1
|
Springer CS, Pike MM, Barbara TM. Metabolic Energy is Stored in a Homeostatic Trans-Membrane Water Barochemical Gradient. J Membr Biol 2025; 258:135-160. [PMID: 40009106 DOI: 10.1007/s00232-024-00332-1] [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: 08/29/2024] [Accepted: 12/13/2024] [Indexed: 02/27/2025]
Abstract
Trans-membrane water transport and co-transport is ubiquitous in cell biology. Integrated over all the cell's H2O transporters and co-transporters, the rate of homeostatic, bidirectional trans-cytolemmal water "exchange" is synchronized with the metabolic rate of the crucial Na+,K+-ATPase (NKA) enzyme: the active trans-membrane water cycling (AWC) phenomenon. Is AWC futile, or is it consequential? Conservatively representative literature metabolomic and proteinomic results enable comprehensive free energy (ΔG) calculations for the many transport reactions with known water stoichiometries. Including established intracellular pressure (Pi) magnitudes, these reveal an outward trans-membrane H2O barochemical ΔG gradient comparable to that of the well-known inward Na+ electrochemical ΔG gradient. For most co-influxers, these two gradients are finely balanced to maintain intracellular metabolite concentration values near their consuming enzyme Michaelis constants. Our analyses include glucose, glutamate-, gamma-aminobutyric acid (GABA), and lactate- transporters. 2%-4% Pi alterations can lead to disastrous metabolite concentrations. For the neurotransmitters glutamate- and GABA, very small astrocytic Pi changes can allow/disallow synaptic transmission. Unlike the Na+ and K+ electrochemical steady-states, the H2O barochemical steady-state is in (or near) chemical equilibrium. The analyses show why the presence of aquaporins (AQPs) does not dissipate trans-membrane pressure gradients. A feedback loop inherent in the opposing Na+ electrochemical and H2O barochemical gradients regulates AQP-catalyzed water flux as integral to AWC. A re-consideration of the underlying nature of Pi is also necessary. AWC is not a futile cycle but is inherent to the cell's "NKA system"-a new, fundamental aspect of biology. Metabolic energy is stored in the trans-membrane water barochemical gradient.
Collapse
Affiliation(s)
- Charles S Springer
- Advanced Imaging Research Center, Oregon Health and Science University, 3181 S. W. Sam Jackson Park Road, L452, Portland, OR, 97239, USA.
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, USA.
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, USA.
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| | - Martin M Pike
- Advanced Imaging Research Center, Oregon Health and Science University, 3181 S. W. Sam Jackson Park Road, L452, Portland, OR, 97239, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Thomas M Barbara
- Advanced Imaging Research Center, Oregon Health and Science University, 3181 S. W. Sam Jackson Park Road, L452, Portland, OR, 97239, USA
| |
Collapse
|
2
|
Orlovskiy S, Gupta PK, Arias-Mendoza F, Singh DK, Nova S, Nelson DS, Narayan V, Koch CJ, Hardy M, You M, Kalyanaraman B, Nath K. Enhancing Radiation Therapy Response in Prostate Cancer Through Metabolic Modulation by Mito-Lonidamine: A 1H and 31P Magnetic Resonance Spectroscopy Study. Int J Mol Sci 2025; 26:509. [PMID: 39859224 PMCID: PMC11765175 DOI: 10.3390/ijms26020509] [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: 11/28/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/27/2025] Open
Abstract
Radiation therapy (RT) is the cornerstone treatment for prostate cancer; however, it frequently induces gastrointestinal and genitourinary toxicities that substantially diminish the patients' quality of life. While many individuals experience transient side effects, a subset endures persistent, long-term complications. A promising strategy to mitigate these toxicities involves enhancing tumor radiosensitivity, potentially allowing for lower radiation doses. In this context, mito-lonidamine (Mito-LND), an antineoplastic agent targeting the mitochondrial electron transport chain's complexes I and II, emerges as a potential radiosensitizer. This study investigated Mito-LND's capacity to augment RT efficacy and reduce adverse effects through comprehensive in vitro and in vivo assessments using hormone-sensitive and hormone-refractory prostate cancer models. Employing a Seahorse analysis and 1H/31P magnetic resonance spectroscopy (MRS), we observed that Mito-LND selectively suppressed lactate production, decreased intracellular pH, and reduced bioenergetics and oxygen consumption levels within tumor cells. These findings suggest that Mito-LND remodels the tumor microenvironment by inducing acidification, metabolic de-energization, and enhanced oxygenation, thereby sensitizing tumors to RT. Our results underscore the potential of Mito-LND as a therapeutic adjunct in RT to improve patient outcomes and reduce radiation-associated toxicities in early-stage prostate cancer.
