1
|
Sandoval TA, Salvagno C, Chae CS, Awasthi D, Giovanelli P, Falco MM, Hwang SM, Teran-Cabanillas E, Suominen L, Yamazaki T, Kuo HH, Moyer JE, Martin ML, Manohar J, Kim K, Sierra MA, Ramos Y, Tan C, Emmanuelli A, Song M, Morales DK, Zamarin D, Frey MK, Cantillo E, Chapman-Davis E, Holcomb K, Mason CE, Galluzzi L, Zhou ZN, Vähärautio A, Cloonan SM, Cubillos-Ruiz JR. Iron Chelation Therapy Elicits Innate Immune Control of Metastatic Ovarian Cancer. Cancer Discov 2024; 14:1901-1921. [PMID: 39073085 PMCID: PMC11452292 DOI: 10.1158/2159-8290.cd-23-1451] [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: 12/01/2023] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024]
Abstract
Iron accumulation in tumors contributes to disease progression and chemoresistance. Although targeting this process can influence various hallmarks of cancer, the immunomodulatory effects of iron chelation in the tumor microenvironment are unknown. Here, we report that treatment with deferiprone, an FDA-approved iron chelator, unleashes innate immune responses that restrain ovarian cancer. Deferiprone reprogrammed ovarian cancer cells toward an immunostimulatory state characterized by the production of type-I IFN and overexpression of molecules that activate NK cells. Mechanistically, these effects were driven by innate sensing of mitochondrial DNA in the cytosol and concomitant activation of nuclear DNA damage responses triggered upon iron chelation. Deferiprone synergized with chemotherapy and prolonged the survival of mice with ovarian cancer by bolstering type-I IFN responses that drove NK cell-dependent control of metastatic disease. Hence, iron chelation may represent an alternative immunotherapeutic strategy for malignancies that are refractory to current T-cell-centric modalities. Significance: This study uncovers that targeting dysregulated iron accumulation in ovarian tumors represents a major therapeutic opportunity. Iron chelation therapy using an FDA-approved agent causes immunogenic stress responses in ovarian cancer cells that delay metastatic disease progression and enhance the effects of first-line chemotherapy. See related commentary by Bell and Zou, p. 1771.
Collapse
Affiliation(s)
- Tito A. Sandoval
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Camilla Salvagno
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Chang-Suk Chae
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Deepika Awasthi
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Paolo Giovanelli
- Weill Cornell Graduate School of Medical Sciences. New York, NY 10065. USA
| | - Matias Marin Falco
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sung-Min Hwang
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Eli Teran-Cabanillas
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Lasse Suominen
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medicine. New York, NY 10065, USA
| | - Hui-Hsuan Kuo
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065. USA
| | - Jenna E. Moyer
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065. USA
| | - M Laura Martin
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065. USA
| | - Jyothi Manohar
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065. USA
| | - Kihwan Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine. New York, NY 10065, USA
| | - Maria A. Sierra
- Weill Cornell Graduate School of Medical Sciences. New York, NY 10065. USA
| | - Yusibeska Ramos
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
| | - Chen Tan
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Alexander Emmanuelli
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
- Weill Cornell Graduate School of Medical Sciences. New York, NY 10065. USA
| | - Minkyung Song
- Departments of Integrative Biotechnology and of Biopharmaceutical Convergence, Sungkyunkwan University. Suwon, Gyeonggi-do, Korea
| | - Diana K. Morales
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
| | - Dmitriy Zamarin
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Melissa K. Frey
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Evelyn Cantillo
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Eloise Chapman-Davis
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Kevin Holcomb
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Christopher E. Mason
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine. New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine. New York, USA
- The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine. New York, NY, USA
| | - Lorenzo Galluzzi
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
- Department of Radiation Oncology, Weill Cornell Medicine. New York, NY 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065. USA
| | - Zhen Ni Zhou
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Anna Vähärautio
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Suzanne M. Cloonan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine. New York, NY 10065, USA
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College. Dublin, Ireland
| | - Juan R. Cubillos-Ruiz
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
- Weill Cornell Graduate School of Medical Sciences. New York, NY 10065. USA
| |
Collapse
|
2
|
Basu T, Kumar B, Shendge AK, Panja S, Chugh H, Gautam HK, Mandal N. An Indian Desert Shrub 'Hiran Chabba', Farsetia hamiltonii Royle, Exhibits Potent Antioxidant and Hepatoprotective Effect Against Iron- Overload Induced Liver Toxicity in Swiss Albino Mice. Curr Drug Discov Technol 2020; 16:210-222. [PMID: 29669498 DOI: 10.2174/1570163815666180418150123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Farsetia hamiltonii Royle, also known as Hiran Chabba grows in desert regions. It is widely used as folk medicine to treat joint pains, diarrhea and diabetes. However, its antioxidant and iron chelation abilities both in vitro and in vivo have not yet been investigated. METHODS The 70% methanolic extract of F. hamiltonii (FHME) was investigated for its free radical scavenging and iron chelation potential, in vitro. An iron-overload situation was established by intraperitoneal injection of iron-dextran in Swiss albino mice, followed by oral administration of FHME. Liver damage and serum parameters due to iron-overload were measured biochemically and histopathologically to test iron-overload remediation and hepatoprotective potential of FHME. Phytochemical analyses were performed to determine its probable bioactive components. RESULTS FHME showed promising antioxidant activity, scavenged various reactive oxygen and nitrogen species and chelated iron in vitro. FHME reduced liver iron, serum ferritin, normalized serum parameters, reduced oxidative stress in liver, serum and improved liver antioxidant status in ironoverloaded mice. It also alleviated liver damage and fibrosis as evident from biochemical parameters and morphological analysis of liver sections. The phytochemical analyses of FHME reflected the presence of alkaloids, phenols, flavonoids and tannins. HPLC analysis indicated presence of tannic acid, quercetin, methyl gallate, catechin, reserpine, ascorbic acid and gallic acid. CONCLUSION Based on the experimental outcome, FHME, an ethnologically important plant can be envisaged as excellent antioxidant and iron chelator drug capable of remediating iron-overload induced hepatotoxicity and the bioactive compounds present in FHME might be responsible for its efficacy.
