1
|
Arias-Betancur A, Fontova P, Alonso-Carrillo D, Carreira-Barral I, Duis J, García-Valverde M, Soto-Cerrato V, Quesada R, Pérez-Tomás R. Deregulation of lactate permeability using a small-molecule transporter (Lactrans-1) disturbs intracellular pH and triggers cancer cell death. Biochem Pharmacol 2024; 229:116469. [PMID: 39117009 DOI: 10.1016/j.bcp.2024.116469] [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: 02/14/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Due to the relevance of lactic acidosis in cancer, several therapeutic strategies have been developed targeting its production and/or regulation. In this matter, inhibition approaches of key proteins such as lactate dehydrogenase or monocarboxylate transporters have showed promising results, however, metabolic plasticity and tumor heterogeneity limits their efficacy. In this study, we explored the anticancer potential of a new strategy based on disturbing lactate permeability independently of monocarboxylate transporters activity using a small molecule ionophore named Lactrans-1. Derived from click-tambjamines, Lactrans-1 facilitates transmembrane lactate transportation in liposome models and reduces cancer cell viability. The results showed that Lactrans-1 triggered both apoptosis and necrosis depending on the cell line tested, displaying a synergistic effect in combination with first-line standard chemotherapeutic cisplatin. The ability of this compound to transport outward lactate anions was confirmed in A549 and HeLa cells, two cancer cell lines having distinct rates of lactate production. In addition, through cell viability reversion experiments it was possible to establish a correlation between the amount of lactate transported and the cytotoxic effect exhibited. The movement of lactate anions was accompanied with intracellular pH disturbances that included basification of lysosomes and acidification of the cytosol and mitochondria. We also observed mitochondrial swelling, increased ROS production and activation of oxidative stress signaling pathways p38-MAPK and JNK/SAPK. Our findings provide evidence that enhancement of lactate permeability is critical for cellular pH homeostasis and effective to trigger cancer cell death, suggesting that Lactrans-1 may be a promising anticancer therapy.
Collapse
Affiliation(s)
- Alain Arias-Betancur
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain; Molecular Signalling, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain; Department of Integral Adult Dentistry, Research Centre for Dental Sciences (CICO), Dental School, Universidad de La Frontera, 4811230 Temuco, Chile
| | - Pere Fontova
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain; Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Daniel Alonso-Carrillo
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Israel Carreira-Barral
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Janneke Duis
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain; Avans University of Applied Science, 4818 AJ Breda, the Netherlands
| | - María García-Valverde
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Vanessa Soto-Cerrato
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain; Molecular Signalling, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Roberto Quesada
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain.
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain; Molecular Signalling, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
2
|
Kumar A, Chithanna S, Li Y, Zhang X, Dodean RA, Caridha D, Madejczyk MS, Lee PJ, Jin X, Chetree R, Blount C, Dennis WE, DeLuca J, Vuong C, Pannone K, Dinh HT, Leed S, Roth A, Reynolds KA, Kelly JX, Kancharla P. Optimization of B-Ring-Functionalized Antimalarial Tambjamines and Prodiginines. J Med Chem 2024. [PMID: 39425665 DOI: 10.1021/acs.jmedchem.4c02093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Malaria has been a deadly enemy of mankind throughout history, affecting over 200 million people annually, along with approximately half a million deaths. Resistance to current therapies is of great concern, and there is a dire need for novel and well-tolerated antimalarials that operate by clinically unexploited mechanisms. We have previously reported that both tambjamines and prodiginines are highly potent novel antiplasmodial agents, but they required rigor optimizations to enhance the oral efficacy, safety, and physicochemical properties. Here, we launched a comprehensive structure-activity relationship study for B-ring-functionalized tambjamines and prodiginines with 54 novel analogues systematically designed and synthesized. A number of compounds exhibited remarkable antiplasmodial activities against asexual erythrocytic Plasmodium parasites, with improved safety and metabolic profiles. Notably, several prodiginines cured erythrocytic Plasmodium yoelii infections after oral 25 mg/kg × 4 days in a murine model and provided partial protection against liver stage Plasmodium berghei sporozoite-induced infection in mice.
Collapse
Affiliation(s)
- Amrendra Kumar
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Sivanna Chithanna
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Yuexin Li
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Xiaowei Zhang
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Rozalia A Dodean
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Diana Caridha
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Michael S Madejczyk
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Patricia J Lee
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Xiannu Jin
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Ravi Chetree
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Cameron Blount
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - William E Dennis
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Jesse DeLuca
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Chau Vuong
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Kristina Pannone
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Hieu T Dinh
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Susan Leed
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Alison Roth
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Kevin A Reynolds
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Jane X Kelly
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Papireddy Kancharla
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| |
Collapse
|
3
|
Hassan AMIA, Zhao Y, Chen X, He C. Blockage of Autophagy for Cancer Therapy: A Comprehensive Review. Int J Mol Sci 2024; 25:7459. [PMID: 39000565 PMCID: PMC11242824 DOI: 10.3390/ijms25137459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The incidence and mortality of cancer are increasing, making it a leading cause of death worldwide. Conventional treatments such as surgery, radiotherapy, and chemotherapy face significant limitations due to therapeutic resistance. Autophagy, a cellular self-degradation mechanism, plays a crucial role in cancer development, drug resistance, and treatment. This review investigates the potential of autophagy inhibition as a therapeutic strategy for cancer. A systematic search was conducted on Embase, PubMed, and Google Scholar databases from 1967 to 2024 to identify studies on autophagy inhibitors and their mechanisms in cancer therapy. The review includes original articles utilizing in vitro and in vivo experimental methods, literature reviews, and clinical trials. Key terms used were "Autophagy", "Inhibitors", "Molecular mechanism", "Cancer therapy", and "Clinical trials". Autophagy inhibitors such as chloroquine (CQ) and hydroxychloroquine (HCQ) have shown promise in preclinical studies by inhibiting lysosomal acidification and preventing autophagosome degradation. Other inhibitors like wortmannin and SAR405 target specific components of the autophagy pathway. Combining these inhibitors with chemotherapy has demonstrated enhanced efficacy, making cancer cells more susceptible to cytotoxic agents. Clinical trials involving CQ and HCQ have shown encouraging results, although further investigation is needed to optimize their use in cancer therapy. Autophagy exhibits a dual role in cancer, functioning as both a survival mechanism and a cell death pathway. Targeting autophagy presents a viable strategy for cancer therapy, particularly when integrated with existing treatments. However, the complexity of autophagy regulation and the potential side effects necessitate further research to develop precise and context-specific therapeutic approaches.
Collapse
Affiliation(s)
| | - Yuxin Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China (X.C.)
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China (X.C.)
- Department of Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China (X.C.)
- Department of Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| |
Collapse
|
4
|
Zhi HT, Lu Z, Chen L, Wu JQ, Li L, Hu J, Chen WH. Anticancer efficacy triggered by synergistically modulating the homeostasis of anions and iron: Design, synthesis and biological evaluation of dual-functional squaramide-hydroxamic acid conjugates. Bioorg Chem 2024; 147:107421. [PMID: 38714118 DOI: 10.1016/j.bioorg.2024.107421] [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: 03/09/2024] [Revised: 04/12/2024] [Accepted: 04/29/2024] [Indexed: 05/09/2024]
Abstract
Targeting the homeostasis of anions and iron has emerged as a promising therapeutic approach for the treatment of cancers. However, single-targeted agents often fall short of achieving optimal treatment efficacy. Herein we designed and synthesized a series of novel dual-functional squaramide-hydroxamic acid conjugates that are capable of synergistically modulating the homeostasis of anions and iron. Among them, compound 16 exhibited the most potent antiproliferative activity against a panel of selected cancer cell lines, and strong in vivo anti-tumor efficacy. This compound effectively elevated lysosomal pH through anion transport, and reduced the levels of intracellular iron. Compound 16 could disturb autophagy in A549 cells and trigger robust apoptosis. This compound caused cell cycle arrest at the G1/S phase, altered the mitochondrial function and elevated ROS levels. The present findings clearly demonstrated that synergistic modulation of anion and iron homeostasis has high potentials in the development of promising chemotherapeutic agents with dual action against cancers.
Collapse
Affiliation(s)
- Hai-Tao Zhi
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Zhonghui Lu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Li Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Jia-Qiang Wu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Lanqing Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Jinhui Hu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China.
| | - Wen-Hua Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China.
| |
Collapse
|
5
|
Kancharla P, Ortiz D, Fargo CM, Zhang X, Li Y, Sanchez M, Kumar A, Yeluguri M, Dodean RA, Caridha D, Madejczyk MS, Martin M, Jin X, Blount C, Chetree R, Pannone K, Dinh HT, DeLuca J, Evans M, Nadeau R, Vuong C, Leed S, Dennis WE, Roncal N, Pybus BS, Lee PJ, Roth A, Reynolds KA, Kelly JX, Landfear SM. Discovery and Optimization of Tambjamines as a Novel Class of Antileishmanial Agents. J Med Chem 2024; 67:8323-8345. [PMID: 38722757 PMCID: PMC11163866 DOI: 10.1021/acs.jmedchem.4c00517] [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] [Indexed: 05/24/2024]
Abstract
Leishmaniasis is a neglected tropical disease that is estimated to afflict over 12 million people. Current drugs for leishmaniasis suffer from serious deficiencies, including toxicity, high cost, modest efficacy, primarily parenteral delivery, and emergence of widespread resistance. We have discovered and developed a natural product-inspired tambjamine chemotype, known to be effective against Plasmodium spp, as a novel class of antileishmanial agents. Herein, we report in vitro and in vivo antileishmanial activities, detailed structure-activity relationships, and metabolic/pharmacokinetic profiles of a large library of tambjamines. A number of tambjamines exhibited excellent potency against both Leishmania mexicana and Leishmania donovani parasites with good safety and metabolic profiles. Notably, tambjamine 110 offered excellent potency and provided partial protection to leishmania-infected mice at 40 and/or 60 mg/kg/10 days of oral treatment. This study presents the first account of antileishmanial activity in the tambjamine family and paves the way for the generation of new oral antileishmanial drugs.
