251
|
Abstract
INTRODUCTION The p90 ribosomal S6 kinases (RSK) are a family of Ser/Thr protein kinases that are downstream effectors of MEK1/2-ERK1/2. Increased RSK activation is implicated in the etiology of multiple pathologies, including numerous types of cancers, cardiovascular disease, liver and lung fibrosis, and infections. AREAS COVERED The review summarizes the patent and scientific literature on small molecule modulators of RSK and their potential use as therapeutics. The patents were identified using World Intellectual Property Organization and United States Patent and Trademark Office databases. The compounds described are predominantly RSK inhibitors, but a RSK activator is also described. The majority of the inhibitors are not RSK-specific. EXPERT OPINION Based on the overwhelming evidence that RSK is involved in a number of diseases that have high mortalities it seems surprising that there are no RSK modulators that have pharmacokinetic properties suitable for in vivo use. MEK1/2 inhibitors are in the clinic, but the efficacy of these compounds appears to be limited by their side effects. We hypothesize that targeting the downstream effectors of MEK1/2, like RSK, are an untapped source of drug targets and that they will generate less side effects than MEK1/2 inhibitors because they regulate fewer effectors.
Collapse
Affiliation(s)
- Katarzyna A Ludwik
- a Department of Pathology, Microbiology & Immunology , Vanderbilt University , Nashville , TN , USA
| | - Deborah A Lannigan
- a Department of Pathology, Microbiology & Immunology , Vanderbilt University , Nashville , TN , USA.,b Department of Cancer Biology , Vanderbilt University , Nashville , TN , USA
| |
Collapse
|
252
|
Hsiao YH, Huang YT, Hung CY, Kuo TC, Luo FJ, Yuan TC. PYK2 via S6K1 regulates the function of androgen receptors and the growth of prostate cancer cells. Endocr Relat Cancer 2016; 23:651-63. [PMID: 27492635 DOI: 10.1530/erc-16-0122] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 06/30/2016] [Indexed: 12/30/2022]
Abstract
Androgen receptor (AR) is a steroid hormone receptor that functions as a transcription factor for regulating cell growth and survival. Aberrant AR function becomes a risk factor for promoting the progression of prostate cancer (PCa). In this study, we examined the roles of proline-rich tyrosine kinase 2 (PYK2) and ribosomal S6 kinase 1 (S6K1) in regulating AR expression and activity and growth properties in PCa cells. Compared with normal prostate tissues, PCa tumors exhibited high levels of PYK2 and S6K1 expression. Furthermore, the expression levels of PYK2 and S6K1 were significantly correlated with nuclear AR expression in PCa tissues. We further found the association between PYK2, S6K1, and AR in their protein expression and phosphorylation levels among normal prostate PZ-HPV-7 cells and prostate cancer LNCaP and 22Rv1 cells. Overexpression of the wild-type PYK2 in PZ-HPV-7 and LNCaP cells promoted AR and S6K1 expression and phosphorylation as well as enhanced cell growth. In contrast, expression of the mutated PYK2 or knockdown of PYK2 expression in LNCaP or 22Rv1 cells caused reduced expression or phosphorylation of AR and S6K1 as well as retarded cell growth. Under an androgen-deprived condition, PYK2-promoted AR expression and phosphorylation and PSA production in LNCaP cells can be abolished by knocking down S6K1 expression. In summary, our data suggested that PYK2 via S6K1 activation modulated AR function and growth properties in PCa cells. Thus, PYK2 and S6K1 may potentially serve as therapeutic targets for PCa treatment.
Collapse
Affiliation(s)
- Yu-Hsuan Hsiao
- Department of Life ScienceNational Dong Hwa University, Hualien, Taiwan, Republic of China
| | - Yu-Ting Huang
- Department of Life ScienceNational Dong Hwa University, Hualien, Taiwan, Republic of China
| | - Chia-Yu Hung
- Department of Life ScienceNational Dong Hwa University, Hualien, Taiwan, Republic of China
| | - Tzu-Chien Kuo
- Department of Life ScienceNational Dong Hwa University, Hualien, Taiwan, Republic of China
| | - Fuh-Jinn Luo
- Department of PathologyMennonite Hospital, Hualien, Taiwan, Republic of China
| | - Ta-Chun Yuan
- Department of Life ScienceNational Dong Hwa University, Hualien, Taiwan, Republic of China
| |
Collapse
|
253
|
Cell-cycle involvement in autophagy and apoptosis in yeast. Mech Ageing Dev 2016; 161:211-224. [PMID: 27450768 DOI: 10.1016/j.mad.2016.07.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/16/2016] [Accepted: 07/17/2016] [Indexed: 12/14/2022]
Abstract
Regulation of the cell cycle and apoptosis are two eukaryotic processes required to ensure maintenance of genomic integrity, especially in response to DNA damage. The ease with which yeast, amongst other eukaryotes, can switch from cellular proliferation to cell death may be the result of a common set of biochemical factors which play dual roles depending on the cell's physiological state. A wide variety of homologues are shared between different yeasts and metazoans and this conservation confirms their importance. This review gives an overview of key molecular players involved in yeast cell-cycle regulation, and those involved in mechanisms which are induced by cell-cycle dysregulation. One such mechanism is autophagy which, depending on the severity and type of DNA damage, may either contribute to the cell's survival or death. Cell-cycle dysregulation due to checkpoint deficiency leads to mitotic catastrophe which in turn leads to programmed cell death. Molecular players implicated in the yeast apoptotic pathway were shown to play important roles in the cell cycle. These include the metacaspase Yca1p, the caspase-like protein Esp1p, the cohesin subunit Mcd1p, as well as the inhibitor of apoptosis protein Bir1p. The roles of these molecular players are discussed.
Collapse
|
254
|
Diamanti S, Nikitakis N, Rassidakis G, Doulis I, Sklavounou A. Immunohistochemical evaluation of the mTOR pathway in intra-oral minor salivary gland neoplasms. Oral Dis 2016; 22:620-9. [PMID: 27177463 DOI: 10.1111/odi.12504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 04/16/2016] [Accepted: 05/04/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The aim of this study was to investigate the expression of upstream and downstream molecules of the oncogenic mTOR signaling pathway in intra-oral minor salivary gland tumors (SGTs). MATERIALS AND METHODS Tissue samples consisted of 39 malignant and 13 benign minor SGTs, and 8 controls of normal minor salivary glands (NMSG). An immunohistochemical analysis for phosphorylated Akt, 4EBP1 and S6 (total and phosphorylated), and eIF4E was performed. RESULTS Expression of pAkt and 4EBP1 was observed in all SGTs and in most NMSG. p4EBP1 was detected in almost all SGT cases, NMSG being negative. S6 immunoreactivity was observed in 37.5% of NMSG, 92.3% of benign and 100% of malignant SGTs, while pS6 expression was observed in 77% of benign and 95% of malignant SGTs, but not in NMSG. Finally, eIF4E was expressed in 12.5% of NMSG, 69.2% of benign, and 76.9% of malignant tumors. All molecules studied had statistically significantly lower expression in NMSG compared with SGTs. Moreover, malignant neoplasms received higher scores compared with benign tumors for all molecules with the exception of eIF4E. CONCLUSION The mTOR signaling pathway is activated in SGTs, especially in malignancies. Therefore, the possible therapeutic role of targeting the mTOR pathway by rapamycin analogs in SGTs needs further investigation.
Collapse
Affiliation(s)
- S Diamanti
- Department of Oral Medicine and Pathology, Dental School, University of Athens, Athens, Greece. , .,Oral Medicine Department, 251 General Air Force and VA Hospital, Athens, Greece. ,
| | - N Nikitakis
- Department of Oral Medicine and Pathology, Dental School, University of Athens, Athens, Greece
| | - G Rassidakis
- Department of Pathology, Medical School, University of Athens, Athens, Greece.,Department of Pathology and Cytology, Carolinska University Hospital and Karolinska Institute, Solna, Sweden
| | - I Doulis
- Oral Medicine Department, 251 General Air Force and VA Hospital, Athens, Greece
| | - A Sklavounou
- Department of Oral Medicine and Pathology, Dental School, University of Athens, Athens, Greece
| |
Collapse
|
255
|
Alteration of protein prenylation promotes spermatogonial differentiation and exhausts spermatogonial stem cells in newborn mice. Sci Rep 2016; 6:28917. [PMID: 27374985 PMCID: PMC4931501 DOI: 10.1038/srep28917] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/10/2016] [Indexed: 12/12/2022] Open
Abstract
Spermatogenesis in adulthood depends on the successful neonatal establishment of the spermatogonial stem cell (SSC) pool and gradual differentiation during puberty. The stage-dependent changes in protein prenylation in the seminiferous epithelium might be important during the first round of spermatogenesis before sexual maturation, but the mechanisms are unclear. We have previous found that altered prenylation in Sertoli cells induced spermatogonial apoptosis in the neonatal testis, resulting in adult infertility. Now we further explored the role of protein prenylation in germ cells, using a conditional deletion of geranylgeranyl diphosphate synthase (Ggpps) in embryonic stage and postmeiotic stage respectively. We observed infertility of Ggpps(-/-) Ddx4-Cre mice that displayed a Sertoli-cell-only syndrome phenotype, which resulted from abnormal spermatogonial differentiation and SSC depletion during the prepubertal stage. Analysis of morphological characteristics and cell-specific markers revealed that spermatogonial differentiation was enhanced from as early as the 7(th) postnatal day in the first round of spermatogenesis. Studies of the molecular mechanisms indicated that Ggpps deletion enhanced Rheb farnesylation, which subsequently activated mTORC1 and facilitated spermatogonial differentiation. In conclusion, the prenylation balance in germ cells is crucial for spermatogonial differentiation fate decision during the prepubertal stage, and the disruption of this process results in primary infertility.
Collapse
|
256
|
Xu J, Li Z, Su Q, Zhao J, Ma J. Embryonic develop-associated gene 1 is overexpressed and acts as a tumor promoter in thyroid carcinoma. Biomed Pharmacother 2016; 81:86-92. [DOI: 10.1016/j.biopha.2016.03.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 11/15/2022] Open
|
257
|
Oleksiewicz MB, Thorup I, Nielsen HS, Andersen HV, Hegelund AC, Iversen L, Guldberg TS, Brinck PR, Sjogren I, Thinggaard UK, Jørgensen L, Jensen MB. Generalized Cellular Hypertrophy is Induced by a Dual-Acting PPAR Agonist in Rat Urinary Bladder Urothelium In Vivo. Toxicol Pathol 2016; 33:552-60. [PMID: 16076770 DOI: 10.1080/01926230500214657] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Some developmental dual-acting PPARα/γ agonists, such as ragaglitazar, have shown carcinogenic effects in the rodent urinary bladder urothelium after months-years of dosing. We examined early (precancerous) changes in the bladder urothelium of rats orally dosed with ragaglitazar, using a newly developed flow cytometric method. Following 3 weeks of oral ragaglitazar dosing, increases in physical size occurred in a generalized fashion in rat bladder urothelial cells, determined by flow cytometry. Protein/DNA measurements confirmed increased protein content of urothelial cells in the bladder, and hypertrophy was observed in the kidney pelvis urothelium by histopathology. In animals exhibiting urothelial hypertrophy, no cell cycle changes were detected in parallel samples of bladder urothelium. Interestingly, urothelial cells from normal rats were found to constitute a unique type of noncycling population, with high G2/M fractions. In summary, our findings showed that in the urothelium of ragaglitazar-treated animals, hypertrophy (increased size and protein content per cell) was an early change, that affected the whole bladder urothelial cell population. The urothelial hypertrophy was primary, i.e., occurred in the absence of similarly pronounced changes in cell cycle distributions. To our knowledge, this is the first report of a direct hypertrophic effect of a PPAR agonist. Urothelial hypertrophy might be a relevant early biological endpoint in mechanistic studies regarding the bladder-carcinogenic effect of PPAR agonists.