Collapse
Affiliation(s)
- Tapasree Basu
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme - VII M, Kolkata 700054, West Bengal, India
| | - Bipul Kumar
- Department of Microbial Biotechnology, CSIR- Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
| | - Anil K Shendge
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme - VII M, Kolkata 700054, West Bengal, India
| | - Sourav Panja
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme - VII M, Kolkata 700054, West Bengal, India
| | - Heerak Chugh
- Department of Microbial Biotechnology, CSIR- Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
| | - Hemant K Gautam
- Department of Microbial Biotechnology, CSIR- Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
| | - Nripendranath Mandal
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme - VII M, Kolkata 700054, West Bengal, India
| |
Collapse
|
3
|
Shakya B, Yadav PN. Thiosemicarbazones as Potent Anticancer Agents and their Modes of Action. Mini Rev Med Chem 2020; 20:638-661. [DOI: 10.2174/1389557519666191029130310] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/17/2019] [Accepted: 10/03/2019] [Indexed: 11/22/2022]
Abstract
:Thiosemicarbazones (TSCs) are a class of Schiff bases usually obtained by the condensation of thiosemicarbazide with a suitable aldehyde or ketone. TSCs have been the focus of chemists and biologists due to their wide range of pharmacological effects. One of the promising areas in which these excellent metal chelators are being developed is their use against cancer. TSCs have a wide clinical antitumor spectrum with efficacy in various tumor types such as leukemia, pancreatic cancer, breast cancer, non-small cell lung cancer, cervical cancer, prostate cancer and bladder cancer. To obtain better activity, different series of TSCs have been developed by modifying the heteroaromatic system in their molecules. These compounds possessed significant antineoplastic activity when the carbonyl attachment of the side chain was located at a position α to the ring nitrogen atom, whereas attachment of the side chain β or γ to the heterocyclic N atom resulted in inactive antitumor agents. In addition, replacement of the heterocyclic ring N with C also resulted in a biologically inactive compound suggesting that a conjugated N,N,S-tridentate donor set is essential for the biological activities of thiosemicarbazones. Several possible mechanisms have been implemented for the anticancer activity of thiosemicarbazones.
Collapse
Affiliation(s)
- Bhushan Shakya
- Amrit Campus, Tribhuvan University, Thamel, Kathmandu, Nepal
| | - Paras Nath Yadav
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| |
Collapse
|
4
|
The Role of the Antioxidant Response in Mitochondrial Dysfunction in Degenerative Diseases: Cross-Talk between Antioxidant Defense, Autophagy, and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6392763. [PMID: 31057691 PMCID: PMC6476015 DOI: 10.1155/2019/6392763] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/18/2019] [Accepted: 02/11/2019] [Indexed: 12/29/2022]
Abstract
The mitochondrion is an essential organelle important for the generation of ATP for cellular function. This is especially critical for cells with high energy demands, such as neurons for signal transmission and cardiomyocytes for the continuous mechanical work of the heart. However, deleterious reactive oxygen species are generated as a result of mitochondrial electron transport, requiring a rigorous activation of antioxidative defense in order to maintain homeostatic mitochondrial function. Indeed, recent studies have demonstrated that the dysregulation of antioxidant response leads to mitochondrial dysfunction in human degenerative diseases affecting the nervous system and the heart. In this review, we outline and discuss the mitochondrial and oxidative stress factors causing degenerative diseases, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and Friedreich's ataxia. In particular, the pathological involvement of mitochondrial dysfunction in relation to oxidative stress, energy metabolism, mitochondrial dynamics, and cell death will be explored. Understanding the pathology and the development of these diseases has highlighted novel regulators in the homeostatic maintenance of mitochondria. Importantly, this offers potential therapeutic targets in the development of future treatments for these degenerative diseases.
Collapse
|
5
|
Basu T, Panja S, Shendge AK, Das A, Mandal N. A natural antioxidant, tannic acid mitigates iron-overload induced hepatotoxicity in Swiss albino mice through ROS regulation. ENVIRONMENTAL TOXICOLOGY 2018; 33:603-618. [PMID: 29446234 DOI: 10.1002/tox.22549] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/24/2018] [Accepted: 02/03/2018] [Indexed: 06/08/2023]
Abstract
Tannic acid (TA), a water soluble natural polyphenol with 8 gallic acids groups, is abundantly present in various medicinal plants. Previously TA has been investigated for its antimicrobial and antifungal properties. Being a large polyphenol, TA chelates more than 1 metal. Hence TA has been explored for potent antioxidant activities against reactive oxygen species (ROS), reactive nitrogen species (RNS) and as iron chelator in vitro thereby mitigating iron-overload induced hepatotoxicity in vivo. Iron dextran was injected intraperitoneally in Swiss albino mice to induce iron-overload triggered hepatotoxicity, followed by oral administration of TA for remediation. After treatment, liver, spleen, and blood samples were processed from sacrificed animals. The liver iron, serum ferritin, serum markers, ROS, liver antioxidant status, and liver damage parameters were assessed, followed by histopathology and protein expression studies. Our results show that TA is a prominent ROS and RNS scavenger as well as iron chelator in vitro. It also reversed the ROS levels in vivo and restricted the liver damage parameters as compared to the standard drug, desirox. Moreover, this natural polyphenol exclusively ameliorates the histopathological and fibrotic changes in liver sections reducing the iron-overload, along with chelation of liver iron and normalization of serum ferritin. The protective role of TA against iron-overload induced apoptosis in liver was further supported by changed levels of caspase 3, PARP as well as Bax/BCl-2 ratio. Thus, TA can be envisaged as a better orally administrable iron chelator to reduce iron-overload induced hepatotoxicity through ROS regulation.
Collapse
Affiliation(s)
- Tapasree Basu
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme-VIIM, Kolkata, West Bengal, 700054, India
| | - Sourav Panja
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme-VIIM, Kolkata, West Bengal, 700054, India
| | - Anil Khushalrao Shendge
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme-VIIM, Kolkata, West Bengal, 700054, India
| | - Abhishek Das
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme-VIIM, Kolkata, West Bengal, 700054, India
| | - Nripendranath Mandal
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme-VIIM, Kolkata, West Bengal, 700054, India
| |
Collapse
|
6
|
Badria FA, Ibrahim AS, Badria AF, Elmarakby AA. Curcumin Attenuates Iron Accumulation and Oxidative Stress in the Liver and Spleen of Chronic Iron-Overloaded Rats. PLoS One 2015; 10:e0134156. [PMID: 26230491 PMCID: PMC4521784 DOI: 10.1371/journal.pone.0134156] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/06/2015] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVES Iron overload is now recognized as a health problem in industrialized countries, as excessive iron is highly toxic for liver and spleen. The potential use of curcumin as an iron chelator has not been clearly identified experimentally in iron overload condition. Here, we evaluate the efficacy of curcumin to alleviate iron overload-induced hepatic and splenic abnormalities and to gain insight into the underlying mechanisms. DESIGN AND METHODS Three groups of male adult rats were treated as follows: control rats, rats treated with iron in a drinking water for 2 months followed by either vehicle or curcumin treatment for 2 more months. Thereafter, we studied the effects of curcumin on iron overload-induced lipid peroxidation and anti-oxidant depletion. RESULTS Treatment of iron-overloaded rats with curcumin resulted in marked decreases in iron accumulation within liver and spleen. Iron-overloaded rats had significant increases in malonyldialdehyde (MDA), a marker of lipid peroxidation and nitric oxide (NO) in liver and spleen when compared to control group. The effects of iron overload on lipid peroxidation and NO levels were significantly reduced by the intervention treatment with curcumin (P<0.05). Furthermore, the endogenous anti-oxidant activities/levels in liver and spleen were also significantly decreased in chronic iron overload and administration of curcumin restored the decrease in the hepatic and splenic antioxidant activities/levels. CONCLUSION Our study suggests that curcumin may represent a new horizon in managing iron overload-induced toxicity as well as in pathological diseases characterized by hepatic iron accumulation such as thalassemia, sickle cell anemia, and myelodysplastic syndromes possibly via iron chelation, reduced oxidative stress derived lipid peroxidation and improving the body endogenous antioxidant defense mechanism.
