1
|
Kamble OS, Chatterjee R, Abishek KG, Chandra J, Alsayari A, Wahab S, Sahebkar A, Kesharwani P, Dandela R. Small molecules targeting mitochondria as an innovative approach to cancer therapy. Cell Signal 2024; 124:111396. [PMID: 39251050 DOI: 10.1016/j.cellsig.2024.111396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Cellular death evasion is a defining characteristic of human malignancies and a significant contributor to therapeutic inefficacy. As a result of oncogenic inhibition of cell death mechanisms, established therapeutic regimens seems to be ineffective. Mitochondria serve as the cellular powerhouses, but they also function as repositories of self-destructive weaponry. Changes in the structure and activities of mitochondria have been consistently documented in cancer cells. In recent years, there has been an increasing focus on using mitochondria as a targeted approach for treating cancer. Considerable attention has been devoted to the development of delivery systems that selectively aim to deliver small molecules called "mitocans" to mitochondria, with the ultimate goal of modulating the physiology of cancer cells. This review summarizes the rationale and mechanism of mitochondrial targeting with small molecules in the treatment of cancer, and their impact on the mitochondria. This paper provides a concise overview of the reasoning and mechanism behind directing treatment towards mitochondria in cancer therapy, with a particular focus on targeting using small molecules. This review also examines diverse small molecule types within each category as potential therapeutic agents for cancer.
Collapse
Affiliation(s)
- Omkar S Kamble
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - Rana Chatterjee
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - K G Abishek
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India.
| |
Collapse
|
2
|
Rainey NE, Armand AS, Petit PX. Sodium arsenite and arsenic trioxide differently affect the oxidative stress of lymphoblastoid cells: An intricate crosstalk between mitochondria, autophagy and cell death. PLoS One 2024; 19:e0302701. [PMID: 38728286 PMCID: PMC11086853 DOI: 10.1371/journal.pone.0302701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Although the toxicity of arsenic depends on its chemical forms, few studies have taken into account the ambiguous phenomenon that sodium arsenite (NaAsO2) acts as a potent carcinogen while arsenic trioxide (ATO, As2O3) serves as an effective therapeutic agent in lymphoma, suggesting that NaAsO2 and As2O3 may act via paradoxical ways to either promote or inhibit cancer pathogenesis. Here, we compared the cellular response of the two arsenical compounds, NaAsO2 and As2O3, on the Burkitt lymphoma cell model, the Epstein Barr Virus (EBV)-positive P3HR1 cells. Using flow cytometry and biochemistry analyses, we showed that a NaAsO2 treatment induces P3HR1 cell death, combined with drastic drops in ΔΨm, NAD(P)H and ATP levels. In contrast, As2O3-treated cells resist to cell death, with a moderate reduction of ΔΨm, NAD(P)H and ATP. While both compounds block cells in G2/M and affect their protein carbonylation and lipid peroxidation, As2O3 induces a milder increase in superoxide anions and H2O2 than NaAsO2, associated to a milder inhibition of antioxidant defenses. By electron microscopy, RT-qPCR and image cytometry analyses, we showed that As2O3-treated cells display an overall autophagic response, combined with mitophagy and an unfolded protein response, characteristics that were not observed following a NaAsO2 treatment. As previous works showed that As2O3 reactivates EBV in P3HR1 cells, we treated the EBV- Ramos-1 cells and showed that autophagy was not induced in these EBV- cells upon As2O3 treatment suggesting that the boost of autophagy observed in As2O3-treated P3HR1 cells could be due to the presence of EBV in these cells. Overall, our results suggest that As2O3 is an autophagic inducer which action is enhanced when EBV is present in the cells, in contrast to NaAsO2, which induces cell death. That's why As2O3 is combined with other chemicals, as all-trans retinoic acid, to better target cancer cells in therapeutic treatments.
Collapse
Affiliation(s)
- Nathan Earl Rainey
- CNRS UMR 8003 Paris University, SSPIN, Neuroscience Institute, Team “Mitochondria, Apoptosis and Autophagy Signaling”, Campus Saint-Germain, Paris, France
| | - Anne-Sophie Armand
- INSERM U1151, Institut Necker Enfants Malades (INEM), Campus Necker, Université Paris Cité, Paris, France
| | - Patrice X. Petit
- CNRS UMR 8003 Paris University, SSPIN, Neuroscience Institute, Team “Mitochondria, Apoptosis and Autophagy Signaling”, Campus Saint-Germain, Paris, France
| |
Collapse
|
3
|
Korotkov SM. Mitochondrial Oxidative Stress Is the General Reason for Apoptosis Induced by Different-Valence Heavy Metals in Cells and Mitochondria. Int J Mol Sci 2023; 24:14459. [PMID: 37833908 PMCID: PMC10572412 DOI: 10.3390/ijms241914459] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
This review analyzes the causes and consequences of apoptosis resulting from oxidative stress that occurs in mitochondria and cells exposed to the toxic effects of different-valence heavy metals (Ag+, Tl+, Hg2+, Cd2+, Pb2+, Al3+, Ga3+, In3+, As3+, Sb3+, Cr6+, and U6+). The problems of the relationship between the integration of these toxic metals into molecular mechanisms with the subsequent development of pathophysiological processes and the appearance of diseases caused by the accumulation of these metals in the body are also addressed in this review. Such apoptosis is characterized by a reduction in cell viability, the activation of caspase-3 and caspase-9, the expression of pro-apoptotic genes (Bax and Bcl-2), and the activation of protein kinases (ERK, JNK, p53, and p38) by mitogens. Moreover, the oxidative stress manifests as the mitochondrial permeability transition pore (MPTP) opening, mitochondrial swelling, an increase in the production of reactive oxygen species (ROS) and H2O2, lipid peroxidation, cytochrome c release, a decline in the inner mitochondrial membrane potential (ΔΨmito), a decrease in ATP synthesis, and reduced glutathione and oxygen consumption as well as cytoplasm and matrix calcium overload due to Ca2+ release from the endoplasmic reticulum (ER). The apoptosis and respiratory dysfunction induced by these metals are discussed regarding their interaction with cellular and mitochondrial thiol groups and Fe2+ metabolism disturbance. Similarities and differences in the toxic effects of Tl+ from those of other heavy metals under review are discussed. Similarities may be due to the increase in the cytoplasmic calcium concentration induced by Tl+ and these metals. One difference discussed is the failure to decrease Tl+ toxicity through metallothionein-dependent mechanisms. Another difference could be the decrease in reduced glutathione in the matrix due to the reversible oxidation of Tl+ to Tl3+ near the centers of ROS generation in the respiratory chain. The latter may explain why thallium toxicity to humans turned out to be higher than the toxicity of mercury, lead, cadmium, copper, and zinc.
Collapse
Affiliation(s)
- Sergey M Korotkov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez pr. 44, 194223 St. Petersburg, Russia
| |
Collapse
|
4
|
Zeng Q, Singh R, Ye Y, Cheng S, Kong F, Zeng Q. Anti‐breast‐cancer activity of self‐fermented
Bovistella sinensis Lloyd
extracts through the mitochondrial
ROS
‐induced apoptosis in vitro. J Food Biochem 2022; 46:e14218. [DOI: 10.1111/jfbc.14218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/11/2022] [Accepted: 04/07/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Qinghua Zeng
- School of Food and Biological Engineering, Engineering Research Center of Bio‐process, Ministry of Education Hefei University of Technology Hefei China
- Department of Food Science and Engineering, College of Agronomy Liaocheng University Liaocheng China
| | - Ragini Singh
- Department of Food Science and Engineering, College of Agronomy Liaocheng University Liaocheng China
| | - Yong Ye
- School of Food and Biological Engineering, Engineering Research Center of Bio‐process, Ministry of Education Hefei University of Technology Hefei China
| | - Shuang Cheng
- Department of Food Science and Engineering, College of Agronomy Liaocheng University Liaocheng China
| | - Feng Kong
- Department of Food Science and Engineering, College of Agronomy Liaocheng University Liaocheng China
| | - Qingmei Zeng
- School of Food and Biological Engineering, Engineering Research Center of Bio‐process, Ministry of Education Hefei University of Technology Hefei China
| |
Collapse
|
5
|
Cantoni O, Zito E, Fiorani M, Guidarelli A. Arsenite impinges on endoplasmic reticulum-mitochondria crosstalk to elicit mitochondrial ROS formation and downstream toxicity. Semin Cancer Biol 2021; 76:132-138. [PMID: 34089843 DOI: 10.1016/j.semcancer.2021.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 02/08/2023]
Abstract
Arsenite is an important carcinogen and toxic compound, causing various deleterious effects through multiple mechanisms. In this review, we focused on mitochondrial ROS (mitoROS) and discussed on the mechanisms mediating their formation. The metalloid promotes direct effects in mitochondria, resulting in superoxide formation only under conditions of increased mitochondrial Ca2+ concentration ([Ca2+]m). In this perspective, the time of exposure and concentration requirements for arsenite were largely conditioned by other effects of the metalloid in specific sites of the endoplasmic reticulum (ER). Arsenite induced a slow and limited mobilization of Ca2+ from IP3R via a saturable mechanism, failing to increase the [Ca2+]m. This effect was however associated with the triggering of an intraluminal crosstalk between the IP3R and the ryanodine receptor (RyR), causing a large and concentration dependent release of Ca2+ from RyR and a parallel increase in [Ca2+]m. Thus, the Ca2+-dependent mitoO2-. formation appears to be conditioned by the spatial/functional organization of the ER/mitochondria network and RyR expression. We also speculate on the possibility that the ER stress response might regulate the above effects on the intraluminal crosstalk between the IP3R and the RyR via oxidation of critical thiols mediated by the H2O2 locally released by oxidoreductin 1α.
Collapse
Affiliation(s)
- Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Mara Fiorani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
6
|
Tsukahara T, Sahara Y, Ribeiro N, Tsukahara R, Gotoh M, Sakamoto S, Handa H, Murakami-Murofushi K. Adenine nucleotide translocase 2, a putative target protein for 2-carba cyclic phosphatidic acid in microglial cells. Cell Signal 2021; 82:109951. [PMID: 33592249 DOI: 10.1016/j.cellsig.2021.109951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/12/2021] [Accepted: 01/30/2021] [Indexed: 11/28/2022]
Abstract
Lipid-protein interactions play essential roles in many biological phenomena. Lysophospholipid mediators, such as cyclic phosphatidic acid (cPA), have been recognized as secondary messengers, yet few cellular targets for cPA have been identified to date. Furthermore, the molecular mechanism that activates these downstream signaling events remains unknown. In this study, using metabolically stabilized cPA carba-derivative (2ccPA)-immobilized magnetic beads, we identified adenine nucleotide translocase 2 (ANT2) as a 2ccPA-interacting protein in microglial cells. 2ccPA was tested for its ability to inhibit apoptosis caused by phenylarsine oxide in microglial cells. This damage was significantly improved upon 2ccPA treatment, along with cell proliferation, apoptosis, reactive oxygen species production, and intracellular ATP levels. This is the first report to suggest the direct binding of 2ccPA to ANT2 in microglial cells and provides evidence for a new benefit of 2ccPA in protecting microglial cells from apoptotic death induced by the ANT2-mediated signaling pathway.
