroenterology, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-Gil, Hwasung, Gyeonggi-Do, 18450, South Korea
| |
Collapse
|
16
|
Wang J, Zhang Y, Xiao Y, Yuan X, Li P, Wang X, Duan Y, Seewaldt VL, Yu D. Boosting immune surveillance by low-dose PI3K inhibitor facilitates early intervention of breast cancer. Am J Cancer Res 2021; 11:2005-2024. [PMID: 34094666 PMCID: PMC8167687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 12/07/2020] [Indexed: 06/12/2023] Open
Abstract
Prevention of estrogen receptor-negative (ER-) breast cancer is an unmet challenge, although tamoxifen and aromatase inhibitors can successfully decrease the incidence of ER-positive (ER+) breast cancer. PI3K pathway activation has been detected in tamoxifen-resistant ER- breast lesions of patients. Here, we further ratified that the PI3K pathway is significantly activated in premalignant ER- breast lesions compared with paired normal tissues of patients, which prompted our assessment of targeting PI3K on inhibition of ER- mammary tumor initiation and progression. Both genetic knockdown of PIK3CA or intervention with low-doses of a PI3K inhibitor (GDC-0941) prevented the dysplasia phenotype of semi-transformed human ER- mammary epithelial cells in 3-dimensional culture in vitro. Importantly, low-dose GDC-0941 treatment significantly delayed mammary tumor initiation in the MMTV-neu mouse model without exhibiting discernable adverse effects. Interestingly, increased CD8+/GZMB+ T-cells were detected in mammary tissue after GDC-0941 treatment, suggesting enhanced immune surveillance. Mechanistically, elevated expression of potent T-cell chemo-attractants, including CCL5 and CXCL10, were detected both in vitro and in vivo after GDC-0941 treatment. Furthermore, inhibition of PI3K significantly increased T-cell recruitment in a CCL5/CXCL10-dependent manner. In human ER- breast cancer, PI3K activation is correlated with significantly reduced CCL5, CXCL10 and CD8A expression, suggesting that the decreased CD8+ T-cell recruitment and escape of immune surveillance may contribute to ER- breast cancer development. In summary, our study indicates that low-dose PI3K inhibitor treatment may intervene early stage ER- breast cancer development by enhancing immune surveillance via CCL5/CXCL10.
Collapse
Affiliation(s)
- Jinyang Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- Current address: Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou, Guangdong, China
| | - Yuan Zhang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Yi Xiao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Xiangliang Yuan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Ping Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Xiao Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Yimin Duan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Victoria L Seewaldt
- Department of Population Sciences, City of HopeDuarte, California, USA
- Comprehensive Cancer Center, City of HopeDuarte, California, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical SciencesHouston, Texas, USA
| |
Collapse
|
17
|
Yang L, Zhao S, Zhu T, Zhang J. GPRC5A Is a Negative Regulator of the Pro-Survival PI3K/Akt Signaling Pathway in Triple-Negative Breast Cancer. Front Oncol 2021; 10:624493. [PMID: 33680947 PMCID: PMC7928339 DOI: 10.3389/fonc.2020.624493] [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: 10/31/2020] [Accepted: 12/30/2020] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is one of the most common types of malignancy worldwide; however, its underlying mechanisms remain unclear. In the present study, we investigated the roles of G-protein-coupled receptor family C, member 5, group A (GPRC5A) in cell apoptosis in triple-negative breast cancer (TNBC). The expression of GPRC5A in breast cancer cell lines was detected by real time PCR and western blot. And the results suggested that GPRC5A was downregulated in breast cancer cell lines compared to normal breast epithelial cell lines. Additionally, the expression of GPRC5A in TCGA database was analyzed in silico. GPRC5A exhibited the lowest expression levels in TNBC compared to ER+ and HER2+ breast cancer. Overexpression of GPRC5A in MDA-MB-231 and MDA-MB-468 cells promoted apoptosis, whereas depletion of GPRC5A in T47D and MCF7 cells inhibited cell apoptosis via the intrinsic apoptotic pathway. We performed RNA-sequencing in GPRC5A overexpressed MDA-MB-231 and the control cells. The results facilitated the identification of a number of signaling pathways involved in this process, and the PI3K/Akt signaling pathway was found to be one the most important. A specific activator of the PI3K/Akt signaling pathway inhibited apoptosis of breast cancer cells, whereas cotreatment of this activator with a GPRC5A-expressing plasmid reduced this effect. Similarly, a specific inhibitor of the PI3K/Akt signaling pathway increased cell apoptosis by activating caspase-3 and caspase-9, whereas co-incubation of the inhibitor with a short hairpin RNA targeting GPRC5A significantly reduced the cell apoptotic rate. Additionally, the overexpression of GPRC5A suppressed tumor growth by inducing cell apoptosis in vivo. Taken together, the present study identified GPRC5A as a protective factor against the progression of human triple-negative breast cancer by increasing cell apoptosis via the regulation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Lu Yang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Shaorong Zhao
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Tong Zhu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Jin Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| |
Collapse
|
18
|
The emerging role of non-coding RNAs in the regulation of PI3K/AKT pathway in the carcinogenesis process. Biomed Pharmacother 2021; 137:111279. [PMID: 33493969 DOI: 10.1016/j.biopha.2021.111279] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
The PI3K/AKT pathway is an intracellular signaling pathway with an indispensable impact on cell cycle control. This pathway is functionally related with cell proliferation, cell survival, metabolism, and quiescence. The crucial role of this pathway in the development of cancer has offered this pathway as a target of novel anti-cancer treatments. Recent researches have demonstrated the role of microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) in controlling the PI3K/AKT pathway. Some miRNAs such as miR-155-5p, miR-328-3p, miR-125b-5p, miR-126, miR-331-3p and miR-16 inactivate this pathway, while miR-182, miR-106a, miR-193, miR-214, miR-106b, miR-93, miR-21 and miR-103/107 enhance activity of this pathway. Expression levels of PI3K/AKT-associated miRNAs could be used to envisage the survival of cancer patients. Numerous lncRNAs such as GAS5, FER1L4, LINC00628, PICART1, LOC101928316, ADAMTS9-AS2, SLC25A5-AS1, MEG3, AB073614 and SNHG6 interplay with this pathway. Identification of the impact of miRNAs and lncRNAs in the control of the activity of PI3K/AKT pathway would enhance the efficacy of targeted therapies against this pathway. Moreover, each of the mentioned miRNAs and lncRNAs could be used as a putative therapeutic candidate for the interfering with the carcinogenesis. In the current study, we review the role of miRNAs and lncRNAs in controlling the PI3K/AKT pathway and their contribution to carcinogenesis.
Collapse
|
19
|
Bakhshinyan D, Savage N, Salim SK, Venugopal C, Singh SK. The Strange Case of Jekyll and Hyde: Parallels Between Neural Stem Cells and Glioblastoma-Initiating Cells. Front Oncol 2021; 10:603738. [PMID: 33489908 PMCID: PMC7820896 DOI: 10.3389/fonc.2020.603738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
During embryonic development, radial glial precursor cells give rise to neural lineages, and a small proportion persist in the adult mammalian brain to contribute to long-term neuroplasticity. Neural stem cells (NSCs) reside in two neurogenic niches of the adult brain, the hippocampus and the subventricular zone (SVZ). NSCs in the SVZ are endowed with the defining stem cell properties of self-renewal and multipotent differentiation, which are maintained by intrinsic cellular programs, and extrinsic cellular and niche-specific interactions. In glioblastoma, the most aggressive primary malignant brain cancer, a subpopulation of cells termed glioblastoma stem cells (GSCs) exhibit similar stem-like properties. While there is an extensive overlap between NSCs and GSCs in function, distinct genetic profiles, transcriptional programs, and external environmental cues influence their divergent behavior. This review highlights the similarities and differences between GSCs and SVZ NSCs in terms of their gene expression, regulatory molecular pathways, niche organization, metabolic programs, and current therapies designed to exploit these differences.
Collapse
Affiliation(s)
- David Bakhshinyan
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Neil Savage
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Sabra Khalid Salim
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Chitra Venugopal
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Sheila K. Singh
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
20
|
Li W, Wu M, Zhang Y, Wei X, Zang J, Liu Y, Wang Y, Gong CX, Wei W. Intermittent fasting promotes adult hippocampal neuronal differentiation by activating GSK-3β in 3xTg-AD mice. J Neurochem 2020; 155:697-713. [PMID: 32578216 DOI: 10.1111/jnc.15105] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 12/26/2022]
Abstract
Moderate dietary restriction can ameliorate age-related chronic diseases such as Alzheimer's disease (AD) by increasing the expression of neurotrophic factors and promoting neurogenesis in the brain. Glycogen synthase kinase-3β (GSK-3β) signaling is essential for the coordination of progenitor cell proliferation and differentiation during brain development. The mechanisms by which GSK-3β is involved in dietary restriction-induced neurogenesis and cognitive improvement remain unclear. Six-month-old male 3xTg-AD and wild-type mice were fed on alternate days (intermittent fasting, IF) or ad libitum (AL) for 3 months. GSK-3β activity was regulated by bilaterally infusing lentiviral vectors carrying siRNA targeting GSK-3β into the dentate gyrus region of the hippocampus. Intermittent fasting promoted neuronal differentiation and maturation in the dentate gyrus and ameliorated recognized dysfunction in 3xTg-AD mice. These effects were reversed by siRNA targeting GSK-3β. After intermittent fasting, the insulin and protein kinase A signaling pathways were inhibited, while the adenosine monophosphate-activated protein kinase and brain-derived neurotrophic factor pathways were activated. These findings suggest that intermittent fasting can promote neuronal differentiation and maturation in the hippocampus by activating GSK-3β, thus improving learning and memory.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China.,Department of Pathology, The first people's hospital of foshan, Foshan, Guangdong, P. R. China
| | - Meijian Wu
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Yilin Zhang
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Xuemin Wei
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Jiankun Zang
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Yinghua Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, P. R. China
| | - Yanping Wang
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Wei Wei
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| |
Collapse
|
21
|
Recent Trends of microRNA Significance in Pediatric Population Glioblastoma and Current Knowledge of Micro RNA Function in Glioblastoma Multiforme. Int J Mol Sci 2020; 21:ijms21093046. [PMID: 32349263 PMCID: PMC7246719 DOI: 10.3390/ijms21093046] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Central nervous system tumors are a significant problem for modern medicine because of their location. The explanation of the importance of microRNA (miRNA) in the development of cancerous changes plays an important role in this respect. The first papers describing the presence of miRNA were published in the 1990s. The role of miRNA has been pointed out in many medical conditions such as kidney disease, diabetes, neurodegenerative disorder, arthritis and cancer. There are several miRNAs responsible for invasiveness, apoptosis, resistance to treatment, angiogenesis, proliferation and immunology, and many others. The research conducted in recent years analyzing this group of tumors has shown the important role of miRNA in the course of gliomagenesis. These particles seem to participate in many stages of the development of cancer processes, such as proliferation, angiogenesis, regulation of apoptosis or cell resistance to cytostatics.