Collapse
Affiliation(s)
- Farid A. Badria
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed S. Ibrahim
- Department of Biochemistry and Clinical Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Adel F. Badria
- Medical Technology Center, Alexandria University, Alexandria 21526, Egypt
- Department of Mechanical and Aeronautical Engineering, University of Patras, Patras, 26500, Greece
| | - Ahmed A. Elmarakby
- Department of Oral Biology, Georgia Regents University, Augusta, Georgia, United States of America
| |
Collapse
|
7
|
Ghate NB, Chaudhuri D, Das A, Panja S, Mandal N. An Antioxidant Extract of the Insectivorous Plant Drosera burmannii Vahl. Alleviates Iron-Induced Oxidative Stress and Hepatic Injury in Mice. PLoS One 2015; 10:e0128221. [PMID: 26010614 PMCID: PMC4444084 DOI: 10.1371/journal.pone.0128221] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/24/2015] [Indexed: 12/02/2022] Open
Abstract
Free iron typically leads to the formation of excess free radicals, and additional iron deposition in the liver contributes to the oxidative pathologic processes of liver disease. Many pharmacological properties of the insectivorous plant Drosera burmannii Vahl. have been reported in previous studies; however, there is no evidence of its antioxidant or hepatoprotective potential against iron overload. The antioxidant activity of 70% methanolic extract of D. burmannii (DBME) was evaluated. DBME showed excellent DPPH, hydroxyl, hypochlorous, superoxide, singlet oxygen, nitric oxide, peroxynitrite radical and hydrogen peroxide scavenging activity. A substantial iron chelation (IC50 = 40.90 ± 0.31 μg/ml) and supercoiled DNA protection ([P]50 = 50.41 ± 0.55 μg) were observed. DBME also displayed excellent in vivo hepatoprotective activity in iron-overloaded Swiss albino mice compared to the standard desirox treatment. Administration of DBME significantly normalized serum enzyme levels and restored liver antioxidant enzymes levels. DBME lowered the raised levels of liver damage parameters, also reflected from the morphological analysis of the liver sections. DBME also reduced liver iron content by 115.90% which is also seen by Perls’ staining. A phytochemical analysis of DBME confirms the presence of various phytoconstituents, including phenols, flavonoids, carbohydrates, tannins, alkaloids and ascorbic acid. Alkaloids, phenols and flavonoids were abundantly found in DBME. An HPLC analysis of DBME revealed the presence of purpurin, catechin, tannic acid, reserpine, methyl gallate and rutin. Purpurin, tannic acid, methyl gallate and rutin displayed excellent iron chelation but exhibited cytotoxicity toward normal (WI-38) cells; while DBME found to be non-toxic to the normal cells. These findings suggest that the constituents present in DBME contributed to its iron chelation activity. Additional studies are needed to determine if DBME can be used as a treatment for iron overload diseases.
Collapse
Affiliation(s)
- Nikhil Baban Ghate
- Division of Molecular Medicine, Bose Institute, P 1/12, Scheme—VIIM, Kolkata, West Bengal, India
| | - Dipankar Chaudhuri
- Division of Molecular Medicine, Bose Institute, P 1/12, Scheme—VIIM, Kolkata, West Bengal, India
| | - Abhishek Das
- Division of Molecular Medicine, Bose Institute, P 1/12, Scheme—VIIM, Kolkata, West Bengal, India
| | - Sourav Panja
- Division of Molecular Medicine, Bose Institute, P 1/12, Scheme—VIIM, Kolkata, West Bengal, India
| | - Nripendranath Mandal
- Division of Molecular Medicine, Bose Institute, P 1/12, Scheme—VIIM, Kolkata, West Bengal, India
- * E-mail:
| |
Collapse
|
8
|
Liu-Snyder P, Webster TJ. Designing drug-delivery systems for the nervous system using nanotechnology: opportunities and challenges. Expert Rev Med Devices 2014; 3:683-7. [PMID: 17280531 DOI: 10.1586/17434440.3.6.683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
9
|
Serda M, Mrozek-Wilczkiewicz A, Jampilek J, Pesko M, Kralova K, Vejsova M, Musiol R, Ratuszna A, Polanski J. Investigation of the biological properties of (hetero)aromatic thiosemicarbazones. Molecules 2012; 17:13483-502. [PMID: 23151918 PMCID: PMC6268061 DOI: 10.3390/molecules171113483] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/07/2012] [Accepted: 11/08/2012] [Indexed: 11/17/2022] Open
Abstract
Two series of thiosemicarbazone-based iron chelators (twenty-seven compounds) were designed and synthesized using a microwave-assisted approach. Quinoline and halogenated phenyl were selected as parent scaffolds on the basis of a similarity search. The lipophilicity of the synthesized compounds was measured using HPLC and then calculated. Primary in vitro screening of the synthesized compounds was performed against eight pathogenic fungal strains. Only a few compounds showed moderate activity against fungi, and (E)-2-(quinolin-2-ylvinyl)-N,N-dimethylhydrazine-carbothioamide appeared to be more effective than fluconazole against most of the fungal strains tested. Antiproliferative activity was measured using a human colon cancer cell line (HCT-116). Several of the tested compounds showed submicromolar antiproliferative activity. Compounds were also tested for their activity related to the inhibition of photosynthetic electron transport (PET) in spinach (Spinacia oleracea L.) chloroplasts. The structure-activity relationships are discussed for all of the compounds.