Collapse
Affiliation(s)
- Tamotsu Tsukahara
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - Yasuka Sahara
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | - Ryoko Tsukahara
- Ochadai Academic Production, Ochanomizu University, Tokyo, Japan
| | - Mari Gotoh
- Institute for Human Life Innovation, Ochanomizu University, Tokyo, Japan
| | - Satoshi Sakamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Hiroshi Handa
- Department of Chemical Biology, Tokyo Medical University, Tokyo, Japan
| | | |
Collapse
|
7
|
Macasoi I, Mioc A, Mioc M, Racoviceanu R, Soica I, Chevereșan A, Dehelean C, Dumitrașcu V. Targeting Mitochondria through the Use of Mitocans as Emerging Anticancer Agents. Curr Med Chem 2020; 27:5730-5757. [DOI: 10.2174/0929867326666190712150638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/19/2019] [Accepted: 06/11/2019] [Indexed: 01/10/2023]
Abstract
Mitochondria are key players with a multi-functional role in many vital cellular processes,
such as energy metabolism, redox regulation, calcium homeostasis, Reactive Oxygen Species
(ROS) as well as in cell signaling, survival and apoptosis. These functions are mainly regulated
through important enzyme signaling cascades, which if altered may influence the outcome of cell
viability and apoptosis. Therefore some of the key enzymes that are vital for these signaling pathways
are emerging as important targets for new anticancer agent development. Mitocans are compounds
aimed at targeting mitochondria in cancer cells by altering mitochondrial functions thus
causing cell growth inhibition or apoptosis. This review summarizes the till present known classes
of mitocans, their mechanism of action and potential therapeutic use in different forms of cancer.
Collapse
Affiliation(s)
- Ioana Macasoi
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, 2 Eftimie Murgu, Timisoara, Romania
| | - Alexandra Mioc
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, 2 Eftimie Murgu, Timisoara, Romania
| | - Marius Mioc
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, 2 Eftimie Murgu, Timisoara, Romania
| | - Roxana Racoviceanu
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, 2 Eftimie Murgu, Timisoara, Romania
| | - Irina Soica
- Earlscliffe Sixth Form, Earlscliffe, 29 Shorncliffe Road, Folkestone, CT20 2NB, United Kingdom
| | - Adelina Chevereșan
- Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2 Eftimie Murgu, Timisoara, Romania
| | - Cristina Dehelean
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, 2 Eftimie Murgu, Timisoara, Romania
| | - Victor Dumitrașcu
- Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2 Eftimie Murgu, Timisoara, Romania
| |
Collapse
|
8
|
Pichler G, Grau-Perez M, Tellez-Plaza M, Umans J, Best L, Cole S, Goessler W, Francesconi K, Newman J, Redon J, Devereux R, Navas-Acien A. Association of Arsenic Exposure With Cardiac Geometry and Left Ventricular Function in Young Adults. Circ Cardiovasc Imaging 2020; 12:e009018. [PMID: 31060373 DOI: 10.1161/circimaging.119.009018] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Arsenic exposure has been related to numerous adverse cardiovascular outcomes. The aim of this study was to investigate the cross-sectional and prospective association between arsenic exposure with echocardiographic measures of left ventricular (LV) geometry and functioning. METHODS A total of 1337 young adult participants free of diabetes mellitus and cardiovascular disease were recruited from the SHFS (Strong Heart Family Study). The sum of inorganic and methylated arsenic concentrations in urine (ΣAs) at baseline was used as a biomarker of arsenic exposure. LV geometry and functioning were assessed using transthoracic echocardiography at baseline and follow-up. RESULTS Mean follow-up was 5.6 years, and median (interquartile range) of ΣAs was 4.2 (2.8-6.9) µg/g creatinine. Increased arsenic exposure was associated with prevalent LV hypertrophy, with an odds ratio (95% CI) per a 2-fold increase in ΣAs of 1.47 (1.05-2.08) in all participants and of 1.58 (1.04-2.41) among prehypertensive or hypertensive individuals. Measures of LV geometry, including LV mass index, left atrial systolic diameter, interventricular septum, and LV posterior wall thickness, were positively and significantly related to arsenic exposure. Among measures of LV functioning, stroke volume, and ejection fraction were associated with arsenic exposure. CONCLUSIONS Arsenic exposure was related to an increase in LV wall thickness and LV hypertrophy in young American Indians with a low burden of cardiovascular risk factors. The relationship was stronger in participants with prehypertension or hypertension, suggesting that potential cardiotoxic effects of arsenic might be more pronounced in individuals already undergoing cardiovascular adaptive mechanisms following elevated systemic blood pressure.
Collapse
Affiliation(s)
- Gernot Pichler
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, NY (G.P., M.G.-P., A.N.-A.).,Area of Cardiometabolic and Renal Risk, Institute for Biomedical Research INCLIVA, Valencia, Spain (G.P., M.G.-P., M.T.-P., J.R.).,Division of Cardiology, Department of Internal Medicine, Hospital Hietzing, Vienna, Austria (G.P.)
| | - Maria Grau-Perez
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, NY (G.P., M.G.-P., A.N.-A.).,Area of Cardiometabolic and Renal Risk, Institute for Biomedical Research INCLIVA, Valencia, Spain (G.P., M.G.-P., M.T.-P., J.R.).,Department of Statistics and Operational Research, University of Valencia, Spain (M.G.-P.)
| | - Maria Tellez-Plaza
- Area of Cardiometabolic and Renal Risk, Institute for Biomedical Research INCLIVA, Valencia, Spain (G.P., M.G.-P., M.T.-P., J.R.).,Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (M.T.-P.).,Department of Chronic Diseases Epidemiology, National Center for Epidemiology, National Institutes for Health Carlos III, Madrid, Spain (M.T.-P.)
| | - Jason Umans
- MedStar Health Research Institute, and Georgetown University (J.U.).,Georgetown-Howard Universities Center for Clinical and Translational Science, Washington DC (J.U.)
| | - Lyle Best
- Missouri Breaks Industries Research, Inc, Timber Lake (L.B.)
| | - Shelley Cole
- Department of Genetics, Texas Biomedical Research Institute, San Antonio (S.C.)
| | - Walter Goessler
- Institute of Chemistry-Analytical Chemistry, University of Graz, Austria (W.G., K.F.)
| | - Kevin Francesconi
- Institute of Chemistry-Analytical Chemistry, University of Graz, Austria (W.G., K.F.)
| | - Jonathan Newman
- Division of Cardiology and Center for the Prevention of Cardiovascular Disease, Department of Medicine, New York University School of Medicine, NY (J.N.)
| | - Josep Redon
- Area of Cardiometabolic and Renal Risk, Institute for Biomedical Research INCLIVA, Valencia, Spain (G.P., M.G.-P., M.T.-P., J.R.).,CIBER 03/06 Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain (J.R.)
| | | | - Ana Navas-Acien
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, NY (G.P., M.G.-P., A.N.-A.)
| |
Collapse
|
9
|
Jahangirnejad R, Goudarzi M, Kalantari H, Najafzadeh H, Rezaei M. Subcellular Organelle Toxicity Caused by Arsenic Nanoparticles in Isolated Rat Hepatocytes. THE INTERNATIONAL JOURNAL OF OCCUPATIONAL AND ENVIRONMENTAL MEDICINE 2020; 11:41-52. [PMID: 31905194 PMCID: PMC7024596 DOI: 10.15171/ijoem.2020.1614] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 11/24/2019] [Indexed: 11/09/2022]
Abstract
BACKGROUND Arsenic, an environmental pollutant, is a carcinogenic metalloid and also an anticancer agent. OBJECTIVE To evaluate the toxicity of arsenic nanoparticles in rat hepatocytes. METHODS Freshly isolated rat hepatocytes were exposed to 0, 20, 40, and 100 μM of arsenic nanoparticles and its bulk counterpart. Their viability, reactive oxygen species level, glutathione depletion, mitochondrial and lysosomal damage, and apoptosis were evaluated. RESULTS By all concentrations, lysosomal damage and apoptosis were clearly evident in hepatocytes exposed to arsenic nanoparticles. Evaluation of mitochondria and lysosomes revealed that lysosomes were highly damaged. CONCLUSION Exposure to arsenic nanoparticles causes apoptosis and organelle impairment. The nanoparticles have potentially higher toxicity than the bulk arsenic. Lysosomes are highly affected. It seems that, instead of mitochondria, lysosomes are the first target organelles involved in the toxicity induced by arsenic nanoparticles.
Collapse
Affiliation(s)
- Rashid Jahangirnejad
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehdi Goudarzi
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Heibatullah Kalantari
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Najafzadeh
- Department of Pharmacology, Faculty of Veterinary Medicine, Shahid Chamran University, Ahvaz, Iran
| | - Mohsen Rezaei
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
10
|
Optimisation of Folate-Mediated Liposomal Encapsulated Arsenic Trioxide for Treating HPV-Positive Cervical Cancer Cells In Vitro. Int J Mol Sci 2019; 20:ijms20092156. [PMID: 31052347 PMCID: PMC6539325 DOI: 10.3390/ijms20092156] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/28/2019] [Accepted: 04/29/2019] [Indexed: 11/28/2022] Open
Abstract
High-risk human papilloma virus (HPV) infection is directly associated with cervical cancer development. Arsenic trioxide (ATO), despite inducing apoptosis in HPV-infected cervical cancer cells in vitro, has been compromised by toxicity and poor pharmacokinetics in clinical trials. Therefore, to improve ATO’s therapeutic profile for HPV-related cancers, this study aims to explore the effects of length of ligand spacers of folate-targeted liposomes on the efficiency of ATO delivery to HPV-infected cells. Fluorescent ATO encapsulated liposomes with folic acid (FA) conjugated to two different PEG lengths (2000 Da and 5000 Da) were synthesised, and their cellular uptake was examined for HPV-positive HeLa and KB and HPV-negative HT-3 cells using confocal microscopy, flow cytometry, and spectrophotometer readings. Cellular arsenic quantification and anti-tumour efficacy was evaluated through inductively coupled plasma-mass spectrometry (ICP-MS) and cytotoxicity studies, respectively. Results showed that liposomes with a longer folic acid-polyethylene glycol (FA-PEG) spacer (5000 Da) displayed a higher efficiency in targeting folate receptor (FR) + HPV-infected cells without increasing any inherent cytotoxicity. Targeted liposomally delivered ATO also displayed superior selectivity and efficiency in inducing higher cell apoptosis in HPV-positive cells per unit of arsenic taken up than free ATO, in contrast to HT-3. These findings may hold promise in improving the management of HPV-associated cancers.