Collapse
Affiliation(s)
- Stepan Orlovskiy
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (P.K.G.); (F.A.-M.); (D.K.S.); (S.N.); (D.S.N.)
| | - Pradeep Kumar Gupta
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (P.K.G.); (F.A.-M.); (D.K.S.); (S.N.); (D.S.N.)
| | - Fernando Arias-Mendoza
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (P.K.G.); (F.A.-M.); (D.K.S.); (S.N.); (D.S.N.)
- Advanced Imaging Research Inc., Cleveland, OH 44114, USA
| | - Dinesh Kumar Singh
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (P.K.G.); (F.A.-M.); (D.K.S.); (S.N.); (D.S.N.)
| | - Skyler Nova
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (P.K.G.); (F.A.-M.); (D.K.S.); (S.N.); (D.S.N.)
| | - David S. Nelson
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (P.K.G.); (F.A.-M.); (D.K.S.); (S.N.); (D.S.N.)
| | - Vivek Narayan
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Cameron J. Koch
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Micael Hardy
- Département de Chimie, Aix-Marseille University, CNRS, ICR, UMR 7273, 13013 Marseille, France;
| | - Ming You
- Center for Cancer Prevention, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA;
| | | | - Kavindra Nath
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (P.K.G.); (F.A.-M.); (D.K.S.); (S.N.); (D.S.N.)
| |
Collapse
|
3
|
Springer CS, Pike MM, Barbara TM. A Futile Cycle?: Tissue Homeostatic Trans-Membrane Water Co-Transport: Kinetics, Thermodynamics, Metabolic Consequences. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589812. [PMID: 38659823 PMCID: PMC11042311 DOI: 10.1101/2024.04.17.589812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The phenomenon of active trans-membrane water cycling (AWC) has emerged in little over a decade. Here, we consider H2O transport across cell membranes from the origins of its study. Historically, trans-membrane water transport processes were classified into: A) compensating bidirectional fluxes ("exchange"), and B) unidirectional flux ("net flow") categories. Recent literature molecular structure determinations and molecular dynamic (MD) simulations indicate probably all the many different hydrophilic substrate membrane co-transporters have membrane-spanning hydrophilic pathways and co-transport water along with their substrates, and that they individually catalyze category A and/or B water flux processes, although usually not simultaneously. The AWC name signifies that, integrated over the all the cell's co-transporters, the rate of homeostatic, bidirectional trans-cytolemmal water exchange (category A) is synchronized with the metabolic rate of the crucial Na+,K+-ATPase (NKA) enzyme. A literature survey indicates the stoichiometric (category B) water/substrate ratios of individual co-transporters are often very large. The MD simulations also suggest how different co-transporter reactions can be kinetically coupled molecularly. Is this (Na+,K+-ATPase rate-synchronized) cycling futile, or is it consequential? Conservatively representative literature metabolomic and proteinomic results enable comprehensive free energy analyses of the many transport reactions with known water stoichiometries. Free energy calculations, using literature intracellular pressure (Pi) values reveals there is an outward trans-membrane H2O barochemical gradient of magnitude comparable to that of the well-known inward Na+ electrochemical gradient. For most co-influxers, these gradients are finely balanced to maintain intracellular metabolite concentration values near their consuming enzyme Michaelis constants. The thermodynamic analyses include glucose, glutamate-, gamma-aminobutyric acid (GABA), and lactate- transporters. 2%-4% Pi alterations can lead to disastrous concentration levels. For the neurotransmitters glutamate- and GABA, very small astrocytic Pi changes can allow/disallow synaptic transmission. Unlike the Na+ and K+ electrochemical steady-states, the H2O barochemical steady-state is in (or near) chemical equilibrium. The analyses show why the presence of aquaporins (AQPs) does not dissipate the trans-membrane pressure gradient. A feedback loop inherent in the opposing Na+ electrochemical and H2O barochemical gradients regulates AQP-catalyzed water flux as an integral AWC aspect. These results also require a re-consideration of the underlying nature of Pi. Active trans-membrane water cycling is not futile, but is inherent to the cell's "NKA system" - a new, fundamental aspect of biology.
