1
|
Rieder GS, Braga MM, Mussulini BHM, Silva ES, Lazzarotto G, Casali EA, Oliveira DL, Franco JL, Souza DOG, Rocha JBT. Diphenyl Diselenide Attenuates Mitochondrial Damage During Initial Hypoxia and Enhances Resistance to Recurrent Hypoxia. Neurotox Res 2024; 42:13. [PMID: 38332435 DOI: 10.1007/s12640-024-00691-6] [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: 08/09/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 02/10/2024]
Abstract
Hypoxia plays a significant role in the development of various cerebral diseases, many of which are associated with the potential risk of recurrence due to mitochondrial damage. Conventional drug treatments are not always effective for hypoxia-related brain diseases, necessitating the exploration of alternative compounds. In this study, we investigated the potential of diphenyl diselenide [(PhSe)2] to ameliorate locomotor impairments and mitigate brain mitochondrial dysfunction in zebrafish subjected to hypoxia. Additionally, we explored whether these improvements could confer resistance to recurrent hypoxia. Through a screening process, an appropriate dose of (PhSe)2 was determined, and animals exposed to hypoxia received a single intraperitoneal injection of 100 mg/kg of the compound or vehicle. After 1 h from the injection, evaluations were conducted on locomotor deficits, (PhSe)2 content, mitochondrial electron transport system, and mitochondrial viability in the brain. The animals were subsequently exposed to recurrent hypoxia to assess the latency time to hypoxia symptoms. The findings revealed that (PhSe)2 effectively crossed the blood-brain barrier, attenuated locomotor deficits induced by hypoxia, and improved brain mitochondrial respiration by modulating complex III. Furthermore, it enhanced mitochondrial viability in the telencephalon, contributing to greater resistance to recurrent hypoxia. These results demonstrate the beneficial effects of (PhSe)2 on both hypoxia and recurrent hypoxia, with cerebral mitochondria being a critical target of its action. Considering the involvement of brain hypoxia in numerous pathologies, (PhSe)2 should be further tested to determine its effectiveness as a potential treatment for hypoxia-related brain diseases.
Collapse
Affiliation(s)
- Guilherme S Rieder
- Programa de Pós Graduação Em Bioquímica Toxicológica, Departamento de Bioquímica E Biologia Molecular, Centro de Ciências Naturais E Exatas, Universidade Federal de Santa Maria, Avenida Roraima 1000, Santa Maria, RS, 97105-900, Brazil
| | - Marcos M Braga
- Programa de Pós Graduação Em Bioquímica Toxicológica, Departamento de Bioquímica E Biologia Molecular, Centro de Ciências Naturais E Exatas, Universidade Federal de Santa Maria, Avenida Roraima 1000, Santa Maria, RS, 97105-900, Brazil
| | - Ben Hur M Mussulini
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Emerson S Silva
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Gabriela Lazzarotto
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Emerson André Casali
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Diogo L Oliveira
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Jeferson L Franco
- Universidade Federal Do Pampa, Campus São Gabriel, São Gabriel, RS, Brazil
| | - Diogo O G Souza
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - João Batista T Rocha
- Programa de Pós Graduação Em Bioquímica Toxicológica, Departamento de Bioquímica E Biologia Molecular, Centro de Ciências Naturais E Exatas, Universidade Federal de Santa Maria, Avenida Roraima 1000, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
2
|
Ji X, Zhu R, Gao C, Xie H, Gong X, Luo J. Hypoxia-Derived Exosomes Promote Lung Adenocarcinoma by Regulating HS3ST1-GPC4-Mediated Glycolysis. Cancers (Basel) 2024; 16:695. [PMID: 38398086 PMCID: PMC10886556 DOI: 10.3390/cancers16040695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE The diagnosis of lung adenocarcinoma (LUAD) is often delayed due to the typically asymptomatic nature of the early-stage disease, causing advanced-stage LUAD diagnosis in most patients. Hypoxia is widely recognized as a driving force in cancer progression. Exosomes originating from hypoxic tumor cells promote tumorigenesis by influencing glycolysis, migration, invasion, and immune infiltration. Given these insights, our study aimed to explore the role of hypoxia-derived exosomal long non-coding RNA (lncRNA) OIP5-AS1 in LUAD cell lines and mouse models. MATERIALS AND METHODS Exosomes were meticulously isolated and authenticated based on their morphology and biomarkers. The interaction between heparan sulfate (glucosamine) 3-O-sulfotransferase 1 (HS3ST1) and Glypican 4 (GPC4) was examined using immunoprecipitation. The influence of the hypoxia-derived exosomal lncRNA OIP5-AS1 on glycolysis was assessed in LUAD cell lines. The effect of the hypoxia-derived exosomal lncRNA OIP5-AS1 on cell proliferation and metastasis was evaluated using colony formation, cell viability, cell cycle, and apoptosis analyses. Its effects on tumor size were confirmed in xenograft animal models. RESULTS Our study revealed the mechanism of the hypoxia-derived exosomal lncRNA OIP5-AS1 in LUAD progression. We discovered that GPC4 promotes HS3ST1-mediated glycolysis and that the hypoxia-derived exosomal lncRNA OIP5-AS1 enhances glycolysis by regulating miR-200c-3p in LUAD cells. Notably, this lncRNA stimulates LUAD cell proliferation and metastasis and fosters LUAD tumor size via miR-200c-3p. Our findings underscore the potential role of the hypoxia-derived exosomal lncRNA OIP5-AS1 in LUAD progression. CONCLUSIONS The hypoxia-derived exosomal lncRNA OIP5-AS1 promotes LUAD by regulating HS3ST1-GPC4-mediated glycolysis via miR-200c-3p.
Collapse
Affiliation(s)
- Xianxiu Ji
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Ren Zhu
- Department of Medical Administration, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Caixia Gao
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Xiaomei Gong
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Jie Luo
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| |
Collapse
|
3
|
Mertens RT, Kim JH, Ofori S, Olelewe C, Kamitsuka PJ, Kwakye GF, Awuah SG. A gold-based inhibitor of oxidative phosphorylation is effective against triple negative breast cancer. Biomed Pharmacother 2024; 170:116010. [PMID: 38128183 PMCID: PMC11254167 DOI: 10.1016/j.biopha.2023.116010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/28/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is associated with metabolic heterogeneity and poor prognosis with limited treatment options. New treatment paradigms for TNBC remains an unmet need. Thus, therapeutics that target metabolism are particularly attractive approaches. We previously designed organometallic Au(III) compounds capable of modulating mitochondrial respiration by ligand tuning with high anticancer potency in vitro and in vivo. Here, we show that an efficacious Au(III) dithiocarbamate (AuDTC) compound induce mitochondrial dysfunction and oxidative damage in cancer cells. Efficacy of AuDTC in TNBC mouse models harboring mitochondrial oxidative phosphorylation (OXPHOS) dependence and metabolic heterogeneity establishes its therapeutic potential following systemic delivery. This provides evidence that AuDTC is an effective modulator of mitochondrial respiration worthy of clinical development in the context of TNBC. ONE SENTENCE SUMMARY: Metabolic-targeting of triple-negative breast cancer by gold anticancer agent may provide efficacious therapy.