Collapse
|
11
|
Yang S, Shergalis A, Lu D, Kyani A, Liu Z, Ljungman M, Neamati N. Design, Synthesis, and Biological Evaluation of Novel Allosteric Protein Disulfide Isomerase Inhibitors. J Med Chem 2019; 62:3447-3474. [PMID: 30759340 DOI: 10.1021/acs.jmedchem.8b01951] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein disulfide isomerase (PDI) is responsible for nascent protein folding in the endoplasmic reticulum (ER) and is critical for glioblastoma survival. To improve the potency of lead PDI inhibitor BAP2 (( E)-3-(3-(4-hydroxyphenyl)-3-oxoprop-1-en-1-yl)benzonitrile), we designed and synthesized 67 analogues. We determined that PDI inhibition relied on the A ring hydroxyl group of the chalcone scaffold and cLogP increase in the sulfonamide chain improved potency. Docking studies revealed that BAP2 and analogues bind to His256 in the b' domain of PDI, and mutation of His256 to Ala abolishes BAP2 analogue activity. BAP2 and optimized analogue 59 have modest thiol reactivity; however, we propose that PDI inhibition by BAP2 analogues depends on the b' domain. Importantly, analogues inhibit glioblastoma cell growth, induce ER stress, increase expression of G2M checkpoint proteins, and reduce expression of DNA repair proteins. Cumulatively, our results support inhibition of PDI as a novel strategy to treat glioblastoma.
Collapse
Affiliation(s)
- Suhui Yang
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center , University of Michigan , North Campus Research Complex, 1600 Huron Parkway , Ann Arbor , Michigan 48109 , United States
| | - Andrea Shergalis
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center , University of Michigan , North Campus Research Complex, 1600 Huron Parkway , Ann Arbor , Michigan 48109 , United States
| | - Dan Lu
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center , University of Michigan , North Campus Research Complex, 1600 Huron Parkway , Ann Arbor , Michigan 48109 , United States
| | - Anahita Kyani
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center , University of Michigan , North Campus Research Complex, 1600 Huron Parkway , Ann Arbor , Michigan 48109 , United States
| | - Ziwei Liu
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center , University of Michigan , North Campus Research Complex, 1600 Huron Parkway , Ann Arbor , Michigan 48109 , United States
| | - Mats Ljungman
- Department of Radiation Oncology Rogel Cancer Center , University of Michigan Medical School and Rogel Cancer Center, School of Public Health , Ann Arbor , Michigan 48109 , United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center , University of Michigan , North Campus Research Complex, 1600 Huron Parkway , Ann Arbor , Michigan 48109 , United States
| |
Collapse
|
12
|
Liang H, Xu J, Wang W. Ran1 is essential for parental macronuclear import of apoptosis-inducing factor and programmed nuclear death in Tetrahymena thermophila. FEBS J 2019; 286:913-929. [PMID: 30663224 DOI: 10.1111/febs.14761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/30/2018] [Accepted: 01/17/2019] [Indexed: 01/30/2023]
Abstract
During programmed nuclear death (PND), apoptosis-inducing factor (AIF) translocates from mitochondria to the parental macronucleus (MAC) in Tetrahymena thermophila. In the degenerating parental MAC, AIF induces chromatin condensation and large-scale DNA fragmentation in a caspase-independent manner. However, the regulation of AIF nuclear translocation and molecular mechanism of PND are less clear. In this study, we demonstrated that the asymmetric distribution of nuclear GDP-bound Ran1-mimetic mutant Ran1T25N and cytoplasmic GTP-bound Ran1-mimetic mutant Ran1Q70L exists across the parental macronuclear-cytoplasmic barrier during PND. Knockdown of RAN1 led to defects in PND progression and failure of parental macronuclear accumulation of AIF. Moreover, AIF parental macronuclear import occurred in Ran1T25N mutants, while it was inhibited in Ran1Q70L mutants. Importantly, artificial accumulation of AIF in the parental MAC rescued PND progression defects in RAN1 knockdown mutants. These data suggest that Ran1 is essential for parental macronuclear import of AIF and PND in T. thermophila.
Collapse
Affiliation(s)
- Haixia Liang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China.,MicroNano System Research Center, Key Laboratory of Advanced Transducers and Intelligent Control System of Ministry of Education and Shanxi Province, College of Information & Computer Engineering, Taiyuan University of Technology, China
| | - Jing Xu
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Wei Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| |
Collapse
|
13
|
Drzeżdżon J, Jacewicz D, Chmurzyński L. The impact of environmental contamination on the generation of reactive oxygen and nitrogen species - Consequences for plants and humans. ENVIRONMENT INTERNATIONAL 2018; 119:133-151. [PMID: 29957355 DOI: 10.1016/j.envint.2018.06.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/15/2018] [Accepted: 06/16/2018] [Indexed: 05/23/2023]
Abstract
Environmental contaminants, such as heavy metals, nanomaterials, and pesticides, induce the formation of reactive oxygen and nitrogen species (RONS). Plants interact closely with the atmosphere, water, and soil, and consequently RONS intensely affect their biochemistry. For the past 30 years researchers have thoroughly examined the role of RONS in plant organisms and oxidative modifications to cellular components. Hydrogen peroxide, superoxide anion, nitrogen(II) oxide, and hydroxyl radicals have been found to take part in many metabolic pathways. In this review the various aspects of the oxidative stress induced by environmental contamination are described based on an analysis of literature. The review reinforces the contention that RONS play a dual role, that is, both a deleterious and a beneficial one, in plants. Environmental contamination affects human health, also, and so we have additionally described the impact of RONS on the coupled human - environment system.
Collapse
Affiliation(s)
- Joanna Drzeżdżon
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Dagmara Jacewicz
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland.
| | - Lech Chmurzyński
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| |
Collapse
|
14
|
Abstract
The field of Traditional Chinese Medicine (TCM) represents a vast and largely untapped resource for modern medicine. Exemplified by the success of the antimalarial artemisinin, the recent years have seen a rapid increase in the understanding and application of TCM-derived herbs and formulations for evidence-based therapy. In this review, we summarise and discuss the developmental history, clinical background and molecular basis of an action for several representative TCM-derived medicines, including artemisinin, arsenic trioxide, berberine and Salvia miltiorrhiza or Danshen. Through this, we highlight important examples of how TCM-derived medicines have already contributed to modern medicine, and discuss potential avenues for further research.
Collapse
|
15
|
Thymoquinone alleviates arsenic induced hippocampal toxicity and mitochondrial dysfunction by modulating mPTP in Wistar rats. Biomed Pharmacother 2018; 102:1152-1160. [PMID: 29710533 DOI: 10.1016/j.biopha.2018.03.159] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022] Open
Abstract
Arsenic is a pervasive environmental pollutant that is found in ground waters globally and is related to numerous morbidities in the high-risk population areas in countries including Bangladesh, India, and the USA. Arsenic exposure has been ubiquitously reported for exacerbating free radical generation, mitochondrial dysfunction, and apoptosis by interfering with the mPTP functioning. Over the past decades, nutraceuticals with antioxidant properties have shown promising efficacy in arsenic poisoning. In the present study, we have examined, the protective efficacy of thymoquinone (TQ), an active component of seed oil of Nigella sativa with antioxidant and anti-inflammatory activity on arsenic-induced toxicity in hippocampi of Wistar rats. In our results, arsenic conditioning (10 mg/kg b.wt.; p.o.) for 8 days has caused a significant increase in intracellular ROS generation, mitochondrial dysfunction and apoptotic events. On the contrary pretreatment with TQ (2.5 and 5 mg/kg b.wt.; p.o.) inhibited arsenic-induced mitochondrial dysfunction such as lowering of mitochondrial membrane potential (Δψm). Our results indicated that the neuroprotective efficacy of TQ in arsenic-induced stress is mediated through or in part by inhibition of mPTP opening. Demonstration of neuroprotective action of TQ provides insight into the pathogenesis of arsenic-related neuropathological morbidities.
Collapse
|
16
|
Bodaghi-Namileh V, Sepand MR, Omidi A, Aghsami M, Seyednejad SA, Kasirzadeh S, Sabzevari O. Acetyl-l-carnitine attenuates arsenic-induced liver injury by abrogation of mitochondrial dysfunction, inflammation, and apoptosis in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 58:11-20. [PMID: 29278859 DOI: 10.1016/j.etap.2017.12.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 12/10/2017] [Accepted: 12/11/2017] [Indexed: 06/07/2023]
Abstract
Industrial and agricultural developments in recent years have resulted in the excessive discharge of arsenic into the environment, making arsenic toxicity a major worldwide concern. Oxidative stress is considered the primary mechanism for arsenic toxicity. The main objective of this study was to evaluate acetyl-l-carnitine's (ALC) protective ability against the arsenic-induced hepatotoxicity. For this purpose, male Wistar rats were distributed randomly into 5 groups of 8 rats each: control, arsenic (5 mg/kg) and arsenic plus ALC (5 mg/kg; 100, 200, 300 mg/kg). The animals were gavaged for 21 consecutive days. Liver tissue samples were extracted 24 h after the last treatment and were later analyzed for biochemical and histological alterations. The arsenic-induced oxidative damage was confirmed by elevation of malondialdehyde (MDA), a lipid peroxidation byproduct, as well as depletion in physiological antioxidant content such as superoxide dismutase (SOD) and catalase (CAT). Furthermore, alterations in mitochondrial functions including a significant decrease of mitochondrial outer membrane potential and reactive oxygen species (ROS) generation increase, mitochondrial swelling, release of cytochrome c and consequent activation of caspase-3 and caspase-9 and initiation of apoptosis, was observed following arsenic administration. Moreover, the inflammation was confirmed by the overexpression of inflammatory mediators such as NF-ĸB and IL-1 and IL-6. The present study demonstrated that ALC ameliorates arsenic-induced oxidative damage, mitochondrial dysfunction, apoptosis, inflammation and histological damage. ALC's protective features against arsenic hepatotoxicity may be due to this agent's antioxidant and anti-inflammatory properties as well as its stabilizing effects on mitochondrial function.
Collapse
Affiliation(s)
- Vida Bodaghi-Namileh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Sepand
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ameneh Omidi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Aghsami
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Afshin Seyednejad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Kasirzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Sabzevari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Toxicology and Poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran; Drug Design and Development Research Centre, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Evaluation of serum arsenic and its effects on antioxidant alterations in relapsing-remitting multiple sclerosis patients. Mult Scler Relat Disord 2017; 19:79-84. [PMID: 29156301 DOI: 10.1016/j.msard.2017.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 11/02/2017] [Accepted: 11/12/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Environmental factors that are involved in the development of autoimmune diseases include bacteria, viruses, and xenobiotics such as chemicals, drugs, and metals. Regarding the metals, a number of studies have demonstrated that oxidative stress is one of the well-directed pathways of arsenic-induced tissue damages. This study was designed to explore the serum concentrations of arsenic and its correlation with markers associated with oxidative stress in relapsing-remitting MS (RRMS) patients. METHODS This case-controlled study comprised 50 patients with RRMS and 50 healthy subjects. Serum arsenic levels, total antioxidant potential, malondialdehyde (MDA), and lactate levels were measured. RESULTS The arsenic value, MDA, and lactate levels were elevated meaningfully while FRAP level significantly was decreased in RRMS patients with respect to healthy subjects (P <0.05). Furthermore, arsenic serum levels were positively correlated with serum concentrations of MDA and lactate. In contrast, serum levels were negatively correlated to FRAP values in RRMS patients. CONCLUSION Taken together, the association between arsenic level and oxidative stress parameters supports the hypothesis that high serum arsenic levels may play a critical role in the pathogenesis of MS progression.