Collapse
Affiliation(s)
- Charles S Springer
- Advanced Imaging Research Center
- Department of Chemical Physiology and Biochemistry
- Department of Biomedical Engineering
- Brenden-Colson Center for Pancreatic Care
- Knight Cancer Institute, Oregon Health & Science University; Portland, Oregon
| | - Martin M Pike
- Advanced Imaging Research Center
- Department of Biomedical Engineering
- Knight Cancer Institute, Oregon Health & Science University; Portland, Oregon
| | | |
Collapse
|
4
|
Koltai T, Fliegel L. Exploring monocarboxylate transporter inhibition for cancer treatment. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:135-169. [PMID: 38464385 PMCID: PMC10918235 DOI: 10.37349/etat.2024.00210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/01/2023] [Indexed: 03/12/2024] Open
Abstract
Cells are separated from the environment by a lipid bilayer membrane that is relatively impermeable to solutes. The transport of ions and small molecules across this membrane is an essential process in cell biology and metabolism. Monocarboxylate transporters (MCTs) belong to a vast family of solute carriers (SLCs) that facilitate the transport of certain hydrophylic small compounds through the bilipid cell membrane. The existence of 446 genes that code for SLCs is the best evidence of their importance. In-depth research on MCTs is quite recent and probably promoted by their role in cancer development and progression. Importantly, it has recently been realized that these transporters represent an interesting target for cancer treatment. The search for clinically useful monocarboxylate inhibitors is an even more recent field. There is limited pre-clinical and clinical experience with new inhibitors and their precise mechanism of action is still under investigation. What is common to all of them is the inhibition of lactate transport. This review discusses the structure and function of MCTs, their participation in cancer, and old and newly developed inhibitors. Some suggestions on how to improve their anticancer effects are also discussed.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires 2199, Argentina
| | - Larry Fliegel
- Department of Biochemistry, Faculty of Medicine, University of Alberta, Edmonton T6G 2R3, Alberta, Canada
| |
Collapse
|
5
|
Oberg AL, Heinzen EP, Hou X, Al Hilli MM, Hurley RM, Wahner Hendrickson AE, Goergen KM, Larson MC, Becker MA, Eckel-Passow JE, Maurer MJ, Kaufmann SH, Haluska P, Weroha SJ. Statistical analysis of comparative tumor growth repeated measures experiments in the ovarian cancer patient derived xenograft (PDX) setting. Sci Rep 2021; 11:8076. [PMID: 33850213 PMCID: PMC8044116 DOI: 10.1038/s41598-021-87470-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
Repeated measures studies are frequently performed in patient-derived xenograft (PDX) models to evaluate drug activity or compare effectiveness of cancer treatment regimens. Linear mixed effects regression models were used to perform statistical modeling of tumor growth data. Biologically plausible structures for the covariation between repeated tumor burden measurements are explained. Graphical, tabular, and information criteria tools useful for choosing the mean model functional form and covariation structure are demonstrated in a Case Study of five PDX models comparing cancer treatments. Power calculations were performed via simulation. Linear mixed effects regression models applied to the natural log scale were shown to describe the observed data well. A straight growth function fit well for two PDX models. Three PDX models required quadratic or cubic polynomial (time squared or cubed) terms to describe delayed tumor regression or initial tumor growth followed by regression. Spatial(power), spatial(power) + RE, and RE covariance structures were found to be reasonable. Statistical power is shown as a function of sample size for different levels of variation. Linear mixed effects regression models provide a unified and flexible framework for analysis of PDX repeated measures data, use all available data, and allow estimation of tumor doubling time.