Collapse
Affiliation(s)
- R Tyler Mertens
- Department of Chemistry, University of Kentucky; Lexington, KY 40506, United States
| | - Jong Hyun Kim
- Department of Chemistry, University of Kentucky; Lexington, KY 40506, United States
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky; Lexington, KY 40506, United States; Department of Neuroscience, Oberlin College, Oberlin, OH 44074, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, United States; University of Kentucky Markey Cancer Center, USA
| | - Chibuzor Olelewe
- Department of Chemistry, University of Kentucky; Lexington, KY 40506, United States; Department of Neuroscience, Oberlin College, Oberlin, OH 44074, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, United States; University of Kentucky Markey Cancer Center, USA
| | - Paul J Kamitsuka
- Department of Neuroscience, Oberlin College, Oberlin, OH 44074, United States
| | - Gunnar F Kwakye
- Department of Neuroscience, Oberlin College, Oberlin, OH 44074, United States
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky; Lexington, KY 40506, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, United States; University of Kentucky Markey Cancer Center, USA.
| |
Collapse
|
4
|
de Andrade Querino AL, de Sousa AM, Thomas SR, de Lima GM, Dittz D, Casini A, do Monte-Neto RL, Silva H. Organogold(III)-dithiocarbamate compounds and their coordination analogues as anti-tumor and anti-leishmanial metallodrugs. J Inorg Biochem 2023; 247:112346. [PMID: 37536162 DOI: 10.1016/j.jinorgbio.2023.112346] [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: 05/31/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023]
Abstract
The limited chemical stability of gold(III)-based compounds in physiological environment has been a challenge in drug discovery, and organometallic chemistry might provide the solution to overcome this issue. In this work, four novel cationic organogold(III)-dithiocarbamate complexes of general structure [(C^N)AuIIIDTC]PF6 (C1a - C4a, DTC = dithiocarbamate, L1 - L4, C^N = 2-anilinopyridine) are presented, and compared to their coordination gold(III)-dithiocarbamate analogues [AuIIIDTCCl2] (C1b - C4b), as potential anti-cancer and anti-leishmanial drugs. Most of the complexes effectively inhibited cancer cell growth, notably C3a presented anti-proliferative effect in the nanomolar range against breast cancer (MCF-7 and MDA-MB-231 cells with moderate selectivity. Pro-apoptotic studies on treated MCF-7 cells showed a high population of cells in early apoptosis. Reactivity studies of C3a towards model thiols (N-acetyl-L-cysteine) refer to a possible mode of action involving bonding between the organogold(III)-core and the thiolate. In the scope of neglected diseases, gold complexes are emerging as promising therapeutic alternatives against leishmaniasis. In this regard, all gold(III)-dithiocarbamate complexes presented anti-leishmanial activity against at least one Leishmania species. Complexes C1a, C4a, C1b, C4b were active against all tested parasites with IC50 values varying between 0.12 and 42 μM, and, overall, organometallic compounds presented more intriguing inhibition profiles. For C4a selectivity over 500-fold for L. braziliensis; even higher than the reference anti-leishmanial drug amphotericin B. Overall, our findings revealed that the organogold(III) moiety significantly amplified the anti-cancer and anti-leishmanial effects with respect to the coordination analogues; thus, showing the great potential of organometallic chemistry in metallodrug-based chemotherapy for cancer and leishmaniasis.
Collapse
Affiliation(s)
- Ana Luiza de Andrade Querino
- Laboratório de Síntese e Interações Bioinorgânicas (SibLab), Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, Technical University of Munich, Garching b. Munich, Germany.
| | - Alessandra Mara de Sousa
- RdM Lab - Biotechnology Applied to Pathogens Research Group, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Sophie R Thomas
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, Technical University of Munich, Garching b. Munich, Germany
| | - Geraldo Magela de Lima
- Laboratório de Síntese e Interações Bioinorgânicas (SibLab), Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Dalton Dittz
- Department of Biochemistry and Pharmacology, Universidade Federal do Piaui, Teresina, Brazil
| | - Angela Casini
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, Technical University of Munich, Garching b. Munich, Germany
| | - Rubens Lima do Monte-Neto
- RdM Lab - Biotechnology Applied to Pathogens Research Group, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Heveline Silva
- Laboratório de Síntese e Interações Bioinorgânicas (SibLab), Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
5
|
Arojojoye AS, Walker B, Dewahare JC, Afrifa MAO, Parkin S, Awuah SG. Circumventing Physicochemical Barriers of Cyclometalated Gold(III) Dithiocarbamate Complexes with Protein-Based Nanoparticle Delivery to Enhance Anticancer Activity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43607-43620. [PMID: 37698293 PMCID: PMC11264193 DOI: 10.1021/acsami.3c10025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Optimizing the bioavailability of drug candidates is crucial to successful drug development campaigns, especially for metal-derived chemotherapeutic agents. Nanoparticle delivery strategies can be deployed to overcome physicochemical limitations associated with drugs to improve bioavailability, pharmacokinetics, efficacy, and minimize toxicity. Biodegradable albumin nanoconstructs offer pragmatic solutions for drug delivery of metallodrugs with translational benefits in the clinic. In this work, we explored a logical approach to investigate and resolve the physicochemical drawbacks of gold(III) complexes with albumin nanoparticle delivery to improve solubility, enhance intracellular accumulation, circumvent premature deactivation, and enhance anticancer activity. We synthesized and characterized stable gold(III) dithiocarbamate complexes with a variable degree of cyclometalation such as phenylpyridine (C^N) or biphenyl (C^C) Au(III) framework and different alkyl chain lengths. We noted that extended alkyl chain lengths impaired the solubility of these complexes in biological media, thus adversely impacting potency. Encapsulation of these complexes in bovine serum albumin (BSA) reversed solubility limitations and improved cancer cytotoxicity by ∼25-fold. Further speciation and mechanism of action studies demonstrate the stability of the compounds and alteration of mitochondria bioenergetics, respectively. We postulate that this nanodelivery strategy is a relevant approach for translational small-molecule gold drug delivery.
Collapse
Affiliation(s)
| | - Breyanna Walker
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
| | - James C. Dewahare
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
| | | | - Sean Parkin
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
| | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
- Center for Pharmaceutical Research and Innovation and Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington KY 40536
| |
Collapse
|
6
|
Gukathasan S, Obisesan OA, Saryazdi S, Ratliff L, Parkin S, Grossman RB, Awuah SG. A Conformationally Restricted Gold(III) Complex Elicits Antiproliferative Activity in Cancer Cells. Inorg Chem 2023; 62:13118-13129. [PMID: 37530672 PMCID: PMC11268950 DOI: 10.1021/acs.inorgchem.3c02066] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Diamine ligands are effective structural scaffolds for tuning the reactivity of transition-metal complexes for catalytic, materials, and phosphorescent applications and have been leveraged for biological use. In this work, we report the synthesis and characterization of a novel class of cyclometalated [C^N] Au(III) complexes bearing secondary diamines including a norbornane backbone, (2R,3S)-N2,N3-dibenzylbicyclo[2.2.1]heptane-2,3-diamine, or a cyclohexane backbone, (1R,2R)-N1,N2-dibenzylcyclohexane-1,2-diamine. X-ray crystallography confirms the square-planar geometry and chirality at nitrogen. The electronic character of the conformationally restricted norbornane backbone influences the electrochemical behavior with redox potentials of -0.8 to -1.1 V, atypical for Au(III) complexes. These compounds demonstrate promising anticancer activity, particularly, complex 1, which bears a benzylpyridine organogold framework, and supported by the bicyclic conformationally restricted diaminonorbornane, shows good potency in A2780 cells. We further show that a cellular response to 1 evokes reactive oxygen species (ROS) production and does not induce mitochondrial dysfunction. This class of complexes provides significant stability and reactivity for different applications in protein modification, catalysis, and therapeutics.
Collapse
Affiliation(s)
| | | | - Setareh Saryazdi
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA
| | - Libby Ratliff
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA
| | - Sean Parkin
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA
| | - Robert B. Grossman
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA
| | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA
- Center for Pharmaceutical Research and Innovation and Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington KY 40536
| |
Collapse
|
7
|
Greif CE, Mertens RT, Berger G, Parkin S, Awuah SG. An anti-glioblastoma gold(i)-NHC complex distorts mitochondrial morphology and bioenergetics to induce tumor growth inhibition. RSC Chem Biol 2023; 4:592-599. [PMID: 37547458 PMCID: PMC10398352 DOI: 10.1039/d3cb00051f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/19/2023] [Indexed: 08/08/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most lethal brain cancer subtype, often advanced by the time of initial diagnosis. Existing treatment modalities including surgery, chemotherapy and radiation have been stymied by recurrence, metastasis, drug resistance and brain targetability. Here, we report a geometrically distinct Au(i) complex ligated by N^N-bidentate ligands and supported by a N-heterocyclic ligand that modulates mitochondrial morphology to inhibit GBM in vitro and in vivo. This work benefits from the facile preparation of anti-GBM Au(i)-NHC complexes.
