1
|
Jain A, Mishra AK, Hurkat P, Shilpi S, Mody N, Jain SK. Navigating liver cancer: Precision targeting for enhanced treatment outcomes. Drug Deliv Transl Res 2025; 15:1935-1961. [PMID: 39847205 DOI: 10.1007/s13346-024-01780-x] [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] [Accepted: 12/18/2024] [Indexed: 01/24/2025]
Abstract
Cancer treatments such as surgery and chemotherapy have several limitations, including ineffectiveness against large or persistent tumors, high relapse rates, drug toxicity, and non-specificity of therapy. Researchers are exploring advanced strategies for treating this life-threatening disease to address these challenges. One promising approach is targeted drug delivery using prodrugs or surface modification with receptor-specific moieties for active or passive targeting. While various drug delivery systems have shown potential for reaching hepatic cells, nano-carriers offer significant size, distribution, and targetability advantages. Engineered nanocarriers can be customized to achieve effective and safe targeting of tumors by manipulating physical characteristics such as particle size or attaching receptor-specific ligands. This method is particularly advantageous in treating liver cancer by targeting specific hepatocyte receptors and enzymatic pathways for both passive and active therapeutic strategies. It highlights the epidemiology of liver cancer and provides an in-depth analysis of the various targeting approaches, including prodrugs, liposomes, magneto-liposomes, micelles, glycol-dendrimers, magnetic nanoparticles, chylomicron-based emulsion, and quantum dots surface modification with receptor-specific moieties. The insights from this review can be immensely significant for preclinical and clinical researchers working towards developing effective treatments for liver cancer. By utilizing these novel strategies, we can overcome the limitations of conventional therapies and offer better outcomes for liver cancer patients.
Collapse
Affiliation(s)
- Ankit Jain
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, 333031, India.
| | - Ashwini Kumar Mishra
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
- Central Ayurveda Research Institute, Jhansi, Uttar Pradesh, 284003, India
| | - Pooja Hurkat
- Dr. Hari Singh Gour Central University, Sagar, 470003, MP, India
| | - Satish Shilpi
- School of Pharmaceuticals and Population Health Informatics, FOP, DIT University, Dehradun, Uttarakahnad, India
| | - Nishi Mody
- Dr. Hari Singh Gour Central University, Sagar, 470003, MP, India
| | | |
Collapse
|
2
|
Sagar B, Gupta S, Verma SK, Reddy YVM, Shukla S. Navigating cancer therapy: Harnessing the power of peptide-drug conjugates as precision delivery vehicles. Eur J Med Chem 2025; 283:117131. [PMID: 39647418 DOI: 10.1016/j.ejmech.2024.117131] [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: 01/30/2024] [Revised: 11/17/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
Cancer treatment is a formidable challenge due to the adverse effects associated with non-selective therapies like chemotherapy and radiotherapy. This review article primarily centers on the application of Peptide-Drug Conjugates (PDCs) for delivering cancer treatment. PDCs represent a promising class of precision medicines, harnessing the unique attributes of peptides in conjunction with non-peptide components. The covalent linking of peptides and drugs through specialized connectors characterizes PDCs. These constructs play a pivotal role in delivering drugs directly to tumor sites with high precision. PDCs encompass three pivotal components: a targeting ligand, a cytotoxic ligand, and a carefully chosen linker. The selection of these elements is crucial to maximize the efficiency of PDCs. PDCs offer a multitude of advantages over conventional drug molecules, including enhanced specificity, reduced off-target effects, and an improved therapeutic profile. The peptide component within PDCs can be customized to specifically adhere to disease-specific receptors or biomarkers, facilitating targeted drug delivery and accumulation in afflicted cells or tissues. This targeted approach enables the controlled release of therapeutic payloads at the localized site, resulting in heightened effectiveness and minimized systemic toxicity. Diverse linker strategies are employed to ensure the stable connection between the peptide and non-peptide components, ensuring controlled drug release at the desired location of action. The peptides utilized in these treatments encompass cell-penetrating peptides, peptides designed to target tumor cells, and those aimed at the nucleus of cancer cells. While certain clinical trials have been conducted, and some PDCs are currently in use for cancer treatment, it's essential to acknowledge that PDCs have their limitations, such as low stability in plasma, fast elimination and limited oral bioavailability. Ongoing research endeavors seek to surmount these challenges and further establish PDCs as potent agents for cancer treatment. This review sheds light on recent advancements in the design, delivery, and applications of PDCs, while also highlighting the prevailing challenges and charting a path for future research directions.
Collapse
Affiliation(s)
- Bulbul Sagar
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016, New Delhi, India
| | - Sarthak Gupta
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016, New Delhi, India
| | - Sarvesh Kumar Verma
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, 302017, Rajasthan, India
| | | | - Shefali Shukla
- Sri Venkateswara College, University of Delhi, New Delhi, India.
| |
Collapse
|
3
|
Martinez P, Baghli I, Gourjon G, Seyfried TN. Mitochondrial-Stem Cell Connection: Providing Additional Explanations for Understanding Cancer. Metabolites 2024; 14:229. [PMID: 38668357 PMCID: PMC11051897 DOI: 10.3390/metabo14040229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/29/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The cancer paradigm is generally based on the somatic mutation model, asserting that cancer is a disease of genetic origin. The mitochondrial-stem cell connection (MSCC) proposes that tumorigenesis may result from an alteration of the mitochondria, specifically a chronic oxidative phosphorylation (OxPhos) insufficiency in stem cells, which forms cancer stem cells (CSCs) and leads to malignancy. Reviewed evidence suggests that the MSCC could provide a comprehensive understanding of all the different stages of cancer. The metabolism of cancer cells is altered (OxPhos insufficiency) and must be compensated by using the glycolysis and the glutaminolysis pathways, which are essential to their growth. The altered mitochondria regulate the tumor microenvironment, which is also necessary for cancer evolution. Therefore, the MSCC could help improve our understanding of tumorigenesis, metastases, the efficiency of standard treatments, and relapses.
Collapse
Affiliation(s)
- Pierrick Martinez
- Scientific and Osteopathic Research Department, Institut de Formation en Ostéopathie du Grand Avignon, 84140 Montfavet, France;
| | - Ilyes Baghli
- International Society for Orthomolecular Medicine, Toronto, ON M4B 3M9, Canada;
| | - Géraud Gourjon
- Scientific and Osteopathic Research Department, Institut de Formation en Ostéopathie du Grand Avignon, 84140 Montfavet, France;
| | | |
Collapse
|
4
|
Casciati A, Taddei AR, Rampazzo E, Persano L, Viola G, Cani A, Bresolin S, Cesi V, Antonelli F, Mancuso M, Merla C, Tanori M. Involvement of Mitochondria in the Selective Response to Microsecond Pulsed Electric Fields on Healthy and Cancer Stem Cells in the Brain. Int J Mol Sci 2024; 25:2233. [PMID: 38396911 PMCID: PMC10889160 DOI: 10.3390/ijms25042233] [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: 11/17/2023] [Revised: 02/02/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
In the last few years, pulsed electric fields have emerged as promising clinical tools for tumor treatments. This study highlights the distinct impact of a specific pulsed electric field protocol, PEF-5 (0.3 MV/m, 40 μs, 5 pulses), on astrocytes (NHA) and medulloblastoma (D283) and glioblastoma (U87 NS) cancer stem-like cells (CSCs). We pursued this goal by performing ultrastructural analyses corroborated by molecular/omics approaches to understand the vulnerability or resistance mechanisms triggered by PEF-5 exposure in the different cell types. Electron microscopic analyses showed that, independently of exposed cells, the main targets of PEF-5 were the cell membrane and the cytoskeleton, causing membrane filopodium-like protrusion disappearance on the cell surface, here observed for the first time, accompanied by rapid cell swelling. PEF-5 induced different modifications in cell mitochondria. A complete mitochondrial dysfunction was demonstrated in D283, while a mild or negligible perturbation was observed in mitochondria of U87 NS cells and NHAs, respectively, not sufficient to impair their cell functions. Altogether, these results suggest the possibility of using PEF-based technology as a novel strategy to target selectively mitochondria of brain CSCs, preserving healthy cells.
Collapse
Affiliation(s)
- Arianna Casciati
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| | - Anna Rita Taddei
- Great Equipment Center-Section of Electron Microscopy, University of Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
| | - Elena Rampazzo
- Department of Women’s and Children’s Health (SDB), University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Luca Persano
- Department of Women’s and Children’s Health (SDB), University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Giampietro Viola
- Department of Women’s and Children’s Health (SDB), University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Alice Cani
- Department of Women’s and Children’s Health (SDB), University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Silvia Bresolin
- Department of Women’s and Children’s Health (SDB), University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Vincenzo Cesi
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| | - Francesca Antonelli
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| | - Mariateresa Mancuso
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| | - Caterina Merla
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| | - Mirella Tanori
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| |
Collapse
|
5
|
Rainho MDA, Siqueira PB, de Amorim ÍSS, Mencalha AL, Thole AA. Mitochondria in colorectal cancer stem cells - a target in drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:273-283. [PMID: 37457136 PMCID: PMC10344721 DOI: 10.20517/cdr.2022.116] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/15/2023] [Accepted: 04/24/2023] [Indexed: 07/18/2023]
Abstract
Colorectal cancer (CRC) is the third most diagnosed cancer and the second most deadly type of cancer worldwide. In late diagnosis, CRC can resist therapy regimens in which cancer stem cells (CSCs) are intimately related. CSCs are a subpopulation of tumor cells responsible for tumor initiation and maintenance, metastasis, and resistance to conventional treatments. In this scenario, colorectal cancer stem cells (CCSCs) are considered an important key for therapeutic failure and resistance. In its turn, mitochondria is an organelle involved in many mechanisms in cancer, including chemoresistance of cytotoxic drugs due to alterations in mitochondrial metabolism, apoptosis, dynamics, and mitophagy. Therefore, it is crucial to understand the mitochondrial role in CCSCs regarding CRC drug resistance. It has been shown that enhanced anti-apoptotic protein expression, mitophagy rate, and addiction to oxidative phosphorylation are the major strategies developed by CCSCs to avoid drug insults. Thus, new mitochondria-targeted drug approaches must be explored to mitigate CRC chemoresistance via the ablation of CCSCs.