Collapse
|
22
|
Zhang L, Li Y, Wang Q, Chen Z, Li X, Wu Z, Hu C, Liao D, Zhang W, Chen ZS. The PI3K subunits, P110α and P110β are potential targets for overcoming P-gp and BCRP-mediated MDR in cancer. Mol Cancer 2020; 19:10. [PMID: 31952518 PMCID: PMC6966863 DOI: 10.1186/s12943-019-1112-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/29/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND PI3K/AKT is a vital signaling pathway in humans. Recently, several PI3K/AKT inhibitors were reported to have the ability to reverse cancer multidrug resistance (MDR); however, specific targets in the PI3K/AKT pathways and the mechanisms associated with MDR have not been found because many of the inhibitors have multiple targets within a large candidate protein pool. AKT activation is one presumed mechanism by which MDR develops during cancer treatment. METHODS The effects of inhibiting PI3K 110α and 110β by BAY-1082439 treatment and CRISPR/Cas9 knockout were examined to determine the possible functions of BAY-1082439 and the roles of PI3K 110α and 110β in the reversal of MDR that is mediated by the downregulation of P-gp and BCRP. Inhibition of AKT with GSK-2110183 showed that the downregulation of P-gp and BCRP is independent of generalized AKT inactivation. Immunofluorescence, immunoprecipitation, MTT, flow cytometry and JC-1 staining analyses were conducted to study the reversal of MDR that is mediated by P-gp and BCRP in cancer cells. An ATPase assay and a structural analysis were also used to analyze the potential mechanisms by which BAY-1082439 specifically targets PI3K 110α and 110β and nonspecifically influences P-gp and BCRP. RESULTS By inhibiting the activation of the PI3K 110α and 110β catalytic subunits through both the administration of BAY-1082439 and the CRISPR/Cas9 deletion of Pik3ca and Pik3cb, the ATP-binding cassette transporters P-gp/ABCB1 and BCRP/ABCG2 were downregulated, thereby reestablishing the drug sensitivity of human epidermoid carcinoma and non-small cell lung cancer (NSCLC) MDR cells. Inhibition of AKT did not reverse the MDR mediated by P-gp or BCRP. The ABC family proteins and AKT may play MDR-enhancing roles independently. CONCLUSIONS The reversal of the dual functions of ABC-transporter-mediated and AKT-activation-enhanced MDR through the inhibition or knockout of PI3K 110α or 110β promises to improve current strategies based on combined drug treatments to overcome MDR challenges.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Proliferation
- Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Gene Expression Regulation, Neoplastic
- Humans
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Protein Kinase Inhibitors/pharmacology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Lei Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, China
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yidong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Qianchao Wang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, China
| | - Zhuo Chen
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, China.
| | - Xiaoyun Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Zhuoxun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Chaohua Hu
- College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Dan Liao
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
- Key Laboratory of Complementary and Alternative Medicine Experimental Animal Models of Guangxi, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Wei Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
- Institute of Plastic Surgery, Weifang Medical University, Weifang, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| |
Collapse
|
23
|
Fan Q, Wang Q, Cai R, Yuan H, Xu M. The ubiquitin system: orchestrating cellular signals in non-small-cell lung cancer. Cell Mol Biol Lett 2020; 25:1. [PMID: 31988639 PMCID: PMC6966813 DOI: 10.1186/s11658-019-0193-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023] Open
Abstract
The ubiquitin system, known as a common feature in eukaryotes, participates in multiple cellular processes, such as signal transduction, cell-cycle progression, receptor trafficking and endocytosis, and even the immune response. In lung cancer, evidence has revealed that aberrant events in ubiquitin-mediated processes can cause a variety of pathological outcomes including tumorigenesis and metastasis. Likewise, ubiquitination on the core components contributing to the activity of cell signaling controls bio-signal turnover and cell final destination. Given this, inhibitors targeting the ubiquitin system have been developed for lung cancer therapies and have shown great prospects for clinical application. However, the exact biological effects and physiological role of the drugs used in lung cancer therapies are still not clearly elucidated, which might seriously impede the progress of treatment. In this work, we summarize current research advances in cell signal regulation processes mediated through the ubiquitin system during the development of lung cancer, with the hope of improving the therapeutic effects by means of aiming at efficient targets.
Collapse
Affiliation(s)
- Qiang Fan
- 1Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China.,2Department of General Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
| | - Qian Wang
- 1Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
| | - Renjie Cai
- 1Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China.,2Department of General Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
| | - Haihua Yuan
- 1Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
| | - Ming Xu
- 1Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
| |
Collapse
|
24
|
Shi H, Li X, Yang J, Zhao Y, Xue C, Wang Y, He Q, Shen M, Zhang Q, Yang Y, Ding F. Bone marrow-derived neural crest precursors improve nerve defect repair partially through secreted trophic factors. Stem Cell Res Ther 2019; 10:397. [PMID: 31852510 PMCID: PMC6921427 DOI: 10.1186/s13287-019-1517-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/03/2019] [Accepted: 11/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Emerging evidence suggests that neural crest-derived cells (NCCs) present important functions in peripheral nerve regeneration to correct the insufficiency of autogenous Schwann cells. Postmigratory NCCs have been successfully isolated from adult rat bone marrow in our previous work. In this study, we aim to provide neural crest-derived Schwann cell precursors (SCPs) for repair of nerve defects in adult rats, and partially reveal the mechanisms involved in neuroregeneration of cell therapy. METHODS A clonal cell line of neural crest precursors of rat bone marrow origin (rBM-NCPs) with SCP identity was expanded in adherent monolayer culture to ensure the stable cell viability of NCPs and potentiate the repair of nerve defects after rBM-NCPs implantation based on tissue engineering nerve grafts (TENG). Here the behavioral, morphological, and electrophysiological detection was performed to evaluate the therapy efficacy. We further investigated the treatment with NCP-conditioned medium (NCP-CM) to sensory neurons after exposure to oxygen-glucose-deprivation (OGD) and partially compared the expression of trophic factor genes in rBM-NCPs with that in mesenchymal stem cells of bone marrow origin (rBM-MSCs). RESULTS It was showed that the constructed TENG with rBM-NCPs loaded into silk fibroin fiber scaffolds/chitosan conduits repaired 10-mm long sciatic nerve defects more efficiently than conduits alone. The axonal regrowth, remyelination promoted the reinnervation of the denervated hind limb muscle and skin and thereby alleviated muscle atrophy and facilitated the rehabilitation of motor and sensory function. Moreover, it was demonstrated that treatment with NCP-CM could restore the cultured primary sensory neurons after OGD through trophic factors including epidermal growth factor (EGF), platelet-derived growth factor alpha (PDGFα), ciliary neurotrophic factor (CNTF), and vascular endothelial growth factor alpha (VEGFα). CONCLUSIONS In summary, our findings indicated that monolayer-cultured rBM-NCPs cell-based therapy might effectively repair peripheral nerve defects partially through secreted trophic factors, which represented the secretome of rBM-NCPs differing from that of rBM-MSCs.
Collapse
Affiliation(s)
- Haiyan Shi
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, 19 Qixiu Road, Nantong, 226001, China
| | - Xiaoli Li
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China.,Department of Pathology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Junling Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Yahong Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, 19 Qixiu Road, Nantong, 226001, China
| | - Chengbin Xue
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, 19 Qixiu Road, Nantong, 226001, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Yaxian Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, 19 Qixiu Road, Nantong, 226001, China
| | - Qianru He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, 19 Qixiu Road, Nantong, 226001, China
| | - Mi Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, 19 Qixiu Road, Nantong, 226001, China
| | - Qi Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, 19 Qixiu Road, Nantong, 226001, China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, 19 Qixiu Road, Nantong, 226001, China. .,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China.