Collapse
Affiliation(s)
- Maciej Serda
- Department of Organic Chemistry, Institute of Chemistry, University of Silesia, PL-40006 Katowice, Poland
| | - Anna Mrozek-Wilczkiewicz
- Department of Organic Chemistry, Institute of Chemistry, University of Silesia, PL-40006 Katowice, Poland
- Institute of Physics, University of Silesia, PL-40007 Katowice, Poland
| | - Josef Jampilek
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho 1/3, CZ-61242 Brno, Czech Republic
- Research Institute for Pharmacy and Biochemistry, Lidicka 1879/48, CZ-60200 Brno, Czech Republic
| | - Matus Pesko
- Department of Ecosozology and Physiotactics, Faculty of Natural Sciences, Comenius University, Mlynska dolina Ch-2, SK-84215 Bratislava, Slovakia
| | - Katarina Kralova
- Institute of Chemistry, Faculty of Natural Sciences, Comenius University, Mlynska dolina Ch-2, SK-84215 Bratislava, Slovakia
| | - Marcela Vejsova
- Department of Clinical Microbiology, Charles University Medical School and Teaching Hospital, Sokolska 581, CZ-50005 Hradec Kralove, Czech Republic
| | - Robert Musiol
- Department of Organic Chemistry, Institute of Chemistry, University of Silesia, PL-40006 Katowice, Poland
| | - Alicja Ratuszna
- Institute of Physics, University of Silesia, PL-40007 Katowice, Poland
| | - Jaroslaw Polanski
- Department of Organic Chemistry, Institute of Chemistry, University of Silesia, PL-40006 Katowice, Poland
| |
Collapse
|
10
|
Serda M, Kalinowski DS, Mrozek-Wilczkiewicz A, Musiol R, Szurko A, Ratuszna A, Pantarat N, Kovacevic Z, Merlot AM, Richardson DR, Polanski J. Synthesis and characterization of quinoline-based thiosemicarbazones and correlation of cellular iron-binding efficacy to anti-tumor efficacy. Bioorg Med Chem Lett 2012; 22:5527-31. [DOI: 10.1016/j.bmcl.2012.07.030] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/05/2012] [Accepted: 07/06/2012] [Indexed: 01/08/2023]
|
11
|
Lanfranchi DA, Cesar-Rodo E, Bertrand B, Huang HH, Day L, Johann L, Elhabiri M, Becker K, Williams DL, Davioud-Charvet E. Synthesis and biological evaluation of 1,4-naphthoquinones and quinoline-5,8-diones as antimalarial and schistosomicidal agents. Org Biomol Chem 2012; 10:6375-87. [PMID: 22777178 PMCID: PMC3423093 DOI: 10.1039/c2ob25812a] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Improving the solubility of polysubstituted 1,4-naphthoquinone derivatives was achieved by introducing nitrogen in two different positions of the naphthoquinone core, at C-5 and at C-8 of menadione through a two-step, straightforward synthesis based on the regioselective hetero-Diels-Alder reaction. The antimalarial and the antischistosomal activities of these polysubstituted aza-1,4-naphthoquinone derivatives were evaluated and led to the selection of distinct compounds for antimalarial versus antischistosomal action. The Ag(II)-assisted oxidative radical decarboxylation of the phenyl acetic acids using AgNO(3) and ammonium peroxodisulfate was modified to generate the 3-picolinyl-menadione with improved pharmacokinetic parameters, high antimalarial effects and capacity to inhibit the formation of β-hematin.
Collapse
Affiliation(s)
- Don Antoine Lanfranchi
- European School of Chemistry, Polymers and Materials (ECPM) University of Strasbourg, UMR CNRS 7509, 25 Rue Becquerel, F-67087 Strasbourg, France. Fax: +33 (0)3 68 85 27 42; Tel: +33 3 68 85 26 20
| | - Elena Cesar-Rodo
- European School of Chemistry, Polymers and Materials (ECPM) University of Strasbourg, UMR CNRS 7509, 25 Rue Becquerel, F-67087 Strasbourg, France. Fax: +33 (0)3 68 85 27 42; Tel: +33 3 68 85 26 20
| | - Benoît Bertrand
- European School of Chemistry, Polymers and Materials (ECPM) University of Strasbourg, UMR CNRS 7509, 25 Rue Becquerel, F-67087 Strasbourg, France. Fax: +33 (0)3 68 85 27 42; Tel: +33 3 68 85 26 20
| | - Hsin-Hung Huang
- Department of Immunology/Microbiology, Rush University Medical Center, 1735 West Harrison Street, Chicago, IL 60612, USA
| | - Latasha Day
- Department of Immunology/Microbiology, Rush University Medical Center, 1735 West Harrison Street, Chicago, IL 60612, USA
| | - Laure Johann
- European School of Chemistry, Polymers and Materials (ECPM) University of Strasbourg, UMR CNRS 7509, 25 Rue Becquerel, F-67087 Strasbourg, France. Fax: +33 (0)3 68 85 27 42; Tel: +33 3 68 85 26 20
| | - Mourad Elhabiri
- European School of Chemistry, Polymers and Materials (ECPM) University of Strasbourg, UMR CNRS 7509, 25 Rue Becquerel, F-67087 Strasbourg, France. Fax: +33 (0)3 68 85 27 42; Tel: +33 3 68 85 26 20
| | - Katja Becker
- Interdisciplinary Research Center, Nutritional Biochemistry, Justus Liebig University Giessen, Heinrich-Buff-Ring 26–32, D-35392 Giessen, Germany
| | - David L. Williams
- Department of Immunology/Microbiology, Rush University Medical Center, 1735 West Harrison Street, Chicago, IL 60612, USA
| | - Elisabeth Davioud-Charvet
- European School of Chemistry, Polymers and Materials (ECPM) University of Strasbourg, UMR CNRS 7509, 25 Rue Becquerel, F-67087 Strasbourg, France. Fax: +33 (0)3 68 85 27 42; Tel: +33 3 68 85 26 20
| |
Collapse
|
12
|
Sarkar R, Hazra B, Mandal N. Hepatoprotective Potential of Caesalpinia crista against Iron-Overload-Induced Liver Toxicity in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2012; 2012:896341. [PMID: 22919421 PMCID: PMC3418686 DOI: 10.1155/2012/896341] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 05/26/2012] [Accepted: 05/28/2012] [Indexed: 11/17/2022]
Abstract
The present study was carried out to evaluate the ameliorating effect of Caesalpinia crista Linn. (CCME) extract on iron-overload-induced liver injury. Iron overload was induced by intraperitoneal administration of iron dextran into mice. CCME attenuated the percentage increase in liver iron and serum ferritin levels when compared to control group. CCME also showed a dose-dependent inhibition of lipid peroxidation, protein oxidation, and liver fibrosis. The serum enzyme markers were found to be less, whereas enhanced levels of liver antioxidant enzymes were detected in CCME-treated group. In presence of CCME, the reductive release of ferritin iron was increased significantly. Furthermore, CCME exhibited DPPH radical scavenging and protection against Fe(2+)-mediated oxidative DNA damage. The current study confirmed the hepatoprotective effect of CCME against the model hepatotoxicant iron overload and the activity is likely related to its potent antioxidant and iron-chelating property.
Collapse
Affiliation(s)
- Rhitajit Sarkar
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Bibhabasu Hazra
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Nripendranath Mandal
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata 700054, India
| |
Collapse
|
13
|
Synthesis, structure and conformational analysis of 2,4-diaryl-3-azabicyclo[3.3.1]nonan-9-one thiosemicarbazones and semicarbazones. J Mol Struct 2010. [DOI: 10.1016/j.molstruc.2010.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
14
|
Abstract
Current therapies for Alzheimer disease (AD) such as the acetylcholinesterase inhibitors and the latest NMDA receptor inhibitor, Namenda, provide moderate symptomatic delay at various stages of the disease, but do not arrest the disease progression or bring in meaningful remission. New approaches to the disease management are urgently needed. Although the etiology of AD is largely unknown, oxidative damage mediated by metals is likely a significant contributor since metals such as iron, aluminum, zinc, and copper are dysregulated and/or increased in AD brain tissue and create a pro-oxidative environment. This role of metal ion-induced free radical formation in AD makes chelation therapy an attractive means of dampening the oxidative stress burden in neurons. The chelator desferrioxamine, FDA approved for iron overload, has shown some benefit in AD, but like many chelators, it has a host of adverse effects and substantial obstacles for tissue-specific targeting. Other chelators are under development and have shown various strengths and weaknesses. Here, we propose a novel system of chelation therapy through the use of nanoparticles. Nanoparticles conjugated to chelators show unique ability to cross the blood-brain barrier (BBB), chelate metals, and exit through the BBB with their corresponding complexed metal ions. This method may provide a safer and more effective means of reducing the metal load in neural tissue, thus attenuating the harmful effects of oxidative damage and its sequelae. Experimental procedures are presented in this chapter.