Collapse
|
18
|
Henderson MW, Madenspacher JH, Whitehead GS, Thomas SY, Aloor JJ, Gowdy KM, Fessler MB. Effects of Orally Ingested Arsenic on Respiratory Epithelial Permeability to Bacteria and Small Molecules in Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:097024. [PMID: 28960179 PMCID: PMC5915208 DOI: 10.1289/ehp1878] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 08/14/2017] [Accepted: 08/16/2017] [Indexed: 05/27/2023]
Abstract
BACKGROUND Arsenic exposure via drinking water impacts millions of people worldwide. Although arsenic has been associated epidemiologically with increased lung infections, the identity of the lung cell types targeted by peroral arsenic and the associated immune mechanisms remain poorly defined. OBJECTIVES We aimed to determine the impact of peroral arsenic on pulmonary antibacterial host defense. METHODS Female C57BL/6 mice were administered drinking water with 0, 250 ppb, or 25 ppm sodium arsenite for 5 wk and then challenged intratracheally with Klebsiella pneumoniae, Streptococcus pneumoniae, or lipopolysaccharide. Bacterial clearance and immune responses were profiled. RESULTS Arsenic had no effect on bacterial clearance in the lung or on the intrapulmonary innate immune response to bacteria or lipopolysaccharide, as assessed by neutrophil recruitment to, and cytokine induction in, the airspace. Alveolar macrophage TNFα production was unaltered. By contrast, arsenic-exposed mice had significantly reduced plasma TNFα in response to systemic lipopolysaccharide challenge, together suggesting that the local airway innate immune response may be relatively preserved from arsenic intoxication. Despite intact intrapulmonary bacterial clearance during pneumonia, arsenic-exposed mice suffered dramatically increased bacterial dissemination to the bloodstream. Mechanistically, this was linked to increased respiratory epithelial permeability, as revealed by intratracheal FITC-dextran tracking, serum Club Cell protein 16 measurement, and other approaches. Consistent with barrier disruption at the alveolar level, arsenic-exposed mice had evidence for alveolar epithelial type 1 cell injury. CONCLUSIONS Peroral arsenic has little effect on local airway immune responses to bacteria but compromises respiratory epithelial barrier integrity, increasing systemic translocation of inhaled pathogens and small molecules. https://doi.org/10.1289/EHP1878.
Collapse
Affiliation(s)
- Michael W Henderson
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services , Research Triangle Park, North Carolina, USA
| | - Jennifer H Madenspacher
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services , Research Triangle Park, North Carolina, USA
| | - Gregory S Whitehead
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services , Research Triangle Park, North Carolina, USA
| | - Seddon Y Thomas
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services , Research Triangle Park, North Carolina, USA
| | - Jim J Aloor
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services , Research Triangle Park, North Carolina, USA
| | - Kymberly M Gowdy
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University , Greenville, North Carolina, USA
| | - Michael B Fessler
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services , Research Triangle Park, North Carolina, USA
| |
Collapse
|
19
|
Yousefsani BS, Pourahmad J, Hosseinzadeh H. The mechanism of protective effect of crocin against liver mitochondrial toxicity caused by arsenic III. Toxicol Mech Methods 2017; 28:105-114. [DOI: 10.1080/15376516.2017.1368054] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Bahareh Sadat Yousefsani
- Department of Pharmacodynamy and Toxicology, School of Pharmacy, Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamy and Toxicology, School of Pharmacy, Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Guzman-Villanueva D, Weissig V. Mitochondria-Targeted Agents: Mitochondriotropics, Mitochondriotoxics, and Mitocans. Handb Exp Pharmacol 2017; 240:423-438. [PMID: 27590226 DOI: 10.1007/164_2016_37] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mitochondria, the powerhouse of the cell, have been known for many years for their central role in the energy metabolism; however, extensive progress has been made and to date substantial evidence demonstrates that mitochondria play a critical role not only in the cell bioenergetics but also in the entire cell metabolome. Mitochondria are also involved in the intracellular redox poise, the regulation of calcium homeostasis, and the generation of reactive oxygen species (ROS), which are crucial for the control of a variety of signaling pathways. Additionally, they are essential for the mitochondrial-mediated apoptosis process. Thus, it is not surprising that disruptions of mitochondrial functions can lead or be associated with human pathologies. Because of diseases like diabetes, Alzheimer, Parkinson's, cancer, and ischemic disease are being increasingly linked to mitochondrial dysfunctions, the interest in mitochondria as a prime pharmacological target has dramatically risen over the last decades and as a consequence a large number of agents, which could potentially impact or modulate mitochondrial functions, are currently under investigation. Based on their site of action, these agents can be classified as mitochondria-targeted and non-mitochondria-targeted agents. As a result of the continuous search for new agents and the design of potential therapeutic agents to treat mitochondrial diseases, terms like mitochondriotropics, mitochondriotoxics, mitocancerotropics, and mitocans have emerged to describe those agents with high affinity to mitochondria that exert a therapeutic or deleterious effect on these organelles. In this chapter, mitochondria-targeted agents and some strategies to deliver agents to and/or into mitochondria will be reviewed.
Collapse
Affiliation(s)
- Diana Guzman-Villanueva
- Department of Pharmaceutical Sciences, Nanomedicine Center of Excellence in Translational Cancer Research, Midwestern University College of Pharmacy-Glendale, Glendale, AZ, 85308, USA.
| | - Volkmar Weissig
- Department of Pharmaceutical Sciences, Nanomedicine Center of Excellence in Translational Cancer Research, Midwestern University College of Pharmacy-Glendale, Glendale, AZ, 85308, USA
| |
Collapse
|
21
|
Parker LJ, Bocedi A, Ascher DB, Aitken JB, Harris HH, Lo Bello M, Ricci G, Morton CJ, Parker MW. Glutathione transferase P1-1 as an arsenic drug-sequestering enzyme. Protein Sci 2016; 26:317-326. [PMID: 27863446 DOI: 10.1002/pro.3084] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/09/2016] [Accepted: 11/14/2016] [Indexed: 11/07/2022]
Abstract
Arsenic-based compounds are paradoxically both poisons and drugs. Glutathione transferase (GSTP1-1) is a major factor in resistance to such drugs. Here we describe using crystallography, X-ray absorption spectroscopy, mutagenesis, mass spectrometry, and kinetic studies how GSTP1-1 recognizes the drug phenylarsine oxide (PAO). In conditions of cellular stress where glutathione (GSH) levels are low, PAO crosslinks C47 to C101 of the opposing monomer, a distance of 19.9 Å, and causes a dramatic widening of the dimer interface by approximately 10 Å. The GSH conjugate of PAO, which forms rapidly in cancerous cells, is a potent inhibitor (Ki = 90 nM) and binds as a di-GSH complex in the active site forming part of a continuous network of interactions from one active site to the other. In summary, GSTP1-1 can detoxify arsenic-based drugs by sequestration at the active site and at the dimer interface, in situations where there is a plentiful supply of GSH, and at the reactive cysteines in conditions of low GSH.
Collapse
Affiliation(s)
- Lorien J Parker
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Alessio Bocedi
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - David B Ascher
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia
| | - Jade B Aitken
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Hugh H Harris
- Department of Chemistry, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Mario Lo Bello
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Giorgio Ricci
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Craig J Morton
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia
| | - Michael W Parker
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
22
|
Zhang X, Zhou T, Frabutt DA, Zheng YH. HIV-1 Vpr increases Env expression by preventing Env from endoplasmic reticulum-associated protein degradation (ERAD). Virology 2016; 496:194-202. [PMID: 27343732 DOI: 10.1016/j.virol.2016.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 06/06/2016] [Indexed: 11/29/2022]
Abstract
Vpr enhances HIV-1 replication in macrophages and dendritic cells, as well as the human CD4(+) CEM.NKR T cell line. Recently, Vpr was reported to increase HIV-1 Env expression in macrophages. Here, we report that Vpr also increases HIV-1 Env expression in dendritic cells and CEM.NKR cells. The Vpr activity depends on its N-terminal region, which was disrupted by a single A30L mutation. Env was rapidly degraded in the absence of Vpr, which was blocked by the ERAD pathway inhibitor kifunesine or the lysosome inhibitor Bafilomycin. As2O3 or PK11195, which reportedly enhances HIV-1 Env folding, also blocked the Env degradation in CEM.NKR cells. Thus, these results not only identify Env as a primary target for Vpr to boost HIV-1 replication, but also suggest that Vpr likely promotes Env folding in the ER, which is otherwise misfolded and targeted by the ERAD pathway to lysosomes for degradation.
Collapse
Affiliation(s)
- Xianfeng Zhang
- Harbin Veterinary Research Institute, CAAS-Michigan State University Joint Laboratory of Innate Immunity, State Key Laboratory of Veterinary Biotechnology, Chinese Academy of Agricultural Sciences, Harbin 150001, China; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Tao Zhou
- Harbin Veterinary Research Institute, CAAS-Michigan State University Joint Laboratory of Innate Immunity, State Key Laboratory of Veterinary Biotechnology, Chinese Academy of Agricultural Sciences, Harbin 150001, China; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Dylan A Frabutt
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Yong-Hui Zheng
- Harbin Veterinary Research Institute, CAAS-Michigan State University Joint Laboratory of Innate Immunity, State Key Laboratory of Veterinary Biotechnology, Chinese Academy of Agricultural Sciences, Harbin 150001, China; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
23
|
Flora SJS. Arsenic and dichlorvos: Possible interaction between two environmental contaminants. J Trace Elem Med Biol 2016; 35:43-60. [PMID: 27049126 DOI: 10.1016/j.jtemb.2016.01.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 01/04/2016] [Accepted: 01/26/2016] [Indexed: 02/01/2023]
Abstract
Metals are ubiquitously present in the environment and pesticides are widely used throughout the world. Environmental and occupational exposure to metal along with pesticide is an area of great concern to both the public and regulatory authorities. Our major concern is that combination of these toxicant present in environment may elicit toxicity either due to additive or synergistic interactions or 'joint toxic actions' among these toxicants. It poses a rising threat to human health. Water contamination particularly ground water contamination with arsenic is a serious problem in today's scenario since arsenic is associated with several kinds of health problems, such arsenic associated health anomalies are commonly called as 'Arsenism'. Uncontrolled use and spillage of pesticides into the environment has resulted in alarming situation. Moreover serious concerns are being addressed due to their persistence in the environmental matrices such as air, soil and surface water runoff resulting in continuous exposure of these harmful chemicals to human beings and animals. Bio-availability of these environmental toxicants has been enhanced much due to anthropological activities. Dreadfully very few studies are available on combined exposures to these toxicants on the animal or human system. Studies on the acute and chronic exposure to arsenic and DDVP are well reported and well defined. Arsenic is a common global ground water contaminant while dichlorvos is one of the most commonly and widely employed organophosphate based insecticide used in agriculture, horticulture etc. There is thus a real situation where a human may get exposed to these toxicants while working in a field. This review highlights the individual and combined exposure to arsenic and dichlorvos on health.