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, 19 Qixiu Road, Nantong, 226001, China. .,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China.
| |
Collapse
|
25
|
Yan Y, Huang H. Interplay Among PI3K/AKT, PTEN/FOXO and AR Signaling in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:319-331. [DOI: 10.1007/978-3-030-32656-2_14] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
26
|
Mohseni R, Ashrafi MR, Ai J, Nikougoftar M, Mohammadi M, Ghahvechi-Akbari M, Shoae-Hassani A, Hamidieh AA. Overexpression of SMN2 Gene in Motoneuron-Like Cells Differentiated from Adipose-Derived Mesenchymal Stem Cells by Ponasterone A. J Mol Neurosci 2018; 67:247-257. [PMID: 30535775 DOI: 10.1007/s12031-018-1232-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 11/25/2018] [Indexed: 01/25/2023]
Abstract
Cell therapy and stem cell transplantation strategies have provided potential therapeutic approaches for the treatment of neurological disorders. Adipose-derived mesenchymal stem cells (ADMSCs) are abundant adult stem cells with low immunogenicity, which can be used for allogeneic cell replacement therapies. Differentiation of ADMSCs into acetylcholine-secreting motoneurons (MNs) is a promising treatment for MN diseases, such as spinal muscular atrophy (SMA), which is associated with the level of SMN1 gene expression. The SMN2 gene plays an important role in MN disorders, as it can somewhat compensate for the lack of SMN1 expression in SMA patients. Although the differentiation potential of ADMSCs into MNs has been previously established, overexpression of SMN2 gene in a shorter period with a longer survival has yet to be elucidated. Ponasterone A (PNA), an ecdysteroid hormone activating the PI3K/Akt pathway, was studied as a new steroid to promote SMN2 overexpression in MNs differentiated from ADMSCs. After induction with retinoic acid, sonic hedgehog, forskolin, and PNA, MN phenotypes were differentiated from ADMSCs, and immunochemical staining, specific for β-tubulin, neuron-specific enolase, and choline acetyltransferase, was performed. Also, the results of real-time PCR assay indicated nestin, Pax6, Nkx2.2, Hb9, Olig2, and SMN2 expression in the differentiated cells. After 2 weeks of treatment, cultures supplemented with PNA showed a longer survival and a 1.2-fold increase in the expression of SMN2 (an overall 5.6-fold increase; *P ≤ 0.05), as confirmed by the Western blot analysis. The PNA treatment increased the levels of ChAT, Isl1, Hb9, and Nkx2 expression in MN-like cells. Our findings highlight the role of PNA in the upregulation of SMN2 genes from MSC-derived MN-like cells, which may serve as a potential candidate in cellular therapy for SMA patients.
Collapse
Affiliation(s)
- Rashin Mohseni
- Applied Cell Sciences and Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Reza Ashrafi
- Pediatric Neurology Division, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Applied Cell Sciences and Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahin Nikougoftar
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Mahmoud Mohammadi
- Pediatric Neurology Division, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Ghahvechi-Akbari
- Pediatric Neurology Division, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Shoae-Hassani
- Applied Cell Sciences and Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Applied Cell Sciences and Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Pediatric Hematology, Oncology and Stem Cell Transplantation Department, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Xie Y, Shi X, Sheng K, Han G, Li W, Zhao Q, Jiang B, Feng J, Li J, Gu Y. PI3K/Akt signaling transduction pathway, erythropoiesis and glycolysis in hypoxia (Review). Mol Med Rep 2018; 19:783-791. [PMID: 30535469 PMCID: PMC6323245 DOI: 10.3892/mmr.2018.9713] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/17/2018] [Indexed: 12/13/2022] Open
Abstract
The purpose of this review is to summarize the research progress of PI3K/Akt signaling pathway in erythropoiesis and glycolysis. Phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) is activated by numerous genes and leads to protein kinase B (Akt) binding to the cell membrane, with the help of phosphoinositide-dependent kinase, in the PI3K/Akt signal transduction pathway. Threonine and serine phosphorylation contribute to Akt translocation from the cytoplasm to the nucleus and further mediates enzymatic biological effects, including those involved in cell proliferation, apoptosis inhibition, cell migration, vesicle transport and cell cancerous transformation. As a key downstream protein of the PI3K/Akt signaling pathway, hypoxia-inducible factor (HIF)-1 is closely associated with the concentration of oxygen in the environment. Maintaining stable levels of HIF-1 protein is critical under normoxic conditions; however, HIF-1 levels quickly increase under hypoxic conditions. HIF-1α is involved in the acute hypoxic response associated with erythropoietin, whereas HIF-2α is associated with the response to chronic hypoxia. Furthermore, PI3K/Akt can reduce the synthesis of glycogen and increase glycolysis. Inhibition of glycogen synthase kinase 3β activity by phosphorylation of its N-terminal serine increases accumulation of cyclin D1, which promotes the cell cycle and improves cell proliferation through the PI3K/Akt signaling pathway. The PI3K/Akt signaling pathway is closely associated with a variety of enzymatic biological effects and glucose metabolism.
Collapse
Affiliation(s)
- Youbang Xie
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Xuefeng Shi
- Department of Respiratory Medicine, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Kuo Sheng
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Guoxiong Han
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Wenqian Li
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Qiangqiang Zhao
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Baili Jiang
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Jianming Feng
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Jianping Li
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Yuhai Gu
- Department of Respiratory Medicine, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| |
Collapse
|
28
|
Wang L, Schlagal CR, Gao J, Hao Y, Dunn TJ, McGrath EL, Labastida JA, Yu Y, Feng SQ, Liu SY, Wu P. Oligodendrocyte differentiation from human neural stem cells: A novel role for c-Src. Neurochem Int 2018; 120:21-32. [PMID: 30041015 DOI: 10.1016/j.neuint.2018.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/28/2018] [Accepted: 07/18/2018] [Indexed: 01/06/2023]
Abstract
Human neural stem cells (hNSCs) can differentiate into an oligodendrocyte lineage to facilitate remyelination in patients. Molecular mechanisms underlying oligodendrocyte fate specification remains unknown, hindering the development of efficient methods to generate oligodendrocytes from hNSCs. We have found that Neurobasal-A medium (NB) is capable of inducing hNSCs to oligodendrocyte progenitor cells (OPCs). We identified several signaling molecules are altered after cultivation in NB medium, including Akt, ERK1/2 and c-Src. While sustained activation of Akt and ERK1/2 during both NB induction and subsequent differentiation was required for OPC differentiation, c-Src phosphorylation was increased temporally during the period of NB induction. Both pharmacological inhibition and RNA interference confirmed that a transient elevation of phospho-c-Src is critical for OPC induction. Furthermore, inactivation of c-Src inhibited phosphorylation of Akt and ERK1/2. In summary, we identified a novel and critical role of c-Src in guiding hNSC differentiation to an oligodendrocyte lineage.
Collapse
Affiliation(s)
- Le Wang
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA; Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Er Rd, Yuexiu Qu, Guangzhou Shi, Guangdong Sheng, China
| | - Caitlin R Schlagal
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Junling Gao
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Yan Hao
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA; Department of Orthopedics, Tianjin Medical University General Hospital, 154 Anshan Rd, Heping Qu, 300051, China
| | - Tiffany J Dunn
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Erica L McGrath
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Javier Allende Labastida
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Yongjia Yu
- Department of Radiation Oncology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Shi-Qing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, 154 Anshan Rd, Heping Qu, 300051, China
| | - Shao-Yu Liu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Er Rd, Yuexiu Qu, Guangzhou Shi, Guangdong Sheng, China
| | - Ping Wu
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA.
| |
Collapse
|
29
|
Tang Y, Pan J, Huang S, Peng X, Zou X, Luo Y, Ren D, Zhang X, Li R, He P, Wa Q. Downregulation of miR-133a-3p promotes prostate cancer bone metastasis via activating PI3K/AKT signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:160. [PMID: 30021600 PMCID: PMC6052526 DOI: 10.1186/s13046-018-0813-4] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/23/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND Bone metastasis is a leading cause of morbidity and mortality in advanced prostate cancer (PCa). Downexpression of miR-133a-3p has been found to contribute to the progression, recurrence and distant metastasis in PCa. However, clinical significance of miR-133a-3p in bone metastasis of PCa, and the biological role of miR-133a-3p and its molecular mechanisms underlying bone metastasis of PCa remain unclear. METHODS miR-133a-3p expression was evaluated in 245 clinical PCa tissues by real-time PCR. Statistical analysis was performed to evaluate the clinical correlation between miR-133a-3p expression and clinicopathological features, and overall and bone metastasis-free survival in PCa patients. The biological roles of miR-133a-3p in the bone metastasis of PCa were investigated both in vitro and in vivo. Bioinformatics analysis, real-time PCR, western blot and luciferase reporter analysis were applied to demonstrate the relationship between miR-133a-3p and its potential targets. Western blotting and luciferase assays were examined to identify the underlying pathway involved in the anti-tumor role of miR-133a-3p. Clinical correlation of miR-133a-3p with its targets was verified in human PCa tissues. RESULTS miR-133a-3p expression is reduced in PCa tissues compared with the adjacent normal tissues and benign prostate lesion tissues, particularly in bone metastatic PCa tissues. Low expression of miR-133a-3p is significantly correlated with advanced clinicopathological characteristics and shorter bone metastasis-free survival in PCa patients by statistical analysis. Moreover, upregulating miR-133a-3p inhibits cancer stem cell-like phenotypes in vitro and in vivo, as well as attenuates anoikis resistance in vitro in PCa cells. Importantly, administration of agomir-133a-3p greatly suppresses the incidence of PCa bone metastasis in vivo. Our results further demonstrate that miR-133a-3p suppresses bone metastasis of PCa via inhibiting PI3K/AKT signaling by directly targeting multiple cytokine receptors, including EGFR, FGFR1, IGF1R and MET. The negative clinical correlation of miR-133a-3p with EGFR, FGFR1, IGF1R, MET and PI3K/AKT signaling activity is determined in clinical PCa tissues. CONCLUSION Our results unveil a novel mechanism by which miR-133a-3p inhibits bone metastasis of PCa, providing the evidence that miR-133a-3p may serve as a potential bone metastasis marker in PCa, and delivery of agomir-133a-3p may be an effective anti-bone metastasis therapeutic strategy in PCa.
