51
|
Kamble VS, Pachpor TA, Khandagale SB, Wagh VV, Khare SP. Translation initiation and dysregulation of initiation factors in rare diseases. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
52
|
Xu B, Li Z, Li S, Ke H, Zhang Q, Qin Y, Guo T. Pathogenic variants in TSC2 might cause premature ovarian insufficiency through activated mTOR induced hyperactivation of primordial follicles. Fertil Steril 2022; 118:1139-1149. [PMID: 36229297 DOI: 10.1016/j.fertnstert.2022.08.853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To investigate the role of tuberous sclerosis complex (TSC) genes, including TSC1 and TSC2, in the pathogenesis of human premature ovarian insufficiency (POI). DESIGN Genetic and functional study. SETTING University-based reproductive medical center. PATIENT(S) Six patients from a cohort of 1,030 cases with idiopathic POI. INTERVENTION(S) Variants in TSC1 and TSC2 were screened through the largest in-house database of whole exome sequencing performed in 1,030 patients with idiopathic POI. The pathogenic effects of the variants were further verified by functional studies. MAIN OUTCOME MEASURE(S) TSC1 or TSC2 variant and functional characteristics. RESULT(S) Five pathogenic heterozygous variants in TSC2 were identified in 6 patients with POI. Functional studies showed these variants impaired the repressive effect of TSC2 on mammalian target of rapamycin (mTOR) pathway by disrupting the formation of TSC complex or its GTPase-activating protein activity. Furthermore, in vitro ovarian culture assay showed that TSC2 p.R98Q led to hyperactivation of mTOR pathway thereby triggering primordial follicle activation. CONCLUSION(S) The present study identified pathogenic variants of TSC2 in patients with POI, firstly suggested defective TSC/mTOR pathway mediated hyperactivation of primordial follicle participating in the pathogenesis of POI, giving insights into new targets of genetic counseling and clinical prevention for POI. Considering the pivotal role of TSC2 variants in diagnosis of TSC syndrome, the present study also highlighted the importance of history collection and long-term follow-up for the TSC2 variants carriers.
Collapse
Affiliation(s)
- Bingying Xu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People's Republic of China; Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People's Republic of China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China
| | - Zhuqing Li
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People's Republic of China; Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People's Republic of China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China
| | - Shan Li
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People's Republic of China; Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People's Republic of China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China
| | - Hanni Ke
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People's Republic of China; Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People's Republic of China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China
| | - Qian Zhang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People's Republic of China; Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People's Republic of China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yingying Qin
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People's Republic of China; Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People's Republic of China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China
| | - Ting Guo
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People's Republic of China; Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People's Republic of China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
53
|
Gonfloni S, Jodice C, Gustavino B, Valentini E. DNA Damage Stress Response and Follicle Activation: Signaling Routes of Mammalian Ovarian Reserve. Int J Mol Sci 2022; 23:14379. [PMID: 36430860 PMCID: PMC9693393 DOI: 10.3390/ijms232214379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Chemotherapy regimens and radiotherapy are common strategies to fight cancer. In women, these therapies may cause side effects such as premature ovarian insufficiency (POI) and infertility. Clinical strategies to protect the ovarian reserve from the lethal effect of cancer therapies needs better understanding of the mechanisms underlying iatrogenic loss of follicle reserve. Recent reports demonstrate a critical role for p53 and CHK2 in the oocyte response to different DNA stressors, which are commonly used to treat cancer. Here we review the molecular mechanisms underlying the DNA damage stress response (DDR) and discuss crosstalk between DDR and signaling pathways implicated in primordial follicle activation.
Collapse
Affiliation(s)
- Stefania Gonfloni
- Department of Biology, University of Rome Tor Vergata, Via Della Ricerca Scientifica, 00133 Rome, Italy
| | - Carla Jodice
- Department of Biology, University of Rome Tor Vergata, Via Della Ricerca Scientifica, 00133 Rome, Italy
| | - Bianca Gustavino
- Department of Biology, University of Rome Tor Vergata, Via Della Ricerca Scientifica, 00133 Rome, Italy
| | - Elvia Valentini
- Department of Biology, University of Rome Tor Vergata, Via Della Ricerca Scientifica, 00133 Rome, Italy
- PhD Program in Cellular and Molecular Biology, 00133 Rome, Italy
| |
Collapse
|
54
|
Selig JI, Krug HV, Küppers C, Ouwens DM, Kraft FA, Adler E, Bauer SJ, Lichtenberg A, Akhyari P, Barth M. Interactive contribution of hyperinsulinemia, hyperglycemia, and mammalian target of rapamycin signaling to valvular interstitial cell differentiation and matrix remodeling. Front Cardiovasc Med 2022; 9:942430. [PMID: 36386326 PMCID: PMC9661395 DOI: 10.3389/fcvm.2022.942430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetes and its major key determinants insulin resistance and hyperglycemia are known risk factors for calcific aortic valve disease (CAVD). The processes leading to molecular and structural alterations of the aortic valve are yet not fully understood. In previous studies, we could show that valvular interstitial cells (VIC) display canonical elements of classical insulin signaling and develop insulin resistance upon hyperinsulinemia and hyperglycemia accompanied by impaired glucose metabolism. Analyses of cultured VIC and aortic valve tissue revealed extracellular matrix remodeling and degenerative processes. Since PI3K signaling through mammalian target of rapamycin (mTOR) is involved in fibrotic processes of the heart, we aim at further functional investigation of this particular Akt-downstream signaling pathway in the context of diabetes-induced CAVD. Primary cultures of VIC were treated with hyperinsulinemia and hyperglycemia. Phosphorylation of mTOR(Ser2448) was determined by Western blot analysis after acute insulin stimulus. Inhibition of mTOR phosphorylation was performed by rapamycin. Phosphorylation of mTOR complex 1 (MTORC1) downstream substrates 4E-BP1(Thr37/46) and P70S6K(Thr389), and MTORC2 downstream substrate Akt(Ser473) as well as the PDK1-dependent phosphorylation of Akt(Thr308) was investigated. Markers for extracellular matrix remodeling, cell differentiation and degenerative changes were analyzed by Western blot analysis, semi-quantitative real-time PCR and colorimetric assays. Hyperinsulinemia and hyperglycemia lead to alterations of VIC activation, differentiation and matrix remodeling as well as to an abrogation of mTOR phosphorylation. Inhibition of mTOR signaling by rapamycin leads to a general downregulation of matrix molecules, but to an upregulation of α-smooth muscle actin expression and alkaline phosphatase activity. Comparison of expression patterns upon diabetic conditions and rapamycin treatment reveal a possible regulation of particular matrix components and key degeneration markers by MTORC1 downstream signaling. The present findings broaden the understanding of mitogenic signaling pathways in VIC triggered by hyperinsulinemia and hyperglycemia, supporting the quest for developing strategies of prevention and tailored treatment of CAVD in diabetic patients.
Collapse
Affiliation(s)
- Jessica I. Selig
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - H. Viviana Krug
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Caroline Küppers
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - D. Margriet Ouwens
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - Felix A. Kraft
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Elena Adler
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sebastian J. Bauer
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Artur Lichtenberg
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Payam Akhyari
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- *Correspondence: Payam Akhyari,
| | - Mareike Barth
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
55
|
Qiao S, Liu C, Sun L, Wang T, Dai H, Wang K, Bao L, Li H, Wang W, Liu SJ, Liu H. Gut Parabacteroides merdae protects against cardiovascular damage by enhancing branched-chain amino acid catabolism. Nat Metab 2022; 4:1271-1286. [PMID: 36253620 DOI: 10.1038/s42255-022-00649-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 08/30/2022] [Indexed: 01/20/2023]
Abstract
Obesity, dyslipidemia and gut dysbiosis are all linked to cardiovascular diseases. A Ganoderma meroterpene derivative (GMD) has been shown to alleviate obesity and hyperlipidemia through modulating the gut microbiota in obese mice. Here we show that GMD protects against obesity-associated atherosclerosis by increasing the abundance of Parabacteroides merdae in the gut and enhancing branched-chain amino acid (BCAA) catabolism. Administration of live P. merdae to high-fat-diet-fed ApoE-null male mice reduces atherosclerotic lesions and enhances intestinal BCAA degradation. The degradation of BCAAs is mediated by the porA gene expressed in P. merdae. Deletion of porA from P. merdae blunts its capacity to degrade BCAAs and leads to inefficacy in fighting against atherosclerosis. We further show that P. merdae inhibits the mTORC1 pathway in atherosclerotic plaques. In support of our preclinical findings, an in silico analysis of human gut metagenomic studies indicates that P. merdae and porA genes are depleted in the gut microbiomes of individuals with atherosclerosis. Our results provide mechanistic insights into the therapeutic potential of GMD through P. merdae in treating obesity-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Shanshan Qiao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, PR China
| | - Chang Liu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China
| | - Li Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, PR China
| | - Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, PR China
| | - Huanqin Dai
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, PR China
| | - Kai Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, PR China
| | - Li Bao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, PR China
| | - Hantian Li
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, PR China
| | - Wenzhao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China
| | - Shuang-Jiang Liu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, PR China.
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China.
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, PR China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, PR China.
| |
Collapse
|
56
|
Park SG, Jo IJ, Park SA, Park MC, Mun YJ. Poria cocos Extract from Mushrooms Stimulates Aquaporin-3 via the PI3K/Akt/mTOR Signaling Pathway. Clin Cosmet Investig Dermatol 2022; 15:1919-1931. [PMID: 36133400 PMCID: PMC9484496 DOI: 10.2147/ccid.s378545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/05/2022] [Indexed: 11/23/2022]
Abstract
Background Poria cocos (PC), a fungus, has been used for more than 2000 years as a food and medicine in China. PC and its components have various pharmacological effects on the skin, including immunomodulatory activities, barrier function improvement, and anti-tumor effects. However, the effect of PC in aquaporin-3 (AQP3) expression, which is essential for epidermal water permeability barrier maintenance, was not reported. Methods This study examined the mechanism through which the ethanol extract of the sclerotium of PC (EPC) promoted the expression of AQP3 in cultured human keratinocytes. Western blotting was used to investigate the expression of AQPs and the activation of phosphoinositide 3-kinase (PI3K)/Akt-related signaling molecules in HaCaT cells. Cells were treated with inhibitors of PI3K/Akt and mechanistic target of rapamycin (mTOR) prior to EPC treatment. Results EPC promoted the expression of AQP3 in HaCaT cells without affecting AQP1 and AQP2 expression. Phosphorylated Akt levels were increased by EPC treatment, and the inhibition of PI3K by LY2940002 resulted in a reduction in EPC-induced AQP3 expression. Furthermore, EPC stimulated the phosphorylation of p70S6K and AktSer473, which are downstream targets of mTORC1 and mTORC2, respectively. The mTOR complex inhibitors, rapamycin and Torin 1, partially reduced EPC-induced AQP3 expression. Conclusion These results suggest that EPC increased expression of AQP3, which is important for skin moisturization, by activating the PI3K/Akt/mTOR signaling pathway in human keratinocytes.