Collapse
Affiliation(s)
- Swaran J S Flora
- Division of Regulatory Toxicology, Defence Research and Development Establishment, Jhansi Road, Gwalior 474002, India.
| |
Collapse
|
24
|
Mao G, Zhou Z, Chen Y, Wang W, Wu X, Feng W, Cobbina SJ, Huang J, Zhang Z, Xu H, Yang L, Wu X. Neurological Toxicity of Individual and Mixtures of Low Dose Arsenic, Mono and Di (n-butyl) Phthalates on Sub-Chronic Exposure to Mice. Biol Trace Elem Res 2016; 170:183-93. [PMID: 26257159 DOI: 10.1007/s12011-015-0457-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/26/2015] [Indexed: 11/24/2022]
Abstract
The objective of this study was to evaluate the toxicity of individual and mixtures of di(n-butyl) phthalates (DBP) and their active metabolite monobutyl phthalate (MBP) and arsenic (As) on spatial cognition associated with hippocampal apoptosis in mice. Mice were exposed, individually or in combination, to DBP (50 mg/kg body weight, intragastrically), MBP (50 mg/kg body weight, intragastrically), and As (10 mg/L, per os) for 8 weeks. The Morris water maze test showed that mice exposed to DBP/MBP combined with As exhibited longer escape latencies and the lower average number of crossing the platform. The As content in the hippocampus after As exposure increased as compared to those without As exposure. In mice exposed to DBP/MBP combined with As, pathological alterations and oxidative damage to the hippocampus were found. Expression of apoptosis-related protein: Bax and caspase-3 were significantly increased in the hippocampus, while there was no significant change in expression of Bcl-2. The results suggested that DBP and MBP combined with As can induce spatial cognitive deficits through altering the expression of apoptosis-related protein and As played a critical role in cognition impairments. And the joint exposure has antagonistic effect.
Collapse
Affiliation(s)
- Guanghua Mao
- School of the Environment and Safety Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China
| | - Zhaoxiang Zhou
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China
| | - Yao Chen
- School of the Environment and Safety Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China
| | - Wei Wang
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China
| | - Xueshan Wu
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China
| | - Weiwei Feng
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China
| | - Samuel Jerry Cobbina
- School of the Environment and Safety Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China
| | - Jing Huang
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China
| | - Zhen Zhang
- School of the Environment and Safety Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China
| | - Hai Xu
- School of the Environment and Safety Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China
| | - Liuqing Yang
- School of the Environment and Safety Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China.
| | - Xiangyang Wu
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Rd., 212013, Zhenjiang, Jiangsu, China.
| |
Collapse
|
25
|
Mandal S, Chaudhuri K. Engineered magnetic core shell nanoprobes: Synthesis and applications to cancer imaging and therapeutics. World J Biol Chem 2016; 7:158-167. [PMID: 26981204 PMCID: PMC4768120 DOI: 10.4331/wjbc.v7.i1.158] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 09/23/2015] [Accepted: 12/04/2015] [Indexed: 02/05/2023] Open
Abstract
Magnetic core shell nanoparticles are composed of a highly magnetic core material surrounded by a thin shell of desired drug, polymer or metal oxide. These magnetic core shell nanoparticles have a wide range of applications in biomedical research, more specifically in tissue imaging, drug delivery and therapeutics. The present review discusses the up-to-date knowledge on the various procedures for synthesis of magnetic core shell nanoparticles along with their applications in cancer imaging, drug delivery and hyperthermia or cancer therapeutics. Literature in this area shows that magnetic core shell nanoparticle-based imaging, drug targeting and therapy through hyperthermia can potentially be a powerful tool for the advanced diagnosis and treatment of various cancers.
Collapse
|
26
|
Alamolhodaei NS, Shirani K, Karimi G. Arsenic cardiotoxicity: An overview. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:1005-14. [PMID: 26606645 DOI: 10.1016/j.etap.2015.08.030] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/25/2015] [Accepted: 08/30/2015] [Indexed: 05/20/2023]
Abstract
Arsenic, a naturally ubiquitous element, is found in foods and environment. Cardiac dysfunction is one of the major causes of morbidity and mortality in the world. Arsenic exposure is associated with various cardiopathologic effects including ischemia, arrhythmia and heart failure. Possible mechanisms of arsenic cardiotoxicity include oxidative stress, DNA fragmentation, apoptosis and functional changes of ion channels. Several evidences have shown that mitochondrial disruption, caspase activation, MAPK signaling and p53 are the pathways for arsenic induced apoptosis. Arsenic trioxide is an effective and potent antitumor agent used in patients with acute promyelocytic leukemia and produces dramatic remissions. As2O3 administration has major limitations such as T wave changes, QT prolongation and sudden death in humans. In this review, we discuss the underlying pathobiology of arsenic cardiotoxicity and provide information about cardiac health effects associated with some medicinal plants in arsenic toxicity.
Collapse
Affiliation(s)
| | - Kobra Shirani
- Department of Pharmacodynamy and Toxicology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Gholamreza Karimi
- Pharmaceutical Research Center and Pharmacy School, Mashhad University of Medical Sciences, Iran.
| |
Collapse
|
27
|
Prakash C, Soni M, Kumar V. Mitochondrial oxidative stress and dysfunction in arsenic neurotoxicity: A review. J Appl Toxicol 2015; 36:179-88. [DOI: 10.1002/jat.3256] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 09/01/2015] [Accepted: 09/28/2015] [Indexed: 01/19/2023]
Affiliation(s)
- Chandra Prakash
- Department of Biochemistry; Maharshi Dayanand University; Rohtak 124001 Haryana India
| | - Manisha Soni
- Department of Biochemistry; Maharshi Dayanand University; Rohtak 124001 Haryana India
| | - Vijay Kumar
- Department of Biochemistry; Maharshi Dayanand University; Rohtak 124001 Haryana India
| |
Collapse
|
28
|
Chau D, Ng K, Chan TSY, Cheng YY, Fong B, Tam S, Kwong YL, Tse E. Azacytidine sensitizes acute myeloid leukemia cells to arsenic trioxide by up-regulating the arsenic transporter aquaglyceroporin 9. J Hematol Oncol 2015; 8:46. [PMID: 25953102 PMCID: PMC4431177 DOI: 10.1186/s13045-015-0143-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/27/2015] [Indexed: 11/23/2022] Open
Abstract
Background The therapeutic efficacy of arsenic trioxide (As2O3) in acute myeloid leukemia (AML) is modest, which is partly related to its limited intracellular uptake into the leukemic cells. As2O3 enters cells via the transmembrane protein aquaglyceroporin 9 (AQP9). Azacytidine, a demethylating agent that is approved for the treatment of AML, has been shown to have synergistic effect with As2O3. We tested the hypothesis that azacytidine might up-regulate AQP9 and enhances As2O3-mediated cytotoxicity in AML. Methods Arsenic-induced cytotoxicity, the expression of AQP9, and the intracellular uptake of As2O3 were determined in AML cell lines and primary AML cells with or without azacytidine pre-treatment. The mechanism of AQP9 up-regulation was then investigated by examining the expression of transcription factors for AQP9 gene and the methylation status of their gene promoters. Results As2O3-induced cytotoxicity in AML cell lines was significantly enhanced after azacytidine pre-treatment as a result of AQP9 up-regulation, leading to increased arsenic uptake and hence intracellular concentration. Blocking AQP9-mediated As2O3 uptake with mercury chloride abrogated the sensitization effect of azacytidine. AQP9 promoter does not contain CpG islands. Instead, azacytidine pre-treatment led to increased expression of HNF1A, a transcription activator of AQP9, through demethylation of HNF1A promoter. HNF1 knockdown abrogated azacytidine-induced AQP9 up-regulation and almost completely blocked intracellular As2O3 entry, confirming that azacytidine enhanced As2O3-mediated cell death via up-regulation of HNF1A and hence increased AQP9 and As2O3 intracellular concentration. Azacytidine sensitization to As2O3 treatment was re-capitulated also in primary AML samples. Finally, azacytidine did not enhance arsenic toxicity in a liver cell line, where HNF1A was largely unmethylated. Conclusions Azacytidine sensitizes AML cells to As2O3 treatment, and our results provide proof-of-principle evidence that pharmacological up-regulation of AQP9 potentially expands the therapeutic spectrum of As2O3. Further clinical trial should evaluate the efficacy of azacytidine in combination with As2O3 in the treatment of AML. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0143-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David Chau
- Division of Haematology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pok Fu Lam, Hong Kong, Hong Kong.
| | - Karen Ng
- Division of Haematology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pok Fu Lam, Hong Kong, Hong Kong.
| | - Thomas Sau-Yan Chan
- Division of Haematology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pok Fu Lam, Hong Kong, Hong Kong.
| | - Yuen-Yee Cheng
- Division of Haematology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pok Fu Lam, Hong Kong, Hong Kong. .,Asbestos Diseases Research Institute, ADRI Bernie Banton Centre, University of Sydney, Concord Hospital, Sydney, Australia.
| | - Bonnie Fong
- Department of Pathology and Clinical Biochemistry, Queen Mary Hospital, Pok Fu Lam, Hong Kong, Hong Kong.
| | - Sidney Tam
- Department of Pathology and Clinical Biochemistry, Queen Mary Hospital, Pok Fu Lam, Hong Kong, Hong Kong.
| | - Yok-Lam Kwong
- Division of Haematology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pok Fu Lam, Hong Kong, Hong Kong.
| | - Eric Tse
- Division of Haematology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pok Fu Lam, Hong Kong, Hong Kong.
| |
Collapse
|
29
|
Guidarelli A, Fiorani M, Azzolini C, Cerioni L, Scotti M, Cantoni O. U937 cell apoptosis induced by arsenite is prevented by low concentrations of mitochondrial ascorbic acid with hardly any effect mediated by the cytosolic fraction of the vitamin. Biofactors 2015; 41:101-10. [PMID: 25809564 DOI: 10.1002/biof.1204] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/12/2015] [Indexed: 02/03/2023]
Abstract
Arsenite directly triggers cytochrome c and Smac/DIABLO release in mitochondria isolated from U937 cells. These effects were not observed in mitochondria pre-exposed for 15 min to 10 µM L-ascorbic acid (AA). In other experiments, intact cells treated for 24-72 h with arsenite were found to die by apoptosis through a mechanism involving mitochondrial permeability transition. Pre-exposure (15 min) to low micromolar concentrations of AA and dehydroascorbic acid (DHA), resulting in identical cytosolic levels of the vitamin, had a diverse impact on cell survival, as cytoprotection was only observed after treatment with AA. Also the mitochondrial accumulation of the vitamin was restricted to AA exposure. An additional indication linking cytoprotection to the mitochondrial fraction of the vitamin was obtained in experiments measuring susceptibility to arsenite in parallel with loss of mitochondrial and cytosolic AA at different times after vitamin exposure. Finally, we took advantage of our recent findings that DHA potently inhibits AA transport to demonstrate that DHA abolishes all the protective effects of AA, under the same conditions in which the mitochondrial accumulation of the vitamin is prevented without affecting the overall cellular accumulation of the vitamin.