Collapse
Affiliation(s)
- Yubo Tang
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Jincheng Pan
- Department of Urology Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuai Huang
- Department of Orthopaedic Surgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.,Department of Orthopaedic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan 2rd Road, Guangzhou, 510080, Guangdong Province, China
| | - Xinsheng Peng
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan 2rd Road, Guangzhou, 510080, Guangdong Province, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.,Department of Orthopaedic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan 2rd Road, Guangzhou, 510080, Guangdong Province, China
| | - Yongxiang Luo
- Department of biomedical engineering, health science center, Shenzhen University, Shenzhen, 518060, China
| | - Dong Ren
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan 2rd Road, Guangzhou, 510080, Guangdong Province, China
| | - Xin Zhang
- Department of Pathology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Ronggang Li
- Department of Pathology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Peiheng He
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan 2rd Road, Guangzhou, 510080, Guangdong Province, China.
| | - Qingde Wa
- Department of Orthopaedic Surgery, the Affiliated Hospital of Zunyi Medical college, 149 Dalian Road, Zunyi, 563003, Guizhou Province, China.
| |
Collapse
|
30
|
Cai B, Zheng Y, Ma S, Xing Q, Wang X, Yang B, Yin G, Guan F. Long non‑coding RNA regulates hair follicle stem cell proliferation and differentiation through PI3K/AKT signal pathway. Mol Med Rep 2018; 17:5477-5483. [PMID: 29393477 DOI: 10.3892/mmr.2018.8546] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/15/2017] [Indexed: 11/06/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are defined as non-coding transcripts (>200 nucleotides) that serve important roles in the proliferation and differentiation of stem cells. Hair follicle stem cells (HFTs) have multidirectional differentiation potential and are able to differentiate into skin, hair follicles and sebaceous glands, serving a role in skin wound healing. The aim of the present study was to analyze the regulatory role of lncRNA AK015322 (IncRNA5322) in HFTs and the potential mechanism of IncRNA5322‑mediated differentiation of HFTs. The results demonstrated that lncRNA5322 transfection promoted proliferation and differentiation in HFTs. It was identified that lncRNA5322 transfection upregulated the expression and phosphorylation of phosphoinositide 3‑kinase (PI3K) and protein kinase B (AKT) in HFTs. It was also observed that lncRNA5322 transfection upregulated microRNA (miR)‑21 and miR‑21 agonist (agomir‑21) eliminated lncRNA5322‑induced expression and phosphorylation of PI3K and AKT. The present study also demonstrated that agomir‑21 blocked IncRNA5322‑induced expression and phosphorylation of PI3K and AKT in HFTs. The results indicated that agomir‑21 transfection also suppressed the IncRNA5322‑induced proliferation and differentiation of HFTs. In conclusion, the results of the present study suggest that lncRNA5322 is able to promote the proliferation and differentiation of HFTs by targeting the miR‑21‑mediated PI3K‑AKT signaling pathway in HFTs.
Collapse
Affiliation(s)
- Bingjie Cai
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yunpeng Zheng
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Qu Xing
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xinxin Wang
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Bo Yang
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Guangwen Yin
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Fangxia Guan
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
31
|
Gray KM, Kaifer KA, Baillat D, Wen Y, Bonacci TR, Ebert AD, Raimer AC, Spring AM, Have ST, Glascock JJ, Gupta K, Van Duyne GD, Emanuele MJ, Lamond AI, Wagner EJ, Lorson CL, Matera AG. Self-oligomerization regulates stability of survival motor neuron protein isoforms by sequestering an SCF Slmb degron. Mol Biol Cell 2018; 29:96-110. [PMID: 29167380 PMCID: PMC5909936 DOI: 10.1091/mbc.e17-11-0627] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022] Open
Abstract
Spinal muscular atrophy (SMA) is caused by homozygous mutations in human SMN1 Expression of a duplicate gene (SMN2) primarily results in skipping of exon 7 and production of an unstable protein isoform, SMNΔ7. Although SMN2 exon skipping is the principal contributor to SMA severity, mechanisms governing stability of survival motor neuron (SMN) isoforms are poorly understood. We used a Drosophila model system and label-free proteomics to identify the SCFSlmb ubiquitin E3 ligase complex as a novel SMN binding partner. SCFSlmb interacts with a phosphor degron embedded within the human and fruitfly SMN YG-box oligomerization domains. Substitution of a conserved serine (S270A) interferes with SCFSlmb binding and stabilizes SMNΔ7. SMA-causing missense mutations that block multimerization of full-length SMN are also stabilized in the degron mutant background. Overexpression of SMNΔ7S270A, but not wild-type (WT) SMNΔ7, provides a protective effect in SMA model mice and human motor neuron cell culture systems. Our findings support a model wherein the degron is exposed when SMN is monomeric and sequestered when SMN forms higher-order multimers.
Collapse
Affiliation(s)
- Kelsey M Gray
- Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Integrative Program in Biological and Genome Sciences, Department of Biology and Department of Genetics, University of North Carolina, Chapel Hill, NC 27599
| | - Kevin A Kaifer
- Molecular Pathogenesis and Therapeutics Program, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211
| | - David Baillat
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77550
| | - Ying Wen
- Integrative Program in Biological and Genome Sciences, Department of Biology and Department of Genetics, University of North Carolina, Chapel Hill, NC 27599
| | - Thomas R Bonacci
- Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Amanda C Raimer
- Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Integrative Program in Biological and Genome Sciences, Department of Biology and Department of Genetics, University of North Carolina, Chapel Hill, NC 27599
| | - Ashlyn M Spring
- Integrative Program in Biological and Genome Sciences, Department of Biology and Department of Genetics, University of North Carolina, Chapel Hill, NC 27599
| | - Sara Ten Have
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD15EH, UK
| | - Jacqueline J Glascock
- Molecular Pathogenesis and Therapeutics Program, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211
| | - Kushol Gupta
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Gregory D Van Duyne
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Michael J Emanuele
- Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599
| | - Angus I Lamond
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD15EH, UK
| | - Eric J Wagner
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77550
| | - Christian L Lorson
- Molecular Pathogenesis and Therapeutics Program, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211
| | - A Gregory Matera
- Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Integrative Program in Biological and Genome Sciences, Department of Biology and Department of Genetics, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
32
|
Radial Extracorporeal Shock Wave Therapy Enhances the Proliferation and Differentiation of Neural Stem Cells by Notch, PI3K/AKT, and Wnt/β-catenin Signaling. Sci Rep 2017; 7:15321. [PMID: 29127399 PMCID: PMC5681501 DOI: 10.1038/s41598-017-15662-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 10/20/2017] [Indexed: 01/05/2023] Open
Abstract
Neural stem cell (NSC) proliferation and differentiation play a pivotal role in the repair of brain function in central nervous system (CNS) diseases. Radial extracorporeal shock wave therapy (rESWT) is a non-invasive and innovative treatment for many conditions, yet little is known about the effects of this treatment on NSCs. Mouse NSCs (NE-4C) were exposed to rESWT with 1.0, 1.5, 2.0, 2.5, 3.0, and 3.5 bar (500 impulses, and 2 Hz) in vitro. Cell viability test results indicated that rESWT, at a dose of 2.5 bar, 500 impulses, and 2 Hz, increased NE-4C viability within 72 h, and that the PI3K/AKT pathway was involved in its mechanisms. Exposure to rESWT also affected proliferation and differentiation of NE-4C after 8 weeks, which may be associated with Wnt/β-catenin and Notch pathways. This assessment is corroborated by the ability of inhibitors of Wnt/β-catenin [Dickkopf-1 (Dkk-1)] and the Notch pathway (DAPT) to weaken proliferation and differentiation of NSCs. In summary, a proper dose of rESWT enhanced NSCs augment via the PI3K/AKT pathway initially. Also, Wnt/β-catenin and the Notch pathway play important roles in regulation of the long-term efficacy of rESWT. This study reveals a novel approach to culture NSCs in vitro and support neurogenesis.
Collapse
|
33
|
Chung TW, Su JH, Lin CC, Li YR, Chao YH, Lin SH, Chan HL. 24-Methyl-Cholesta-5,24(28)-Diene-3β,19-diol-7β-Monoacetate Inhibits Human Small Cell Lung Cancer Growth In Vitro and In Vivo via Apoptosis Induction. Mar Drugs 2017; 15:md15070210. [PMID: 28671570 PMCID: PMC5532652 DOI: 10.3390/md15070210] [Citation(s) in RCA: 10] [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: 04/19/2017] [Revised: 06/13/2017] [Accepted: 06/26/2017] [Indexed: 01/06/2023] Open
Abstract
24-methyl-cholesta-5,24(28)-diene-3β,19-diol-7β-monoacetate (MeCDDA) is a natural steroid compound isolated from a wild-type soft coral (Nephthea erecta). The present study aimed to investigate the anti-small cell lung cancer (SCLC) effects of MeCDDA in vitro and in vivo, as well as to elucidate its underlying mechanism. Our results indicated that H1688 and H146 cells show relevant sensitivity to MeCDDA, and the exposure to MeCDDA in SCLC cells caused dose-dependent growth inhibitory responses. In addition, MeCDDA treatment promoted cell apoptosis and increased the activities of caspases in H1688 cells, reducing the mitochondrial membrane potential and stimulating the release of cytochrome c into the cytosol. Along with the increase in Bax expression and reduction in Bcl-2, the MeCDDA treatment also significantly decreased Akt and mTOR phosphorylation. Finally, MeCDDA treatment in the mouse xenograft model of H1688 cells exhibited significant inhibition of tumor growth, corroborating MeCDDA as a potential pre-clinical candidate for the treatment of SCLC. Overall, our results demonstrate that the cytotoxic effects of MeCDDA towards H1688 and H146 cells, possibly through the activation of the mitochondrial apoptotic pathway and inhibition of the PI3K/Akt/mTOR pathway, merit further studies for its possible clinical application in chemotherapy.