Collapse
Affiliation(s)
- Mateus de Almeida Rainho
- Laboratory of Stem Cell Research, Histology and Embryology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil
| | - Priscyanne Barreto Siqueira
- Laboratory of Cancer Biology, Biometry and Biophysics Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil
| | - Ísis Salviano Soares de Amorim
- Laboratory of Cancer Biology, Biometry and Biophysics Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil
| | - Andre Luiz Mencalha
- Laboratory of Cancer Biology, Biometry and Biophysics Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil
| | - Alessandra Alves Thole
- Laboratory of Stem Cell Research, Histology and Embryology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil
| |
Collapse
|
6
|
Wang Q, Chai D, Sobhani N, Sun N, Neeli P, Zheng J, Tian H. C1QBP regulates mitochondrial plasticity to impact tumor progression and antitumor immune response. Front Physiol 2022; 13:1012112. [PMID: 36467687 PMCID: PMC9713694 DOI: 10.3389/fphys.2022.1012112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/02/2022] [Indexed: 12/22/2024] Open
Abstract
Mitochondrial plasticity including mitochondrial dynamics, metabolic flexibility, and mitochondrial quality control, impact tumor cells' progression and determine immune cells' fate. Complement C1q binding protein (C1QBP) plays an indispensable role through regulating mitochondrial morphology, metabolism, and autophagy. C1QBP promotes mitochondrial plasticity to impact tumor metastasis and their therapeutic response. At the same time, C1QBP is involved in regulating immune cells' maturation, differentiation, and effector function through the enhancement of mitochondrial function. In this regard, manipulation of C1QBP has been shown to adjust the competitive balance between tumor cells and immune cells. In the course of evolution, mitochondrial plasticity has endowed numerous advantages against the relentless microenvironment of tumors. In this current review, we summarize the current knowledge of the mechanism of C1QBP regulation of cancer and immunity. We explain this process in vision of potentially new anticancer therapies.
Collapse
Affiliation(s)
- Qiping Wang
- Jiangyin Clinical Medical College, Jiangsu University, Jiangyin, Jiangsu, China
| | - Dafei Chai
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Navid Sobhani
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Nan Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Praveen Neeli
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hui Tian
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
7
|
McCartin C, Mathieu E, Dontenwill M, Herold-Mende C, Idbaih A, Bonfiglio A, Mauro M, Fournel S, Kichler A. An N-heterocyclic carbene iridium(III) complex as a potent anti-cancer stem cell therapeutic. Chem Biol Interact 2022; 367:110167. [PMID: 36087816 DOI: 10.1016/j.cbi.2022.110167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/12/2022] [Accepted: 09/02/2022] [Indexed: 12/20/2022]
Abstract
Cancer stem cells (CSCs) represent a difficult to treat cellular niche within tumours due to their unique characteristics, which give them a high propensity for resistance to classical anti-cancer treatments and the ability to repopulate the tumour mass. An attribute that may be implicated in the high rates of recurrence of certain tumours. However, other characteristics specific to these cells, such as their high dependence on mitochondria, may be exploited for the development of new therapeutic agents that are effective against the niche. As such, a previously described phosphorescent N-heterocyclic carbene iridium(III) compound which showed a high level of cytotoxicity against classical tumour cell lines with mitochondria-specific effects was studied for its potential against CSCs. The results showed a significantly higher level of activity against several CSC lines compared to non-CSCs. Mitochondrial localisation and superoxide production were confirmed. Although the cell death involved caspase activation, their role in cell death was not definitive, with a potential implication of other, non-apoptotic pathways shown. A cytostatic effect of the compound was also displayed at low mortality doses. This study thus provides important insights into the mechanisms and the potential for this class of molecule in the domain of anti-CSC therapeutics.
Collapse
Affiliation(s)
- Conor McCartin
- 3Bio Team, CAMB UMR7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, F-67401 Illkirch cedex, France
| | - Eric Mathieu
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR_S 1121 Biomaterials and Bioengineering, Strasbourg, F-67085, France; Université de Strasbourg, Faculté de Chirurgie Dentaire de Strasbourg, Strasbourg, F-67000, France
| | - Monique Dontenwill
- Laboratoire de bioimagerie et pathologies UMR CNRS 7021 (LBP), Faculté de Pharmacie, 74 route du Rhin, F-67401, Illkirch cedex, France
| | - Christel Herold-Mende
- Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Ahmed Idbaih
- Sorbonne Université, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, DMU Neurosciences, Service de Neurologie 2-Mazarin, F-75013, Paris, France
| | - Anna Bonfiglio
- Institut de Physique et Chimie des Matériaux de Strasbourg (IPCMS) UMR7504, Université de Strasbourg & CNRS 23 rue du Loess, 67083, Strasbourg, France
| | - Matteo Mauro
- Institut de Physique et Chimie des Matériaux de Strasbourg (IPCMS) UMR7504, Université de Strasbourg & CNRS 23 rue du Loess, 67083, Strasbourg, France
| | - Sylvie Fournel
- 3Bio Team, CAMB UMR7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, F-67401 Illkirch cedex, France.
| | - Antoine Kichler
- 3Bio Team, CAMB UMR7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, F-67401 Illkirch cedex, France.
| |
Collapse
|
8
|
Investigating the role of peptides in effective therapies against cancer. Cancer Cell Int 2022; 22:139. [PMID: 35346211 PMCID: PMC8962089 DOI: 10.1186/s12935-022-02553-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/14/2022] [Indexed: 12/03/2022] Open
Abstract
Early diagnosis and effective treatment of cancer are challenging. To diagnose and treat cancer effectively and to overcome these challenges, fundamental innovations in traditional diagnosis and therapy are necessary. Peptides can be very helpful in this regard due to their potential and diversity. To enhance the therapeutic potential of peptides, their limitations must be properly identified and their structures engineered and modified for higher efficiency. Promoting the bioavailability and stability of peptides is one of the main concerns. Peptides can also be effective in different areas of targeting, alone or with the help of other therapeutic agents. There has been a lot of research in this area, and the potential for variability of peptides will continue to improve this process. Another promising area in which peptides can help treat cancer is peptide vaccines, which are undergoing promising research, and high throughput technologies can lead to fundamental changes in this area. Peptides have been effective in almost all areas of cancer treatment, and some have even gone through clinical phases. However, many barriers need to be overcome to reach the desired point. The purpose of this review is to evaluate the mechanisms associated with peptides in the diagnosis and treatment of cancer. Therefore, related studies in this area will be discussed.
Collapse
|
9
|
Mitra S, Anand U, Jha NK, Shekhawat MS, Saha SC, Nongdam P, Rengasamy KRR, Proćków J, Dey A. Anticancer Applications and Pharmacological Properties of Piperidine and Piperine: A Comprehensive Review on Molecular Mechanisms and Therapeutic Perspectives. Front Pharmacol 2022; 12:772418. [PMID: 35069196 PMCID: PMC8776707 DOI: 10.3389/fphar.2021.772418] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/18/2021] [Indexed: 12/26/2022] Open
Abstract
Piperine and piperidine are the two major alkaloids extracted from black pepper (Piper nigrum); piperidine is a heterocyclic moiety that has the molecular formula (CH2)5NH. Over the years, many therapeutic properties including anticancer potential of these two compounds have been observed. Piperine has therapeutic potential against cancers such as breast cancer, ovarian cancer, gastric cancer, gliomal cancer, lung cancer, oral squamous, chronic pancreatitis, prostate cancer, rectal cancer, cervical cancer, and leukemia. Whereas, piperidine acts as a potential clinical agent against cancers, such as breast cancer, prostate cancer, colon cancer, lung cancer, and ovarian cancer, when treated alone or in combination with some novel drugs. Several crucial signalling pathways essential for the establishment of cancers such as STAT-3, NF-κB, PI3k/Aκt, JNK/p38-MAPK, TGF-ß/SMAD, Smac/DIABLO, p-IκB etc., are regulated by these two phytochemicals. Both of these phytochemicals lead to inhibition of cell migration and help in cell cycle arrest to inhibit survivability of cancer cells. The current review highlights the pharmaceutical relevance of both piperine and piperidine against different types of cancers.
Collapse
Affiliation(s)
- Sicon Mitra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Mahipal S Shekhawat
- Department of Plant Biology and Biotechnology, Kanchi Mamunivar Government Institute for Postgraduate Studies and Research, Lawspet, India
| | - Suchismita Chatterjee Saha
- Department of Zoology, Nabadwip Vidyasagar College (Affiliated to the University of Kalyani), Nabadwip, India
| | | | - Kannan R R Rengasamy
- Green Biotechnologies Research Centre of Excellence, University of Limpopo, Sovenga, South Africa
| | - Jarosław Proćków
- Department of Plant Biology, Institute of Environmental Biology, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Abhijit Dey
- Ethnopharmacology and Natural Product Research Laboratory, Department of Life Sciences, Presidency University, Kolkata, India
| |
Collapse
|
10
|
Crous A, Abrahamse H. Aluminium (III) phthalocyanine chloride tetrasulphonate is an effective photosensitizer for the eradication of lung cancer stem cells. ROYAL SOCIETY OPEN SCIENCE 2021; 8:210148. [PMID: 34527268 PMCID: PMC8424323 DOI: 10.1098/rsos.210148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 08/19/2021] [Indexed: 05/04/2023]
Abstract
Cancer stem cells (CSCs) are considered to contribute to the recurrence of lung cancer due to their stem-like nature and the involvement of genetic markers associated with drug efflux, regeneration and metastases. Photodynamic therapy (PDT) is a cost-effective and non-invasive therapeutic application that can act as an alternative therapy for lung cancer when considering CSC involvement. Stem-like cells derived from the A549 lung cancer cell line, positive for CD133, CD56 and CD44 antigen markers, were characterized, intracellular localization of aluminium (III) phthalocyanine chloride tetrasulphonate (AlPcS4Cl) determined and its anti-cancer PDT effects were evaluated. Results confirmed that isolated cells were stem cell-like and subcellular localization of AlPcS4Cl in integral organelles involved in cell homeostasis supported the destruction of CSC. AlPcS4Cl's effectivity was demonstrated with CSC eradication showing a significant increase in cytotoxicity and cell death via apoptosis, caused by a decrease in mitochondrial membrane potential. PDT could serve as a palliative treatment for lung cancer and improve prognosis by elimination of lung CSCs.
Collapse
Affiliation(s)
- Anine Crous
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, PO Box 17011, Johannesburg 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, PO Box 17011, Johannesburg 2028, South Africa
| |
Collapse
|
11
|
Shen YA, Chen CC, Chen BJ, Wu YT, Juan JR, Chen LY, Teng YC, Wei YH. Potential Therapies Targeting Metabolic Pathways in Cancer Stem Cells. Cells 2021; 10:1772. [PMID: 34359941 PMCID: PMC8304173 DOI: 10.3390/cells10071772] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) are heterogeneous cells with stem cell-like properties that are responsible for therapeutic resistance, recurrence, and metastasis, and are the major cause for cancer treatment failure. Since CSCs have distinct metabolic characteristics that plays an important role in cancer development and progression, targeting metabolic pathways of CSCs appears to be a promising therapeutic approach for cancer treatment. Here we classify and discuss the unique metabolisms that CSCs rely on for energy production and survival, including mitochondrial respiration, glycolysis, glutaminolysis, and fatty acid metabolism. Because of metabolic plasticity, CSCs can switch between these metabolisms to acquire energy for tumor progression in different microenvironments compare to the rest of tumor bulk. Thus, we highlight the specific conditions and factors that promote or suppress CSCs properties to portray distinct metabolic phenotypes that attribute to CSCs in common cancers. Identification and characterization of the features in these metabolisms can offer new anticancer opportunities and improve the prognosis of cancer. However, the therapeutic window of metabolic inhibitors used alone or in combination may be rather narrow due to cytotoxicity to normal cells. In this review, we present current findings of potential targets in these four metabolic pathways for the development of more effective and alternative strategies to eradicate CSCs and treat cancer more effectively in the future.