Collapse
Affiliation(s)
- Ann L Oberg
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Ethan P Heinzen
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Xiaonan Hou
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Mariam M Al Hilli
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Division of Subspecialty Care for Women's Health, Gynecologic Oncology, Women's Health Institute, Cleveland Clinic, Cleveland, OH, 44126, USA
| | - Rachel M Hurley
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Andrea E Wahner Hendrickson
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Krista M Goergen
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Melissa C Larson
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Marc A Becker
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Blueprint Medicines, 45 Sidney St., Cambridge, MA, 02139, USA
| | - Jeanette E Eckel-Passow
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Matthew J Maurer
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, 55905, MN, USA
| | - Paul Haluska
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Bristol-Meyers Squibb, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA
| | - S John Weroha
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
6
|
The Potential of Lonidamine in Combination with Chemotherapy and Physical Therapy in Cancer Treatment. Cancers (Basel) 2020; 12:cancers12113332. [PMID: 33187214 PMCID: PMC7696079 DOI: 10.3390/cancers12113332] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The unique characteristics of tumor energy metabolism (highly dependent on aerobic glycolysis, namely, the Warburg effect) make it an interesting and attractive target for drug discovery. Radio- and chemoresistance are closely associated with the Warburg effect. Lonidamine (LND), as a glycolytic inhibitor, although having low anticancer activity when used alone, exhibits selectivity to various tumors, and its adverse effects do not overlap when combined with other chemotherapeutic drugs. Therefore, LND may be very promising as a sensitizer of tumors to chemotherapeutic agents and physical therapies. This review summarizes the advance of LND in combination with chemotherapy and physical therapy over the past several decades, as well as the promising LND derivative adjudin (ADD). The underlying sensitizing mechanisms were also analyzed and discussed, which may contribute to an improved therapeutic effect in future clinical cancer treatment. Abstract Lonidamine (LND) has the ability to resist spermatogenesis and was first used as an anti-spermatogenic agent. Later, it was found that LND has a degree of anticancer activity. Currently, LND is known to target energy metabolism, mainly involving the inhibition of monocarboxylate transporter (MCT), mitochondrial pyruvate carrier (MPC), respiratory chain complex I/II, mitochondrial permeability transition (PT) pore, and hexokinase II (HK-II). However, phase II clinical studies showed that LND alone had a weak therapeutic effect, and the effect was short and reversible. Interestingly, LND does not have the common side effects of traditional chemotherapeutic drugs, such as alopecia and myelosuppression. In addition, LND has selective activity toward various tumors, and its toxic and side effects do not overlap when combined with other chemotherapeutic drugs. Therefore, LND is commonly used as a chemosensitizer to enhance the antitumor effects of chemotherapeutic drugs based on its disruption of energy metabolism relating to chemo- or radioresistance. In this review, we summarized the combination treatments of LND with several typical chemotherapeutic drugs and several common physical therapies, such as radiotherapy (RT), hyperthermia (HT), and photodynamic therapy (PDT), and discussed the underlying mechanisms of action. Meanwhile, the development of novel formulations of LND in recent years and the research progress of LND derivative adjudin (ADD) as an anticancer drug were also discussed.