Collapse
Affiliation(s)
- Ting-Wen Chung
- Department of Medical Sciences, Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 300, Taiwan.
| | - Jui-Hsin Su
- Taiwan Coral Research Center, National Museum of Marine Biology & Aquarium, Pingtung 944, Taiwan.
| | - Chi-Chen Lin
- Department of Life Sciences, Institute of Biomedical Science, National Chung Hsing University, Taichung 402, Taiwan.
| | - Yi-Rong Li
- Department of Life Sciences, Institute of Biomedical Science, National Chung Hsing University, Taichung 402, Taiwan.
- Department of Internal Medicine, Changhua Christian Hospital, Changhua Division of Chest Medicine, Changhua 500, Taiwan.
| | - Ya-Hsuan Chao
- Department of Life Sciences, Institute of Biomedical Science, National Chung Hsing University, Taichung 402, Taiwan.
| | - Sheng-Hao Lin
- Department of Internal Medicine, Changhua Christian Hospital, Changhua Division of Chest Medicine, Changhua 500, Taiwan.
| | - Hong-Lin Chan
- Department of Medical Sciences, Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 300, Taiwan.
| |
Collapse
|
34
|
Jablonka S, Sendtner M. Developmental regulation of SMN expression: pathophysiological implications and perspectives for therapy development in spinal muscular atrophy. Gene Ther 2017; 24:506-513. [DOI: 10.1038/gt.2017.46] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/16/2017] [Accepted: 05/19/2017] [Indexed: 12/18/2022]
|
35
|
Wang LY, Tang ZJ, Han YZ. Neuroprotective effects of caffeic acid phenethyl ester against sevoflurane‑induced neuronal degeneration in the hippocampus of neonatal rats involve MAPK and PI3K/Akt signaling pathways. Mol Med Rep 2016; 14:3403-12. [PMID: 27498600 DOI: 10.3892/mmr.2016.5586] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 06/24/2016] [Indexed: 11/06/2022] Open
Abstract
Millions of infants and children are exposed to anesthesia every year during medical care. Sevoflurane is a volatile anesthetic that is frequently used for pediatric anesthesia. However, previous reports have suggested that the administration of sevoflurane promotes neurodegeneration, raising concerns regarding the safety of its usage. The present study aimed to investigate caffeic acid phenethyl ester (CAPE) and its protective effect against sevoflurane‑induced neurotoxicity in neonatal rats. Rat pups were administered with CAPE at 10, 20 or 40 mg/kg body weight from postnatal day 1 (P1) to P15. The P7 rats were exposed to sevoflurane (2.9%) for 6 h. Control group rats received no sevoflurane or CAPE. Neuronal apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick‑end labeling assay. The expression levels of caspases (caspase‑3, ‑8 and ‑9), apoptotic pathway proteins [Bcl‑2‑associated X protein (Bax), B cell CCL/lymphoma 2 (Bcl‑2), Bcl‑2‑like 1 (Bcl‑xL), Bcl‑2‑associated agonist of cell death (Bad) and phosphorylated (p)‑Bad], mitogen‑activated protein kinases (MAPK) signaling pathway proteins [c‑Jun N‑terminal kinase (JNK), p‑JNK, extracellular signal‑regulated kinase (ERK)1/2, p‑ERK1/2, p38, p‑p38 and p‑c‑Jun] and the phosphoinositide 3‑kinase (PI3K)/Akt cascade were evaluated by western blotting following sevoflurane and CAPE treatment. In addition, the expression of cleaved caspase‑3 was analyzed by immunohistochemistry. CAPE significantly reduced sevoflurane‑induced apoptosis, downregulated the expression levels of caspases and pro‑apoptotic proteins (Bax and Bad) and elevated the expression levels of Bcl‑2 and Bcl‑xL when compared with sevoflurane treatment. Furthermore, CAPE appeared to modify the expression levels of MAPKs and activate the PI3K/Akt signaling pathway. Thus, the present study demonstrated that CAPE effectively inhibited sevoflurane‑induced neuroapoptosis by modulating the expression and phosphorylation of apoptotic pathway proteins and MAPKs, and by regulating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Li-Yan Wang
- Department of Pediatric Surgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Zhi-Jun Tang
- Department of Orthopedics in Repair and Reconstruction, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Yu-Zeng Han
- Department of Pediatric Internal Medicine, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| |
Collapse
|
36
|
Zhang YH, Zhang J, Song JN, Xu X, Cai JS, Zhou Y, Gao JG. The PI3K-AKT-mTOR pathway activates recovery from general anesthesia. Oncotarget 2016; 7:40939-40952. [PMID: 27340771 PMCID: PMC5173033 DOI: 10.18632/oncotarget.10172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/16/2016] [Indexed: 12/29/2022] Open
Abstract
We investigated roles of PI3K-AKT-mTOR pathway in recovery from general anesthesia. Sprague-Dawley rats divided into five groups: saline+artificial cerebrospinal fluid (ACSF; Group A), ketamine+ACSF (Group B), ketamine+IGF-1 (Group C), ketamine+PI3K inhibitor (Group D), and PI3K/Akt agonists (Group E). Proportion of δ waves on ECoGs was recorded. Rats were tested for duration of loss of righting reflex (LORR), ataxic period and behavior in Morris water maze. mRNA and protein expression of members of PI3K-AKT-mTOR pathway were measured by RT-qPCR and Western blots. Histopathologic changes in hippocampal tissues observed by HE staining. We found that the proportion of δ waves decreased in Group C, while increased in Group D compared with Group B; the durations of LORR and ataxic period were shorter in Group C, but longer in Group D. In Morris water maze, escape latency (EL) and duration and frequency of staying on platform was shorter in Group C and longer in Group D than in Group B. Group A exhibited low expression of proteins in PI3K-AKT-mTOR pathway, while p-AKT, p-mTOR and p-P70S6K expression increased in cerebral cortex, brain stem, and thalamus in Group C. By contrast, expression of those proteins was lower in Group D than Group B. Those proteins expressions were higher in Group E than in Group A. HE staining showed that anesthesia may induce cell apoptosis in rat hippocampal CA1 areas, and PI3K/Akt agonists could inhibit apoptosis. Our results suggest that activation of PI3K-AKT-mTOR pathway may promote recovery from general anesthesia and enhance spatial learning and memory.
Collapse
Affiliation(s)
- Yun-Hui Zhang
- Department of Anesthesia, the Second Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Jin Zhang
- Department of Anesthesia, Shijiazhuang Obstetrics and Gynecology Hospital, Shijiazhuang, P. R. China
| | - Jian-Nan Song
- Department of Anesthesia, Chifeng Municipal Hospital, Chifeng, P. R. China
| | - Xue Xu
- Department of Anesthesia, the Second Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Jin-Song Cai
- Department of Anesthesia, the Second Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Yang Zhou
- Department of Anesthesia, Hebei Medical University, Shijiazhuang, P. R. China
| | - Jin-Gui Gao
- Department of Anesthesia, the Second Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| |
Collapse
|
37
|
Praveen P, Hülsmann H, Sültmann H, Kuner R, Fröhlich H. Cross-talk between AMPK and EGFR dependent Signaling in Non-Small Cell Lung Cancer. Sci Rep 2016; 6:27514. [PMID: 27279498 PMCID: PMC4899720 DOI: 10.1038/srep27514] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 05/16/2016] [Indexed: 01/25/2023] Open
Abstract
Lung cancers globally account for 12% of new cancer cases, 85% of these being Non Small Cell Lung Cancer (NSCLC). Therapies like erlotinib target the key player EGFR, which is mutated in about 10% of lung adenocarcinoma. However, drug insensitivity and resistance caused by second mutations in the EGFR or aberrant bypass signaling have evolved as a major challenge in controlling these tumors. Recently, AMPK activation was proposed to sensitize NSCLC cells against erlotinib treatment. However, the underlying mechanism is largely unknown. In this work we aim to unravel the interplay between 20 proteins that were previously associated with EGFR signaling and erlotinib drug sensitivity. The inferred network shows a high level of agreement with protein-protein interactions reported in STRING and HIPPIE databases. It is further experimentally validated with protein measurements. Moreover, predictions derived from our network model fairly agree with somatic mutations and gene expression data from primary lung adenocarcinoma. Altogether our results support the role of AMPK in EGFR signaling and drug sensitivity.