Collapse
Affiliation(s)
- Martin B Oleksiewicz
- Novo Nordisk A/S, Novo Nordisk Park, Department of Virology and Molecular Toxicology, Maalov, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
258
|
van Geldermalsen M, Wang Q, Nagarajah R, Marshall AD, Thoeng A, Gao D, Ritchie W, Feng Y, Bailey CG, Deng N, Harvey K, Beith JM, Selinger CI, O'Toole SA, Rasko JEJ, Holst J. ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer. Oncogene 2016; 35:3201-8. [PMID: 26455325 PMCID: PMC4914826 DOI: 10.1038/onc.2015.381] [Citation(s) in RCA: 420] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 09/01/2015] [Accepted: 09/04/2015] [Indexed: 12/31/2022]
Abstract
Alanine, serine, cysteine-preferring transporter 2 (ASCT2; SLC1A5) mediates uptake of glutamine, a conditionally essential amino acid in rapidly proliferating tumour cells. Uptake of glutamine and subsequent glutaminolysis is critical for activation of the mTORC1 nutrient-sensing pathway, which regulates cell growth and protein translation in cancer cells. This is of particular interest in breast cancer, as glutamine dependence is increased in high-risk breast cancer subtypes. Pharmacological inhibitors of ASCT2-mediated transport significantly reduced glutamine uptake in human breast cancer cell lines, leading to the suppression of mTORC1 signalling, cell growth and cell cycle progression. Notably, these effects were subtype-dependent, with ASCT2 transport critical only for triple-negative (TN) basal-like breast cancer cell growth compared with minimal effects in luminal breast cancer cells. Both stable and inducible shRNA-mediated ASCT2 knockdown confirmed that inhibiting ASCT2 function was sufficient to prevent cellular proliferation and induce rapid cell death in TN basal-like breast cancer cells, but not in luminal cells. Using a bioluminescent orthotopic xenograft mouse model, ASCT2 expression was then shown to be necessary for both successful engraftment and growth of HCC1806 TN breast cancer cells in vivo. Lower tumoral expression of ASCT2 conferred a significant survival advantage in xenografted mice. These responses remained intact in primary breast cancers, where gene expression analysis showed high expression of ASCT2 and glutamine metabolism-related genes, including GLUL and GLS, in a cohort of 90 TN breast cancer patients, as well as correlations with the transcriptional regulators, MYC and ATF4. This study provides preclinical evidence for the feasibility of novel therapies exploiting ASCT2 transporter activity in breast cancer, particularly in the high-risk basal-like subgroup of TN breast cancer where there is not only high expression of ASCT2, but also a marked reliance on its activity for sustained cellular proliferation.
Collapse
Affiliation(s)
- M van Geldermalsen
- Origins of Cancer Program, Centenary Institute, Camperdown, New South Wales, Australia
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Q Wang
- Origins of Cancer Program, Centenary Institute, Camperdown, New South Wales, Australia
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - R Nagarajah
- Origins of Cancer Program, Centenary Institute, Camperdown, New South Wales, Australia
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - A D Marshall
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - A Thoeng
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - D Gao
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Bioinformatics Laboratory, Centenary Institute, Camperdown, New South Wales, Australia
| | - W Ritchie
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Bioinformatics Laboratory, Centenary Institute, Camperdown, New South Wales, Australia
| | - Y Feng
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - C G Bailey
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - N Deng
- The Kinghorn Cancer Centre and Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - K Harvey
- The Kinghorn Cancer Centre and Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - J M Beith
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - C I Selinger
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - S A O'Toole
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- The Kinghorn Cancer Centre and Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - J E J Rasko
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - J Holst
- Origins of Cancer Program, Centenary Institute, Camperdown, New South Wales, Australia
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Associate, Origins of Cancer Program, Centenary Institute, Locked Bag 6, Newtown, New South Wales 2042, Australia. E-mail:
| |
Collapse
|
259
|
Khaliullina H, Love NK, Harris WA. Nutrient-Deprived Retinal Progenitors Proliferate in Response to Hypoxia: Interaction of the HIF-1 and mTOR Pathway. J Dev Biol 2016; 4. [PMID: 27280081 PMCID: PMC4894462 DOI: 10.3390/jdb4020017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
At a cellular level, nutrients are sensed by the mechanistic Target of Rapamycin (mTOR). The response of cells to hypoxia is regulated via action of the oxygen sensor Hypoxia-Inducible Factor 1 (HIF-1). During development, injury and disease, tissues might face conditions of both low nutrient supply and low oxygen, yet it is not clear how cells adapt to both nutrient restriction and hypoxia, or how mTOR and HIF-1 interact in such conditions. Here we explore this question in vivo with respect to cell proliferation using the ciliary marginal zone (CMZ) of Xenopus. We found that both nutrient-deprivation and hypoxia cause retinal progenitors to decrease their proliferation, yet when nutrient-deprived progenitors are exposed to hypoxia there is an unexpected rise in cell proliferation. This increase, mediated by HIF-1 signalling, is dependent on glutaminolysis and reactivation of the mTOR pathway. We discuss how these findings in non-transformed tissue may also shed light on the ability of cancer cells in poorly vascularised solid tumours to proliferate.
Collapse
Affiliation(s)
- Helena Khaliullina
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK;
| | - Nicola K Love
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK;
| | - William A Harris
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK;
| |
Collapse
|
260
|
Jujuboside A Protects H9C2 Cells from Isoproterenol-Induced Injury via Activating PI3K/Akt/mTOR Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:9593716. [PMID: 27293469 PMCID: PMC4884826 DOI: 10.1155/2016/9593716] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/26/2016] [Indexed: 11/17/2022]
Abstract
Jujuboside A is a kind of the saponins isolated from the seeds of Ziziphus jujuba, which possesses multiple biological effects, such as antianxiety, antioxidant, and anti-inflammatory effects; however, its mediatory effect on isoproterenol-stimulated cardiomyocytes has not been investigated yet. In this study, we tried to detect the protective effect and potential mechanism of JUA on ISO-induced cardiomyocytes injury. H9C2 cells were treated with ISO to induce cell damage. Cells were pretreated with JUA to investigate the effects on the cell viability, morphological changes, light chain 3 conversion, and the activation of PI3K/Akt/mTOR signaling pathway. Results showed that ISO significantly inhibited the cell viability in a time- and dose-dependent manner. JUA pretreatment could reverse the reduction of cell viability and better the injury of H9C2 cells induced by ISO. Western blot analysis showed that JUA could accelerate the phosphorylation of PI3K, Akt, and mTOR. Results also indicated that JUA could significantly decrease the ratio of microtubule-associated protein LC3-II/I in H9C2 cells. Taken together, our research showed that JUA could notably reduce the damage cause by ISO via promoting the phosphorylation of PI3K, Akt, and mTOR and inhibiting LC3 conversion, which may be a potential choice for the treatment of heart diseases.
Collapse
|
261
|
Erythropoietin Pathway: A Potential Target for the Treatment of Depression. Int J Mol Sci 2016; 17:ijms17050677. [PMID: 27164096 PMCID: PMC4881503 DOI: 10.3390/ijms17050677] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 04/05/2016] [Accepted: 04/27/2016] [Indexed: 12/21/2022] Open
Abstract
During the past decade, accumulating evidence from both clinical and experimental studies has indicated that erythropoietin may have antidepressant effects. In addition to the kidney and liver, many organs have been identified as secretory tissues for erythropoietin, including the brain. Its receptor is expressed in cerebral and spinal cord neurons, the hypothalamus, hippocampus, neocortex, dorsal root ganglia, nerve axons, and Schwann cells. These findings may highlight new functions for erythropoietin, which was originally considered to play a crucial role in the progress of erythroid differentiation. Erythropoietin and its receptor signaling through JAK2 activate multiple downstream signaling pathways including STAT5, PI3K/Akt, NF-κB, and MAPK. These factors may play an important role in inflammation and neuroprogression in the nervous system. This is particularly true for the hippocampus, which is possibly related to learning, memory, neurocognitive deficits and mood alterations. Thus, the influence of erythropoietin on the downstream pathways known to be involved in the treatment of depression makes the erythropoietin-related pathway an attractive target for the development of new therapeutic approaches. Focusing on erythropoietin may help us understand the pathogenic mechanisms of depression and the molecular basis of its treatment.
Collapse
|
262
|
Ruggenenti P, Gentile G, Perico N, Perna A, Barcella L, Trillini M, Cortinovis M, Ferrer Siles CP, Reyes Loaeza JA, Aparicio MC, Fasolini G, Gaspari F, Martinetti D, Carrara F, Rubis N, Prandini S, Caroli A, Sharma K, Antiga L, Remuzzi A, Remuzzi G. Effect of Sirolimus on Disease Progression in Patients with Autosomal Dominant Polycystic Kidney Disease and CKD Stages 3b-4. Clin J Am Soc Nephrol 2016; 11:785-794. [PMID: 26912555 PMCID: PMC4858487 DOI: 10.2215/cjn.09900915] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 01/26/2016] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND OBJECTIVES The effect of mammalian target of rapamycin (mTOR) inhibitors has never been tested in patients with autosomal dominant polycystic kidney disease (ADPKD) and severe renal insufficiency. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS In this academic, prospective, randomized, open label, blinded end point, parallel group trial (ClinicalTrials.gov no. NCT01223755), 41 adults with ADPKD, CKD stage 3b or 4, and proteinuria ≤0.5 g/24 h were randomized between September of 2010 and March of 2012 to sirolimus (3 mg/d; serum target levels of 5-10 ng/ml) added on to conventional therapy (n=21) or conventional treatment alone (n=20). Primary outcome was GFR (iohexol plasma clearance) change at 1 and 3 years versus baseline. RESULTS At the 1-year preplanned interim analysis, GFR fell from 26.7±5.8 to 21.3±6.3 ml/min per 1.73 m(2) (P<0.001) and from 29.6±5.6 to 24.9±6.2 ml/min per 1.73 m(2) (P<0.001) in the sirolimus and conventional treatment groups, respectively. Albuminuria (73.8±81.8 versus 154.9±152.9 μg/min; P=0.02) and proteinuria (0.3±0.2 versus 06±0.4 g/24 h; P<0.01) increased with sirolimus. Seven patients on sirolimus versus one control had de novo proteinuria (P=0.04), ten versus three patients doubled proteinuria (P=0.02), 18 versus 11 patients had peripheral edema (P=0.04), and 14 versus six patients had upper respiratory tract infections (P=0.03). Three patients on sirolimus had angioedema, 14 patients had aphthous stomatitis, and seven patients had acne (P<0.01 for both versus controls). Two patients progressed to ESRD, and two patients withdrew because of worsening of proteinuria. These events were not observed in controls. Thus, the independent data and safety monitoring board recommend early trial termination for safety reasons. At 1 year, total kidney volume (assessed by contrast-enhanced computed tomography imaging) increased by 9.0% from 2857.7±1447.3 to 3094.6±1519.5 ml on sirolimus and 4.3% from 3123.4±1695.3 to 3222.6±1651.4 ml on conventional therapy (P=0.12). On follow-up, 37% and 7% of serum sirolimus levels fell below or exceeded the therapeutic range, respectively. CONCLUSIONS Finding that sirolimus was unsafe and ineffective in patients with ADPKD and renal insufficiency suggests that mTOR inhibitor therapy may be contraindicated in this context.