Collapse
|
7
|
Muhammad N, Tan CP, Nawaz U, Wang J, Wang FX, Nasreen S, Ji LN, Mao ZW. Multiaction Platinum(IV) Prodrug Containing Thymidylate Synthase Inhibitor and Metabolic Modifier against Triple-Negative Breast Cancer. Inorg Chem 2020; 59:12632-12642. [PMID: 32838518 DOI: 10.1021/acs.inorgchem.0c01736] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multifunctional platinumIV anticancer prodrugs have the potential to enrich the anticancer properties and overcome the clinical problems of drug resistance and side effects of platinumII anticancer agents. Herein, we develop dual and triple action platinumIV complexes with targeted and biological active functionalities. One complex (PFL) that consists of cisplatin, tegafur, and lonidamine exhibits strong cytotoxicity against triple negative breast cancer (TNBC) cells. Cellular uptake and distribution studies reveal that PFL mainly accumulates in mitochondria. As a result, PFL disrupts the mitochondrial ultrastructure and induces significant alterations in the mitochondrial membrane potential, which further leads to an increase in production of reactive oxygen species (ROS) and a decrease in ATP synthesis in MDA-MB-231 TNBCs. Western blot analysis reveals the formation of ternary complex of thymidylate synthase, which shows the intracellular conversion of tegafur into 5-FU after its release from PFL. Furthermore, treatment with PFL impairs the mitochondrial function, leading to the inhibition of glycolysis and mitochondrial respiration and induction of apoptosis through the mitochondrial pathway. The RNA-sequencing experiment shows that PFL can perturb the pathways involved in DNA synthesis, DNA damage, metabolism, and transcriptional activity. These findings demonstrate that PFL intervenes in several cellular processes including DNA damage, thymidylate synthase inhibition, and perturbation of the mitochondrial bioenergetics to kill the cancer cells. The results highlight the significance of a triple-action prodrug for efficient anticancer therapy for TNBCs.
Collapse
Affiliation(s)
- Nafees Muhammad
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Uroosa Nawaz
- Department of Surgery, P.O.F. Hospital, Wah Cantt 47040, Pakistan
| | - Jie Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Fang-Xin Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Sadia Nasreen
- Department of Environmental Engineering, University of Engineering & Technology (UET), Taxila 47080, Pakistan
| | - Liang-Nian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, P. R. China
| |
Collapse
|
8
|
Abstract
PURPOSE Fluorescence of co-enzyme reduced nicotinamide adenine dinucleotide (NADH) and oxidized flavoproteins (Fp) provides a sensitive measure of the mitochondrial redox state and cellular metabolism. By imaging NADH and Fp, we investigated the utility of optical redox imaging (ORI) to monitor cellular metabolism and detect early metabolic response to cancer drugs. PROCEDURES We performed ORI of human melanoma DB-1 cells in culture and DB-1 mouse xenografts to detect the redox response to lonidamine (LND) treatment. RESULTS For cultured cells, LND treatment for 45 min significantly lowered NADH levels with no significant change in Fp, resulting in a significant increase in the Fp redox ratio (Fp/(NADH+Fp)); 3-h prolonged treatment led to a decrease in NADH and an increase in Fp and a more oxidized redox state compared to control. Significant decrease in the mitochondrial redox capacity of LND-treated cells was observed for the first time. For xenografts, 45-min LND treatment resulted in a significant reduction of NADH content, no significant changes in Fp content, and a trend of increase in the Fp redox ratio. Intratumor redox heterogeneity was observed in both control and LND-treated groups. CONCLUSION Our results support the utility of ORI for evaluating cellular metabolism and monitoring early metabolic response to cancer drugs.
Collapse
|
9
|
Cohen-Erez I, Issacson C, Lavi Y, Shaco-Levy R, Milam J, Laster B, Gheber LA, Rapaport H. Antitumor Effect of Lonidamine-Polypeptide-Peptide Nanoparticles in Breast Cancer Models. ACS APPLIED MATERIALS & INTERFACES 2019; 11:32670-32678. [PMID: 31414594 DOI: 10.1021/acsami.9b09886] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Biomaterials folded into nanoparticles (NPs) can be utilized as targeted drug delivery systems for cancer therapy. NPs may provide a vehicle for the anticancer drug lonidamine (LND), which inhibits glycolysis but was suspended from use at the clinical trial stage because of its hepatotoxicity due to poor solubility and pharmacokinetic properties. The NPs prepared by coassembly of the anionic polypeptide poly gamma glutamic acid (γ-PGA) and a designed amphiphilic and positively charged peptide (designated as mPoP-NPs) delivered LND to the mitochondria in cell cultures. In this study, we demonstrate that LND-mPoP-NP effective drug concentrations can be increased to reach therapeutically relevant concentrations. The self-assembled NP solution was subjected to snap-freezing and lyophilization and the resultant powder was redissolved in a tenth of the original volume. The NP size and their ability to target the proximity of the mitochondria of breast cancer cells were both maintained in this new formulation, C-LND-mPoP-NPs. Furthermore, these NPs exhibited 40% better cytotoxicity, relative to the nonlyophilized LND-mPoP-NPs and led to tumor growth inhibition with no adverse side effects upon intravenous administration in a xenograft breast cancer murine model.