Collapse
Affiliation(s)
- Papireddy Kancharla
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Diana Ortiz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, 97239, United States
| | - Corinne M. Fargo
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, 97239, United States
| | - Xiaowei Zhang
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Yuexin Li
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Marco Sanchez
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, 97239, United States
| | - Amrendra Kumar
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Monish Yeluguri
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Rozalia A. Dodean
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Diana Caridha
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Michael S. Madejczyk
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Monica Martin
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Xiannu Jin
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Cameron Blount
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Ravi Chetree
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Kristina Pannone
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Hieu T. Dinh
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Jesse DeLuca
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Martin Evans
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Robert Nadeau
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Chau Vuong
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Susan Leed
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - William E. Dennis
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Norma Roncal
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Brandon S. Pybus
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Patricia J. Lee
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Alison Roth
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Kevin A. Reynolds
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Jane X. Kelly
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Scott M. Landfear
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, 97239, United States
| |
Collapse
|
6
|
Zheng Z, Wei X, Lin Y, Tao S, Li H, Ji Z, Wei H, Jin J, Zhao F, Lang C, Liu J, Chen J. In vivo therapy of osteosarcoma using anion transporters-based supramolecular drugs. J Nanobiotechnology 2024; 22:29. [PMID: 38216937 PMCID: PMC10787436 DOI: 10.1186/s12951-023-02270-x] [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: 09/27/2023] [Accepted: 12/14/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Osteosarcoma represents a serious clinical challenge due to its widespread genomic alterations, tendency for drug resistance and distant metastasis. New treatment methods are urgently needed to address those treatment difficulties in osteosarcoma to improve patient prognoses. In recent years, small-molecule based anion transporter have emerged as innovative and promising therapeutic compound with various biomedical applications. However, due to a lack of efficient delivery methods, using ion transporters as therapeutic drugs in vivo remains a major challenge. RESULT Herein, we developed self-assembled supramolecular drugs based on small-molecule anion transporters, which exhibited potent therapeutic effect towards osteosarcoma both in vitro and in vivo. The anion transporters can disrupt intracellular ion homeostasis, inhibit proliferation, migration, epithelial-mesenchymal transition process, and lead to osteosarcoma cell death. RNA sequencing, western blot and flow cytometry indicated reprogramming of HOS cells and induced cell death through multiple pathways. These pathways included activation of endoplasmic reticulum stress, autophagy, apoptosis and cell cycle arrest, which avoided the development of drug resistance in osteosarcoma cells. Functionalized with osteosarcoma targeting peptide, the assembled supramolecular drug showed excellent targeted anticancer therapy against subcutaneous xenograft tumor and lung metastasis models. Besides good tumor targeting capability and anti-drug resistance, the efficacy of the assembly was also attributed to its ability to regulate the tumor immune microenvironment in vivo. CONCLUSIONS In summary, we have demonstrated for the first time that small-molecule anion transporters are capable of killing osteosarcoma cells through multiple pathways. The assemblies, OTP-BP-L, show excellent targeting and therapeutic effect towards osteosarcoma tumors. Furthermore, the supramolecular drug shows a strong ability to regulate the tumor immune microenvironment in vivo. This work not only demonstrated the biomedical value of small-molecule anion transporters in vivo, but also provided an innovative approach for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Zeyu Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiaoan Wei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Yangyang Lin
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Siyue Tao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hui Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhongyin Ji
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hongxin Wei
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Jiayan Jin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Chao Lang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China.
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China.
| | - Junhui Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Jian Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
7
|
Alonso-Carrillo D, Carreira-Barral I, Mielczarek M, Sancho-Medina A, Herran E, Vairo C, Del Pozo A, Luzuriaga I, Lazcanoiturburu N, Ibarrola O, Ponce S, Villar-Vidal M, García-Valverde M, Quesada R. Formulation and evaluation of anion transporters in nanostructured lipid carriers. Org Biomol Chem 2023; 21:7753-7757. [PMID: 37691616 DOI: 10.1039/d3ob01182h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Six novel click-tambjamines (1-6) bearing an alkyl chain of varying length linked to the imine moiety have been formulated in nanostructured lipid carriers (NLCs) to evaluate their transmembrane anion transport activity both when free (i.e., not encapsulated) and nanoformulated. Nanostructured lipid carriers (NLCs) are an example of drug delivery systems (DDSs) that stand out because of their versatility. In this work we show that NLCs can be used to efficiently formulate highly lipophilic anionophores and experiments conducted in model liposomes reveal that these formulations are adequate to deliver anionophores without compromising their transport activity. This result paves the way to facilitate the study of highly lipophilic anionophores and their potential use as future drugs.
Collapse
Affiliation(s)
| | | | - Marcin Mielczarek
- Departamento de Química, Universidad de Burgos, Burgos 09001, Spain.
| | | | - Enara Herran
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | - Claudia Vairo
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | - Angel Del Pozo
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | - Iris Luzuriaga
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | | | - Oihane Ibarrola
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | - Sara Ponce
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | - María Villar-Vidal
- Biokeralty Research Institute AIE, Hermanos Lumière 5, 01510 Miñano, Spain
| | | | - Roberto Quesada
- Departamento de Química, Universidad de Burgos, Burgos 09001, Spain.
| |
Collapse
|
8
|
Mondal A, Ahmad M, Mondal D, Talukdar P. Progress and prospects toward supramolecular bioactive ion transporters. Chem Commun (Camb) 2023; 59:1917-1938. [PMID: 36691926 DOI: 10.1039/d2cc06761g] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The majority of cellular physiological processes depend on natural ion channels, which are pore-forming membrane-embedded proteins that let ions flow across the cell membranes selectively. This selective movement of ions across the membranes balances the osmolality within and outside the cell. However, mutations in the genes that encode essential membrane transport proteins or structural reorganisation of these proteins can cause life-threatening diseases like cystic fibrosis. Artificial ion transport systems have opened up a way to replace dysfunctional natural ion channels to cure such diseases through channel replacement therapy. Moreover, recent research has also demonstrated the ability of these systems to kill cancer cells, reigniting interest in the field among scientists. Our contributions to the recent progress in the design and development of artificial chloride ion transporters and their effect on biological systems have been discussed in this review. This review would provide current vistas and future directions toward the development of novel ion transporters with improved biocompatibility and desired anti-cancer properties. Additionally, it strongly emphasises stimuli-responsive ion transport systems, which are crucial for obtaining target-specificity and may speed up the application of these systems in clinical therapeutics.
Collapse
Affiliation(s)
- Abhishek Mondal
- Chemistry Department, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| | - Manzoor Ahmad
- Chemistry Department, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India. .,Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Debashis Mondal
- Chemistry Department, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India. .,Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Zwirkii Wigury 101, Warsaw 02-089, Poland
| | - Pinaki Talukdar
- Chemistry Department, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.
| |
Collapse
|
9
|
S. M. S, Naveen NR, Rao GSNK, Gopan G, Chopra H, Park MN, Alshahrani MM, Jose J, Emran TB, Kim B. A spotlight on alkaloid nanoformulations for the treatment of lung cancer. Front Oncol 2022; 12:994155. [PMID: 36330493 PMCID: PMC9623325 DOI: 10.3389/fonc.2022.994155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/12/2022] [Indexed: 07/30/2023] Open
Abstract
Numerous naturally available phytochemicals have potential anti-cancer activities due to their vast structural diversity. Alkaloids have been extensively used in cancer treatment, especially lung cancers, among the plant-based compounds. However, their utilization is limited by their poor solubility, low bioavailability, and inadequacies such as lack of specificity to cancer cells and indiscriminate distribution in the tissues. Incorporating the alkaloids into nanoformulations can overcome the said limitations paving the way for effective delivery of the alkaloids to the site of action in sufficient concentrations, which is crucial in tumor targeting. Our review attempts to assess whether alkaloid nanoformulation can be an effective tool in lung cancer therapy. The mechanism of action of each alkaloid having potential is explored in great detail in the review. In general, Alkaloids suppress oncogenesis by modulating several signaling pathways involved in multiplication, cell cycle, and metastasis, making them significant component of many clinical anti-cancerous agents. The review also explores the future prospects of alkaloid nanoformulation in lung cancer. So, in conclusion, alkaloid based nanoformulation will emerge as a potential gamechanger in treating lung cancer in the near future.
Collapse
Affiliation(s)
- Sindhoor S. M.
