7201
|
Kumar A, Das G, Bose B. Recombinant receptor-binding domain of diphtheria toxin increases the potency of curcumin by enhancing cellular uptake. Mol Pharm 2013; 11:208-17. [PMID: 24224661 DOI: 10.1021/mp400378x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Diphtheria toxin (DT) binds to a specific cell surface receptor, gets internalized, and causes cytotoxicity through its catalytic domain. The toxicity of DT is used in several therapeutic molecules. Here, we have exploited the receptor-binding ability of DT to increase cellular uptake of curcumin, a hydrophobic molecule with low bioavailability and cellular uptake. We have expressed only the receptor-binding domain of DT (RDT) in Escherichia coli. Purified RDT binds to the receptor with an affinity equivalent to that of full-length DT. It also binds to curcumin forming a curcumin-RDT complex, and this increases the fluorescence intensity and fluorescence lifetime of curcumin. The curcumin-RDT complex binds to the receptor and associates with human glioblastoma cells (U-87 MG) expressing the receptor. The cellular uptake of curcumin is higher for the curcumin-RDT complex than curcumin alone. This increase in uptake enhances the antiproliferative effect of curcumin and induces apoptosis of these cells even at a lower dose.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Biotechnology and ‡Department of Chemistry, Indian Institute of Technology Guwahati , Guwahati, 781039 India
| | | | | |
Collapse
|
7202
|
Pourhassan-Moghaddam M, Rahmati-Yamchi M, Akbarzadeh A, Daraee H, Nejati-Koshki K, Hanifehpour Y, Joo SW. Protein detection through different platforms of immuno-loop-mediated isothermal amplification. NANOSCALE RESEARCH LETTERS 2013; 8:485. [PMID: 24237767 PMCID: PMC3835475 DOI: 10.1186/1556-276x-8-485] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 11/05/2013] [Indexed: 05/14/2023]
Abstract
Different immunoassay-based methods have been devised to detect protein targets. These methods have some challenges that make them inefficient for assaying ultra-low-amounted proteins. ELISA, iPCR, iRCA, and iNASBA are the common immunoassay-based methods of protein detection, each of which has specific and common technical challenges making it necessary to introduce a novel method in order to avoid their problems for detection of target proteins. Here we propose a new method nominated as 'immuno-loop-mediated isothermal amplification' or 'iLAMP'. This new method is free from the problems of the previous methods and has significant advantages over them. In this paper we also offer various configurations in order to improve the applicability of this method in real-world sample analyses. Important potential applications of this method are stated as well.
Collapse
Affiliation(s)
- Mohammad Pourhassan-Moghaddam
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51656, Iran
- Ian Wark Research Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Mohammad Rahmati-Yamchi
- Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 51656, Iran
| | - Abolfazl Akbarzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51656, Iran
| | - Hadis Daraee
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51656, Iran
- Ian Wark Research Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Kazem Nejati-Koshki
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51656, Iran
| | - Younes Hanifehpour
- School of Mechanical Engineering, WCU Nanoresearch Center, Yeungnam University, Gyeongsan 712-749, South Korea
| | - Sang Woo Joo
- School of Mechanical Engineering, WCU Nanoresearch Center, Yeungnam University, Gyeongsan 712-749, South Korea
| |
Collapse
|
7203
|
Histone deacetylase inhibitors decrease intra-abdominal adhesions with one intraoperative dose by reducing peritoneal fibrin deposition pathways. Surgery 2013; 155:234-44. [PMID: 24239397 DOI: 10.1016/j.surg.2013.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 08/13/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND We previously demonstrated that postoperative peritoneal injury and inflammation contribute to adhesiogenesis. Recent evidence suggests that in addition to their role of interfering with the acetylation status of nuclear histone proteins, histone deacetylase inhibitors (HDACIs) including valproic acid (VPA) can target nonhistone proteins to resolve inflammation and modulate immune cells. We hypothesized that HDACIs could reduce adhesions. METHODS Seventy-two rats underwent laparotomy with creation of 6 peritoneal ischemic buttons to induce adhesions. A single intraperitoneal (IP) dose of 50 mg/kg VPA was administered intraoperatively, whereas controls received vehicle. To evaluate the timing, 25 rats underwent ischemic button creation with either an intraoperative or a delayed IP dose of VPA at 1, 3, or 6 hours postoperatively. On postoperative day 7, adhesions were quantified. To investigate mechanisms, ischemic buttons were created in 24 rats and either VPA or saline was administered in 1 intraoperative dose. At 3 or 24 hours later, peritoneal fluid was collected and fibrinolytic activity measured. Alternatively, button tissue was collected 30 minutes postoperatively to measure tissue factor, fibrinogen, and vascular endothelial growth factor (VEGF) by real-time polymerase chain reaction or Western blot. RESULTS A single intraoperative dose of VPA reduced adhesions by 50% relative to controls (P < .001). Delayed dosing did not reduce adhesions. In operated animals, peritoneal fibrinolytic activity was not different between groups. Tissue factor mRNA was downregulated by 50% (P = .02) and protein by 34% (P < .01) in animals administered VPA versus saline. VPA decreased fibrinogen protein by 56% and VEGF protein by 25% compared with saline (P = .03). CONCLUSION These findings suggest that VPA rapidly reduces the extravasation of key adhesiogenic substrates into the peritoneum. A single, intraoperative intervention provides an ideal dosing strategy and indicates an exciting new role for HDACIs in adhesion prevention.
Collapse
|
7204
|
Inflammatory and microenvironmental factors involved in breast cancer progression. Arch Pharm Res 2013; 36:1419-31. [PMID: 24222504 DOI: 10.1007/s12272-013-0271-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 10/21/2013] [Indexed: 12/20/2022]
Abstract
The primary reason for the high mortality rate of breast cancer is metastasis, which can result in a poor survival rate. The tumor environment is important for promotion and invasion of cancer cells. Recent studies have shown that inflammation is associated with breast cancer. Therefore, it is important to investigate the role of the inflammatory and microenvironment in breast cancer progression and metastasis. The present review summarizes some of the markers for inflammation and breast cancer invasion, which may aid in the design of an appropriate therapy for metastatic breast cancer. The following four inflammatory markers are discussed in this review: (1) Tumor associated macrophages (TAMs); (2) Matrix metalloproteinases (MMPs); (3) Sphingosine 1-phosphate (S1P); (4) C-reactive protein (CRP). TAMs are commonly found in breast cancer patients, and high infiltration is positively correlated with poor prognosis and low survival rate. MMPs are well-known for their roles in the degradation of ECM components when cancer cells invade and migrate. MMPs are also associated with inflammation through recruitment of a variety of stromal cells such as fibroblasts and leukocytes. S1P is an inflammatory lipid and is involved in various cellular processes such as proliferation, survival, and migration. Recent studies indicate that S1P participates in breast cancer invasion in various ways. CRP is used clinically to indicate the outcome of cancer patients as well as acute inflammatory status. This review summarizes the current understanding on the role of S1P in CRP expression which promotes the breast epithelial cell invasion, suggesting a specific mechanism linking inflammation and breast cancer. The present review might be useful for understanding the relationship between inflammation and breast cancer for the development of pharmacological interventions that may control the primary molecules involved in the breast cancer microenvironment.
Collapse
|
7205
|
Di Lorenzo C, Coppola G, La Salvia V, Pierelli F. Nasal decongestant and chronic headache: a case of naphazoline overuse headache? F1000Res 2013; 2:237. [PMID: 25110575 PMCID: PMC4111119 DOI: 10.12688/f1000research.2-237.v1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2013] [Indexed: 11/29/2022] Open
Abstract
Background: Chronic headache is an incapacitating condition afflicting patients at least for 15 days per month. In the most cases it is developed as a consequence of an excessive use of symptomatic drugs. Case: Here we report the case of a 34 year-old man suffering from chronic headache possibly related to the overuse of naphazoline nitrate nasal decongestant, used to treat a supposed chronic sinusitis. However, the patient did not suffer from sinusitis, but from a medication overuse headache (ICHD-II 8.3; ICD-10 44.41) that appeared to be due to excessive use of naphazoline. Conclusion: The use of naphazoline nitrate may result in an analgesic effect upon first use, through activation of adrenergic and opioidergic systems, followed by a pro-migraine effect via a late induction of an inflammatory cascade, modulated by nitric oxide and arachidonic acid. The observation that naphazoline detoxification relieved the patient’s headache, indicates that prolonged use of naphazoline may cause chronic headaches. Therefore, physicians should ask for details on the use of nasal decongestants in patients complaining of chronic headache, as they could potentially be suffering from a medication-overuse headache.
Collapse
Affiliation(s)
| | - Gianluca Coppola
- Department of Neurophysiology of Vision and Neurophthalmology, G.B. Bietti Foundation IRCCS, Rome, 00100, Italy
| | - Valeria La Salvia
- Department of Medical and Surgical Sciences and Biotechnologies and ICOT, "Sapienza" University of Rome Polo Pontino, Latina, 04100, Italy
| | | |
Collapse
|
7206
|
Lajiness JD, Conway SJ. Origin, development, and differentiation of cardiac fibroblasts. J Mol Cell Cardiol 2013; 70:2-8. [PMID: 24231799 DOI: 10.1016/j.yjmcc.2013.11.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 10/23/2013] [Accepted: 11/04/2013] [Indexed: 01/14/2023]
Abstract
Cardiac fibroblasts are the most abundant cell in the mammalian heart. While they have been historically underappreciated in terms of their functional contributions to cardiac development and physiology, they and their activated form, myofibroblasts, are now known to play key roles in both development and disease through structural, paracrine, and electrical interactions with cardiomyocytes. The lack of specific markers for fibroblasts currently convolutes the study of this dynamic cell lineage, but advances in marker analysis and lineage mapping technologies are continuously being made. Understanding how to best utilize these tools, both individually and in combination, will help to elucidate the functional significance of fibroblast-cardiomyocyte interactions in vivo. Here we review what is currently known about the diverse roles played by cardiac fibroblasts and myofibroblasts throughout development and periods of injury with the intent of emphasizing the duality of their nature. This article is part of a Special Issue entitled "Myocyte-Fibroblast Signalling in Myocardium ".
Collapse
Affiliation(s)
- Jacquelyn D Lajiness
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Simon J Conway
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
7207
|
Zhang B, Elmabsout AA, Khalaf H, Basic VT, Jayaprakash K, Kruse R, Bengtsson T, Sirsjö A. The periodontal pathogen Porphyromonas gingivalis changes the gene expression in vascular smooth muscle cells involving the TGFbeta/Notch signalling pathway and increased cell proliferation. BMC Genomics 2013; 14:770. [PMID: 24209892 PMCID: PMC3827841 DOI: 10.1186/1471-2164-14-770] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 11/05/2013] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Porphyromonas gingivalis is a gram-negative bacterium that causes destructive chronic periodontitis. In addition, this bacterium is also involved in the development of cardiovascular disease. The aim of this study was to investigate the effects of P. gingivalis infection on gene and protein expression in human aortic smooth muscle cells (AoSMCs) and its relation to cellular function. RESULTS AoSMCs were exposed to viable P. gingivalis for 24 h, whereafter confocal fluorescence microscopy was used to study P. gingivalis invasion of AoSMCs. AoSMCs proliferation was evaluated by neutral red assay. Human genome microarray, western blot and ELISA were used to investigate how P. gingivalis changes the gene and protein expression of AoSMCs. We found that viable P. gingivalis invades AoSMCs, disrupts stress fiber structures and significantly increases cell proliferation. Microarray results showed that, a total of 982 genes were identified as differentially expressed with the threshold log2 fold change > |1| (adjust p-value <0.05). Using bioinformatic data mining, we demonstrated that up-regulated genes are enriched in gene ontology function of positive control of cell proliferation and down-regulated genes are enriched in the function of negative control of cell proliferation. The results from pathway analysis revealed that all the genes belonging to these two categories induced by P. gingivalis were enriched in 25 pathways, including genes of Notch and TGF-beta pathways. CONCLUSIONS This study demonstrates that P. gingivalis is able to invade AoSMCs and stimulate their proliferation. The activation of TGF-beta and Notch signaling pathways may be involved in the bacteria-mediated proliferation of AoSMCs. These findings further support the association between periodontitis and cardiovascular diseases.