Collapse
Affiliation(s)
| | | | | | - Ruthy Shaco-Levy
- Pathology Institute , Soroka Medical Center , Beer-Sheva 84105 , Israel
| | | | | | | | | |
Collapse
|
10
|
Effect of Differences in Metabolic Activity of Melanoma Models on Response to Lonidamine plus Doxorubicin. Sci Rep 2018; 8:14654. [PMID: 30279592 PMCID: PMC6168452 DOI: 10.1038/s41598-018-33019-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/19/2018] [Indexed: 11/16/2022] Open
Abstract
Lonidamine (LND), a metabolic modulator, sensitizes DB-1 human melanoma to doxorubicin (DOX) chemotherapy by acidifying and de-energizing the tumor. This report compares the effects of LND on two human melanoma lines, DB-1 and WM983B, which exhibit different metabolic properties. Using liquid chromatography mass spectrometry and Seahorse analysis, we show that DB-1 was more glycolytic than WM983B in vitro. 31P magnetic resonance spectroscopy (MRS) indicates that LND (100 mg/kg, i.p.) induces similar selective acidification and de-energization of WM983B xenografts in immunosuppressed mice. Over three hours, intracellular pH (pHi) of WM983B decreased from 6.91 ± 0.03 to 6.59 ± 0.10 (p = 0.03), whereas extracellular pH (pHe) of this tumor changed from 7.03 ± 0.05 to 6.89 ± 0.06 (p = 0.19). A decline in bioenergetics (β-NTP/Pi) of 55 ± 5.0% (p = 0.03) accompanied the decline in pHi of WM983B. Using 1H MRS with a selective multiquantum pulse sequence and Hadamard localization, we show that LND induced a significant increase in tumor lactate levels (p < 0.01). LND pre-treatment followed by DOX (10 mg/kg, i.v.) produced a growth delay of 13.7 days in WM983B (p < 0.01 versus control), a growth delay significantly smaller than the 25.4 days that occurred with DB-1 (p = 0.03 versus WM983B). Differences in relative levels of glycolysis may produce differential therapeutic responses of DB-1 and WM983B melanomas.
Collapse
|
11
|
Synthesis, antitumor evaluation and molecular docking study of a novel podophyllotoxin-lonidamine hybrid. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2230-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
12
|
Guerra F, Arbini AA, Moro L. Mitochondria and cancer chemoresistance. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:686-699. [DOI: 10.1016/j.bbabio.2017.01.012] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 01/07/2023]
|
13
|
White KA, Grillo-Hill BK, Barber DL. Cancer cell behaviors mediated by dysregulated pH dynamics at a glance. J Cell Sci 2017; 130:663-669. [PMID: 28202602 PMCID: PMC5339414 DOI: 10.1242/jcs.195297] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dysregulated pH is a common characteristic of cancer cells, as they have an increased intracellular pH (pHi) and a decreased extracellular pH (pHe) compared with normal cells. Recent work has expanded our knowledge of how dysregulated pH dynamics influences cancer cell behaviors, including proliferation, metastasis, metabolic adaptation and tumorigenesis. Emerging data suggest that the dysregulated pH of cancers enables these specific cell behaviors by altering the structure and function of selective pH-sensitive proteins, termed pH sensors. Recent findings also show that, by blocking pHi increases, cancer cell behaviors can be attenuated. This suggests ion transporter inhibition as an effective therapeutic approach, either singly or in combination with targeted therapies. In this Cell Science at a Glance article and accompanying poster, we highlight the interconnected roles of dysregulated pH dynamics in cancer initiation, progression and adaptation.