Collapse
Affiliation(s)
- Sung-Gu Park
- Department of Oriental Medical Ophthalmology & Otolaryngology & Dermatology, College of Korean Medicine, Wonkwang University, Iksan, South Korea
| | - Il-Joo Jo
- Central Stroke Center of Korean medicine, College of Korean Medicine, Wonkwang University, Iksan, South Korea
| | - Seol-A Park
- Department of Beauty Design Graduate School, Wonkwang University, Iksan, South Korea
| | - Min-Cheol Park
- Department of Oriental Medical Ophthalmology & Otolaryngology & Dermatology, College of Korean Medicine, Wonkwang University, Iksan, South Korea
| | - Yeun-Ja Mun
- Department of Anatomy, College of Korean Medicine, Wonkwang University, Iksan, South Korea
| |
Collapse
|
57
|
Oncolytic Avian Reovirus p17-Modulated Inhibition of mTORC1 by Enhancement of Endogenous mTORC1 Inhibitors Binding to mTORC1 To Disrupt Its Assembly and Accumulation on Lysosomes. J Virol 2022; 96:e0083622. [PMID: 35946936 PMCID: PMC9472607 DOI: 10.1128/jvi.00836-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mechanism by which avian reovirus (ARV)-modulated suppression of mTORC1 triggers autophagy remains largely unknown. In this work, we determined that p17 functions as a negative regulator of mTORC1. This study suggest novel mechanisms whereby p17-modulated inhibition of mTORC1 occurs via upregulation of p53, inactivation of Akt, and enhancement of binding of the endogenous mTORC1 inhibitors (PRAS40, FKBP38, and FKPP12) to mTORC1 to disrupt its assembly and accumulation on lysosomes. p17-modulated inhibition of Akt leads to activation of the downstream targets PRAS40 and TSC2, which results in mTORC1 inhibition, thereby triggering autophagy and translation shutoff, which is favorable for virus replication. p17 impairs the interaction of mTORC1 with its activator Rheb, which promotes FKBP38 interaction with mTORC1. It is worth noting that p17 activates ULK1 and Beclin1 and increases the formation of the Beclin 1/class III PI3K complex. These effects could be reversed in the presence of insulin or depletion of p53. Furthermore, we found that p17 induces autophagy in cancer cell lines by upregulating the p53/PTEN pathway, which inactivates Akt and mTORC1. This study highlights p17-modulated inhibition of Akt and mTORC1, which triggers autophagy and translation shutoff by positively modulating the tumor suppressors p53 and TSC2 and endogenous mTORC1 inhibitors. IMPORTANCE The mechanisms by which p17-modulated inhibition of mTORC1 induces autophagy and translation shutoff is elucidated. In this work, we determined that p17 serves as a negative regulator of mTORC1. This study provides several lines of conclusive evidence demonstrating that p17-modulated inhibition of mTORC1 occurs via upregulation of the p53/PTEN pathway, downregulation of the Akt/Rheb/mTORC1 pathway, enhancement of binding of the endogenous mTORC1 inhibitors to mTORC1 to disrupt its assembly, and suppression of mTORC1 accumulation on lysosomes. This work provides valuable information for better insights into p17-modulated inhibition of mTORC1, which induces autophagy and translation shutoff to benefit virus replication.
Collapse
|
58
|
Codenotti S, Zizioli D, Mignani L, Rezzola S, Tabellini G, Parolini S, Giacomini A, Asperti M, Poli M, Mandracchia D, Vezzoli M, Bernardi S, Russo D, Mitola S, Monti E, Triggiani L, Tomasini D, Gastaldello S, Cassandri M, Rota R, Marampon F, Fanzani A. Hyperactive Akt1 Signaling Increases Tumor Progression and DNA Repair in Embryonal Rhabdomyosarcoma RD Line and Confers Susceptibility to Glycolysis and Mevalonate Pathway Inhibitors. Cells 2022; 11:cells11182859. [PMID: 36139434 PMCID: PMC9497225 DOI: 10.3390/cells11182859] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022] Open
Abstract
In pediatric rhabdomyosarcoma (RMS), elevated Akt signaling is associated with increased malignancy. Here, we report that expression of a constitutively active, myristoylated form of Akt1 (myrAkt1) in human RMS RD cells led to hyperactivation of the mammalian target of rapamycin (mTOR)/70-kDa ribosomal protein S6 kinase (p70S6K) pathway, resulting in the loss of both MyoD and myogenic capacity, and an increase of Ki67 expression due to high cell mitosis. MyrAkt1 signaling increased migratory and invasive cell traits, as detected by wound healing, zymography, and xenograft zebrafish assays, and promoted repair of DNA damage after radiotherapy and doxorubicin treatments, as revealed by nuclear detection of phosphorylated H2A histone family member X (γH2AX) through activation of DNA-dependent protein kinase (DNA-PK). Treatment with synthetic inhibitors of phosphatidylinositol-3-kinase (PI3K) and Akt was sufficient to completely revert the aggressive cell phenotype, while the mTOR inhibitor rapamycin failed to block cell dissemination. Furthermore, we found that pronounced Akt1 signaling increased the susceptibility to cell apoptosis after treatments with 2-deoxy-D-glucose (2-DG) and lovastatin, enzymatic inhibitors of hexokinase, and 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR), especially in combination with radiotherapy and doxorubicin. In conclusion, these data suggest that restriction of glucose metabolism and the mevalonate pathway, in combination with standard therapy, may increase therapy success in RMS tumors characterized by a dysregulated Akt signaling.
Collapse
Affiliation(s)
- Silvia Codenotti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Daniela Zizioli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Luca Mignani
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Giovanna Tabellini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Silvia Parolini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Michela Asperti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Maura Poli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Delia Mandracchia
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Marika Vezzoli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Simona Bernardi
- Department of Clinical and Experimental Sciences, ASST Spedali Civili di Brescia, University of Brescia, 25123 Brescia, Italy
| | - Domenico Russo
- Department of Clinical and Experimental Sciences, ASST Spedali Civili di Brescia, University of Brescia, 25123 Brescia, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Eugenio Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Luca Triggiani
- Radiation Oncology Department, ASST Spedali Civili di Brescia, University of Brescia, 25123 Brescia, Italy
| | - Davide Tomasini
- Radiation Oncology Department, ASST Spedali Civili di Brescia, University of Brescia, 25123 Brescia, Italy
| | - Stefano Gastaldello
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
- Precision Medicine Research Center, School of Pharmacy, Binzhou Medical University, Laishan District, Guanhai Road 346, Yantai 264003, China
| | - Matteo Cassandri
- Department of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
- Department of Radiotherapy, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Rossella Rota
- Department of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Francesco Marampon
- Department of Radiotherapy, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Alessandro Fanzani
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Correspondence: ; Tel.: +39-030-3717567
| |
Collapse
|
59
|
Renal Transcriptome and Metabolome in Mice with Principal Cell-Specific Ablation of the Tsc1 Gene: Derangements in Pathways Associated with Cell Metabolism, Growth and Acid Secretion. Int J Mol Sci 2022; 23:ijms231810601. [PMID: 36142537 PMCID: PMC9502912 DOI: 10.3390/ijms231810601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is caused by mutations in the hamartin (TSC1) or tuberin (TSC2) genes. Using a mouse model of TSC renal cystogenesis that we have previously described, the current studies delineate the metabolic changes in the kidney and their relation to alterations in renal gene expression. To accomplish this, we compared the metabolome and transcriptome of kidneys from 28-day-old wildtype (Wt) and principal cell-specific Tsc1 KO (Tsc1 KO) mice using targeted 1H nuclear magnetic resonance targeted metabolomic and RNA-seq analyses. The significant changes in the kidney metabolome of Tsc1 KO mice included reductions in the level of several amino acids and significant decreases in creatine, NADH, inosine, UDP-galactose, GTP and myo-inositol levels. These derangements may affect energy production and storage, signal transduction and synthetic pathways. The pertinent derangement in the transcriptome of Tsc1 KO mice was associated with increased collecting duct acid secretion, active cell division and the up-regulation of signaling pathways (e.g., MAPK and AKT/PI3K) that suppress the TSC2 GTPase-activating function. The combined renal metabolome and transcriptome alterations observed in these studies correlate with the unregulated growth and predominance of genotypically normal A-intercalated cells in the epithelium of renal cysts in Tsc1 KO mice.
Collapse
|
60
|
Lin WH, Jacobs-Wagner C. Connecting single-cell ATP dynamics to overflow metabolism, cell growth, and the cell cycle in Escherichia coli. Curr Biol 2022; 32:3911-3924.e4. [PMID: 35961315 DOI: 10.1016/j.cub.2022.07.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/21/2022] [Accepted: 07/14/2022] [Indexed: 10/15/2022]
Abstract
Adenosine triphosphate (ATP) is an abundant and essential metabolite that cells consume and regenerate in large amounts to support growth. Although numerous studies have inferred the intracellular concentration of ATP in bacterial cultures, what happens in individual bacterial cells under stable growth conditions is less clear. Here, we use the QUEEN-2m biosensor to quantify ATP dynamics in single Escherichia coli cells in relation to their growth rate, metabolism, cell cycle, and cell lineage. We find that ATP dynamics are more complex than expected from population studies and are associated with growth-rate variability. Under stable nutrient-rich condition, cells can display large fluctuations in ATP level that are partially coordinated with the cell cycle. Abrogation of aerobic acetate fermentation (overflow metabolism) through genetic deletion considerably reduces both the amplitude of ATP level fluctuations and the cell-cycle trend. Similarly, growth in media in which acetate fermentation is lower or absent results in the reduction of ATP level fluctuation and cell-cycle trend. This suggests that overflow metabolism exhibits temporal dynamics, which contributes to fluctuating ATP levels during growth. Remarkably, at the single-cell level, growth rate negatively correlates with the amplitude of ATP fluctuation for each tested condition, linking ATP dynamics to growth-rate heterogeneity in clonal populations. Our work highlights the importance of single-cell analysis in studying metabolism and its implication to phenotypic diversity and cell growth.
Collapse
Affiliation(s)
- Wei-Hsiang Lin
- Department of Biology, Stanford University, Palo Alto, CA 94305, USA; Chemistry, Engineering, Medicine for Human Health Institute, Stanford University, Palo Alto, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305, USA
| | - Christine Jacobs-Wagner
- Department of Biology, Stanford University, Palo Alto, CA 94305, USA; Chemistry, Engineering, Medicine for Human Health Institute, Stanford University, Palo Alto, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305, USA.
| |
Collapse
|
61
|
Li B, Wang W, Huang Y, Han L, Li J, Zheng N, Wu Z, Zhang X, Li X, Deng L, Lin M, Chen X, Zhang M. Lithium treatment promotes the activation of primordial follicles through PI3K/Akt signaling. Biol Reprod 2022; 107:1059-1071. [PMID: 35871551 DOI: 10.1093/biolre/ioac150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 04/20/2022] [Accepted: 07/17/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
In mammals, dormant primordial follicles represent the ovarian reserve throughout reproductive life. In vitro activation of dormant primordial follicles has been used to treat patients with premature ovarian insufficiency (POI). However, there remains a lack of effective strategies to stimulate follicle activation in vivo. In this study, we used an in vitro ovarian culture system and intraperitoneal injection to study the effect of lithium treatment on primordial follicle activation. Lithium increased the number of growing follicles in cultured mouse ovaries and promoted pre-granulosa cell proliferation. Furthermore, lithium significantly increased the levels of phosphorylated protein kinase B (Akt) and the number of oocytes with forkhead Box O3a (FOXO3a) nuclear export. Inhibition of the phosphatidylinositol 3 kinase (PI3K)/Akt pathway by LY294002 reversed lithium-promoted mouse primordial follicle activation. These results suggest that lithium promotes mouse primordial follicle activation by the PI3K/Akt signaling. Lithium also promoted primordial follicle activation and increased the levels of p-Akt in mouse ovaries in vivo and in human ovarian tissue cultured in vitro. Taken together, lithium promotes primordial follicle activation in mice and humans by the PI3K/Akt signaling. Lithium might be a potential oral drug for treating infertility in POI patients with residual dormant primordial follicles.