Collapse
Affiliation(s)
- Paurush Praveen
- University of Bonn, Bonn-Aachen International Center for IT, Dahlmannstr. 2, Bonn Germany
- The Microsoft Research-University of Trento Center for Computational and Systems Biology, 38068 Rovereto, Italy
| | - Helen Hülsmann
- Cancer Genome Research Group, German Cancer Consortium (DKTK), German Center for Lung Research (DZL), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460,D-69120 Heidelberg, Germany
| | - Holger Sültmann
- Cancer Genome Research Group, German Cancer Consortium (DKTK), German Center for Lung Research (DZL), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460,D-69120 Heidelberg, Germany
| | - Ruprecht Kuner
- Cancer Genome Research Group, German Cancer Consortium (DKTK), German Center for Lung Research (DZL), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460,D-69120 Heidelberg, Germany
| | - Holger Fröhlich
- University of Bonn, Bonn-Aachen International Center for IT, Dahlmannstr. 2, Bonn Germany
| |
Collapse
|
38
|
YU YANG, YU XIAOFENG, MA JIANXIA, TONG YILI, YAO JIANFENG. Effects of NVP-BEZ235 on the proliferation, migration, apoptosis and autophagy in HT-29 human colorectal adenocarcinoma cells. Int J Oncol 2016; 49:285-93. [DOI: 10.3892/ijo.2016.3507] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/21/2016] [Indexed: 11/05/2022] Open
|
39
|
Wu T, Wang C, Ding L, Shen Y, Cui H, Wang M, Wang H. Arginine Relieves the Inflammatory Response and Enhances the Casein Expression in Bovine Mammary Epithelial Cells Induced by Lipopolysaccharide. Mediators Inflamm 2016; 2016:9618795. [PMID: 27110069 PMCID: PMC4821974 DOI: 10.1155/2016/9618795] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 02/24/2016] [Indexed: 12/31/2022] Open
Abstract
As one of functional active amino acids, L-arginine holds a key position in immunity. However, the mechanism that arginine modulates cow mammary inflammatory response in ruminant is unclear. Therefore, this study was conducted to investigate the effects of L-arginine on inflammatory response and casein expression after challenging the bovine mammary epithelial cells (BMECs) with lipopolysaccharide (LPS). The cells were divided into four groups, stimulated with or without LPS (10 μg/mL) and treated with or without arginine (100 μg/mL) for 12 h. The concentration of proinflammatory cytokines, inducible nitric oxide synthase (iNOS), mammalian target of rapamycin (mTOR), and Toll-like receptor 4 (TLR4) signaling pathways as well as the casein was determined. The results showed that arginine reduced the LPS-induced production like IL-1β, IL-6, TNF-α, and iNOS. Though the expression of NF-κB was attenuated and the mTOR signaling pathway was upregulated, arginine had no effect on TLR4 expression. In addition, our results show that the content of β-casein and the total casein were enhanced after arginine was supplemented in LPS-induced BMECs. In conclusion, arginine could relieve the inflammatory reaction induced by LPS and enhance the concentration of β-casein and the total casein in bovine mammary epithelial cells.
Collapse
Affiliation(s)
- Tianyou Wu
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Chao Wang
- Cell Signaling Group, School of Pathology and Laboratory Medicine, The University of Western Australia, M Block QEII Medical Center, Monash Avenue, Nedlands, WA 6009, Australia
| | - Luoyang Ding
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yizhao Shen
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Huihui Cui
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Hongrong Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
40
|
Differentiation therapy of hepatocellular carcinoma by inhibiting the activity of AKT/GSK-3β/β-catenin axis and TGF-β induced EMT with sophocarpine. Cancer Lett 2016; 376:95-103. [PMID: 26945965 DOI: 10.1016/j.canlet.2016.01.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 01/03/2016] [Accepted: 01/06/2016] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma progression is thought to be driven by cancer stem cells (CSCs). No clinical trial has, as yet, shown convincing long-term disease free survival results for the majority of patients in HCC. So it is important to discover new anti-cancer agents. In our study, we chose sophocarpine, which is derived from the foxtail-like sophora herb, for its efficacy to inhibit HCC including CSCs and potential mechanism study. Our results show that sophocarpine could not only reduce HCC cell viability, eliminate HCC and reverse hepatoma cells malignant phenotype, but also reduce the ratio of CSCs and inhibit the sphere formation of CSCs in vitro. In vivo, sophocarpine significantly displayed antitumor effects in subcutaneous xenograft HCC models and orthotopic transplantation tumor models. Further studies showed that sophocarpine could exert anti-tumor effects partly via downregulating the activity of the cancer stem cell related pathways and inhibiting EMT induced by TGF-β.
Collapse
|
41
|
Willems C, Fu Q, Roose H, Mertens F, Cox B, Chen J, Vankelecom H. Regeneration in the Pituitary After Cell-Ablation Injury: Time-Related Aspects and Molecular Analysis. Endocrinology 2016; 157:705-21. [PMID: 26653762 DOI: 10.1210/en.2015-1741] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We recently showed that the mouse pituitary holds regenerative competence. Young-adult GHCre/iDTR mice, expressing diphtheria toxin (DT) receptor in GH-producing cells, regenerate the GH(+) cells, as ablated by 3-day DT treatment (3DT), up to 60% after 5 months. The pituitary's stem cells participate in this restoration process. Here, we characterized this regenerative capacity in relation to age and recovery period and started to search for underlying molecular mechanisms. Extending the recovery period (up to 19 mo) does not result in higher regeneration levels. In addition, the regenerative competence disappears at older age, coinciding with a reduction in pituitary stem cell number and fitness. Surprisingly, prolonging DT treatment of young-adult mice to 10 days (10DT) completely blocks the regeneration, although the stem cell compartment still reacts by promptly expanding, and retains in vitro stem cell functionality. To obtain a first broad view on molecular grounds underlying reparative capacity and/or failure, the stem cell-clustering side population was analyzed by whole-genome expression analysis. A number of stemness factors and components of embryonic, epithelial-mesenchymal transition, growth factor and Hippo pathways are higher expressed in the stem cell-clustering side population of the regenerating pituitary (after 3DT) when compared with the basal gland and to the nonregenerating pituitary (after 10DT). Together, the regenerative capacity of the pituitary is limited both in age-related terms and final efficacy, and appears to rely on stem cell-associated pathway activation. Dissection of the molecular profiles may eventually identify targets to induce or boost regeneration in situations of (injury-related) pituitary deficiency.
Collapse
Affiliation(s)
- Christophe Willems
- Department of Development and Regeneration (C.W., Q.F., H.R., F.M., B.C., J.C., H.V.), Cluster Stem Cell Biology and Embryology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven 3000, Belgium; Eye Center (Q.F.), Second Affiliated Hospital, School of Medicine, Zhejiang University, and Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou 310009, China; and Department of Hand Surgery (J.C.), Tongji Medical College, Union Hospital, Huazhong University of Science and Technology (HUST), Wuhan, Hubei 430022, China
| | - Qiuli Fu
- Department of Development and Regeneration (C.W., Q.F., H.R., F.M., B.C., J.C., H.V.), Cluster Stem Cell Biology and Embryology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven 3000, Belgium; Eye Center (Q.F.), Second Affiliated Hospital, School of Medicine, Zhejiang University, and Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou 310009, China; and Department of Hand Surgery (J.C.), Tongji Medical College, Union Hospital, Huazhong University of Science and Technology (HUST), Wuhan, Hubei 430022, China
| | - Heleen Roose
- Department of Development and Regeneration (C.W., Q.F., H.R., F.M., B.C., J.C., H.V.), Cluster Stem Cell Biology and Embryology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven 3000, Belgium; Eye Center (Q.F.), Second Affiliated Hospital, School of Medicine, Zhejiang University, and Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou 310009, China; and Department of Hand Surgery (J.C.), Tongji Medical College, Union Hospital, Huazhong University of Science and Technology (HUST), Wuhan, Hubei 430022, China
| | - Freya Mertens
- Department of Development and Regeneration (C.W., Q.F., H.R., F.M., B.C., J.C., H.V.), Cluster Stem Cell Biology and Embryology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven 3000, Belgium; Eye Center (Q.F.), Second Affiliated Hospital, School of Medicine, Zhejiang University, and Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou 310009, China; and Department of Hand Surgery (J.C.), Tongji Medical College, Union Hospital, Huazhong University of Science and Technology (HUST), Wuhan, Hubei 430022, China
| | - Benoit Cox
- Department of Development and Regeneration (C.W., Q.F., H.R., F.M., B.C., J.C., H.V.), Cluster Stem Cell Biology and Embryology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven 3000, Belgium; Eye Center (Q.F.), Second Affiliated Hospital, School of Medicine, Zhejiang University, and Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou 310009, China; and Department of Hand Surgery (J.C.), Tongji Medical College, Union Hospital, Huazhong University of Science and Technology (HUST), Wuhan, Hubei 430022, China
| | - Jianghai Chen
- Department of Development and Regeneration (C.W., Q.F., H.R., F.M., B.C., J.C., H.V.), Cluster Stem Cell Biology and Embryology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven 3000, Belgium; Eye Center (Q.F.), Second Affiliated Hospital, School of Medicine, Zhejiang University, and Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou 310009, China; and Department of Hand Surgery (J.C.), Tongji Medical College, Union Hospital, Huazhong University of Science and Technology (HUST), Wuhan, Hubei 430022, China
| | - Hugo Vankelecom
- Department of Development and Regeneration (C.W., Q.F., H.R., F.M., B.C., J.C., H.V.), Cluster Stem Cell Biology and Embryology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven 3000, Belgium; Eye Center (Q.F.), Second Affiliated Hospital, School of Medicine, Zhejiang University, and Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou 310009, China; and Department of Hand Surgery (J.C.), Tongji Medical College, Union Hospital, Huazhong University of Science and Technology (HUST), Wuhan, Hubei 430022, China
| |
Collapse
|
42
|
Nakano R, Edamura K, Nakayama T, Narita T, Okabayashi K, Sugiya H. Fibroblast Growth Factor Receptor-2 Contributes to the Basic Fibroblast Growth Factor-Induced Neuronal Differentiation in Canine Bone Marrow Stromal Cells via Phosphoinositide 3-Kinase/Akt Signaling Pathway. PLoS One 2015; 10:e0141581. [PMID: 26523832 PMCID: PMC4629880 DOI: 10.1371/journal.pone.0141581] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/09/2015] [Indexed: 11/26/2022] Open
Abstract
Bone marrow stromal cells (BMSCs) are considered as candidates for regenerative therapy and a useful model for studying neuronal differentiation. The role of basic fibroblast growth factor (bFGF) in neuronal differentiation has been previously studied; however, the signaling pathway involved in this process remains poorly understood. In this study, we investigated the signaling pathway in the bFGF-induced neuronal differentiation of canine BMSCs. bFGF induced the mRNA expression of the neuron marker, microtubule associated protein-2 (MAP2) and the neuron-like morphological change in canine BMSCs. In the presence of inhibitors of fibroblast growth factor receptors (FGFR), phosphatidylinositol 3-kinase (PI3K) and Akt, i.e., SU5402, LY294002, and MK2206, respectively, bFGF failed to induce the MAP2 mRNA expression and the neuron-like morphological change. bFGF induced Akt phosphorylation, but it was attenuated by the FGFR inhibitor SU5402 and the PI3K inhibitor LY294002. In canine BMSCs, expression of FGFR-1 and FGFR-2 was confirmed, but only FGFR-2 activation was detected by cross-linking and immunoprecipitation analysis. Small interfering RNA-mediated knockdown of FGFR-2 in canine BMSCs resulted in the attenuation of bFGF-induced Akt phosphorylation. These results suggest that the FGFR-2/PI3K/Akt signaling pathway is involved in the bFGF-induced neuronal differentiation of canine BMSCs.