Collapse
Affiliation(s)
- Gang Liu
- Department of Radiology, University of Utah, Salt Lake City, Utah, USA
| | | | | | | |
Collapse
|
15
|
Grazul M, Budzisz E. Biological activity of metal ions complexes of chromones, coumarins and flavones. Coord Chem Rev 2009. [DOI: 10.1016/j.ccr.2009.06.015] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
16
|
Liu G, Men P, Perry G, Smith MA. Metal chelators coupled with nanoparticles as potential therapeutic agents for Alzheimer's disease. ACTA ACUST UNITED AC 2009; 1:42-55. [PMID: 19936278 DOI: 10.1166/jns.2009.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Alzheimer's disease (AD) is a devastating neuro-degenerative disorder characterized by the progressive and irreversible loss of memory followed by complete dementia. Despite the disease's high prevalence and great economic and social burden, an explicative etiology or viable cure is not available. Great effort has been made to better understand the disease's pathogenesis, and to develop more effective therapeutic agents. However, success is greatly hampered by the presence of the blood-brain barrier that limits a large number of potential therapeutics from entering the brain. Nanoparticle-mediated drug delivery is one of the few valuable tools for overcoming this impediment and its application as a potential AD treatment shows promise. In this review, the current studies on nanoparticle delivery of chelation agents as possible therapeutics for AD are discussed because several metals are found excessive in the AD brain and may play a role in the disease development. Specifically, a novel approach involving transport of iron chelation agents into and out of the brain by nanoparticles is highlighted. This approach may provide a safer and more effective means of simultaneously reducing several toxic metals in the AD brain. It may also provide insights into the mechanisms of AD pathophysiology, and prove useful in treating other iron-associated neurodegenerative diseases such as Friedreich's ataxia, Parkinson's disease, Huntington's disease and Hallervorden-Spatz Syndrome. It is important to note that the use of nanoparticle-mediated transport to facilitate toxicant excretion from diseased sites in the body may advance nanoparticle technology, which is currently focused on targeted drug delivery for disease prevention and treatment. The application of nanoparticle-mediated drug transport in the treatment of AD is at its very early stages of development and, therefore, more studies are warranted.
Collapse
Affiliation(s)
- Gang Liu
- Department of Radiology, University of Utah, Salt Lake City, UT 84108, USA
| | | | | | | |
Collapse
|
17
|
Pardo-Andreu GL, Barrios MF, Curti C, Hernández I, Merino N, Lemus Y, Martínez I, Riaño A, Delgado R. Protective effects of Mangifera indica L extract (Vimang), and its major component mangiferin, on iron-induced oxidative damage to rat serum and liver. Pharmacol Res 2008; 57:79-86. [DOI: 10.1016/j.phrs.2007.12.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 12/19/2007] [Accepted: 12/20/2007] [Indexed: 01/06/2023]
|
18
|
Matoga D, Szklarzewicz J, Stadnicka K, Shongwe MS. Iron(III) Complexes with a Biologically Relevant Aroylhydrazone: Crystallographic Evidence for Coordination Versatility. Inorg Chem 2007; 46:9042-4. [PMID: 17892285 DOI: 10.1021/ic701435x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Complexation of iron(III) with the heterodonor chelating agent 3,5-di-tert-butylsalicylidene benzoylhydrazine, H2(3,5-tBu2)salbh, in the absence or presence of a base affords the complex cation [Fe{H(3,5-tBu2)salbh}2]+ or the neutral compound [Fe{H(3,5-tBu2)salbh}{(3,5-tBu2)salbh}], respectively, as revealed by single-crystal X-ray analyses. Such a synthetic and crystallographic demonstration of the coordination versatility of an aroylhydrazone toward iron is uncommon. The oxidation and spin states of the iron have been verified with magnetic and spectroscopic measurements.
Collapse
Affiliation(s)
- Dariusz Matoga
- Faculty of Chemistry, Jagiellonian University, Ingardena 3, 30-060 Kraków, Poland.
| | | | | | | |
Collapse
|
19
|
Richardson DR, Sharpe PC, Lovejoy DB, Senaratne D, Kalinowski DS, Islam M, Bernhardt PV. Dipyridyl thiosemicarbazone chelators with potent and selective antitumor activity form iron complexes with redox activity. J Med Chem 2006; 49:6510-21. [PMID: 17064069 DOI: 10.1021/jm0606342] [Citation(s) in RCA: 317] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
There has been much interest in the development of iron (Fe) chelators for the treatment of cancer. We developed a series of di-2-pyridyl ketone thiosemicarbazone (HDpT) ligands which show marked and selective antitumor activity in vitro and in vivo. In this study, we assessed chemical and biological properties of these ligands and their Fe complexes in order to understand their marked activity. This included examination of their solution chemistry, electrochemistry, ability to mediate redox reactions, and antiproliferative activity against tumor cells. The higher antiproliferative efficacy of the HDpT series of chelators relative to the related di-2-pyridyl ketone isonicotinoyl hydrazone (HPKIH) analogues can be ascribed, in part, to the redox potentials of their Fe complexes which lead to the generation of reactive oxygen species. The most effective HDpT ligands as antiproliferative agents possess considerable lipophilicity and were shown to be charge neutral at physiological pH, allowing access to intracellular Fe pools.
Collapse
Affiliation(s)
- Des R Richardson
- Iron Metabolism and Chelation Program, Department of Pathology, University of Sydney, Sydney, New South Wales 2006, Australia.
