1
|
Garcés P, Amaro A, Montecino M, van Zundert B. Inorganic polyphosphate: from basic research to diagnostic and therapeutic opportunities in ALS/FTD. Biochem Soc Trans 2024; 52:123-135. [PMID: 38323662 DOI: 10.1042/bst20230257] [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: 11/22/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/08/2024]
Abstract
Inorganic polyphosphate (polyP) is a simple, negatively charged biopolymer with chain lengths ranging from just a few to over a thousand ortho-phosphate (Pi) residues. polyP is detected in every cell type across all organisms in nature thus far analyzed. Despite its structural simplicity, polyP has been shown to play important roles in a remarkably broad spectrum of biological processes, including blood coagulation, bone mineralization and inflammation. Furthermore, polyP has been implicated in brain function and the neurodegenerative diseases amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Alzheimer's disease and Parkinson's disease. In this review, we first address the challenges associated with identifying mammalian polyP metabolizing enzymes, such as Nudt3, and quantifying polyP levels in brain tissue, cultured neural cells and cerebrospinal fluid. Subsequently, we focus on recent studies that unveil how the excessive release of polyP by human and mouse ALS/FTD astrocytes contributes to these devastating diseases by inducing hyperexcitability, leading to motoneuron death. Potential implications of elevated polyP levels in ALS/FTD patients for innovative diagnostic and therapeutic approaches are explored. It is emphasized, however, that caution is required in targeting polyP in the brain due to its diverse physiological functions, serving as an energy source, a chelator for divalent cations and a scaffold for amyloidogenic proteins. Reducing polyP levels, especially in neurons, might thus have adverse effects in brain functioning. Finally, we discuss how activated mast cells and platelets also can significantly contribute to ALS progression, as they can massively release polyP.
Collapse
Affiliation(s)
- Polett Garcés
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences (ICB), Universidad Andres Bello, Santiago, Chile
| | - Armando Amaro
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences (ICB), Universidad Andres Bello, Santiago, Chile
| | - Martin Montecino
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences (ICB), Universidad Andres Bello, Santiago, Chile
- Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Brigitte van Zundert
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences (ICB), Universidad Andres Bello, Santiago, Chile
- Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
- Department of Neurology, University of Massachusetts Chan Medical School (UMMS), Worcester, MA, U.S.A
| |
Collapse
|
2
|
Jesus RLC, Araujo FA, Alves QL, Dourado KC, Silva DF. Targeting temperature-sensitive transient receptor potential channels in hypertension: far beyond the perception of hot and cold. J Hypertens 2023; 41:1351-1370. [PMID: 37334542 DOI: 10.1097/hjh.0000000000003487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Transient receptor potential (TRP) channels are nonselective cation channels and participate in various physiological roles. Thus, changes in TRP channel function or expression have been linked to several disorders. Among the many TRP channel subtypes, the TRP ankyrin type 1 (TRPA1), TRP melastatin type 8 (TRPM8), and TRP vanilloid type 1 (TRPV1) channels are temperature-sensitive and recognized as thermo-TRPs, which are expressed in the primary afferent nerve. Thermal stimuli are converted into neuronal activity. Several studies have described the expression of TRPA1, TRPM8, and TRPV1 in the cardiovascular system, where these channels can modulate physiological and pathological conditions, including hypertension. This review provides a complete understanding of the functional role of the opposing thermo-receptors TRPA1/TRPM8/TRPV1 in hypertension and a more comprehensive appreciation of TRPA1/TRPM8/TRPV1-dependent mechanisms involved in hypertension. These channels varied activation and inactivation have revealed a signaling pathway that may lead to innovative future treatment options for hypertension and correlated vascular diseases.