Collapse
Affiliation(s)
- Andrea Guidarelli
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino, Italy
| | | | | | | | | | | |
Collapse
|
30
|
|
31
|
Liu G, Middleton RJ, Hatty CR, Kam WW, Chan R, Pham T, Harrison‐Brown M, Dodson E, Veale K, Banati RB. The 18 kDa translocator protein, microglia and neuroinflammation. Brain Pathol 2014; 24:631-53. [PMID: 25345894 PMCID: PMC8029074 DOI: 10.1111/bpa.12196] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/19/2014] [Indexed: 12/17/2022] Open
Abstract
The 18 kDa translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is expressed in the injured brain. It has become known as an imaging marker of "neuroinflammation" indicating active disease, and is best interpreted as a nondiagnostic biomarker and disease staging tool that refers to histopathology rather than disease etiology. The therapeutic potential of TSPO as a drug target is mostly based on the understanding that it is an outer mitochondrial membrane protein required for the translocation of cholesterol, which thus regulates the rate of steroid synthesis. This pivotal role together with the evolutionary conservation of TSPO has underpinned the belief that any loss or mutation of TSPO should be associated with significant physiological deficits or be outright incompatible with life. However, against prediction, full Tspo knockout mice are viable and across their lifespan do not show the phenotype expected if cholesterol transport and steroid synthesis were significantly impaired. Thus, the "translocation" function of TSPO remains to be better substantiated. Here, we discuss the literature before and after the introduction of the new nomenclature for TSPO and review some of the newer findings. In light of the controversy surrounding the function of TSPO, we emphasize the continued importance of identifying compounds with confirmed selectivity and suggest that TSPO expression is analyzed within specific disease contexts rather than merely equated with the reified concept of "neuroinflammation."
Collapse
Affiliation(s)
- Guo‐Jun Liu
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Ryan J. Middleton
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Claire R. Hatty
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Winnie Wai‐Ying Kam
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Ronald Chan
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Tien Pham
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Meredith Harrison‐Brown
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Eoin Dodson
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Kelly Veale
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Richard B. Banati
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
- National Imaging Facility and Ramaciotti Brain Imaging CentreSydneyNSWAustralia
| |
Collapse
|
32
|
Miltonprabu S, Sumedha NC. Arsenic-induced hepatic mitochondrial toxicity in rats and its amelioration by diallyl trisulfide. Toxicol Mech Methods 2014; 24:124-35. [PMID: 24295472 DOI: 10.3109/15376516.2013.869778] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The present investigation was aimed to investigate the possible protective role of diallyl trisulfide (DATS) against arsenic (As)-induced hepatic mitochondrial toxicity in rats. Mitochondria were isolated from the liver tissue of rats from all the groups. Lipid profile, lipid peroxidation, antioxidant enzyme activities, hepatic function enzymes, mitochondrial swelling, cytochrome c oxidase activity, mitochondrial Ca(+)-ATPase and Na(+)/K(+)-ATPase activity, mitochondrial calcium content and mitochondrial enzyme activities were measured. Short-term As exposure (5 mg/kg bw/d for 28 d) caused liver damage as evidenced by changes in activities of liver enzymes. The effects of As were coupled with enhanced reactive oxygen species generation, mitochondrial swelling, inhibition of cytochrome c oxidase, complex I-mediated electron transfer, decreased Ca(2+)-ATPase and Na(+)/K(+)-ATPase activity, a reduction in mitochondrial calcium content, changes in indices of hepatic mitochondrial oxidative stress, significant increase in mitochondrial lipid peroxidation products and alterations in mitochondrial lipid profile. Significant decreases in mitochondrial antioxidants and tricarboxylic acid cycle enzymes were also found in the liver mitochondria of As-induced hepatic mitochondrial toxicity in rats. As also increased hepatic caspase-3 activity and DNA fragmentation. All these apoptosis-related molecular changes caused by As could be alleviated by supplementation with DATS, which likely suggests a protective role against As-induced hepatotoxic changes and hepatic mitochondrial toxicity. The protective effect of DATS on the liver mitochondria was evidenced by altering all the changes induced by As. Free radical scavenging and metal chelating activities of DATS may be the mechanism, responsible for the protective action against As-induced mitochondrial damage in liver.
Collapse
Affiliation(s)
- S Miltonprabu
- Department of Zoology, Faculty of Science, Annamalai University , Annamalainagar, Tamilnadu , India
| | | |
Collapse
|
33
|
Abstract
The field of mitochondrial ion channels has recently seen substantial progress, including the molecular identification of some of the channels. An integrative approach using genetics, electrophysiology, pharmacology, and cell biology to clarify the roles of these channels has thus become possible. It is by now clear that many of these channels are important for energy supply by the mitochondria and have a major impact on the fate of the entire cell as well. The purpose of this review is to provide an up-to-date overview of the electrophysiological properties, molecular identity, and pathophysiological functions of the mitochondrial ion channels studied so far and to highlight possible therapeutic perspectives based on current information.
Collapse
|
34
|
Todorova T, Vuilleumier S, Kujumdzieva A. Role of Glutathione S-Transferases and Glutathione in Arsenic and Peroxide Resistance inSaccharomyces Cerevisiae:A Reverse Genetic Analysis Approach. BIOTECHNOL BIOTEC EQ 2014. [DOI: 10.1080/13102818.2007.10817472] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
35
|
Wu Y, Dai J, Zhang W, Yan R, Zhang Y, Ruan C, Dai K. Arsenic trioxide induces apoptosis in human platelets via C-Jun NH2-terminal kinase activation. PLoS One 2014; 9:e86445. [PMID: 24466103 PMCID: PMC3899281 DOI: 10.1371/journal.pone.0086445] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 12/10/2013] [Indexed: 01/18/2023] Open
Abstract
Arsenic trioxide (ATO), one of the oldest drugs in both Western and traditional Chinese medicine, has become an effective anticancer drug, especially in the treatment of acute promyelocytic leukemia (APL). However, thrombocytopenia occurred in most of ATO-treated patients with APL or other malignant diseases, and the pathogenesis remains unclear. Here we show that ATO dose-dependently induces depolarization of mitochondrial inner transmembrane potential (ΔΨm), up-regulation of Bax and down-regulation of Bcl-2 and Bcl-XL, caspase-3 activation, and phosphotidylserine (PS) exposure in platelets. ATO did not induce surface expression of P-selectin and PAC-1 binding, whereas, obviously reduced collagen, ADP, and thrombin induced platelet aggregation. ATO dose-dependently induced c-Jun NH2-terminal kinase (JNK) activation, and JNK specific inhibitor dicumarol obviously reduced ATO-induced ΔΨm depolarization in platelets. Clinical therapeutic dosage of ATO was intraperitoneally injected into C57 mice, and the numbers of circulating platelets were significantly reduced after five days of continuous injection. The data demonstrate that ATO induces caspase-dependent apoptosis via JNK activation in platelets. ATO does not incur platelet activation, whereas, it not only impairs platelet function but also reduces circulating platelets in vivo, suggesting the possible pathogenesis of thrombocytopenia in patients treated with ATO.
Collapse
Affiliation(s)
- Yicun Wu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| | - Jin Dai
- School of Life Sciences, Peking University, Beijing, China
| | - Weilin Zhang
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| | - Rong Yan
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| | - Yiwen Zhang
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| | - Changgeng Ruan
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| | - Kesheng Dai
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
- * E-mail:
| |
Collapse
|
36
|
The mitochondrial translocator protein, TSPO, inhibits HIV-1 envelope glycoprotein biosynthesis via the endoplasmic reticulum-associated protein degradation pathway. J Virol 2014; 88:3474-84. [PMID: 24403586 DOI: 10.1128/jvi.03286-13] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The HIV-1 Env glycoprotein is folded in the endoplasmic reticulum (ER), which is necessary for viral entry and replication. Currently, it is still unclear how this process is regulated. The glycoprotein folding in the ER is controlled by the ER-associated protein degradation (ERAD) pathway, which specifically targets misfolded proteins for degradation. Previously, we reported that HIV-1 replication is restricted in the human CD4(+) T cell line CEM.NKR (NKR). To understand this mechanism, we first analyzed cellular protein expression in NKR cells and discovered that levels of the mitochondrial translocator protein TSPO were upregulated by ∼64-fold. Notably, when NKR cells were treated with TSPO antagonist PK-11195, Ro5-4864, or diazepam, HIV restriction was completely disrupted, and TSPO knockdown by short hairpin RNAs (shRNAs) achieved a similar effect. We next analyzed viral protein expression, and, interestingly, we discovered that Env expression was specifically inhibited. Both TSPO knockdown and treatment with TSPO antagonist could restore Env expression in NKR cells. We further discovered that Env proteins were rapidly degraded and that kifunensine, an ERAD pathway inhibitor, could restore Env expression and viral replication, indicating that Env proteins were misfolded and degraded through the ERAD pathway in NKR cells. We also knocked out the TSPO gene in 293T cells using CRISPR/Cas9 (clustered, regularly interspaced, short palindromic repeat [CRISPR]/CRISPR-associated-9) technology and found that TSPO could similarly inhibit Env expression in these cells. Taken together, these results demonstrate that TSPO inhibits Env protein expression through the ERAD pathway and suggest that mitochondria play an important role in regulating the Env folding process. IMPORTANCE The HIV-1 Env glycoprotein is absolutely required for viral infection, and an understanding of its expression pathway in infected cells will identify new targets for antiretroviral therapies. Env proteins are folded in the ER and secreted through the classical secretory pathway. The Env folding process involves extensive cross-linking of 10 Cys residues by disulfide bond formation and heavy N-glycosylation on ∼30 Asn residues. Currently, it is still unclear how this process is regulated. Here, we studied this mechanism in the HIV nonpermissive human CD4(+) T cell line CEM.NKR. We found that Env proteins were rapidly degraded through a cellular pathway that specifically targets misfolded proteins, resulting in inhibition of Env expression. Importantly, we have identified a mitochondrial translocator protein, TSPO, which could trigger this degradation by interfering with the Env folding process. Further characterization of TSPO antiviral activity will reveal a novel antiretroviral mechanism that targets the Env protein.
Collapse
|
37
|
Mandal S, Chatterjee N, Das S, Saha KD, Chaudhuri K. Magnetic core–shell nanoprobe for sensitive killing of cancer cells via induction with a strong external magnetic field. RSC Adv 2014. [DOI: 10.1039/c4ra01407c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The title system, composed of a highly magnetic core surrounded by a thin arsenite shell, has been synthesized and applied to the magnetically facilitated targeting of anticancer agent (sodium arsenite) at lower dose with minimal side effects and higher efficacy in a biocompatible manner.
Collapse
Affiliation(s)
- Samir Mandal
- Molecular and Human Genetics Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700 032, India
| | - Nabanita Chatterjee
- Cancer Biology and Inflammatory Disorder Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata 700032, India
| | - Subhadip Das
- Cancer Biology and Inflammatory Disorder Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata 700032, India
| | - Krishna Das Saha
- Cancer Biology and Inflammatory Disorder Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata 700032, India
| | - Keya Chaudhuri
- Molecular and Human Genetics Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700 032, India
| |
Collapse
|
38
|
Swindell EP, Hankins PL, Chen H, Miodragović ÐU, O'Halloran TV. Anticancer activity of small-molecule and nanoparticulate arsenic(III) complexes. Inorg Chem 2013; 52:12292-304. [PMID: 24147771 PMCID: PMC3893798 DOI: 10.1021/ic401211u] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Starting in ancient China and Greece, arsenic-containing compounds have been used in the treatment of disease for over 3000 years. They were used for a variety of diseases in the 20th century, including parasitic and sexually transmitted illnesses. A resurgence of interest in the therapeutic application of arsenicals has been driven by the discovery that low doses of a 1% aqueous solution of arsenic trioxide (i.e., arsenous acid) lead to complete remission of certain types of leukemia. Since Food and Drug Administration (FDA) approval of arsenic trioxide (As2O3) for treatment of acute promyelocytic leukemia in 2000, it has become a front-line therapy in this indication. There are currently over 100 active clinical trials involving inorganic arsenic or organoarsenic compounds registered with the FDA for the treatment of cancers. New generations of inorganic and organometallic arsenic compounds with enhanced activity or targeted cytotoxicity are being developed to overcome some of the shortcomings of arsenic therapeutics, namely, short plasma half-lives and a narrow therapeutic window.