Collapse
Affiliation(s)
- Charles E Greif
- Department of Chemistry, University of Kentucky Lexington Kentucky 40506 USA
| | - R Tyler Mertens
- Department of Chemistry, University of Kentucky Lexington Kentucky 40506 USA
| | - Gilles Berger
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School Boston MA 02115 USA
- Microbiology, Bioorganic & Macromolecular Chemistry, Faculté de Pharmacie, Université libre de Bruxelles (ULB), Boulevard du Triomphe 1050 Brussels Belgium
| | - Sean Parkin
- Department of Chemistry, University of Kentucky Lexington Kentucky 40506 USA
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky Lexington Kentucky 40506 USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky Lexington Kentucky 40536 USA
- University of Kentucky Markey Cancer Center, University of Kentucky Lexington KY 40536 USA
| |
Collapse
|
8
|
Mitchell RJ, Gowda AS, Olivelli AG, Huckaba AJ, Parkin S, Unrine JM, Oza V, Blackburn JS, Ladipo F, Heidary DK, Glazer EC. Triarylphosphine-Coordinated Bipyridyl Ru(II) Complexes Induce Mitochondrial Dysfunction. Inorg Chem 2023; 62:10940-10954. [PMID: 37405779 DOI: 10.1021/acs.inorgchem.3c00736] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
While cancer cells rely heavily upon glycolysis to meet their energetic needs, reducing the importance of mitochondrial oxidative respiration processes, more recent studies have shown that their mitochondria still play an active role in the bioenergetics of metastases. This feature, in combination with the regulatory role of mitochondria in cell death, has made this organelle an attractive anticancer target. Here, we report the synthesis and biological characterization of triarylphosphine-containing bipyridyl ruthenium (Ru(II)) compounds and found distinct differences as a function of the substituents on the bipyridine and phosphine ligands. 4,4'-Dimethylbipyridyl-substituted compound 3 exhibited especially high depolarizing capabilities, and this depolarization was selective for the mitochondrial membrane and occurred within minutes of treatment in cancer cells. The Ru(II) complex 3 exhibited an 8-fold increase in depolarized mitochondrial membranes, as determined by flow cytometry, which compares favorably to the 2-fold increase observed by carbonyl cyanide chlorophenylhydrazone (CCCP), a proton ionophore that shuttles protons across membranes, depositing them into the mitochondrial matrix. Fluorination of the triphenylphosphine ligand provided a scaffold that maintained potency against a range of cancer cells but avoided inducing toxicity in zebrafish embryos at higher concentrations, displaying the potential of these Ru(II) compounds for anticancer applications. This study provides essential information regarding the role of ancillary ligands for the anticancer activity of Ru(II) coordination compounds that induce mitochondrial dysfunction.
Collapse
Affiliation(s)
- Richard J Mitchell
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506, United States
| | - Anitha S Gowda
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506, United States
| | - Alexander G Olivelli
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506, United States
| | - Aron J Huckaba
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506, United States
| | - Sean Parkin
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506, United States
| | - Jason M Unrine
- Department of Plant and Soil Sciences, University of Kentucky, 1100 S. Limestone Street, Lexington, Kentucky 40546, United States
| | - Viral Oza
- Department of Molecular and Cell Biology, University of Kentucky, 741 S. Limestone Street, Lexington, Kentucky 40536, United States
| | - Jessica S Blackburn
- Department of Molecular and Cell Biology, University of Kentucky, 741 S. Limestone Street, Lexington, Kentucky 40536, United States
| | - Folami Ladipo
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506, United States
| | - David K Heidary
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506, United States
| | - Edith C Glazer
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506, United States
| |
Collapse
|
9
|
Mertens RT, Gukathasan S, Arojojoye AS, Olelewe C, Awuah SG. Next Generation Gold Drugs and Probes: Chemistry and Biomedical Applications. Chem Rev 2023; 123:6612-6667. [PMID: 37071737 PMCID: PMC10317554 DOI: 10.1021/acs.chemrev.2c00649] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
The gold drugs, gold sodium thiomalate (Myocrisin), aurothioglucose (Solganal), and the orally administered auranofin (Ridaura), are utilized in modern medicine for the treatment of inflammatory arthritis including rheumatoid and juvenile arthritis; however, new gold agents have been slow to enter the clinic. Repurposing of auranofin in different disease indications such as cancer, parasitic, and microbial infections in the clinic has provided impetus for the development of new gold complexes for biomedical applications based on unique mechanistic insights differentiated from auranofin. Various chemical methods for the preparation of physiologically stable gold complexes and associated mechanisms have been explored in biomedicine such as therapeutics or chemical probes. In this Review, we discuss the chemistry of next generation gold drugs, which encompasses oxidation states, geometry, ligands, coordination, and organometallic compounds for infectious diseases, cancer, inflammation, and as tools for chemical biology via gold-protein interactions. We will focus on the development of gold agents in biomedicine within the past decade. The Review provides readers with an accessible overview of the utility, development, and mechanism of action of gold-based small molecules to establish context and basis for the thriving resurgence of gold in medicine.
Collapse
Affiliation(s)
- R Tyler Mertens
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Sailajah Gukathasan
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Adedamola S Arojojoye
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Chibuzor Olelewe
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
- University of Kentucky Markey Cancer Center, Lexington, Kentucky 40536, United States
| |
Collapse
|
10
|
Eswari S, Lakshmanan P, Thirumaran S. Synthesis, spectral, crystal structure, DFT and Hirshfeld surface analysis of Zn(II) complexes involving dithiocarbamate and imine ligands and usage as precursor to prepare ZnS for photodegradation of dyes. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
11
|
Au(III) Cyclometallated Compounds with 2-Arylpyridines and Their Derivatives or Analogues: 34 Years (1989–2022) of NMR and Single Crystal X-ray Studies. INORGANICS 2023. [DOI: 10.3390/inorganics11030100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
A review paper on Au(III) cyclometallated compounds with 2-arylpyridines (2-phenylpyridine, 2-benzylpyridine, 2-benzoylpyridine, 2-phenoxypyridine, 2-phenylsulfanylpyridine, 2-anilinopyridine, 2-(naphth-2-yl)pyridine, 2-(9,9-dialkylfluoren-2-yl)pyridines, 2-(dibenzofuran-4-yl)pyridine, and their derivatives) and their analogues (2-arylquinolines, 1- and 3-arylisoquinolines, 7,8-benzoquinoline), with 113 references. A total of 554 species, containing κ2-N(1),C(6′)*-Au(III), or analogous moiety (i.e., chelated by nitrogen of the pyridine-like ring and the deprotonated ortho- carbon of the phenyl-like ring) and, thus, possessing a character intermediate between metal complexes and organometallics, studied in the years 1989–2022 by NMR spectroscopy and/or single crystal X-ray diffraction (207 X-ray structures), are described. The compounds for which biological or catalytic activity and the luminescence properties were studied are also quoted.