Collapse
Affiliation(s)
- Boxi Zhang
- Department of Clinical Medicine, School of Health and Medical Sciences, Örebro University, Örebro, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
7208
|
Engler FA, Zheng B, Balthasar JP. Investigation of the influence of nephropathy on monoclonal antibody disposition: a pharmacokinetic study in a mouse model of diabetic nephropathy. Pharm Res 2013; 31:1185-93. [PMID: 24203494 DOI: 10.1007/s11095-013-1241-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 10/20/2013] [Indexed: 01/06/2023]
Abstract
PURPOSE This study employed a mouse model to evaluate the effects of diabetic nephropathy on the pharmacokinetics of 8C2, a murine monoclonal antibody (mAb). METHODS Streptozotocin (STZ) was administered to mice to induce diabetic nephropathy (125 mg/kg/day × 2). Mice were grouped (n = 8-10) based on time after STZ-treatment (control, 1, 2, 3, 4, or 6 weeks), and injected intravenously with 10 mg/kg 8C2. Blood samples were collected up to 7 days, and 8C2 plasma concentrations were determined via immunoassay. Inulin clearance and urinary albumin excretion rate (UAE) were determined to assess renal function. RESULTS UAE, inulin clearance, and 8C2 clearance increased significantly following STZ. Comparing control and 6 week STZ-treatment groups, UAE and inulin clearance increased from 25.7 ± 3.3 to 99.3 ± 13.7 μg/day, and from 421 ± 31 to 584 ± 78 μl/min. 8C2 clearance increased from 121 ± 12.5 to 228 ± 61 μl/hr/kg (p < 0.01). 8C2 clearance was highly correlated with UAE (r(2): 0.731). Inclusion of UAE as a covariate in population modeling explained significant residual variability in 8C2 clearance. CONCLUSIONS The clearance of 8C2 increased significantly in STZ-treated mice. Population pharmacokinetic modeling suggests that UAE has potential for use in predicting mAb clearance in subjects with diabetic nephropathy, possibly assisting in the individualization of mAb dosing.
Collapse
Affiliation(s)
- Frank A Engler
- Department of Pharmaceutical Sciences School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 452 Kapoor Hall, Buffalo, New York, 14260, USA
| | | | | |
Collapse
|
7209
|
Yang Z, Xu Y, Xu L, Maccauro G, Rossi B, Chen Y, Li H, Zhang J, Sun H, Yang Y, Xu D, Liu X. Regulation of autophagy via PERK-eIF2α effectively relieve the radiation myelitis induced by iodine-125. PLoS One 2013; 8:e76819. [PMID: 24223705 PMCID: PMC3818370 DOI: 10.1371/journal.pone.0076819] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 08/28/2013] [Indexed: 11/18/2022] Open
Abstract
Radiation myelitis is the most serious complication in clinical radiotherapy for spinal metastases. We previously showed that 125I brachytherapy induced apoptosis of spinal cord neurons accompanied by autophagy. In this study, we further investigated the mechanism by which 125I radiation triggered autophagy in neural cells. We found that autophagy induced by 125I radiation was involved in endoplasmic reticulum (ER) stress and mainly dependent on PERK-eIF2α pathway. The expressions of LC3II, ATG12 and PI3K were significantly suppressed in PERK knockout neural cells. Meanwhile, the expressions of phosphorylated-Akt s473 and caspase3/8 all significantly increased in neural cells transfected with a PERK siRNA and which enhanced apoptosis of neurons after 125I radiation. The results were consistent with that by MTT and Annexin-FITC/PT staining. In annimal model of banna pigs with radiation myelitis caused by 125I brachytherapy, we have successfully decreased PERK expression by intrathecal administration of the lentivirus vector. The apoptosis rate was significantly higher than that in control group and which deteriorated radiation myelitis of banna pigs. Thus, autophagy caused by 125I radiation was mainly as an attempt of cell survival at an early stage, but it would be a self-destructive process and promoted the process of apoptosis and necrosis radiated by 125I for more than 72 hours. The study would be useful and helpful to maximize efficiency of radiation therapy in clinical therapy.
Collapse
Affiliation(s)
- Zuozhang Yang
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P. R. China
- Department of Orthopaedic Oncology, Agostino Gemelli Hospital, Catholic University of Rome, Largo Francesco Vito 1, Rome, Italy
- * E-mail:
| | - Yongqing Xu
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Lei Xu
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Giulio Maccauro
- Department of Orthopaedic Oncology, Agostino Gemelli Hospital, Catholic University of Rome, Largo Francesco Vito 1, Rome, Italy
| | - Barbara Rossi
- Department of Orthopaedic Oncology, Agostino Gemelli Hospital, Catholic University of Rome, Largo Francesco Vito 1, Rome, Italy
| | - Yanjin Chen
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Hongjun Li
- Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Science, Kunming, Yunnan, P. R. China
| | - Jing Zhang
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Hongpu Sun
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Yihao Yang
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Da Xu
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P. R. China
| | - Xuefeng Liu
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P. R. China
| |
Collapse
|
7210
|
Scotland S, Saland E, Skuli N, de Toni F, Boutzen H, Micklow E, Sénégas I, Peyraud R, Peyriga L, Théodoro F, Dumon E, Martineau Y, Danet-Desnoyers G, Bono F, Rocher C, Levade T, Manenti S, Junot C, Portais JC, Alet N, Récher C, Selak MA, Carroll M, Sarry JE. Mitochondrial energetic and AKT status mediate metabolic effects and apoptosis of metformin in human leukemic cells. Leukemia 2013; 27:2129-38. [PMID: 23568147 PMCID: PMC10869165 DOI: 10.1038/leu.2013.107] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 03/29/2013] [Accepted: 04/02/2013] [Indexed: 12/16/2022]
Abstract
Previous reports demonstrate that metformin, an anti-diabetic drug, can decrease the risk of cancer and inhibit cancer cell growth. However, its mechanism in cancer cells is still unknown. Metformin significantly blocks cell cycle and inhibits cell proliferation and colony formation of leukemic cells. However, the apoptotic response to metformin varies. Furthermore, daily treatment with metformin induces apoptosis and reduces tumor growth in vivo. While metformin induces early and transient activation of AMPK, inhibition of AMPKα1/2 does not abrogate anti-proliferative or pro-apoptotic effects of metformin. Metformin decreases electron transport chain complex I activity, oxygen consumption and mitochondrial ATP synthesis, while stimulating glycolysis for ATP and lactate production, pentose phosphate pathway for purine biosynthesis, fatty acid metabolism, as well as anaplerotic and mitochondrial gene expression. Importantly, leukemic cells with high basal AKT phosphorylation, glucose consumption or glycolysis exhibit a markedly reduced induction of the Pasteur effect in response to metformin and are resistant to metformin-induced apoptosis. Accordingly, glucose starvation or treatment with deoxyglucose or an AKT inhibitor induces sensitivity to metformin. Overall, metformin elicits reprogramming of intermediary metabolism leading to inhibition of cell proliferation in all leukemic cells and apoptosis only in leukemic cells responding to metformin with AKT phosphorylation and a strong Pasteur effect.
Collapse
Affiliation(s)
- S Scotland
- INSERM, U1037, Cancer Research Center of Toulouse, CHU Purpan, Toulouse, France
- Université de Paul Sabatier, Toulouse III, Toulouse, France
| | - E Saland
- INSERM, U1037, Cancer Research Center of Toulouse, CHU Purpan, Toulouse, France
- Université de Paul Sabatier, Toulouse III, Toulouse, France
| | - N Skuli
- INSERM, U1037, Cancer Research Center of Toulouse, CHU Purpan, Toulouse, France
- Université de Paul Sabatier, Toulouse III, Toulouse, France
| | - F de Toni
- INSERM, U1037, Cancer Research Center of Toulouse, CHU Purpan, Toulouse, France
- Université de Paul Sabatier, Toulouse III, Toulouse, France
| | - H Boutzen
- INSERM, U1037, Cancer Research Center of Toulouse, CHU Purpan, Toulouse, France
- Université de Paul Sabatier, Toulouse III, Toulouse, France
| | - E Micklow
- Department of Medicine, Division of Hematology & Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - I Sénégas
- Sanofi R&D, Early-to-Candidate Unit, Toulouse, France
| | - R Peyraud
- Université de Toulouse, INSA, UPS, INP, LISBP, Toulouse, France
- INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, Toulouse, France
| | - L Peyriga
- Université de Toulouse, INSA, UPS, INP, LISBP, Toulouse, France
- INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, Toulouse, France
- CNRS, UMR5504, Toulouse, France
| | - F Théodoro
- CEA/DSV/iBiTec-S/SPI, Bâtiment 136, CEA/Saclay, Gif-sur-Yvette, Fontenay-aux-Roses, France
| | - E Dumon
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, Bordeaux, France
| | - Y Martineau
- INSERM, U1037, Cancer Research Center of Toulouse, CHU Purpan, Toulouse, France
- Université de Paul Sabatier, Toulouse III, Toulouse, France
| | - G Danet-Desnoyers
- Department of Medicine, Division of Hematology & Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - F Bono
- Sanofi R&D, Early-to-Candidate Unit, Toulouse, France
| | - C Rocher
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, Bordeaux, France
| | - T Levade
- INSERM, U1037, Cancer Research Center of Toulouse, CHU Purpan, Toulouse, France
- Université de Paul Sabatier, Toulouse III, Toulouse, France
| | - S Manenti
- INSERM, U1037, Cancer Research Center of Toulouse, CHU Purpan, Toulouse, France
- Université de Paul Sabatier, Toulouse III, Toulouse, France
| | - C Junot
- CEA/DSV/iBiTec-S/SPI, Bâtiment 136, CEA/Saclay, Gif-sur-Yvette, Fontenay-aux-Roses, France
| | - J-C Portais
- Université de Toulouse, INSA, UPS, INP, LISBP, Toulouse, France
- INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, Toulouse, France
- CNRS, UMR5504, Toulouse, France
| | - N Alet
- Sanofi R&D, Early-to-Candidate Unit, Toulouse, France
| | - C Récher
- INSERM, U1037, Cancer Research Center of Toulouse, CHU Purpan, Toulouse, France
- Université de Paul Sabatier, Toulouse III, Toulouse, France
- Service d’Hématologie, CHU de Toulouse, Hôpital Purpan, Toulouse, France
| | - MA Selak
- Department of Medicine, Division of Hematology & Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - M Carroll
- Department of Medicine, Division of Hematology & Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - J-E Sarry
- INSERM, U1037, Cancer Research Center of Toulouse, CHU Purpan, Toulouse, France
- Université de Paul Sabatier, Toulouse III, Toulouse, France
| |
Collapse
|
7211
|
Abstract
Bioanalytical laboratories develop and validate ligand-binding assays (LBA) used to quantify the concentration of analytes of interest in various buffers and relevant biological matrices. The building blocks of LBA are reagents that recognize molecular and structural motifs on ligands, which are combined in various LBA formats to minimize biological matrix interferences and specifically detect and quantify the analyte of interest. The use of these LBA-requiring critical reagents, can span decades as programs mature to commercialization. Since critical reagents are generated mostly from biological systems, attention to their life cycle management, quality, characterization and sustainability are vital to the success of bioanalytical laboratories. Integrating de novo reagent generation, reagent biophysical characterization, LBA development, validation, and use, with reagent resupply processes leverages interdisciplinary activities and ensures smooth operations of a bioanalytical laboratory.