Collapse
Affiliation(s)
- Biao Li
- State Key Laboratory for Agrobiotechnology , College of Biological Sciences, China Agricultural University, Beijing 100193 , China
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Weiyong Wang
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Yingying Huang
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Lincheng Han
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Jia Li
- State Key Laboratory for Agrobiotechnology , College of Biological Sciences, China Agricultural University, Beijing 100193 , China
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Nana Zheng
- State Key Laboratory for Agrobiotechnology , College of Biological Sciences, China Agricultural University, Beijing 100193 , China
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Zhanying Wu
- State Key Laboratory for Agrobiotechnology , College of Biological Sciences, China Agricultural University, Beijing 100193 , China
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Xiaodan Zhang
- State Key Laboratory for Agrobiotechnology , College of Biological Sciences, China Agricultural University, Beijing 100193 , China
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Xuelan Li
- The Center for Reproductive Medicine , Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528300, Guangdong , China
| | - Ling Deng
- The Center for Reproductive Medicine , Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528300, Guangdong , China
| | - Min Lin
- The Center for Reproductive Medicine , Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528300, Guangdong , China
| | - Xin Chen
- The Center for Reproductive Medicine , Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528300, Guangdong , China
| | - Meijia Zhang
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| |
Collapse
|
62
|
Guerbette T, Boudry G, Lan A. Mitochondrial function in intestinal epithelium homeostasis and modulation in diet-induced obesity. Mol Metab 2022; 63:101546. [PMID: 35817394 PMCID: PMC9305624 DOI: 10.1016/j.molmet.2022.101546] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022] Open
Abstract
Background Systemic low-grade inflammation observed in diet-induced obesity has been associated with dysbiosis and disturbance of intestinal homeostasis. This latter relies on an efficient epithelial barrier and coordinated intestinal epithelial cell (IEC) renewal that are supported by their mitochondrial function. However, IEC mitochondrial function might be impaired by high fat diet (HFD) consumption, notably through gut-derived metabolite production and fatty acids, that may act as metabolic perturbators of IEC. Scope of review This review presents the current general knowledge on mitochondria, before focusing on IEC mitochondrial function and its role in the control of intestinal homeostasis, and featuring the known effects of nutrients and metabolites, originating from the diet or gut bacterial metabolism, on IEC mitochondrial function. It then summarizes the impact of HFD on mitochondrial function in IEC of both small intestine and colon and discusses the possible link between mitochondrial dysfunction and altered intestinal homeostasis in diet-induced obesity. Major conclusions HFD consumption provokes a metabolic shift toward fatty acid β-oxidation in the small intestine epithelial cells and impairs colonocyte mitochondrial function, possibly through downstream consequences of excessive fatty acid β-oxidation and/or the presence of deleterious metabolites produced by the gut microbiota. Decreased levels of ATP and concomitant O2 leaks into the intestinal lumen could explain the alterations of intestinal epithelium dynamics, barrier disruption and dysbiosis that contribute to the loss of epithelial homeostasis in diet-induced obesity. However, the effect of HFD on IEC mitochondrial function in the small intestine remains unknown and the precise mechanisms by which HFD induces mitochondrial dysfunction in the colon have not been elucidated so far.
Collapse
Affiliation(s)
| | - Gaëlle Boudry
- Institut Numecan, INSERM, INRAE, Univ Rennes, Rennes, France.
| | - Annaïg Lan
- Institut Numecan, INSERM, INRAE, Univ Rennes, Rennes, France; Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Paris, France
| |
Collapse
|
63
|
Zhao Y, Zhang Y, Liu D, Feng H, Wang X, Su J, Yao Y, Ng EHY, Yeung WSB, Li RHW, Rodriguez-Wallberg KA, Liu K. Identification of curcumin as a novel potential drug for promoting the development of small ovarian follicles for infertility treatment. PNAS NEXUS 2022; 1:pgac108. [PMID: 36741430 PMCID: PMC9896916 DOI: 10.1093/pnasnexus/pgac108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 07/02/2022] [Indexed: 02/07/2023]
Abstract
In-vitro fertilization is an effective treatment for various causes of infertility. However, management of women with poor ovarian response or premature ovarian insufficiency remains challenging because these women have underdeveloped small ovarian follicles that do not respond to hormone treatment. In-vitro activation of small follicles has been developed but its efficiency has much room for improvement. In the current study, we provide several lines of evidence showing that curcumin, an FDA-approved traditional medicine, can specifically promote the development of mouse ovarian follicles from the primary to secondary stage, which greatly potentiates these small follicles for subsequent in-vivo development into antral follicles that can be ovulated. Mechanistically, we show that curcumin promotes the proliferation and differentiation of granulosa cells and the growth of oocytes by activating the phosphatidylinositol 3 kinase (PI3K) signaling pathway. Most importantly, we show that in-vitro treatment of human ovarian tissues with curcumin can promote the in-vivo survival and development of small human ovarian follicles, showing that curcumin can be used as a potential drug to increase the success rate of in-vitro activation of small human follicles. We thus identify curcumin as a novel potential drug for promoting the development of small human ovarian follicles for infertility treatment.
Collapse
Affiliation(s)
- Yu Zhao
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong—Shenzhen Hospital, Haiyuan First Road 1, Shenzhen, Guangdong 518053, China,Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Yihui Zhang
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong—Shenzhen Hospital, Haiyuan First Road 1, Shenzhen, Guangdong 518053, China,Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Dongteng Liu
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong—Shenzhen Hospital, Haiyuan First Road 1, Shenzhen, Guangdong 518053, China,Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Haiwei Feng
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong—Shenzhen Hospital, Haiyuan First Road 1, Shenzhen, Guangdong 518053, China
| | - Xiaohui Wang
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong—Shenzhen Hospital, Haiyuan First Road 1, Shenzhen, Guangdong 518053, China
| | - Jiajun Su
- Department of Anatomical Pathology, The University of Hong Kong—Shenzhen Hospital, Haiyuan First Road 1, Shenzhen, Guangdong 518053, China
| | - Yuanqing Yao
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong—Shenzhen Hospital, Haiyuan First Road 1, Shenzhen, Guangdong 518053, China
| | - Ernest H Y Ng
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong—Shenzhen Hospital, Haiyuan First Road 1, Shenzhen, Guangdong 518053, China,Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - William S B Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong—Shenzhen Hospital, Haiyuan First Road 1, Shenzhen, Guangdong 518053, China,Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Raymond H W Li
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong—Shenzhen Hospital, Haiyuan First Road 1, Shenzhen, Guangdong 518053, China,Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | | | - Kui Liu
- To whom correspondence should be addressed:
| |
Collapse
|
64
|
Haq SIU, Shang J, Xie H, Qiu QS. Roles of TOR signaling in nutrient deprivation and abiotic stress. JOURNAL OF PLANT PHYSIOLOGY 2022; 274:153716. [PMID: 35597106 DOI: 10.1016/j.jplph.2022.153716] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/25/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
In living organisms, nutrient, energy, and environmental stimuli sensing and signaling are considered as the most primordial regulatory networks governing growth and development. Target of Rapamycin (TOR) is a diversified Serine/Threonine protein kinase existing in all eukaryotes that regulates distinct salient growth and developmental signaling pathways. TOR signaling acts as a central hub in plants that allows a variety of nutrients, energy, hormones, and environmental stimuli to be integrated. TOR is activated by several nutrients and promotes energy-consuming processes such as cell division, protein translation, mRNA translation and ribosome biogenesis. We summarized the recent findings on the TOR function in regulating the dynamic networks of nutrients, including sugar, sulfur, nitrogen, carbon, phosphorus, potassium, and amino acids. TOR's role in abiotic stress was discussed, in which TOR orchestrating stress signaling, including heat, cold, salt, and osmotic stress, to regulate transcriptional and metabolic reprogramming, as well as growth and development. The interconnections between TOR and SnRK1 kinase were discussed in controlling nutrient deprivation and abiotic stress.
Collapse
Affiliation(s)
- Syed Inzimam Ul Haq
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, 73000, China
| | - Jun Shang
- Academy of Plateau Science and Sustainability, School of Life Sciences, Qinghai Normal University, Xining, Qinghai, 810000, China; Qinghai Provincial Key Laboratory of Medicinal Plant and Animal Resources of Qinghai-Tibet Plateau, Xining, Qinghai, 810008, China
| | - Huichun Xie
- Academy of Plateau Science and Sustainability, School of Life Sciences, Qinghai Normal University, Xining, Qinghai, 810000, China; Qinghai Provincial Key Laboratory of Medicinal Plant and Animal Resources of Qinghai-Tibet Plateau, Xining, Qinghai, 810008, China
| | - Quan-Sheng Qiu
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, 73000, China; Academy of Plateau Science and Sustainability, School of Life Sciences, Qinghai Normal University, Xining, Qinghai, 810000, China.
| |
Collapse
|
65
|
Rab22a Promotes Epithelial-Mesenchymal Transition in Papillary Thyroid Carcinoma by Activating PI3K/AKT/mTOR Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1874550. [PMID: 35757470 PMCID: PMC9217539 DOI: 10.1155/2022/1874550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/30/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022]
Abstract
Background Rab22a is a member of the RAS superfamily, involved in early endosome formation and intracellular vesicle transport. Rab22a is significantly upregulated in a variety of malignant tumors. However, its function in thyroid cancer has never been addressed. Methods The expression of Rab22a in paraffin sections of 101 patients was detected by immunohistochemical staining. By upregulating and downregulating the expression of Rab22a in thyroid cancer cell lines, the effect of Rab22a on cell proliferation, invasion, and migration was analyzed. Co-IP was employed, and the interaction between Rab22a and PI3Kp85α was shown. The function of Rab22a on PI3K/AKT/mTOR signaling and epithelial-mesenchymal transition (EMT) was further studied by western blot analysis. Results Immunostaining showed that Rab22a was significantly overexpressed in thyroid cancer tissues but negative in adjacent normal tissues or nodular goiters. The proliferation, migration, invasion, and EMT in papillary thyroid carcinoma cell lines were enhanced upon Rab22a overexpression but inhibited after knocking down Rab22a. The co-IP assay demonstrated an interaction between Rab22a and PI3K85α, an effector of PI3K. We further found that Rab22a can activate the PI3K/AKT/mTOR signaling pathway. However, the ability of Rab22a to promote the proliferation, invasion, migration, and EMT of papillary thyroid carcinoma cells was significantly inhibited after being treated with LY294002, a PI3K inhibitor. Conclusions Rab22a can promote the EMT process and enhance proliferation, migration, and invasion of papillary thyroid carcinoma cells by activating the PI3K/AKT/mTOR signaling pathway. Our study provides new pathological diagnosis clues and clinical treatment targets for thyroid cancer.
Collapse
|
66
|
Wang M, Wang L, Tan X, Wang L, Xiong X, Wang Y, Wang Q, Yang H, Yin Y. The developmental changes in intestinal epithelial cell proliferation, differentiation, and shedding in weaning piglets. ANIMAL NUTRITION 2022; 9:214-222. [PMID: 35600553 PMCID: PMC9092860 DOI: 10.1016/j.aninu.2021.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/20/2021] [Accepted: 11/07/2021] [Indexed: 10/24/2022]
|
67
|
Zhai Y, Zhou X, Wang X. Novel insights into the biomarkers and therapies for primary central nervous system lymphoma. Ther Adv Med Oncol 2022; 14:17588359221093745. [PMID: 35558005 PMCID: PMC9087239 DOI: 10.1177/17588359221093745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/24/2022] [Indexed: 11/17/2022] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare and highly aggressive extranodal type of non-Hodgkin lymphoma. After the introduction and widespread use of high-dose-methotrexate (HD-MTX)-based polychemotherapy, treatment responses of PCNSL have been improved. However, long-term prognosis for patients who have failed first-line therapy and relapsed remains poor. Less invasive diagnostic markers, including the circulating tumor DNAs (ctDNAs), microRNAs, metabolomic markers, and other novel biomarkers, such as a proliferation inducing ligand (APRIL) and B-cell activating factor of the TNF family (BAFF), have shown potential to distinguish PCNSL at an early stage, and some of them are related with prognosis to a certain extent. Recent insights into novel therapies, including Bruton tyrosine kinase (BTK) inhibitors, immunomodulatory drugs, immune checkpoint inhibitors, PI3K/mTOR inhibitors, and chimeric antigen receptor (CAR) T cells, have revealed encouraging efficacy in treatment response, whereas the duration of response and long-term survival of patients with relapsed or refractory PCNSL (r/r PCNSL) need further improvement. In addition, the diagnostic efficiency of novel markers and the antitumor efficacy of novel therapies are needed to be assessed further in larger clinical trials. This review provides an overview of recent research on novel diagnostic markers and therapeutic strategies for PCNSL.