- Department of Pharmaceutics, P.A. College of Pharmacy, Mangalore, Karnataka, India
| | - N. Raghavendra Naveen
- Department of Pharmaceutics, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G. Nagar, Karnataka, India
| | - GSN Koteswara Rao
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Gopika Gopan
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Moon Nyeo Park
- Department of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Mohammed Merae Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Bonglee Kim
- Department of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
10
|
Zhang T, Hong XQ, Zhi HT, Hu J, Chen WH. Synthesis and mechanism of biological action of morpholinyl-bearing arylsquaramides as small-molecule lysosomal pH modulators. RSC Adv 2022; 12:22748-22759. [PMID: 36105976 PMCID: PMC9376937 DOI: 10.1039/d2ra02146c] [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: 04/03/2022] [Accepted: 08/03/2022] [Indexed: 11/21/2022] Open
Abstract
Lysosomal pH is an important modulator for many cellular processes. An agent that is capable of regulating lysosomal pH may find a wide range of potential applications in the field of biomedicine. In this study, we describe the synthesis of a family of morpholinyl-bearing arylsquaramides as small-molecule lysosomal pH modulators. These compounds are able to efficiently facilitate the transmembrane transport of chloride anions as mobile carriers across vesicular and cellular phospholipid membranes. They are capable of specifically alkalizing liposomes, disrupting the homeostasis of lysosomal pH and inactivivating lysosomal Cathepsin B enzyme. Anion transport is considered as the probable mechanism of action for the high efficiency of these compounds to modulate lysosomal pH. The present findings present a novel means to efficiently regulate lysosomal pH, which is in contrast to the methods shown by conventional lysosomal pH modulators that generally function by either acting as a weak base/acid, or releasing a basic/acidic component in lysosomal environments to change lysosomal pH.
Collapse
Affiliation(s)
- Tao Zhang
- School of Biotechnology and Health Sciences, Wuyi University Jiangmen 529020 Guangdong P. R. China
| | - Xiao-Qiao Hong
- School of Pharmaceutical Sciences, Tsinghua University Haidian Dist Beijing 100084 P. R. China
| | - Hai-Tao Zhi
- School of Biotechnology and Health Sciences, Wuyi University Jiangmen 529020 Guangdong P. R. China
| | - Jinhui Hu
- School of Biotechnology and Health Sciences, Wuyi University Jiangmen 529020 Guangdong P. R. China
| | - Wen-Hua Chen
- School of Biotechnology and Health Sciences, Wuyi University Jiangmen 529020 Guangdong P. R. China
| |
Collapse
|
11
|
Díaz-Cabrera S, Carreira-Barral I, García-Valverde M, Quesada R. Roseophilin-inspired derivatives as transmembrane anion carriers. Supramol Chem 2022. [DOI: 10.1080/10610278.2022.2099277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
| | | | | | - Roberto Quesada
- Departamento de Química, Universidad de Burgos, Burgos, Spain
| |
Collapse
|
12
|
A Novel Late-Stage Autophagy Inhibitor That Efficiently Targets Lysosomes Inducing Potent Cytotoxic and Sensitizing Effects in Lung Cancer. Cancers (Basel) 2022; 14:cancers14143387. [PMID: 35884450 PMCID: PMC9324127 DOI: 10.3390/cancers14143387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/28/2022] [Accepted: 07/07/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Lung cancer is the main cause of cancer-related deaths worldwide, mainly due to treatment resistance. For that reason, it is necessary to develop novel therapeutic strategies to overcome this phenomenon. The aim of our study was to design and characterize a synthetic anionophore, LAI-1, that would be able to efficiently disrupt lysosomal activity, leading to autophagy blockage, one of the most important resistance mechanisms in cancer cells. We confirmed that LAI-1 selectively localized in lysosomes, deacidifying them. This effect produced a blockage of autophagy, characterized by an abrogation of autophagosomes and lysosomes fusion. Moreover, LAI-1 produced cell death in lung cancer cells from different histological subtypes, inducing cytotoxicity more efficiently than other known autophagy inhibitors. Finally, LAI-1 was evaluated in combination therapy, showing sensitization to the first-line chemotherapeutic agent cisplatin. Altogether, LAI-1 is a novel late-stage autophagy inhibitor with potential therapeutic applications in tumors with cytoprotective autophagy. Abstract Overcoming resistance is one of the most challenging features in current anticancer therapy. Autophagy is a cellular process that confers resistance in some advanced tumors, since it enables cancer cells to adapt to stressful situations, such as anticancer treatments. Hence, the inhibition of this cytoprotective autophagy leads to tumor cells sensitization and death. In this regard, we designed a novel potent anionophore compound that specifically targets lysosomes, called LAI-1 (late-stage autophagy inhibitor-1), and evaluated its role in blocking autophagy and its potential anticancer effects in three lung cancer cell lines from different histological subtypes. Compared to other autophagy inhibitors, such as chloroquine and 3-Methyladenine, the LAI-1 treatment induced more potent anticancer effects in all tested cancer cells. LAI-1 was able to efficiently target and deacidify lysosomes, while acidifying cytoplasmic pH. Consequently, LAI-1 efficiently blocked autophagy, indicated by the increased LC3-II/I ratio and p62/SQSTM1 levels. Moreover, no colocalization was observed between autophagosomes, marked with LC3 or p62/SQSTM1, and lysosomes, stained with LAMP-1, after the LAI-1 treatment, indicating the blockage of autophagolysosome formation. Furthermore, LAI-1 induced cell death by activating apoptosis (enhancing the cleavage of caspase-3 and PARP) or necrosis, depending on the cancer cell line. Finally, LAI-1 sensitized cancer cells to the first-line chemotherapeutic agent cisplatin. Altogether, LAI-1 is a new late-stage autophagy inhibitor that causes lysosomal dysfunction and the blockage of autophagolysosome formation, as well as potently induces cancer cell death and sensitization to conventional treatments at lower concentrations than other known autophagy inhibitors, appearing as a potential new therapeutic approach to overcome cancer resistance.
Collapse
|
13
|
Picci G, Marchesan S, Caltagirone C. Ion Channels and Transporters as Therapeutic Agents: From Biomolecules to Supramolecular Medicinal Chemistry. Biomedicines 2022; 10:biomedicines10040885. [PMID: 35453638 PMCID: PMC9032600 DOI: 10.3390/biomedicines10040885] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/07/2022] [Accepted: 04/09/2022] [Indexed: 12/13/2022] Open
Abstract
Ion channels and transporters typically consist of biomolecules that play key roles in a large variety of physiological and pathological processes. Traditional therapies include many ion-channel blockers, and some activators, although the exact biochemical pathways and mechanisms that regulate ion homeostasis are yet to be fully elucidated. An emerging area of research with great innovative potential in biomedicine pertains the design and development of synthetic ion channels and transporters, which may provide unexplored therapeutic opportunities. However, most studies in this challenging and multidisciplinary area are still at a fundamental level. In this review, we discuss the progress that has been made over the last five years on ion channels and transporters, touching upon biomolecules and synthetic supramolecules that are relevant to biological use. We conclude with the identification of therapeutic opportunities for future exploration.
Collapse
Affiliation(s)
- Giacomo Picci
- Chemical and Geological Sciences Department, University of Cagliari, 09042 Cagliari, Italy;
| | - Silvia Marchesan
- Chemical and Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
- Correspondence: (S.M.); (C.C.)
| | - Claudia Caltagirone
- Chemical and Geological Sciences Department, University of Cagliari, 09042 Cagliari, Italy;
- Correspondence: (S.M.); (C.C.)
| |
Collapse
|
14
|
Wezenberg SJ, Chen LJ, Bos JE, Feringa BL, Howe ENW, Wu X, Siegler MA, Gale PA. Photomodulation of Transmembrane Transport and Potential by Stiff-Stilbene Based Bis(thio)ureas. J Am Chem Soc 2022; 144:331-338. [PMID: 34932344 PMCID: PMC8759083 DOI: 10.1021/jacs.1c10034] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Indexed: 12/14/2022]
Abstract
Membrane transport proteins fulfill important regulatory functions in biology with a common trait being their ability to respond to stimuli in the environment. Various small-molecule receptors, capable of mediating transmembrane transport, have been successfully developed. However, to confer stimuli-responsiveness on them poses a fundamental challenge. Here we demonstrate photocontrol of transmembrane transport and electric potential using bis(thio)ureas derived from stiff-stilbene. UV-vis and 1H NMR spectroscopy are used to monitor E-Z photoisomerization of these bis(thio)ureas and 1H NMR titrations reveal stronger binding of chloride to the (Z)-form than to the (E)-form. Additional insight into the binding properties is provided by single crystal X-ray crystallographic analysis and DFT geometry optimization. Importantly, the (Z)-isomers are much more active in transmembrane transport than the respective (E)-isomers as shown through various assays. As a result, both membrane transport and depolarization can be modulated upon irradiation, opening up new prospects toward light-based therapeutics as well as physiological and optopharmacological tools for studying anion transport-associated diseases and to stimulate neuronal activity, respectively.