Collapse
|
7212
|
Damayanti NP, Fang Y, Parikh MR, Craig AP, Kirshner J, Irudayaraj J. Differentiation of cancer cells in two-dimensional and three-dimensional breast cancer models by Raman spectroscopy. JOURNAL OF BIOMEDICAL OPTICS 2013; 18:117008. [PMID: 24247810 PMCID: PMC3832300 DOI: 10.1117/1.jbo.18.11.117008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 10/10/2013] [Accepted: 10/15/2013] [Indexed: 05/25/2023]
Abstract
We demonstrate the first application of Raman spectroscopy in diagnosing nonmalignant, premalignant, malignant, and metastatic stages of breast cancer in a three-dimensional (3-D) cell culture model that closely mimics an in vivo environment. Comprehensive study comparing classification in two-dimensional (2-D) and 3-D cell models was performed using statistical methods composed of principal component analysis for exploratory analysis and outlier removal, partial least squares discriminant analysis, and elastic net regularized regression for classification. Our results show that Raman spectroscopy with an appropriate classification tool has excellent resolution to discriminate the four stages of breast cancer progression, with a near 100% accuracy for both 2-D and 3-D cell models. The diversity in chemical groups related to nucleic acids, proteins, and lipids, among other chemicals, were identified by appropriate peaks in the Raman spectra that correspond to the correct classification of the different stages of tumorigenesis model comprising of MCF10A, MCF10AneoT, MCF10CA1h, and MCF10CA1a cell lines. An explicit relationship between wavenumber and the stages of cancer progression was identified by the elastic net variable selection.
Collapse
Affiliation(s)
- Nur P. Damayanti
- Purdue University, Department of Agricultural and Biological Engineering, 225 S. University Street, West Lafayette, Indiana 47907
- Purdue University, Bindley Bioscience Center, West Lafayette, Indiana 47907
| | - Yi Fang
- Purdue University, Department of Agricultural and Biological Engineering, 225 S. University Street, West Lafayette, Indiana 47907
- Purdue University, Bindley Bioscience Center, West Lafayette, Indiana 47907
| | - Mukti R. Parikh
- Purdue University, Department of Biological Sciences, 915 W. State Street, West Lafayette, Indiana 47907
| | - Ana Paula Craig
- Purdue University, Bindley Bioscience Center, West Lafayette, Indiana 47907
| | - Julia Kirshner
- Purdue University, Department of Biological Sciences, 915 W. State Street, West Lafayette, Indiana 47907
| | - Joseph Irudayaraj
- Purdue University, Department of Agricultural and Biological Engineering, 225 S. University Street, West Lafayette, Indiana 47907
- Purdue University, Bindley Bioscience Center, West Lafayette, Indiana 47907
| |
Collapse
|
7213
|
Characterization of fragmented 3-phosphoinsitide-dependent protein kinase-1 (PDK1) by phosphosite-specific antibodies. Life Sci 2013; 93:700-6. [DOI: 10.1016/j.lfs.2013.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 07/31/2013] [Accepted: 09/05/2013] [Indexed: 11/18/2022]
|
7214
|
Laggai S, Simon Y, Ranssweiler T, Kiemer AK, Kessler SM. Rapid chromatographic method to decipher distinct alterations in lipid classes in NAFLD/NASH. World J Hepatol 2013; 5:558-567. [PMID: 24179615 PMCID: PMC3812458 DOI: 10.4254/wjh.v5.i10.558] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 09/10/2013] [Accepted: 10/12/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish a simple method to quantify lipid classes in liver diseases and to decipher the lipid profile in p62/IMP2-2/IGF2BP2-2 transgenic mice.
METHODS: Liver-specific overexpression of the insulin-like growth factor 2 mRNA binding protein p62/IMP2-2/IGF2BP2-2 was used as a model for steatosis. Steatohepatitis was induced by feeding a methionine-choline deficient diet. Steatosis was assessed histologically. For thin layer chromatographic analysis, lipids were extracted from freeze-dried tissues by hexane/2-propanol, dried, redissolved, and chromatographically separated by a two-solvent system. Dilution series of lipid standards were chromatographed, detected, and quantified. The detection was performed by either 2’,7’-dichlorofluoresceine or a sulfuric acid/ethanol mixture.
RESULTS: Histological analyses confirmed steatosis and steatohepatitis development. The extraction, chromatographic, and detection method showed high inter-assay reproducibility and allowed quantification of the different lipid classes. The analyses confirmed an increase of triglycerides and phosphatidylethanolamine and a decrease in phosphatidylcholine in the methionine-choline deficient diet. The method was used for the first time to asses the lipid classes induced in the p62-overexpressing mouse model and showed a significant increase in all detected lipid species with a prominent increase of triglycerides by 2-fold. Interestingly, the ratio of phosphatidylcholine to phosphatidylethanolamine was decreased, as previously suggested as a marker in the progression from steatosis to steatohepatitis.
CONCLUSION: The thin layer chromatography analysis allows a reliable quantification of lipid classes and provides detailed insight into the lipogenic effect of p62.
Collapse
|
7215
|
Shih PC, Yang YR, Wang RY. Effects of exercise intensity on spatial memory performance and hippocampal synaptic plasticity in transient brain ischemic rats. PLoS One 2013; 8:e78163. [PMID: 24205142 PMCID: PMC3808358 DOI: 10.1371/journal.pone.0078163] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 09/17/2013] [Indexed: 12/17/2022] Open
Abstract
Memory impairment is commonly noted in stroke survivors, and can lead to delay of functional recovery. Exercise has been proved to improve memory in adult healthy subjects. Such beneficial effects are often suggested to relate to hippocampal synaptic plasticity, which is important for memory processing. Previous evidence showed that in normal rats, low intensity exercise can improve synaptic plasticity better than high intensity exercise. However, the effects of exercise intensities on hippocampal synaptic plasticity and spatial memory after brain ischemia remain unclear. In this study, we investigated such effects in brain ischemic rats. The middle cerebral artery occlusion (MCAO) procedure was used to induce brain ischemia. After the MCAO procedure, rats were randomly assigned to sedentary (Sed), low-intensity exercise (Low-Ex), or high-intensity exercise (High-Ex) group. Treadmill training began from the second day post MCAO procedure, 30 min/day for 14 consecutive days for the exercise groups. The Low-Ex group was trained at the speed of 8 m/min, while the High-Ex group at the speed of 20 m/min. The spatial memory, hippocampal brain-derived neurotrophic factor (BDNF), synapsin-I, postsynaptic density protein 95 (PSD-95), and dendritic structures were examined to document the effects. Serum corticosterone level was also quantified as stress marker. Our results showed the Low-Ex group, but not the High-Ex group, demonstrated better spatial memory performance than the Sed group. Dendritic complexity and the levels of BDNF and PSD-95 increased significantly only in the Low-Ex group as compared with the Sed group in bilateral hippocampus. Notably, increased level of corticosterone was found in the High-Ex group, implicating higher stress response. In conclusion, after brain ischemia, low intensity exercise may result in better synaptic plasticity and spatial memory performance than high intensity exercise; therefore, the intensity is suggested to be considered during exercise training.
Collapse
Affiliation(s)
- Pei-Cheng Shih
- Department of Physical Therapy and Assistive Technology, National Yang-Ming University, Taipei, Taiwan
- Department of Physical Therapy, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yea-Ru Yang
- Department of Physical Therapy and Assistive Technology, National Yang-Ming University, Taipei, Taiwan
| | - Ray-Yau Wang
- Department of Physical Therapy and Assistive Technology, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
7216
|
Samokhvalov V, Alsaleh N, El-Sikhry HE, Jamieson KL, Chen CB, Lopaschuk DG, Carter C, Light PE, Manne R, Falck JR, Seubert JM. Epoxyeicosatrienoic acids protect cardiac cells during starvation by modulating an autophagic response. Cell Death Dis 2013; 4:e885. [PMID: 24157879 PMCID: PMC3920965 DOI: 10.1038/cddis.2013.418] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 09/21/2013] [Accepted: 09/26/2013] [Indexed: 12/25/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) are cytochrome P450 epoxygenase metabolites of arachidonic acid involved in regulating pathways promoting cellular protection. We have previously shown that EETs trigger a protective response limiting mitochondrial dysfunction and reducing cellular death. Considering it is unknown how EETs regulate cell death processes, the major focus of the current study was to investigate their role in the autophagic response of HL-1 cells and neonatal cardiomyocytes (NCMs) during starvation. We employed a dual-acting synthetic analog UA-8 (13-(3-propylureido)tridec-8-enoic acid), possessing both EET-mimetic and soluble epoxide hydrolase (sEH) inhibitory properties, or 14,15-EET as model EET molecules. We demonstrated that EETs significantly improved viability and recovery of starved cardiac cells, whereas they lowered cellular stress responses such as caspase-3 and proteasome activities. Furthermore, treatment with EETs resulted in preservation of mitochondrial functional activity in starved cells. The protective effects of EETs were abolished by autophagy-related gene 7 (Atg7) short hairpin RNA (shRNA) or pharmacological inhibition of autophagy. Mechanistic evidence demonstrated that sarcolemmal ATP-sensitive potassium channels (pmKATP) and enhanced activation of AMP-activated protein kinase (AMPK) played a crucial role in the EET-mediated effect. Our data suggest that the protective effects of EETs involve regulating the autophagic response, which results in a healthier pool of mitochondria in the starved cardiac cells, thereby representing a novel mechanism of promoting survival of cardiac cells. Thus, we provide new evidence highlighting a central role of the autophagic response in linking EETs with promoting cell survival during deep metabolic stress such as starvation.