Collapse
Affiliation(s)
- Yujia Zhai
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, ChinaSchool of Medicine, Shandong University, Jinan, China
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, China
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No. 324, Jingwu Road, Jinan 250021, Shandong, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- School of Medicine, Shandong University, Jinan, China
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, China
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, China
| |
Collapse
|
68
|
Cardiovascular protection associated with cilostazol, colchicine and target of rapamycin inhibitors. J Cardiovasc Pharmacol 2022; 80:31-43. [PMID: 35384911 DOI: 10.1097/fjc.0000000000001276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/06/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT An alteration in extracellular matrix production by vascular smooth muscle cells is a crucial event in the pathogenesis of vascular diseases such as aging-related, atherosclerosis and allograft vasculopathy. The human target of rapamycin (TOR) is involved in the synthesis of extracellular matrix by vascular smooth muscle cells. TOR inhibitors reduce arterial stiffness, blood pressure, and left ventricle hypertrophy and decrease cardiovascular risk in kidney graft recipients and patients with coronary artery disease and heart allograft vasculopathy. Other drugs that modulate extracellular matrix production such as cilostazol and colchicine have also demonstrated a beneficial cardiovascular effect. Clinical studies have consistently shown that cilostazol confers cardiovascular protection in peripheral vascular disease, coronary artery disease, and cerebrovascular disease. In patients with type 2 diabetes, cilostazol prevents the progression of subclinical coronary atherosclerosis. Colchicine reduces arterial stiffness in patients with Familial Mediterranean Fever and patients with coronary artery disease. Pathophysiological mechanisms underlying the cardioprotective effect of these drugs may be related to interactions between the cytoskeleton, TOR signaling and cyclic AMP synthesis that remain to be fully elucidated. Adult vascular smooth muscle cells exhibit a contractile phenotype and produce little extracellular matrix. Conditions that upregulate extracellular matrix synthesis induce a phenotypic switch toward a synthetic phenotype. TOR inhibition with rapamycin reduces extracellular matrix production by promoting the change to the contractile phenotype. Cilostazol increases the cytosolic level of cyclic AMP, which in turn leads to a reduction in extracellular matrix synthesis. Colchicine is a microtubule-destabilizing agent that may enhance the synthesis of cyclic AMP.
Collapse
|
69
|
Wang K, Qian R, Li H, Wang C, Ding Y, Gao Z. Interpreting the Pharmacological Mechanisms of Sho-saiko-to on Thyroid Carcinoma through Combining Network Pharmacology and Experimental Evaluation. ACS OMEGA 2022; 7:11166-11176. [PMID: 35415320 PMCID: PMC8991932 DOI: 10.1021/acsomega.1c07335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/14/2022] [Indexed: 06/14/2023]
Abstract
Sho-saiko-to is a well-known traditional Chinese medicine compound and is considered to have therapeutic effects against many diseases, including thyroid cancer (TC). However, the mechanisms and therapeutic targets of Sho-saiko-to against TC remain unclear. In this study, network pharmacology, molecular docking, and cell experiments were combined to predict and verify the targets and mechanisms of the active ingredients of Sho-saiko-to against TC. The results demonstrated that the main chemical ingredients of Sho-saiko-to could suppress the viability and proliferation of TC cells, promote apoptosis through the caspase3 pathway, and induce autophagy through the PI3K-AKT pathway. In addition, Sho-saiko-to could also induce the redifferentiation of anaplastic thyroid cancer. Our study provides a novel approach for treating differentiated thyroid cancer (DTC) or radioactive iodine refractory differentiated thyroid cancer (RAIR-DTC).
Collapse
Affiliation(s)
- Kun Wang
- Department
of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei
Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Ruijie Qian
- Department
of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei
Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Hongyan Li
- Department
of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei
Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Chenyang Wang
- Department
of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei
Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Ying Ding
- Department
of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei
Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Zairong Gao
- Department
of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei
Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
70
|
eIF3a regulation of mTOR signaling and translational control via HuR in cellular response to DNA damage. Oncogene 2022; 41:2431-2443. [PMID: 35279705 PMCID: PMC9035104 DOI: 10.1038/s41388-022-02262-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 01/29/2023]
Abstract
eIF3a (eukaryotic translation initiation factor 3a), a subunit of the eIF3 complex, has been suggested to play a regulatory role in protein synthesis and in cellular response to DNA-damaging treatments. S6K1 is an effector and a mediator of mTOR complex 1 (mTORC1) in regulating protein synthesis and integrating diverse signals into control of cell growth and response to stress. Here, we show that eIF3a regulates S6K1 activity by inhibiting mTORC1 kinase via regulating Raptor synthesis. The regulation of Raptor synthesis is via eIF3a interaction with HuR (human antigen R) and binding of the eIF3a-HuR complex to the 5'-UTR of Raptor mRNA. Furthermore, mTORC1 may mediate eIF3a function in cellular response to cisplatin by regulating synthesis of NER proteins and NER activity. Taken together, we conclude that the mTOR signaling pathway may also be regulated by translational control and mediate eIF3a regulation of cancer cell response to cisplatin by regulating NER protein synthesis.
Collapse
|
71
|
In vivo CRISPR screening identifies BAZ2 chromatin remodelers as druggable regulators of mammalian liver regeneration. Cell Stem Cell 2022; 29:372-385.e8. [PMID: 35090595 PMCID: PMC8897233 DOI: 10.1016/j.stem.2022.01.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 09/17/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022]
Abstract
Identifying new pathways that regulate mammalian regeneration is challenging due to the paucity of in vivo screening approaches. We employed pooled CRISPR knockout and activation screening in the regenerating liver to evaluate 165 chromatin regulatory proteins. Both screens identified the imitation-SWI chromatin remodeling components Baz2a and Baz2b, not previously implicated in regeneration. In vivo sgRNA, siRNA, and knockout strategies against either paralog confirmed increased regeneration. Distinct BAZ2-specific bromodomain inhibitors, GSK2801 and BAZ2-ICR, resulted in accelerated liver healing after diverse injuries. Inhibitor-treated mice also exhibited improved healing in an inflammatory bowel disease model, suggesting multi-tissue applicability. Transcriptomics on regenerating livers showed increases in ribosomal and cell cycle mRNAs. Surprisingly, CRISPRa screening to define mechanisms showed that overproducing Rpl10a or Rpl24 was sufficient to drive regeneration, whereas Rpl24 haploinsufficiency was rate limiting for BAZ2 inhibition-mediated regeneration. The discovery of regenerative roles for imitation-SWI components provides immediate strategies to enhance tissue repair.
Collapse
|
72
|
An H, Gan T, Tang M, Chen H. Molecular Mechanism of Overcoming Host Resistance by the Target of Rapamycin Gene in Leptographium qinlingensis. Microorganisms 2022; 10:microorganisms10030503. [PMID: 35336079 PMCID: PMC8954470 DOI: 10.3390/microorganisms10030503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023] Open
Abstract
Leptographium qinlingensis is a fungal symbiont of the Chinese white pine beetle (Dendroctonus armandi) and a pathogen of the Chinese white pine (Pinus armandii) that must overcome the terpenoid oleoresin defenses of host trees to invade and colonize. L. qinlingensis responds to monoterpene flow with abundant mechanisms that include the decomposing and use of these compounds as a nitrogen source. Target of Rapamycin (TOR) is an evolutionarily conserved protein kinase that plays a central role in both plants and animals through integration of nutrients, energies, hormones, growth factors and environmental inputs to control proliferation, growth and metabolism in diverse multicellular organisms. In this study, in order to explore the relationship between TOR gene and carbon sources, nitrogen sources, host nutrients and host volatiles (monoterpenoids) in L. qinlingensis, we set up eight carbon source treatments, ten nitrogen source treatments, two host nutrients and six monoterpenoids (5%, 10% and 20%) treatments, and prepared different media conditions. By measuring the biomass and growth rate of mycelium, the results revealed that, on the whole, the response of L. qinlingensis to nitrogen sources was better than carbon sources, and the fungus grew well in maltose (carbon source), (NH4)2C2O4 (inorganic nitrogen source), asparagine (organic nitrogen source) and P. armandii (host nutrient) versus other treatments. Then, by analyzing the relationship between TOR expression and different nutrients, the data showed that: (i) TOR expression exhibited negative regulation in response to carbon sources and host nutrition. (ii) The treatments of nitrogen sources and terpenoids had positively regulatory effects on TOR gene; moreover, the fungus was most sensitive to β-pinene and 3-carene. In conclusion, our findings reveal that TOR in L. qinlingensis plays a key role in the utilization of host volatiles as nutrient intake, overcoming the physical and chemical host resistances and successful colonization.
Collapse
|
73
|
Zhou Y, Xu X. Application of new targeted drugs in relapsed/refractory primary central nervous system lymphoma. Hematology 2022; 27:105-112. [PMID: 35068379 DOI: 10.1080/16078454.2021.2019363] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ying Zhou
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, People’s Republic of China
| | - Xiaohong Xu
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, People’s Republic of China
| |
Collapse
|
74
|
Mohseni AH, Casolaro V, Bermúdez-Humarán LG, Keyvani H, Taghinezhad-S S. Modulation of the PI3K/Akt/mTOR signaling pathway by probiotics as a fruitful target for orchestrating the immune response. Gut Microbes 2022; 13:1-17. [PMID: 33615993 PMCID: PMC7899637 DOI: 10.1080/19490976.2021.1886844] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) and the phosphatidylinositol-3-kinase (PI3K)/protein kinase B or Akt (PKB/Akt) signaling pathways are considered as two but somewhat interconnected significant immune pathways which play complex roles in a variety of physiological processes as well as pathological conditions. Aberrant activation of PI3K/Akt/mTOR signaling pathways has been reported to be associated in a wide variety of human diseases. Over the past few years, growing evidence in in vitro and in vivo models suggest that this sophisticated and subtle cascade mediates the orchestration of the immune response in health and disease through exposure to probiotics. An expanding body of literature has highlighted the contribution of probiotics and PI3K/Akt/mTOR signaling pathways in gastrointestinal disorders, metabolic syndrome, skin diseases, allergy, salmonella infection, and aging. However, longitudinal human studies are possibly required to verify more conclusively whether the investigational tools used to understand the regulation of these pathways might provide effective approaches in the prevention and treatment of various disorders. In this Review, we summarize the experimental evidence from recent peer-reviewed studies and provide a brief overview of the causal relationship between the effects of probiotics and their metabolites on the components of PI3K/Akt/mTOR signaling pathways and human disease.