Collapse
Affiliation(s)
- Sander J. Wezenberg
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
- Stratingh
Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747 AG, Groningen, The Netherlands
| | - Li-Jun Chen
- School
of Chemistry, The University of Sydney, Sydney NSW 2006, Australia
| | - Jasper E. Bos
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Ben L. Feringa
- Stratingh
Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747 AG, Groningen, The Netherlands
| | - Ethan N. W. Howe
- School
of Chemistry, The University of Sydney, Sydney NSW 2006, Australia
| | - Xin Wu
- School
of Chemistry, The University of Sydney, Sydney NSW 2006, Australia
| | - Maxime A. Siegler
- Department
of Chemistry, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Philip A. Gale
- School
of Chemistry, The University of Sydney, Sydney NSW 2006, Australia
- The
University of Sydney Nano Institute (SydneyNano), The University of
Sydney, Sydney NSW 2006, Australia
| |
Collapse
|
15
|
Yang J, Yu G, Sessler JL, Shin I, Gale PA, Huang F. Artificial transmembrane ion transporters as potential therapeutics. Chem 2021. [DOI: 10.1016/j.chempr.2021.10.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Abstract
Several life-threatening diseases, also known as 'Channelopathies' are linked to irregularities in ion transport proteins. Significant research efforts have fostered the development of artificial transport systems that facilitates to restore the functions of impaired natural transport proteins. Indeed, a few of these artificial ionophores demonstrate the rare combination of transmembrane ion transport and important biological activity, offering early promises of suitability in 'channel replacement therapy'. In this review, structural facets and functions of both cationophores and anionophores are discussed. Ionophores that are toxic to various bacteria and yeast, could be exploited as antimicrobial agent. Nevertheless, few non-toxic ionophores offer the likelihood of treating a wide range of genetic diseases caused by the gene mutations. In addition, their ability to disrupt cellular homeostasis and to alter lysosomal pH endow ionophores as promising candidates for cancer treatment. Overall, critically outlining the advances in artificial ionophores in terms of in vitro ion transport, possible modes of action and biological activities enables us to propose possible future roadmaps in this research area.
Collapse
Affiliation(s)
- Arundhati Roy
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, 81377, Munich, Germany
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| |
Collapse
|
17
|
Chen H, Liu Y, Cheng X, Fang S, Sun Y, Yang Z, Zheng W, Ji X, Wu Z. Self‐Assembly of Size‐Controlled
m
‐Pyridine–Urea Oligomers and Their Biomimetic Chloride Ion Channels. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Hualong Chen
- Beijing Institute of Brain Disorders Laboratory of Brain Disorders Ministry of Science and Technology Collaborative Innovation Center for Brain Disorders Beijing Advanced Innovation Center for Big Data-based Precision Medicine Capital Medical University Beijing 100069 China
| | - Yajing Liu
- School of Pharmaceutical Science Capital Medical University Beijing 100069 China
| | - Xuebo Cheng
- Beijing Institute of Brain Disorders Laboratory of Brain Disorders Ministry of Science and Technology Collaborative Innovation Center for Brain Disorders Beijing Advanced Innovation Center for Big Data-based Precision Medicine Capital Medical University Beijing 100069 China
| | - Senbiao Fang
- School of Computer Science and Engineering Central South University Changsha 410012 China
| | - Yuli Sun
- Beijing Institute of Brain Disorders Laboratory of Brain Disorders Ministry of Science and Technology Collaborative Innovation Center for Brain Disorders Beijing Advanced Innovation Center for Big Data-based Precision Medicine Capital Medical University Beijing 100069 China
| | - Zequn Yang
- Beijing Institute of Brain Disorders Laboratory of Brain Disorders Ministry of Science and Technology Collaborative Innovation Center for Brain Disorders Beijing Advanced Innovation Center for Big Data-based Precision Medicine Capital Medical University Beijing 100069 China
| | - Wei Zheng
- Beijing Institute of Brain Disorders Laboratory of Brain Disorders Ministry of Science and Technology Collaborative Innovation Center for Brain Disorders Beijing Advanced Innovation Center for Big Data-based Precision Medicine Capital Medical University Beijing 100069 China
| | - Xunming Ji
- Beijing Institute of Brain Disorders Laboratory of Brain Disorders Ministry of Science and Technology Collaborative Innovation Center for Brain Disorders Beijing Advanced Innovation Center for Big Data-based Precision Medicine Capital Medical University Beijing 100069 China
- Institute of Hypoxia Medicine Xuanwu Hospital Capital Medical University Beijing 100053 China
| | - Zehui Wu
- Beijing Institute of Brain Disorders Laboratory of Brain Disorders Ministry of Science and Technology Collaborative Innovation Center for Brain Disorders Beijing Advanced Innovation Center for Big Data-based Precision Medicine Capital Medical University Beijing 100069 China
| |
Collapse
|
18
|
Chen H, Liu Y, Cheng X, Fang S, Sun Y, Yang Z, Zheng W, Ji X, Wu Z. Self-Assembly of Size-Controlled m-Pyridine-Urea Oligomers and Their Biomimetic Chloride Ion Channels. Angew Chem Int Ed Engl 2021; 60:10833-10841. [PMID: 33624345 DOI: 10.1002/anie.202102174] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Indexed: 01/06/2023]
Abstract
The m-pyridine urea (mPU) oligomer was constructed by using the intramolecular hydrogen bond formed by the pyridine nitrogen atom and the NH of urea and the intermolecular hydrogen bond of the terminal carbonyl group and the NH of urea. Due to the synergistic effect of hydrogen bonds, mPU oligomer folds and exhibits strong self-assembly behaviour. Affected by folding, mPU oligomer generates a twisted plane, and one of its important features is that the carbonyl group of the urea group orientates outwards from the twisted plane, while the NHs tend to direct inward. This feature is beneficial to NH attraction for electron-rich species. Among them, the trimer self-assembles into helical nanotubes, and can efficiently transport chloride ions. This study provides a novel and efficient strategy for constructing self-assembled biomimetic materials for electron-rich species transmission.
Collapse
Affiliation(s)
- Hualong Chen
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Yajing Liu
- School of Pharmaceutical Science, Capital Medical University, Beijing, 100069, China
| | - Xuebo Cheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Senbiao Fang
- School of Computer Science and Engineering, Central South University, Changsha, 410012, China
| | - Yuli Sun
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Zequn Yang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Wei Zheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, 100069, China.,Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Zehui Wu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
19
|
Barros-Nepomuceno FWA, de Araújo Viana D, Pinheiro DP, de Cássia Evangelista de Oliveira F, Magalhães Ferreira J, R de Queiroz MG, Ma X, Cavalcanti BC, Pessoa C, Banwell MG. The Effects of the Alkaloid Tambjamine J on Mice Implanted with Sarcoma 180 Tumor Cells. ChemMedChem 2020; 16:420-428. [PMID: 32886437 DOI: 10.1002/cmdc.202000387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/18/2020] [Indexed: 12/12/2022]
Abstract
The tambjamines are a small group of bipyrrolic alkaloids that, collectively, display a significant range of biological activities including antitumor, antimicrobial and immunosuppressive properties. The key objective of the present study was to undertake preclinical assessments of tambjamine J (T-J) so as to determine its in vivo antitumor effects. To that end, sarcoma 180 cells were transplanted in mice and the impacts of the title compound then evaluated using a range of protocols including hematological, biochemical, histopathological, genotoxic and clastogenic assays. As a result it was established that this alkaloid has a significant therapeutic window and effectively reduces tumor growth (by 40 % and 79 % at doses of 10 and 20 mg/kg/day, respectively). In this regard it displays similar antitumor activity to the anticancer agent cyclophosphamide and alters animal weight in an analogous manner.
Collapse
Affiliation(s)
- Francisco Washington A Barros-Nepomuceno
- Institute of Health Sciences, University for International Integration of the Afro-Brazilian Lusophony, Acarape, 62.785-000, CE, Brazil.,Center for Research and Drug Development, Federal University of Ceará, Fortaleza, 60.430.275, CE, Brazil
| | - Daniel de Araújo Viana
- PATHOVET Laboratory, Pathological Anatomy and Veterinary Clinic, Fortaleza, 60.020.001, CE, Brazil
| | - Daniel Pascoalino Pinheiro
- Center for Research and Drug Development, Federal University of Ceará, Fortaleza, 60.430.275, CE, Brazil
| | | | - Jamile Magalhães Ferreira
- Institute of Health Sciences, University for International Integration of the Afro-Brazilian Lusophony, Acarape, 62.785-000, CE, Brazil.,Clinical and Toxicological Analysis Department, Faculty of Pharmacy, Odontology and Nursing, Federal University of Ceará, Fortaleza, 60.714.903, CE, Brazil
| | - Maria Goretti R de Queiroz
- Clinical and Toxicological Analysis Department, Faculty of Pharmacy, Odontology and Nursing, Federal University of Ceará, Fortaleza, 60.714.903, CE, Brazil
| | - Xinghua Ma
- Research School of Chemistry, Institute of Advanced Studies, The Australian National University, Canberra, ACT, 2601, Australia
| | - Bruno Coêlho Cavalcanti
- Center for Research and Drug Development, Federal University of Ceará, Fortaleza, 60.430.275, CE, Brazil
| | - Claudia Pessoa
- Center for Research and Drug Development, Federal University of Ceará, Fortaleza, 60.430.275, CE, Brazil
| | - Martin G Banwell
- Research School of Chemistry, Institute of Advanced Studies, The Australian National University, Canberra, ACT, 2601, Australia.,Institute for Advanced and Applied Chemical Synthesis, Jinan University, Zhuhai, 519070, Guangdong, China
| |
Collapse
|
20
|
Fares M, Wu X, Ramesh D, Lewis W, Keller PA, Howe ENW, Pérez‐Tomás R, Gale PA. Stimuli‐Responsive Cycloaurated “OFF‐ON” Switchable Anion Transporters. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202006392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Mohamed Fares
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
- School of Chemistry & Molecular Bioscience, Molecular Horizons University of Wollongong Illawarra Health & Medical Research Institute Wollongong NSW 2522 Australia
| | - Xin Wu
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| | - Deepthi Ramesh
- Faculty of Medicine & Health Sciences Department of Pathology and Experimental Therapeutics Cancer Cell Biology Research Group University of Barcelona Barcelona Spain
| | - William Lewis
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| | - Paul A. Keller
- School of Chemistry & Molecular Bioscience, Molecular Horizons University of Wollongong Illawarra Health & Medical Research Institute Wollongong NSW 2522 Australia
| | - Ethan N. W. Howe
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
- GlaxoSmithKline GSK Jurong 1 Pioneer Sector 1 Singapore 628413 Singapore
| | - Ricardo Pérez‐Tomás
- Faculty of Medicine & Health Sciences Department of Pathology and Experimental Therapeutics Cancer Cell Biology Research Group University of Barcelona Barcelona Spain
| | - Philip A. Gale
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| |
Collapse
|
21
|
Fares M, Wu X, Ramesh D, Lewis W, Keller PA, Howe ENW, Pérez-Tomás R, Gale PA. Stimuli-Responsive Cycloaurated "OFF-ON" Switchable Anion Transporters. Angew Chem Int Ed Engl 2020; 59:17614-17621. [PMID: 32583552 DOI: 10.1002/anie.202006392] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Indexed: 01/28/2023]
Abstract
Anion transporters have shown potential application as anti-cancer agents that function by disrupting homeostasis and triggering cell death. In this research article we report switchable anion transport by gold complexes of anion transporters that are "switched on" in situ in the presence of the reducing agent GSH by decomplexation of gold. GSH is found in higher concentrations in tumors than in healthy tissue and hence this approach offers a strategy to target these systems to tumors.