Collapse
Affiliation(s)
- Yao-An Shen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chang-Cyuan Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Bo-Jung Chen
- Department of Pathology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
| | - Yu-Ting Wu
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 50046, Taiwan;
| | - Jiun-Ru Juan
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Liang-Yun Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Yueh-Chun Teng
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Yau-Huei Wei
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 50046, Taiwan;
| |
Collapse
|
12
|
Abstract
Cancer stem cells (CSCs) are heterogeneous cells with stem cell-like properties that are responsible for therapeutic resistance, recurrence, and metastasis, and are the major cause for cancer treatment failure. Since CSCs have distinct metabolic characteristics that plays an important role in cancer development and progression, targeting metabolic pathways of CSCs appears to be a promising therapeutic approach for cancer treatment. Here we classify and discuss the unique metabolisms that CSCs rely on for energy production and survival, including mitochondrial respiration, glycolysis, glutaminolysis, and fatty acid metabolism. Because of metabolic plasticity, CSCs can switch between these metabolisms to acquire energy for tumor progression in different microenvironments compare to the rest of tumor bulk. Thus, we highlight the specific conditions and factors that promote or suppress CSCs properties to portray distinct metabolic phenotypes that attribute to CSCs in common cancers. Identification and characterization of the features in these metabolisms can offer new anticancer opportunities and improve the prognosis of cancer. However, the therapeutic window of metabolic inhibitors used alone or in combination may be rather narrow due to cytotoxicity to normal cells. In this review, we present current findings of potential targets in these four metabolic pathways for the development of more effective and alternative strategies to eradicate CSCs and treat cancer more effectively in the future.
Collapse
|
13
|
Biagioni A, Chillà A, Del Rosso M, Fibbi G, Scavone F, Andreucci E, Peppicelli S, Bianchini F, Calorini L, Li Santi A, Ragno P, Margheri F, Laurenzana A. CRISPR/Cas9 uPAR Gene Knockout Results in Tumor Growth Inhibition, EGFR Downregulation and Induction of Stemness Markers in Melanoma and Colon Carcinoma Cell Lines. Front Oncol 2021; 11:663225. [PMID: 34055629 PMCID: PMC8163229 DOI: 10.3389/fonc.2021.663225] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
uPAR is a globular protein, tethered to the cell membrane by a GPI-anchor involved in several cancer-related properties and its overexpression commonly correlates with poor prognosis and metastasis. We investigated the consequences of uPAR irreversible loss in human melanoma and colon cancer cell lines, knocking out its expression by CRISPR/Cas9. We analyzed through flow cytometry, western blotting and qPCR, the modulation of the most known cancer stem cells-associated genes and the EGFR while we observed the proliferation rate exploiting 2D and 3D cellular models. We also generated uPAR “rescue” expression cell lines as well as we promoted the expression of only its 3’UTR to demonstrate the involvement of uPAR mRNA in tumor progression. Knocking out PLAUR, uPAR-encoding gene, we observed an inhibited growth ratio unexpectedly coupled with a significant percentage of cells acquiring a stem-like phenotype. In vivo experiments demonstrated that uPAR loss completely abrogates tumorigenesis despite the gained stem-like profile. Nonetheless, we proved that the reintroduction of the 3’UTR of PLAUR gene was sufficient to restore the wild-type status validating the hypothesis that such a region may act as a “molecular sponge”. In particular miR146a, by binding PLAUR 3’ UTR region might be responsible for uPAR-dependent inhibition of EGFR expression.
Collapse
Affiliation(s)
- Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Francesca Scavone
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Anna Li Santi
- Department of Chemistry and Biology, University of Salerno, Fisciano, Italy
| | - Pia Ragno
- Department of Chemistry and Biology, University of Salerno, Fisciano, Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| |
Collapse
|
14
|
Prasad P, Ghosh S, Roy SS. Glutamine deficiency promotes stemness and chemoresistance in tumor cells through DRP1-induced mitochondrial fragmentation. Cell Mol Life Sci 2021; 78:4821-4845. [PMID: 33895866 PMCID: PMC11073067 DOI: 10.1007/s00018-021-03818-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/11/2021] [Accepted: 03/20/2021] [Indexed: 12/11/2022]
Abstract
Glutamine is essential for maintaining the TCA cycle in cancer cells yet they undergo glutamine starvation in the core of tumors. Cancer stem cells (CSCs), responsible for tumor recurrence are often found in the nutrient limiting cores. Our study uncovers the molecular basis and cellular links between glutamine deprivation and stemness in the cancer cells. We showed that glutamine is dispensable for the survival of ovarian and colon cancer cells while it is required for their proliferation. Glutamine starvation leads to the metabolic reprogramming in tumor cells with enhanced glycolysis and unaltered oxidative phosphorylation. Production of reactive oxygen species (ROS) in glutamine limiting condition induces MAPK-ERK1/2 signaling pathway to phosphorylate dynamin-related protein-1(DRP1) at Ser616. Moreover, p-DRP1 promotes mitochondrial fragmentation and enhances numbers of CD44 and CD117/CD45 positive CSCs. Besides the established features of cancer stem cells, glutamine deprivation induces perinuclear localization of fragmented mitochondria and reduction in proliferation rate which are usually observed in CSCs. Treatment with glutaminase inhibitor (L-DON) mimics the effects of glutamine starvation without altering cell survival in in vitro as well as in in vivo model. Interestingly, the combinatorial treatment of L-DON with DRP1 inhibitor (MDiVi-1) reduces the stem cell population in tumor tissue in mouse model. Collectively our data suggest that glutamine deficiency in the core of tumors can increase the cancer stem cell population and the combination therapy with MDiVi-1 and L-DON is a useful approach to reduce CSCs population in tumor.
Collapse
Affiliation(s)
- Parash Prasad
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India
| | - Sampurna Ghosh
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India
| | - Sib Sankar Roy
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India.
- Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata, 700032, India.
| |
Collapse
|
15
|
Raghav PK, Mann Z. Cancer stem cells targets and combined therapies to prevent cancer recurrence. Life Sci 2021; 277:119465. [PMID: 33831426 DOI: 10.1016/j.lfs.2021.119465] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/01/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022]
Abstract
Cancer stem cells (CSCs) control the dynamics of tumorigenesis by self-renewal ability and differentiation potential. These properties contribute towards tumor malignancy, metastasis, cellular heterogeneity, and immune escape, which are regulated by multiple signaling pathways. The CSCs are chemoresistant and cause cancer recurrence, generally recognized as a small side-population that eventually leads to tumor relapse. Despite many treatment options available, none can be considered entirely efficient due to a lack of specificity and dose limitation. This review primarily highlights the processes involved in CSCs development and maintenance. Secondly, the current effective therapies based on stem cells, cell-free therapies that involve exosomes and miRNAs, and photodynamic therapy have been discussed. Also, the inhibitors that specifically target various signaling pathways, which can be used in combination to control CSCs kinetics have been highlighted. Conclusively, this comprehensive review is a detailed study of recently developed novel treatment strategies that will facilitate in coming up with better-targeted approaches against CSCs.
Collapse
Affiliation(s)
| | - Zoya Mann
- Independent Researcher, New Delhi, India
| |
Collapse
|
16
|
Abstract
Accumulating evidence strongly indicates that the presence of cancer stem cells (CSCs) leads to the emergence of worse clinical scenarios, such as chemo- and radiotherapy resistance, metastasis, and cancer recurrence. CSCs are a highly tumorigenic population characterized by self-renewal capacity and differentiation potential. Thus, CSCs establish a hierarchical intratumor organization that enables tumor adaptation to evade the immune response and resist anticancer therapy. YY1 functions as a transcription factor, RNA-binding protein, and 3D chromatin regulator. Thus, YY1 has multiple effects and regulates several molecular processes. Emerging evidence indicates that the development of lethal YY1-mediated cancer phenotypes is associated with the presence of or enrichment in cancer stem-like cells. Therefore, it is necessary to investigate whether and to what extent YY1 regulates the CSC phenotype. Since CSCs mirror the phenotypic behavior of stem cells, we initially describe the roles played by YY1 in embryonic and adult stem cells. Next, we scrutinize evidence supporting the contributions of YY1 in CSCs from a number of various cancer types. Finally, we identify new areas for further investigation into the YY1-CSCs axis, including the participation of YY1 in the CSC niche.
Collapse
|
17
|
Wu S, Tseng IC, Huang WC, Su CW, Lai YH, Lin C, Lee AYL, Kuo CY, Su LY, Lee MC, Hsu TC, Yu CH. Establishment of an Immunocompetent Metastasis Rat Model with Hepatocyte Cancer Stem Cells. Cancers (Basel) 2020; 12:cancers12123721. [PMID: 33322441 PMCID: PMC7764036 DOI: 10.3390/cancers12123721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/30/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer mortality. Cancer stem cells (CSCs) are responsible for the maintenance, metastasis, and relapse of various tumors. The effects of CSCs on the tumorigenesis of HCC are still not fully understood, however. We have recently established two new rat HCC cell lines HTC and TW-1, which we isolated from diethylnitrosamine-induced rat liver cancer. Results showed that TW-1 expressed the genetic markers of CSCs, including CD133, GSTP1, CD44, CD90, and EpCAM. Moreover, TW-1 showed higher tolerance to sorafenib than HTC did. In addition, tumorigenesis and metastasis were observed in nude mice and wild-type rats with TW-1 xenografts. Finally, we combined highly expressed genes in TW-1/HTC with well-known biomarkers from recent HCC studies to predict HCC-related biomarkers and able to identify HCC with AUCs > 0.9 after machine learning. These results indicated that TW-1 was a novel rat CSC line, and the mice or rat models we established with TW-1 has great potential on HCC studies in the future.