Collapse
Affiliation(s)
- Amir Hossein Mohseni
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Salerno, Italy
| | | | - Hossein Keyvani
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran,Hossein Keyvani Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran, Tel +98 21 88715350
| | - Sedigheh Taghinezhad-S
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran,CONTACT Sedigheh Taghinezhad-S Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, 1477893855, Iran
| |
Collapse
|
75
|
Ince W, Eisen T. Combination therapies in clinical trials for renal cell carcinoma: how could they impact future treatments? Expert Opin Investig Drugs 2022; 30:1221-1229. [PMID: 34875200 DOI: 10.1080/13543784.2021.2014814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Pharmacological combinations using immune checkpoint inhibition (ICI), tyrosine kinase inhibition (TKIs), and mammalian target of rapamycin inhibitors (mTOR) have improved survival in metastatic clear cell renal cell cancer (mccRCC). Despite improvements in survival, complete durable responses are rare. AREAS COVERED Molecular pathways involved in mccRCC and drugs targets are highlighted. The background and rationale for combination therapy are covered. Results from combination trials are reviewed and potential approaches with biomarker-stratified treatment and novel experimental agents are examined. PubMed Central and ClinicalTrials.gov were searched. Search terms used to identify clinical trials were '(metastatic renal cell cancer OR renal cell carcinoma OR mccRCC OR mRCC OR RCC OR kidney cancer) AND (combination OR combined).' EXPERT OPINION First-line standard of care has moved to combination therapy with ICI-ICI and TKI-ICI combinations; VEGF-mTORi is available in subsequent lines. Combining targeted treatments without validated biomarkers is imprecise, and combinations may lead to overtreatment of a subset of patients, exposing them to unnecessary toxicity. The aim of combinations must be clear: improvement in overall survival (OS) and complete response (CR). Recent data suggest a role for novel biomarker stratification rather traditional risk groups. Further combination approaches with triplets and quadruplets should be biomarker directed.
Collapse
Affiliation(s)
- Will Ince
- Department of Oncology, Addenbrookes's Hospital, Cambridge, UK
| | - Tim Eisen
- Department of Oncology, Addenbrookes's Hospital, Cambridge, UK
| |
Collapse
|
76
|
Cebey-López M, Currás-Tuala MJ, Gómez-Rial J, Rivero-Calle I, Pardo-Seco J, Mendez-Gallart R, Pischedda S, Gómez-Carballa A, Barral-Arca R, Justicia-Grande A, Viz-Lasheras S, Rodríguez-Tenreiro C, Gómez R, Salas A, Martinón-Torres F. Case Report: Everolimus reduced bone turnover markers but showed no clinical benefit in a patient with severe progressive osseous heteroplasia. Front Pediatr 2022; 10:936780. [PMID: 36483469 PMCID: PMC9723155 DOI: 10.3389/fped.2022.936780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 10/24/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Progressive osseous heteroplasia (POH) is an ultrarare genetic disorder characterized by an inactivating mutation in the GNAS gene that causes heterotopic ossification. Inhibition of the mammalian target of the rapamycin (mTOR) signalling pathway has been proposed as a therapy for progressive bone fibrodysplasia and non-genetic forms of bone heteroplasia. Herein, we describe the impact of using Everolimus as a rescue therapy for an identical twin girl exhibiting an aggressive clinical phenotype of POH. METHODS Clinical evaluation of the progression of the disease during Everolimus treatment was performed periodically. Cytokine markers involved in bone metabolism and protein markers related to bone activity were analyzed to explore bone turnover activity. RESULTS The patient received Everolimus therapy for 36 weeks. During treatment, no clinical improvement of the disease was perceived. Analysis of biochemical parameters, namely, β-CTX (r 2 = -0.576, P-value = 0.016) and PNIP (r 2 = -0.598, P-value = 0.011), indicated that bone turnover activity was significantly reduced. Additionally, bone metabolism-related biomarkers showed only a significant positive correlation with PTH levels. CONCLUSIONS Everolimus treatment did not modify the clinical progression of the disease in an aggressive form of POH, although an impact on the protein markers studied was observed.
Collapse
Affiliation(s)
- M Cebey-López
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - M J Currás-Tuala
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - J Gómez-Rial
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,Servicio de inmunologia, Servicio de Análisis Clínicos. Hospital Clínico Universitario (SERGAS), Santiago de Compostela, Spain
| | - I Rivero-Calle
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - J Pardo-Seco
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - R Mendez-Gallart
- Pediatric Surgery, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - S Pischedda
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - A Gómez-Carballa
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - R Barral-Arca
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - A Justicia-Grande
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - S Viz-Lasheras
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - C Rodríguez-Tenreiro
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - R Gómez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital (SERGAS), Santiago de Compostela, Spain
| | - A Salas
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigación Sanitaria, Hospital Clínico Universitario de Santiago (SERGAS), Santiago de Compostela, Spain
| | - F Martinón-Torres
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
77
|
Synergistic effects of Rapamycin and Fluorouracil to treat a gastric tumor in a PTEN conditional deletion mouse model. Gastric Cancer 2022; 25:96-106. [PMID: 34370147 DOI: 10.1007/s10120-021-01229-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023]
Abstract
The tumor suppressor gene phosphatase and tensin homolog (PTEN) in PI3K/Akt/mTOR pathway is essential in inhibiting tumor growth and metastasis. However, whether the mutation of PTEN gene could induce tumorigenesis and impact the treatment of gastric cancer is still unclear. The purpose of the study was to investigate the combined treatment of gastric tumorigenesis using Rapamycin and Fluorouracil (5-Fu) through interfering with the Akt/mTOR pathway in a mouse model with PTEN conditional deletion. Three groups of mice were exposed for 5 days to Rapamycin and 5-Fu separately and together. The gene expression of the Akt/mTOR pathway, the protein expression of caspase-3 and p-Akt, p-S6K and p-4EBP1, and the pathological changes in stomachs were analyzed. Our study demonstrates that the conditional PTEN deletion in the cells of glandular stomach induces hyperplastic gastric tumors in mice. The combined Rapamycin administration with 5-Fu resulted in better outcomes than their separate administration for the treatment of gastric cancer by inhibiting the mTOR signal pathway. Our study indicates that Rapamycin has a synergistic interaction with chemotherapeutic 5-Fu, and demonstrates a potential therapeutic combination treatment on glandular stomach tumor with PTEN functional absence or aberrantly activated Akt/mTOR pathway. It provides important insights into the inhibition of the Akt/mTOR pathway in gastric cancer clinical therapy.
Collapse
|
78
|
Musiol R, Malecki P, Pacholczyk M, Mularski J. Terpyridines as promising antitumor agents: an overview of their discovery and development. Expert Opin Drug Discov 2021; 17:259-271. [PMID: 34928186 DOI: 10.1080/17460441.2022.2017877] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The fused aromatic system of terpyridines makes them good, innocent ligands for various metals. The resulting complexes have been extensively studied for both their biological activity and physico-chemical properties. However, although free ligands also have an interesting biological activity, their share in recent research is considerably limited. AREAS COVERED This review covers the literature on the anticancer activity of terpyridines with special attention being paid to their use as free ligands. Whenever possible, the mechanism of action has been discussed, thereby providing evidence of the substantial differences between sole ligands or less stable complexes and those that have heavier elements. EXPERT OPINION The existing literature indicates that there is a specific attitude for investigating terpyridines and their transition metal complexes. While the latter have been well explored and recognized in the scientific community, the free terpyridines are considered to be useful solely due to their complexing ability. At the same time, terpyridines could have similar or even higher anticancer potency than their complexes. Moreover, a mechanistic analysis of the stability and intracellular activity would provide information that would be useful for designing new drugs.
Collapse
Affiliation(s)
- Robert Musiol
- Faculty of Science and Technology, University of Silesia in Katowice, Szkolna 7, Katowice, Poland
| | | | - Marcin Pacholczyk
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, Poland
| | - Jacek Mularski
- Faculty of Science and Technology, University of Silesia in Katowice, Szkolna 7, Katowice, Poland
| |
Collapse
|
79
|
Das R, Choithramani A, Shard A. A molecular perspective for the use of type IV tyrosine kinase inhibitors as anticancer therapeutics. Drug Discov Today 2021; 27:808-821. [PMID: 34920095 DOI: 10.1016/j.drudis.2021.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/21/2021] [Accepted: 12/10/2021] [Indexed: 11/03/2022]
Abstract
Tyrosine kinases are enzymes that can transfer a phosphate group from ATP to a specific protein tyrosine, serine or threonine residue within a cell, operating as a switch that can turn 'on' and 'off' causing different physiological alterations in the body. Mutated kinases have been shown to display an equilibrium shift toward the activated state. Types I-III have been studied intensively leading to drugs like imatinib (type II), cobimetinib (type III), among others. It is the same scenario for types V-VII; however, there is a lacuna in information regarding type IV inhibitors, although recently some advances have surfaced. This review aims to accumulate the knowledge gained so far about type IV inhibitors.
Collapse
Affiliation(s)
- Rudradip Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 380054, India
| | - Asmita Choithramani
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 380054, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 380054, India.
| |
Collapse
|
80
|
A Phase II Study to Assess the Safety and Efficacy of the Dual mTORC1/2 and PI3K Inhibitor Bimiralisib (PQR309) in Relapsed, Refractory Lymphoma. Hemasphere 2021; 5:e656. [PMID: 34901759 PMCID: PMC8660000 DOI: 10.1097/hs9.0000000000000656] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/07/2021] [Indexed: 11/28/2022] Open
Abstract
Bimiralisib is an orally bioavailable pan-phosphatidylinositol 3-kinase and mammalian target of rapamycin inhibitor which has shown activity against lymphoma in preclinical models. This phase I/II study evaluated the response rate to bimiralisib at 2 continuous dose levels (60 mg and 80 mg) in patients with relapsed/refractory lymphoma. Fifty patients were enrolled and started treatment. The most common histologies were diffuse large B-cell lymphoma (n = 17), follicular lymphoma (n = 9), T-cell lymphoma (n = 8), and others (mostly indolent). Patients had been treated with a median of 5 prior lines of treatment and 44% were considered refractory to their last treatment. Mean duration of treatment (and standard deviations) with 60 mg once daily (o.d.) was 1.3 ± 1.2 months, and with 80 mg o.d. 3.7 ± 3.9 months. On an intention to treat analysis, the overall response rate was 14% with 10% achieving a partial response and 4% a complete response. Thirty-six percent of patients were reported as having stable disease. No dose-limiting toxicities were observed during the phase I portion of the study. Overall, 70% of patients had a grade 3 treatment emergent adverse events (TEAE) and 34% had a grade 4 TEAE; 28% of patients discontinued treatment due to toxicity. The most frequent TEAEs grade ≥3 was hyperglycemia (24%), neutropenia (20%), thrombocytopenia (22%), and diarrhea (12%). Per protocol, hyperglycemia required treatment with oral antihyperglycemic agents in 28% and with insulin in 14%. At 60 mg or 80 mg continuous dosing, bimiralisib showed modest efficacy with significant toxicity in heavily pretreated patients with various histological subtypes of lymphoma.