Collapse
Affiliation(s)
- V Samokhvalov
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7217
|
James AD, Chan A, Erice O, Siriwardena AK, Bruce JIE. Glycolytic ATP fuels the plasma membrane calcium pump critical for pancreatic cancer cell survival. J Biol Chem 2013; 288:36007-19. [PMID: 24158437 PMCID: PMC3861649 DOI: 10.1074/jbc.m113.502948] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Pancreatic cancer is an aggressive cancer with poor prognosis and limited treatment options. Cancer cells rapidly proliferate and are resistant to cell death due, in part, to a shift from mitochondrial metabolism to glycolysis. We hypothesized that this shift is important in regulating cytosolic Ca2+ ([Ca2+]i), as the ATP-dependent plasma membrane Ca2+ ATPase (PMCA) is critical for maintaining low [Ca2+]i and thus cell survival. The present study aimed to determine the relative contribution of mitochondrial versus glycolytic ATP in fuelling the PMCA in human pancreatic cancer cells. We report that glycolytic inhibition induced profound ATP depletion, PMCA inhibition, [Ca2+]i overload, and cell death in PANC1 and MIA PaCa-2 cells. Conversely, inhibition of mitochondrial metabolism had no effect, suggesting that glycolytic ATP is critical for [Ca2+]i homeostasis and thus survival. Targeting the glycolytic regulation of the PMCA may, therefore, be an effective strategy for selectively killing pancreatic cancer while sparing healthy cells.
Collapse
Affiliation(s)
- Andrew D James
- From the Faculty of Life Sciences, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, United Kingdom and
| | | | | | | | | |
Collapse
|
7218
|
Zwolak I. Vanadium carcinogenic, immunotoxic and neurotoxic effects: a review ofin vitrostudies. Toxicol Mech Methods 2013; 24:1-12. [DOI: 10.3109/15376516.2013.843110] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
7219
|
Nauczyciel C, Robic S, Dondaine T, Verin M, Robert G, Drapier D, Naudet F, Millet B. The nucleus accumbens: a target for deep brain stimulation in resistant major depressive disorder. J Mol Psychiatry 2013; 1:17. [PMID: 26019865 PMCID: PMC4445576 DOI: 10.1186/2049-9256-1-17] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 09/18/2013] [Indexed: 01/21/2023] Open
Abstract
Objective This review aimed to investigate the therapeutic potential of Deep Brain Stimulation (DBS) for treating resistant Major Depressive Disorder (MDD). We explored the role of Nucleus accumbens (Nac) as a target for treatment. Method We made a systematic review of all studies examining the mechanisms of action of high frequency brain stimulation and the pathophysiology of MDD. We also reported all the studies exploring the therapeutic potential of DBS in MDD. Results As a central relay-structure, the Nac seems to play a central role in MDD symptomatology. We investigated its role as a primary target for DBS in depressed patients. Anatomically the Nac is at the centre of the interactions between dopaminergic, serotoninergic and glutamatergic systems. Functionally, the Nac is involved in both normal and abnormal reward processes and in anhedonia and loss of motivation. Due to its central location between the emotional system, the cognitive system and motor control system, the Nac seems to have a central role in mood and feeling regulation. Conclusion According to encouraging recent studies, DBS seems to be a promising technique in resistant MDD treatment.
Collapse
Affiliation(s)
- Cecilia Nauczyciel
- Academic Department of Adult Psychiatry, Guillaume Régnier Hospital, EA 4712 35000, Rennes, France
| | - Suzanne Robic
- Lyon Neuroscience Research Center INSERM U1028 - CNRS UMR 5292 Dynamics and Brain Cognition, 69675, Bron, France
| | - Thibaut Dondaine
- Rennes University Hospital Centre Research Unit EA 4712 Behavior and Basal Ganglia, 35000, Rennes, France
| | - Marc Verin
- Rennes University Hospital Centre Research Unit EA 4712 Behavior and Basal Ganglia, 35000, Rennes, France
| | - Gabriel Robert
- Academic Department of Adult Psychiatry, Guillaume Régnier Hospital, EA 4712 35000, Rennes, France ; Rennes University Hospital Centre Research Unit EA 4712 Behavior and Basal Ganglia, 35000, Rennes, France
| | - Dominique Drapier
- Academic Department of Adult Psychiatry, Guillaume Régnier Hospital, EA 4712 35000, Rennes, France ; Rennes University Hospital Centre Research Unit EA 4712 Behavior and Basal Ganglia, 35000, Rennes, France
| | - Florian Naudet
- Academic Department of Adult Psychiatry, Guillaume Régnier Hospital, EA 4712 35000, Rennes, France ; Rennes University Hospital Centre Research Unit EA 4712 Behavior and Basal Ganglia, 35000, Rennes, France
| | - Bruno Millet
- Academic Department of Adult Psychiatry, Guillaume Régnier Hospital, EA 4712 35000, Rennes, France ; Rennes University Hospital Centre Research Unit EA 4712 Behavior and Basal Ganglia, 35000, Rennes, France
| |
Collapse
|
7220
|
Chen WT, Zhu G, Pfaffenbach K, Kanel G, Stiles B, Lee AS. GRP78 as a regulator of liver steatosis and cancer progression mediated by loss of the tumor suppressor PTEN. Oncogene 2013; 33:4997-5005. [PMID: 24141775 PMCID: PMC3994182 DOI: 10.1038/onc.2013.437] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 08/30/2013] [Accepted: 09/05/2013] [Indexed: 02/06/2023]
Abstract
Glucose-regulated protein 78 (GRP78), a molecular chaperone widely elevated in human cancers, is critical for endoplasmic reticulum (ER) protein folding, stress signaling and PI3K/AKT activation. Genetic knockout models of GRP78 revealed that GRP78 maintains homeostasis of metabolic organs, including liver, pancreas and adipose tissues. Hepatocellular carcinoma (HCC) and cholangiocarcinoma (CC) are the most common liver cancers. There is a lack of effective therapeutics for HCC and CC, highlighting the need to further understand liver tumorigenic mechanisms. PTEN, a tumor suppressor that antagonizes the PI3K/AKT pathway, is inactivated in a wide range of tumors, including 40–50% of human liver cancers. To elucidate the role of GRP78 in liver cancer, we created a mouse model with biallelic liver-specific deletion of Pten and Grp78 mediated by Albumin-Cre-recombinase (cPf/f78f/f). Interestingly, in contrast to PTEN, deletion of GRP78 was progressive but incomplete. At 3 months, cPf/f78f/f livers showed hepatomegaly, activation of lipogenic genes, exacerbated steatosis and liver injury, implying that GRP78 protects the liver against PTEN-null mediated pathogenesis. Furthermore, in response to liver injury, we observed increased proliferation and expansion of bile duct and liver progenitor cells in cPf/f78f/f livers. Strikingly, bile duct cells in cPf/f78f/f livers maintained wild-type (WT) GRP78 level while adjacent areas showed GRP78 reduction. Analysis of signaling pathways revealed selective JNK activation, β-catenin downregulation, along with PDGFRα upregulation, which was unique to cPf/f78f/f livers at 6 months. Development of both HCC and CC was accelerated and evident in cPf/f78f/f livers at 8–9 months, coinciding with intense GRP78 expression in the cancer lesions, and GRP78 expression in adjacent normal areas reverted back to the WT level. In contrast, c78f/f livers showed no malignancy even at 14 months. These studies reveal GRP78 is a novel regulator for PTEN-loss mediated liver injury and cancer progression.
Collapse
Affiliation(s)
- W-T Chen
- Department of Biochemistry and Molecular Biology, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - G Zhu
- Department of Biochemistry and Molecular Biology, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - K Pfaffenbach
- Department of Biology, Eastern Oregon University, La Grande, OR, USA
| | - G Kanel
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - B Stiles
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy, Los Angeles, CA, USA
| | - A S Lee
- Department of Biochemistry and Molecular Biology, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
7221
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
7222
|
Detection and Discrimination of Non-Melanoma Skin Cancer by Multimodal Imaging. Healthcare (Basel) 2013; 1:64-83. [PMID: 27429131 PMCID: PMC4934506 DOI: 10.3390/healthcare1010064] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/30/2013] [Accepted: 09/30/2013] [Indexed: 01/18/2023] Open
Abstract
Non-melanoma skin cancer (NMSC) belongs to the most frequent human neoplasms. Its exposed location facilitates a fast ambulant treatment. However, in the clinical practice far more lesions are removed than necessary, due to the lack of an efficient pre-operational examination procedure: Standard imaging methods often do not provide a sufficient spatial resolution. The demand for an efficient in vivo imaging technique might be met in the near future by non-linear microscopy. As a first step towards this goal, the appearance of NMSC in various microspectroscopic modalities has to be defined and approaches have to be derived to distinguish healthy skin from NMSC using non-linear optical microscopy. Therefore, in this contribution the appearance of ex vivo NMSC in a combination of coherent anti-Stokes Raman scattering (CARS), second harmonic generation (SHG) and two photon excited fluorescence (TPEF) imaging—referred as multimodal imaging—is described. Analogous to H&E staining, an overview of the distinct appearances and features of basal cell and squamous cell carcinoma in the complementary modalities is derived, and is expected to boost in vivo studies of this promising technological approach.
Collapse
|
7223
|
Cytosolic calcium regulation in rat afferent vagal neurons during anoxia. Cell Calcium 2013; 54:416-27. [PMID: 24189167 DOI: 10.1016/j.ceca.2013.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 09/30/2013] [Accepted: 10/06/2013] [Indexed: 12/12/2022]
Abstract
Sensory neurons are able to detect tissue ischaemia and both transmit information to the brainstem as well as release local vasoactive mediators. Their ability to sense tissue ischaemia is assumed to be primarily mediated through proton sensing ion channels, lack of oxygen however may also affect sensory neuron function. In this study we investigated the effects of anoxia on isolated capsaicin sensitive neurons from rat nodose ganglion. Acute anoxia triggered a reversible increase in [Ca2+]i that was mainly due to Ca2+-efflux from FCCP sensitive stores and from caffeine and CPA sensitive ER stores. Prolonged anoxia resulted in complete depletion of ER Ca2+-stores. Mitochondria were partially depolarised by acute anoxia but mitochondrial Ca2+-uptake/buffering during voltage-gated Ca2+-influx was unaffected. The process of Ca2+-release from mitochondria and cytosolic Ca2+-clearance following Ca2+ influx was however significantly slowed. Anoxia was also found to inhibit SERCA activity and, to a lesser extent, PMCA activity. Hence, anoxia has multiple influences on [Ca2+]i homeostasis in vagal afferent neurons, including depression of ATP-driven Ca2+-pumps, modulation of the kinetics of mitochondrial Ca2+ buffering/release and Ca2+-release from, and depletion of, internal Ca2+-stores. These effects are likely to influence sensory neuronal function during ischaemia.
Collapse
|
7224
|
Zheng M, Jiang J, Tang YL, Liang XH. Oncogene and non-oncogene addiction in inflammation-associated cancers. Future Oncol 2013; 9:561-73. [PMID: 23560378 DOI: 10.2217/fon.12.202] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Many cancers originate in tissues that are chronically inflamed, and the inflammatory microenvironment is considered to promote the progression of malignancy, including initiation, growth, angiogenesis, invasion and metastasis. The molecular mechanism of inflammation-induced progression of cancers has been widely discussed. Oncogene and non-oncogene addiction have been proposed as two distinct but complementary theories to explain the initiation and development of cancers. Furthermore, they also play a role in cancer-associated inflammation. A solid understanding of oncogene and non-oncogene addiction in cancer-associated inflammatory microenvironments will help to exploit cancer drug targets for cancer prevention and clinical treatment.