Collapse
Affiliation(s)
- Elden P. Swindell
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208-3113
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208-3113
| | - Patrick L. Hankins
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208-3113
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208-3113
| | - Haimei Chen
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208-3113
| | - Ðenana U. Miodragović
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208-3113
| | - Thomas V. O'Halloran
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208-3113
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208-3113
| |
Collapse
|
39
|
Ivanov VN, Wen G, Hei TK. Sodium arsenite exposure inhibits AKT and Stat3 activation, suppresses self-renewal and induces apoptotic death of embryonic stem cells. Apoptosis 2013; 18:188-200. [PMID: 23143138 DOI: 10.1007/s10495-012-0779-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Sodium arsenite exposure at concentration >5 μM may induce embryotoxic and teratogenic effects in animal models. Long-term health effects of sodium arsenite from contaminated drinking water may result in different forms of cancer and neurological abnormalities. As cancer development processes seem to be originated in stem cells, we have chosen to examine the effects of sodium arsenite on signaling pathways and the corresponding transcription factors that regulate cell viability and self-renewal in mouse embryonic stem cells (ESC) and mouse neural stem/precursor cells. We demonstrated that the crucial signaling pathway, which was substantially suppressed by sodium arsenite exposure (4 μM) in ESC, was the PI3K-AKT pathway linked with numerous downstream targets that control cell survival and apoptosis. Furthermore, the whole core transcription factor circuitry that control self-renewal of mouse ESC (Stat3-P-Tyr705, Oct4, Sox2 and Nanog) was strongly down-regulated by sodium arsenite (4 μM) exposure. This was followed by G2/M arrest and induction of the mitochondrial apoptotic pathway that might be suppressed by caspase-9 and caspase-3 inhibitors. In contrast to mouse ESC with very low endogenous IL6, mouse neural stem/precursor cells (C17.2 clone immortalized by v-myc) with high endogenous production of IL6 exhibited a strong resistance to cytotoxic effects of sodium arsenite that could be decreased by inhibitory anti-IL6 antibody or Stat3 inhibition. In summary, our data demonstrated suppression of self-renewal and induction of apoptosis in mouse ESC by sodium arsenite exposure, which was further accelerated due to simultaneous inhibition of the protective PI3K-AKT and Stat3-dependent pathways.
Collapse
Affiliation(s)
- Vladimir N Ivanov
- Department of Radiation Oncology, Center for Radiological Research, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| | | | | |
Collapse
|
40
|
Affiliation(s)
- Shengwen Shen
- Department
of Laboratory Medicine
and Pathology, 10-102 Clinical Sciences Building, University
of Alberta, Edmonton, Alberta, Canada, T6G 2G3
| | - Xing-Fang Li
- Department
of Laboratory Medicine
and Pathology, 10-102 Clinical Sciences Building, University
of Alberta, Edmonton, Alberta, Canada, T6G 2G3
| | - William R. Cullen
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver,
British Columbia, Canada, V6T 1Z1
| | - Michael Weinfeld
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta, Canada, T6G 1Z2
| | - X. Chris Le
- Department
of Laboratory Medicine
and Pathology, 10-102 Clinical Sciences Building, University
of Alberta, Edmonton, Alberta, Canada, T6G 2G3
| |
Collapse
|
41
|
Bhujade A, Gupta G, Talmale S, Das SK, Patil MB. Induction of apoptosis in A431 skin cancer cells by Cissus quadrangularis Linn stem extract by altering Bax-Bcl-2 ratio, release of cytochrome c from mitochondria and PARP cleavage. Food Funct 2013; 4:338-46. [PMID: 23175101 DOI: 10.1039/c2fo30167a] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Skin is generally damaged through genetic and environmental factors such as smoking, exposure to xenobiotics, heat, hormonal changes, and ultraviolet light. These factors can cause skin diseases. Cissus quadrangularis Linn. (CQ) has been used in folk medicine for the treatment of skin diseases since ancient times. Taking in to consideration the medicinal properties exhibited by this genus, it was decided to investigate the anti-cancer activity of CQ. Extracts obtained from CQ and their phenolic contents were subjected to in vitro evaluation of anticancer activity by using A431 (skin epidermoid carcinoma, human) cell line. The A431 cells were treated with different extracts of CQ in a dose dependent manner. Out of five extracts, the acetone extract demonstrated significant anti-cancer activity in the A431 cell line. Hexane, chloroform, ethyl acetate and methanol extracts also exhibited cytotoxicity but to a comparatively lesser extent than the acetone extract. The GI(50) value of the acetone extract was found to be 8 μg mL(-1), whereas GI(50) value of purified fraction of acetone extract, termed as AFCQ (active acetone fraction of CQ) with respect to A431 cells, was found to be 4.8 μg mL(-1). Furthermore, the mechanism of anticancer activity exhibited by AFCQ was investigated by comparing its effect with the standard anticancer drug Doxorubicin (DOX) by evaluating the status of apoptotic markers after treatment of A431 cells with AFCQ and DOX. Bax-Bcl-2 ratio along with the release of cytochrome c from mitochondria to cytoplasm, which is a hallmark of apoptosis, was also evaluated. Cleavage of PARP revealed that AFCQ induces apoptosis in A431 cells with reference to DOX.
Collapse
Affiliation(s)
- Arti Bhujade
- Department of Biochemistry, Nagpur University, LIT Premises, Nagpur, Maharashtra, India.
| | | | | | | | | |
Collapse
|
42
|
Suh DH, Kim MK, Kim HS, Chung HH, Song YS. Mitochondrial permeability transition pore as a selective target for anti-cancer therapy. Front Oncol 2013; 3:41. [PMID: 23483560 PMCID: PMC3592197 DOI: 10.3389/fonc.2013.00041] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 02/12/2013] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial outer membrane permeabilization (MOMP) is the ultimate step in dozens of lethal apoptotic signal transduction pathways which converge on mitochondria. One of the representative systems proposed to be responsible for the MOMP is the mitochondrial permeability transition pore (MPTP). Although the molecular composition of the MPTP is not clearly understood, the MPTP attracts much interest as a promising target for resolving two conundrums regarding cancer treatment: tumor selectivity and resistance to treatment. The regulation of the MPTP is closely related to metabolic reprogramming in cancer cells including mitochondrial alterations. Restoration of deregulated apoptotic machinery in cancer cells by tumor-specific modulation of the MPTP could therefore be a promising anti-cancer strategy. Currently, a number of MPTP-targeting agents are under pre-clinical and clinical studies. Here, we reviewed the structure and regulation of the MPTP as well as the current status of the development of promising MPTP-targeting drugs.
Collapse
Affiliation(s)
- Dong H. Suh
- Department of Obstetrics and Gynecology, Seoul National University College of MedicineSeoul, South Korea
| | - Mi-Kyung Kim
- Department of Obstetrics and Gynecology, Seoul National University College of MedicineSeoul, South Korea
| | - Hee S. Kim
- Department of Obstetrics and Gynecology, Seoul National University College of MedicineSeoul, South Korea
| | - Hyun H. Chung
- Department of Obstetrics and Gynecology, Seoul National University College of MedicineSeoul, South Korea
| | - Yong S. Song
- Department of Obstetrics and Gynecology, Seoul National University College of MedicineSeoul, South Korea
- Cancer Research Institute, Seoul National University College of MedicineSeoul, South Korea
- Major in Biomodulation, World Class University, Seoul National UniversitySeoul, South Korea
| |
Collapse
|
43
|
Arsenic trioxide stabilizes accumulations of adeno-associated virus virions at the perinuclear region, increasing transduction in vitro and in vivo. J Virol 2013; 87:4571-83. [PMID: 23408604 DOI: 10.1128/jvi.03443-12] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Interactions with cellular stress pathways are central to the life cycle of many latent viruses. Here, we utilize adeno-associated virus (AAV) as a model to study these interactions, as previous studies have demonstrated that cellular stressors frequently increase transduction of recombinant AAV (rAAV) vectors and may even substitute for helper virus functions. Since several chemotherapeutic drugs are known to increase rAAV transduction, we investigated the effect of arsenic trioxide (As(2)O(3)), an FDA-approved chemotherapeutic agent with known effects on several other virus life cycles, on the transduction of rAAV. In vitro, As(2)O(3) caused a dose-dependent increase in rAAV2 transduction over a broad range of cell lines from various cell types and species (e.g., HEK-293, HeLa, HFF hTERT, C-12, and Cos-1). Mechanistically, As(2)O(3) treatment acted to prevent loss of virions from the perinuclear region, which correlated with increased cellular vector genome retention, and was distinguishable from proteasome inhibition. To extend our investigation of the cellular mechanism, we inhibited reactive oxygen species formation and determined that the As(2)O(3)-mediated increase in rAAV2 transduction was dependent upon production of reactive oxygen species. To further validate our in vitro data, we tested the effect of As(2)O(3) on rAAV transduction in vivo and determined that treatment initiated transgene expression as early as 2 days posttransduction and increased reporter expression by up to 10-fold. Moreover, the transduction of several other serotypes of rAAV was also enhanced in vivo, suggesting that As(2)O(3) affects a pathway used by several AAV serotypes. In summary, our data support a model wherein As(2)O(3) increases rAAV transduction both in vitro and in vivo and maintains perinuclear accumulations of capsids, facilitating productive nuclear trafficking.
Collapse
|
44
|
Bhattacharjee P, Chatterjee D, Singh KK, Giri AK. Systems biology approaches to evaluate arsenic toxicity and carcinogenicity: an overview. Int J Hyg Environ Health 2013; 216:574-86. [PMID: 23340121 DOI: 10.1016/j.ijheh.2012.12.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 12/11/2012] [Accepted: 12/13/2012] [Indexed: 01/08/2023]
Abstract
Long term exposure to arsenic, either through groundwater, food stuff or occupational sources, results in a plethora of dermatological and non-dermatological health effects including multi-organ cancer and early mortality. Several epidemiological studies, across the globe have reported arsenic-induced health effects and cancerous outcomes; but the prevalence of such diseases varies depending on environmental factors (geographical location, exposure level), and genetic makeup (and variants thereof); which is further modulated by several other factors like ethnicity, age-sex, smoking status, diet, etc. It is also interesting to note that, chronic arsenic exposure to a similar extent, even among the same family members, result in wide inter-individual variations. To understand the adverse effect of this toxic metabolite on biological system (cellular targets), and to unravel the underlying molecular basis (at the level of transcript, proteome, or metabolite), a holistic, systems biology approach was taken. Due to the paradoxical nature and unavailability of any suitable animal model system; the literature review is primarily based on cell line and population based studies. Thus, here we present a comprehensive review on the systems biology approaches to explore the underlying mechanism of arsenic-induced carcinogenicity, along with our own observations and an overview of mitigation strategies and their effectiveness till date.