Collapse
Affiliation(s)
- Rei Nakano
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
| | - Kazuya Edamura
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
| | - Tomohiro Nakayama
- Laboratory of Veterinary Radiology, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
| | - Takanori Narita
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
| | - Ken Okabayashi
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
| | - Hiroshi Sugiya
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
43
|
Li T, Li Z, Nan F, Dong J, Deng Y, Yu Q, Zhang T. Construction of a novel inducing system with multi-layered alginate microcapsules to regulate differentiation of neural precursor cells from bone mesenchymal stem cells. Med Hypotheses 2015; 85:910-3. [PMID: 26386487 DOI: 10.1016/j.mehy.2015.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/09/2015] [Indexed: 01/08/2023]
Abstract
Neural precursor cells (NPCs) are a promising cell source for the treatment of nervous system diseases; however, they are limited in their applications due to source-related ethical considerations or legislations. Therefore, a novel approach is necessary to obtain sufficient NPCs. Recently, the usage of bone marrow-derived mesenchymal stem cells (BMSCs) differentiated into neural cells has become a potential method to obtain NPCs. Moreover, growth factors (GFs) are emerging as inducers to evoke the differentiation of BMSCs into NPCs. For example, GFs may activate various signaling pathways related to neural differentiation, such as phosphatidylinositol 3 kinase/protein kinase B, cyclic adenosine monophosphate/protein kinase A, and Janus kinase/signal transducer activator of transcription. However, the utilization of growth factors still has some limitations such as high costs and low rates of neural differentiation. Neuroblastoma cells have been characterized as a potential pool for growth factors. Additionally, basic fibroblast growth factor (bFGF), a type of growth factor, has been demonstrated to be able to increase the differentiation and survival rate of NPCs. For better use of neuroblastoma cells and bFGF, we established a novel system involving multi-layered alginate-polylysine-alginate (APA) microcapsules to encapsulate neuroblastoma cells and bFGF, which may not only provide sufficient growth factors in a sustained manner but also avoid the carcinogenicity caused by neuroblastoma cells. Above all, we hypothesized that neuroblastoma cells and bFGF encapsulated in multilayered alginate microcapsules may efficiently induce the differentiation of BMSCs into NPCs.
Collapse
Affiliation(s)
- Tao Li
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| | - Zhengwei Li
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| | - Feng Nan
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China.
| | - Jianli Dong
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| | - Yushuang Deng
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| | - Qing Yu
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| | - Teng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| |
Collapse
|
44
|
Wang S, Jiang B, Zhang T, Liu L, Wang Y, Wang Y, Chen X, Lin H, Zhou L, Xia Y, Chen L, Yang C, Xiong Y, Ye D, Guan KL. Insulin and mTOR Pathway Regulate HDAC3-Mediated Deacetylation and Activation of PGK1. PLoS Biol 2015; 13:e1002243. [PMID: 26356530 PMCID: PMC4565669 DOI: 10.1371/journal.pbio.1002243] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 08/04/2015] [Indexed: 01/07/2023] Open
Abstract
Phosphoglycerate kinase 1 (PGK1) catalyzes the reversible transfer of a phosphoryl group from 1, 3-bisphosphoglycerate (1, 3-BPG) to ADP, producing 3-phosphoglycerate (3-PG) and ATP. PGK1 plays a key role in coordinating glycolytic energy production with one-carbon metabolism, serine biosynthesis, and cellular redox regulation. Here, we report that PGK1 is acetylated at lysine 220 (K220), which inhibits PGK1 activity by disrupting the binding with its substrate, ADP. We have identified KAT9 and HDAC3 as the potential acetyltransferase and deacetylase, respectively, for PGK1. Insulin promotes K220 deacetylation to stimulate PGK1 activity. We show that the PI3K/AKT/mTOR pathway regulates HDAC3 S424 phosphorylation, which promotes HDAC3-PGK1 interaction and PGK1 K220 deacetylation. Our study uncovers a previously unknown mechanism for the insulin and mTOR pathway in regulation of glycolytic ATP production and cellular redox potential via HDAC3-mediated PGK1 deacetylation.
Collapse
Affiliation(s)
- Shiwen Wang
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Bowen Jiang
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Tengfei Zhang
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Lixia Liu
- Key Laboratory of Synthetic Biology, Bioinformatics Center and Laboratory of Systems Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Wang
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Yiping Wang
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Xiufei Chen
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Huaipeng Lin
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Lisha Zhou
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Yukun Xia
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Leilei Chen
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Chen Yang
- Key Laboratory of Synthetic Biology, Bioinformatics Center and Laboratory of Systems Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yue Xiong
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
- Lineberger Comprehensive Cancer Center, Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Dan Ye
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
| | - Kun-Liang Guan
- Key Laboratory of Molecular Medicine of Ministry of Education and Institutes of Biomedical Sciences, Shanghai Medical College, College of Life Science, Fudan University, Shanghai, China
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
45
|
El-Hamoly T, El-Denshary ES, Saad SM, El-Ghazaly MA. 3-aminobenzamide, a poly (ADP ribose) polymerase inhibitor, enhances wound healing in whole body gamma irradiated model. Wound Repair Regen 2015; 23:672-84. [PMID: 26080614 DOI: 10.1111/wrr.12330] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 06/15/2015] [Indexed: 01/09/2023]
Abstract
The custom use of radiotherapy was found to participate in the development of chronic unhealed wounds. In general, exposure to gamma radiation stimulates the production of reactive oxygen species (ROS) that eventually leads to damaging effect. Conversely, overexpression of a nuclear poly (ADP-ribose) polymerase enzyme (PARP) after oxidative insult extremely brings about cellular injury due to excessive consumption of NAD and ATP. Here, we dedicated our study to investigate the role of 3-aminobenzamide (3-AB), a PARP inhibitor, on pregamma irradiated wounds. Two full-thickness (6 mm diameter) wounds were created on the dorsum of Swiss albino mouse. The progression of wound contraction was monitored by capturing daily photo images. Exposure to gamma radiation (6Gy) exacerbated the normal healing of excisional wounds. Remarkably, topical application of 3-AB cream (50 µM) revealed a marked acceleration in the rate of wound contraction. Likewise, PARP inhibition ameliorated the unbalanced oxidative/nitrosative status of granulated skin tissues. Such effect was significantly revealed by the correction of the reduced antioxidant capacity and the enhanced lipid peroxidation, hydrogen peroxide, and myeloperoxidase contents. Moreover, application of 3-AB modified the cutaneous nitrite content throughout healing process. Conversely, the expressions of pro-inflammatory cytokines were down-regulated by PARP inhibition. The mitochondrial ATP content showed a lower consumption rate on 3-AB-treated wound bed as well. In parallel, the mRNA expressions of Sirt-1 and acyl-COA oxidase-2 (ACOX-2) were up-regulated; whom functions control the mitochondrial ATP synthesis and lipid metabolism. The current data suggested that inhibition of PARP-1 enzyme may accelerate the delayed wound healing in whole body gamma irradiated mice by early modifying the oxidative stress as well as the inflammatory response.
Collapse
Affiliation(s)
- Tarek El-Hamoly
- Cyclotron Project, Nuclear Physics Department, Nuclear Research Center, Egyptian Atomic Energy Authority, Cairo, Egypt.,Drug Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ezzeddin S El-Denshary
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Shokry Mohamed Saad
- Nuclear Physics Department, Nuclear Research Center, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mona A El-Ghazaly
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
46
|
Trans-Activation between EphA and FGFR Regulates Self-Renewal and Differentiation of Mouse Embryonic Neural Stem/Progenitor Cells via Differential Activation of FRS2α. PLoS One 2015; 10:e0128826. [PMID: 26024354 PMCID: PMC4449203 DOI: 10.1371/journal.pone.0128826] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/30/2015] [Indexed: 12/05/2022] Open
Abstract
Ephs and FGFRs belong to a superfamily of receptor tyrosine kinases, playing important roles in stem cell biology. We previously reported that EphA4 and FGFR form a heterodimer following stimulation with ligands, trans-activating each other and signaling through a docking protein, FRS2α, that binds to both receptors. Here, we investigated whether the interaction between EphA4 and FGFRs can be generalized to other Ephs and FGFRs, and, in addition, examined the downstream signal mediating their function in embryonic neural stem/progenitor cells. We revealed that various Ephs and FGFRs interact with each other through similar molecular domains. When neural stem/progenitor cells were stimulated with FGF2 and ephrin-A1, the signal transduced from the EphA4/FGFR/FRS2α complex enhanced self-renewal, while stimulation with ephrin-A1 alone induced neuronal differentiation. The downstream signal required for neuronal differentiation appears to be MAP kinase mainly linked to the Ras family of G proteins. MAP kinase activation was delayed and sustained, distinct from the transient activation induced by FGF2. Interestingly, this effect on neuronal differentiation required the presence of FGFRs. Specific FGFR inhibitor almost completely abolished the function of ephrin-A1 stimulation. These findings suggest that the ternary complex of EphA, FGFR and FRS2α formed by ligand stimulation regulates self-renewal and differentiation of mouse embryonic neural stem/progenitor cells by ligand-specific fine tuning of the downstream signal via FRS2α.