Collapse
Affiliation(s)
- Rafael Leonne C Jesus
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Fênix A Araujo
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Bahia, Brazil
| | - Quiara L Alves
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Keina C Dourado
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Darizy F Silva
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Bahia, Brazil
| |
Collapse
|
3
|
Ciaglia T, Vestuto V, Bertamino A, González-Muñiz R, Gómez-Monterrey I. On the modulation of TRPM channels: Current perspectives and anticancer therapeutic implications. Front Oncol 2023; 12:1065935. [PMID: 36844925 PMCID: PMC9948629 DOI: 10.3389/fonc.2022.1065935] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/15/2022] [Indexed: 02/11/2023] Open
Abstract
The transient melastatin receptor potential (TRPM) ion channel subfamily functions as cellular sensors and transducers of critical biological signal pathways by regulating ion homeostasis. Some members of TRPM have been cloned from cancerous tissues, and their abnormal expressions in various solid malignancies have been correlated with cancer cell growth, survival, or death. Recent evidence also highlights the mechanisms underlying the role of TRPMs in tumor epithelial-mesenchymal transition (EMT), autophagy, and cancer metabolic reprogramming. These implications support TRPM channels as potential molecular targets and their modulation as an innovative therapeutic approach against cancer. Here, we discuss the general characteristics of the different TRPMs, focusing on current knowledge about the connection between TRPM channels and critical features of cancer. We also cover TRPM modulators used as pharmaceutical tools in biological trials and an indication of the only clinical trial with a TRPM modulator about cancer. To conclude, the authors describe the prospects for TRPM channels in oncology.
Collapse
Affiliation(s)
- Tania Ciaglia
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | - Vincenzo Vestuto
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | - Alessia Bertamino
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | | | | |
Collapse
|
4
|
Plaza‐Cayón A, González‐Muñiz R, Martín‐Martínez M. Mutations of TRPM8 channels: Unraveling the molecular basis of activation by cold and ligands. Med Res Rev 2022; 42:2168-2203. [PMID: 35976012 PMCID: PMC9805079 DOI: 10.1002/med.21920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 01/09/2023]
Abstract
The cation nonselective channel TRPM8 is activated by multiple stimuli, including moderate cold and various chemical compounds (i.e., menthol and icilin [Fig. 1], among others). While research continues growing on the understanding of the physiological involvement of TRPM8 channels and their role in various pathological states, the information available on its activation mechanisms has also increased, supported by mutagenesis and structural studies. This review compiles known information on specific mutations of channel residues and their consequences on channel viability and function. Besides, the comparison of sequence of animals living in different environments, together with chimera and mutagenesis studies are helping to unravel the mechanism of adaptation to different temperatures. The results of mutagenesis studies, grouped by different channel regions, are compared with the current knowledge of TRPM8 structures obtained by cryo-electron microscopy. Trying to make this review self-explicative and highly informative, important residues for TRPM8 function are summarized in a figure, and mutants, deletions and chimeras are compiled in a table, including also the observed effects by different methods of activation and the corresponding references. The information provided by this review may also help in the design of new ligands for TRPM8, an interesting biological target for therapeutic intervention.
Collapse
|
5
|
Synthesis, structural and spectroscopic characterization, in silico study, and antinociceptive effect in adult zebrafish of 2-(4-isobutylphenyl) -N'-phenylpropanohydrazide. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
6
|
Izquierdo C, Martín-Martínez M, Gómez-Monterrey I, González-Muñiz R. TRPM8 Channels: Advances in Structural Studies and Pharmacological Modulation. Int J Mol Sci 2021; 22:ijms22168502. [PMID: 34445208 PMCID: PMC8395166 DOI: 10.3390/ijms22168502] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
The transient receptor potential melastatin subtype 8 (TRPM8) is a cold sensor in humans, activated by low temperatures (>10, <28 °C), but also a polymodal ion channel, stimulated by voltage, pressure, cooling compounds (menthol, icilin), and hyperosmolarity. An increased number of experimental results indicate the implication of TRPM8 channels in cold thermal transduction and pain detection, transmission, and maintenance in different tissues and organs. These channels also have a repercussion on different kinds of life-threatening tumors and other pathologies, which include urinary and respiratory tract dysfunctions, dry eye disease, and obesity. This compendium firstly covers newly described papers on the expression of TRPM8 channels and their correlation with pathological states. An overview on the structural knowledge, after cryo-electron microscopy success in solving different TRPM8 structures, as well as some insights obtained from mutagenesis studies, will follow. Most recently described families of TRPM8 modulators are also covered, along with a section of molecules that have reached clinical trials. To finalize, authors provide an outline of the potential prospects in the TRPM8 field.