Collapse
Affiliation(s)
- Semon Wu
- Department of Life Science, Chinese Culture University, Taipei 11114, Taiwan;
- Correspondence: (S.W.); (C.-H.Y.); Tel.: +886-2-2861-0511(ext. 26234) (S.W.); +886-2-66289779 (C.-H.Y.); Fax: +886-2-2862-3724 (S.W.); +886-2-66289009 (C.-H.Y.)
| | - I-Chieh Tseng
- Department of Life Science, Chinese Culture University, Taipei 11114, Taiwan;
| | - Wen-Cheng Huang
- License Biotech, Co., Ltd., Taipei 10690, Taiwan; (W.-C.H.); (C.-W.S.)
| | - Cheng-Wen Su
- License Biotech, Co., Ltd., Taipei 10690, Taiwan; (W.-C.H.); (C.-W.S.)
| | - Yu-Heng Lai
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan;
| | - Che Lin
- Department of Electrical Engineering and Graduate Institute of Communication Engineering, National Taiwan University, Taipei 10617, Taiwan;
| | - Alan Yueh-Luen Lee
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan;
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei 23142, Taiwan; (C.-Y.K.); (L.-Y.S.); (M.-C.L.)
| | - Li-Yu Su
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei 23142, Taiwan; (C.-Y.K.); (L.-Y.S.); (M.-C.L.)
| | - Ming-Cheng Lee
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei 23142, Taiwan; (C.-Y.K.); (L.-Y.S.); (M.-C.L.)
| | - Te-Cheng Hsu
- Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taipei 30013, Taiwan;
| | - Chun-Hsien Yu
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei 23142, Taiwan
- Department of Pediatrics, School of Medicine, Tzu Chi University, Hualien 97071, Taiwan
- Correspondence: (S.W.); (C.-H.Y.); Tel.: +886-2-2861-0511(ext. 26234) (S.W.); +886-2-66289779 (C.-H.Y.); Fax: +886-2-2862-3724 (S.W.); +886-2-66289009 (C.-H.Y.)
| |
Collapse
|
18
|
Tian H, Zhang B, Li L, Wang G, Li H, Zheng J. Manipulation of Mitochondrial Plasticity Changes the Metabolic Competition Between "Foe" and "Friend" During Tumor Malignant Transformation. Front Oncol 2020; 10:1692. [PMID: 32974209 PMCID: PMC7471250 DOI: 10.3389/fonc.2020.01692] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/29/2020] [Indexed: 11/16/2022] Open
Abstract
Mitochondria as the cellular energy powerhouses provide a common site for multiple metabolic reactions in order to cover energy and biomolecule demands, thus integrating the diverse metabolic pathways to endow cells with metabolic adaptation. Mitochondrial plasticity is normally regulated by mitochondrial dynamics, mitochondrial metabolism and mitochondrial biogenesis. Given that tumor cells and T cells share the metabolic similarities of survival, proliferation, expansion as well as effector function, manipulation of mitochondrial plasticity would change the metabolic competition between “foe” and “friend” during tumor malignant progression. On the one hand, for “foe” tumor cells, mitochondrial plasticity provides the enhancement of tumor metastasis and the development of resistance to‘ diverse antitumor drugs. On the other hand, for “friend” T cells, mitochondrial plasticity promotes the generation of long-term memory T (TM) cells and alleviates the exhaustion of tumor-infiltrating lymphocytes (TILs). Therefore, downregulation of mitochondrial plasticity of tumor cells through engineering tumor-targeting nanoparticles may effectively potentiate metabolic vulnerability and re-sensitize tumor to relevant therapeutic treatment. On the contrary, upregulation of mitochondrial plasticity of T cells through optimizing adoptive cellular immunotherapy (ACI) or chimeric antigen receptor (CAR)-T cell therapy would provide T cells with the robust metabolic fitness and the persistent immune function, thus blocking tumor metastasis and reoccurrence.
Collapse
Affiliation(s)
- Hui Tian
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Baofu Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Liantao Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - JunNian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
19
|
Shen FF, Dai SY, Wong NK, Deng S, Wong AST, Yang D. Mediating K +/H + Transport on Organelle Membranes to Selectively Eradicate Cancer Stem Cells with a Small Molecule. J Am Chem Soc 2020; 142:10769-10779. [PMID: 32441923 DOI: 10.1021/jacs.0c02134] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Molecules that are capable of disrupting cellular ion homeostasis offer unique opportunities to treat cancer. However, previously reported synthetic ion transporters showed limited value, as promiscuous ionic disruption caused toxicity to both healthy cells and cancer cells indiscriminately. Here we report a simple yet efficient synthetic K+ transporter that takes advantage of the endogenous subcellular pH gradient and membrane potential to site-selectively mediate K+/H+ transport on the mitochondrial and lysosomal membranes in living cells. Consequent mitochondrial and lysosomal damages enhanced cytotoxicity to chemo-resistant ovarian cancer stem cells (CSCs) via apoptosis induction and autophagy suppression with remarkable selectivity (up to 47-fold). The eradication of CSCs blunted tumor formation in mice. We believe this strategy can be exploited in the structural design and applications of next-generation synthetic cation transporters for the treatment of cancer and other diseases related to dysfunctional K+ channels.
Collapse
Affiliation(s)
- Fang-Fang Shen
- Morningside Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Sheng-Yao Dai
- Morningside Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Nai-Kei Wong
- Morningside Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China.,Department of Infectious Diseases, Shenzhen Third People's Hospital, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen 518112, China
| | - Shan Deng
- Morningside Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China.,School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Alice Sze-Tsai Wong
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Dan Yang
- Morningside Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| |
Collapse
|
20
|
Klepinin A, Zhang S, Klepinina L, Rebane-Klemm E, Terzic A, Kaambre T, Dzeja P. Adenylate Kinase and Metabolic Signaling in Cancer Cells. Front Oncol 2020; 10:660. [PMID: 32509571 PMCID: PMC7248387 DOI: 10.3389/fonc.2020.00660] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/08/2020] [Indexed: 12/23/2022] Open
Abstract
A hallmark of cancer cells is the ability to rewire their bioenergetics and metabolic signaling circuits to fuel their uncontrolled proliferation and metastasis. Adenylate kinase (AK) is the critical enzyme in the metabolic monitoring of cellular adenine nucleotide homeostasis. It also directs AK→ AMP→ AMPK signaling controlling cell cycle and proliferation, and ATP energy transfer from mitochondria to distribute energy among cellular processes. The significance of AK isoform network in the regulation of a variety of cellular processes, which include cell differentiation and motility, is rapidly growing. Adenylate kinase 2 (AK2) isoform, localized in intermembrane and intra-cristae space, is vital for mitochondria nucleotide exchange and ATP export. AK2 deficiency disrupts cell energetics, causes severe human diseases, and is embryonically lethal in mice, signifying the importance of catalyzed phosphotransfer in cellular energetics. Suppression of AK phosphotransfer and AMP generation in cancer cells and consequently signaling through AMPK could be an important factor in the initiation of cancerous transformation, unleashing uncontrolled cell cycle and growth. Evidence also builds up that shift in AK isoforms is used later by cancer cells for rewiring energy metabolism to support their high proliferation activity and tumor progression. As cell motility is an energy-consuming process, positioning of AK isoforms to increased energy consumption sites could be an essential factor to incline cancer cells to metastases. In this review, we summarize recent advances in studies of the significance of AK isoforms involved in cancer cell metabolism, metabolic signaling, metastatic potential, and a therapeutic target.
Collapse
Affiliation(s)
- Aleksandr Klepinin
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Song Zhang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ljudmila Klepinina
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Egle Rebane-Klemm
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Andre Terzic
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Tuuli Kaambre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Petras Dzeja
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
21
|
Leng Z, Li Y, Zhou G, Lv X, Ai W, Li J, Hou L. Krüppel-like factor 4 regulates stemness and mesenchymal properties of colorectal cancer stem cells through the TGF-β1/Smad/snail pathway. J Cell Mol Med 2020; 24:1866-1877. [PMID: 31830379 PMCID: PMC6991673 DOI: 10.1111/jcmm.14882] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Krüppel-like factor 4 (KLF4) was closely associated with epithelial-mesenchymal transition and stemness in colorectal cancer stem cells (CSCs)-enriched spheroid cells. Nonetheless, the underlying molecular mechanism is unclear. This study showed that KLF4 overexpression was accompanied with stemness and mesenchymal features in Lgr5+ CD44+ EpCAM+ colorectal CSCs. KLF4 knockdown suppressed stemness, mesenchymal features and activation of the TGF-β1 pathway, whereas enforced KLF4 overexpression activated TGF-β1, phosphorylation of Smad 2/3 and Snail expression, and restored stemness and mesenchymal phenotypes. Furthermore, TGF-β1 pathway inhibition invalidated KLF4-facilitated stemness and mesenchymal features without affecting KLF4 expression. The data from the current study are the first to demonstrate that KLF4 maintains stemness and mesenchymal properties through the TGF-β1/Smad/Snail pathway in Lgr5+ CD44+ EpCAM+ colorectal CSCs.
Collapse
Affiliation(s)
- Zhengwei Leng
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
- Cancer Stem Cells Research CenterAffiliated Hospital of North Sichuan Medical CollegeSichuanChina
| | - Yong Li
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
| | - Guojun Zhou
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
| | - Xiaojiang Lv
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
| | - Walden Ai
- Department of Biology, Chemistry and Environmental Health ScienceBenedict CollegeColumbiaSCUSA
| | - Jianshui Li
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
| | - Lingmi Hou
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
- Cancer Stem Cells Research CenterAffiliated Hospital of North Sichuan Medical CollegeSichuanChina
- Thyriod and Breast SurgeryAffiliated Hospital of North Sichuan Medical CollegeSichuanChina
| |
Collapse
|
22
|
Prieto I, Alarcón CR, García-Gómez R, Berdún R, Urgel T, Portero M, Pamplona R, Martínez-Ruiz A, Ruiz-Sanz JI, Ruiz-Larrea MB, Jove M, Cerdán S, Monsalve M. Metabolic adaptations in spontaneously immortalized PGC-1α knock-out mouse embryonic fibroblasts increase their oncogenic potential. Redox Biol 2019; 29:101396. [PMID: 31926622 PMCID: PMC6921228 DOI: 10.1016/j.redox.2019.101396] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022] Open
Abstract
PGC-1α controls, to a large extent, the capacity of cells to respond to changing nutritional requirements and energetic demands. The key role of metabolic reprogramming in tumor development has highlighted the potential role of PGC-1α in cancer. To investigate how loss of PGC-1α activity in primary cells impacts the oncogenic characteristics of spontaneously immortalized cells, and the mechanisms involved, we used the classic 3T3 protocol to generate spontaneously immortalized mouse embryonic fibroblasts (iMEFs) from wild-type (WT) and PGC-1α knockout (KO) mice and analyzed their oncogenic potential in vivo and in vitro. We found that PGC-1α KO iMEFs formed larger and more proliferative primary tumors than WT counterparts, and fostered the formation of lung metastasis by B16 melanoma cells. These characteristics were associated with the reduced capacity of KO iMEFs to respond to cell contact inhibition, in addition to an increased ability to form colonies in soft agar, an enhanced migratory capacity, and a reduced growth factor dependence. The mechanistic basis of this phenotype is likely associated with the observed higher levels of nuclear β-catenin and c-myc in KO iMEFs. Evaluation of the metabolic adaptations of the immortalized cell lines identified a decrease in oxidative metabolism and an increase in glycolytic flux in KO iMEFs, which were also more dependent on glutamine for their survival. Furthermore, glucose oxidation and tricarboxylic acid cycle forward flux were reduced in KO iMEF, resulting in the induction of compensatory anaplerotic pathways. Indeed, analysis of amino acid and lipid patterns supported the efficient use of tricarboxylic acid cycle intermediates to synthesize lipids and proteins to support elevated cell growth rates. All these characteristics have been observed in aggressive tumors and support a tumor suppressor role for PGC-1α, restraining metabolic adaptations in cancer.