Collapse
|
81
|
Chandra S, Mannino PJ, Thaller DJ, Ader NR, King MC, Melia TJ, Lusk CP. Atg39 selectively captures inner nuclear membrane into lumenal vesicles for delivery to the autophagosome. J Cell Biol 2021; 220:e202103030. [PMID: 34714326 PMCID: PMC8575018 DOI: 10.1083/jcb.202103030] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/26/2021] [Accepted: 09/29/2021] [Indexed: 12/26/2022] Open
Abstract
Mechanisms that turn over components of the nucleus and inner nuclear membrane (INM) remain to be fully defined. We explore how components of the INM are selected by a cytosolic autophagy apparatus through a transmembrane nuclear envelope-localized cargo adaptor, Atg39. A split-GFP reporter showed that Atg39 localizes to the outer nuclear membrane (ONM) and thus targets the INM across the nuclear envelope lumen. Consistent with this, sequence elements that confer both nuclear envelope localization and a membrane remodeling activity are mapped to the Atg39 lumenal domain; these lumenal motifs are required for the autophagy-mediated degradation of integral INM proteins. Interestingly, correlative light and electron microscopy shows that the overexpression of Atg39 leads to the expansion of the ONM and the enclosure of a network of INM-derived vesicles in the nuclear envelope lumen. Thus, we propose an outside-in model of nucleophagy where INM is delivered into vesicles in the nuclear envelope lumen, which can be targeted by the autophagosome.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas J. Melia
- Department of Cell Biology, Yale School of Medicine, New Haven, CT
| | - C. Patrick Lusk
- Department of Cell Biology, Yale School of Medicine, New Haven, CT
| |
Collapse
|
82
|
Chakraborty S, Utter MB, Frias MA, Foster DA. Cancer cells with defective RB and CDKN2A are resistant to the apoptotic effects of rapamycin. Cancer Lett 2021; 522:164-170. [PMID: 34563639 DOI: 10.1016/j.canlet.2021.09.020] [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: 06/23/2021] [Revised: 08/28/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
Inhibition of mammalian target of rapamycin complex 1 (mTORC1) with rapamycin in the absence of transforming growth factor-β (TGFβ) signaling induces apoptosis in many cancer cell lines. In the presence of TGFβ, rapamycin induces G1 cell cycle arrest; however, in the absence of TGFβ, cells do not arrest in G1 and progress into S-phase where rapamycin is cytotoxic rather than cytostatic. However, we observed that DU145 prostate and NCI-H2228 lung cancer cells were resistant to the cytotoxic effect of rapamycin. Of interest, the rapamycin-resistant DU145 and NCI-H2228 cells have mutations in the RB and CDKN2A tumor suppressor genes. The gene products of RB and CDKN2A (pRb and p14ARF) suppress E2F family transcription factors that promote cell cycle progression from G1 into S. Restoration of wild type RB or inhibition of E2F activity in DU145 and NCI-H2228 cells led to rapamycin sensitivity. These data provide evidence that the combination of mutant RB and mutant CDKN2A in cancer cells leads to rapamycin resistance, which has implications for precision medicine approaches to anti-cancer therapies.
Collapse
Affiliation(s)
- Sohag Chakraborty
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, USA; Biochemistry Program, Graduate Center of the City University of New York, NY, New York, USA
| | - Matthew B Utter
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, USA; Biochemistry Program, Graduate Center of the City University of New York, NY, New York, USA
| | - Maria A Frias
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, USA
| | - David A Foster
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, USA; Biochemistry Program, Graduate Center of the City University of New York, NY, New York, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
83
|
Paez-Nova M, Andaur K, García-Ballestas E, Bustos-Salazar D, Moscote-Salazar LR, Koller O, Valenzuela S. Primary intracranial smooth muscle tumor associated with Epstein-Barr virus in immunosuppressed children: two cases report and review of literature. Childs Nerv Syst 2021; 37:3923-3932. [PMID: 33884483 DOI: 10.1007/s00381-021-05173-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 04/15/2021] [Indexed: 11/29/2022]
Abstract
Primary intracranial smooth muscle tumors are rare. Most cases are related to Epstein-Barr virus proliferation in immunocompromised patients such as organ solid recipients. Only a few cases have been reported in pediatric patients. The clinical features are very variable depending mainly on the location and size of the smooth muscle tumor (SMT) and the pathogenesis is poorly understood. We describe two cases of intracranial SMT localized in the temporal lobe and associated with EBV in immunosuppressed children. A review of the literature associated with intracranial leiomyomas was also done.
Collapse
Affiliation(s)
- Maximiliano Paez-Nova
- Pediatric Neurosurgery Department, Dr. Asenjo Neurosurgical Institute, Santiago, Chile. .,Radioneurosurgery and Functional Neurosurgery Unit, Sheba Medical Center, Ramat Gan, Israel. .,Sheba Medical Center at Tel HaShomer, Tel Aviv University, Tel Aviv, Israel.
| | - Karem Andaur
- Pediatric Neurology Department, University of Santiago de Chile, Santiago, Chile
| | - Ezequiel García-Ballestas
- Latin American Council of Neurocritical Care-CLaNi, Cartagena, Colombia.,Center of Biomedical Research-CIB, Faculty of Medicine, University of Cartagena, Cartagena, Colombia
| | - Diego Bustos-Salazar
- Center of Biomedical Research-CIB, Faculty of Medicine, University of Cartagena, Cartagena, Colombia
| | - Luis Rafael Moscote-Salazar
- Latin American Council of Neurocritical Care-CLaNi, Cartagena, Colombia.,Center of Biomedical Research-CIB, Faculty of Medicine, University of Cartagena, Cartagena, Colombia
| | - Osvaldo Koller
- Pediatric Neurosurgery Department, Dr. Asenjo Neurosurgical Institute, Santiago, Chile.,Pediatric Neurosurgery Department, Alemana Clinic, Santiago, Chile
| | - Sergio Valenzuela
- Pediatric Neurosurgery Department, Dr. Asenjo Neurosurgical Institute, Santiago, Chile.,Pediatric Neurosurgery Department, Alemana Clinic, Santiago, Chile
| |
Collapse
|
84
|
Champagne J, Pataskar A, Blommaert N, Nagel R, Wernaart D, Ramalho S, Kenski J, Bleijerveld OB, Zaal EA, Berkers CR, Altelaar M, Peeper DS, Faller WJ, Agami R. Oncogene-dependent sloppiness in mRNA translation. Mol Cell 2021; 81:4709-4721.e9. [PMID: 34562372 DOI: 10.1016/j.molcel.2021.09.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/13/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022]
Abstract
mRNA translation is a highly conserved and tightly controlled mechanism for protein synthesis. Despite protein quality control mechanisms, amino acid shortage in melanoma induces aberrant proteins by ribosomal frameshifting. The extent and the underlying mechanisms related to this phenomenon are yet unknown. Here, we show that tryptophan depletion-induced ribosomal frameshifting is a widespread phenomenon in cancer. We termed this event sloppiness and strikingly observed its association with MAPK pathway hyperactivation. Sloppiness is stimulated by RAS activation in primary cells, suppressed by pharmacological inhibition of the oncogenic MAPK pathway in sloppy cells, and restored in cells with acquired resistance to MAPK pathway inhibition. Interestingly, sloppiness causes aberrant peptide presentation at the cell surface, allowing recognition and specific killing of drug-resistant cancer cells by T lymphocytes. Thus, while oncogenes empower cancer progression and aggressiveness, they also expose a vulnerability by provoking the production of aberrant peptides through sloppiness.
Collapse
Affiliation(s)
- Julien Champagne
- Division of Oncogenomics, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Abhijeet Pataskar
- Division of Oncogenomics, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Naomi Blommaert
- Division of Oncogenomics, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Remco Nagel
- Division of Oncogenomics, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Demi Wernaart
- Division of Oncogenomics, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Sofia Ramalho
- Division of Oncogenomics, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Juliana Kenski
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Onno B Bleijerveld
- Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Esther A Zaal
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University and Netherlands Proteomics Centre, Utrecht, the Netherlands
| | - Celia R Berkers
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University and Netherlands Proteomics Centre, Utrecht, the Netherlands
| | - Maarten Altelaar
- Proteomics Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University and Netherlands Proteomics Centre, Utrecht, the Netherlands
| | - Daniel S Peeper
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - William J Faller
- Division of Oncogenomics, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Reuven Agami
- Division of Oncogenomics, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; Erasmus MC, Rotterdam University, Rotterdam, the Netherlands.
| |
Collapse
|
85
|
Update on Novel Therapeutics for Primary CNS Lymphoma. Cancers (Basel) 2021; 13:cancers13215372. [PMID: 34771535 PMCID: PMC8582401 DOI: 10.3390/cancers13215372] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Primary central nervous system lymphoma is a rare and aggressive form of non-Hodgkin lymphoma. While it is highly responsive to first-line chemo and radiation treatments, rates of relapse are high, demonstrating the need for improved therapeutic strategies. Recent advancements in the understanding of the pathophysiology of this disease have led to the identification of new potential treatment targets and the development of novel agents. This review aims to discuss different targeted strategies and review some of the data supporting these approaches, and discusses recently completed and ongoing clinical trials using these novel agents. Abstract Primary central nervous system lymphoma (PCNSL) is a rare lymphoma isolated to the central nervous system or vitreoretinal space. Standard treatment consists of cytotoxic methotrexate-based chemotherapy, with or without radiation. Despite high rates of response, relapse is common, highlighting the need for novel therapeutic approaches. Recent advances in the understanding of PCNSL have elucidated mechanisms of pathogenesis and resistance including activation of the B-cell receptor and mammalian target of rapamycin pathways. Novel treatment strategies such as the Bruton’s tyrosine kinase (BTK) inhibitor ibrutinib, phosphatidylinositol-3 kinase (PI3K) inhibitors, and immunomodulatory drugs are promising. Increasingly, evidence suggests immune evasion plays a role in PCNSL pathogenesis and several immunotherapeutic strategies including checkpoint inhibition and targeted chimeric antigen receptor T (CAR-T) cells are under investigation. This review provides a discussion on the challenges in development of targeted therapeutic strategies, an update on recent treatment advances, and offers a look toward ongoing clinical studies.
Collapse
|
86
|
Pang AP, Zhang F, Hu X, Luo Y, Wang H, Durrani S, Wu FG, Li BZ, Zhou Z, Lu Z, Lin F. Glutamine involvement in nitrogen regulation of cellulase production in fungi. BIOTECHNOLOGY FOR BIOFUELS 2021; 14:199. [PMID: 34645509 PMCID: PMC8513308 DOI: 10.1186/s13068-021-02046-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/23/2021] [Indexed: 05/27/2023]
Abstract
BACKGROUND Cellulase synthesized by fungi can environment-friendly and sustainably degrades cellulose to fermentable sugars for producing cellulosic biofuels, biobased medicine and fine chemicals. Great efforts have been made to study the regulation mechanism of cellulase biosynthesis in fungi with the focus on the carbon sources, while little attention has been paid to the impact and regulation mechanism of nitrogen sources on cellulase production. RESULTS Glutamine displayed the strongest inhibition effect on cellulase biosynthesis in Trichoderma reesei, followed by yeast extract, urea, tryptone, ammonium sulfate and L-glutamate. Cellulase production, cell growth and sporulation in T. reesei RUT-C30 grown on cellulose were all inhibited with the addition of glutamine (a preferred nitrogen source) with no change for mycelium morphology. This inhibition effect was attributed to both L-glutamine itself and the nitrogen excess induced by its presence. In agreement with the reduced cellulase production, the mRNA levels of 44 genes related to the cellulase production were decreased severely in the presence of glutamine. The transcriptional levels of genes involved in other nitrogen transport, ribosomal biogenesis and glutamine biosynthesis were decreased notably by glutamine, while the expression of genes relevant to glutamate biosynthesis, amino acid catabolism, and glutamine catabolism were increased noticeably. Moreover, the transcriptional level of cellulose signaling related proteins ooc1 and ooc2, and the cellular receptor of rapamycin trFKBP12 was increased remarkably, whose deletion exacerbated the cellulase depression influence of glutamine. CONCLUSION Glutamine may well be the metabolite effector in nitrogen repression of cellulase synthesis, like the role of glucose plays in carbon catabolite repression. Glutamine under excess nitrogen condition repressed cellulase biosynthesis significantly as well as cell growth and sporulation in T. reesei RUT-C30. More importantly, the presence of glutamine notably impacted the transport and metabolism of nitrogen. Genes ooc1, ooc2, and trFKBP12 are associated with the cellulase repression impact of glutamine. These findings advance our understanding of nitrogen regulation of cellulase production in filamentous fungi, which would aid in the rational design of strains and fermentation strategies for cellulase production in industry.