Collapse
Affiliation(s)
- Mohamed Fares
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.,School of Chemistry & Molecular Bioscience, Molecular Horizons, University of Wollongong, Illawarra Health & Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - Xin Wu
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Deepthi Ramesh
- Faculty of Medicine & Health Sciences, Department of Pathology and Experimental Therapeutics, Cancer Cell Biology Research Group, University of Barcelona, Barcelona, Spain
| | - William Lewis
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Paul A Keller
- School of Chemistry & Molecular Bioscience, Molecular Horizons, University of Wollongong, Illawarra Health & Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - Ethan N W Howe
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.,GlaxoSmithKline, GSK Jurong, 1 Pioneer Sector 1, Singapore, 628413, Singapore
| | - Ricardo Pérez-Tomás
- Faculty of Medicine & Health Sciences, Department of Pathology and Experimental Therapeutics, Cancer Cell Biology Research Group, University of Barcelona, Barcelona, Spain
| | - Philip A Gale
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
22
|
Hong XQ, He XY, Tam KY, Chen WH. Synthesis and biological effect of lysosome-targeting fluorescent anion transporters with enhanced anionophoric activity. Bioorg Med Chem Lett 2020; 30:127461. [PMID: 32755679 DOI: 10.1016/j.bmcl.2020.127461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 11/27/2022]
Abstract
Two lysosome-targeting fluorescent anion transporters derived from coumarins, trifluoromethylated arylsquaramides and morpholines were synthesized, and their specificity and efficiency to target and alkalize lysosomes were investigated. They are able to target lysosomes specifically. Compared with the previous analogue without trifluoromethyl substituents, these two conjugates, in particular the one having a 3,5-bis(trifluoromethyl) substituent, exhibit significantly higher ability to facilitate the transport of chloride anions, alkalize lysosomes and reduce the activity of lysosomal Cathepsin B enzyme. The present finding suggests that improving the anionophoric activity of lysosome-targeting fluorescent anion transporters is favorable to the efficiency to alkalize lysosomes and deactivate lysosomal Cathepsin B enzyme.
Collapse
Affiliation(s)
- Xiao-Qiao Hong
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Xiang-Yu He
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Kin Yip Tam
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, PR China
| | - Wen-Hua Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; School of Biotechnology and Health Sciences, International Healthcare Innovation Institute (Jiangmen), Wuyi University, Jiangmen 529020, PR China.
| |
Collapse
|
23
|
Davis JT, Gale PA, Quesada R. Advances in anion transport and supramolecular medicinal chemistry. Chem Soc Rev 2020; 49:6056-6086. [PMID: 32692794 DOI: 10.1039/c9cs00662a] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Advances in anion transport by synthetic supramolecular systems are discussed in this article. Developments in the design of discrete molecular carriers for anions and supramolecular anion channels are reviewed followed by an overview of the use of these systems in biological systems as putative treatments for diseases such as cystic fibrosis and cancer.
Collapse
Affiliation(s)
- Jeffery T Davis
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA.
| | - Philip A Gale
- School of Chemistry (F11), The University of Sydney, NSW 2006, Australia.
| | - Roberto Quesada
- Departmento de Química, Universidad de Burgos, 09001 Burgos, Spain.
| |
Collapse
|
24
|
Bąk KM, van Kolck B, Maslowska-Jarzyna K, Papadopoulou P, Kros A, Chmielewski MJ. Oxyanion transport across lipid bilayers: direct measurements in large and giant unilamellar vesicles. Chem Commun (Camb) 2020; 56:4910-4913. [PMID: 32238998 DOI: 10.1039/c9cc09888g] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A simple di(thioamido)carbazole 1 serves as a potent multispecific transporter for various biologically relevant oxyanions, such as drugs, metabolites and model organic phosphate. The transport kinetics of a wide range of oxyanions can be easily quantified by a modified lucigenin assay in both large and giant unilamellar vesicles.
Collapse
Affiliation(s)
- Krzysztof M Bąk
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089 Warszawa, Poland.
| | - Bartjan van Kolck
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Krystyna Maslowska-Jarzyna
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089 Warszawa, Poland.
| | - Panagiota Papadopoulou
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Alexander Kros
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Michał J Chmielewski
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089 Warszawa, Poland.
| |
Collapse
|
25
|
|
26
|
Akhtar N, Biswas O, Manna D. Biological applications of synthetic anion transporters. Chem Commun (Camb) 2020; 56:14137-14153. [DOI: 10.1039/d0cc05489e] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Transmembrane transport of anions by small molecules has recently been used to reduce the viability of cancer cells and fight against antibiotic-resistant and clinically relevant bacterial strains.
Collapse
Affiliation(s)
- Nasim Akhtar
- Department of Chemistry
- Indian Institute of Technology Guwahati
- India
| | - Oindrila Biswas
- Department of Chemistry
- Indian Institute of Technology Guwahati
- India
| | - Debasis Manna
- Department of Chemistry
- Indian Institute of Technology Guwahati
- India
| |
Collapse
|
27
|
Development of a Library of Thiophene‐Based Drug‐Like Lego Molecules: Evaluation of Their Anion Binding, Transport Properties, and Cytotoxicity. Chemistry 2019; 26:888-899. [DOI: 10.1002/chem.201904255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Indexed: 12/15/2022]
|
28
|
Yu XH, Hong XQ, Mao QC, Chen WH. Biological effects and activity optimization of small-molecule, drug-like synthetic anion transporters. Eur J Med Chem 2019; 184:111782. [DOI: 10.1016/j.ejmech.2019.111782] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 12/27/2022]
|
29
|
Pérez-Hernández M, Arias A, Martínez-García D, Pérez-Tomás R, Quesada R, Soto-Cerrato V. Targeting Autophagy for Cancer Treatment and Tumor Chemosensitization. Cancers (Basel) 2019; 11:E1599. [PMID: 31635099 PMCID: PMC6826429 DOI: 10.3390/cancers11101599] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/14/2019] [Accepted: 10/16/2019] [Indexed: 12/15/2022] Open
Abstract
Autophagy is a tightly regulated catabolic process that facilitates nutrient recycling from damaged organelles and other cellular components through lysosomal degradation. Deregulation of this process has been associated with the development of several pathophysiological processes, such as cancer and neurodegenerative diseases. In cancer, autophagy has opposing roles, being either cytoprotective or cytotoxic. Thus, deciphering the role of autophagy in each tumor context is crucial. Moreover, autophagy has been shown to contribute to chemoresistance in some patients. In this regard, autophagy modulation has recently emerged as a promising therapeutic strategy for the treatment and chemosensitization of tumors, and has already demonstrated positive clinical results in patients. In this review, the dual role of autophagy during carcinogenesis is discussed and current therapeutic strategies aimed at targeting autophagy for the treatment of cancer, both under preclinical and clinical development, are presented. The use of autophagy modulators in combination therapies, in order to overcome drug resistance during cancer treatment, is also discussed as well as the potential challenges and limitations for the use of these novel therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Marta Pérez-Hernández
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08905 Barcelona, Spain.
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain.
| | - Alain Arias
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08905 Barcelona, Spain.
- Department of Integral Adult Dentistry, Research Centre for Dental Sciences (CICO), Universidad de La Frontera, Temuco 4811230, Chile.
- Research Group of Health Sciences, Faculty of Health Sciences, Universidad Adventista de Chile, Chillán 3780000, Chile.
| | - David Martínez-García
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08905 Barcelona, Spain.
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain.
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08905 Barcelona, Spain.