| | | | | | | | | | | | | |
Collapse
|
20
|
Liu G, Men P, Harris PLR, Rolston RK, Perry G, Smith MA. Nanoparticle iron chelators: a new therapeutic approach in Alzheimer disease and other neurologic disorders associated with trace metal imbalance. Neurosci Lett 2006; 406:189-93. [PMID: 16919875 DOI: 10.1016/j.neulet.2006.07.020] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 06/27/2006] [Accepted: 07/08/2006] [Indexed: 01/19/2023]
Abstract
Accumulating evidence suggests that oxidative stress may be a major etiologic factor in initiating and promoting neurodegeneration in Alzheimer disease. Contributing to this, there is a dyshomeostasis of metal ions in Alzheimer disease with abnormally high levels of redox-active metals, particularly iron, in affected areas of the brain. Although it is unclear whether metal excesses are the sole cause of oxidative stress and neurodegeneration or a by-product of neuronal loss, the finding that metal chelators can partially solubilize amyloid-beta deposits in Alzheimer disease suggests a promising therapeutic role for chelating agents. However, the blood-brain barrier and toxicity of known chelators limit their utility. In this study, we suggest that covalent conjugation of iron chelators with nanoparticles may help overcome the limitations in blood-brain barrier permeability of existing chelation therapy. Using in vitro studies, we have shown that a chelator-nanoparticle system and the chelator-nanoparticle system complexed with iron, when incubated with human plasma, preferentially adsorb apolipoprotein E and apolipoprotein A-I, that would facilitate transport into and out of the brain via mechanisms used for transporting low-density lipoprotein. Our studies suggest a unique approach, utilizing nanoparticles, to transport chelators and chelator-metal complexes in both directions across the blood-brain barrier, thus providing safer and more effective chelation treatment in Alzheimer disease and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Gang Liu
- Department of Radiology, University of Utah, Salt Lake City, UT 84102, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Zhang Y, Li H, Zhao Y, Gao Z. Dietary supplementation of baicalin and quercetin attenuates iron overload induced mouse liver injury. Eur J Pharmacol 2006; 535:263-9. [PMID: 16527270 DOI: 10.1016/j.ejphar.2006.01.067] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Revised: 01/22/2006] [Accepted: 01/25/2006] [Indexed: 10/24/2022]
Abstract
The introduction of new iron chelating drugs may ultimately improve iron-chelation therapy for patients with iron overload diseases such as thalassaemia and other disorders. In this paper, the in vivo effects of baicalin and quercetin on iron overload induced liver injury were studied on mice. It was found that when iron-dextran induced iron overload mice were fed baicalin or quercetin containing diet (1% w/w) for 45 days, both flavonoids significantly inhibited iron overload induced lipid peroxidation and protein oxidation of liver, decreased hepatic iron and hepatic collagen content, increased the serum non-heme iron level but not serum ferritin level. Flavonoids supplementation also increased the excretion of iron through feces. In vitro study demonstrated that both flavonoids could release iron from ferritin. These results indicate that besides acting as antioxidants, both flavonoids can also release iron from liver and finally excrete it through feces. The present study provides further support for flavonoids to be medicines for iron overload diseases.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Chemistry, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | | | | | | |
Collapse
|
22
|
Melnyk P, Leroux V, Sergheraert C, Grellier P. Design, synthesis and in vitro antimalarial activity of an acylhydrazone library. Bioorg Med Chem Lett 2006; 16:31-5. [PMID: 16263280 DOI: 10.1016/j.bmcl.2005.09.058] [Citation(s) in RCA: 174] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2005] [Revised: 09/21/2005] [Accepted: 09/22/2005] [Indexed: 10/25/2022]
Abstract
A library of acylhydrazone iron chelators was synthesized and tested for its ability to inhibit the growth of a chloroquine-resistant strain of Plasmodium falciparum. Some of these new compounds are significantly more active than desferrioxamine DFO, the iron chelator in widespread clinical use and also than the most effective chelators.
Collapse
Affiliation(s)
- Patricia Melnyk
- Institut de Biologie et Institut Pasteur de Lille, UMR CNRS 8525, Université de Lille II, 1 rue du Professeur Calmette, BP 447, 59021 Lille, France.
| | | | | | | |
Collapse
|
23
|
Liu G, Garrett MR, Men P, Zhu X, Perry G, Smith MA. Nanoparticle and other metal chelation therapeutics in Alzheimer disease. Biochim Biophys Acta Mol Basis Dis 2005; 1741:246-52. [PMID: 16051470 DOI: 10.1016/j.bbadis.2005.06.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Revised: 06/21/2005] [Accepted: 06/29/2005] [Indexed: 11/16/2022]
Abstract
Current therapies for Alzheimer disease (AD) such as the anticholinesterase inhibitors and the latest NMDA receptor inhibitor, Namenda, provide moderate symptomatic delay at various stages of disease, but do not arrest disease progression or supply meaningful remission. As such, new approaches to disease management are urgently needed. Although the etiology of AD is largely unknown, oxidative damage mediated by metals is likely a significant contributor since metals such as iron, aluminum, zinc, and copper are dysregulated and/or increased in AD brain tissue and create a pro-oxidative environment. This role of metal ion-induced free radical formation in AD makes chelation therapy an attractive means of dampening the oxidative stress burden in neurons. The chelator desferioxamine, FDA approved for iron overload, has shown some benefit in AD, but like many chelators, it has a host of adverse effects and substantial obstacles for tissue-specific targeting. Other chelators are under development and have shown various strengths and weaknesses. In this review, we propose a novel system of chelation therapy through the use of nanoparticles. Nanoparticles conjugated to chelators show a unique ability to cross the blood-brain barrier (BBB), chelate metals, and exit through the BBB with their corresponding complexed metal ions. This method may prove to be a safe and effective means of reducing the metal load in neural tissue thus staving off the harmful effects of oxidative damage and its sequelae.
Collapse
Affiliation(s)
- Gang Liu
- Department of Radiology, University of Utah, Salt Lake City, UT 84102, USA
| | | | | | | | | | | |
Collapse
|
24
|
Zhao Y, Li H, Gao Z, Xu H. Effects of dietary baicalin supplementation on iron overload-induced mouse liver oxidative injury. Eur J Pharmacol 2005; 509:195-200. [PMID: 15733556 DOI: 10.1016/j.ejphar.2004.11.060] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2004] [Accepted: 11/26/2004] [Indexed: 11/18/2022]
Abstract
Iron overload is one of the most common metal related toxicity. Under this circumstance, excessive iron deposition in liver will lead to further injury such as hepatocellular necrosis, inflammation, fibrosis, and in some cases even to carcinoma. In this paper, the effect of a nature flavonoid, baicalin, on iron overload-induced mouse liver oxidative injury has been studied. It was found that when iron-dextran-induced iron overload, mice were fed baicalin-containing diet (0.25% and 1%) for 50 days, hepatic iron, liver-to-body weight ratio, and hepatic lipid peroxidation were dose-dependently decreased; while catalase activity, total antioxidant status, and serum iron content were dose dependently increased. The protective effect of baicalin on liver of iron overload mouse may due to both the antioxidant and iron chelation activities of baicalin. These data provide preliminary experimental support for baicalin as medicine for iron overload diseases.
Collapse
Affiliation(s)
- Yuling Zhao
- Department of Chemistry, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | | | | | | |
Collapse
|
25
|
Chaston TB, Watts RN, Yuan J, Richardson DR. Potent antitumor activity of novel iron chelators derived from di-2-pyridylketone isonicotinoyl hydrazone involves fenton-derived free radical generation. Clin Cancer Res 2005; 10:7365-74. [PMID: 15534113 DOI: 10.1158/1078-0432.ccr-04-0865] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The development of novel and potent iron chelators as clinically useful antitumor agents is an area of active interest. Antiproliferative activity of chelators often relates to iron deprivation or stimulation of iron-dependent free radical damage. Recently, we showed that novel iron chelators of the di-2-pyridylketone isonicotinoyl hydrazone (PKIH) class have potent and selective antineoplastic activity (E. Becker, et al., Br. J. Pharmacol., 138: 819-30, 2003). In this study, we assessed the effects of the PKIH analogues on the redox activity of iron in terms of understanding their antitumor activity. EXPERIMENTAL DESIGN We tested the PKIH analogues for their ability to promote iron-mediated ascorbate oxidation, benzoate hydroxylation, and plasmid degradation. Subsequent experiments assessed their ability to bind DNA, inhibit topoisomerase I, and cause DNA damage. To measure intracellular reactive oxygen species, we used the redox-sensitive probe, 2',7'-dichloro-fluorescein-diacetate, to measure intracellular PKIH-dependent redox activity. RESULTS The PKIH analogues had relatively little effect on ascorbate oxidation in the presence of Fe(III) but stimulated benzoate hydroxylation and plasmid DNA degradation in the presence of Fe(II) and H2O2. These ligands could not inhibit DNA topoisomerase I or cause DNA damage in intact cells. PKIH markedly increased the intracellular generation of reactive oxygen species, and this was inhibited by catalase. This enzyme also decreased the antiproliferative effect of PKIH, indicating H2O2 played a role in its cytotoxic activity. CONCLUSIONS Our results suggest that the antiproliferative effects of these chelators relates to intracellular iron chelation, followed by the stimulation of iron-mediated free radical generation via the so-formed iron complex.