Collapse
Affiliation(s)
- Ai-Ping Pang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Funing Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Xin Hu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yongsheng Luo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Haiyan Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Samran Durrani
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Bing-Zhi Li
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Zhihua Zhou
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zuhong Lu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Fengming Lin
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
87
|
Liu D, Gu Y, Pang Q, Yu H, Zhang J. Dietary betaine regulates the synthesis of fatty acids through mTOR signaling in the muscle of zebrafish. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
88
|
Song G, Chen F, Chen S, Ye S. Polysaccharides from Premna microphylla turcz ameliorate inflammation via the enhancement of intestinal resistance in host. JOURNAL OF ETHNOPHARMACOLOGY 2021; 276:114208. [PMID: 34010697 DOI: 10.1016/j.jep.2021.114208] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Premna microphylla turcz is traditionally used as a folk remedy. Its roots, stems and leaves can be invoked as medicines, which have the functions of detoxification, swelling and hemostasis. It belongs to the Premna in the Verbenaceae and is mainly distributed in the mountains of southeastern China. However, there are few reports of in-depth studies on the anti-inflammatory effects of polysaccharide, which was the main component in Premna microphylla turcz. MATERIALS AND METHODS The flies were fed with standard corn flour-yeast medium to cause inflammation by sodium lauryl sulfate (SDS). The treatment group contained Premna microphylla turcz polysaccharide (pPMTLs) extract. The survival rate was obtained by feeding a vial containing five layers of filter paper, which was infiltrated with the 5% sucrose solution contaminated with SDS or SDS polysaccharide. The microvilli and nucleus of the midgut epithelial cells of different treatments were observed by transmission electron microscope, and the expression of inflammation-related genes was detected by real-time quantitative PCR (qRT-PCR). Finally, 16S rDNA analysis was conducted on the differences in the composition of the intestinal microbes of Drosophila. RESULTS In the current study, we showed that pPMTLs significantly prolonged the life span of SDS-inflamed flies from 5 days to 6 days. And pPMTLs reduced the rupture of microvilli in the midgut and restored the nuclear structure. In addition, pPMTLs significantly improved expression level of immune-related genes in Inflammation Drosophila especially the defensin (4.32 ± 0.75 vs 9.97 ± 0.52 SDS-polysaccharide group: SDS group, p < 0.001). The analysis of intestinal microbiota showed that pPMTLs decreased the relative abundance of Raoultella while Wolbachia increased (p < 0.05). CONCLUSIONS Collectively, our results revealed the potential application of pPMTLs in enhancing inflammation defense, which would be enormous significance for the inflammation-related disorders treatment.
Collapse
Affiliation(s)
- Guanglei Song
- School of Food Science and Biotechnology, Institute of Jinhua Food Industry, Zhejiang Gongshang University, 18 Xuezheng str., Hangzhou, Zhejiang, 310018, China.
| | - Fangyuan Chen
- School of Food Science and Biotechnology, Institute of Jinhua Food Industry, Zhejiang Gongshang University, 18 Xuezheng str., Hangzhou, Zhejiang, 310018, China.
| | - Shubo Chen
- School of Food Science and Biotechnology, Institute of Jinhua Food Industry, Zhejiang Gongshang University, 18 Xuezheng str., Hangzhou, Zhejiang, 310018, China.
| | - Shuhui Ye
- School of Food Science and Biotechnology, Institute of Jinhua Food Industry, Zhejiang Gongshang University, 18 Xuezheng str., Hangzhou, Zhejiang, 310018, China.
| |
Collapse
|
89
|
Zhao Q, Li Y, Du X, Chen X, Jiao Q, Jiang H. Effects of deubiquitylases on the biological behaviors of neural stem cells. Dev Neurobiol 2021; 81:847-858. [PMID: 34241974 DOI: 10.1002/dneu.22844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/03/2021] [Accepted: 05/16/2021] [Indexed: 11/11/2022]
Abstract
New neurons are generated throughout life in distinct regions of the mammalian brain due to the proliferation and differentiation of neural stem cells (NSCs). Ubiquitin, a post-translational modification of cellular proteins, is an important factor in regulating neurogenesis. Deubiquitination is a biochemical process that mediates the removal of ubiquitin moieties from ubiquitin-conjugated substrates. Recent studies have provided growing evidence that deubiquitylases (DUBs) which reverse ubiquitylation process play critical roles in NSCs maintenance, differentiation and maturation. This review mainly focused on the relationship of DUBs and NSCs, and further summarized recent advances in our understanding of DUBs on regulating NSCs biological behaviors.
Collapse
Affiliation(s)
- Qiqi Zhao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Yixin Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
90
|
Allosteric Modulation of GSK-3β as a New Therapeutic Approach in Limb Girdle Muscular Dystrophy R1 Calpain 3-Related. Int J Mol Sci 2021; 22:ijms22147367. [PMID: 34298987 PMCID: PMC8308041 DOI: 10.3390/ijms22147367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022] Open
Abstract
Limb-girdle muscular dystrophy R1 calpain 3-related (LGMDR1) is an autosomal recessive muscular dystrophy produced by mutations in the CAPN3 gene. It is a rare disease and there is no cure or treatment for the disease while the pathophysiological mechanism by which the absence of calpain 3 provokes the dystrophy in muscles is not clear. However, key proteins implicated in Wnt and mTOR signaling pathways, which regulate muscle homeostasis, showed a considerable reduction in their expression and in their phosphorylation in LGMDR1 patients' muscles. Finally, the administration of tideglusib and VP0.7, ATP non-competitive inhibitors of glycogen synthase kinase 3β (GSK-3β), restore the expression and phosphorylation of these proteins in LGMDR1 cells, opening the possibility of their use as therapeutic options.
Collapse
|
91
|
The effects of glucagon and the target of rapamycin (TOR) on skeletal muscle protein synthesis and age-dependent sarcopenia in humans. Clin Nutr ESPEN 2021; 44:15-25. [PMID: 34330459 DOI: 10.1016/j.clnesp.2021.06.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS Human target of rapamycin (TOR) is a kinase that stimulates protein synthesis in the skeletal muscle in response to amino acids and physical activity. METHODS A comprehensive literature search was conducted on the PubMed database from its inception up to May 2021 to retrieve information on the effects of TOR and glucagon on muscle function. Articles written in English regarding human subjects were included. RESULTS l-leucine activates TOR to initiate protein synthesis in the skeletal muscle. Glucagon has a crucial role suppressing skeletal muscle protein synthesis by increasing l-leucine oxidation and the irreversible loss of this amino acid. Glucagon-induced l-leucine oxidation suppresses TOR and attenuates the ability of skeletal muscle to synthesize proteins. Conditions associated with increased glucagon secretion typically feature reduced ability to synthesize proteins in the skeletal muscle that may evolve into sarcopenia. Animal protein ingestion, unlike vegetable protein, stimulates glucagon secretion. High intake of animal protein increases l-leucine oxidation and promotes the use of amino acids as fuel. Sarcopenia and arterial stiffness characteristically occur together in conditions featuring insulin resistance, such as aging. Insulin resistance mediates the relationship between aging and sarcopenia and arterial stiffness. The loss of skeletal muscle fibers that characterizes sarcopenia is followed by collagen and lipid accumulation. Likewise, insulin resistance is associated with arterial stiffness and intima-media thickening due to adaptive accretion of collagen and lipids in the arterial wall. CONCLUSIONS Human TOR participates in the pathogenesis of sarcopenia and arterial stiffness, although its effects remain to be fully elucidated.
Collapse
|
92
|
Kidney intercalated cells and the transcription factor FOXi1 drive cystogenesis in tuberous sclerosis complex. Proc Natl Acad Sci U S A 2021; 118:2020190118. [PMID: 33536341 PMCID: PMC8017711 DOI: 10.1073/pnas.2020190118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is caused by mutations in TSC1 or TSC2 gene and affects multiple organs, including the kidney, where it presents with angiomyolipomata and cysts that can result in kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Current studies demonstrate that kidney cyst epithelia in TSC mouse models and in humans with TSC are composed of hyperproliferating intercalated cells, along with activation of H+-ATPase and carbonic anhydrase 2. Interfering with intercalated cell proliferation completely inhibited and inactivating carbonic anhydrase 2 significantly protected against cyst formation in TSC. Targeting the acid base and/or electrolyte transporters of intercalated cells may provide a therapeutic approach for the treatment of kidney cysts in TSC. Tuberous sclerosis complex (TSC) is caused by mutations in either TSC1 or TSC2 genes and affects multiple organs, including kidney, lung, and brain. In the kidney, TSC presents with the enlargement of benign tumors (angiomyolipomata) and cysts, which eventually leads to kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Here, we report that mice with principal cell-specific inactivation of Tsc1 develop numerous cortical cysts, which are overwhelmingly composed of hyperproliferating A-intercalated (A-IC) cells. RNA sequencing and confirmatory expression studies demonstrated robust expression of Forkhead Transcription Factor 1 (Foxi1) and its downstream targets, apical H+-ATPase and cytoplasmic carbonic anhydrase 2 (CAII), in cyst epithelia in Tsc1 knockout (KO) mice but not in Pkd1 mutant mice. In addition, the electrogenic 2Cl−/H+ exchanger (CLC-5) is significantly up-regulated and shows remarkable colocalization with H+-ATPase on the apical membrane of cyst epithelia in Tsc1 KO mice. Deletion of Foxi1, which is vital to intercalated cells viability and H+-ATPase expression, completely abrogated the cyst burden in Tsc1 KO mice, as indicated by MRI images and histological analysis in kidneys of Foxi1/Tsc1 double-knockout (dKO) mice. Deletion of CAII, which is critical to H+-ATPase activation, caused significant reduction in cyst burden and increased life expectancy in CAII/Tsc1 dKO mice vs. Tsc1 KO mice. We propose that intercalated cells and their acid/base/electrolyte transport machinery (H+-ATPase/CAII/CLC-5) are critical to cystogenesis, and their inhibition or inactivation is associated with significant protection against cyst generation and/or enlargement in TSC.
Collapse
|
93
|
Current Understandings of Core Pathways for the Activation of Mammalian Primordial Follicles. Cells 2021; 10:cells10061491. [PMID: 34199299 PMCID: PMC8231864 DOI: 10.3390/cells10061491] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
The mammalian ovary has two main functions-producing mature oocytes for fertilization and secreting hormones for maintaining the ovarian endocrine functions. Both functions are vital for female reproduction. Primordial follicles are composed of flattened pre-granulosa cells and a primary oocyte, and activation of primordial follicles is the first step in follicular development and is the key factor in determining the reproductive capacity of females. The recent identification of the phosphatidylinositol 3 kinase (PI3K)/phosphatase and tensin homolog deleted on chromosome 10 (PTEN) signaling pathway as the key controller for follicular activation has made the study of primordial follicle activation a hot research topic in the field of reproduction. This review systematically summarizes the roles of the PI3K/PTEN signaling pathway in primordial follicle activation and discusses how the pathway interacts with various other molecular networks to control follicular activation. Studies on the activation of primordial follicles have led to the development of methods for the in vitro activation of primordial follicles as a treatment for infertility in women with premature ovarian insufficiency or poor ovarian response, and these are also discussed along with some practical applications of our current knowledge of follicular activation.