Collapse
Affiliation(s)
- Carolina Izquierdo
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
- Programa de Doctorado en Química Orgánica, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Mercedes Martín-Martínez
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
| | - Isabel Gómez-Monterrey
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
- Correspondence: (I.G.-M.); (R.G.-M.)
| | - Rosario González-Muñiz
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
- Correspondence: (I.G.-M.); (R.G.-M.)
| |
Collapse
|
7
|
Amodeo GF, Pavlov EV. Amyloid β, α-synuclein and the c subunit of the ATP synthase: Can these peptides reveal an amyloidogenic pathway of the permeability transition pore? BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183531. [PMID: 33309700 DOI: 10.1016/j.bbamem.2020.183531] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/26/2020] [Accepted: 11/09/2020] [Indexed: 01/29/2023]
Abstract
Mitochondrial Permeability Transition (PT) is a phenomenon of increased permeability of the inner mitochondrial membrane in response to high levels of Ca2+ and/or reactive oxygen species (ROS) in the matrix. PT occurs upon the opening of a pore, namely the permeability transition pore (PTP), which dissipates the membrane potential uncoupling the respiratory chain. mPT activation and PTP formation can occur through multiple molecular pathways. The specific focus of this review is to discuss the possible molecular mechanisms of PTP that involve the participation of mitochondrially targeted amyloid peptides Aβ, α-synuclein and c subunit of the ATP synthase (ATPase). As activators of PTP, amyloid peptides are uniquely different from other activators because they are capable of forming channels in lipid bilayers. This property rises the possibility that in this permeabilization pathway the formation of the channel involves the direct participation of peptides, making it uniquely different from other PTP induction mechanisms. In this pathway, a critical step of PTP activation involves the import of amyloidogenic peptides from the cytosol into the matrix. In the matrix these peptides, which would fold into α-helical structure in native conditions, interact with cyclophilin D (CypD) and upon stimulation by elevated ROS and/or the Ca2+ spontaneously misfold into β-sheet ion conducting pores, causing PTP opening.
Collapse
Affiliation(s)
- Giuseppe F Amodeo
- Department of Molecular Pathobiology, New York University, United States of America.
| | - Evgeny V Pavlov
- Department of Molecular Pathobiology, New York University, United States of America.
| |
Collapse
|
8
|
Mohandass A, Krishnan V, Gribkova ED, Asuthkar S, Baskaran P, Nersesyan Y, Hussain Z, Wise LM, George RE, Stokes N, Alexander BM, Cohen AM, Pavlov EV, Llano DA, Zhu MX, Thyagarajan B, Zakharian E. TRPM8 as the rapid testosterone signaling receptor: Implications in the regulation of dimorphic sexual and social behaviors. FASEB J 2020; 34:10887-10906. [PMID: 32609392 PMCID: PMC7496617 DOI: 10.1096/fj.202000794r] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/20/2020] [Accepted: 06/08/2020] [Indexed: 01/19/2023]
Abstract
Testosterone regulates dimorphic sexual behaviors in all vertebrates. However, the molecular mechanism underlying these behaviors remains unclear. Here, we report that a newly identified rapid testosterone signaling receptor, Transient Receptor Potential Melastatin 8 (TRPM8), regulates dimorphic sexual and social behaviors in mice. We found that, along with higher steroid levels in the circulation, TRPM8-/- male mice exhibit increased mounting frequency indiscriminate of sex, delayed sexual satiety, and increased aggression compared to wild-type controls, while TRPM8-/- females display an increased olfaction-exploratory behavior. Furthermore, neuronal responses to acute testosterone application onto the amygdala were attenuated in TRPM8-/- males but remained unchanged in females. Moreover, activation of dopaminergic neurons in the ventral tegmental area following mating was impaired in TRPM8-/- males. Together, these results demonstrate that TRPM8 regulates dimorphic sexual and social behaviors, and potentially constitutes a signalosome for mediation of sex-reward mechanism in males. Thus, deficiency of TRPM8 might lead to a delayed sexual satiety phenomenon.