Collapse
|
12
|
Moreno-Alcántar G, Picchetti P, Casini A. Gold Complexes in Anticancer Therapy: From New Design Principles to Particle-Based Delivery Systems. Angew Chem Int Ed Engl 2023; 62:e202218000. [PMID: 36847211 DOI: 10.1002/anie.202218000] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 02/28/2023]
Abstract
The discovery of the medicinal properties of gold complexes has fuelled the design and synthesis of new anticancer metallodrugs, which have received special attention due to their unique modes of action. Current research in the development of gold compounds with therapeutic properties is predominantly focused on the molecular design of drug leads with superior pharmacological activities, e.g., by introducing targeting features. Moreover, intensive research aims at improving the physicochemical properties of gold compounds, such as chemical stability and solubility in the physiological environment. In this regard, the encapsulation of gold compounds in nanocarriers or their chemical grafting onto targeted delivery vectors could lead to new nanomedicines that eventually reach clinical applications. Herein, we provide an overview of the state-of-the-art progress of gold anticancer compounds, andmore importantly we thoroughly revise the development of nanoparticle-based delivery systems for gold chemotherapeutics.
Collapse
Affiliation(s)
- Guillermo Moreno-Alcántar
- Chair of Medicinal and Bioinorganic Chemistry, School of Natural Sciences, Department of Chemistry, Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching b. München, Germany
| | - Pierre Picchetti
- Karlsruhe Institute of Technology (KIT), Institute of Nanotechnology, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Angela Casini
- Chair of Medicinal and Bioinorganic Chemistry, School of Natural Sciences, Department of Chemistry, Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching b. München, Germany
| |
Collapse
|
13
|
Johnson A, Olelewe C, Kim JH, Northcote-Smith J, Mertens RT, Passeri G, Singh K, Awuah SG, Suntharalingam K. The anti-breast cancer stem cell properties of gold(i)-non-steroidal anti-inflammatory drug complexes. Chem Sci 2023; 14:557-565. [PMID: 36741517 PMCID: PMC9847679 DOI: 10.1039/d2sc04707a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
The anti-breast cancer stem cell (CSC) properties of a series of gold(i) complexes comprising various non-steroidal anti-inflammatory drugs (NSAIDs) and triphenylphosphine 1-8 are reported. The most effective gold(i)-NSAID complex 1, containing indomethacin, exhibits greater potency for breast CSCs than bulk breast cancer cells (up to 80-fold). Furthermore, 1 reduces mammosphere viability to a better extent than a panel of clinically used breast cancer drugs and salinomycin, an established anti-breast CSC agent. Mechanistic studies suggest 1-induced breast CSC death results from breast CSC entry, cytoplasm localisation, an increase in intracellular reactive oxygen species levels, cyclooxygenase-2 downregulation and inhibition, and apoptosis. Remarkably, 1 also significantly inhibits tumour growth in a murine metastatic triple-negative breast cancer model. To the best of our knowledge, 1 is the first gold complex of any geometry or oxidation state to demonstrate anti-breast CSC properties.
Collapse
Affiliation(s)
- Alice Johnson
- School of Chemistry, University of LeicesterLeicesterUK,Biomolecular Sciences Research Centre, Sheffield Hallam UniversitySheffieldUK
| | - Chibuzor Olelewe
- Department of Chemistry, University of KentuckyLexingtonKentuckyUSA
| | - Jong Hyun Kim
- Department of Chemistry, University of KentuckyLexingtonKentuckyUSA
| | | | - R. Tyler Mertens
- Department of Chemistry, University of KentuckyLexingtonKentuckyUSA
| | | | - Kuldip Singh
- School of Chemistry, University of LeicesterLeicesterUK
| | - Samuel G. Awuah
- Department of Chemistry, University of KentuckyLexingtonKentuckyUSA,Department of Pharmaceutical Sciences, University of KentuckyLexingtonKentuckyUSA
| | | |
Collapse
|
14
|
Molecular subtypes based on cuproptosis-related genes and tumor microenvironment infiltration characteristics in pancreatic adenocarcinoma. Cancer Cell Int 2023; 23:7. [PMID: 36647100 PMCID: PMC9844034 DOI: 10.1186/s12935-022-02836-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 12/14/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Multiple molecular subtypes with distinct clinical outcomes in pancreatic adenocarcinoma (PAAD) have been identified in recent years. Cuproptosis is a new form of cell death that likely involved in tumor progression. However, the cuproptosis-related molecular subtypes as well as its mediated tumor microenvironment (TME) cell infiltration characteristics largely remain unclear. METHODS Expression profiles of 10 cuproptosis-related genes (CRGs) and their association with patient survival, TME, cancer stemness and drug resistance were studied in 33 cancer types using the TCGA pan-cancer data. Using 437 PAAD samples from five cohorts (TCGA-PAAD cohort and four GEO cohorts), we explored the molecular subtypes mediated by CRGs, along with the associated TME cell infiltration. Unsupervised methods were utilized to perform cuproptosis subtype clustering. The cuproptosis score was constructed using the COX regression model with least absolute shrinkage and selection operator regression (LASSO) algorithm to quantify the cuproptosis characteristics of a single tumor. RESULTS The expression of 10 CRGs varies in different cancer types with striking inter- and intra- cancer heterogeneity. We integrated the genomic profiling of the CRGs and identified three distinct cuproptosis subtypes, and found that multi-layer CRG alterations were correlated with patient prognosis and TME cell infiltration characteristics. In addition, a cuproptosis score signature was constructed to predict prognosis, and its clinical impacts were characterized in the TCGA-PAAD cohort. The cuproptosis signature was significantly associated with prognosis, tumor subtypes, CD8 T-cell infiltration, response to immune checkpoint inhibitors (ICIs) and chemotherapeutic drug sensitivity. Furthermore, the expression patterns of CRGs in pancreatic cancer cells and normal controls were validated, which was almost consistent with the results from the public database. The expression level and prognostic predictive capability of DLAT were verified in 97 PAAD patients from our patient cohort. CONCLUSIONS These findings may help understand the roles of CRGs in PAAD and the molecular characterization of cuproptosis subtypes. In addition, the cuproptosis score could serve as a promising biomarker for predicting prognosis and response to immunotherapy in PAAD patients.
Collapse
|
15
|
Abdel Aal S. DFT study of the therapeutic potential of borospherene and metalloborospherenes as a new drug-delivery system for the 5-fluorouracil anticancer drug. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
16
|
Maity S, Tomar MS, Wasnik K, Patra S, Modak MD, Gupta PS, Pareek D, Singh M, Paik P. Azadirachta indica Seed Derived Carbon Nanocapsules: Cell Imaging, Depolarization of Mitochondrial Membrane Potential, and Dose-Dependent Control Death of Breast Cancer. ACS Biomater Sci Eng 2022; 8:3608-3622. [PMID: 35892286 DOI: 10.1021/acsbiomaterials.2c00463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this work, a series of mesoporous carbon nanocapsules (mCNS) of size below 10 nm have been prepared from Azadirachta indica seeds with a very easy and cost-effective approach. These nanocapsules can emit red and green light and are effective for cell imaging. Further, these carbon nanocapsules are biocompatible toward the normal healthy cells, however, they possess modest cytotoxicity against the MCF-7 (human breast cancer) and triple-negative breast cancer (TNBC) (MDA- MB-231 breast cancer cells), and the rate of killing cancer cells strongly depends on the dose of mCNCs. Further, the mitochondrial membrane potential and apoptosis assay were performed to analyze the therapeutic significance of these nanocapsules to kill breast cancer. Results showed that these carbon nanocapsules can depolarize the mitochondrial membrane potential alone (without using conventional drugs) and can change the physiological parameters and cellular metabolic energy of the cancer cells and kill them. The apoptosis results confirmed the death of breast cancer cells in the form of apoptosis and necrosis. Moreover, the results suggested that the porous carbon nanocapsules (mCNCs) reported herein can be used as a potential candidate and useful for the theranostic applications such as for cancer cell detection and therapy without using any conventional drugs.