Collapse
Affiliation(s)
- Katharine A White
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Bree K Grillo-Hill
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
14
|
Nath K, Guo L, Nancolas B, Nelson DS, Shestov AA, Lee SC, Roman J, Zhou R, Leeper DB, Halestrap AP, Blair IA, Glickson JD. Mechanism of antineoplastic activity of lonidamine. Biochim Biophys Acta Rev Cancer 2016; 1866:151-162. [PMID: 27497601 DOI: 10.1016/j.bbcan.2016.08.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/26/2016] [Accepted: 08/03/2016] [Indexed: 12/19/2022]
Abstract
Lonidamine (LND) was initially introduced as an antispermatogenic agent. It was later found to have anticancer activity sensitizing tumors to chemo-, radio-, and photodynamic-therapy and hyperthermia. Although the mechanism of action remained unclear, LND treatment has been known to target metabolic pathways in cancer cells. It has been reported to alter the bioenergetics of tumor cells by inhibiting glycolysis and mitochondrial respiration, while indirect evidence suggested that it also inhibited l-lactic acid efflux from cells mediated by members of the proton-linked monocarboxylate transporter (MCT) family and also pyruvate uptake into the mitochondria by the mitochondrial pyruvate carrier (MPC). Recent studies have demonstrated that LND potently inhibits MPC activity in isolated rat liver mitochondria (Ki 2.5μM) and cooperatively inhibits l-lactate transport by MCT1, MCT2 and MCT4 expressed in Xenopus laevis oocytes with K0.5 and Hill coefficient values of 36-40μM and 1.65-1.85, respectively. In rat heart mitochondria LND inhibited the MPC with similar potency and uncoupled oxidation of pyruvate was inhibited more effectively (IC50~7μM) than other substrates including glutamate (IC50~20μM). LND inhibits the succinate-ubiquinone reductase activity of respiratory Complex II without fully blocking succinate dehydrogenase activity. LND also induces cellular reactive oxygen species through Complex II and has been reported to promote cell death by suppression of the pentose phosphate pathway, which resulted in inhibition of NADPH and glutathione generation. We conclude that MPC inhibition is the most sensitive anti-tumour target for LND, with additional inhibitory effects on MCT-mediated l-lactic acid efflux, Complex II and glutamine/glutamate oxidation.
Collapse
Affiliation(s)
- Kavindra Nath
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Lili Guo
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Bethany Nancolas
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD, UK
| | - David S Nelson
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Alexander A Shestov
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Seung-Cheol Lee
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jeffrey Roman
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Rong Zhou
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Dennis B Leeper
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrew P Halestrap
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD, UK
| | - Ian A Blair
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jerry D Glickson
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
Nath K, Nelson DS, Putt ME, Leeper DB, Garman B, Nathanson KL, Glickson JD. Comparison of the Lonidamine Potentiated Effect of Nitrogen Mustard Alkylating Agents on the Systemic Treatment of DB-1 Human Melanoma Xenografts in Mice. PLoS One 2016; 11:e0157125. [PMID: 27285585 PMCID: PMC4902256 DOI: 10.1371/journal.pone.0157125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 05/25/2016] [Indexed: 11/18/2022] Open
Abstract
Previous NMR studies demonstrated that lonidamine (LND) selectively diminishes the intracellular pH (pHi) of DB-1 melanoma and mouse xenografts of a variety of other prevalent human cancers while decreasing their bioenergetic status (tumor βNTP/Pi ratio) and enhancing the activities of melphalan and doxorubicin in these cancer models. Since melphalan and doxorubicin are highly toxic agents, we have examined three other nitrogen (N)-mustards, chlorambucil, cyclophosphamide and bendamustine, to determine if they exhibit similar potentiation by LND. As single agents LND, melphalan and these N-mustards exhibited the following activities in DB-1 melanoma xenografts; LND: 100% tumor surviving fraction (SF); chlorambucil: 100% SF; cyclophosphamide: 100% SF; bendamustine: 79% SF; melphalan: 41% SF. When combined with LND administered 40 min prior to administration of the N-mustard (to maximize intracellular acidification) the following responses were obtained; chlorambucil: 62% SF; cyclophosphamide: 42% SF; bendamustine: 36% SF; melphalan: 10% SF. The effect of LND on the activities of these N-mustards is generally attributed to acid stabilization of the aziridinium active intermediate, acid inhibition of glutathione-S-transferase, which acts as a scavenger of aziridinium, and acid inhibition of DNA repair by O6-alkyltransferase. Depletion of ATP by LND may also decrease multidrug resistance and increase tumor response. At similar maximum tolerated doses, our data indicate that melphalan is the most effective N-mustard in combination with LND when treating DB-1 melanoma in mice, but the choice of N-mustard for coadministration with LND will also depend on the relative toxicities of these agents, and remains to be determined.