Collapse
|
47
|
Guo Q, Ning F, Fang R, Wang HS, Zhang G, Quan MY, Cai SH, Du J. Endogenous Nodal promotes melanoma undergoing epithelial-mesenchymal transition via Snail and Slug in vitro and in vivo. Am J Cancer Res 2015; 5:2098-2112. [PMID: 26269769 PMCID: PMC4529629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/10/2015] [Indexed: 06/04/2023] Open
Abstract
Nodal, an important embryonic morphogen, has been reported to modulate tumorigenesis. Epithelial-mesenchymal transition (EMT) plays an important role in cancer metastasis. We have previously reported that recombinant Nodal treatment can promote melanoma undergoing EMT, but the effects of endogenous Nodal on EMT are still unknown. Here we generated both Nodal-overexpression and -knockdown stable cell lines to investigate the in vitro and in vivo characteristics of Nodal-induced EMT in murine melanoma cells. Nodal-overexpression cells displayed increased migration ability, accompanied by typical phenotype changes of EMT. In contrast, Nodal-knockdown stable cells repressed the EMT phenotype as well as reduced cell motility. Results of animal experiments confirmed that overexpression of Nodal can promote the metastasis of melanoma tumor in vivo. Mechanistically, we found that Nodal-induced expression of Snail and Slug involves its activation of ALK/Smads and PI3k/AKT pathways, which is an important process in the Nodal-induced EMT. However, we also found that the EMT phenotype was not completely inhibited by blocking the paracrine activity of Nodal in Nodal overexpression cell line suggesting the presence of additional mechanism(s) in the Nodal-induced EMT. This study provides a better understanding of Nodal function in melanoma, and suggests targeting Nodal as a potential strategy for melanoma therapey.
Collapse
Affiliation(s)
- Qiang Guo
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
| | - Fen Ning
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
| | - Rui Fang
- Department of Pharmacy, Guangdong Women and Children HospitalGuangzhou 511400, Guangdong, China
| | - Hong-Sheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
| | - Ge Zhang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
| | - Mei-Yu Quan
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
| | - Shao-Hui Cai
- Department of Pharmacology, College of Pharmacy, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen UniversityGuangzhou 510006, Guangdong, China
| |
Collapse
|
48
|
Di Liddo R, Bertalot T, Schuster A, Schrenk S, Tasso A, Zanusso I, Conconi MT, Schäfer KH. Anti-inflammatory activity of Wnt signaling in enteric nervous system: in vitro preliminary evidences in rat primary cultures. J Neuroinflammation 2015; 12:23. [PMID: 25644719 PMCID: PMC4332439 DOI: 10.1186/s12974-015-0248-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/14/2015] [Indexed: 01/22/2023] Open
Abstract
Background In the last years, Wnt signaling was demonstrated to regulate inflammatory processes. In particular, an increased expression of Wnts and Frizzled receptors was reported in inflammatory bowel disease (IBD) and ulcerative colitis to exert both anti- and pro-inflammatory functions regulating the intestinal activated nuclear factor κB (NF-кB), TNFa release, and IL10 expression. Methods To investigate the role of Wnt pathway in the response of the enteric nervous system (ENS) to inflammation, neurons and glial cells from rat myenteric plexus were treated with exogenous Wnt3a and/or LPS with or without supporting neurotrophic factors such as basic fibroblast growth factor (bFGF), epithelial growth factor (EGF), and glial cell-derived neurotrophic factor (GDNF). The immunophenotypical characterization by flow cytometry and the protein and gene expression analysis by qPCR and Western blotting were carried out. Results Flow cytometry and immunofluorescence staining evidenced that enteric neurons coexpressed Frizzled 9 and toll-like receptor 4 (TLR4) while glial cells were immunoreactive to TLR4 and Wnt3a suggesting that canonical Wnt signaling is active in ENS. Under in vitro LPS treatment, Western blot analysis demonstrated an active cross talk between canonical Wnt signaling and NF-кB pathway that is essential to negatively control enteric neuronal response to inflammatory stimuli. Upon costimulation with LPS and Wnt3a, a significant anti-inflammatory activity was detected by RT-PCR based on an increased IL10 expression and a downregulation of pro-inflammatory cytokines TNFa, IL1B, and interleukin 6 (IL6). When the availability of neurotrophic factors in ENS cultures was abolished, a changed cell reactivity by Wnt signaling was observed at basal conditions and after LPS treatment. Conclusions The results of this study suggested the existence of neuronal surveillance through FZD9 and Wnt3a in enteric myenteric plexus. Moreover, experimental evidences were provided to clarify the correlation among soluble trophic factors, Wnt signaling, and anti-inflammatory protection of ENS.
Collapse
Affiliation(s)
- Rosa Di Liddo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy.
| | - Thomas Bertalot
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy.
| | - Anne Schuster
- Department of Biotechnology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany.
| | - Sandra Schrenk
- Department of Biotechnology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany.
| | - Alessia Tasso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy.
| | - Ilenia Zanusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy.
| | - Maria Teresa Conconi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy.
| | - Karl Herbert Schäfer
- Department of Biotechnology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany.
| |
Collapse
|
49
|
Iyer S, Viernes DR, Chisholm JD, Margulies BS, Kerr WG. SHIP1 regulates MSC numbers and their osteolineage commitment by limiting induction of the PI3K/Akt/β-catenin/Id2 axis. Stem Cells Dev 2014; 23:2336-51. [PMID: 24857423 DOI: 10.1089/scd.2014.0122] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Here, we show that Src homology 2-domain-containing inositol 5'-phosphatase 1 (SHIP1) is required for the efficient development of osteoblasts from mesenchymal stem cells (MSCs) such that bone growth and density are reduced in mice that lack SHIP1 expression in MSCs. We find that SHIP1 promotes the osteogenic output of MSCs by limiting activation of the PI3K/Akt/β-catenin pathway required for induction of the MSC stemness factor Id2. In parallel, we demonstrate that mice with myeloid-restricted ablation of SHIP1, including osteoclasts (OCs), show no reduction in bone mass or density. Hence, diminished bone mass and density in the SHIP1-deficient mice results from SHIP deficiency in MSC and osteolineage progenitors. Intriguingly, mice with a SHIP-deficient MSC compartment also exhibit decreased OC numbers. In agreement with our genetic findings we also show that treatment of mice with an SHIP1 inhibitor (SHIPi) significantly reduces bone mass. These findings demonstrate a novel role for SHIP1 in MSC fate determination and bone growth. Further, SHIPi may represent a novel therapeutic approach to limit bone development in osteopetrotic and sclerotic bone diseases.
Collapse
Affiliation(s)
- Sonia Iyer
- 1 Department of Microbiology and Immunology, SUNY Upstate Medical University , Syracuse, New York
| | | | | | | | | |
Collapse
|
50
|
Natarajan R, Singal V, Benes R, Gao J, Chan H, Chen H, Yu Y, Zhou J, Wu P. STAT3 modulation to enhance motor neuron differentiation in human neural stem cells. PLoS One 2014; 9:e100405. [PMID: 24945434 PMCID: PMC4063761 DOI: 10.1371/journal.pone.0100405] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/27/2014] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury or amyotrophic lateral sclerosis damages spinal motor neurons and forms a glial scar, which prevents neural regeneration. Signal transducer and activator of transcription 3 (STAT3) plays a critical role in astrogliogenesis and scar formation, and thus a fine modulation of STAT3 signaling may help to control the excessive gliogenic environment and enhance neural repair. The objective of this study was to determine the effect of STAT3 inhibition on human neural stem cells (hNSCs). In vitro hNSCs primed with fibroblast growth factor 2 (FGF2) exhibited a lower level of phosphorylated STAT3 than cells primed by epidermal growth factor (EGF), which correlated with a higher number of motor neurons differentiated from FGF2-primed hNSCs. Treatment with STAT3 inhibitors, Stattic and Niclosamide, enhanced motor neuron differentiation only in FGF2-primed hNSCs, as shown by increased homeobox gene Hb9 mRNA levels as well as HB9+ and microtubule-associated protein 2 (MAP2)+ co-labeled cells. The increased motor neuron differentiation was accompanied by a decrease in the number of glial fibrillary acidic protein (GFAP)-positive astrocytes. Interestingly, Stattic and Niclosamide did not affect the level of STAT3 phosphorylation; rather, they perturbed the nuclear translocation of phosphorylated STAT3. In summary, we demonstrate that FGF2 is required for motor neuron differentiation from hNSCs and that inhibition of STAT3 further increases motor neuron differentiation at the expense of astrogliogenesis. Our study thus suggests a potential benefit of targeting the STAT3 pathway for neurotrauma or neurodegenerative diseases.
Collapse
Affiliation(s)
- Rajalaxmi Natarajan
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Vinamrata Singal
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Richard Benes
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Junling Gao
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Hoi Chan
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Haijun Chen
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yongjia Yu
- Department of Radiation Oncology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jia Zhou
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ping Wu
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|