Collapse
Affiliation(s)
- Somedutta Maity
- School of Engineering Sciences and Technology, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Munendra Singh Tomar
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221 005, Uttar Pradesh, India.,Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Kirti Wasnik
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221 005, Uttar Pradesh, India
| | - Sukanya Patra
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221 005, Uttar Pradesh, India
| | - Monami Das Modak
- School of Engineering Sciences and Technology, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Prem Shankar Gupta
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221 005, Uttar Pradesh, India
| | - Divya Pareek
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221 005, Uttar Pradesh, India
| | - Monika Singh
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221 005, Uttar Pradesh, India
| | - Pradip Paik
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221 005, Uttar Pradesh, India
| |
Collapse
|
17
|
Olelewe C, Kim JH, Ofori S, Mertens RT, Gukathasan S, Awuah SG. Gold(III)-P-chirogenic complex induces mitochondrial dysfunction in triple-negative breast cancer. iScience 2022; 25:104340. [PMID: 35602949 PMCID: PMC9117869 DOI: 10.1016/j.isci.2022.104340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/14/2022] [Accepted: 04/27/2022] [Indexed: 12/19/2022] Open
Abstract
Chemical agents that specifically exploit metabolic vulnerabilities of cancer cells will be beneficial but are rare. The role of oxidative phosphorylation (OXPHOS) in promoting and maintaining triple-negative breast cancer (TNBC) growth provides new treatment opportunity. In this work, we describe AuPhos-19, a small-molecule gold(III)-based agent bearing a chiral phosphine ligand that selectively disrupts mitochondrial metabolism in murine and human TNBC cells but not normal epithelial cells. AuPhos-19 induces potent cytotoxic effect with half maximal inhibitory concentration (IC50) in the nanomolar range (220-650 nM) across different TNBC cell lines. The lipophilic cationic character of AuPhos-19 facilitates interaction with mitochondrial OXPHOS. AuPhos-19 inhibits mitochondria respiration and induces significant AMPK activation. Depolarization of the mitochondria membrane, mitochondria ROS accumulation, and mitochondria DNA depletion provided further indication that AuPhos-19 perturbs mitochondria function. AuPhos-19 inhibits tumor growth in tumor-bearing mice. This study highlights the development of gold-based compounds targeting mitochondrial pathways for efficacious cancer treatment.
Collapse
Affiliation(s)
- Chibuzor Olelewe
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Jong Hyun Kim
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Randall T. Mertens
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | | | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
18
|
Novel bifluorescent Zn(II)–cryptolepine–cyclen complexes trigger apoptosis induced by nuclear and mitochondrial DNA damage in cisplatin-resistant lung tumor cells. Eur J Med Chem 2022; 238:114418. [DOI: 10.1016/j.ejmech.2022.114418] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 12/15/2022]
|
19
|
Ma L, Xue X, Zhang X, Yu K, Xu X, Tian X, Miao Y, Meng F, Liu X, Guo S, Qiu S, Wang Y, Cui J, Guo W, Li Y, Xia J, Yu Y, Wang J. The essential roles of m 6A RNA modification to stimulate ENO1-dependent glycolysis and tumorigenesis in lung adenocarcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:36. [PMID: 35078505 PMCID: PMC8788079 DOI: 10.1186/s13046-021-02200-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/26/2021] [Indexed: 12/31/2022]
Abstract
Background Lung adenocarcinoma (LUAD) is the most common subtype of lung cancer. Patient prognosis is poor, and the existing therapeutic strategies for LUAD are far from satisfactory. Recently, targeting N6-methyladenosine (m6A) modification of RNA has been suggested as a potential strategy to impede tumor progression. However, the roles of m6A modification in LUAD tumorigenesis is unknown. Methods Global m6A levels and expressions of m6A writers, erasers and readers were evaluated by RNA methylation assay, dot blot, immunoblotting, immunohistochemistry and ELISA in human LUAD, mouse models and cell lines. Cell viability, 3D-spheroid generation, in vivo LUAD formation, experiments in cell- and patient-derived xenograft mice and survival analysis were conducted to explore the impact of m6A on LUAD. The RNA-protein interactions, translation, putative m6A sites and glycolysis were explored in the investigation of the mechanism underlying how m6A stimulates tumorigenesis. Results The elevation of global m6A level in most human LUAD specimens resulted from the combined upregulation of m6A writer methyltransferase 3 (METTL3) and downregulation of eraser alkB homolog 5 (ALKBH5). Elevated global m6A level was associated with a poor overall survival in LUAD patients. Reducing m6A levels by knocking out METTL3 and overexpressing ALKBH5 suppressed 3D-spheroid generation in LUAD cells and intra-pulmonary tumor formation in mice. Mechanistically, m6A-dependent stimulation of glycolysis and tumorigenesis occurred via enolase 1 (ENO1). ENO1 mRNA was m6A methylated at 359 A, which facilitated it’s binding with the m6A reader YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) and resulted in enhanced translation of ENO1. ENO1 positively correlated with METTL3 and global m6A levels, and negatively correlated with ALKBH5 in human LUAD. In addition, m6A-dependent elevation of ENO1 was associated with LUAD progression. In preclinical models, tumors with a higher global m6A level showed a more sensitive response to the inhibition of pan-methylation, glycolysis and ENO activity in LUAD. Conclusions The m6A-dependent stimulation of glycolysis and tumorigenesis in LUAD is at least partially orchestrated by the upregulation of METTL3, downregulation of ALKBH5, and stimulation of YTHDF1-mediated ENO1 translation. Blocking this mechanism may represent a potential treatment strategy for m6A-dependent LUAD. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02200-5.
Collapse
Affiliation(s)
- Lifang Ma
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China.,Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - Xiangfei Xue
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, 200072, Shanghai, China
| | - Xiao Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - Keke Yu
- Department of Bio-bank, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Xin Xu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - Xiaoting Tian
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - Yayou Miao
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - Fanyu Meng
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - Xiaoxin Liu
- Nursing Department, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Susu Guo
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, 200072, Shanghai, China
| | - Shiyu Qiu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - Yikun Wang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - Jiangtao Cui
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - Wanxin Guo
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - You Li
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China
| | - Jinjing Xia
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China.
| | - Yongchun Yu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China.
| | - Jiayi Wang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China. .,Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, 200030, Shanghai, China. .,Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, 200072, Shanghai, China.
| |
Collapse
|
20
|
Marloye M, Inam H, Moore CJ, Mertens TR, Ingels A, Koch M, Nowicki MO, Mathieu V, Pritchard JR, Awuah SG, Lawler SE, Meyer F, Dufrasne F, Berger G. Self-assembled ruthenium and osmium nanosystems display a potent anticancer profile by interfering with metabolic activity. Inorg Chem Front 2022; 9:2594-2607. [PMID: 36311556 PMCID: PMC9610622 DOI: 10.1039/d2qi00423b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Amphiphilic ruthenium and osmium complexes auto-assemble to nanosystems that poison mitochondria and show highly promising in vitro and in vivo anticancer activity.