Collapse
Affiliation(s)
- Ignacio Prieto
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
| | - Carmen Rubio Alarcón
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
| | - Raquel García-Gómez
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
| | - Rebeca Berdún
- Institut de Recerca Biomédica Lleida, Avda, Alcalde Rovira Roure 80, 25198, Lleida, Spain.
| | - Tamara Urgel
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
| | - Manuel Portero
- Institut de Recerca Biomédica Lleida, Avda, Alcalde Rovira Roure 80, 25198, Lleida, Spain.
| | - Reinald Pamplona
- Institut de Recerca Biomédica Lleida, Avda, Alcalde Rovira Roure 80, 25198, Lleida, Spain.
| | - Antonio Martínez-Ruiz
- Unidad de Ivestigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP). Maestro Vives 3, 28009, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain.
| | - José Ignacio Ruiz-Sanz
- Departamento de Fisiología, Facultad de Medicina y Enfermería, Universidad del País Vasco, Euskal Herriko Unibertsitea, Barrio Sarriena s/n, 48940, Leioa, Spain.
| | - M Begoña Ruiz-Larrea
- Departamento de Fisiología, Facultad de Medicina y Enfermería, Universidad del País Vasco, Euskal Herriko Unibertsitea, Barrio Sarriena s/n, 48940, Leioa, Spain.
| | - Mariona Jove
- Institut de Recerca Biomédica Lleida, Avda, Alcalde Rovira Roure 80, 25198, Lleida, Spain.
| | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
| | - María Monsalve
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.
| |
Collapse
|
23
|
Peng W, Hegazy AM, Jiang N, Chen X, Qi HX, Zhao XD, Pu J, Ye RR, Li RT. Identification of two mitochondrial-targeting cyclometalated iridium(III) complexes as potent anti-glioma stem cells agents. J Inorg Biochem 2019; 203:110909. [PMID: 31689591 DOI: 10.1016/j.jinorgbio.2019.110909] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/24/2019] [Accepted: 10/27/2019] [Indexed: 02/05/2023]
Abstract
Glioma stem cells (GSCs) are thought to be responsible for the recurrence and invasion of glioblastoma multiform (GBM), which have been evaluated and exploited as the therapeutic target for GBM. Cyclometalated iridium(III) complexes have been demonstrated as the potential anticancer agents, however, their antitumor efficacies against GSCs are still unknown. Herein, we investigated the antitumor activity of two cyclometalated iridium(III) complexes [Ir(ppy)2L](PF6) (Ir1) and [Ir(thpy)2L](PF6) (Ir2) (ppy = 2-phenylpyridine, thpy = 2-(2-thienyl)pyridine and L = 4,4'-Bis(hydroxymethyl)-2,2'-bipyridine) against GSCs. The results clearly indicate that Ir1 and Ir2 kill GSCs selectively with IC50 values ranging from 5.26-9.05 μM. Further mechanism research display that Ir1 and Ir2 can suppress the proliferation of GSCs, penetrate into GSCs efficiently, localize to mitochondria, and induce mitochondria-mediated apoptosis, including the loss of mitochondrial membrane (MMP), elevation of intracellular reactive oxygen species (ROS) and caspases activation. Moreover, Ir1 and Ir2 can destroy the GSCs self-renewal and unlimited proliferation capacity by affecting the GSCs colony formation. According our knowledge, this is the first study to investigate the anti-GSCs properties of cyclometalated iridium(III) complexes.
Collapse
Affiliation(s)
- Wan Peng
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Ahmed M Hegazy
- The First Department of Neurosurgery, The Second Affiliated Hospital, Kunming Medical University, Kunming 650101, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; Zoology Department, Faculty of Science, Minia University, Minia 61519, Egypt
| | - Ning Jiang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Xi Chen
- The First Department of Neurosurgery, The Second Affiliated Hospital, Kunming Medical University, Kunming 650101, China; Kunming Medical University, Kunming 650101, China
| | - Hua-Xin Qi
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Xu-Dong Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; Laboratory of Animal Tumor Models, Department of Thoracic Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jun Pu
- The First Department of Neurosurgery, The Second Affiliated Hospital, Kunming Medical University, Kunming 650101, China; Kunming Medical University, Kunming 650101, China.
| | - Rui-Rong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| |
Collapse
|
24
|
Novel therapeutic interventions in cancer treatment using protein and peptide-based targeted smart systems. Semin Cancer Biol 2019; 69:249-267. [PMID: 31442570 DOI: 10.1016/j.semcancer.2019.08.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 01/12/2023]
Abstract
Cancer, being the most prevalent and resistant disease afflicting any gender, age or social status, is the ultimate challenge for the scientific community. The new generation therapeutics for cancer management has shifted the approach to personalized/precision medicine, making use of patient- and tumor-specific markers for specifying the targeted therapies for each patient. Peptides targeting these cancer-specific signatures hold enormous potential for cancer therapy and diagnosis. The rapid advancements in the combinatorial peptide libraries served as an impetus to the development of multifunctional peptide-based materials for targeted cancer therapy. The present review outlines benefits and shortcomings of peptides as cancer therapeutics and the potential of peptide modified nanomedicines for targeted delivery of anticancer agents.
Collapse
|
25
|
Bao Z, Dai X, Wang P, Tao Y, Chai D. Capsaicin induces cytotoxicity in human osteosarcoma MG63 cells through TRPV1-dependent and -independent pathways. Cell Cycle 2019; 18:1379-1392. [PMID: 31095448 DOI: 10.1080/15384101.2019.1618119] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
An accumulating body of evidence has shown that capsaicin induces apoptosis in various tumor cells as a mechanism of its anti-tumor activity. However, the effects of capsaicin on osteosarcoma have not been studied extensively. In the current study, we explore the molecular mechanism of capsaicin-mediated tumor suppressive function in osteosarcoma. We found that capsaicin-induced apoptosis and the activation of transient receptor potential receptor vanilloid 1 (TRPV1) in a dose- and time-dependent manner in human osteosarcoma MG63 cells in vitro. Blocking TRPV1 using capsazepine attenuated the capsaicin-induced cytotoxicity, mitochondrial dysfunction, overproduction of reactive oxygen species (ROS) and decrease in superoxide dismutase (SOD) activity. In addition, the results demonstrated that capsaicin induced the activation of adenosine 5'-monophosphate-activated protein kinase (AMPK), p53 and C-jun N-terminal kinase (JNK). In addition, Compound C (antagonist of AMPK) attenuated the activation of p53, which appeared to be TRPV1 independent. Taken together, the present study suggests that capsaicin effectively causes cell death in human osteosarcoma MG63 cells via the activation of TRPV1-dependent (mitochondrial dysfunction, and overproduction of ROS and JNK) and TRPV1-independent (AMPK-p53) pathways. Thus, capsaicin may be a potential anti-osteosarcoma agent.
Collapse
Affiliation(s)
- Zhengqi Bao
- a Department of Orthopedics , The First Affiliated Hospital of Bengbu Medical College , Bengbu , China
| | - Xiusong Dai
- a Department of Orthopedics , The First Affiliated Hospital of Bengbu Medical College , Bengbu , China
| | - Peter Wang
- b Department of Biochemistry and Molecular Biology, School of Laboratory Medicine , Bengbu Medical College , Anhui , China
| | - Yisheng Tao
- c Department of Pathology , the First Affiliated Hospital of Bengbu Medical College , Bengbu , China
| | - Damin Chai
- c Department of Pathology , the First Affiliated Hospital of Bengbu Medical College , Bengbu , China
| |
Collapse
|
26
|
Wang RQ, Geng J, Sheng WJ, Liu XJ, Jiang M, Zhen YS. The ionophore antibiotic gramicidin A inhibits pancreatic cancer stem cells associated with CD47 down-regulation. Cancer Cell Int 2019; 19:145. [PMID: 31139022 PMCID: PMC6532126 DOI: 10.1186/s12935-019-0862-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/15/2019] [Indexed: 01/10/2023] Open
Abstract
Background Pancreatic cancer stem cells (CSCs), a special population of cells, renew themselves infinitely and resist to various treatment. Gramicidin A (GrA), an ionophore antibiotic derived from microorganism, can form channels across the cell membrane and disrupt cellular ionic homeostasis, leading to cell dysfunction and death. As reported, the ionophore antibiotic salinomycin (Sal) has been proved to kill CSCs effectively. Whether GrA owns the potential as a therapeutic drug for CSCs still remains unknown. This study investigated the effect of GrA on pancreatic CSCs and the mechanism. Methods Tumorsphere formation assay was performed to assess pancreatic CSCs self-renewal potential. In vitro hemolysis assay was determined to test the borderline concentration of GrA. CCK-8 assay was used to detect pancreatic cancer cell proliferation capability. Flow cytometry was performed to detect cell apoptosis and mitochondrial membrane potential. Scanning and transmission electron microscopy was used to observe ultrastructural morphological changes on cell membrane surface and mitochondria, respectively. Western blot analysis was used to determine relative protein expression levels. Immunofluorescence staining was performed to observe CD47 re-distribution. Results GrA at 0.05 μM caused tumorspheres disintegration and decrease in number of pancreatic cancer BxPC-3 and MIA PaCa-2 cells. GrA and Sal both inhibited cancer cell proliferation. The IC50 values of GrA and Sal for BxPC-3 cells were 0.025 μM and 0.363 μM; while for MIA PaCa-2 cells were 0.032 μM and 0.163 μM, respectively. Compared on equal concentrations, the efficacy of GrA was stronger than that of Sal. GrA at 0.1 μM or lower did not cause hemolysis. GrA induced ultrastructural changes, such as the decrease of microvilli-like protrusions on cell surface membrane and the swelling of mitochondria. GrA down-regulated the expression levels of CD133, CD44, and CD47; in addition, CD47 re-distribution was observed on cell surface. Moreover, GrA showed synergism with gemcitabine in suppressing cancer cell proliferation. Conclusions The study found that GrA was highly active against pancreatic CSCs. It indicates that GrA exerts inhibitory effects against pancreatic CSCs associated with CD47 down-regulation, implying that GrA might play a positive role in modulating the interaction between macrophages and tumor cells.