Collapse
Affiliation(s)
- Pritha Bhattacharjee
- Molecular and Human Genetics Division, Indian Institute of Chemical Biology, Kolkata, India
| | | | | | | |
Collapse
|
45
|
Shoshan-Barmatz V, Mizrachi D. VDAC1: from structure to cancer therapy. Front Oncol 2012; 2:164. [PMID: 23233904 PMCID: PMC3516065 DOI: 10.3389/fonc.2012.00164] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Accepted: 10/24/2012] [Indexed: 12/14/2022] Open
Abstract
Here, we review current evidence pointing to the function of VDAC1 in cell life and death, and highlight these functions in relation to cancer. Found at the outer mitochondrial membrane, VDAC1 assumes a crucial position in the cell, controlling the metabolic cross-talk between mitochondria and the rest of the cell. Moreover, its location at the boundary between the mitochondria and the cytosol enables VDAC1 to interact with proteins that mediate and regulate the integration of mitochondrial functions with other cellular activities. As a metabolite transporter, VDAC1 contributes to the metabolic phenotype of cancer cells. This is reflected by VDAC1 over-expression in many cancer types, and by inhibition of tumor development upon silencing VDAC1 expression. Along with regulating cellular energy production and metabolism, VDAC1 is also a key protein in mitochondria-mediated apoptosis, participating in the release of apoptotic proteins and interacting with anti-apoptotic proteins. The involvement of VDAC1 in the release of apoptotic proteins located in the inter-membranal space is discussed, as is VDAC1 oligomerization as an important step in apoptosis induction. VDAC also serves as an anchor point for mitochondria-interacting proteins, some of which are also highly expressed in many cancers, such as hexokinase (HK), Bcl2, and Bcl-xL. By binding to VDAC, HK provides both metabolic benefit and apoptosis-suppressive capacity that offers the cell a proliferative advantage and increases its resistance to chemotherapy. VDAC1-based peptides that bind specifically to HK, Bcl2, or Bcl-xL abolished the cell’s abilities to bypass the apoptotic pathway. Moreover, these peptides promote cell death in a panel of genetically characterized cell lines derived from different human cancers. These and other functions point to VDAC1 as a rational target for the development of a new generation of therapeutics.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev Beer-Sheva, Israel ; The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev Beer-Sheva, Israel
| | | |
Collapse
|
46
|
Evidence for Vpr-dependent HIV-1 replication in human CD4+ CEM.NKR T-cells. Retrovirology 2012; 9:93. [PMID: 23134572 PMCID: PMC3528630 DOI: 10.1186/1742-4690-9-93] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Accepted: 10/11/2012] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Vpr is exclusively expressed in primate lentiviruses and contributes to viral replication and disease progression in vivo. HIV-1 Vpr has two major activities in vitro: arrest of cell cycle in the G2 phase (G2 arrest), and enhancement of viral replication in macrophages. Previously, we reported a potent HIV-1 restriction in the human CD4+ CEM.NKR (NKR) T cells, where wild-type (WT) HIV-1 replication was inhibited by almost 1,000-fold. From the parental NKR cells, we isolated eight clones by limiting dilution. These clones showed three levels of resistance to the WT HIV-1 infection: non-permissive (NP), semi-permissive (SP), and permissive (P). Here, we compared the replication of WT, Vif-defective, Vpr-defective, and Vpu-defective viruses in these cells. RESULTS Although both WT and Vpu-defective viruses could replicate in the permissive and semi-permissive clones, the replication of Vif-defective and Vpr-defective viruses was completely restricted. The expression of APOBEC3G (A3G) cytidine deaminase in NKR cells explains why Vif, but not Vpr, was required for HIV-1 replication. When the Vpr-defective virus life cycle was compared with the WT virus life cycle in the semi-permissive cells, it was found that the Vpr-defective virus could enter the cell and produce virions containing properly processed Gag and Env proteins, but these virions showed much less efficiency for reverse transcription during the next-round of infection. In addition, although viral replication was restricted in the non-permissive cells, treatment with arsenic trioxide (As2O3) could completely restore WT, but not Vpr-defective virus replication. Moreover, disruption of Vpr binding to its cofactor DCAF1 and/or induction of G2 arrest activity did not disrupt the Vpr activity in enhancing HIV-1 replication in NKR cells. CONCLUSIONS These results demonstrate that HIV-1 replication in NKR cells is Vpr-dependent. Vpr promotes HIV-1 replication from the 2nd cycle likely by overcoming a block at early stage of viral replication; and this activity does not require DCAF1 and G2 arrest. Further studies of this mechanism should provide new understanding of Vpr function in the HIV-1 life cycle.
Collapse
|
47
|
Naranmandura H, Chen X, Tanaka M, Wang WW, Rehman K, Xu S, Chen Z, Chen SQ, Suzuki N. Release of Apoptotic Cytochrome c From Mitochondria by Dimethylarsinous Acid Occurs Through Interaction With Voltage-Dependent Anion Channel In Vitro. Toxicol Sci 2012; 128:137-46. [DOI: 10.1093/toxsci/kfs154] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
48
|
Majumdar S, Maiti A, Karmakar S, Das AS, Mukherjee S, Das D, Mitra C. Antiapoptotic efficacy of folic acid and vitamin B₁₂ against arsenic-induced toxicity. ENVIRONMENTAL TOXICOLOGY 2012; 27:351-363. [PMID: 22566245 DOI: 10.1002/tox.20648] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 07/17/2010] [Accepted: 07/20/2010] [Indexed: 05/31/2023]
Abstract
Earlier, we proposed that the ability of folic acid and vitamin B₁₂ to preserve systemic and mitochondrial function after short-term exposure to arsenic may prevent further progression to more permanent injury and pathological changes leading to cell death. To elucidate its mechanism, the present study examined the antiapoptotic efficacy of folic acid and vitamin B₁₂ against short-term arsenic exposure-induced hepatic mitochondria oxidative stress and dysfunction. Sixteen to eighteen weeks old male albino rats weighing 140-150 × g were divided into five groups: Control (A), Arsenic-treated (B), Arsenic + folic acid (C), Arsenic +vitamin B₁₂ (D), and Arsenic + folic acid + vitamin B₁₂ (E). Data generated indicated that folic acid and vitamin B₁₂ separately or in combination can give significant protection against alterations in oxidative stress and apoptotic marker parameters and downstream changes in mitochondria, namely pro-oxidative (NO, TBARS, OH⁻) and antioxidative defense (SOD, CAT, GSH) markers, iNOS protein expression, mitochondrial swelling, cytochrome c oxidase and Ca²⁺-ATPase activity, Ca²⁺ content, caspase-3 activity. Additionally, results of hepatic cell DNA fragmentation, arsenic load of blood, hepatic tissue and urine, and histological observations, all strongly support that both these supplements have efficacy in preventing apoptotic changes and cellular damage. As the mechanisms of actions of both of these supplements are methylation related, a combined application was more effective. Results further reveal new molecular targets through which folic acid and vitamin B₁₂ separately or in combination work to alleviate one critical component of arsenic-induced liver injury: mitochondria dysfunction.
Collapse
Affiliation(s)
- Sangita Majumdar
- Institute of Genetic Medicine and Genomic Science, Madhyamgram, Kolkata, India
| | | | | | | | | | | | | |
Collapse
|
49
|
Breccia M, Lo-Coco F. Arsenic trioxide for management of acute promyelocytic leukemia: current evidence on its role in front-line therapy and recurrent disease. Expert Opin Pharmacother 2012; 13:1031-43. [PMID: 22468778 DOI: 10.1517/14656566.2012.677436] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Acute promyelocytic leukemia (APL), the most rapidly fatal leukemia only two decades ago, has been converted into the most frequently curable leukemia by the advent of all-trans retinoic acid (ATRA) and its combination with anthracycline-based chemotherapy. More recently, arsenic trioxide (ATO) has been shown to be the most effective single agent in this disease and has been approved for the treatment of relapsed patients both in the United States and Europe. Moreover, ATO has been included in the design of several front-line studies, with the aim to reduce therapy-related toxicity while maintaining the potential of cure. AREAS COVERED First, this review briefly discusses the mechanisms of action and the toxicity profile of ATO. Furthermore, the reported experience on the use of ATO as single agent or in combinatorial schemes both in relapsed and in newly diagnosed patients with APL is critically reviewed. Finally, the use of this agent in special subsets of patients unfit to receive conventional chemotherapy is discussed, along with its potential role in maintenance therapy. EXPERT OPINION While the role of ATO as single agent or in combination with ATRA is well established and recommended by the European LeukemiaNet guidelines as a first option for relapsed patients, the role of the drug in newly diagnosed patients is still uncertain and based only on evidence levels mostly originating from non-randomized trials. The results of ongoing randomized studies should better define the role of ATO in front-line therapy.
Collapse
Affiliation(s)
- Massimo Breccia
- Sapienza University, Department of Cellular Biotechnologies and Hematology, Via Benevento 6, 00161 Rome, Italy.
| | | |
Collapse
|
50
|
Wang S, Wu X, Tan M, Gong J, Tan W, Bian B, Chen M, Wang Y. Fighting fire with fire: poisonous Chinese herbal medicine for cancer therapy. JOURNAL OF ETHNOPHARMACOLOGY 2012; 140:33-45. [PMID: 22265747 DOI: 10.1016/j.jep.2011.12.041] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 12/22/2011] [Accepted: 12/23/2011] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Following the known principle of "fighting fire with fire", poisonous Chinese herbal medicine (PCHM) has been historically used in cancer therapies by skilled Chinese practitioners for thousands of years. In fact, most of the marketed natural anti-cancer compounds (e.g., camptothecin derivatives, vinca alkaloids, etc.) are often known in traditional Chinese medicine (TCM) and recorded as poisonous herbs as well. Inspired by the encouraging precedents, significant researches into the potential of novel anticancer drugs from other PCHM-derived natural products have been ongoing for several years and PCHM is increasingly being recognized as a gathering place for promising anti-cancer drugs. The present review aimed at giving a rational understanding of the toxicity of PCHM and, especially, providing the most recent developments on PCHM-derived anti-cancer compounds. MATERIALS AND METHODS Information on the toxicity and safety control of PCHM, as well as PCHM-derived anti-cancer compounds, was gathered from the articles, books and monographs published in the past 20 years. RESULTS Based on an objective introduction to the CHM toxicity, we clarified the general misconceptions about the safety of CHM and summarized the traditional experiences in dealing with the toxicity. Several PCHM-derived compounds, namely gambogic acid, triptolide, arsenic trioxide, and cantharidin, were selected as representatives, and their traditional usage and mechanism of anti-cancer actions were discussed. CONCLUSIONS Natural products derived from PCHM are of extreme importance in devising new drugs and providing unique ideas for the war against cancer. To fully exploit the potential of PCHM in cancer therapy, more attentions are advocated to be focused on their safety evaluation and mechanism exploration.
Collapse
Affiliation(s)
- Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | | | | | | | | | | | | | | |
Collapse
|