Collapse
Affiliation(s)
- Timothy B Chaston
- Children's Cancer Institute Australia for Medical Research, Iron Metabolism and Chelation Program, Sydney, New South Wales, Australia
| | | | | | | |
Collapse
|
26
|
Walcourt A, Loyevsky M, Lovejoy DB, Gordeuk VR, Richardson DR. Novel aroylhydrazone and thiosemicarbazone iron chelators with anti-malarial activity against chloroquine-resistant and -sensitive parasites. Int J Biochem Cell Biol 2004; 36:401-7. [PMID: 14687919 DOI: 10.1016/s1357-2725(03)00248-6] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Iron (Fe) is crucial for cellular proliferation, and Fe chelators have shown activity at preventing the growth of the malarial parasite in cell culture and in animal and human studies. We investigated the anti-malarial activity of novel aroylhydrazone and thiosemicarbazone Fe chelators that show high activity at inhibiting the growth of tumour cells in cell culture [Blood 100 (2002) 666]. Experiments with the chelators were performed using the chloroquine-sensitive, 3D7, and chloroquine-resistant, 7G8, strains of Plasmodium falciparum in vitro. The new ligands were significantly more active in both strains than the Fe chelator in widespread clinical use, desferrioxamine (DFO). The most effective chelators examined were 2-hydroxy-1-naphthylaldehyde isonicotinoyl hydrazone and 2-hydroxy-1-naphthylaldehyde-4-phenyl-3-thiosemicarbazone. The anti-malarial activity correlates with anti-proliferative activity against neoplastic cells demonstrated in a previous study. Our studies suggest that this class of lipophilic chelators may have potential as useful agents for the treatment of malaria.
Collapse
Affiliation(s)
- Asikiya Walcourt
- Center for Sickle Cell Disease, Howard University, 2121 Georgia Avenue, Washington, DC 20060, USA.
| | | | | | | | | |
Collapse
|
27
|
Chaston TB, Richardson DR. Iron chelators for the treatment of iron overload disease: relationship between structure, redox activity, and toxicity. Am J Hematol 2003; 73:200-10. [PMID: 12827659 DOI: 10.1002/ajh.10348] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The success of the iron (Fe) chelator desferrioxamine (DFO) in the treatment of beta-thalassemia is limited by its lack of bioavailability. The design and characterization of synthetic alternatives to DFO has attracted much scientific interest and has led to the discovery of orally active chelators that can remove pathological Fe deposits. However, chelators that access intracellular Fe pools can be toxic by either inhibiting Fe-containing enzymes or promoting Fe-mediated free radical damage. Interestingly, toxicity does not necessarily correlate with Fe-binding affinity or with chelation efficacy, suggesting that other factors may promote the cytopathic effects of chelators. In this review, we discuss the interactions of chelators and their Fe complexes with biomolecules that can lead to toxicity and tissue damage.
Collapse
Affiliation(s)
- Timothy B Chaston
- Children's Cancer Institute Australia for Medical Research, The Iron Metabolism and Chelation Program, Randwick, Sydney, New South Wales, Australia
| | | |
Collapse
|
28
|
Wong C, Richardson DR. Beta-thalassaemia: emergence of new and improved iron chelators for treatment. Int J Biochem Cell Biol 2003; 35:1144-9. [PMID: 12672484 DOI: 10.1016/s1357-2725(03)00046-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Beta-thalassaemia is an inherited blood disorder which through repeated blood transfusions and enhanced iron uptake from the gastrointestinal tract, results in marked iron overload. Untreated, the iron accumulation results in the dysfunction of vital organs such as the heart and liver. At present, the most effective treatment for beta-thalassaemia is the use of the iron chelator, desferrioxamine, which is expensive, orally inactive and requires long subcutaneous infusions. In this concise review, we will focus on novel chelators which show therapeutic potential to replace desferrioxamine. Furthermore, we will discuss the potential of combined iron chelation therapy and the principle that, in the future, the use of more than just one chelator may be beneficial in tailoring individual iron chelation regimens.
Collapse
Affiliation(s)
- Cynthia Wong
- The Iron Metabolism and Chelation Program, Children's Cancer Institute Australia for Medical Research, PO Box 81, High Street, Randwick, New South Wales Sydney 2031, Australia
| | | |
Collapse
|
29
|
Chaston TB, Richardson DR. Interactions of the pyridine-2-carboxaldehyde isonicotinoyl hydrazone class of chelators with iron and DNA: implications for toxicity in the treatment of iron overload disease. J Biol Inorg Chem 2003; 8:427-38. [PMID: 12761664 DOI: 10.1007/s00775-002-0434-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2002] [Accepted: 11/15/2002] [Indexed: 10/18/2022]
Abstract
Iron chelation therapy for the management of iron-overload disease is dominated by desferrioxamine (DFO). However, treatment using DFO is very arduous. Recently, novel Fe chelators of the pyridine-2-carboxaldehyde isonicotinoyl hydrazone (PCIH) class have shown high chelation efficacy and the potential to replace DFO. A critical consideration in the design of alternatives to DFO is that the chelator forms a redox-inert Fe complex. In the present study, the participation of Fe complexes in redox reactions has been investigated. Ascorbate oxidation in the presence of Fe(III) or benzoate hydroxylation in the presence of Fe(II) was not enhanced by the PCIH analogues. However, redox-induced DNA strand breaks were observed with these ligands under highly oxidizing conditions in the presence of Fe(II) and hydrogen peroxide. Experiments then examined the interactions of the PCIH analogues with DNA, and this was found to be weak. Considering this, we suggest that under extreme conditions seen in the DNA-strand break assay, weak DNA-binding may potentiate the redox activity of the PCIH analogues. However, importantly, in contrast to naked plasmid DNA, DNA damage by these chelators using intact human cells was not significant. Collectively, our results support the potential of the PCIH analogues for the treatment of Fe overload.