Collapse
Affiliation(s)
- Rui-Qi Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| | - Jing Geng
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| | - Wei-Jin Sheng
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| | - Xiu-Jun Liu
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| | - Min Jiang
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| | - Yong-Su Zhen
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| |
Collapse
|
27
|
Jafri A, Bano S, Rais J, Khan F, Shivnath N, Sharma AK, Arshad M. Phytochemical screening of Sterculia foetida seed extract for anti-oxidant, anti-microbial activity, and detection of apoptosis through reactive oxygen species (ROS) generation, mitochondrial membrane potential (MMP) decrease, and nuclear fragmentation in human osteosarcoma cells. J Histotechnol 2019. [DOI: 10.1080/01478885.2019.1592832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Asif Jafri
- Molecular Endocrinology Lab, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh, India
| | - Shabana Bano
- Protozoology Research Lab, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh, India
| | - Juhi Rais
- Molecular Endocrinology Lab, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh, India
| | - Fahad Khan
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Neelam Shivnath
- Molecular Endocrinology Lab, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh, India
| | - AK Sharma
- Protozoology Research Lab, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh, India
| | - Md Arshad
- Molecular Endocrinology Lab, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh, India
| |
Collapse
|
28
|
Mascarenhas S, Mutnuri S, Ganguly A. Silica - A trace geogenic element with emerging nephrotoxic potential. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 645:297-317. [PMID: 30029111 DOI: 10.1016/j.scitotenv.2018.07.075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/14/2018] [Accepted: 07/06/2018] [Indexed: 06/08/2023]
Abstract
Silica is a trace-geogenic compound with limited-bioavailability. It inflicts health-perils like pulmonary-silicosis and chronic kidney disease (CKD), when available via anthropogenic-disturbances. Amidst silica-imposed pathologies, pulmonary toxicological-mechanisms are well-described, ignoring the renal-pathophysiological mechanisms. Hence, the present-study aimed to elucidate cellular-cum-molecular toxicological-mechanisms underlying silica-induced renal-pathology in-vitro. Various toxicity-assessments were used to study effects of silica on the physiological-functions of HK-cells (human-kidney proximal-tubular cells - the toxin's prime target) on chronic (1-7 days) sub-toxic (80 mg/L) and toxic (100-120 mg/L) dosing. Results depicted that silica triggered dose-cum-time dependent cytotoxicity/cell-death (MTT-assay) that significantly increased on long-term dosing with ≥100 mg/L silica; establishing the nephrotoxic-potential of this dose. Contrarily, insignificant cell-death on sub-toxic (80 mg/L) dosing was attributed to rapid intracellular toxin-clearance at lower-doses preventing toxic-effects. The proximal-tubular (HK-cells) cytotoxicity was found to be primarily mediated by silica-triggered incessant oxidative-stress (elevated ROS).·This enhanced ROS inflicted severe inflammation and subsequent fibrosis, evident from increased pro-inflammatory-cum-fibrogenic cytokines generation (IL-1β, IL-2, IL-6, TNF-α and TGF-β). Simultaneously, ROS induced persistent DNA-damage (Comet-assay) that stimulated G2/M arrest for p53-mediated damage-repair, aided by checkpoint-promoter (Chk1) activation and mitotic-inducers (i.e. Cdc-25, Cdk1, cyclinB1) inhibition. However, DNA-injuries surpassed the cellular-repair, which provoked the p53-gene to induce mitochondrial-mediated apoptotic cell-death via activation of Bax, cytochrome-c and caspase-cascade (9/3). This persistent apoptotic cell-death and simultaneous incessant inflammation culminated in the development of tubular-atrophy and fibrosis, the major pathological-manifestations of CKD. These findings provided novel-insights into the pathological-mechanisms (cellular and molecular) of silica-induced CKD, inflicted on chronic toxic-dosing (≥100 mg/L).Thereby, encouraging the development of therapeutic-strategies (e.g. anti-oxidant treatment) for specific molecular-targets (e.g. ROS) to retard silica-induced CKD-progression, for reduction in the global-CKD burden.
Collapse
Affiliation(s)
- Starlaine Mascarenhas
- Department of Biological Sciences, BITS Pilani, K K Birla Goa Campus, NH 17 B, Zuarinagar, Goa 403 726, India.
| | - Srikanth Mutnuri
- Department of Biological Sciences, BITS Pilani, K K Birla Goa Campus, NH 17 B, Zuarinagar, Goa 403 726, India.
| | - Anasuya Ganguly
- Department of Biological Sciences, BITS Pilani, K K Birla Goa Campus, NH 17 B, Zuarinagar, Goa 403 726, India.
| |
Collapse
|
29
|
Laws K, Suntharalingam K. The Next Generation of Anticancer Metallopharmaceuticals: Cancer Stem Cell-Active Inorganics. Chembiochem 2018; 19:2246-2253. [PMID: 30109911 DOI: 10.1002/cbic.201800358] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Indexed: 12/17/2022]
Abstract
Cancer stem cells (CSCs) are heavily linked to fatal incidences of cancer relapse and metastasis. Conventional cancer therapies such as surgery, chemotherapy and radiation are largely futile against CSCs. Therefore, highly original approaches are needed to overcome CSCs and to provide durable, long-term clinical outcomes. Many academia- and pharmaceutical-led studies aimed at developing chemical or biological anti-CSC agents are ongoing; however, the application of inorganic compounds is rare. In this minireview, we discuss how the chemical diversity and versatility offered by metals has been harnessed to develop an unprecedented, emerging class of metallopharmaceuticals: CSC-active inorganics. A detailed account of their mechanism(s) of action is provided, and possible future directions for exploration are also put forward.
Collapse
Affiliation(s)
- Kristine Laws
- Department of Chemistry, King's College London, Trinity Street, London, SE1 1DB, UK
| | | |
Collapse
|
30
|
Sotgia F, Ozsvari B, Fiorillo M, De Francesco EM, Bonuccelli G, Lisanti MP. A mitochondrial based oncology platform for targeting cancer stem cells (CSCs): MITO-ONC-RX. Cell Cycle 2018; 17:2091-2100. [PMID: 30257595 PMCID: PMC6226227 DOI: 10.1080/15384101.2018.1515551] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Here, we wish to propose a new systematic approach to cancer therapy, based on the targeting of mitochondrial metabolism, especially in cancer stem cells (CSCs). In the future, we envision that anti-mitochondrial therapy would ultimately be practiced as an add-on to more conventional therapy, largely for the prevention of tumor recurrence and cancer metastasis. This mitochondrial based oncology platform would require a panel of FDA-approved therapeutics (e.g. Doxycycline) that can safely be used to inhibit mitochondrial OXPHOS and/or biogenesis in CSCs. In addition, new therapeutics that target mitochondria could also be developed, to optimize their ability to eradicate CSCs. Finally, in this context, mitochondrial-based biomarkers (i.e. "Mito-signatures") could be utilized as companion diagnostics, to identify high-risk cancer patients at diagnosis, facilitating the early detection of tumor recurrence and the prevention of treatment failure. In summary, we suggest that new clinical trials are warranted to test and possibly implement this emerging treatment strategy, in a variety of human cancer types. This general approach, using FDA-approved antibiotics to target mitochondria, was effective in killing CSCs originating from many different cancer types, including DCIS, breast (ER(+) and ER(-)), prostate, ovarian, lung and pancreatic cancers, as well as melanoma and glioblastoma, among others. Thus, we propose the term MITO-ONC-RX, to describe this anti-mitochondrial platform for targeting CSCs. The use of re-purposed FDA-approved drugs will undoubtedly help to accelerate the clinical evaluation of this approach, as these drugs can move directly into Phase II clinical trials, saving considerable amounts of time (10-15 y) and billions in financial resources.
Collapse
Affiliation(s)
- Federica Sotgia
- a Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC) , University of Salford , Greater Manchester , UK
| | - Bela Ozsvari
- a Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC) , University of Salford , Greater Manchester , UK
| | - Marco Fiorillo
- a Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC) , University of Salford , Greater Manchester , UK.,b Department of Pharmacy, Health and Nutritional Sciences , University of Calabria , Rende , Italy
| | - Ernestina Marianna De Francesco
- a Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC) , University of Salford , Greater Manchester , UK.,b Department of Pharmacy, Health and Nutritional Sciences , University of Calabria , Rende , Italy
| | - Gloria Bonuccelli
- a Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC) , University of Salford , Greater Manchester , UK
| | - Michael P Lisanti
- a Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC) , University of Salford , Greater Manchester , UK
| |
Collapse
|
31
|
Highly Charged, Cytotoxic, Cyclometalated Iridium(III) Complexes as Cancer Stem Cell Mitochondriotropics. Chemistry 2018; 24:15205-15210. [PMID: 30052298 DOI: 10.1002/chem.201803521] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 07/26/2018] [Indexed: 11/07/2022]
|
32
|
Cuyàs E, Verdura S, Folguera-Blasco N, Bastidas-Velez C, Martin ÁG, Alarcón T, Menendez JA. Mitostemness. Cell Cycle 2018; 17:918-926. [PMID: 29886796 DOI: 10.1080/15384101.2018.1467679] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Unraveling the key mechanisms governing the retention versus loss of the cancer stem cell (CSC) state would open new therapeutic avenues to eradicate cancer. Mitochondria are increasingly recognized key drivers in the origin and development of CSC functional traits. We here propose the new term "mitostemness" to designate the mitochondria-dependent signaling functions that, evolutionary rooted in the bacterial origin of mitochondria, regulate the maintenance of CSC self-renewal and resistance to differentiation. Mitostemness traits, namely mitonuclear communication, mitoproteome components, and mitochondrial fission/fusion dynamics, can be therapeutically exploited to target the CSC state. We briefly review the pre-clinical evidence of action of investigational compounds on mitostemness traits and discuss ongoing strategies to accelerate the clinical translation of new mitostemness drugs. The recognition that the bacterial origin of present-day mitochondria can drive decision-making signaling phenomena may open up a new therapeutic dimension against life-threatening CSCs. New therapeutics aimed to target mitochondria not only as biochemical but also as biophysical and morpho-physiological hallmarks of CSC might certainly guide improvements to cancer treatment.
Collapse
Affiliation(s)
- Elisabet Cuyàs
- a Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group , Catalan Institute of Oncology , Girona , Spain.,b Girona Biomedical Research Institute (IDIBGI) , Girona , Spain
| | - Sara Verdura
- a Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group , Catalan Institute of Oncology , Girona , Spain.,b Girona Biomedical Research Institute (IDIBGI) , Girona , Spain
| | | | | | | | - Tomás Alarcón
- c Centre de Recerca Matemàtica , Barcelona , Spain.,e Barcelona Graduate School of Mathematics (BGSMath) , Barcelona , Spain.,f ICREA , Barcelona , Spain.,g Departament de Matemàtiques , Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Javier A Menendez
- a Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group , Catalan Institute of Oncology , Girona , Spain.,b Girona Biomedical Research Institute (IDIBGI) , Girona , Spain
| |
Collapse
|
33
|
Cancer stem cells (CSCs): metabolic strategies for their identification and eradication. Biochem J 2018; 475:1611-1634. [PMID: 29743249 PMCID: PMC5941316 DOI: 10.1042/bcj20170164] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/12/2018] [Accepted: 04/12/2018] [Indexed: 02/08/2023]
Abstract
Phenotypic and functional heterogeneity is one of the most relevant features of cancer cells within different tumor types and is responsible for treatment failure. Cancer stem cells (CSCs) are a population of cells with stem cell-like properties that are considered to be the root cause of tumor heterogeneity, because of their ability to generate the full repertoire of cancer cell types. Moreover, CSCs have been invoked as the main drivers of metastatic dissemination and therapeutic resistance. As such, targeting CSCs may be a useful strategy to improve the effectiveness of classical anticancer therapies. Recently, metabolism has been considered as a relevant player in CSC biology, and indeed, oncogenic alterations trigger the metabolite-driven dissemination of CSCs. More interestingly, the action of metabolic pathways in CSC maintenance might not be merely a consequence of genomic alterations. Indeed, certain metabotypic phenotypes may play a causative role in maintaining the stem traits, acting as an orchestrator of stemness. Here, we review the current studies on the metabolic features of CSCs, focusing on the biochemical energy pathways involved in CSC maintenance and propagation. We provide a detailed overview of the plastic metabolic behavior of CSCs in response to microenvironment changes, genetic aberrations, and pharmacological stressors. In addition, we describe the potential of comprehensive metabolic approaches to identify and selectively eradicate CSCs, together with the possibility to 'force' CSCs within certain metabolic dependences, in order to effectively target such metabolic biochemical inflexibilities. Finally, we focus on targeting mitochondria to halt CSC dissemination and effectively eradicate cancer.