Collapse
Affiliation(s)
- Piero Ruggenenti
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
- Units of Nephrology and Dialysis
| | - Giorgio Gentile
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
- Units of Nephrology and Dialysis
| | - Norberto Perico
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Annalisa Perna
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | | | - Matias Trillini
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Monica Cortinovis
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Claudia Patricia Ferrer Siles
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Jorge Arturo Reyes Loaeza
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Maria Carolina Aparicio
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Giorgio Fasolini
- Radiology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy; and
| | - Flavio Gaspari
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Davide Martinetti
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Fabiola Carrara
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Nadia Rubis
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Silvia Prandini
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Anna Caroli
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Kanishka Sharma
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Luca Antiga
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Andrea Remuzzi
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
| | - Giuseppe Remuzzi
- Clinical Research Center for Rare Diseases “Aldo e Cele Daccò,” IRCCS—Istituto di Ricerche Farmacologiche “Mario Negri,” Bergamo, Italy
- Units of Nephrology and Dialysis
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
263
|
Thiyagarajan V, Lin SH, Chang YC, Weng CF. Identification of novel FAK and S6K1 dual inhibitors from natural compounds via ADMET screening and molecular docking. Biomed Pharmacother 2016; 80:52-62. [DOI: 10.1016/j.biopha.2016.02.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/21/2016] [Accepted: 02/22/2016] [Indexed: 01/26/2023] Open
|
264
|
Considine MJ, Considine JA. On the language and physiology of dormancy and quiescence in plants. JOURNAL OF EXPERIMENTAL BOTANY 2016; 67:3189-203. [PMID: 27053719 DOI: 10.1093/jxb/erw138] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The language of dormancy is rich and poetic, as researchers spanning disciplines and decades have attempted to understand the spell that entranced 'Sleeping Beauty', and how she was gently awoken. The misleading use of 'dormancy', applied to annual axillary buds, for example, has confounded progress. Language is increasingly important as genetic and genomic approaches become more accessible to species of agricultural and ecological importance. Here we examine how terminology has been applied to different eco-physiological states in plants, and with pertinent reference to quiescent states described in other domains of life, in order to place plant quiescence and dormancy in a more complete context than previously described. The physiological consensus defines latency or quiescence as opportunistic avoidance states, where growth resumes in favourable conditions. In contrast, the dormant state in higher plants is entrained in the life history of the organism. Competence to resume growth requires quantitative and specific conditioning. This definition applies only to the embryo of seeds and specialized meristems in higher plants; however, mechanistic control of dormancy extends to mobile signals from peripheral tissues and organs, such as the endosperm of seed or subtending leaf of buds. The distinction between dormancy, quiescence, and stress-hardiness remains poorly delineated, most particularly in buds of winter perennials, which comprise multiple meristems of differing organogenic states. Studies in seeds have shown that dormancy is not a monogenic trait, and limited study has thus far failed to canalize dormancy as seen in seeds and buds. We argue that a common language, based on physiology, is central to enable further dissection of the quiescent and dormant states in plants. We direct the topic largely to woody species showing a single cycle of growth and reproduction per year, as these bear the majority of global timber, fruit, and nut production, as well being of great ecological value. However, for context and hypotheses, we draw on knowledge from annuals and other specialized plant conditions, from a perspective of the major physical, metabolic, and molecular cues that regulate cellular activity.
Collapse
Affiliation(s)
- Michael J Considine
- School of Plant Biology, and The Institute of Agriculture, The University of Western Australia, Perth, WA 6009 Australia Department of Agriculture and Food Western Australia, South Perth, WA 6151 Australia Centre for Plant Sciences, University of Leeds, Leeds, Yorkshire LS2 9JT, UK
| | - John A Considine
- School of Plant Biology, and The Institute of Agriculture, The University of Western Australia, Perth, WA 6009 Australia
| |
Collapse
|
265
|
Hoxhaj G, Caddye E, Najafov A, Houde VP, Johnson C, Dissanayake K, Toth R, Campbell DG, Prescott AR, MacKintosh C. The E3 ubiquitin ligase ZNRF2 is a substrate of mTORC1 and regulates its activation by amino acids. eLife 2016; 5. [PMID: 27244671 PMCID: PMC4889327 DOI: 10.7554/elife.12278] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/28/2016] [Indexed: 12/22/2022] Open
Abstract
The mechanistic Target of Rapamycin complex 1 (mTORC1) senses intracellular amino acid levels through an intricate machinery, which includes the Rag GTPases, Ragulator and vacuolar ATPase (V-ATPase). The membrane-associated E3 ubiquitin ligase ZNRF2 is released into the cytosol upon its phosphorylation by Akt. In this study, we show that ZNRF2 interacts with mTOR on membranes, promoting the amino acid-stimulated translocation of mTORC1 to lysosomes and its activation in human cells. ZNRF2 also interacts with the V-ATPase and preserves lysosomal acidity. Moreover, knockdown of ZNRF2 decreases cell size and cell proliferation. Upon growth factor and amino acid stimulation, mTORC1 phosphorylates ZNRF2 on Ser145, and this phosphosite is dephosphorylated by protein phosphatase 6. Ser145 phosphorylation stimulates vesicle-to-cytosol translocation of ZNRF2 and forms a novel negative feedback on mTORC1. Our findings uncover ZNRF2 as a component of the amino acid sensing machinery that acts upstream of Rag-GTPases and the V-ATPase to activate mTORC1. DOI:http://dx.doi.org/10.7554/eLife.12278.001 During digestion, proteins are broken down into their constituent parts called amino acids. Amino acids are transported in the bloodstream and are used to build up new cells and repair old ones. Optimal regulation of the cellular rates of amino acid uptake and protein synthesis is critical to the overall health of our bodies. Inside each of our cells is a molecule called mammalian target of rapamycin (mTOR for short), which acts as a controller that receives information about amino acid availability. mTOR also senses how much of each amino acid the cell needs and calibrates the cell’s amino acid uptake and protein synthesis machineries accordingly. When investigating an enzyme named ZNRF2, Hoxhaj et al. discovered that it interacts with mTOR on membranes inside cells. This raised questions about how ZNRF2 might work with mTOR to sense amino acid supplies and regulate cell growth. Hoxhaj et al. found that when cells are provided with amino acids and growth-stimulating hormones, mTOR is activated and attaches a phosphate group to ZNRF2. This chemical modification promotes the release of ZNRF2 from membranes so that ZNRF2 separates from mTOR. In contrast, when cells are starved of amino acids, this phosphate group is removed from ZNRF2, which then returns to the membranes. On membranes, ZNRF2 also influences the activity of a pump called V-ATPase, which controls the internal acidity of the membrane-enclosed vesicles named lysosomes that help to recycle amino acids inside cells. The action of ZNRF2 on the pump may help to prime mTOR so that it is ready to sense amino acids. These findings by Hoxhaj et al. suggest that ZNRF2 and mTOR may ‘tune’ each other, making constant to-and-fro adjustments to help ensure that levels of amino acid uptake and cell growth are set just right. However, many questions about ZNRF2 still remain to be addressed. For example, are genetic mutations in ZNRF2 involved in cancers, developmental disorders or growth syndromes? Is ZNRF2 most important in the brain, where it is particularly abundant? And how does ZNRF2 affect acidity within the lysosomes? DOI:http://dx.doi.org/10.7554/eLife.12278.002
Collapse
Affiliation(s)
- Gerta Hoxhaj
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom.,Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, United States
| | - Edward Caddye
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Ayaz Najafov
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Vanessa P Houde
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Catherine Johnson
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Kumara Dissanayake
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom.,Cell and Developmental Biology Division, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Rachel Toth
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - David G Campbell
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Alan R Prescott
- Cell Signalling and Immunology Division, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Carol MacKintosh
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom.,Cell and Developmental Biology Division, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
266
|
Law BYK, Mok SWF, Wu AG, Lam CWK, Yu MXY, Wong VKW. New Potential Pharmacological Functions of Chinese Herbal Medicines via Regulation of Autophagy. Molecules 2016; 21:359. [PMID: 26999089 PMCID: PMC6274228 DOI: 10.3390/molecules21030359] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/29/2016] [Accepted: 03/09/2016] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a universal catabolic cellular process for quality control of cytoplasm and maintenance of cellular homeostasis upon nutrient deprivation and environmental stimulus. It involves the lysosomal degradation of cellular components such as misfolded proteins or damaged organelles. Defects in autophagy are implicated in the pathogenesis of diseases including cancers, myopathy, neurodegenerations, infections and cardiovascular diseases. In the recent decade, traditional drugs with new clinical applications are not only commonly found in Western medicines, but also highlighted in Chinese herbal medicines (CHM). For instance, pharmacological studies have revealed that active components or fractions from Chaihu (Radix bupleuri), Hu Zhang (Rhizoma polygoni cuspidati), Donglingcao (Rabdosia rubesens), Hou po (Cortex magnoliae officinalis) and Chuan xiong (Rhizoma chuanxiong) modulate cancers, neurodegeneration and cardiovascular disease via autophagy. These findings shed light on the potential new applications and formulation of CHM decoctions via regulation of autophagy. This article reviews the roles of autophagy in the pharmacological actions of CHM and discusses their new potential clinical applications in various human diseases.
Collapse
Affiliation(s)
- Betty Yuen Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Simon Wing Fai Mok
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - An Guo Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Christopher Wai Kei Lam
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Margaret Xin Yi Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
267
|
Impact of rapamycin on status epilepticus induced hippocampal pathology and weight gain. Exp Neurol 2016; 280:1-12. [PMID: 26995324 DOI: 10.1016/j.expneurol.2016.03.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 02/07/2023]
Abstract
Growing evidence implicates the dentate gyrus in temporal lobe epilepsy (TLE). Dentate granule cells limit the amount of excitatory signaling through the hippocampus and exhibit striking neuroplastic changes that may impair this function during epileptogenesis. Furthermore, aberrant integration of newly-generated granule cells underlies the majority of dentate restructuring. Recently, attention has focused on the mammalian target of rapamycin (mTOR) signaling pathway as a potential mediator of epileptogenic change. Systemic administration of the mTOR inhibitor rapamycin has promising therapeutic potential, as it has been shown to reduce seizure frequency and seizure severity in rodent models. Here, we tested whether mTOR signaling facilitates abnormal development of granule cells during epileptogenesis. We also examined dentate inflammation and mossy cell death in the dentate hilus. To determine if mTOR activation is necessary for abnormal granule cell development, transgenic mice that harbored fluorescently-labeled adult-born granule cells were treated with rapamycin following pilocarpine-induced status epilepticus. Systemic rapamycin effectively blocked phosphorylation of S6 protein (a readout of mTOR activity) and reduced granule cell mossy fiber axon sprouting. However, the accumulation of ectopic granule cells and granule cells with aberrant basal dendrites was not significantly reduced. Mossy cell death and reactive astrocytosis were also unaffected. These data suggest that anti-epileptogenic effects of mTOR inhibition may be mediated by mechanisms other than inhibition of these common dentate pathologies. Consistent with this conclusion, rapamycin prevented pathological weight gain in epileptic mice, suggesting that rapamycin might act on central circuits or even peripheral tissues controlling weight gain in epilepsy.