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain.
| | - Roberto Quesada
- Department of Chemistry, Universidad de Burgos, 09001 Burgos, Spain.
| | - Vanessa Soto-Cerrato
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08905 Barcelona, Spain.
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain.
| |
Collapse
|
30
|
Wu X, Small JR, Cataldo A, Withecombe AM, Turner P, Gale PA. Voltage‐Switchable HCl Transport Enabled by Lipid Headgroup–Transporter Interactions. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201907466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Xin Wu
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| | - Jennifer R. Small
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
- Chemistry University of Southampton Southampton SO17 1BJ UK
| | - Alessio Cataldo
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
- Department of Chemistry University of Copenhagen Universitetsparken 5 2100 Copenhagen Ø Denmark
| | - Anne M. Withecombe
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| | - Peter Turner
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| | - Philip A. Gale
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| |
Collapse
|
31
|
Wu X, Small JR, Cataldo A, Withecombe AM, Turner P, Gale PA. Voltage‐Switchable HCl Transport Enabled by Lipid Headgroup–Transporter Interactions. Angew Chem Int Ed Engl 2019; 58:15142-15147. [DOI: 10.1002/anie.201907466] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/20/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Xin Wu
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| | - Jennifer R. Small
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
- Chemistry University of Southampton Southampton SO17 1BJ UK
| | - Alessio Cataldo
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
- Department of Chemistry University of Copenhagen Universitetsparken 5 2100 Copenhagen Ø Denmark
| | - Anne M. Withecombe
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| | - Peter Turner
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| | - Philip A. Gale
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| |
Collapse
|
32
|
|
33
|
Sakai-Kawada FE, Ip CG, Hagiwara KA, Awaya JD. Biosynthesis and Bioactivity of Prodiginine Analogs in Marine Bacteria, Pseudoalteromonas: A Mini Review. Front Microbiol 2019; 10:1715. [PMID: 31396200 PMCID: PMC6667630 DOI: 10.3389/fmicb.2019.01715] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/11/2019] [Indexed: 11/28/2022] Open
Abstract
The Prodiginine family consists of primarily red-pigmented tripyrrole secondary metabolites that were first characterized in the Gram-negative bacterial species Serratia marcescens and demonstrates a wide array of biological activities and applications. Derivatives of prodiginine have since been characterized in the marine γ-proteobacterium, Pseudoalteromonas. Although biosynthetic gene clusters involved in prodiginine synthesis display homology among genera, there is an evident structural difference in the resulting metabolites. This review will summarize prodiginine biosynthesis, bioactivity, and gene regulation in Pseudoalteromonas in comparison to the previously characterized species of Serratia, discuss the ecological contributions of Pseudoalteromonas in the marine microbiome and their eukaryotic hosts, and consider the importance of modern functional genomics and classic DNA manipulation to understand the overall prodiginine biosynthesis pathway.
Collapse
Affiliation(s)
- Francis E. Sakai-Kawada
- Department of Molecular Biosciences and Bioengineering, University of Hawai´i at Mānoa, Honolulu, HI, United States
| | - Courtney G. Ip
- Department of Biology, University of Hawai´i at Hilo, Hilo, HI, United States
| | - Kehau A. Hagiwara
- Institute of Marine and Environmental Technology, University of Maryland, Baltimore, Baltimore, MD, United States
- Chemical Sciences Division, National Institute of Standards and Technology, Hollings Marine Laboratory, Charleston, SC, United States
| | - Jonathan D. Awaya
- Department of Molecular Biosciences and Bioengineering, University of Hawai´i at Mānoa, Honolulu, HI, United States
- Department of Biology, University of Hawai´i at Hilo, Hilo, HI, United States
| |
Collapse
|
34
|
Abstract
Our work on the complexation of fluoride anions using group 15 Lewis acids has led us to investigate the use of these main group compounds as anion transporters. In this paper, we report on the anion transport properties of tetraarylstibonium and tetraarylbismuthonium cations of the general formula [Ph3PnAr]+ with Pn = Sb or Bi and with Ar = phenyl, naphthyl, anthryl, or pyrenyl. Using EYPC-based large unilamellar vesicles, we show that these main group cations transport hydroxide, fluoride and chloride anions across phospholipid bilayers. A comparison of the properties of [Ph3SbAnt]+ and [Ph3BiAnt]+ (Ant = 9-anthryl) illustrates the favorable role played by the Lewis acidity of the central pnictogen element with respect to the anion transport. Finally, we show that [Ph3SbAnt]+ accelerates the fluoride-induced hemolysis of human red blood cells, an effect that we assign to the transporter-facilitated influx of toxic fluoride anions.
Collapse
|
35
|
Mondal A, Gandhi A, Fimognari C, Atanasov AG, Bishayee A. Alkaloids for cancer prevention and therapy: Current progress and future perspectives. Eur J Pharmacol 2019; 858:172472. [PMID: 31228447 DOI: 10.1016/j.ejphar.2019.172472] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022]
Abstract
Alkaloids are important chemical compounds that serve as a rich source for drug discovery. Numerous alkaloids screened from medicinal plants and herbs showed antiproliferative and anticancer effects on wide category of cancers both in vitro and in vivo. Vinblastine, vinorelbine, vincristine, and vindesine have already been successfully developed as anticancer drugs. The available and up-to-date information on the ethnopharmacological uses in traditional medicine, phytochemistry, pharmacology and clinical utility of alkaloids were collected using various resources (PubMed, ScienceDirect, Google Scholar and Springerlink). In this article, we provide a comprehensive and critical overview on naturally-occurring alkaloids with anticancer activities and highlight the molecular mechanisms of action of these secondary metabolites. Furthermore, this review also presents a summary of synthetic derivatives and pharmacological profiles useful to researchers for the therapeutic development of alkaloids. Based on the literature survey compiled in this review, alkaloids represent an important group of anticancer drugs of plant origin with enormous potential for future development of drugs for cancer therapy and management.
Collapse
Affiliation(s)
- Arijit Mondal
- Department of Pharmacy, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, 700 053, West Bengal, India.
| | - Arijit Gandhi
- Department of Pharmaceutics, Bengal College of Pharmaceutical Science and Research, Durgapur, 713 212, West Burdwan, West Bengal, India
| | - Carmela Fimognari
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto 237, 47921, Rimini, Italy
| | - Atanas G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552, Jastrzebiec, Poland; Department of Pharmacognosy, University of Vienna, 1090, Vienna, Austria; Institute of Neurobiology, Bulgarian Academy of Sciences, 23 Acad. G. Bonchev Street, Sofia, 1113, Bulgaria
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
36
|
Marchetti PM, Kelly V, Simpson JP, Ward M, Campopiano DJ. The carbon chain-selective adenylation enzyme TamA: the missing link between fatty acid and pyrrole natural product biosynthesis. Org Biomol Chem 2019; 16:2735-2740. [PMID: 29594310 PMCID: PMC5939613 DOI: 10.1039/c8ob00441b] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
TamA is the adenylating enzyme that selects and activates fatty acids for tambjamine biosynthesis.
The marine bacterium Pseudoalteromonas tunicata produces the bipyrrole antibiotic tambjamine YP1. This natural product is built from common amino acid and fatty acid building blocks in a biosynthetic pathway that is encoded in the tam operon which contains 19 genes. The exact role that each of these Tam proteins plays in tambjamine biosynthesis is not known. Here, we provide evidence that TamA initiates the synthesis and controls the chain length of the essential tambjamine fatty amine tail. Sequence analysis suggests the unusual TamA is comprised of an N-terminal adenylation (ANL) domain fused to a C-terminal acyl carrier protein (ACP). Mass spectrometry analysis of recombinant TamA revealed the surprising presence of bound C11 and C12 acyl-adenylate intermediates. Acylation of the ACP domain was observed upon attachment of the phosphopantetheine (4′-PP) arm to the ACP. We also show that TamA can transfer fatty acids ranging in chain length from C6–C13 to an isolated ACP domain. Thus TamA bridges the gap between primary and secondary metabolism by linking fatty acid and pyrrole biosynthetic pathways.
Collapse
Affiliation(s)
- Piera M Marchetti
- EaStCHEM School of Chemistry, David Brewster Road, University of Edinburgh, Edinburgh, EH9 3FJ, UK.
| | | | | | | | | |
Collapse
|
37
|
Abstract
Recently, we showed that synthetic anion transporters DSC4P-1 and SA-3 had activity related to cancer cell death. They were found to increase intracellular chloride and sodium ion concentrations. They were also found to induce apoptosis (DSC4P-1) and both induce apoptosis and inhibit autophagy (SA-3). However, determinants underlying these phenomenological findings were not elucidated. The absence of mechanistic understanding has limited the development of yet-improved systems. Here, we show that three synthetic anion transporters, DSC4P-1, SA-3, and 8FC4P, induce osmotic stress in cells by increasing intracellular ion concentrations. This triggers the generation of reactive oxygen species via a sequential process and promotes caspase-dependent apoptosis. In addition, two of the transporters, SA-3 and 8FC4P, induce autophagy by increasing the cytosolic calcium ion concentration promoted by osmotic stress. However, they eventually inhibit the autophagy process as a result of their ability to disrupt lysosome function through a transporter-mediated decrease in a lysosomal chloride ion concentration and an increase in the lysosomal pH.