Collapse
Affiliation(s)
- Min Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Sec. 3, Renminnan Road, Chengdu Sichuan 610041, People's Republic of China
| | | | | | | |
Collapse
|
7225
|
Shi N, Chen SY. Cell division cycle 7 mediates transforming growth factor-β-induced smooth muscle maturation through activation of myocardin gene transcription. J Biol Chem 2013; 288:34336-42. [PMID: 24133205 DOI: 10.1074/jbc.m113.498238] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Smooth muscle (SM) development consists of several processes, including cell fate determination, differentiation, and maturation. The molecular mechanisms controlling SM early differentiation have been studied extensively. However, little is known about the mechanism underlying SM maturation. Cell division cycle 7 (Cdc7) has been shown to regulate cell fate determination in the initial phase of transforming growth factor-β (TGF-β)-induced SM differentiation. Our present study indicates that Cdc7 also regulates SM maturation. Knockdown of Cdc7 suppresses TGF-β-induced expression of SM myosin heavy chain, a late marker of SM differentiation. Cdc7 overexpression, on the other hand, enhances SM myosin heavy chain expression. Interestingly, Cdc7 activates the mRNA expression and promoter activity of myocardin (Myocd), a master regulator of SM differentiation, whose transcription is blocked in the initial phase of the differentiation because TGF-β does not induce Myocd mRNA until after the early SM markers are induced. These data suggest that Cdc7 mediates TGF-β-induced SM maturation via activation of Myocd transcription. Mechanistically, Cdc7 physically and functionally interacts with Nkx2.5 to regulate Myocd promoter activity. Cdc7 appears to enhance Nkx2.5 binding to Myocd promoter, leading to Myocd activation. Taken together, our studies demonstrate that Cdc7 regulates the initial and late phase of SM differentiation through distinct mechanisms.
Collapse
Affiliation(s)
- Ning Shi
- From the Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia 30602
| | | |
Collapse
|
7226
|
Borthakur A, Bhattacharyya S, Kumar A, Anbazhagan AN, Tobacman JK, Dudeja PK. Lactobacillus acidophilus alleviates platelet-activating factor-induced inflammatory responses in human intestinal epithelial cells. PLoS One 2013; 8:e75664. [PMID: 24130731 PMCID: PMC3794005 DOI: 10.1371/journal.pone.0075664] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 08/16/2013] [Indexed: 02/06/2023] Open
Abstract
Probiotics have been used as alternative prevention and therapy modalities in intestinal inflammatory disorders including inflammatory bowel diseases (IBD) and necrotizing enterocolitis (NEC). Pathophysiology of IBD and NEC includes the production of diverse lipid mediators, including platelet-activating factor (PAF) that mediate inflammatory responses in the disease. PAF is known to activate NF-κB, however, the mechanisms of PAF-induced inflammation are not fully defined. We have recently described a novel PAF-triggered pathway of NF-κB activation and IL-8 production in intestinal epithelial cells (IECs), requiring the pivotal role of the adaptor protein Bcl10 and its interactions with CARMA3 and MALT1. The current studies examined the potential role of the probiotic Lactobacillus acidophilus in reversing the PAF-induced, Bcl10-dependent NF-κB activation and IL-8 production in IECs. PAF treatment (5 µM×24 h) of NCM460 and Caco-2 cells significantly increased nuclear p65 NF-κB levels and IL-8 secretion (2-3-fold, P<0.05), compared to control, which were blocked by pretreatment of the cells for 6 h with L. acidophilus (LA) or its culture supernatant (CS), followed by continued treatments with PAF for 24 h. LA-CS also attenuated PAF-induced increase in Bcl10 mRNA and protein levels and Bcl10 promoter activity. LA-CS did not alter PAF-induced interaction of Bcl10 with CARMA3, but attenuated Bcl10 interaction with MALT1 and also PAF-induced ubiquitination of IKKγ. Efficacy of bacteria-free CS of LA in counteracting PAF-induced inflammatory cascade suggests that soluble factor(s) in the CS of LA mediate these effects. These results define a novel mechanism by which probiotics counteract PAF-induced inflammation in IECs.
Collapse
Affiliation(s)
- Alip Borthakur
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Sumit Bhattacharyya
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Arivarasu Natarajan Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Joanne K. Tobacman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Pradeep K. Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| |
Collapse
|
7227
|
Berra-Romani R, Avelino-Cruz JE, Raqeeb A, Della Corte A, Cinelli M, Montagnani S, Guerra G, Moccia F, Tanzi F. Ca²⁺-dependent nitric oxide release in the injured endothelium of excised rat aorta: a promising mechanism applying in vascular prosthetic devices in aging patients. BMC Surg 2013; 13 Suppl 2:S40. [PMID: 24266895 PMCID: PMC3851245 DOI: 10.1186/1471-2482-13-s2-s40] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background Nitric oxide is key to endothelial regeneration, but it is still unknown whether endothelial cell (EC) loss results in an increase in NO levels at the wound edge. We have already shown that endothelial damage induces a long-lasting Ca2+ entry into surviving cells though connexin hemichannels (CxHcs) uncoupled from their counterparts on ruptured cells. The physiological outcome of injury-induced Ca2+ inflow is, however, unknown. Methods In this study, we sought to determine whether and how endothelial scraping induces NO production (NOP) in the endothelium of excised rat aorta by exploiting the NO-sensitive fluorochrome, DAF-FM diacetate and the Ca2+-sensitive fluorescent dye, Fura-2/AM. Results We demonstrated that injury-induced NOP at the lesion site is prevented in presence of the endothelial NO synthase inhibitor, L-NAME, and in absence of extracellular Ca2+. Unlike ATP-dependent NO liberation, the NO response to injury is insensitive to BTP-2, which selectively blocks store-operated Ca2+ inflow. However, injury-induced NOP is significantly reduced by classic gap junction blockers, and by connexin mimetic peptides specifically targeting Cx37Hcs, Cx40HCs, and Cx43Hcs. Moreover, disruption of caveolar integrity prevents injury-elicited NO signaling, but not the accompanying Ca2+ response. Conclusions The data presented provide the first evidence that endothelial scraping stimulates NO synthesis at the wound edge, which might both exert an immediate anti-thrombotic and anti-inflammatory action and promote the subsequent re-endothelialization.
Collapse
|
7228
|
Moccia F, Dragoni S, Cinelli M, Montagnani S, Amato B, Rosti V, Guerra G, Tanzi F. How to utilize Ca²⁺ signals to rejuvenate the repairative phenotype of senescent endothelial progenitor cells in elderly patients affected by cardiovascular diseases: a useful therapeutic support of surgical approach? BMC Surg 2013; 13 Suppl 2:S46. [PMID: 24267290 PMCID: PMC3851045 DOI: 10.1186/1471-2482-13-s2-s46] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Endothelial dysfunction or loss is the early event that leads to a host of severe cardiovascular diseases, such as atherosclerosis, hypertension, brain stroke, myocardial infarction, and peripheral artery disease. Ageing is regarded among the most detrimental risk factor for vascular endothelium and predisposes the subject to atheroscleorosis and inflammatory states even in absence of traditional comorbid conditions. Standard treatment to restore blood perfusion through stenotic arteries are surgical or endovascular revascularization. Unfortunately, ageing patients are not the most amenable candidates for such interventions, due to high operative risk or unfavourable vascular involvement. It has recently been suggested that the transplantation of autologous bone marrow-derived endothelial progenitor cells (EPCs) might constitute an alternative and viable therapeutic option for these individuals. Albeit pre-clinical studies demonstrated the feasibility of EPC-based therapy to recapitulate the diseased vasculature of young and healthy animals, clinical studies provided less impressive results in old ischemic human patients. One hurdle associated to this kind of approach is the senescence of autologous EPCs, which are less abundant in peripheral blood and display a reduced pro-angiogenic activity. Conversely, umbilical cord blood (UCB)-derived EPCs are more suitable for cellular therapeutics due to their higher frequency and sensitivity to growth factors, such as vascular endothelial growth factor (VEGF). An increase in intracellular Ca2+ concentration is central to EPC activation by VEGF. We have recently demonstrated that the Ca2+ signalling machinery driving the oscillatory Ca2+ response to this important growth factor is different in UCB-derived EPCs as compared to their peripheral counterparts. In particular, we focussed on the so-called endothelial colony forming cells (ECFCs), which are the only EPC population belonging to the endothelial lineage and able to form capillary-like structures in vitro and stably integrate with host vasculature in vivo. The present review provides a brief description of how exploiting the Ca2+ toolkit of juvenile EPCs to restore the repairative phenotype of senescent EPCs to enhance their regenerative outcome in therapeutic settings.
Collapse
|
7229
|
Aliwaini S, Swarts AJ, Blanckenberg A, Mapolie S, Prince S. A novel binuclear palladacycle complex inhibits melanoma growth in vitro and in vivo through apoptosis and autophagy. Biochem Pharmacol 2013; 86:1650-63. [PMID: 24099796 DOI: 10.1016/j.bcp.2013.09.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 12/23/2022]
Abstract
Malignant melanoma is an aggressive skin cancer and it is reported to be the most treatment-resistant human cancer. Here we describe the anti-tumour activity of a novel binuclear palladacycle complex (AJ-5) in vertical growth phase (ME1402) and metastatic (WM1158) melanoma cell lines. We show that compared to normal control cell lines, AJ-5 is more effective in inhibiting the proliferation of ME1402 and WM1158 melanoma cells with IC50 values of 0.19 and 0.20μM, respectively. Flow cytometry analyses showed that AJ-5 induced apoptosis (sub-G1 peak) which was confirmed by Annexin V-FITC/propidium iodide double-staining, nuclear fragmentation and an increase in the levels of PARP cleavage. Furthermore, AJ-5 was shown to induce both intrinsic and extrinsic apoptotic pathways as measured by PUMA, Bax and active caspases. Interestingly, AJ-5 treatment also simultaneously induced the formation of autophagosomes and led to an increase in the autophagy markers LC3II and Beclin1. Inhibition of autophagy reduced AJ-5 cytotoxicity suggesting that AJ-5 induced autophagy was a cell death and not cell survival mechanism. Moreover we show that AJ-5 induces the ATM-CHK2 DNA damage pathway and that its anti-tumour function is mediated by the p38 and ERK1/2 signalling pathways. Importantly, AJ-5 treatment efficiently reduced tumour growth in melanoma bearing mice and induced high levels of autophagy and apoptosis markers. Together these findings suggest that AJ-5 may be an effective chemotherapeutic drug in the treatment of melanoma, a highly aggressive and intractable cancer.