Collapse
|
268
|
Korfel A, Schlegel U, Herrlinger U, Dreyling M, Schmidt C, von Baumgarten L, Pezzutto A, Grobosch T, Kebir S, Thiel E, Martus P, Kiewe P. Phase II Trial of Temsirolimus for Relapsed/Refractory Primary CNS Lymphoma. J Clin Oncol 2016; 34:1757-63. [PMID: 26976424 DOI: 10.1200/jco.2015.64.9897] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
PURPOSE In this phase II study (NCT00942747), temsirolimus was tested in patients with relapsed or refractory primary CNS lymphoma (PCNSL). PATIENTS AND METHODS Immunocompetent adults with histologically confirmed PCNSL after experiencing high-dose methotrexate-based chemotherapy failure who were not eligible for or had experienced high-dose chemotherapy with autologous stem-cell transplant failure were included. The first cohort (n = 6) received 25 mg temsirolimus intravenously once per week. All consecutive patients received 75 mg intravenously once per week. RESULTS Thirty-seven eligible patients (median age, 70 years) were included whose median time since their last treatment was 3.9 months (range, 0.1 to 14.6 months). Complete response was seen in five patients (13.5%), complete response unconfirmed in three (8%), and partial response in 12 (32.4%) for an overall response rate of 54%. Median progression-free survival was 2.1 months (95% CI, 1.1 to 3.0 months). The most frequent Common Toxicity Criteria ≥ 3° adverse event was hyperglycemia in 11 (29.7%) patients, thrombocytopenia in eight (21.6%), infection in seven (19%), anemia in four (10.8%), and rash in three (8.1%). Fourteen blood/CSF pairs were collected in nine patients (10 pairs in five patients in the 25-mg cohort and four pairs in four patients in the 75-mg cohort). The mean maximum blood concentration was 292 ng/mL for temsirolimus and 37.2 ng/mL for its metabolite sirolimus in the 25-mg cohort and 484 ng/mL and 91.1 ng/mL, respectively, in the 75-mg cohort. Temsirolimus CSF concentration was 2 ng/mL in one patient in the 75-mg cohort; in all others, no drug was found in their CSF. CONCLUSION Single-agent temsirolimus at a weekly dose of 75 mg was found to be active in relapsed/refractory patients with PCNSL; however, responses were usually short lived.
Collapse
Affiliation(s)
- Agnieszka Korfel
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany.
| | - Uwe Schlegel
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| | - Ulrich Herrlinger
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| | - Martin Dreyling
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| | - Christian Schmidt
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| | - Luisa von Baumgarten
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| | - Antonio Pezzutto
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| | - Thomas Grobosch
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| | - Sied Kebir
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| | - Eckhard Thiel
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| | - Peter Martus
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| | - Philipp Kiewe
- Agnieszka Korfel, Antonio Pezzutto, Eckhard Thiel, and Philipp Kiewe, Charité University Medicine Berlin; Thomas Grobosch, Labor Berlin - Charité Vivantes, Berlin; Uwe Schlegel, Ruhr-Universität Bochum, Bochum; Ulrich Herrlinger and Sied Kebir, University Hospital Bonn, Bonn; Martin Dreyling, Christian Schmidt, and Luisa von Baumgarten, Hospital of the Ludwig Maximilian University München, Munich; and Peter Martus, University Tuebingen, Tuebingen, Germany
| |
Collapse
|
269
|
Mukhopadhyay S, Frias MA, Chatterjee A, Yellen P, Foster DA. The Enigma of Rapamycin Dosage. Mol Cancer Ther 2016; 15:347-53. [PMID: 26916116 DOI: 10.1158/1535-7163.mct-15-0720] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/03/2015] [Indexed: 12/20/2022]
Abstract
The mTOR pathway is a critical regulator of cell growth, proliferation, metabolism, and survival. Dysregulation of mTOR signaling has been observed in most cancers and, thus, the mTOR pathway has been extensively studied for therapeutic intervention. Rapamycin is a natural product that inhibits mTOR with high specificity. However, its efficacy varies by dose in several contexts. First, different doses of rapamycin are needed to suppress mTOR in different cell lines; second, different doses of rapamycin are needed to suppress the phosphorylation of different mTOR substrates; and third, there is a differential sensitivity of the two mTOR complexes mTORC1 and mTORC2 to rapamycin. Intriguingly, the enigmatic properties of rapamycin dosage can be explained in large part by the competition between rapamycin and phosphatidic acid (PA) for mTOR. Rapamycin and PA have opposite effects on mTOR whereby rapamycin destabilizes and PA stabilizes both mTOR complexes. In this review, we discuss the properties of rapamycin dosage in the context of anticancer therapeutics.
Collapse
Affiliation(s)
- Suman Mukhopadhyay
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York
| | - Maria A Frias
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York
| | - Amrita Chatterjee
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York
| | - Paige Yellen
- Molecular Pharmacology & Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - David A Foster
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York. Department of Pharmacology, Weill-Cornell Medical College, New York, New York.
| |
Collapse
|
270
|
Pusceddu S, Buzzoni R, Vernieri C, Concas L, Marceglia S, Giacomelli L, Milione M, Leuzzi L, Femia D, Formisano B, Mazzaferro V, de Braud F. Metformin with everolimus and octreotide in pancreatic neuroendocrine tumor patients with diabetes. Future Oncol 2016; 12:1251-60. [PMID: 26890290 DOI: 10.2217/fon-2015-0077] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
A bidirectional relationship seems to exist between diabetes mellitus and development of pancreatic tumors. Metformin, the most widely used drug in the treatment of Type 2 diabetes mellitus, has recently emerged as a potentially active agent in cancer chemoprevention and treatment. In this article, we discuss the potential correlation between glycemic status, administration of antiglycemic treatments, such as metformin or insulin, and prognosis of pancreatic neuroendocrine tumors patients treated with everolimus and octreotide, on the basis of existing evidence and our experience.
Collapse
Affiliation(s)
- Sara Pusceddu
- Medical Oncology Unit 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roberto Buzzoni
- Day Hospital/Outpatient Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudio Vernieri
- Medical Oncology Unit 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,IFOM, FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Laura Concas
- Day Hospital/Outpatient Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Marceglia
- Department of Information & Bioengineering, Politecnico University, Milan, Italy
| | - Luca Giacomelli
- Department of Surgical Sciences & Integrated Diagnostics, School of Medicine, Genova University, Genoa, Italy
| | - Massimo Milione
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Livia Leuzzi
- Day Hospital/Outpatient Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Daniela Femia
- Day Hospital/Outpatient Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Barbara Formisano
- Medical Oncology Unit 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Vincenzo Mazzaferro
- Gastro-Intestinal Surgery, Liver Transplantation & Hepatology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Unit 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
271
|
Abstract
The excitement surrounding checkpoint inhibitors in the treatment of patients with cancer exemplifies a triumph of the long-term value of investing in basic science and fundamental questions of T-cell signaling. The pharmaceutical future actively embraces ways of making more patients’ cancers responsive to these inhibitors. Such a process will be aided by elucidation of signaling and regulation. With thousands of articles spread across almost 30 years, this commentary can touch only on portions of the canonical picture of T-cell signaling and provide a few parables from work on mammalian (or mechanistic) target of rapamycin (mTOR) pathways as they link to early and later phases of lymphocyte activation. The piece will turn a critical eye to some issues with models about these pathways in T cells. Many of the best insights lie in the future despite all that is uncovered already, but a contention is that further therapeutic successes will be fostered by dealing with disparities among findings and attention to the temporal, spatial, and stochastic aspects of T-cell responses. Finally, thoughts on some (though not all) items urgently needed for future progress will be mooted.
Collapse
Affiliation(s)
- Mark Boothby
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
272
|
Francois-Vaughan H, Adebayo AO, Brilliant KE, Parry NMA, Gruppuso PA, Sanders JA. Persistent effect of mTOR inhibition on preneoplastic foci progression and gene expression in a rat model of hepatocellular carcinoma. Carcinogenesis 2016; 37:408-419. [PMID: 26905589 DOI: 10.1093/carcin/bgw016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/30/2016] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a heterogeneous disease in which tumor subtypes can be identified based on the presence of adult liver progenitor cells. Having previously identified the mTOR pathway as critical to progenitor cell proliferation in a model of liver injury, we investigated the temporal activation of mTOR signaling in a rat model of hepatic carcinogenesis. The model employed chemical carcinogens and partial hepatectomy to induce progenitor marker-positive HCC. Immunohistochemical staining for phosphorylated ribosomal protein S6 indicated robust mTOR complex 1 (mTORC1) activity in early preneoplastic lesions that peaked during the first week and waned over the subsequent 10 days. Continuous administration of rapamycin by subcutaneous pellet for 70 days markedly reduced the development of focal lesions, but resulted in activation of the PI3K signaling pathway. To test the hypothesis that early mTORC1 activation was critical to the development and progression of preneoplastic foci, we limited rapamycin administration to the 3-week period at the start of the protocol. Focal lesion burden was reduced to a degree indistinguishable from that seen with continuous administration. Short-term rapamycin did not result in the activation of PI3K or mTORC2 pathways. Microarray analysis revealed a persistent effect of short-term mTORC1 inhibition on gene expression that resulted in a genetic signature reminiscent of normal liver. We conclude that mTORC1 activation during the early stages of hepatic carcinogenesis may be critical due to the development of preneoplastic focal lesions in progenitor marker-positive HCC. mTORC1 inhibition may represent an effective chemopreventive strategy for this form of liver cancer.
Collapse
Affiliation(s)
- Heather Francois-Vaughan
- Division of Pediatric Endocrinology , Department of Pediatrics , Rhode Island Hospital , Providence , RI 02903 , USA
| | - Adeola O Adebayo
- Division of Pediatric Endocrinology, Department of Pediatrics, Rhode Island Hospital, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02905, USA
| | - Kate E Brilliant
- Division of Hematology/Oncology , Department of Medicine , Rhode Island Hospital , Providence , RI 02903USA
| | - Nicola M A Parry
- Midwest Veterinary Pathology , LLC , Lafayette , IN 47909 , USA and
| | - Philip A Gruppuso
- Division of Pediatric Endocrinology, Department of Pediatrics, Rhode Island Hospital, Providence, RI 02903, USA.,Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Jennifer A Sanders
- Division of Pediatric Endocrinology, Department of Pediatrics, Rhode Island Hospital, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02905, USA
| |
Collapse
|
273
|
Eukaryotic initiation factor 4E-binding protein 1 (4E-BP1): a master regulator of mRNA translation involved in tumorigenesis. Oncogene 2016; 35:4675-88. [DOI: 10.1038/onc.2015.515] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/11/2015] [Accepted: 12/11/2015] [Indexed: 01/17/2023]
|
274
|
mTOR/P70S6K promotes spermatogonia proliferation and spermatogenesis in Sprague Dawley rats. Reprod Biomed Online 2016; 32:207-17. [DOI: 10.1016/j.rbmo.2015.11.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/07/2015] [Accepted: 11/10/2015] [Indexed: 01/14/2023]
|
275
|
Fischer S, Gillessen S, Rothermundt C. Sequence of treatment in locally advanced and metastatic renal cell carcinoma. Transl Androl Urol 2016; 4:310-25. [PMID: 26816832 PMCID: PMC4708238 DOI: 10.3978/j.issn.2223-4683.2015.04.07] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The spectrum of drugs that have shown activity in advanced or metastatic renal cell carcinoma (RCC) has led to a debate on the optimal sequence of treatments. There is agreement on recommending targeted agents as the standard of care in this disease. Uncertainty, however, remains on the best first-line drug choice. Physicians and patients may select sunitinib, bevacizumab in combination with interferon-alpha (IFN-α), pazopanib, or-in poor risk patients-temsirolimus. There are also a variety of therapies with proven efficacy on hand in the second-line setting: sorafenib, pazopanib, axitinib, and everolimus. While most randomized RCC trials assessed progression free survival (PFS) as primary endpoint, some agents were shown to improve median overall survival (OS), and given in sequence they have extended the life expectancy of RCC patients from 13 months in the cytokine era to over 30 months. Despite the progress made, there are sobering aspects to the oncologic success story in RCC, as the new treatments do not obtain an objective response or disease stabilization (SD) in all patients. There are also as yet no predictors to select patients who might benefit and those who are primary resistant to specific drugs, and ultimately almost all patients will experience disease progression. Bearing inevitable treatment failure in mind, availability of further drugs and switching therapy while the patient is in a condition to continue pharmacotherapy is essential. Of note, depending on the setting, only 33-59% of patients receive second-line treatment. In this review we present data on first-, second-, and third-line treatment in RCC, and discuss the difficulties in their interpretation in the context of treatment sequence. We summarize biological aspects and discuss mechanisms of resistance to anti-angiogenic therapy and their implications for treatment selection.