Collapse
Affiliation(s)
- Kavindra Nath
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - David S. Nelson
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mary E. Putt
- Biostatistics & Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Dennis B. Leeper
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Bradley Garman
- Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Katherine L. Nathanson
- Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jerry D. Glickson
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
16
|
Nancolas B, Guo L, Zhou R, Nath K, Nelson DS, Leeper DB, Blair IA, Glickson JD, Halestrap AP. The anti-tumour agent lonidamine is a potent inhibitor of the mitochondrial pyruvate carrier and plasma membrane monocarboxylate transporters. Biochem J 2016; 473:929-36. [PMID: 26831515 PMCID: PMC4814305 DOI: 10.1042/bj20151120] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/01/2016] [Indexed: 12/25/2022]
Abstract
Lonidamine (LND) is an anti-tumour drug particularly effective at selectively sensitizing tumours to chemotherapy, hyperthermia and radiotherapy, although its precise mode of action remains unclear. It has been reported to perturb the bioenergetics of cells by inhibiting glycolysis and mitochondrial respiration, whereas indirect evidence suggests it may also inhibit L-lactic acid efflux from cells mediated by members of the proton-linked monocarboxylate transporter (MCT) family and also pyruvate uptake into the mitochondria by the mitochondrial pyruvate carrier (MPC). In the present study, we test these possibilities directly. We demonstrate that LND potently inhibits MPC activity in isolated rat liver mitochondria (Ki2.5 μM) and co-operatively inhibits L-lactate transport by MCT1, MCT2 and MCT4 expressed in Xenopus laevisoocytes with K0.5 and Hill coefficient values of 36-40 μM and 1.65-1.85 respectively. In rat heart mitochondria LND inhibited the MPC with similar potency and uncoupled oxidation of pyruvate was inhibited more effectively (IC50~ 7 μM) than other substrates including glutamate (IC50~ 20 μM). In isolated DB-1 melanoma cells 1-10 μM LND increased L-lactate output, consistent with MPC inhibition, but higher concentrations (150 μM) decreased L-lactate output whereas increasing intracellular [L-lactate] > 5-fold, consistent with MCT inhibition. We conclude that MPC inhibition is the most sensitive anti-tumour target for LND, with additional inhibitory effects on MCT-mediated L-lactic acid efflux and glutamine/glutamate oxidation. Together these actions can account for published data on the selective tumour effects of LND onL-lactate, intracellular pH (pHi) and ATP levels that can be partially mimicked by the established MPC and MCT inhibitor α-cyano-4-hydroxycinnamate (CHC).
Collapse
Affiliation(s)
- Bethany Nancolas
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, U.K
| | - Lili Guo
- Penn Superfund Research and Training Program Center, Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, U.S.A
| | - Rong Zhou
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania School of Medicine, B6 Blockley Hall, Philadelphia, PA 19104, U.S.A
| | - Kavindra Nath
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania School of Medicine, B6 Blockley Hall, Philadelphia, PA 19104, U.S.A
| | - David S Nelson
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania School of Medicine, B6 Blockley Hall, Philadelphia, PA 19104, U.S.A
| | - Dennis B Leeper
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, U.S.A
| | - Ian A Blair
- Penn Superfund Research and Training Program Center, Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, U.S.A
| | - Jerry D Glickson
- Laboratory of Molecular Imaging, Department of Radiology, University of Pennsylvania School of Medicine, B6 Blockley Hall, Philadelphia, PA 19104, U.S.A
| | - Andrew P Halestrap
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, U.K.
| |
Collapse
|
17
|
Dewhirst MW, Lee CT, Ashcraft KA. The future of biology in driving the field of hyperthermia. Int J Hyperthermia 2016; 32:4-13. [DOI: 10.3109/02656736.2015.1091093] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|