Collapse
|
38
|
Synthesis and biological evaluation of aza-crown ether–squaramide conjugates as anion/cation symporters. Future Med Chem 2019; 11:1091-1106. [DOI: 10.4155/fmc-2018-0595] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: Anion/cation symport across cellular membranes may lead to cell apoptosis and be developed as a strategy for new anticancer drug discovery. Methodology: Four aza-crown ether–squaramide conjugates were synthesized and characterized. Their anion recognition, anion/cation symport, cytotoxicity and probable mechanism of action were investigated in details. Conclusion: These conjugates are able to form ion-pairing complexes with chloride anions and facilitate the transmembrane transport of anions via an anion/cation symport process. They can disrupt the cellular homeostasis of chloride anions and sodium cations and induce the basification of acidic organelles in live cells. These conjugates exhibit moderate cytotoxicity toward the tested cancer cells and trigger cell apoptosis by mediating the influx of chloride anions and sodium cations into live cells.
Collapse
|
39
|
Fiore M, Cossu C, Capurro V, Picco C, Ludovico A, Mielczarek M, Carreira-Barral I, Caci E, Baroni D, Quesada R, Moran O. Small molecule-facilitated anion transporters in cells for a novel therapeutic approach to cystic fibrosis. Br J Pharmacol 2019; 176:1764-1779. [PMID: 30825185 DOI: 10.1111/bph.14649] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/05/2019] [Accepted: 02/08/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Cystic fibrosis (CF) is a lethal autosomal recessive genetic disease that originates from the defective function of the CF transmembrane conductance regulator (CFTR) protein, a cAMP-dependent anion channel involved in fluid transport across epithelium. Because small synthetic transmembrane anion transporters (anionophores) can replace the biological anion transport mechanisms, independent of genetic mutations in the CFTR, such anionophores are candidates as new potential treatments for CF. EXPERIMENTAL APPROACH In order to assess their effects on cell physiology, we have analysed the transport properties of five anionophore compounds, three prodigiosines and two tambjamines. Chloride efflux was measured in large uni-lamellar vesicles and in HEK293 cells with chloride-sensitive electrodes. Iodide influx was evaluated in FRT cells transfected with iodide-sensitive YFP. Transport of bicarbonate was assessed by changes of pH after a NH4 + pre-pulse using the BCECF fluorescent probe. Assays were also carried out in FRT cells permanently transfected with wild type and mutant human CFTR. KEY RESULTS All studied compounds are capable of transporting halides and bicarbonate across the cell membrane, with a higher transport capacity at acidic pH. Interestingly, the presence of these anionophores did not interfere with the activation of CFTR and did not modify the action of lumacaftor (a CFTR corrector) or ivacaftor (a CFTR potentiator). CONCLUSION AND IMPLICATIONS These anionophores, at low concentrations, transported chloride and bicarbonate across cell membranes, without affecting CFTR function. They therefore provide promising starting points for the development of novel treatments for CF.
Collapse
Affiliation(s)
| | | | - Valeria Capurro
- U.O.C. Genetica Medica, Istituto Giannina Gaslini, Genova, Italy
| | | | | | - Marcin Mielczarek
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, Burgos, Spain
| | | | - Emanuela Caci
- U.O.C. Genetica Medica, Istituto Giannina Gaslini, Genova, Italy
| | | | - Roberto Quesada
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, Burgos, Spain
| | | |
Collapse
|
40
|
Zhang S, Wang Y, Xie W, Howe ENW, Busschaert N, Sauvat A, Leduc M, Gomes-da-Silva LC, Chen G, Martins I, Deng X, Maiuri L, Kepp O, Soussi T, Gale PA, Zamzami N, Kroemer G. Squaramide-based synthetic chloride transporters activate TFEB but block autophagic flux. Cell Death Dis 2019; 10:242. [PMID: 30858361 PMCID: PMC6411943 DOI: 10.1038/s41419-019-1474-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 01/08/2019] [Accepted: 02/22/2019] [Indexed: 02/06/2023]
Abstract
Cystic fibrosis is a disease caused by defective function of a chloride channel coupled to a blockade of autophagic flux. It has been proposed to use synthetic chloride transporters as pharmacological agents to compensate insufficient chloride fluxes. Here, we report that such chloride anionophores block autophagic flux in spite of the fact that they activate the pro-autophagic transcription factor EB (TFEB) coupled to the inhibition of the autophagy-suppressive mTORC1 kinase activity. Two synthetic chloride transporters (SQ1 and SQ2) caused a partially TFEB-dependent relocation of the autophagic marker LC3 to the Golgi apparatus. Inhibition of TFEB activation using a calcium chelator or calcineurin inhibitors reduced the formation of LC3 puncta in cells, yet did not affect the cytotoxic action of SQ1 and SQ2 that could be observed after prolonged incubation. In conclusion, the squaramide-based synthetic chloride transporters studied in this work (which can also dissipate pH gradients) are probably not appropriate for the treatment of cystic fibrosis yet might be used for other indications such as cancer.
Collapse
Affiliation(s)
- Shaoyi Zhang
- Department of Surgery, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Faculty of Medicine, University of Paris Sud-Saclay, Kremlin-Bicêtre, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Sorbonne Université, UPMC Univ Paris, Paris, France
| | - Yan Wang
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Sorbonne Université, UPMC Univ Paris, Paris, France.,Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wei Xie
- Faculty of Medicine, University of Paris Sud-Saclay, Kremlin-Bicêtre, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Sorbonne Université, UPMC Univ Paris, Paris, France
| | - Ethan N W Howe
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
| | | | - Allan Sauvat
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Sorbonne Université, UPMC Univ Paris, Paris, France
| | - Marion Leduc
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Sorbonne Université, UPMC Univ Paris, Paris, France
| | - Lígia C Gomes-da-Silva
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Sorbonne Université, UPMC Univ Paris, Paris, France.,Chemistry Department, University of Coimbra, Coimbra, Portugal
| | - Guo Chen
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Sorbonne Université, UPMC Univ Paris, Paris, France
| | - Isabelle Martins
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Sorbonne Université, UPMC Univ Paris, Paris, France
| | - Xiaxing Deng
- Department of Surgery, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy.,Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Sorbonne Université, UPMC Univ Paris, Paris, France
| | - Thierry Soussi
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Sorbonne Université, UPMC Univ Paris, Paris, France.,Department of Oncology-Pathology, Cancer Center Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Philip A Gale
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Naoufal Zamzami
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France. .,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France. .,Université Paris Descartes, Sorbonne Paris Cité, Paris, France. .,Gustave Roussy Comprehensive Cancer Center, Villejuif, France. .,Sorbonne Université, UPMC Univ Paris, Paris, France.
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France. .,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France. .,Université Paris Descartes, Sorbonne Paris Cité, Paris, France. .,Gustave Roussy Comprehensive Cancer Center, Villejuif, France. .,Sorbonne Université, UPMC Univ Paris, Paris, France. .,Pôle de Biologie, Hôpital Européen Georges Pompidou, APsupp-HP, Paris, France. .,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
41
|
Yu XH, Hong XQ, Chen WH. Fluorinated bisbenzimidazoles: a new class of drug-like anion transporters with chloride-mediated, cell apoptosis-inducing activity. Org Biomol Chem 2019; 17:1558-1571. [DOI: 10.1039/c8ob03036g] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Fluorinated bisbenzimidazoles were synthesized as a new class of drug-like anion transporters with chloride-mediated, cell apoptosis-inducing activity.
Collapse
Affiliation(s)
- Xi-Hui Yu
- Guangdong Provincial Key Laboratory of New Drug Screening
- School of Pharmaceutical Sciences
- Southern Medical University
- Guangzhou 510515
- P. R. China
| | - Xiao-Qiao Hong
- Guangdong Provincial Key Laboratory of New Drug Screening
- School of Pharmaceutical Sciences
- Southern Medical University
- Guangzhou 510515
- P. R. China
| | - Wen-Hua Chen
- Guangdong Provincial Key Laboratory of New Drug Screening
- School of Pharmaceutical Sciences
- Southern Medical University
- Guangzhou 510515
- P. R. China
| |
Collapse
|
42
|
Synthesis, anionophoric activity and apoptosis-inducing bioactivity of benzimidazolyl-based transmembrane anion transporters. Eur J Med Chem 2018; 152:115-125. [DOI: 10.1016/j.ejmech.2018.04.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 11/18/2022]
|
43
|
Cheung S, Wu D, Daly HC, Busschaert N, Morgunova M, Simpson JC, Scholz D, Gale PA, O'Shea DF. Real-Time Recording of the Cellular Effects of the Anion Transporter Prodigiosin. Chem 2018. [DOI: 10.1016/j.chempr.2018.02.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
44
|
Hernando E, Capurro V, Cossu C, Fiore M, García-Valverde M, Soto-Cerrato V, Pérez-Tomás R, Moran O, Zegarra-Moran O, Quesada R. Small molecule anionophores promote transmembrane anion permeation matching CFTR activity. Sci Rep 2018; 8:2608. [PMID: 29422673 PMCID: PMC5805763 DOI: 10.1038/s41598-018-20708-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/22/2018] [Indexed: 12/30/2022] Open
Abstract
Anion selective ionophores, anionophores, are small molecules capable of facilitating the transmembrane transport of anions. Inspired in the structure of natural product prodigiosin, four novel anionophores 1a-d, including a 1,2,3-triazole group, were prepared. These compounds proved highly efficient anion exchangers in model phospholipid liposomes. The changes in the hydrogen bond cleft modified the anion transport selectivity exhibited by these compounds compared to prodigiosin and suppressed the characteristic high toxicity of the natural product. Their activity as anionophores in living cells was studied and chloride efflux and iodine influx from living cells mediated by these derivatives was demonstrated. These compounds were shown to permeabilize cellular membranes to halides with efficiencies close to the natural anion channel CFTR at doses that do not compromise cellular viability. Remarkably, optimal transport efficiency was measured in the presence of pH gradients mimicking those found in the airway epithelia of Cystic Fibrosis patients. These results support the viability of developing small molecule anionophores as anion channel protein surrogates with potential applications in the treatment of conditions such as Cystic Fibrosis derived from the malfunction of natural anion transport mechanisms.