Collapse
Affiliation(s)
- Stefanie Fischer
- Division of Oncology/Haematology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| | - Silke Gillessen
- Division of Oncology/Haematology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| | - Christian Rothermundt
- Division of Oncology/Haematology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| |
Collapse
|
276
|
McLeod M, Breen L, Hamilton DL, Philp A. Live strong and prosper: the importance of skeletal muscle strength for healthy ageing. Biogerontology 2016; 17:497-510. [PMID: 26791164 PMCID: PMC4889643 DOI: 10.1007/s10522-015-9631-7] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/22/2015] [Indexed: 12/19/2022]
Abstract
Due to improved health care, diet and infrastructure in developed countries, since 1840 life expectancy has increased by approximately 2 years per decade. Accordingly, by 2050, a quarter of Europe’s population will be over 65 years, representing a 10 % rise in half a century. With this rapid rise comes an increased prevalence of diseases of ageing and associated healthcare expenditure. To address the health consequences of global ageing, research in model systems (worms, flies and mice) has indicated that reducing the rate of organ growth, via reductions in protein synthetic rates, has multi-organ health benefits that collectively lead to improvements in lifespan. In contrast, human pre-clinical, clinical and large cohort prospective studies demonstrate that ageing leads to anabolic (i.e. growth) impairments in skeletal muscle, which in turn leads to reductions in muscle mass and strength, factors directly associated with mortality rates in the elderly. As such, increasing muscle protein synthesis via exercise or protein-based nutrition maintains a strong, healthy muscle mass, which in turn leads to improved health, independence and functionality. The aim of this review is to critique current literature relating to the maintenance of muscle mass across lifespan and discuss whether maintaining or reducing protein synthesis is the most logical approach to support musculoskeletal function and by extension healthy human ageing.
Collapse
Affiliation(s)
- Michael McLeod
- MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, B15 2TT, UK.,School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Leigh Breen
- MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, B15 2TT, UK.,School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | | | - Andrew Philp
- MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, B15 2TT, UK. .,School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
277
|
He B, Zhang N, Zhao R. Dexamethasone Downregulates SLC7A5 Expression and Promotes Cell Cycle Arrest, Autophagy and Apoptosis in BeWo Cells. J Cell Physiol 2016; 231:233-42. [PMID: 26094588 DOI: 10.1002/jcp.25076] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 06/08/2015] [Indexed: 12/14/2022]
Abstract
Synthetic glucocorticoids (GCs) such as dexamethasone (Dex) are widely given to pregnant women to induce maturation and improve viability of preterm infants. Despite the beneficial effects, synthetic GCs have adverse effects on placental growth and nutrient transport system. However, the molecular mechanisms involved in these events remain unknown. Here we use a human placental choriocarcinoma cell line (BeWo) as model to explore the pathway linking amino acids transport with cell viability under Dex challenge. BeWo cells treated with Dex (100 nM) for 24 h demonstrated G1/S cell cycle arrest together with enhanced autophagy and apoptosis. Concurrently, the amino acid carrier SLC7A5 was down-regulated in association with impaired cellular amino acids uptake and inhibition of mammalian target of rapamycin (mTOR) signaling. Similar cellular responses were observed in BeWo cells treated with BCH, a classical System L inhibitor which inactivates SLC7A5. The glucocorticoid receptor (GR) antagonist RU486 was able to diminish Dex-induced translocation of GR into nucleus and to abolish these effects. Furthermore, Dex treatment significantly promoted the binding of GR to the proximal promoter sequence of SLC7A5 gene. Taken together, our results show that Dex downregulates SLC7A5 expression via GR-mediated transrepression. The impaired amino acids uptake leads to inhibition of mTOR signaling which in turn causes inhibited proliferation and enhanced autophagy and apoptosis in BeWo cells. These findings indicate that SLC7A5 mediates the effect of Dex on cell viability, thus providing a novel molecular target for the prevention and treatment of Dex-induced cell cycle arrest and apoptosis in placental cells.
Collapse
Affiliation(s)
- Bin He
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| | - Nana Zhang
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
278
|
Costales J, Kolevzon A. The therapeutic potential of insulin-like growth factor-1 in central nervous system disorders. Neurosci Biobehav Rev 2016; 63:207-22. [PMID: 26780584 DOI: 10.1016/j.neubiorev.2016.01.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 11/09/2015] [Accepted: 01/04/2016] [Indexed: 12/13/2022]
Abstract
Central nervous system (CNS) development is a finely tuned process that relies on multiple factors and intricate pathways to ensure proper neuronal differentiation, maturation, and connectivity. Disruption of this process can cause significant impairments in CNS functioning and lead to debilitating disorders that impact motor and language skills, behavior, and cognitive functioning. Recent studies focused on understanding the underlying cellular mechanisms of neurodevelopmental disorders have identified a crucial role for insulin-like growth factor-1 (IGF-1) in normal CNS development. Work in model systems has demonstrated rescue of pathophysiological and behavioral abnormalities when IGF-1 is administered, and several clinical studies have shown promise of efficacy in disorders of the CNS, including autism spectrum disorder (ASD). In this review, we explore the molecular pathways and downstream effects of IGF-1 and summarize the results of completed and ongoing pre-clinical and clinical trials using IGF-1 as a pharmacologic intervention in various CNS disorders. This aim of this review is to provide evidence for the potential of IGF-1 as a treatment for neurodevelopmental disorders and ASD.
Collapse
Affiliation(s)
| | - Alexander Kolevzon
- Department of Psychiatry, United States; Department of Pediatrics, United States; Seaver Autism Center for Research and Treatment, United States; Friedman Brain Institute, United States; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
279
|
Walker NM, Belloli EA, Stuckey L, Chan KM, Lin J, Lynch W, Chang A, Mazzoni SM, Fingar DC, Lama VN. Mechanistic Target of Rapamycin Complex 1 (mTORC1) and mTORC2 as Key Signaling Intermediates in Mesenchymal Cell Activation. J Biol Chem 2016; 291:6262-71. [PMID: 26755732 DOI: 10.1074/jbc.m115.672170] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Indexed: 01/05/2023] Open
Abstract
Fibrotic diseases display mesenchymal cell (MC) activation with pathologic deposition of matrix proteins such as collagen. Here we investigate the role of mTOR complex 1 (mTORC1) and mTORC2 in regulating MC collagen expression, a hallmark of fibrotic disease. Relative to normal MCs (non-Fib MCs), MCs derived from fibrotic human lung allografts (Fib-MCs) demonstrated increased phosphoinositide-3kinase (PI3K) dependent activation of both mTORC1 and mTORC2, as measured by increased phosphorylation of S6K1 and 4E-BP1 (mTORC1 substrates) and AKT (an mTORC2 substrate). Dual ATP-competitive TORC1/2 inhibitor AZD8055, in contrast to allosteric mTORC1-specific inhibitor rapamycin, strongly inhibited 4E-BP1 phosphorylation and collagen I expression in Fib-MCs. In non-Fib MCs, increased mTORC1 signaling was shown to augment collagen I expression. mTORC1/4E-BP1 pathway was identified as an important driver of collagen I expression in Fib-MCs in experiments utilizing raptor gene silencing and overexpression of dominant-inhibitory 4E-BP1. Furthermore, siRNA-mediated knockdown of rictor, an mTORC2 partner protein, reduced mTORC1 substrate phosphorylation and collagen expression in Fib-, but not non-Fib MCs, revealing a dependence of mTORC1 signaling on mTORC2 function in activated MCs. Together these studies suggest a novel paradigm where fibrotic activation in MCs increases PI3K dependent mTORC1 and mTORC2 signaling and leads to increased collagen I expression via the mTORC1-dependent 4E-BP1/eIF4E pathway. These data provide rationale for targeting specific components of mTORC pathways in fibrotic states and underscore the need to further delineate mTORC2 signaling in activated cell states.
Collapse
Affiliation(s)
- Natalie M Walker
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine
| | - Elizabeth A Belloli
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine
| | | | - Kevin M Chan
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine
| | | | | | | | - Serina M Mazzoni
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine
| | - Diane C Fingar
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Vibha N Lama
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine,
| |
Collapse
|
280
|
Chen Y, Wang W, Wang H, Li Y, Shi M, Li H, Yan J. Rapamycin Attenuates Splenomegaly in both Intrahepatic and Prehepatic Portal Hypertensive Rats by Blocking mTOR Signaling Pathway. PLoS One 2016; 11:e0141159. [PMID: 26734934 PMCID: PMC4703391 DOI: 10.1371/journal.pone.0141159] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/03/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Spleen enlargement is often detected in patients with liver cirrhosis, but the precise pathogenetic mechanisms behind the phenomenon have not been clearly elucidated. We investigated the pathogenetic mechanisms of splenomegaly in both portal hypertensive patients and rats, and tried to identify the possible therapy for this disease. METHODS Spleen samples were collected from portal hypertensive patients after splenectomy. Rat models of portal hypertension were induced by common bile duct ligation and partial portal vein ligation. Spleen samples from patients and rats were used to study the characteristics of splenomegaly by histological, immunohistochemical, and western blot analyses. Rapamycin or vehicle was administered to rats to determine the contribution of mTOR signaling pathway in the development of splenomegaly. RESULTS We found that not only spleen congestion, but also increasing angiogenesis, fibrogenesis, inflammation and proliferation of splenic lymphoid tissue contributed to the development of splenomegaly in portal hypertensive patients and rats. Intriguingly, splenomegaly developed time-dependently in portal hypertensive rat that accompanied with progressive activation of mTOR signaling pathway. mTOR blockade by rapamycin profoundly ameliorated splenomegaly by limiting lymphocytes proliferation, angiogenesis, fibrogenesis and inflammation as well as decreasing portal pressure. CONCLUSIONS This study provides compelling evidence indicating that mTOR signaling activation pathway plays a key role in the pathogenesis of splenomegaly in both portal hypertensive patients and rats. Therapeutic intervention targeting mTOR could be a promising strategy for patients with portal hypertension and splenomegaly.