Collapse
Affiliation(s)
- Saeb Aliwaini
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925 Cape Town, South Africa.
| | | | | | | | | |
Collapse
|
7230
|
Zhang W, An J, Jawadi H, Siow DL, Lee JF, Zhao J, Gartung A, Maddipati KR, Honn KV, Wattenberg BW, Lee MJ. Sphingosine-1-phosphate receptor-2 mediated NFκB activation contributes to tumor necrosis factor-α induced VCAM-1 and ICAM-1 expression in endothelial cells. Prostaglandins Other Lipid Mediat 2013; 106:62-71. [PMID: 23770055 PMCID: PMC3844125 DOI: 10.1016/j.prostaglandins.2013.06.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 05/29/2013] [Accepted: 06/03/2013] [Indexed: 10/26/2022]
Abstract
Sphingosine-1-phosphate (S1P) regulates a wide array of biological functions in endothelial cells. We previously showed that S1P receptor subtype 2 (S1P2) is significantly up-regulated in the atherosclerotic endothelium (J. Biol. Chem. 283:30363, 2008). In this study, we investigated the roles of S1P2-mediated signaling in the proinflammatory responses of endothelial cells. Treatment with tumor necrosis factor-α (TNFα), a proinflammatory cytokine, increased the expression of S1P2 receptors in endothelial cells. TNFα treatment also enhanced sphingosine kinase 1 expression and increased S1P production. Pharmacological inhibition or knockdown of S1P2 receptors completely abrogated the TNFα-induced VCAM-1 (vascular cell adhesion molecule 1) and ICAM-1 (intercellular adhesion molecule 1) expression in endothelial cells. In contrast, pharmacological inhibition or knockdown of other S1P receptor subtypes had no effect on the TNFα-stimulated ICAM-1 and VCAM-1 expression. Moreover, ectopic expression of S1P2 receptors increased VCAM-1 and ICAM-1 expression in endothelial cells in response to S1P stimulation. Mechanistically, we show that antagonizing S1P2 signaling markedly inhibited the TNFα-stimulated NFκB activation. Utilizing the NFκB reporter luciferase assay, the S1P/S1P2 signaling was shown to stimulate NFκB activation. Moreover, the S1P/S1P2-stimulated VCAM-1/ICAM-1 expression was completely abolished by the pharmacological inhibitor of NFκB. Collectively, our data suggest that TNFα treatment activates autocrine S1P/S1P2 signaling, which subsequently activates NFκB and leads to the proinflammatory responses in endothelial cells.
Collapse
Affiliation(s)
- WenLiang Zhang
- Bioactive Lipid Research Program, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48202
| | - Jin An
- Bioactive Lipid Research Program, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48202
| | - Hiba Jawadi
- Bioactive Lipid Research Program, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Nutrition and Food Science, Wayne State University School of Medicine, Detroit, Michigan 48202
| | - Deanna L. Siow
- James Graham Brown Cancer Center, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Jen-Fu Lee
- Bioactive Lipid Research Program, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48202
| | - Jiawei Zhao
- Bioactive Lipid Research Program, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48202
| | - Allison Gartung
- Bioactive Lipid Research Program, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48202
| | - Krishna Rao Maddipati
- Bioactive Lipid Research Program, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48202
| | - Kenneth V. Honn
- Bioactive Lipid Research Program, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48202
| | - Binks W. Wattenberg
- James Graham Brown Cancer Center, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Menq-Jer Lee
- Bioactive Lipid Research Program, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48202
- Department of Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan 48202
| |
Collapse
|
7231
|
Nelson MP, Shacka JJ. Autophagy Modulation in Disease Therapy: Where Do We Stand? CURRENT PATHOBIOLOGY REPORTS 2013; 1:239-245. [PMID: 24470989 DOI: 10.1007/s40139-013-0032-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Since it was first described more than 50 years ago autophagy has been examined in many contexts, from cell survival to pathogen sequestration and removal. In more recent years our understanding of autophagy has developed sufficiently to allow effective targeted therapeutics to be developed against various diseases. The field of autophagy research is expanding rapidly, demonstrated by increases in both numbers of investigators in the field and the breadth of topics being addressed. Some diseases, such as the many cancers, have come to the fore in autophagy therapeutics research as a better understanding of their underlying mechanisms has surfaced. Numerous treatments are being developed and explored, from creative applications of the classic autophagy modulators chloroquine and rapamycin, to repurposing drugs approved for other treatments, such as astemizole, which is currently in use as an antimalarial and chronic rhinitis treatment. The landscape of autophagy modulation in disease therapy is rapidly changing and this review hopes to provide a cross-section of the current state of the field.
Collapse
Affiliation(s)
- Michael P Nelson
- Department of Pathology, Neuropathology Division, University of Alabama at Birmingham, Sparks Clinics Room SC 930B, 1720 7 Ave S., Birmingham, AL 35294, USA
| | - John J Shacka
- Department of Pathology, Neuropathology Division, University of Alabama at Birmingham, Birmingham VA Medical Center, Sparks Clinics Room SC 930B, 1720 7 Ave S., Birmingham, AL 35294, USA
| |
Collapse
|
7232
|
Prevalence and predictors of worsened left ventricular diastolic dysfunction after catheter ablation of atrial fibrillation. Int J Cardiol 2013; 168:3613-5. [DOI: 10.1016/j.ijcard.2013.05.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 05/04/2013] [Indexed: 01/01/2023]
|
7233
|
Minicucci MF, Santos PPD, Rafacho BPM, Gonçalves AF, Ardisson LP, Batista DF, Azevedo PS, Polegato BF, Okoshi K, Pereira EJ, Paiva SAR, Zornoff LAM. Periostin as a modulator of chronic cardiac remodeling after myocardial infarction. Clinics (Sao Paulo) 2013; 68:1344-9. [PMID: 24212842 PMCID: PMC3798673 DOI: 10.6061/clinics/2013(10)09] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/16/2013] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE After acute myocardial infarction, during the cardiac repair phase, periostin is released into the infarct and activates signaling pathways that are essential for the reparative process. However, the role of periostin in chronic cardiac remodeling after myocardial infarction remains to be elucidated. Therefore, the objective of this study was to investigate the relationship between tissue periostin and cardiac variables in the chronic cardiac remodeling induced by myocardial infarction. METHODS Male Wistar rats were assigned to 2 groups: a simulated surgery group (SHAM; n = 8) and a myocardial infarction group (myocardial infarction; n = 13). After 3 months, morphological, functional and biochemical analyses were performed. The data are expressed as means±SD or medians (including the lower and upper quartiles). RESULTS Myocardial infarctions induced increased left ventricular diastolic and systolic areas associated with a decreased fractional area change and a posterior wall shortening velocity. With regard to the extracellular matrix variables, the myocardial infarction group presented with higher values of periostin and types I and III collagen and higher interstitial collagen volume fractions and myocardial hydroxyproline concentrations. In addition, periostin was positively correlated with type III collagen levels (r = 0.673, p = 0.029) and diastolic (r = 0.678, p = 0.036) and systolic (r = 0.795, p = 0.006) left ventricular areas. Considering the relationship between periostin and the cardiac function variables, periostin was inversely correlated with both the fractional area change (r = -0.783, p = 0.008) and the posterior wall shortening velocity (r = -0.767, p = 0.012). CONCLUSIONS Periostin might be a modulator of deleterious cardiac remodeling in the chronic phase after myocardial infarction in rats.
Collapse
Affiliation(s)
- Marcos F Minicucci
- Universidade Estadual Paulista (UNESP), Botucatu Medical School, Internal Medicine Department, BotucatuSP, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7234
|
Yamashita M, Yamashita Y, Suzuki T, Kani Y, Mizusawa N, Imamura S, Takemoto K, Hara T, Hossain MA, Yabu T, Touhata K. Selenoneine, a novel selenium-containing compound, mediates detoxification mechanisms against methylmercury accumulation and toxicity in zebrafish embryo. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2013; 15:559-70. [PMID: 23709046 PMCID: PMC3742965 DOI: 10.1007/s10126-013-9508-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 03/19/2013] [Indexed: 05/15/2023]
Abstract
The selenium (Se)-containing antioxidant selenoneine (2-selenyl-N α,N α,N α-trimethyl-L-histidine) has recently been discovered to be the predominant form of organic Se in tuna blood. Although dietary intake of fish Se has been suggested to reduce methylmercury (MeHg) toxicity, the molecular mechanism of MeHg detoxification by Se has not yet been determined. Here, we report evidence that selenoneine accelerates the excretion and demethylation of MeHg, mediated by a selenoneine-specific transporter, organic cations/carnitine transporter-1 (OCTN1). Selenoneine was incorporated into human embryonic kidney HEK293 cells transiently overexpressing OCTN1 and zebrafish blood cells by OCTN1. The K m for selenoneine uptake was 13.0 μM in OCTN1-overexpressing HEK293 cells and 9.5 μM in zebrafish blood cells, indicating high affinity of OCTN1 for selenoneine in human and zebrafish cells. When such OCTN1-expressing cells and embryos were exposed to MeHg-cysteine (MeHgCys), MeHg accumulation was decreased and the excretion and demethylation of MeHg were enhanced by selenoneine. In addition, exosomal secretion vesicles were detected in the culture water of embryos that had been microinjected with MeHgCys, suggesting that these may be responsible for MeHg excretion and demethylation. In contrast, OCTN1-deficient embryos accumulated MeHg, and MeHg excretion and demethylation were decreased. Furthermore, Hg accumulation was decreased in OCTN1-overexpressing HEK293 cells, but not in mock vector-transfected cells, indicating that selenoneine and OCTN1 can regulate MeHg detoxification in human cells. Thus, the selenoneine-mediated OCTN1 system regulates secretory lysosomal vesicle formation and MeHg demethylation.
Collapse
Affiliation(s)
- Michiaki Yamashita
- National Research Institute of Fisheries Science, 2-12-4 Fukuura, Yokohama, Kanagawa, 236-8648, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7235
|
Morris MJ, Monteggia LM. Unique functional roles for class I and class II histone deacetylases in central nervous system development and function. Int J Dev Neurosci 2013; 31:370-81. [PMID: 23466417 PMCID: PMC3726026 DOI: 10.1016/j.ijdevneu.2013.02.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Revised: 01/11/2013] [Accepted: 02/15/2013] [Indexed: 01/04/2023] Open
Abstract
Non-specific pharmacological inhibition of the histone deacetylase (HDAC) family of enzymes has largely beneficial effects in a variety of diverse contexts including cancer, cognitive function, and neurodegeneration. This review will discuss the role of individual HDAC isoforms in brain function during development and in the adult. Importantly class I and class II HDACs exhibit distinct cellular and subcellular expression patterns and utilize different signaling pathways to influence their substrates. Moreover, dissociable phenotypic outcomes emerge following manipulation of individual HDACs in the brain. To date, pharmacological inhibitors capable of targeting individual HDACs have proven difficult to develop, an obstacle that must be overcome to unlock the substantial clinical promise of manipulating endogenous HDAC isoforms in the central nervous system.