Collapse
Affiliation(s)
- Elsa Hernando
- Departamento de Química, Universidad de Burgos, 09001, Burgos, Spain
| | - Valeria Capurro
- U.O.C. Genetica Medica, Instituto Giannina Gaslini, Genoa, Italy
| | | | | | | | - Vanessa Soto-Cerrato
- Cancer Cell Biology Research Group, Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Ricardo Pérez-Tomás
- Cancer Cell Biology Research Group, Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | | | | | - Roberto Quesada
- Departamento de Química, Universidad de Burgos, 09001, Burgos, Spain.
| |
Collapse
|
45
|
Hong XQ, Yu XH, Zhang K, Chen WH. Synthesis and properties of a lysosome-targeting fluorescent ionophore based on coumarins and squaramides. Org Biomol Chem 2018; 16:8025-8029. [DOI: 10.1039/c8ob01957f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A squaramide derivative bearing a coumarin fluorophore and a morpholinyl group was synthesized and found to be able to target and efficiently deacidify lysosomes.
Collapse
Affiliation(s)
- Xiao-Qiao Hong
- Guangdong Provincial Key Laboratory of New Drug Screening
- School of Pharmaceutical Sciences
- Southern Medical University
- Guangzhou 510515
- P. R. China
| | - Xi-Hui Yu
- Guangdong Provincial Key Laboratory of New Drug Screening
- School of Pharmaceutical Sciences
- Southern Medical University
- Guangzhou 510515
- P. R. China
| | - Kun Zhang
- School of Biotechnology and Health Sciences
- Wuyi University
- Jiangmen 529020
- P. R. China
- International Healthcare Innovation Institute (Jiangmen)
| | - Wen-Hua Chen
- Guangdong Provincial Key Laboratory of New Drug Screening
- School of Pharmaceutical Sciences
- Southern Medical University
- Guangzhou 510515
- P. R. China
| |
Collapse
|
46
|
Behera H, Madhavan N. Anion-Selective Cholesterol Decorated Macrocyclic Transmembrane Ion Carriers. J Am Chem Soc 2017; 139:12919-12922. [DOI: 10.1021/jacs.7b07479] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Harekrushna Behera
- Department
of Chemistry, Indian Institute of Technology, Madras, Tamil Nadu 600036, India
| | - Nandita Madhavan
- Department
of Chemistry, Indian Institute of Technology, Madras, Tamil Nadu 600036, India
- Indian Institute of Technology, Bombay, Maharashtra 400076, India
| |
Collapse
|
47
|
Jowett LA, Howe ENW, Soto-Cerrato V, Van Rossom W, Pérez-Tomás R, Gale PA. Indole-based perenosins as highly potent HCl transporters and potential anti-cancer agents. Sci Rep 2017; 7:9397. [PMID: 28839192 PMCID: PMC5570892 DOI: 10.1038/s41598-017-09645-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/24/2017] [Indexed: 11/19/2022] Open
Abstract
Prodigiosin is one of the most potent anion transporters in lipid bilayer membranes reported to date. Inspired by the structure of this natural product, we have recently designed and synthesised a new class of H+/Cl− cotransporters named ‘perenosins’. Here we report a new library of indole-based perenosins and their anion transport properties. The new transporters demonstrated superior transmembrane transport efficiency when compared to other indole-based transporters, due to favourable encapsulating effects from the substituents on the perenosin backbone. Anion transport assays were used to determine the mechanism of chloride transport revealing that the compounds function as ‘strict’ HCl cotransporters. Cell viability studies showed that some compounds specifically trigger late-onset cell death after 72 h with a unique correlation to the position of alkyl chains on the perenosins. Further investigations of cell death mechanism showed a mixture of cell cycle arrest and apoptosis was responsible for the observed decrease in cell viability.
Collapse
Affiliation(s)
- Laura A Jowett
- School of Chemistry (F11), The University of Sydney, 2006, Sydney, NSW, Australia
| | - Ethan N W Howe
- School of Chemistry (F11), The University of Sydney, 2006, Sydney, NSW, Australia
| | - Vanessa Soto-Cerrato
- Department of Pathology and Experimental Therapeutics, Cancer Cell Biology Research Group, University of Barcelona, Barcelona, Spain
| | - Wim Van Rossom
- Chemistry, University of Southampton, Highfield, Southampton, SO17 1BJ, UK
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapeutics, Cancer Cell Biology Research Group, University of Barcelona, Barcelona, Spain
| | - Philip A Gale
- School of Chemistry (F11), The University of Sydney, 2006, Sydney, NSW, Australia.
| |
Collapse
|
48
|
Kotev M, Manuel-Manresa P, Hernando E, Soto-Cerrato V, Orozco M, Quesada R, Pérez-Tomás R, Guallar V. Inhibition of Human Enhancer of Zeste Homolog 2 with Tambjamine Analogs. J Chem Inf Model 2017; 57:2089-2098. [PMID: 28763207 DOI: 10.1021/acs.jcim.7b00178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Combining computational modeling, de novo compound synthesis, and in vitro and cellular assays, we have performed an inhibition study against the enhancer of zeste homolog 2 (EZH2) histone-lysine N-methyltransferase. This enzyme is an important catalytic component of the PRC2 complex whose alterations have been associated with different cancers. We introduce here several tambjamine-inspired derivatives with low micromolar in vitro activity that produce a significant decrease in histone 3 trimethylation levels in cancer cells. We demonstrate binding at the methyl transfer active site, showing, in addition, that the EZH2 isolated crystal structure is capable of being used in molecular screening studies. Altogether, this work provides a successful molecular model that will help in the identification of new specific EZH2 inhibitors and identify a novel class of tambjamine-derived EZH2 inhibitors with promising activities for their use in cancer treatment.
Collapse
Affiliation(s)
- Martin Kotev
- Joint BSC-CRG-IRB Research Program in Computational Biology. Barcelona Supercomputing Center, c/Jordi Girona 29, 08034 Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), Baldiri i Reixac 8, Barcelona 08028, Spain
| | - Pilar Manuel-Manresa
- Cancer Cell Biology Research Group, Department of Pathology and Experimental Therapeutics, University of Barcelona , E-08907 Barcelona, Spain
| | - Elsa Hernando
- Department of Chemistry, Faculty of Science, University of Burgos , 09001 Burgos, Spain
| | - Vanessa Soto-Cerrato
- Cancer Cell Biology Research Group, Department of Pathology and Experimental Therapeutics, University of Barcelona , E-08907 Barcelona, Spain
| | - Modesto Orozco
- Institute for Research in Biomedicine (IRB Barcelona), Baldiri i Reixac 8, Barcelona 08028, Spain
| | - Roberto Quesada
- Department of Chemistry, Faculty of Science, University of Burgos , 09001 Burgos, Spain
| | - Ricardo Pérez-Tomás
- Cancer Cell Biology Research Group, Department of Pathology and Experimental Therapeutics, University of Barcelona , E-08907 Barcelona, Spain
| | - Victor Guallar
- Joint BSC-CRG-IRB Research Program in Computational Biology. Barcelona Supercomputing Center, c/Jordi Girona 29, 08034 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
49
|
Gale PA, Davis JT, Quesada R. Anion transport and supramolecular medicinal chemistry. Chem Soc Rev 2017; 46:2497-2519. [DOI: 10.1039/c7cs00159b] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
New approaches to the transmembrane transport of anions are discussed in this review.
Collapse
Affiliation(s)
- Philip A. Gale
- School of Chemistry (F11)
- The University of Sydney
- Australia
| | - Jeffery T. Davis
- Department of Chemistry and Biochemistry
- University of Maryland
- College Park
- USA
| | - Roberto Quesada
- Departmento de Química
- Universidad de Burgos
- 09001 Burgos
- Spain
| |
Collapse
|
50
|
Yang Y, Wu X, Busschaert N, Furuta H, Gale PA. Dissecting the chloride–nitrate anion transport assay. Chem Commun (Camb) 2017; 53:9230-9233. [DOI: 10.1039/c7cc04912a] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The chloride/nitrate selectivity of anion transporters in both binding and membrane transport is examined revealing the limitations of chloride–nitrate anion exchange assay.
Collapse
Affiliation(s)
- Yufeng Yang
- Chemistry
- University of Southampton
- Southampton
- UK
- Department of Chemistry and Biochemistry
| | - Xin Wu
- School of Chemistry
- The University of Sydney
- Australia
| | | | - Hiroyuki Furuta
- Department of Chemistry and Biochemistry
- Kyushu University
- Fukuoka
- Japan
| | | |
Collapse
|