Collapse
Affiliation(s)
- Yunyang Chen
- Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weijie Wang
- Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Huakai Wang
- Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yongjian Li
- Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minmin Shi
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongwei Li
- Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiqi Yan
- Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
281
|
Liu TM, Lee EH, Lim B, Shyh-Chang N. Concise Review: Balancing Stem Cell Self-Renewal and Differentiation with PLZF. Stem Cells 2016; 34:277-87. [DOI: 10.1002/stem.2270] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/21/2015] [Accepted: 11/29/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Tong Ming Liu
- Cancer Stem Cell Biology, Genome Institute of Singapore; Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery; National University of Singapore; Singapore
- NUS Tissue Engineering Program (NUSTEP); National University of Singapore; Singapore
| | - Bing Lim
- Cancer Stem Cell Biology, Genome Institute of Singapore; Singapore
| | - Ng Shyh-Chang
- Stem Cell and Regenerative Biology; Genome Institute of Singapore; Singapore
| |
Collapse
|
282
|
The role of CAPE in PI3K/AKT/mTOR activation and oxidative stress on testis torsion. Acta Histochem 2016; 118:31-7. [PMID: 26651953 DOI: 10.1016/j.acthis.2015.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 01/13/2023]
Abstract
Ischemia reperfusion injury arises from testicular torsion resulting in a loss of spermatogenesis and significant germ cell apoptosis. This study evaluates the prooxidant/antioxidant effects of caffeic acid phenethyl ester (CAPE) through PI3K/AKT/mTOR signal pathways on testis torsion. A total of (28) male Wistar rats were divided randomly into 4 groups (n=7 for each group):group A (sham) group,group B torsion/detorsion group, group C (saturation group, during four days of CAPE, one dose (10 μmol/kg, i.p)) and group D (a single dose of CAPE 2h after torsion and before detorsion). At the end of the study, unilateral orchiectomies were performed for measurements of MDA and 8OHdG levels, histopathologic and immunohistochemical and TUNEL apoptotic cell examination. Testicular torsion-detorsion led to a significant decrease in the mean values of the Johnsen's scores and a significant increase in the apoptotic cell values of group B. There were no significant differences between group D and group A. In addition, the MDA and 8OHdG levels increased significantly in group B. The MDA and 8OHdG values were lower in group D. However, the 8OHdG levels were higher in group C than the groups A and D. On the other hand, CAPE suppresses mTOR activation and reduces the apoptosis on ischemia/reperfusion damage in rat testis. These results demonstrate that CAPE suppresses mTOR activation and reduces the apoptosis on ischemia/reperfusion damage in rat testis.
Collapse
|
283
|
Autophagic lysosomal reformation depends on mTOR reactivation in H2O2-induced autophagy. Int J Biochem Cell Biol 2016; 70:76-81. [DOI: 10.1016/j.biocel.2015.11.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 11/09/2015] [Accepted: 11/10/2015] [Indexed: 12/19/2022]
|
284
|
Lodjak J, Tilgar V, Mägi M. Does the interaction between glucocorticoids and insulin-like growth factor 1 predict nestling fitness in a wild passerine? Gen Comp Endocrinol 2016; 225:149-154. [PMID: 26519758 DOI: 10.1016/j.ygcen.2015.10.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/29/2015] [Accepted: 10/22/2015] [Indexed: 12/20/2022]
Abstract
The crucial question in evolutionary ecology is to find out how physiological traits have coevolved so animals fit their stochastic environments. The plasticity of these different physiological mechanisms is largely mediated by hormones, like glucocorticoids and insulin-like growth factor 1 (IGF-1). Brood size manipulation with nestlings of free-living great tits (Parus major) was carried out to see the way in which plasma IGF-1 and feather corticosterone, a predictor of long-term sustained plasma corticosterone level, are associated across different nutritional conditions and how this association predicts survival during the nestling phase. We showed that the association between levels of IGF-1 and corticosterone depended on physiological condition of nestlings. Namely, there was a positive association between the hormones in nestlings from the decreased broods and a negative association in nestlings from the enlarged broods. Furthermore, we showed that the interaction between levels of IGF-1 and corticosterone was also related with the survival of the nestlings. Our results suggest that signalling pathways of IGF-1 and corticosterone most likely interact with each other in a nutrition-dependent way to maximize the rate of development and survival of nestlings in their stochastic environment.
Collapse
Affiliation(s)
- Jaanis Lodjak
- Department of Zoology, Institute of Ecology and Earth Sciences, University of Tartu, 46 Vanemuise Street, Tartu 51014, Estonia.
| | - Vallo Tilgar
- Department of Zoology, Institute of Ecology and Earth Sciences, University of Tartu, 46 Vanemuise Street, Tartu 51014, Estonia
| | - Marko Mägi
- Department of Zoology, Institute of Ecology and Earth Sciences, University of Tartu, 46 Vanemuise Street, Tartu 51014, Estonia
| |
Collapse
|
285
|
Ma Y, Ma N, Liu Q, Qi Y, Manabe RI, Furuyashiki T. Tor Signaling Regulates Transcription of Amino Acid Permeases through a GATA Transcription Factor Gaf1 in Fission Yeast. PLoS One 2015; 10:e0144677. [PMID: 26689777 PMCID: PMC4686964 DOI: 10.1371/journal.pone.0144677] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/20/2015] [Indexed: 01/17/2023] Open
Abstract
In the fission yeast, two Tor isoforms, Tor1 and Tor2, oppositely regulate gene expression of amino acid permeases. To elucidate the transcriptional machinery for these regulations, here we have employed the cap analysis of gene expression (CAGE), a method of analyzing expression profiles and identifying transcriptional start sites (TSSs). The loss of Tor1 decreased, and Tor2 inhibition by its temperature sensitive mutation increased, mRNA expression of isp5+, per1+, put4+ and SPBPB2B2.01. In contrast, the loss of Tor1 increased, and Tor2 inhibition decreased, the expression of cat1+. These changes were confirmed by semi-quantitative RT-PCR. These opposite effects by the loss of Tor1 and Tor2 inhibition appeared to occur evenly across multiple TSSs for the respective genes. The motif discovery analysis based on the CAGE results identified the GATA motifs as a potential cis-regulatory element for Tor-mediated regulation. In the luciferase reporter assay, the loss of Tor1 reduced, and Tor2 inhibition and nitrogen depletion increased, the activity of isp5+ promoter as well as that of a GATAAG reporter. One of the GATAAG motifs in isp5+ promoter was critical for its transcriptional activity, and a GATA transcription factor Gaf1 was critical for the activities of isp5+ promoter and the GATAAG reporter. Furthermore, Tor2 inhibition and nitrogen depletion induced nuclear localization of Gaf1 from the cytosol and its dephosphorylation. These results suggest that Tor2 inhibition, which is known to be induced by nitrogen depletion, promotes nuclear localization of Gaf1, thereby inducing isp5+ transcription through Gaf1 binding to the GATAAG motif in its promoter. Since Gaf1 was also critical for transcription of per1+ and put4+, Tor-Gaf1 signaling may coordinate transcription of multiple amino acid permeases according to nutrient availability.
Collapse
Affiliation(s)
- Yan Ma
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
- * E-mail:
| | - Ning Ma
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Qingbin Liu
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yao Qi
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ri-ichiroh Manabe
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | - Tomoyuki Furuyashiki
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
286
|
Papa A, Zaccarelli E, Caruso D, Vici P, Benedetti Panici P, Tomao F. Targeting angiogenesis in endometrial cancer - new agents for tailored treatments. Expert Opin Investig Drugs 2015; 25:31-49. [PMID: 26560489 DOI: 10.1517/13543784.2016.1116517] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Endometrial carcinoma represents the most frequent gynecologic tumor in developed countries. The majority of women presents with low-grade tumors but a significant subset of women experience recurrence and do not survive their disease. Patients with stage III/ IV or recurrent endometrial cancer have a poor prognosis. Identification of active and tolerable new targeted agents versus specific molecular targets is a priority objective. Angiogenesis is a complex process that plays a crucial role in the development of many types of cancer and in particular endometrial cancer. AREAS COVERED In this review, the authors highlight the main angiogenetic molecular pathways and the anti-angiogenic agents in Phase II clinical trials for endometrial cancer treatment. The authors focus on reports from recent years on angiogenesis inhibitors used in endometrial cancer, including anti- vascular endothelial growth factor (VEGF) monoclonal antibodies (bevacizumab and aflibercept), mammalian target of rapamycin inhibitors (mTORi) (everolimus, temsirolimus and ridaforolimus), PI3 K inhibitors (BKM120), tyrosine kinase inhibitors (brivanib, sunitinib, dovitinib and nintedanib) and thalidomide. EXPERT OPINION These anti-angiogenic drugs, while used either alone or in combination with chemotherapy, have presented mixed results in treating endometrial cancer patients. Challenges for the future include the identification of new pathways, early identification and overcoming resistance and the use of these molecules in combination with old and new chemotherapeutic and targeted agents.
Collapse
Affiliation(s)
- Anselmo Papa
- a Department of Medico-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome, Oncology Unit , Istituto Chirurgico Ortopedico Traumatologico , 04100 Latina , Italy
| | - Eleonora Zaccarelli
- a Department of Medico-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome, Oncology Unit , Istituto Chirurgico Ortopedico Traumatologico , 04100 Latina , Italy
| | - Davide Caruso
- a Department of Medico-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome, Oncology Unit , Istituto Chirurgico Ortopedico Traumatologico , 04100 Latina , Italy
| | - Patrizia Vici
- b Division of Medical Oncology B , Regina Elena National Cancer Institute , 00144 Rome , Italy
| | - Pierluigi Benedetti Panici
- c Department of Gynecological, Obstetrical and Urologic Sciences , "Sapienza" University of Rome , 00186 Rome , Italy
| | - Federica Tomao
- c Department of Gynecological, Obstetrical and Urologic Sciences , "Sapienza" University of Rome , 00186 Rome , Italy
| |
Collapse
|
287
|
Yaba A, Bozkurt ER, Demir N. mTOR expression in human testicular seminoma. Andrologia 2015; 48:702-7. [DOI: 10.1111/and.12504] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2015] [Indexed: 11/30/2022] Open
Affiliation(s)
- A. Yaba
- Institute of Health Sciences; Yeditepe University; İstanbul Turkey
| | - E. R. Bozkurt
- Department of Pathology; İstanbul Samatya Education and Research Hospital; İstanbul Turkey
| | - N. Demir
- Department of Histology and Embryology; Faculty of Medicine; Akdeniz University; Antalya Turkey
| |
Collapse
|
288
|
PATLOLLA JAGANM, KOPELOVICH LEVY, QIAN LI, ZHANG YUTING, KUMAR GAURAV, MADKA VENKATESHWAR, MOHAMMED ALTAF, BIDDICK LAURA, SADEGHI MICHAEL, LIGHTFOOT STAN, RAO CHINTHALAPALLYV. Early and delayed intervention with rapamycin prevents NNK-induced lung adenocarcinoma in A/J mice. Oncol Rep 2015; 34:2925-34. [PMID: 26397133 PMCID: PMC4735698 DOI: 10.3892/or.2015.4277] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 07/09/2015] [Indexed: 12/18/2022] Open
Abstract
In tobacco-associated lung cancers, the protein kinase B/mammalian target of rapamycin (Akt/mTOR) pathway frequently is activated by nicotine and its metabolite 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). The aim of the present study was to examine the effects of early or late intervention with rapamycin in NNK-induced lung adenoma and progression to adenocarcinoma in female A/J mice. At 7 weeks of age, 40 mice/each carcinogen group received one dose of 10 μmol NNK i.p. Three weeks later, the early intervention groups (25/group) were fed diets containing 0, 8 or 16 ppm rapamycin. The mice were sacrificed after 17 or 34 weeks of drug exposure and tumors were evaluated via histopathology. For late intervention (late adenoma and adenocarcinoma stage), groups of 15 mice were administered diets containing 8 or 16 ppm rapamycin starting 20 weeks after NNK treatment and continuing for 17 weeks before evaluation of tumor progression. Administration of 8 or 16 ppm rapamycin as an early or a late stage intervention significantly suppressed lung adenoma and adenocarcinoma formation (p<0.01-0.0001) after 17 or 34 weeks of exposure. The effect was more pronounced (>50‑60% tumor inihibition; p<0.0001) at the early intervention and the size of NNK-induced tumors decreased from >2.10 to <~0.75 mm3 (p=0.0056). Lung tumors harvested from mice exposed to rapamycin showed a significant decrease in p-mTOR, p-S6K1, PCNA and Bcl-xL as compared with controls in the early and late stage intervention studies. These observations suggest that rapamycin is highly effective even with administration after dysplastic adenoma or early adenocarcinoma stages and is useful for high-risk lung cancer patients.