Collapse
Affiliation(s)
- Michael J Morris
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9070, USA
| | | |
Collapse
|
7236
|
Minicucci MF, dos Santos PP, Rafacho BPM, Gonçalves AF, Silva RAC, Chiuso-Minicucci F, Azevedo PS, Polegato BF, Okoshi K, Pereira EJ, Paiva SAR, Zornoff LAM. Mechanisms involved in the beneficial effects of spironolactone after myocardial infarction. PLoS One 2013; 8:e76866. [PMID: 24098808 PMCID: PMC3786966 DOI: 10.1371/journal.pone.0076866] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 08/23/2013] [Indexed: 11/18/2022] Open
Abstract
Introduction Our objective was to analyze the effect of spironolactone on cardiac remodeling after experimental myocardial infarction (MI), assessed by matricellular proteins levels, cardiac collagen amount and distribution, myocardial tissue metalloproteinase inhibitor-1(TIMP-1) concentration, myocyte hypertrophy, left ventricular architecture, and invitro and invivo cardiac function. Methods Wistar rats were assigned to 4 groups: control group, in which animals were submitted to simulated surgery (SHAM group; n=9); group that received spironolactone and in which animals were submitted to simulated surgery (SHAM-S group, n=9); myocardial infarction group, in which animals were submitted to coronary artery ligation (MI group, n=15); and myocardial infarction group with spironolactone supplementation (MI-S group, n=15). The rats were observed for 3 months. Results The MI group had higher values of left cardiac chambers and mass index and lower relative wall thicknesses compared with the SHAM group. In addition, diastolic and systolic functions were worse in the MI groups. However, spironolactone did not influence any of these variables. The MI-S group had a lower myocardial hydroxyproline concentration and myocyte cross-sectional area compared with the MI group. Myocardial periostin and collagen type III were lower in the MI-S group compared with the MI-group. In addition, TIMP-1 concentration in myocardium was higher in the MI-S group compared with the MI group. Conclusions The predominant consequence of spironolactone supplementation after MI is related to reductions in collagens, with discrete attenuation of other remodeling variables. Importantly, this effect may be modulated by periostin and TIMP-1 levels.
Collapse
Affiliation(s)
- Marcos F. Minicucci
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
- * E-mail:
| | - Priscila P. dos Santos
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Bruna P. M. Rafacho
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Andrea F. Gonçalves
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Renata A. C. Silva
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Fernanda Chiuso-Minicucci
- Department of Microbiology and Immunology, Institute of Biosciences, University Estadual, Paulista, Botucatu, São Paulo, Brazil
| | - Paula S. Azevedo
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Bertha F. Polegato
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Katashi Okoshi
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Elenize J. Pereira
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Sergio A. R. Paiva
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Leonardo A. M. Zornoff
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| |
Collapse
|
7237
|
The plasma membrane Ca2+-ATPase2 (PMCA2) is involved in the regulation of Purkinje cell dendritic growth in cerebellar organotypic slice cultures. Neural Plast 2013; 2013:321685. [PMID: 24288624 PMCID: PMC3830849 DOI: 10.1155/2013/321685] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 08/30/2013] [Accepted: 09/03/2013] [Indexed: 11/17/2022] Open
Abstract
Purkinje cells are the principal neurons of the cerebellar cortex and have an extensive and elaborate dendritic tree. Chronic activation of type I metabotropic glutamate receptors inhibits Purkinje cell dendritic growth in organotypic cerebellar slice cultures. This effect is mediated by calcium influx through P/Q-type and T-type Ca2+ channels. We have now studied the role of the plasma membrane Ca2+-ATPase2 (PMCA2), a major calcium extrusion pump, for Purkinje cell dendritic development. We found that PMCA2 is strongly expressed in the plasma membrane and dendritic spines of Purkinje cells in organotypic slice cultures compatible with a role for controlling the local dendritic calcium equilibrium. Inhibition of PMCA2 activity by carboxyeosin resulted in a moderate reduction of Purkinje cell dendritic tree size indicating that the extrusion of calcium by PMCA2 is important for maintaining the dendritic calcium concentration and controlling dendritic growth. When inhibition of PMCA2 was combined with stimulation of type I metabotropic glutamate receptors, it partially rescued dendritic morphology. This protection can be explained by a compensatory inactivation of voltage-gated calcium channels in Purkinje cells after PMCA2 inhibition. Our results demonstrate that PMCA2 activity is an important regulator of the dendritic calcium equilibrium controlling Purkinje cell dendritic growth.
Collapse
|
7238
|
Calabrò E, Condello S, Currò M, Ferlazzo N, Caccamo D, Magazù S, Ientile R. Effects of low intensity static magnetic field on FTIR spectra and ROS production in SH-SY5Y neuronal-like cells. Bioelectromagnetics 2013; 34:618-29. [PMID: 24217848 DOI: 10.1002/bem.21815] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 08/12/2013] [Indexed: 12/28/2022]
Abstract
Biological effects of man-made electromagnetic fields (EMFs) have been studied so far by experimental approaches exposing animals and cell cultures to EMFs. However, the evidence for cell toxicity induced by static magnetic field (SMF) is still uncertain. We investigated the effects produced by the exposure of human SH-SY5Y neuronal-like cells to a uniform magnetic field at intensities of 2.2 mT, which is less than the recommended public exposure limits set by the International Commission on Non-Ionizing Radiation Protection (ICNIRP). A decrease of membrane mitochondrial potential up to 30% was measured after 24 h of exposure to SMF in SH-SY5Y cells, and this effect was associated with reactive oxygen species production increase. Fourier transform infrared spectroscopy (FTIR) analysis showed that exposure to a static magnetic intensity around 2.2 mT changed the secondary structure of cellular proteins and lipid components. The vibration bands relative to the methylene group increased significantly after 4 h of exposure, whereas further exposure up to 24 h produced evident shifts of amide I and II modes and a relative increase in β-sheet contents with respect to α-helix components. Our study demonstrated that a moderate SMF causes alteration in cell homeostasis, as indicated by FTIR spectroscopy observations of changes in protein structures that are part of cell response to magnetic field exposure.
Collapse
|
7239
|
Dynamic Cross Talk between S1P and CXCL12 Regulates Hematopoietic Stem Cells Migration, Development and Bone Remodeling. Pharmaceuticals (Basel) 2013; 6:1145-69. [PMID: 24276423 PMCID: PMC3818832 DOI: 10.3390/ph6091145] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 08/18/2013] [Accepted: 09/04/2013] [Indexed: 12/23/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are mostly retained in a quiescent non-motile mode in their bone marrow (BM) niches, shifting to a migratory cycling and differentiating state to replenish the blood with mature leukocytes on demand. The balance between the major chemo-attractants CXCL12, predominantly in the BM, and S1P, mainly in the blood, dynamically regulates HSC recruitment to the circulation versus their retention in the BM. During alarm situations, stress-signals induce a decrease in CXCL12 levels in the BM, while S1P levels are rapidly and transiently increased in the circulation, thus favoring mobilization of stem cells as part of host defense and repair mechanisms. Myeloid cytokines, including G-CSF, up-regulate S1P signaling in the BM via the PI3K pathway. Induced CXCL12 secretion from stromal cells via reactive oxygen species (ROS) generation and increased S1P1 expression and ROS signaling in HSCs, all facilitate mobilization. Bone turnover is also modulated by both CXCL12 and S1P, regulating the dynamic BM stromal microenvironment, osteoclasts and stem cell niches which all functionally express CXCL12 and S1P receptors. Overall, CXCL12 and S1P levels in the BM and circulation are synchronized to mutually control HSC motility, leukocyte production and osteoclast/osteoblast bone turnover during homeostasis and stress situations.
Collapse
|
7240
|
Das A, Segar CE, Hughley BB, Bowers DT, Botchwey EA. The promotion of mandibular defect healing by the targeting of S1P receptors and the recruitment of alternatively activated macrophages. Biomaterials 2013; 34:9853-62. [PMID: 24064148 DOI: 10.1016/j.biomaterials.2013.08.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/07/2013] [Indexed: 02/07/2023]
Abstract
Endogenous signals originating at the site of injury are involved in the paracrine recruitment, proliferation, and differentiation of circulating progenitor and diverse inflammatory cell types. Here, we investigate a strategy to exploit endogenous cell recruitment mechanisms to regenerate injured bone by local targeting and activation of sphingosine-1-phosphate (S1P) receptors. A mandibular defect model was selected for evaluating regeneration of bone following trauma or congenital disease. The particular challenges of mandibular reconstruction are inherent in the complex anatomy and function of the bone given that the area is highly vascularized and in close proximity to muscle. Nanofibers composed of poly(DL-lactide-co-glycolide) (PLAGA) and polycaprolactone (PCL) were used to delivery FTY720, a targeted agonist of S1P receptors 1 and 3. In vitro culture of bone progenitor cells on drug-loaded constructs significantly enhanced SDF1α mediated chemotaxis of bone marrow mononuclear cells. In vivo results show that local delivery of FTY720 from composite nanofibers enhanced blood vessel ingrowth and increased recruitment of M2 alternatively activated macrophages, leading to significant osseous tissue ingrowth into critical sized defects after 12 weeks of treatment. These results demonstrate that local activation of S1P receptors is a regenerative cue resulting in recruitment of wound healing or anti-inflammatory macrophages and bone healing. Use of such small molecule therapy can provide an alternative to biological factors for the clinical treatment of critical size craniofacial defects.
Collapse
Affiliation(s)
- Anusuya Das
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
| | | | | | | | | |
Collapse
|
7241
|
Gnerlich JL, Yao KA, Fitchev PS, Goldschmidt RA, Bond MC, Cornwell M, Crawford SE. Peritumoral Expression of Adipokines and Fatty Acids in Breast Cancer. Ann Surg Oncol 2013; 20 Suppl 3:S731-8. [DOI: 10.1245/s10434-013-3274-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Indexed: 12/16/2022]
|
7242
|
Ran Y, Malmirchegini GR, Clubb RT, Lei B. Axial ligand replacement mechanism in heme transfer from streptococcal heme-binding protein Shp to HtsA of the HtsABC transporter. Biochemistry 2013; 52:6537-47. [PMID: 23980583 PMCID: PMC3815476 DOI: 10.1021/bi400965u] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The heme-binding protein Shp of Group A Streptococcus rapidly transfers its heme to HtsA, the lipoprotein component of the HtsABC transporter, in a concerted two-step process with one kinetic phase. Heme axial residue-to-alanine replacement mutant proteins of Shp and HtsA (Shp(M66A), Shp(M153A), HtsA(M79A), and HtsA(H229A)) were used to probe the axial displacement mechanism of this heme transfer reaction. Ferric Shp(M66A) at high pH and Shp(M153A) have a pentacoordinate heme iron complex with a methionine axial ligand. ApoHtsA(M79A) efficiently acquires heme from ferric Shp but alters the reaction mechanism to two kinetic phases from a single phase in the wild-type protein reactions. In contrast, apoHtsA(H229A) cannot assimilate heme from ferric Shp. The conversion of pentacoordinate holoShp(M66A) into pentacoordinate holoHtsA(H229A) involves an intermediate, whereas holoHtsA(H229A) is directly formed from pentacoordinate holoShp(M153A). Conversely, apoHtsA(M79A) reacts with holoShp(M66A) and holoShp(M153A) in mechanisms with one and two kinetic phases, respectively. These results imply that the Met79 and His229 residues of HtsA displace the Met66 and Met153 residues of Shp, respectively. Structural docking analysis supports this mechanism of the specific axial residue displacement. Furthermore, the rates of the cleavage of the axial bond in Shp in the presence of a replacing HtsA axial residue are greater than that in the absence of a replacing HtsA axial residue. These findings reveal a novel heme transfer mechanism of the specific displacement of the Shp axial residues with the HtsA axial residues and the involvement of the HtsA axial residues in the displacement.