Collapse
Affiliation(s)
- Timothy B Chaston
- Iron Metabolism and Chelation Program, Children's Cancer Institute Australia for Medical Research, High Street, Randwick, PO Box 81, 2031, Sydney, New South Wales, Australia
| | | |
Collapse
|
30
|
Affiliation(s)
- Des R Richardson
- Children's Cancer Research Institute Australia, Iron Metabolism and Chelation Program, Randwick, Sydney, NSW 2031, Australia.
| |
Collapse
|
31
|
Eybl V, Kotyzová D, Kolek M, Koutenský J, Nielsen P. The influence of deferiprone (L1) and deferoxamine on iron and essential element tissue level and parameters of oxidative status in dietary iron-loaded mice. Toxicol Lett 2002; 128:169-75. [PMID: 11869827 DOI: 10.1016/s0378-4274(01)00541-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The seven week feeding of a diet enriched with 0.5% TMH-ferrocene to male mice was used in this study to produce an iron-overload model in experimental animals for evaluating the effect of deferoxamine (DFO) and deferiprone (L1) on tissue-stored iron, induced lipid peroxidation (LP) and parameters of oxidative status. The iron concentration in the liver reached 600% of the level in control animals. The administration of seven doses of deferoxamine (DFO) i.p. and deferiprone (L1) p.o. (0.72 mmol/kg b.w., every 48 h) during 9th and 10th week significantly decreased the liver, kidneys and heart iron level in both iron-loaded and control mice. The DFO and L1 treatment also equally attenuated lipid peroxidation and increased the GSH level in the liver of iron loaded mice. The glutathione peroxidase (GSH-Px) activity and catalase activity were not affected by iron loading, however, both DFO and L1 caused a decrease of GSH-Px activity.
Collapse
Affiliation(s)
- Vladislav Eybl
- Department of Pharmacology and Toxicology, Charles University Faculty of Medicine in Pilsen, Karlovarská 48, 301 66 Pilsen, Czech Republic.
| | | | | | | | | |
Collapse
|
32
|
Rodgers GP, Saunthararajah Y. Advances in experimental treatment of beta-thalassaemia. Expert Opin Investig Drugs 2001; 10:925-34. [PMID: 11322866 DOI: 10.1517/13543784.10.5.925] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Beta-thalassaemia is highly prevalent and world wide in its distribution. The gene to modify the clinical course of patients with transfusion-dependent thalassaemia (thalassaemia major), the gamma-globin gene, is already present in these patients but silenced in the course of development. During erythropoiesis, progenitors are believed to go through a phase where the milieu favours gamma-globin production. One pharmacological strategy to increase gamma-globin production is directed at recruiting such early progenitors through the use of cytotoxic agents (+/- erythropoietin) that presumably deplete more mature progenitors. Another promising strategy is to use chromatin-modifying agents that prevent the silencing of the gamma-globin gene that occurs during development. These agents, the methyl-transferase inhibitors and histone deacetylase inhibitors, either alone or in combination, may be able to produce the robust increase in gamma-globin and hence fetal haemoglobin and total haemoglobin, needed to successfully treat thalassaemia major. Studies of these agents, which are already available for clinical trials, should be encouraged.
Collapse
Affiliation(s)
- G P Rodgers
- Molecular and Clinical Hematology Branch, National Institutes of Diabetes, Digestive and Kidney Diseases, Bethesda, MD 20892, USA.
| | | |
Collapse
|
33
|
Richardson DR. The controversial role of deferiprone in the treatment of thalassemia. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2001; 137:324-9. [PMID: 11329529 DOI: 10.1067/mlc.2001.114105] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of the orally active iron (Fe) chelator deferiprone in the treatment of beta-thalassemia remains a controversial subject. Despite initial studies showing high Fe chelation efficacy in vitro and also in animals and human subjects, several latter studies have not been so successful. In fact, it has been reported in several clinical trials that deferiprone after long-term treatment had either little effect or actually increased hepatic Fe loading. In addition, an increase in liver fibrosis was noted in one study. However, more recently, results by other investigators have suggested that the drug may be used under some circumstances without marked toxicity. In particular, it has been demonstrated that the combination of deferoxamine (DFO) and deferiprone results in more Fe excretion than when either chelator is used alone. Moreover, a combination of both drugs led to a decrease in deferiprone-mediated toxicity. Other studies performed in patients for up to 10 years showed no progressive fibrosis after deferiprone therapy, while a possible trend toward increasing fibrosis was noted in another investigation. Additional studies using larger numbers of deferiprone-treated patients are essential to determine the efficacy and safety of this drug, particularly in relation to the development of fibrosis. The present review discusses the possible role of deferiprone in the treatment of Fe overload.
Collapse
Affiliation(s)
- D R Richardson
- Heart Research Institute, The Iron Metabolism and Chelation Group, Camperdown, NSW 2050, Australia
| |
Collapse
|
34
|
Whittaker P, Seifried HE, San RH, Clarke JJ, Dunkel VC. Genotoxicity of iron chelators in L5178Y mouse lymphoma cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2001; 38:347-356. [PMID: 11774367 DOI: 10.1002/em.10033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
To further study the mechanism of observed iron mutagenicity and cellular toxicity, a number of different iron chelators were evaluated to select a compound that was not mutagenic and had limited toxicity to mouse lymphoma cells. A series of iron chelators including those used clinically, those under development for clinical applications, and those used in nonclinical applications were evaluated. The mutagenic activity of the iron chelators was assessed in L5178Y mouse lymphoma cells. Eight of the 12 iron chelators that were tested induced mutagenic responses both with and without the addition of S9. Among those chelators used clinically or developed for clinical use, the only compound that did not induce a mutagenic response was the starch deferoxamine conjugate. In contrast, deferoxamine mesylate showed the highest toxicity in this group of chemicals and the concentrations leading to toxicity and mutagenicity between the activated and nonactivated assays were not significantly different. The other three chelators that were not mutagenic were Na2EDTA, phytic acid, and ferrozine.
Collapse
Affiliation(s)
- P Whittaker
- Center for Food Safety and Applied Nutrition, Food and Drug Administration, Washington, DC 20204, USA.
| | | | | | | | | |
Collapse
|
35
|
Becker E, Richardson DR. Frataxin: its role in iron metabolism and the pathogenesis of Friedreich's ataxia. Int J Biochem Cell Biol 2001; 33:1-10. [PMID: 11167127 DOI: 10.1016/s1357-2725(00)00067-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Friedreich's ataxia (FA) is a severe neurodegenerative condition with an incidence of 1:50000 in the European population. In 97% of patients this disease is due to an intronic GAA triplet repeat expansion in the FRDA gene resulting in a marked decrease in its expression. The protein encoded by this gene is known as frataxin which is found within the mitochondrion. Upon deletion of the homologous gene (YFH1) in the yeast, there was an accumulation of iron (Fe) within the mitochondrion. When the YFH1 gene was reintroduced back into the yeast cell Fe was exported out of the mitochondrion and into the cytosol. Evidence that human frataxin is also involved in mitochondrial Fe-overload comes from studies in FA patients that have shown an accumulation of Fe within the heart. While the precise role of human frataxin remains to be determined, the molecule appears to be involved indirectly in regulating the export and/or import of mitochondrial Fe. The finding of mitochondrial Fe-overload suggests that the use of specific Fe chelators which can permeate the mitochondrion may have potential in the treatment of this disease.
Collapse
Affiliation(s)
- E Becker
- The Heart Research Institute, 145 Missenden Road, Camperdown, Sydney, NSW, 2050, Australia
| | | |
Collapse
|