Collapse
|
34
|
Ma J, Salamoun J, Wipf P, Edwards R, Van Houten B, Qian W. Combination of a thioxodihydroquinazolinone with cisplatin eliminates ovarian cancer stem cell-like cells (CSC-LCs) and shows preclinical potential. Oncotarget 2017; 9:6042-6054. [PMID: 29464053 PMCID: PMC5814193 DOI: 10.18632/oncotarget.23679] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 12/08/2017] [Indexed: 12/17/2022] Open
Abstract
Cancer stem cell-like cells (CSC-LCs) contribute to drug resistance and recurrence of ovarian cancer. Strategies that can eradicate CSC-LCs are expected to substantially improve the outcome of ovarian cancer treatment. We have previously identified a class of thioxodihydroquinazolinone small molecules, which have strong synergistic antitumor activity with platinum drugs, the standard chemotherapeutic agents for ovarian cancer treatment. In the current study, using the activity of aldehyde dehydrogenase (ALDH) as a marker of CSC-LCs, we demonstrated that the combination of thioxodihydroquinazolinone compound 19 with cisplatin is able to diminish ALDH-high CSC-LC populations in both platinum-resistant ovarian cancer cell lines and primary ovarian cancer cells from metastatic ascites of a cisplatin-resistant patient. Compound 19 enhanced the accumulation of intracellular cisplatin in ALDH-high ovarian CSC-LCs. The combination of compound 19 with cisplatin was also able to reduce the sphere-forming capability of cisplatin-resistant ovarian cancer cells. Using a spheroid-based in vitro metastasis model of ovarian cancer, we demonstrated that the co-administration of compound 19 with cisplatin prevents ovarian cancer spheroid cells from attaching to substratum and spreading. In a cisplatin-resistant in vivo intraperitoneal xenograft mouse model, the combination of compound 19 with cisplatin significantly reduced tumor burden, as compared to cisplatin alone. Taken together, our study demonstrated that thioxodihydroquinazolinones represent a new class of agents that in combination with cisplatin are capable of eliminating CSC-LCs in ovarian cancer. Further development of thioxodihydroquinazolinone small molecules may yield a more effective treatment for cisplatin-resistant metastatic ovarian cancer.
Collapse
Affiliation(s)
- Jing Ma
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, and UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College of HuaZhong University of Science and Technology, Wuhan 430030, China
| | - Joseph Salamoun
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA.,Accelerated Chemical Discovery Center, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Robert Edwards
- Department of Obstetrics and Gynecology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Bennett Van Houten
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, and UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Wei Qian
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, and UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
35
|
Paolicchi E, Gemignani F, Krstic-Demonacos M, Dedhar S, Mutti L, Landi S. Targeting hypoxic response for cancer therapy. Oncotarget 2017; 7:13464-78. [PMID: 26859576 PMCID: PMC4924654 DOI: 10.18632/oncotarget.7229] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/17/2016] [Indexed: 12/21/2022] Open
Abstract
Hypoxic tumor microenvironment (HTM) is considered to promote metabolic changes, oncogene activation and epithelial mesenchymal transition, and resistance to chemo- and radio-therapy, all of which are hallmarks of aggressive tumor behavior. Cancer cells within the HTM acquire phenotypic properties that allow them to overcome the lack of energy and nutrients supply within this niche. These phenotypic properties include activation of genes regulating glycolysis, glucose transport, acidosis regulators, angiogenesis, all of which are orchestrated through the activation of the transcription factor, HIF1A, which is an independent marker of poor prognosis. Moreover, during the adaptation to a HTM cancer cells undergo deep changes in mitochondrial functions such as “Warburg effect” and the “reverse Warburg effect”. This review aims to provide an overview of the characteristics of the HTM, with particular focus on novel therapeutic strategies currently in clinical trials, targeting the adaptive response to hypoxia of cancer cells.
Collapse
Affiliation(s)
- Elisa Paolicchi
- Genetics-Department of Biology, University of Pisa, Pisa, Italy
| | | | - Marija Krstic-Demonacos
- School of Environment and Life Sciences, College of Science and Technology, University of Salford, Salford, UK
| | - Shoukat Dedhar
- Department of Integrative Oncology, BC Cancer Research Centre, BC Cancer Agency and Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Luciano Mutti
- School of Environment and Life Sciences, College of Science and Technology, University of Salford, Salford, UK
| | - Stefano Landi
- Genetics-Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
36
|
A Copper(II) Phenanthroline Metallopeptide That Targets and Disrupts Mitochondrial Function in Breast Cancer Stem Cells. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201710910] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
37
|
Laws K, Bineva-Todd G, Eskandari A, Lu C, O'Reilly N, Suntharalingam K. A Copper(II) Phenanthroline Metallopeptide That Targets and Disrupts Mitochondrial Function in Breast Cancer Stem Cells. Angew Chem Int Ed Engl 2017; 57:287-291. [DOI: 10.1002/anie.201710910] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Kristine Laws
- Department of Chemistry; King's College London; London SE1 1DB UK
| | | | - Arvin Eskandari
- Department of Chemistry; King's College London; London SE1 1DB UK
| | - Chunxin Lu
- Department of Chemistry; King's College London; London SE1 1DB UK
| | - Nicola O'Reilly
- Peptide Chemistry; The Francis Crick Institute; London NW1 1AT UK
| | | |
Collapse
|
38
|
Yenugonda V, Nomura N, Kouznetsova V, Tsigelny I, Fogal V, Nurmemmedov E, Kesari S, Babic I. A novel small molecule inhibitor of p32 mitochondrial protein overexpressed in glioma. J Transl Med 2017; 15:210. [PMID: 29047383 PMCID: PMC5648515 DOI: 10.1186/s12967-017-1312-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/06/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The mitochondrial protein p32 is a validated therapeutic target of cancer overexpressed in glioma. Therapeutic targeting of p32 with monoclonal antibody or p32-binding LyP-1 tumor-homing peptide can limit tumor growth. However, these agents do not specifically target mitochondrial-localized p32 and would not readily cross the blood-brain barrier to target p32-overexpressing gliomas. Identifying small molecule inhibitors of p32 overexpressed in cancer is a more rational therapeutic strategy. Thus, in this study we employed a pharmacophore modeling strategy to identify small molecules that could bind and inhibit mitochondrial p32. METHODS A pharmacophore model of C1q and LyP-1 peptide association with p32 was used to screen a virtual compound library. A primary screening assay for inhibitors of p32 was developed to identify compounds that could rescue p32-dependent glutamine-addicted glioma cells from glutamine withdrawal. Inhibitors from this screen were analyzed for direct binding to p32 by fluorescence polarization assay and protein thermal shift. Affect of the p32 inhibitor on glioma cell proliferation was assessed by Alamar Blue assay, and affect on metabolism was examined by measuring lactate secretion. RESULTS Identification of a hit compound (M36) validates the pharmacophore model. M36 binds directly to p32 and inhibits LyP-1 tumor homing peptide association with p32 in vitro. M36 effectively inhibits the growth of p32 overexpressing glioma cells, and sensitizes the cells to glucose depletion. CONCLUSIONS This study demonstrates a novel screening strategy to identify potential inhibitors of mitochondrial p32 protein overexpressed in glioma. High throughput screening employing this strategy has potential to identify highly selective, potent, brain-penetrant small molecules amenable for further drug development.
Collapse
Affiliation(s)
- Venkata Yenugonda
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Natsuko Nomura
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | | | - Igor Tsigelny
- University of California San Diego, La Jolla, CA, USA
| | | | - Elmar Nurmemmedov
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Santosh Kesari
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John's Health Center, Santa Monica, CA, USA.
| | - Ivan Babic
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John's Health Center, Santa Monica, CA, USA.
| |
Collapse
|
39
|
Palmirotta R, Silvestris E, D'Oronzo S, Cardascia A, Silvestris F. Ovarian cancer: Novel molecular aspects for clinical assessment. Crit Rev Oncol Hematol 2017; 117:12-29. [PMID: 28807232 DOI: 10.1016/j.critrevonc.2017.06.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/13/2017] [Accepted: 06/15/2017] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer is a very heterogeneous tumor which has been traditionally characterized according to the different histological subtypes and differentiation degree. In recent years, innovative molecular screening biotechnologies have allowed to identify further subtypes of this cancer based on gene expression profiles, mutational features, and epigenetic factors. These novel classification systems emphasizing the molecular signatures within the broad spectrum of ovarian cancer have not only allowed a more precise prognostic prediction, but also proper therapeutic strategies for specific subgroups of patients. The bulk of available scientific data and the high refinement of molecular classifications of ovarian cancers can today address the research towards innovative drugs with the adoption of targeted therapies tailored for single molecular profiles leading to a better prediction of therapeutic response. Here, we summarize the current state of knowledge on the molecular bases of ovarian cancer, from the description of its molecular subtypes derived from wide high-throughput analyses to the latest discoveries of the ovarian cancer stem cells. The latest personalized treatment options are also presented with recent advances in using PARP inhibitors, anti-angiogenic, anti-folate receptor and anti-cancer stem cells treatment approaches.
Collapse
Affiliation(s)
- Raffaele Palmirotta
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Bari, Italy
| | - Erica Silvestris
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Bari, Italy
| | - Stella D'Oronzo
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Bari, Italy
| | - Angela Cardascia
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Bari, Italy
| | - Franco Silvestris
- Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Bari, Italy.
| |
Collapse
|
40
|
Kim HK, Noh YH, Nilius B, Ko KS, Rhee BD, Kim N, Han J. Current and upcoming mitochondrial targets for cancer therapy. Semin Cancer Biol 2017. [PMID: 28627410 DOI: 10.1016/j.semcancer.2017.06.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondria are essential intracellular organelles that regulate energy metabolism, cell death, and signaling pathways that are important for cell proliferation and differentiation. Therefore, mitochondria are fundamentally implicated in cancer biology, including initiation, growth, metastasis, relapse, and acquired drug resistance. Based on these implications, mitochondria have been proposed as a major therapeutic target for cancer treatment. In addition to classical view of mitochondria in cancer biology, recent studies found novel pathophysiological roles of mitochondria in cancer. In this review, we introduce recent concepts of mitochondrial roles in cancer biology including mitochondrial DNA mutation and epigenetic modulation, energy metabolism reprogramming, mitochondrial channels, involvement in metastasis and drug resistance, and cancer stem cells. We also discuss the role of mitochondria in emerging cancer therapeutic strategies, especially cancer immunotherapy and CRISPR-Cas9 system gene therapy.