Collapse
Affiliation(s)
- JAGAN M.R. PATLOLLA
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - LEVY KOPELOVICH
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD 20892-9788, USA
| | - LI QIAN
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - YUTING ZHANG
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - GAURAV KUMAR
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - VENKATESHWAR MADKA
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - ALTAF MOHAMMED
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - LAURA BIDDICK
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - MICHAEL SADEGHI
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - STAN LIGHTFOOT
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - CHINTHALAPALLY V. RAO
- Center for Chemoprevention and Cancer Drug Development, Department of Medicine, Hem-Onc Section, PCS Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
289
|
Rapamycin Protects from Type-I Peritoneal Membrane Failure Inhibiting the Angiogenesis, Lymphangiogenesis, and Endo-MT. BIOMED RESEARCH INTERNATIONAL 2015; 2015:989560. [PMID: 26688823 PMCID: PMC4673327 DOI: 10.1155/2015/989560] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 08/27/2015] [Accepted: 10/13/2015] [Indexed: 01/16/2023]
Abstract
Preservation of peritoneal membrane (PM) is essential for long-term survival in peritoneal dialysis (PD). Continuous presence of PD fluids (PDF) in the peritoneal cavity generates chronic inflammation and promotes changes of the PM, such as fibrosis, angiogenesis, and lymphangiogenesis. Mesothelial-to-mesenchymal transition (MMT) and endothelial-to-mesenchymal transition (Endo-MT) seem to play a central role in this pathogenesis. We speculated that Rapamycin, a potent immunosuppressor, could be beneficial by regulating blood and lymphatic vessels proliferation. We demonstrate that mice undergoing a combined PD and Rapamycin treatment (PDF + Rapa group) presented a reduced PM thickness and lower number of submesothelial blood and lymphatic vessels, as well as decreased MMT and Endo-MT, comparing with their counterparts exposed to PD alone (PDF group). Peritoneal water transport in the PDF + Rapa group remained at control level, whereas PD effluent levels of VEGF, TGF-β, and TNF-α were lower than in the PDF group. Moreover, the treatment of mesothelial cells with Rapamycin in vitro significantly decreased VEGF synthesis and selectively inhibited the VEGF-C and VEGF-D release when compared with control cells. Thus, Rapamycin has a protective effect on PM in PD through an antifibrotic and antiproliferative effect on blood and lymphatic vessels. Moreover, it inhibits Endo-MT and, at least partially, MMT.
Collapse
|
290
|
mTORC1 directly phosphorylates and activates ERα upon estrogen stimulation. Oncogene 2015; 35:3535-43. [PMID: 26522726 PMCID: PMC4853282 DOI: 10.1038/onc.2015.414] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 09/09/2015] [Accepted: 09/28/2015] [Indexed: 01/08/2023]
Abstract
Breast cancer is the leading cause of cancer-related deaths among women. Approximately 75% of breast cancers are estrogen receptor α (ERα) positive, underscoring the dependence of cancer cells on estrogen for growth and survival. Patients treated with endocrine therapy often develop resistance, either de novo or acquired, which in some cases is caused by aberrations within the growth factor signaling pathways. The mechanistic target of rapamycin complex 1 (mTORC1) has emerged as a critical node in estrogenic signaling. We have previously shown that mTORC1 can phosphorylate and activate ERα on S167 via its effector the 40S ribosomal S6 kinase 1 (S6K1). Presently, we have uncovered a direct link between mTORC1 and ERα. We found that ERα binds to regulatory-associated protein of mTOR (Raptor) and causes it to translocate to the nucleus upon estrogen stimulation. Additionally, we identified mTOR as the kinase that phosphorylates ERα on S104/106 and activates transcription of ER target genes. Our findings show a direct link between mTORC1 and ERα, which further implicates mTORC1 signaling in the pathogenesis of ER-positive breast cancer and provides rationale for FDA-approved use of mTORC1 inhibitors in combination with endocrine agents for treatment of this disease.
Collapse
|
291
|
Yoshimura A, Nishimura S, Otsuka S, Hattori A, Kakeya H. Structure Elucidation of Verucopeptin, a HIF-1 Inhibitory Polyketide-Hexapeptide Hybrid Metabolite from an Actinomycete. Org Lett 2015; 17:5364-7. [PMID: 26484856 DOI: 10.1021/acs.orglett.5b02718] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The transcriptional factor, hypoxia inducible factor-1 (HIF-1), is a promising target for cancer chemotherapy. From an actinomycete, verucopeptin (1) was identified as a HIF-1 signaling inhibitor. By a combination of chemical degradation and spectroscopic analyses, the absolute stereochemistry of metabolite 1 was determined to be 10R, 15S, 16S, 23S, 27S, 28R, 31S, 33S, 35R. Moreover, metabolite 1 was revealed to attenuate the HIF-1α and mTORC1 pathway, indicating that verucopeptin (1) would be a potent lead compound for anticancer chemotherapy.
Collapse
Affiliation(s)
- Aya Yoshimura
- Department of System Chemotherapy and Molecular Sciences, Division of Bioinformatics and Chemical Genomics, Graduate School of Pharmaceutical Sciences, Kyoto University , Sakyo-ku, Kyoto 606-8501, Japan
| | - Shinichi Nishimura
- Department of System Chemotherapy and Molecular Sciences, Division of Bioinformatics and Chemical Genomics, Graduate School of Pharmaceutical Sciences, Kyoto University , Sakyo-ku, Kyoto 606-8501, Japan
| | - Saori Otsuka
- Department of System Chemotherapy and Molecular Sciences, Division of Bioinformatics and Chemical Genomics, Graduate School of Pharmaceutical Sciences, Kyoto University , Sakyo-ku, Kyoto 606-8501, Japan
| | - Akira Hattori
- Department of System Chemotherapy and Molecular Sciences, Division of Bioinformatics and Chemical Genomics, Graduate School of Pharmaceutical Sciences, Kyoto University , Sakyo-ku, Kyoto 606-8501, Japan
| | - Hideaki Kakeya
- Department of System Chemotherapy and Molecular Sciences, Division of Bioinformatics and Chemical Genomics, Graduate School of Pharmaceutical Sciences, Kyoto University , Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
292
|
GAO JIAYIN, LIU MEIMEI, ZOU YITING, MAO MIN, SHEN TINGTING, ZHANG CHEN, SONG SHASHA, SUN MEILING, ZHANG SONG, WANG BEIDI, ZHU DALING, LI PEILING. Long non-coding RNA growth arrest-specific transcript 5 is involved in ovarian cancer cell apoptosis through the mitochondria-mediated apoptosis pathway. Oncol Rep 2015; 34:3212-21. [DOI: 10.3892/or.2015.4318] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 08/18/2015] [Indexed: 11/06/2022] Open
|
293
|
Effect of Nourishing "Yin" Removing "Fire" Chinese Herbal Mixture on Hypothalamic Mammalian Target of Rapamycin Expression during Onset of Puberty in Female Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:157846. [PMID: 26457106 PMCID: PMC4589583 DOI: 10.1155/2015/157846] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/03/2015] [Accepted: 09/06/2015] [Indexed: 11/17/2022]
Abstract
Aim. The present study aims to investigate the effects of nourishing "Yin" removing "Fire" (NYRF) Chinese herbal mixture on puberty onset and hypothalamic mTOR expression in female rats. Materials and Methods. Forty female 20-day-old Sprague-Dawley rats were randomly divided into Chinese herbal mixture (CHM) and normal saline (NS) groups. Rats in CHM and NS were treated with NYRF mixture and normal saline, respectively, from d22. Rats in each group were sacrificed on d28, d31, and d34. Serum luteinizing hormone (LH), follicle stimulating hormone (FSH), and estradiol (E2) levels were analyzed by ELISA. Hypothalamic mTOR mRNA expression levels were determined by RT-PCR and hypothalamic p-mTOR protein levels were assayed by western blot. Results. The vaginal opening time in CHM group was significantly delayed (P < 0.05). On d31, in comparison with NS group, the coefficients of uteri and ovaries, levels of serum LH and E2, and the expression levels of hypothalamic mTOR mRNA and p-mTOR protein were significantly lower in CHM group (P < 0.05). Conclusion. The mechanism by which the nourishing "Yin" removing "Fire" Chinese herbal mixture delays puberty onset may be associated with the inhibition of the hypothalamic mTOR signaling.
Collapse
|
294
|
Peng M, Ostrovsky J, Kwon YJ, Polyak E, Licata J, Tsukikawa M, Marty E, Thomas J, Felix CA, Xiao R, Zhang Z, Gasser DL, Argon Y, Falk MJ. Inhibiting cytosolic translation and autophagy improves health in mitochondrial disease. Hum Mol Genet 2015; 24:4829-47. [PMID: 26041819 PMCID: PMC4527487 DOI: 10.1093/hmg/ddv207] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 04/29/2015] [Accepted: 06/01/2015] [Indexed: 01/03/2023] Open
Abstract
Mitochondrial respiratory chain (RC) disease therapies directed at intra-mitochondrial pathology are largely ineffective. Recognizing that RC dysfunction invokes pronounced extra-mitochondrial transcriptional adaptations, particularly involving dysregulated translation, we hypothesized that translational dysregulation is itself contributing to the pathophysiology of RC disease. Here, we investigated the activities, and effects from direct inhibition, of a central translational regulator (mTORC1) and its downstream biological processes in diverse genetic and pharmacological models of RC disease. Our data identify novel mechanisms underlying the cellular pathogenesis of RC dysfunction, including the combined induction of proteotoxic stress, the ER stress response and autophagy. mTORC1 inhibition with rapamycin partially ameliorated renal disease in B6.Pdss2(kd/kd) mice with complexes I-III/II-III deficiencies, improved viability and mitochondrial physiology in gas-1(fc21) nematodes with complex I deficiency, and rescued viability across a variety of RC-inhibited human cells. Even more effective was probucol, a PPAR-activating anti-lipid drug that we show also inhibits mTORC1. However, directly inhibiting mTORC1-regulated downstream activities yielded the most pronounced and sustained benefit. Partial inhibition of translation by cycloheximide, or of autophagy by lithium chloride, rescued viability, preserved cellular respiratory capacity and induced mitochondrial translation and biogenesis. Cycloheximide also ameliorated proteotoxic stress via a uniquely selective reduction of cytosolic protein translation. RNAseq-based transcriptome profiling of treatment effects in gas-1(fc21) mutants provide further evidence that these therapies effectively restored altered translation and autophagy pathways toward that of wild-type animals. Overall, partially inhibiting cytosolic translation and autophagy offer novel treatment strategies to improve health across the diverse array of human diseases whose pathogenesis involves RC dysfunction.