Collapse
|
94
|
Luo Z, Zeng A, Chen Y, He S, He S, Jin X, Li C, Mei W, Lu Q. Ligustilide inhibited Angiotensin II induced A7r5 cell autophagy via Akt/mTOR signaling pathway. Eur J Pharmacol 2021; 905:174184. [PMID: 34004211 DOI: 10.1016/j.ejphar.2021.174184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022]
Abstract
Autophagy is essential to vessel homeostasis and function in the cardiovascular system. Ligustilide (LIG) is one of the main active ingredients extracted from traditional Chinese medicines, such as Ligusticum chuanxiong, Angelica, and other umbelliferous plants, and reported to have cardiovascular protective effects. In this study, we explore the effects and the potential mechanism of ligustilide on the Ang II-induced autophagy in A7r5 cells. Our results showed that ligustilide inhibited the Ang II-induced autophagy in A7r5 cells and down regulated the expression of autophagy-related proteins LC3, ULK1, and Beclin-1. Ligustilide exerted a protective effect on the reduction of the concentrations of reactive oxygen species and Ca2+ and upregulated the nitric oxide concentration in A7r5 cells with Ang II-induced autophagy. Additionally, the analyses of network pharmacological targets and potential signal pathways indicated that the target of ligustilide to regulate autophagy was related to the Akt/mTOR signaling pathway. Furthermore, ligustilide could upregulate the expression of p-Akt and p-mTOR and inhibit the expression of LC3II in A7r5 cells with Ang II-induced autophagy. These findings showed that ligustilide inhibited the autophagic flux in A7r5 cells induced by Ang II via the activation of the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Zhenhui Luo
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ao Zeng
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuankun Chen
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shumiao He
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Siqing He
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaobao Jin
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chunmei Li
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenjie Mei
- Guangdong Province Engineering and Technology Center for Molecular Probe and Bio-medicine Imaging, Guangzhou, China
| | - Qun Lu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Province Engineering and Technology Center for Molecular Probe and Bio-medicine Imaging, Guangzhou, China.
| |
Collapse
|
95
|
Wang W, Zhou Y, Cai Y, Wang S, Shao F, Du J, Fang J, Liu J, Shao X, Liu B, Fang J, Liang Y. Phosphoproteomic Profiling of Rat's Dorsal Root Ganglia Reveals mTOR as a Potential Target in Bone Cancer Pain and Electro-Acupuncture's Analgesia. Front Pharmacol 2021; 12:593043. [PMID: 33995007 PMCID: PMC8117331 DOI: 10.3389/fphar.2021.593043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 04/08/2021] [Indexed: 01/18/2023] Open
Abstract
Bone cancer pain (BCP) is a clinical refractory mixed pain involving neuropathic and inflammatory pain, with the underlying mechanisms remaining largely unknown. Electro-acupuncture (EA) can partly alleviate BCP according to previous research. We aim to explore the proteins and major pathways involved in BCP and EA treatment through phosphoproteomic profiling. BCP rat model was built by tibial inoculation of MRMT-1 mammary gland carcinoma cells. Mechanical hyperalgesia determined by paw withdrawal thresholds (PWTs) and bone destruction manifested on the radiographs confirmed the success of modeling, which were attenuated by EA treatment. The differentially expressed phosphorylated proteins (DEPs) co-regulated by BCP modeling and EA treatment in rat dorsal root ganglions (DRGs) were analyzed through PEX100 Protein microarray. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that DEPs were significantly enriched in mammalian target of rapamycin (mTOR) signaling pathway. The phosphorylations of mTOR at Ser2448 and Thr2446 were increased in BCP and downregulated by EA. In addition, the phosphorylation of S6K and Akt, markers of the mTOR complex, were also increased in BCP and downregulated by EA. Inhibition of mTOR signaling alleviated the PWTs of BCP rats, while the mTOR agonist impaired the analgesic effect of EA. Thus, our study provided a landscape of protein phosphorylation changes in DRGs of EA-treated BCP rats and revealed that mTOR signaling can be potentially targeted to alleviate BCP by EA treatment.
Collapse
Affiliation(s)
- Wen Wang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.,Quzhou Municipal Hospital of Traditional Chinese Medicine, Quzhou, China
| | - You Zhou
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yangqian Cai
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Sisi Wang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangbing Shao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Junying Du
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Junfan Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinggen Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaomei Shao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Boyi Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Liang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
96
|
Jeon HB, Roh H, Ahn HM, Lee JH, Yun CO, Roh TS, Lee WJ. Metformin Inhibits Transforming Growth Factor β-Induced Fibrogenic Response of Human Dermal Fibroblasts and Suppresses Fibrosis in Keloid Spheroids. Ann Plast Surg 2021; 86:406-411. [PMID: 33141769 DOI: 10.1097/sap.0000000000002574] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Accumulation of excessive extracellular matrix (ECM) and aberrant transforming growth factor β (TGF-β) signaling pathway function can be potential therapeutic targets for keloid treatment. In this study, we examined the antifibrotic effect of metformin as a suppressor of TGF-β signaling pathways in human dermal fibroblasts (HDFs) and keloid spheroids. Human dermal fibroblasts were stimulated with TGF-β (10 ng/mL) and treated with metformin (10 mM). The mRNA and protein expression of ECM components were evaluated by quantitative polymerase chain reaction, western blot, and immunofluorescence assay. In addition, we immunohistochemically examined the expression levels of ECM proteins in keloid spheroids. After addition of metformin (10 mM), collagen types I and III and elastin mRNA levels were significantly decreased in HDFs, and collagen type I protein level was significantly decreased. In addition, the expression levels of collagen types I and III, fibronectin, and elastin were significantly reduced in keloid spheroids after treatment with metformin (100 mM). Collagen types I and III and p-Smad2/3 complex proteins were decreased in metformin-treated keloid spheroids. These findings indicated that metformin inhibits the expression of ECM components in TGF-β-stimulated HDFs and keloid spheroids. Therefore, we suggest the potential of metformin as an effective agent for the treatment of keloids.
Collapse
Affiliation(s)
- Hong Bae Jeon
- From the Department of Medicine, Yonsei University Graduate School
| | - Hyun Roh
- Institute for Human Tissue Restoration and Department of Plastic & Reconstructive Surgery, Severance Hospital, Yonsei University College of Medicine
| | - Hyo Min Ahn
- Department of Bioengineering, College of Engineering, Hanyang University
| | - Ju Hee Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Chae Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University
| | - Tai Suk Roh
- Institute for Human Tissue Restoration and Department of Plastic & Reconstructive Surgery, Severance Hospital, Yonsei University College of Medicine
| | - Won Jai Lee
- Institute for Human Tissue Restoration and Department of Plastic & Reconstructive Surgery, Severance Hospital, Yonsei University College of Medicine
| |
Collapse
|
97
|
Williams MC, Patel JH, Kakebeen AD, Wills AE. Nutrient availability contributes to a graded refractory period for regeneration in Xenopus tropicalis. Dev Biol 2021; 473:59-70. [PMID: 33484704 DOI: 10.1016/j.ydbio.2021.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/17/2020] [Accepted: 01/12/2021] [Indexed: 11/28/2022]
Abstract
Xenopus tadpoles are a unique model for regeneration in that they exhibit two distinct phases of age-specific regenerative competence. In Xenopus laevis, young tadpoles fully regenerate following major injuries such as tail transection, then transiently lose regenerative competence during the "refractory period" from stages 45-47. Regenerative competence is then regained in older tadpoles before being permanently lost during metamorphosis. Here we show that a similar refractory period exists in X. tropicalis. Notably, tadpoles lose regenerative competence gradually in X. tropicalis, with full regenerative competence lost at stage 47. We find that the refractory period coincides closely with depletion of maternal yolk stores and the onset of independent feeding, and so we hypothesized that it might be caused in part by nutrient stress. In support of this hypothesis, we find that cell proliferation declines throughout the tail as the refractory period approaches. When we block nutrient mobilization by inhibiting mTOR signaling, we find that tadpole growth and regeneration are reduced, while yolk stores persist. Finally, we are able to restore regenerative competence and cell proliferation during the refractory period by abundantly feeding tadpoles. Our study argues that nutrient stress contributes to lack of regenerative competence and introduces the X. tropicalis refractory period as a valuable new model for interrogating how metabolic constraints inform regeneration.
Collapse
Affiliation(s)
| | - Jeet H Patel
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Program in Molecular and Cellular Biology, University of Washington School of Medicine, Seattle, WA, USA
| | - Anneke D Kakebeen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Andrea E Wills
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
98
|
Nishioka S, Ishimura T, Endo T, Yokoyama N, Ogawa S, Fujisawa M. Suppression of Allograft Fibrosis by Regulation of Mammalian Target of Rapamycin-Related Protein Expression in Kidney-Transplanted Recipients Treated with Everolimus and Reduced Tacrolimus. Ann Transplant 2021; 26:e926476. [PMID: 33431785 PMCID: PMC7812696 DOI: 10.12659/aot.926476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Although renoprotective effects of everolimus (EVR) in kidney transplantation (KTx) have been widely reported, its pathophysiological mechanism remains unclear. Material/Methods We compared changes in eGFR (ΔGFR, ml/min/1.73 m2) and the ratio of the fibrotic area in biopsy specimens (ΔFI,%) from 3 months to 3 years after KTx between the EVR+ group (EVR addition and Tac reduction early after KTx, n=32), and the EVR− group (normal Tac without EVR, n=28). We also immunohistochemically evaluated mTOR-related protein expression. Results ΔGFR and ΔFI in the EVR+ vs. EVR− groups were −0.27±6.8 vs. −9.8±12.8 (p<0.001) and 2.4±4.9 vs. 9.5±10.5 (p<0.001), respectively. Phosphorylated mTOR and phosphorylated 4EBP1 expression at 3 years in the EVR+ group was significantly lower than that in the EVR− group. Moreover, in the subgroup analysis comparing ΔGFR and ΔFI among groups stratified by immunosuppressive regimen and mTOR signal enhancement, the ΔFI in patients with EVR+ with decreased mTOR signal enhancement was significantly milder than that in other patients. In addition, in the multivariate analysis, EVR addition was the only independent predictor for allograft fibrosis, whereas the Tac C0 concentration at neither 1 nor 3 years proved to be a risk factor. Conclusions These results suggested that EVR addition and Tac reduction may attenuate kidney allograft fibrosis, and that the suppression of mTOR signaling process may be involved in the anti-fibrotic effect of this immunosuppressive regimen. These results provide suggestions of how to utilize EVR for patients with KTx and improve graft function.
Collapse
Affiliation(s)
- Shun Nishioka
- Division of Urology, Department of Surgery, Faculty of Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Takeshi Ishimura
- Division of Urology, Department of Surgery, Faculty of Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Takahito Endo
- Division of Urology, Department of Surgery, Faculty of Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Naoki Yokoyama
- Division of Urology, Department of Surgery, Faculty of Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Satoshi Ogawa
- Division of Urology, Department of Surgery, Faculty of Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Masato Fujisawa
- Division of Urology, Department of Surgery, Faculty of Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| |
Collapse
|
99
|
Chen K, Zhang Z, Li J, Xie S, Shi LJ, He YH, Liang XF, Zhu QS, He S. Different regulation of branched-chain amino acid on food intake by TOR signaling in Chinese perch (Siniperca chuatsi). AQUACULTURE 2021; 530:735792. [DOI: 10.1016/j.aquaculture.2020.735792] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
100
|
Lodjak J, Verhulst S. Insulin-like growth factor 1 of wild vertebrates in a life-history context. Mol Cell Endocrinol 2020; 518:110978. [PMID: 32798584 DOI: 10.1016/j.mce.2020.110978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022]
Abstract
Broad variation in intra- and interspecific life-history traits is largely shaped by resource limitation and the ensuing allocation trade-offs that animals are forced to make. Insulin-like growth factor 1 (IGF-1), a growth-hormone-dependent peptide, may be a key player in the regulation of allocation processes. In laboratory animals, the effects of IGF-1 on growth- and development (positive), reproduction (positive), and longevity (negative) are well established. We here review the evidence on these effects in wild vertebrates, where animals are more likely to face resource limitation and other challenges. We point out the similarities and dissimilarities in patterns of IGF-1 functions obtained in these two different study settings and discuss the knowledge we need to develop a comprehensive picture of the role of IGF-1 in mediating life-history variation of wild vertebrates.
Collapse
Affiliation(s)
- Jaanis Lodjak
- Department of Zoology, Institute of Ecology and Earth Sciences, University of Tartu, 46 Vanemuise Street, Tartu, 51014, Estonia; Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands.
| | - Simon Verhulst
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands
| |
Collapse
|