Collapse
Affiliation(s)
- Hyoung Kyu Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea; Department of Integrated Biomedical Science, College of Medicine, Inje University, Busan, Republic of Korea
| | - Yeon Hee Noh
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Bernd Nilius
- KU Leuven, Department Cell Mol Medicine, Leuven, 3000, Belgium
| | - Kyung Soo Ko
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Byoung Doo Rhee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Nari Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea.
| |
Collapse
|
41
|
Siddiqui S, Ahamad MS, Jafri A, Afzal M, Arshad M. Piperine Triggers Apoptosis of Human Oral Squamous Carcinoma Through Cell Cycle Arrest and Mitochondrial Oxidative Stress. Nutr Cancer 2017; 69:791-799. [PMID: 28426244 DOI: 10.1080/01635581.2017.1310260] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Piperine is a nitrogenous pungent substance exhibiting multifunctional pharmacological properties. However, the mechanism underlying its anticancer potential is not well elucidated in human oral squamous carcinoma (KB) cell line. The anticancer potential of piperine was evaluated through potent biomarkers viz. reactive oxygen species (ROS), cellular apoptosis, and loss of mitochondrial membrane potential (MMP). In addition, cell cycle kinetics and caspases-3 activity were also carried out to confirm anticancer activity of piperine. Results showed that various concentrations (25-300 μM) of piperine exposure reduced the cell viability of KB cells significantly (P < 0.01). Piperine induced significant (P < 0.01) dose-related increment in ROS production and nuclear condensation. Moreover, piperine stimulated cell death by inducing loss of MMP, and caspase-3 activation. Cell cycle study revealed that piperine arrested the cells in G2/M phase and decreased the DNA content. Findings of this study suggest the efficacy of piperine in inducing cell death via the decrease in MMP and ROS liberation followed by caspase-3 activation and cell cycle arrest. Further assessment of the anticancer potency of piperine is needed for anticancer drug development.
Collapse
Affiliation(s)
- Sahabjada Siddiqui
- a Molecular Endocrinology Laboratory, Department of Zoology , University of Lucknow , Lucknow , India
| | - Md Sultan Ahamad
- b Department of Zoology , Shibli National (PG) College , Azamgarh , India
| | - Asif Jafri
- a Molecular Endocrinology Laboratory, Department of Zoology , University of Lucknow , Lucknow , India
| | - Mohd Afzal
- c Human Genetics and Toxicology Laboratory, Department of Zoology , Aligarh Muslim University , Aligarh , India
| | - Md Arshad
- a Molecular Endocrinology Laboratory, Department of Zoology , University of Lucknow , Lucknow , India
| |
Collapse
|
42
|
Johnson SC, Gonzalez B, Zhang Q, Milholland B, Zhang Z, Suh Y. Network analysis of mitonuclear GWAS reveals functional networks and tissue expression profiles of disease-associated genes. Hum Genet 2017; 136:55-65. [PMID: 27704213 PMCID: PMC5214989 DOI: 10.1007/s00439-016-1736-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/28/2016] [Indexed: 11/02/2022]
Abstract
While mitochondria have been linked to many human diseases through genetic association and functional studies, the precise role of mitochondria in specific pathologies, such as cardiovascular, neurodegenerative, and metabolic diseases, is often unclear. Here, we take advantage of the catalog of human genome-wide associations, whole-genome tissue expression and expression quantitative trait loci datasets, and annotated mitochondrial proteome databases to examine the role of common genetic variation in mitonuclear genes in human disease. Through pathway-based analysis we identified distinct functional pathways and tissue expression profiles associated with each of the major human diseases. Among our most striking findings, we observe that mitonuclear genes associated with cancer are broadly expressed among human tissues and largely represent one functional process, intrinsic apoptosis, while mitonuclear genes associated with other diseases, such as neurodegenerative and metabolic diseases, show tissue-specific expression profiles and are associated with unique functional pathways. These results provide new insight into human diseases using unbiased genome-wide approaches.
Collapse
Affiliation(s)
- Simon C Johnson
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Brenda Gonzalez
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Quanwei Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Brandon Milholland
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zhengdong Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Medicine, Endocrinology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
43
|
ROS homeostasis and metabolism: a critical liaison for cancer therapy. Exp Mol Med 2016; 48:e269. [PMID: 27811934 PMCID: PMC5133371 DOI: 10.1038/emm.2016.119] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 07/27/2016] [Accepted: 08/04/2016] [Indexed: 12/17/2022] Open
Abstract
Evidence indicates that hypoxia and oxidative stress can control metabolic reprogramming of cancer cells and other cells in tumor microenvironments and that the reprogrammed metabolic pathways in cancer tissue can also alter the redox balance. Thus, important steps toward developing novel cancer therapy approaches would be to identify and modulate critical biochemical nodes that are deregulated in cancer metabolism and determine if the therapeutic efficiency can be influenced by changes in redox homeostasis in cancer tissues. In this review, we will explore the molecular mechanisms responsible for the metabolic reprogramming of tumor microenvironments, the functional modulation of which may disrupt the effects of or may be disrupted by redox homeostasis modulating cancer therapy.
Collapse
|
44
|
Lettini G, Sisinni L, Condelli V, Matassa DS, Simeon V, Maddalena F, Gemei M, Lopes E, Vita G, Del Vecchio L, Esposito F, Landriscina M. TRAP1 regulates stemness through Wnt/β-catenin pathway in human colorectal carcinoma. Cell Death Differ 2016; 23:1792-1803. [PMID: 27662365 DOI: 10.1038/cdd.2016.67] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/30/2016] [Accepted: 06/08/2016] [Indexed: 12/14/2022] Open
Abstract
Colorectal carcinoma (CRC) is a common cause of cancer-related death worldwide. Indeed, treatment failures are triggered by cancer stem cells (CSCs) that give rise to tumor repopulation upon initial remission. Thus, the role of the heat shock protein TRAP1 in stemness was investigated in CRC cell lines and human specimens, based on its involvement in colorectal carcinogenesis, through regulation of apoptosis, protein homeostasis and bioenergetics. Strikingly, co-expression between TRAP1 and stem cell markers was observed in stem cells located at the bottom of intestinal crypts and in CSCs sorted from CRC cell lines. Noteworthy, TRAP1 knockdown reduced the expression of stem cell markers and impaired colony formation, being the CSC phenotype and the anchorage-independent growth conserved in TRAP1-rich cancer cells. Consistently, the gene expression profiling of HCT116 cells showed that TRAP1 silencing results in the loss of the stem-like signature with acquisition of a more-differentiated phenotype and the downregulation of genes encoding for activating ligands and target proteins of Wnt/β-catenin pathway. Mechanistically, TRAP1 maintenance of stemness is mediated by the regulation of Wnt/β-catenin signaling, through the modulation of the expression of frizzled receptor ligands and the control of β-catenin ubiquitination/phosphorylation. Remarkably, TRAP1 is associated with higher expression of β-catenin and several Wnt/β-catenin target genes in human CRCs, thus supporting the relevance of TRAP1 regulation of β-catenin in human pathology. This study is the first demonstration that TRAP1 regulates stemness and Wnt/β-catenin pathway in CRC and provides novel landmarks in cancer biology and therapeutics.
Collapse
Affiliation(s)
- Giacomo Lettini
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Lorenza Sisinni
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Valentina Condelli
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Danilo Swann Matassa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Vittorio Simeon
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Francesca Maddalena
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Marica Gemei
- CEINGE, Biotecnologie Avanzate, University of Naples Federico II, Naples, Italy
| | - Elvira Lopes
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Giulia Vita
- Pathology Unit, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Luigi Del Vecchio
- CEINGE, Biotecnologie Avanzate, University of Naples Federico II, Naples, Italy
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Matteo Landriscina
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy.,Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
45
|
Liu X, Secombe J. The Histone Demethylase KDM5 Activates Gene Expression by Recognizing Chromatin Context through Its PHD Reader Motif. Cell Rep 2015; 13:2219-31. [PMID: 26673323 DOI: 10.1016/j.celrep.2015.11.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/05/2015] [Accepted: 10/31/2015] [Indexed: 12/22/2022] Open
Abstract
KDM5 family proteins are critically important transcriptional regulators whose physiological functions in the context of a whole animal remain largely unknown. Using genome-wide gene expression and binding analyses in Drosophila adults, we demonstrate that KDM5 (Lid) is a direct regulator of genes required for mitochondrial structure and function. Significantly, this occurs independently of KDM5's well-described JmjC domain-encoded histone demethylase activity. Instead, it requires the PHD motif of KDM5 that binds to histone H3 that is di- or trimethylated on lysine 4 (H3K4me2/3). Genome-wide, KDM5 binding overlaps with the active chromatin mark H3K4me3, and a fly strain specifically lacking H3K4me2/3 binding shows defective KDM5 promoter recruitment and gene activation. KDM5 therefore plays a central role in regulating mitochondrial function by utilizing its ability to recognize specific chromatin contexts. Importantly, KDM5-mediated regulation of mitochondrial activity is likely to be key in human diseases caused by dysfunction of this family of proteins.
Collapse
Affiliation(s)
- Xingyin Liu
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Julie Secombe
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
46
|
Tong L, Chuang CC, Wu S, Zuo L. Reactive oxygen species in redox cancer therapy. Cancer Lett 2015; 367:18-25. [DOI: 10.1016/j.canlet.2015.07.008] [Citation(s) in RCA: 285] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 07/08/2015] [Accepted: 07/10/2015] [Indexed: 12/21/2022]
|
47
|
Mitochondrial Transcription Factor A and Mitochondrial Genome as Molecular Targets for Cisplatin-Based Cancer Chemotherapy. Int J Mol Sci 2015; 16:19836-50. [PMID: 26307971 PMCID: PMC4581328 DOI: 10.3390/ijms160819836] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 07/28/2015] [Accepted: 08/07/2015] [Indexed: 12/30/2022] Open
Abstract
Mitochondria are important cellular organelles that function as control centers of the energy supply for highly proliferative cancer cells and regulate apoptosis after cancer chemotherapy. Cisplatin is one of the most important chemotherapeutic agents and a key drug in therapeutic regimens for a broad range of solid tumors. Cisplatin may directly interact with mitochondria, which can induce apoptosis. The direct interactions between cisplatin and mitochondria may account for our understanding of the clinical activity of cisplatin and development of resistance. However, the basis for the roles of mitochondria under treatment with chemotherapy is poorly understood. In this review, we present novel aspects regarding the unique characteristics of the mitochondrial genome in relation to the use of platinum-based chemotherapy and describe our recent work demonstrating the importance of the mitochondrial transcription factor A (mtTFA) expression in cancer cells.
Collapse
|