Collapse
Affiliation(s)
- Min Peng
- Division of Human Genetics, Department of Pediatrics and Department of Genetics and
| | | | | | | | - Joseph Licata
- Division of Human Genetics, Department of Pediatrics and
| | - Mai Tsukikawa
- Division of Human Genetics, Department of Pediatrics and
| | - Eric Marty
- Division of Human Genetics, Department of Pediatrics and
| | - Jeffrey Thomas
- Division of Cell Pathology, Department of Pathology, The Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA and
| | - Carolyn A Felix
- Division of Oncology, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Rui Xiao
- Department of Biostatistics and Epidemiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Zhe Zhang
- Center for Biomedical Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Yair Argon
- Division of Cell Pathology, Department of Pathology, The Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA and
| | - Marni J Falk
- Division of Human Genetics, Department of Pediatrics and
| |
Collapse
|
295
|
Huang WR, Chiu HC, Liao TL, Chuang KP, Shih WL, Liu HJ. Avian Reovirus Protein p17 Functions as a Nucleoporin Tpr Suppressor Leading to Activation of p53, p21 and PTEN and Inactivation of PI3K/AKT/mTOR and ERK Signaling Pathways. PLoS One 2015; 10:e0133699. [PMID: 26244501 PMCID: PMC4526660 DOI: 10.1371/journal.pone.0133699] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 06/30/2015] [Indexed: 11/19/2022] Open
Abstract
Avian reovirus (ARV) protein p17 has been shown to regulate cell cycle and autophagy by activation of p53/PTEN pathway; nevertheless, it is still unclear how p53 and PTEN are activated by p17. Here, we report for the first time that p17 functions as a nucleoporin Tpr suppressor that leads to p53 nuclear accumulation and consequently activates p53, p21, and PTEN. The nuclear localization signal (119IAAKRGRQLD128) of p17 has been identified for Tpr binding. This study has shown that Tpr suppression occurs by p17 interacting with Tpr and by reducing the transcription level of Tpr, which together inhibit Tpr function. In addition to upregulation of PTEN by activation of p53 pathway, this study also suggests that ARV protein p17 acts as a positive regulator of PTEN. ARV p17 stabilizes PTEN by stimulating phosphorylation of cytoplasmic PTEN and by elevating Rak-PTEN association to prevent it from E3 ligase NEDD4-1 targeting. To activate PTEN, p17 is able to promote β-arrestin-mediated PTEN translocation from the cytoplasm to the plasma membrane via a Rock-1-dependent manner. The accumulation of p53 in the nucleus induces the PTEN- and p21-mediated downregulation of cyclin D1 and CDK4. Furthermore, Tpr and CDK4 knockdown increased virus production in contrast to depletion of p53, PTEN, and LC3 reducing virus yield. Taken together, our data suggest that p17-mediated Tpr suppression positively regulates p53, PTEN, and p21 and negatively regulates PI3K/AKT/mTOR and ERK signaling pathways, both of which are beneficial for virus replication.
Collapse
Affiliation(s)
- Wei-Ru Huang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402, Taiwan
| | - Hung-Chuan Chiu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402, Taiwan
| | - Tsai-Ling Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, 402, Taiwan
| | - Kuo-Pin Chuang
- Graduate Institute of Animal Vaccine Technology, National Pingtung University of Science and Technology, Pingtung, 912, Taiwan
| | - Wing-Ling Shih
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung, 912, Taiwan
| | - Hung-Jen Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402, Taiwan
- Agricultural Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan
- * E-mail:
| |
Collapse
|
296
|
Ribosomal Protein S6 Phosphorylation: Four Decades of Research. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 320:41-73. [PMID: 26614871 DOI: 10.1016/bs.ircmb.2015.07.006] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The phosphorylation of ribosomal protein S6 (rpS6) has been described for the first time about four decades ago. Since then, numerous studies have shown that this modification occurs in response to a wide variety of stimuli on five evolutionarily conserved serine residues. However, despite a large body of information on the respective kinases and the signal transduction pathways, the physiological role of rpS6 phosphorylation remained obscure until genetic manipulations were applied in both yeast and mammals in an attempt to block this modification. Thus, studies based on both mice and cultured cells subjected to disruption of the genes encoding rpS6 and the respective kinases, as well as the substitution of the phosphorylatable serine residues in rpS6, have laid the ground for the elucidation of the multiple roles of this protein and its posttranslational modification. This review focuses primarily on newly identified kinases that phosphorylate rpS6, pathways that transduce various signals into rpS6 phosphorylation, and the recently established physiological functions of this modification. It should be noted, however, that despite the significant progress made in the last decade, the molecular mechanism(s) underlying the diverse effects of rpS6 phosphorylation on cellular and organismal physiology are still poorly understood.
Collapse
|
297
|
Mammalian target of rapamycin complex 1 (mTORC1) Is required for mouse spermatogonial differentiation in vivo. Dev Biol 2015; 407:90-102. [PMID: 26254600 DOI: 10.1016/j.ydbio.2015.08.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/02/2015] [Accepted: 08/03/2015] [Indexed: 12/19/2022]
Abstract
Spermatogonial stem cells (SSCs) must balance self-renewal with production of transit-amplifying progenitors that differentiate in response to retinoic acid (RA) before entering meiosis. This self-renewal vs. differentiation spermatogonial fate decision is critical for maintaining tissue homeostasis, as imbalances cause spermatogenesis defects that can lead to human testicular cancer or infertility. A great deal of effort has been exerted to understand how the SSC population is maintained. In contrast, little is known about the essential program of differentiation initiated by retinoic acid (RA) that precedes meiosis, and the pathways and proteins involved are poorly defined. We recently reported a novel role for RA in stimulating the PI3/AKT/mTOR kinase signaling pathway to activate translation of repressed mRNAs such as Kit. Here, we examined the requirement for mTOR complex 1 (mTORC1) in mediating the RA signal to direct spermatogonial differentiation in the neonatal testis. We found that in vivo inhibition of mTORC1 by rapamycin blocked spermatogonial differentiation, which led to an accumulation of undifferentiated spermatogonia. In addition, rapamycin also blocked the RA-induced translational activation of mRNAs encoding KIT, SOHLH1, and SOHLH2 without affecting expression of STRA8. These findings highlight dual roles for RA in germ cell development - transcriptional activation of genes, and kinase signaling to stimulate translation of repressed messages required for spermatogonial differentiation.
Collapse
|
298
|
Wang X, Lin Z, Liu H, Lu W, Liu H, Lu W, Qian Y. [The traditional Chinese medicine Lulongzaisheng decoction may affect the expression of Foxp3 by regulation of Akt and Stat3 phosphorylation in CD4+CD25+ regulatory T cells from severe aplastic anemia mouse models]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:689-91. [PMID: 26462641 PMCID: PMC7348270 DOI: 10.3760/cma.j.issn.0253-2727.2015.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Indexed: 12/03/2022]
Affiliation(s)
- Xuli Wang
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Zenghua Lin
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Wei Lu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Haiyan Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Wenping Lu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yu Qian
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
299
|
Wang W, Wang H, Geng QX, Wang HT, Miao W, Cheng B, Zhao D, Song GM, Leanne G, Zhao Z. Augmentation of autophagy by atorvastatin via Akt/mTOR pathway in spontaneously hypertensive rats. Hypertens Res 2015. [PMID: 26224487 DOI: 10.1038/hr.2015.85] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Autophagy is activated in hypertension-induced cardiac hypertrophy. However, the mechanisms and significance of an activated autophagy are not clear. This study was designed to determine the role of atorvastatin (ATO) in cardiac autophagy and associated benefits on cardiac remodeling and left ventricular function in spontaneously hypertensive rats (SHRs). Twenty-eight male SHRs at 8 weeks of age were randomized to treatment with vehicle (saline solution; SHR+V) or ATO (SHR+ATO; 50 mg kg(-1) per day) for 6 or 12 months. Age-matched male Wistar-Kyoto (WKY) rats were used as normotensive controls. Cardiac magnetic resonance was used to evaluate cardiac function and structure. Compared with WKY rats, SHRs showed significant left ventricle (LV) dysfunction, remodeling and increases in cardiomyocyte size, which were all attenuated by 6 and 12 months of ATO treatment. Compared with WKY rats, autophagy was activated in the hearts of SHRs and this effect was amplified by chronic ATO treatment, particularly following 12 months of treatment. Protein expression levels of microtubule-associated protein-1 light chain 3-II and beclin-1, the biomarkers of an activated cardiac autophagy, were significantly elevated in ATO-treated versus vehicle-treated SHRs and control WKY rats. Cardiac Akt and phosphorylated mammalian target of rapamycin (mTOR) expression were also increased in the hearts of SHR versus WKY rats, and this effect was attenuated by ATO treatment. These findings suggest that ATO-mediated improvements in LV function and structure in SHRs may be, in part, through its regulation of cardiac autophagy via the Akt/mTOR pathway.
Collapse
Affiliation(s)
- Wei Wang
- Department of Cardiovascular Surgery, Shandong University Qilu Hospital, Shandong, China.,Department of Cardiology, Shandong Provincial Chest Hospital, Shandong, China
| | - Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Qing-Xin Geng
- Department of Cardiology, Jinan Central Hospital, Affiliated with Shandong University, Shandong, China
| | - Hua-Ting Wang
- Department of Cardiology, Jinan Central Hospital, Affiliated with Shandong University, Shandong, China
| | - Wei Miao
- Department of Cardiology, Jinan Central Hospital, Affiliated with Shandong University, Shandong, China
| | - Bo Cheng
- Department of Cardiology, Shandong Provincial Chest Hospital, Shandong, China
| | - Di Zhao
- Shandong University of Traditional Chinese Medicine, Shandong, China
| | - Guang-Min Song
- Department of Cardiovascular Surgery, Shandong University Qilu Hospital, Shandong, China
| | - Groban Leanne
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Zhuo Zhao
- Department of Cardiology, Jinan Central Hospital, Affiliated with Shandong University, Shandong, China
| |
Collapse
|
300
|
Herboso L, Oliveira MM, Talamillo A, Pérez C, González M, Martín D, Sutherland JD, Shingleton AW, Mirth CK, Barrio R. Ecdysone promotes growth of imaginal discs through the regulation of Thor in D. melanogaster. Sci Rep 2015. [PMID: 26198204 PMCID: PMC4510524 DOI: 10.1038/srep12383] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Animals have a determined species-specific body size that results from the combined action of hormones and signaling pathways regulating growth rate and duration. In Drosophila, the steroid hormone ecdysone controls developmental transitions, thereby regulating the duration of the growth period. Here we show that ecdysone promotes the growth of imaginal discs in mid-third instar larvae, since imaginal discs from larvae with reduced or no ecdysone synthesis are smaller than wild type due to smaller and fewer cells. We show that insulin-like peptides are produced and secreted normally in larvae with reduced ecdysone synthesis, and upstream components of insulin/insulin-like signaling are activated in their discs. Instead, ecdysone appears to regulate the growth of imaginal discs via Thor/4E-BP, a negative growth regulator downstream of the insulin/insulin-like growth factor/Tor pathways. Discs from larvae with reduced ecdysone synthesis have elevated levels of Thor, while mutations in Thor partially rescue their growth. The regulation of organ growth by ecdysone is evolutionarily conserved in hemimetabolous insects, as shown by our results obtained using Blattella germanica. In summary, our data provide new insights into the relationship between components of the insulin/insulin-like/Tor and ecdysone pathways in the control of organ growth.
Collapse
Affiliation(s)
- Leire Herboso
- CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Bizkaia, Spain
| | - Marisa M Oliveira
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal
| | - Ana Talamillo
- CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Bizkaia, Spain
| | - Coralia Pérez
- CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Bizkaia, Spain
| | - Monika González
- CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Bizkaia, Spain
| | - David Martín
- Institute of Evolutionary Biology, CSIC-Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | | | | | - Christen K Mirth
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal
| | - Rosa Barrio
- CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Bizkaia, Spain
| |
Collapse
|