Collapse
Affiliation(s)
- Mickaël Marloye
- Microbiology, Bioorganic & Macromolecular Chemistry Unit, Faculté de Pharmacie, Université libre de Bruxelles (ULB), Boulevard du Triomphe, 1050 Brussels, Belgium
| | - Haider Inam
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Connor J. Moore
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Tyler R. Mertens
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Aude Ingels
- Department of Pharmacotherapy and Pharmaceutics, Faculté de Pharmacie, Université libre de Bruxelles (ULB), Boulevard du Triomphe, 1050 Brussels, Belgium
| | - Marilin Koch
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michal O. Nowicki
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Véronique Mathieu
- Department of Pharmacotherapy and Pharmaceutics, Faculté de Pharmacie, Université libre de Bruxelles (ULB), Boulevard du Triomphe, 1050 Brussels, Belgium
- ULB Cancer Research Center (UCRC), Université libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - Justin R. Pritchard
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Sean E. Lawler
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Franck Meyer
- Microbiology, Bioorganic & Macromolecular Chemistry Unit, Faculté de Pharmacie, Université libre de Bruxelles (ULB), Boulevard du Triomphe, 1050 Brussels, Belgium
| | - François Dufrasne
- Microbiology, Bioorganic & Macromolecular Chemistry Unit, Faculté de Pharmacie, Université libre de Bruxelles (ULB), Boulevard du Triomphe, 1050 Brussels, Belgium
| | - Gilles Berger
- Microbiology, Bioorganic & Macromolecular Chemistry Unit, Faculté de Pharmacie, Université libre de Bruxelles (ULB), Boulevard du Triomphe, 1050 Brussels, Belgium
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
21
|
Pramanik SK, Das A. Fluorescent probes for imaging bioactive species in subcellular organelles. Chem Commun (Camb) 2021; 57:12058-12073. [PMID: 34706371 DOI: 10.1039/d1cc04273d] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Luminescent molecular probes and nanoscale materials have become important tools in biosensing and bioimaging applications because of their high sensitivity, fast response, specificity, and methodological simplicity. In recent years, there has been a notable advancement in fluorescent probes that respond to the subtle changes in subcellular microenvironments (e.g., polarity, pH, and viscosity) or distribution of certain crucial biomarkers (e.g., reactive oxygen species, ions, amino acids, and enzymes). The dynamic fluctuations of these bio-molecules in subcellular microenvironments control cellular homeostasis, immunity, signal conduction, and metabolism. Their abnormal expressions are linked to various biological disorders and disease states. Thus, the real-time monitoring of such bioactive species is intimately linked to clinical diagnostics. Appropriately designed luminescent probes are ideally suited for desired organelle specificity, as well as for reporting intracellular changes in biochemicals/microenvironmental factors with the luminescence ON response. In this perspective, we review our recent work on the development of fluorescent probes for sensing and imaging within sub-cellular organelles. We have also discussed the design aspects for developing a prodrug with a fluorescent probe as an integral part of possible theranostic applications. An overview of the design principles, photophysical properties, detection mechanisms, current challenges, and potential future directions of fluorescent probes is presented in this feature article. We have also discussed the limitations and challenges of developing the solution platform for sensing technologies in clinical diagnostics.
Collapse
Affiliation(s)
- Sumit Kumar Pramanik
- CSIR-Central Salt and Marine Chemicals Research Institute, Gijubhai Badheka Marg, Bhavnagar, Gujarat, 364002, India.
| | - Amitva Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741 246, West Bengal, India.
| |
Collapse
|
22
|
Khodjoyan S, Remadna E, Dossmann H, Lesage D, Gontard G, Forté J, Hoffmeister H, Basu U, Ott I, Spence P, Waller ZAE, Salmain M, Bertrand B. [(C C)Au(N N)] + Complexes as a New Family of Anticancer Candidates: Synthesis, Characterization and Exploration of the Antiproliferative Properties. Chemistry 2021; 27:15773-15785. [PMID: 34436799 DOI: 10.1002/chem.202102751] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Indexed: 12/31/2022]
Abstract
A library of eleven cationic gold(III) complexes of the general formula [(C C)Au(N N)]+ when C C is either biphenyl or 4,4'-ditertbutyldiphenyl and N N is a bipyridine, phenanthroline or dipyridylamine derivative have been synthesized and characterized. Contrasting effects on the viability of the triple negative breast cancer cells MDA-MB-231 was observed from a preliminary screening. The antiproliferative activity of the seven most active complexes were further assayed on a larger panel of human cancer cells as well as on non-cancerous cells for comparison. Two complexes stood out for being either highly active or highly selective. Eventually, reactivity studies with biologically meaningful amino acids, glutathione, higher order DNA structures and thioredoxin reductase (TrxR) revealed a markedly different behavior from that of the well-known coordinatively isomeric [(C N C)Au(NHC)]+ structure. This makes the [(C C)Au(N N)]+ complexes a new class of organogold compounds with an original mode of action.
Collapse
Affiliation(s)
- Silva Khodjoyan
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, 75005, Paris, France
| | - Edwyn Remadna
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, 75005, Paris, France
| | - Héloïse Dossmann
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, 75005, Paris, France
| | - Denis Lesage
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, 75005, Paris, France
| | - Geoffrey Gontard
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, 75005, Paris, France
| | - Jérémy Forté
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, 75005, Paris, France
| | - Henrik Hoffmeister
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstrasse 55, 38106, Braunschweig, Germany
| | - Uttara Basu
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstrasse 55, 38106, Braunschweig, Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstrasse 55, 38106, Braunschweig, Germany
| | - Philip Spence
- School of Pharmacy, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Zoë A E Waller
- School of Pharmacy, University of East Anglia, Norwich, NR4 7TJ, UK.,UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1 N, UK
| | - Michèle Salmain
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, 75005, Paris, France
| | - Benoît Bertrand
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, 75005, Paris, France
| |
Collapse
|
23
|
Nayeem N, Yeasmin A, Cobos SN, Younes A, Hubbard K, Contel M. Investigation of the Effects and Mechanisms of Anticancer Action of a Ru(II)-Arene Iminophosphorane Compound in Triple Negative Breast Cancer Cells. ChemMedChem 2021; 16:3280-3292. [PMID: 34329530 PMCID: PMC8571052 DOI: 10.1002/cmdc.202100325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/23/2021] [Indexed: 12/22/2022]
Abstract
Triple negative breast cancer (TNBC) is one of the breast cancers with poorer prognosis and survival rates. TNBC has a disproportionally high incidence and mortality in women of African descent. We report on the evaluation of Ru-IM (1), a water-soluble organometallic ruthenium compound, in TNBC cell lines derived from patients of European (MDA-MB-231) and African (HCC-1806) ancestry (including IC50 values, cellular and organelle uptake, cell death pathways, cell cycle, effects on migration, invasion, and angiogenesis, a preliminary proteomic analysis, and an NCI 60 cell-line panel screen). 1 was previously found highly efficacious in MDA-MB-231 cells and xenografts, with little systemic toxicity and preferential accumulation in the tumor. We observe a similar profile for this compound in the two cell lines studied, which includes high cytotoxicity, apoptotic behavior and potential antimetastatic and antiangiogenic properties. Cytokine M-CSF, involved in the PI3/AKT pathway, shows protein expression inhibition with exposure to 1. We also demonstrate a p53 independent mechanism of action.
Collapse
Affiliation(s)
- Nazia Nayeem
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| | - Arefa Yeasmin
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Samantha N Cobos
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| | - Ali Younes
- Department of Chemistry, Hunter College, The City University of New York, 695 Park Avenue, New York, NY, 10065, USA
| | - Karen Hubbard
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
- Biology Department, The City College of New York, The City University of New York, 160 Covent Avenue, New York, NY, 10031, USA
| | - Maria Contel
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| |
Collapse
|
24
|
Hyun Kim J, Ofori S, Mertens RT, Parkin S, Awuah SG. Water-Soluble Gold(III)-Metformin Complex Alters Mitochondrial Bioenergetics in Breast Cancer Cells. ChemMedChem 2021; 16:3222-3230. [PMID: 34159760 PMCID: PMC8526394 DOI: 10.1002/cmdc.202100233] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/16/2021] [Indexed: 12/30/2022]
Abstract
Chemical control of mitochondrial dynamics and bioenergetics can unravel fundamental biological mechanisms and therapeutics for several diseases including, diabetes and cancer. We synthesized stable, water-soluble gold(III) complexes (Auraformin) supported by biguanide metformin or phenylmetformin for efficacious inhibition of mitochondrial respiration. The new compounds were characterized following the reaction of [C N]-cyclometalated gold(III) compounds with respective biguanides. Auraformin is solution stable in a physiologically relevant environment. We show that auraformin decreases mitochondrial respiration efficiently in comparison to the clinically used metformin by 100-fold. The compound displays significant mitochondrial uptake and induces antiproliferative activity in the micromolar range. Our results shed light on the development of new scaffolds as improved inhibitors of mitochondrial respiration.