Collapse
Affiliation(s)
- Yanchao Ran
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, MT 59718
| | - G. Reza Malmirchegini
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Robert T. Clubb
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Benfang Lei
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, MT 59718
| |
Collapse
|
7243
|
Protective efficacy of orally administered, heat-killed Lactobacillus pentosus b240 against influenza A virus. Sci Rep 2013; 3:1563. [PMID: 23535544 PMCID: PMC3610098 DOI: 10.1038/srep01563] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 03/04/2013] [Indexed: 12/30/2022] Open
Abstract
Influenza A(H1N1)pdm virus caused the first human pandemic of the 21st century. Although various probiotic Lactobacillus species have been shown to have anti-microbial effects against pneumonia-inducing pathogens, the prophylactic efficacy and mechanisms behind their protection remain largely unknown. Here, we evaluated the prophylactic efficacy of heat-killed Lactobacillus pentosus b240 against lethal influenza A(H1N1)pdm virus infection in a mouse model. To further define the protective responses induced by b240, we performed virologic, histopathologic, and transcriptomic analyses on the mouse lungs. Although we did not observe an appreciable effect of b240 on virus growth, cytokine production, or histopathology, gene expressional analysis revealed that oral administration of b240 differentially regulates antiviral gene expression in mouse lungs. Our results unveil the possible mechanisms behind the protection mediated by b240 against influenza virus infection and provide new insights into probiotic therapy.
Collapse
|
7244
|
Sharma A, Sisodia R, Bhatnagar D, Saxena VK. Spatial memory and learning performance and its relationship to protein synthesis of Swiss albino mice exposed to 10 GHz microwaves. Int J Radiat Biol 2013; 90:29-35. [PMID: 23952535 DOI: 10.3109/09553002.2013.835883] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE To study the possible role of microwave (MW) exposure on spatial memory of Swiss albino mice and its relationship to protein concentration in whole brain. MATERIALS AND METHODS Mice were exposed to 10 GHz (Giga Hertz) microwaves with the power density of 0.25 mW/cm(2) (milliwatt per centimeter square) with average whole body specific absorption rate (SAR) 0.1790 W/kg daily for 2 hours per day (h/day) for 30 days. After exposure mice were tested for spatial memory performance using Morris water maze test (MWT). For this purpose mice (6-8 weeks old) were divided into two groups: (i) Sham exposed, and (ii) microwaves exposed. After initial training for two days, MWT was performed for another 6 days. Protein was estimated 48 h after exposure and immediately after completion of MWT. RESULTS Both sham-exposed and microwaves-exposed animals showed a significant decrease in escape time with training. Microwaves-exposed animals had statistically significant higher mean latency to reach the target quadrant compared to sham exposed. A concurrent decrease in protein levels was estimated in whole brain of the exposed mice compared to sham-exposed mice. CONCLUSIONS It can be concluded from the current study that exposure to microwave radiation caused decrements in the ability of mice to learn the special memory task, this may be due to simultaneous decrease in protein levels in the brain of mice.
Collapse
|
7245
|
Zhao H, Gu H, Zhang H, Li JH, Zhao WE. PPARγ-dependent pathway in the growth-inhibitory effects of K562 cells by carotenoids in combination with rosiglitazone. Biochim Biophys Acta Gen Subj 2013; 1840:545-55. [PMID: 24036327 DOI: 10.1016/j.bbagen.2013.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 09/03/2013] [Accepted: 09/04/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Carotenoids have been found to play roles in the prevention and therapy of some cancers which PPARγ was also discovered to be involved in. The present studies were directed to determine the inhibitory effects of carotenoids in combination with rosiglitazone, a synthetic PPARγ agonist, on K562 cell proliferation and elucidate the contribution of PPARγ-dependent pathway to cell proliferation suppression. METHODS The effects of carotenoid and rosiglitazone combination on K562 cell proliferation were evaluated by trypan blue dye exclusion assay and MTT assay. When PPARγ has been inhibited by GW9662 and siRNA, cycle-related regulator expression in K562 cells treated with carotenoid and rosiglitazone combination was analyzed by Western blotting. RESULTS Rosiglitazone inhibited K562 cell proliferation and augmented the inhibitory effects of carotenoids on the cell proliferation greatly. Specific PPARγ inhibition attenuated the cell growth suppression induced by carotenoid and rosiglitazone combination. GW9662 pre-treatment attenuated the enhanced up-regulation of PPARγ expression caused by the combination treatment. Moreover, GW9662 and PPARγ siRNA also significantly attenuated the up-regulation of p21 and down-regulation of cyclin D1 caused by carotenoids and rosiglitazone. CONCLUSIONS PPARγ signaling pathway, via stimulating p21 and inhibiting cyclin D1, may play an important role in the anti-proliferative effects of carotenoid and rosiglitazone combination on K562 cells. GENERAL SIGNIFICANCE Carotenoids in combination with rosiglitazone are hopeful to provide attractive dietary or supplementation-based and pharmaceutical strategies to treat cancer diseases.
Collapse
Affiliation(s)
- Han Zhao
- School of Chemical Engineering and Energy, Zhengzhou University, No. 100 Science Road, Zhengzhou 450001, PR China
| | | | | | | | | |
Collapse
|
7246
|
Vantaggiato C, Crimella C, Airoldi G, Polishchuk R, Bonato S, Brighina E, Scarlato M, Musumeci O, Toscano A, Martinuzzi A, Santorelli FM, Ballabio A, Bresolin N, Clementi E, Bassi MT. Defective autophagy in spastizin mutated patients with hereditary spastic paraparesis type 15. ACTA ACUST UNITED AC 2013; 136:3119-39. [PMID: 24030950 DOI: 10.1093/brain/awt227] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hereditary spastic paraparesis type 15 is a recessive complicated form of the disease clinically characterized by slowly progressive spastic paraparesis and mental deterioration with onset between the first and second decade of life. Thinning of corpus callosum is the neuroradiological distinctive sign frequently associated with white matter abnormalities. The causative gene, ZFYVE26, encodes a large protein of 2539 amino acid residues, termed spastizin, containing three recognizable domains: a zinc finger, a leucine zipper and a FYVE domain. Spastizin protein has a diffuse cytoplasmic distribution and co-localizes partially with early endosomes, the endoplasmic reticulum, microtubules and vesicles involved in protein trafficking. In addition, spastizin localizes to the mid-body during the final step of mitosis and contributes to successful cytokinesis. Spastizin interacts with Beclin 1, a protein required for cytokinesis and autophagy, which is the major lysosome-mediated degradation process in the cell. In view of the Beclin 1-spastizin interaction, we investigated the possible role of spastizin in autophagy. We carried out this analysis by using lymphoblast and fibroblast cells derived from four different spastizin mutated patients (p.I508N, p.L243P, p.R1209fsX, p.S1312X) and from control subjects. Of note, the truncating p.R1209fsX and p.S1312X mutations lead to loss of spastizin protein. The results obtained indicate that spastizin interacts with the autophagy related Beclin 1-UVRAG-Rubicon multiprotein complex and is required for autophagosome maturation. In cells lacking spastizin or with mutated forms of the protein, spastizin interaction with Beclin 1 is lost although the formation of the Beclin 1-UVRAG-Rubicon complex can still be observed. However, in these cells we demonstrate an impairment of autophagosome maturation and an accumulation of immature autophagosomes. Autophagy defects with autophagosome accumulation can be observed also in neuronal cells upon spastizin silencing. These results indicate that autophagy is a central process in the pathogenesis of complicated forms of hereditary spastic paraparesis with thin corpus callosum.
Collapse
Affiliation(s)
- Chiara Vantaggiato
- 1 Scientific Institute IRCCS E. Medea, Laboratory of Molecular Biology, 23842 Bosisio Parini, Lecco, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7247
|
|
7248
|
Tomar D, Prajapati P, Sripada L, Singh K, Singh R, Singh AK, Singh R. TRIM13 regulates caspase-8 ubiquitination, translocation to autophagosomes and activation during ER stress induced cell death. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3134-3144. [PMID: 24021263 DOI: 10.1016/j.bbamcr.2013.08.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 08/28/2013] [Accepted: 08/30/2013] [Indexed: 01/06/2023]
Abstract
The emerging evidences suggest that endoplasmic (ER) stress is involved in onset of many pathological conditions like cancer and neurodegeneration. The persistent ER stress results in misfolded protein aggregates, which are degraded through the process of autophagy or lead to cell death through activation of caspases. The regulation of crosstalk of autophagy and cell death during ER stress is emerging. Ubiquitination plays regulatory role in crosstalk of autophagy and cell death. In the current study, we describe the role of TRIM13, RING E3 ubiquitin ligase, in regulation of ER stress induced cell death. The expression of TRIM13 sensitizes cells to ER stress induced death. TRIM13 induced autophagy is essential for ER stress induced caspase activation and cell death. TRIM13 induces K63 linked poly-ubiquitination of caspase-8, which results in its stabilization and activation during ER stress. TRIM13 regulates translocation of caspase-8 to autophagosome and its fusion with lysosome during ER stress. This study first time demonstrated the role of TRIM13 as novel regulator of caspase-8 activation and cell death during ER stress.
Collapse
Affiliation(s)
- Dhanendra Tomar
- Department of Cell Biology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Paresh Prajapati
- Department of Cell Biology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Lakshmi Sripada
- Department of Cell Biology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Kritarth Singh
- Department of Cell Biology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Rochika Singh
- Department of Cell Biology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Arun Kumar Singh
- Department of Cell Biology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Rajesh Singh
- Department of Cell Biology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India.
| |
Collapse
|
7249
|
Abstract
The putative applications of poly-, oligo- and mono-oxometalates in biochemistry, biology, pharmacology and medicine are rapidly attracting interest. In particular, these compounds may act as potent ion pump inhibitors and have the potential to play a role in the treatment of e.g. ulcers, cancer and ischemic heart disease. However, the mechanism of action is not completely understood in most cases, and even remains largely unknown in other cases. In the present review we discuss the most recent insights into the interaction between mono- and polyoxometalate ions with ion pumps, with particular focus on the interaction of decavanadate with Ca(2+)-ATPase. We also compare the proposed mode of action with those of established ion pump inhibitors which are currently in therapeutic use. Of the 18 classes of compounds which are known to act as ion pump inhibitors, the complete mechanism of inhibition is only known for a handful. It has, however, been established that most ion pump inhibitors bind mainly to the E2 ion pump conformation within the membrane domain from the extracellular side and block the cation release. Polyoxometalates such as decavanadate, in contrast, interact with Ca(2+)-ATPase near the nucleotide binding site domain or at a pocket involving several cytoplasmic domains, and therefore need to cross through the membrane bilayer. In contrast to monomeric vanadate, which only binds to the E2 conformation, decavanadate binds to all protein conformations, i.e. E1, E1P, E2 and E2P. Moreover, the specific interaction of decavanadate with sarcoplasmic reticulum Ca(2+)-ATPase has been shown to be non-competitive with respect to ATP and induces protein cysteine oxidation with concomitant vanadium reduction which might explain the high inhibitory capacity of V10 (IC50 = 15 μM) which is quite similar to the majority of the established therapeutic drugs.
Collapse
|
7250
|
Villa R, Ferrari F, Gorini A. ATP-ases of synaptic plasma membranes in striatum: Enzymatic systems for synapses functionality by in vivo administration of l-acetylcarnitine in relation to Parkinson’s Disease. Neuroscience 2013; 248:414-26. [DOI: 10.1016/j.neuroscience.2013.06.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 06/13/2013] [Indexed: 11/17/2022]
|