Collapse
Affiliation(s)
- Jong Hyun Kim
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, KY 40506, USA
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, KY 40506, USA
| | - R Tyler Mertens
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, KY 40506, USA
| | - Sean Parkin
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, KY 40506, USA
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, KY 40506, USA
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone Street, Lexington, KY 40506, USA
| |
Collapse
|
25
|
Wang T, Ashrafi A, Konduri PC, Ghosh P, Dey S, Modareszadeh P, Salamat N, Alemi PS, Berisha E, Zhang L. Heme Sequestration as an Effective Strategy for the Suppression of Tumor Growth and Progression. Mol Cancer Ther 2021; 20:2506-2518. [PMID: 34552010 DOI: 10.1158/1535-7163.mct-21-0033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/14/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022]
Abstract
Heme is an essential nutritional, metabolic, and signaling molecule in living organisms. Pathogenic microbes extract heme from hosts to obtain metallonutrient, while heme fuels mitochondrial respiration and ATP generation in lung tumor cells. Here, we generated small heme-sequestering proteins (HeSPs) based on bacterial hemophores. These HeSPs contain neutral mutations in the heme-binding pocket and hybrid sequences from hemophores of different bacteria. We showed that HeSPs bind to heme and effectively extracted heme from hemoglobin. They strongly inhibited heme uptake and cell proliferation and induced apoptosis in non-small cell lung cancer (NSCLC) cells, while their effects on nontumorigenic cell lines representing normal lung cells were not significant. HeSPs strongly suppressed the growth of human NSCLC tumor xenografts in mice. HeSPs decreased oxygen consumption rates and ATP levels in tumor cells isolated from treated mice, while they did not affect liver and blood cell functions. IHC, along with data from Western blotting and functional assays, revealed that HeSPs reduced the levels of key proteins involved in heme uptake, as well as the consumption of major fuels for tumor cells, glucose, and glutamine. Further, we found that HeSPs reduced the levels of angiogenic and vascular markers, as well as vessel density in tumor tissues. Together, these results demonstrate that HeSPs act via multiple mechanisms, including the inhibition of oxidative phosphorylation, to suppress tumor growth and progression. Evidently, heme sequestration can be a powerful strategy for suppressing lung tumors and likely drug-resistant tumors that rely on oxidative phosphorylation for survival.
Collapse
Affiliation(s)
- Tianyuan Wang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Adnin Ashrafi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | | | - Poorva Ghosh
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Sanchareeka Dey
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Parsa Modareszadeh
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Narges Salamat
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Parinaz Sadat Alemi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Eranda Berisha
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Li Zhang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas.
| |
Collapse
|
26
|
Spectroscopic analysis and molecular structures of mononuclear bis(t-butyltrithiocarbonato)-nickel, -palladium and (t-butyltrithiocarbonato)(t-butylthiolato)platinum dimer. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2021.120382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
27
|
Pettenuzzo A, Vezzù K, Di Paolo ML, Fotopoulou E, Marchiò L, Via LD, Ronconi L. Design, physico-chemical characterization and in vitro biological activity of organogold(III) glycoconjugates. Dalton Trans 2021; 50:8963-8979. [PMID: 34110336 DOI: 10.1039/d1dt01100f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To develop new metal-based glycoconjugates as potential anticancer agents, four organometallic gold(iii)-dithiocarbamato glycoconjugates of the type [AuIII(2-Bnpy)(SSC-Inp-GlcN)](PF6) (2-Bnpy: 2-benzylpyridine; Inp: isonipecotic moiety; GlcN: amino-glucose scaffold; Au3-Au6) and the corresponding model non-glycosylated counterparts [AuIII(2-Bnpy)(SSC-Inp-R)](PF6) (R: OEt (Au1), NH2 (Au2)) have been generated and characterized by means of several analytical techniques (elemental analysis, FT-IR, 1H-/13C-NMR, ESI-MS, UV-Vis, X-ray crystallography). Their stability under physiologically-relevant conditions (PBS solution) and n-octanol/PBS distribution coefficient (D7.4) have also been evaluated. Gold(iii) glycoconjugates showed an antiproliferative effect against ovarian carcinoma A2780 cells, with GI50 values in the low micromolar range. Remarkably, their cell growth inhibitory effect increases upon the addition of a glucose transporter 1 (GLUT1) inhibitor, thus ruling out the involvement of GLUT1 in their transport inside the cell. Additional mechanistic studies have been carried out in A2780 cells, supporting the hypothesis of a facilitated diffusion mechanism (possibly mediated by glucose transporters other than GLUT1), and revealing their capability to act as topoisomerase I and II inhibitors and to disrupt mitochondrial membrane integrity, leading to the generation of ROS, thus resulting in the promotion of oxidative stress and, eventually, cell death.
Collapse
Affiliation(s)
- Andrea Pettenuzzo
- National University of Ireland Galway, School of Chemistry, University Road, H91 TK33 Galway, Co. Galway, Ireland.
| | - Keti Vezzù
- University of Padova, Department of Industrial Engineering, Via F. Marzolo 8, 35131 Padova, Italy
| | - Maria Luisa Di Paolo
- University of Padova, Department of Molecular Medicine, Via G. Colombo 3, 35131 Padova, Italy
| | - Eirini Fotopoulou
- National University of Ireland Galway, School of Chemistry, University Road, H91 TK33 Galway, Co. Galway, Ireland.
| | - Luciano Marchiò
- University of Parma, Department of Chemistry, Life Sciences and Environmental Sustainability, Parco Area delle Scienze 11/a, 43124 Parma, Italy
| | - Lisa Dalla Via
- University of Padova, Department of Pharmaceutical and Pharmacological Sciences, Via F. Marzolo 5, 35131 Padova, Italy.
| | - Luca Ronconi
- National University of Ireland Galway, School of Chemistry, University Road, H91 TK33 Galway, Co. Galway, Ireland.
| |
Collapse
|
28
|
Zhang J, Jiang M, Li S, Zhang Z, Sun H, Yang F, Liang H. Developing a Novel Anticancer Gold(III) Agent to Integrate Chemotherapy and Immunotherapy. J Med Chem 2021; 64:6777-6791. [PMID: 34000198 DOI: 10.1021/acs.jmedchem.1c00050] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
To effectively treat gastric cancer, we innovatively attempted to develop a metal agent to integrate immunotherapy and chemotherapy by dual targeting the cellular components in the tumor microenvironment (TME) based on the specific residue of human serum albumin (HSA) nanoparticles (NPs). We synthesized a series of Au(III) α-N-heterocyclic thiosemicarbazone compounds and obtained a Au agent (5b) with remarkable cytotoxicity to gastric cancer cells; moreover, we successfully constructed a novel HSA-5b complex NP delivery system. Importantly, the in vivo results showed that 5b/HSA-5b NPs effectively inhibited gastric tumor growth and HSA-5b NPs enhanced the therapeutic efficiency, bioavailability, and targeting ability compared with those of 5b alone. Furthermore, the in vitro/in vivo results revealed that 5b/HSA-5b NPs could integrate chemotherapy and immunotherapy by synergistically attacking two different cellular components in TME at the same time, namely, polarizing the tumor-associated macrophages and inducing apoptosis of gastric cancer cells.
Collapse
Affiliation(s)
- Juzheng Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Ming Jiang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Shanhe Li
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Zhenlei Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China
| | - Feng Yang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| |
Collapse
|
29
|
Kim JH, Ofori S, Parkin S, Vekaria H, Sullivan PG, Awuah SG. Anticancer gold(iii)-bisphosphine complex alters the mitochondrial electron transport chain to induce in vivo tumor inhibition. Chem Sci 2021; 12:7467-7479. [PMID: 34163837 PMCID: PMC8171344 DOI: 10.1039/d1sc01418h] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/16/2021] [Indexed: 01/10/2023] Open
Abstract
Expanding the chemical diversity of metal complexes provides a robust platform to generate functional bioactive reagents. To access an excellent repository of metal-based compounds for probe/drug discovery, we capitalized on the rich chemistry of gold to create organometallic gold(iii) compounds by ligand tuning. We obtained novel organogold(iii) compounds bearing a 1,2-bis(diphenylphosphino)benzene ligand, providing structural diversity with optimal physiological stability. Biological evaluation of the lead compound AuPhos-89 demonstrates mitochondrial complex I-mediated alteration of the mitochondrial electron transport chain (ETC) to drive respiration and diminish cellular energy in the form of adenosine triphosphate (ATP). Mechanism-of-action efforts, RNA-Seq, quantitative proteomics, and NCI-60 screening reveal a highly potent anticancer agent that modulates mitochondrial ETC. AuPhos-89 inhibits the tumor growth of metastatic triple negative breast cancer and represents a new strategy to study the modulation of mitochondrial respiration for the treatment of aggressive cancer and other disease states where mitochondria play a pivotal role in the pathobiology.
Collapse
Affiliation(s)
- Jong Hyun Kim
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Sean Parkin
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Hemendra Vekaria
- Spinal Cord and Brain Injury Research Center, University of Kentucky USA
- Department of Neuroscience, University of Kentucky USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky USA
- Department of Neuroscience, University of Kentucky USA
- Lexington Veterans' Affairs Healthcare System USA
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky Lexington Kentucky 40536 USA
| |
Collapse
|
30
|
Mertens RT, Jennings WC, Ofori S, Kim JH, Parkin S, Kwakye GF, Awuah SG. Synthetic Control of Mitochondrial Dynamics: Developing Three-Coordinate Au(I) Probes for Perturbation of Mitochondria Structure and Function. JACS AU 2021; 1:439-449. [PMID: 34467306 PMCID: PMC8395693 DOI: 10.1021/jacsau.1c00051] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Indexed: 06/13/2023]
Abstract
Mitochondrial structure and organization is integral to maintaining mitochondrial homeostasis and an emerging biological target in aging, inflammation, neurodegeneration, and cancer. The study of mitochondrial structure and its functional implications remains challenging in part because of the lack of available tools for direct engagement, particularly in a disease setting. Here, we report a gold-based approach to perturb mitochondrial structure in cancer cells. Specifically, the design and synthesis of a series of tricoordinate Au(I) complexes with systematic modifications to group 15 nonmetallic ligands establish structure-activity relationships (SAR) to identify physiologically relevant tools for mitochondrial perturbation. The optimized compound, AuTri-9 selectively disrupts breast cancer mitochondrial structure rapidly as observed by transmission electron microscopy with attendant effects on fusion and fission proteins. This phenomenon triggers severe depolarization of the mitochondrial membrane in cancer cells. The high in vivo tolerability of AuTri-9 in mice demonstrates its preclinical utility. This work provides a basis for rational design of gold-based agents to control mitochondrial structure and dynamics.
Collapse
Affiliation(s)
- R. Tyler Mertens
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - William C. Jennings
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel Ofori
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Jong Hyun Kim
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Sean Parkin
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Gunnar F. Kwakye
- Department
of Neuroscience, Oberlin College, Oberlin, Ohio 44074, United States
| | - Samuel G. Awuah
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| |
Collapse
|
31
|
Arojojoye AS, Mertens RT, Ofori S, Parkin SR, Awuah SG. Synthesis, Characterization, and Antiproliferative Activity of Novel Chiral [QuinoxP*AuCl 2] + Complexes. Molecules 2020; 25:E5735. [PMID: 33291802 PMCID: PMC7730091 DOI: 10.3390/molecules25235735] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 11/17/2022] Open
Abstract
Herein is reported the synthesis of two Au(III) complexes bearing the (R,R)-(-)-2,3-Bis(tert-butylmethylphosphino)quinoxaline (R,R-QuinoxP*) or (S,S)-(+)-2,3-Bis(tert-butylmethylphosphino)quinoxaline (S,S-QuinoxP*) ligands. By reacting two stoichiometric equivalents of HAuCl4.3H2O to one equivalent of the corresponding QuinoxP* ligand, (R,R)-(-)-2,3-Bis(tert-butylmethylphosphino)quinoxalinedichlorogold(III) tetrachloroaurates(III) (1) and (S,S)-(+)-2,3-Bis(tert-butylmethylphosphino)quinoxalinedichlorogold(III) tetrachloroaurates(III) (2) were formed, respectively, in moderate yields. The structure of (S,S)-(+)-2,3-Bis(tert-butylmethylphosphino)quinoxalinedichlorogold(III) tetrachloroaurates(III) (2) was further confirmed by X-ray crystallography. The antiproliferative activities of the two compounds were evaluated in a panel of cell lines and exhibited promising results comparable to auranofin and cisplatin with IC50 values between 1.08 and 4.83 µM. It is noteworthy that in comparison to other platinum and ruthenium enantiomeric complexes, the two enantiomers (1 and 2) do not exhibit different cytotoxic effects. The compounds exhibited stability in biologically relevant media over 48 h as well as inert reactivity to excess glutathione at 37 °C. These results demonstrate that the Au(III) atom, stabilized by the QuinoxP* ligand, can provide exciting compounds for novel anticancer drugs. These complexes provide a new scaffold to further develop a robust and diverse library of chiral phosphorus Au(III) complexes.
Collapse
Affiliation(s)
- Adedamola S. Arojojoye
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; (A.S.A.); (R.T.M.); (S.O.); (S.R.P.)
| | - R. Tyler Mertens
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; (A.S.A.); (R.T.M.); (S.O.); (S.R.P.)
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; (A.S.A.); (R.T.M.); (S.O.); (S.R.P.)
| | - Sean R. Parkin
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; (A.S.A.); (R.T.M.); (S.O.); (S.R.P.)
| | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; (A.S.A.); (R.T.M.); (S.O.); (S.R.P.)
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
32
|
Zhang J, Zhang Z, Jiang M, Li S, Yuan H, Sun H, Yang F, Liang H. Developing a Novel Gold(III) Agent to Treat Glioma Based on the Unique Properties of Apoferritin Nanoparticles: Inducing Lethal Autophagy and Apoptosis. J Med Chem 2020; 63:13695-13708. [PMID: 33185442 DOI: 10.1021/acs.jmedchem.0c01257] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Effective delivery of anticancer agents across the blood-brain barrier (BBB) required innovative strategies to achieve glioma regression. To resolve this problem, we proposed to develop a metal agent that target and treat glioma based on the unique property of apoferritin (AFt) nanoparticles (NPs). Thus, we synthesized a series of Au(III) 3-(4-metyl piperidine)thiosemicarbazides compounds and analyzed their structure-activity relationships, obtaining a Au agent (C6) with remarkable cytotoxicity in glioma. Moreover, we confirmed that C6 kills glioma cells by inducing lethal autophagy and apoptosis. Importantly, our results revealed that the successfully constructed apoferritin-C6 NPs (AFt-C6 NPs) can effectively cross the BBB, inhibit glioma growth, and selectively accumulate in tumors.
Collapse
Affiliation(s)
- Juzheng Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China. Guangxi Normal University, Guilin, Guangxi 541003, China
| | - Zhenlei Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China. Guangxi Normal University, Guilin, Guangxi 541003, China
| | - Ming Jiang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China. Guangxi Normal University, Guilin, Guangxi 541003, China
| | - Shanhe Li
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China. Guangxi Normal University, Guilin, Guangxi 541003, China
| | - Haoliang Yuan
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Feng Yang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China. Guangxi Normal University, Guilin, Guangxi 541003, China
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China. Guangxi Normal University, Guilin, Guangxi 541003, China
| |
Collapse
|