Collapse
Affiliation(s)
- Adithya Mohandass
- College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Vivek Krishnan
- College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Ekaterina D Gribkova
- Department of Molecular and Integrative Physiology, Neuroscience Program and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Swapna Asuthkar
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Padmamalini Baskaran
- College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Yelena Nersesyan
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Zahir Hussain
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA.,Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Leslie M Wise
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Robert E George
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Nadarra Stokes
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | | | - Alejandro M Cohen
- Biological Mass Spectrometry Core Facility, Life Sciences Research Institute, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Evgeny V Pavlov
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| | - Daniel A Llano
- Department of Molecular and Integrative Physiology, Neuroscience Program and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Baskaran Thyagarajan
- College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Eleonora Zakharian
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| |
Collapse
|
9
|
Negative Modulation of TRPM8 Channel Function by Protein Kinase C in Trigeminal Cold Thermoreceptor Neurons. Int J Mol Sci 2020; 21:ijms21124420. [PMID: 32580281 PMCID: PMC7352406 DOI: 10.3390/ijms21124420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/30/2020] [Accepted: 06/12/2020] [Indexed: 01/19/2023] Open
Abstract
TRPM8 is the main molecular entity responsible for cold sensing. This polymodal ion channel is activated by cold, cooling compounds such as menthol, voltage, and rises in osmolality. In corneal cold thermoreceptor neurons (CTNs), TRPM8 expression determines not only their sensitivity to cold, but also their role as neural detectors of ocular surface wetness. Several reports suggest that Protein Kinase C (PKC) activation impacts on TRPM8 function; however, the molecular bases of this functional modulation are still poorly understood. We explored PKC-dependent regulation of TRPM8 using Phorbol 12-Myristate 13-Acetate to activate this kinase. Consistently, recombinant TRPM8 channels, cultured trigeminal neurons, and free nerve endings of corneal CTNs revealed a robust reduction of TRPM8-dependent responses under PKC activation. In corneal CTNs, PKC activation decreased ongoing activity, a key parameter in the role of TRPM8-expressing neurons as humidity detectors, and also the maximal cold-evoked response, which were validated by mathematical modeling. Biophysical analysis indicated that PKC-dependent downregulation of TRPM8 is mainly due to a decreased maximal conductance value, and complementary noise analysis revealed a reduced number of functional channels at the cell surface, providing important clues to understanding the molecular mechanisms of how PKC activity modulates TRPM8 channels in CTNs.
Collapse
|
10
|
Miyahara Y, Hiroe A, Tsuge T, Taguchi S. Microbial Secretion Platform for 3‐Hydroxybutyrate Oligomer and Its End‐Capped Forms Using Chain Transfer Reaction‐Mediated Polyhydroxyalkanoate Synthases. Biotechnol J 2019; 14:e1900201. [DOI: 10.1002/biot.201900201] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 10/27/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Yuki Miyahara
- Department of Chemistry for Life Sciences and Agriculture, Faculty of Life Sciences Tokyo University of Agriculture 1‐1‐1 Sakuragaoka, Setagaya Tokyo 156–8502 Japan
- MIRAI, JST 4‐1‐8 Honcho Kawaguchi Saitama 332‐0012 Japan
| | - Ayaka Hiroe
- Department of Chemistry for Life Sciences and Agriculture, Faculty of Life Sciences Tokyo University of Agriculture 1‐1‐1 Sakuragaoka, Setagaya Tokyo 156–8502 Japan
- MIRAI, JST 4‐1‐8 Honcho Kawaguchi Saitama 332‐0012 Japan
| | - Takeharu Tsuge
- Department of Materials Science and Engineering, Major in Human Centered Science and Biomedical Engineering School of Materials and Chemical Technology, Tokyo Institute of Technology 4259 J2‐47 Nagatsuta‐cho, Midori‐ku Yokohama‐shi Kanagawa 226–8502 Japan
- MIRAI, JST 4‐1‐8 Honcho Kawaguchi Saitama 332‐0012 Japan
| | - Seiichi Taguchi
- Department of Chemistry for Life Sciences and Agriculture, Faculty of Life Sciences Tokyo University of Agriculture 1‐1‐1 Sakuragaoka, Setagaya Tokyo 156–8502 Japan
- CREST, JST 4‐1‐8 Honcho Kawaguchi Saitama 332‐0012 Japan
| |
Collapse
|
11
|
Voinova V, Bonartseva G, Bonartsev A. Effect of poly(3-hydroxyalkanoates) as natural polymers on mesenchymal stem cells. World J Stem Cells 2019; 11:764-786. [PMID: 31692924 PMCID: PMC6828591 DOI: 10.4252/wjsc.v11.i10.764] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/17/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are stromal multipotent stem cells that can differentiate into multiple cell types, including fibroblasts, osteoblasts, chondrocytes, adipocytes, and myoblasts, thus allowing them to contribute to the regeneration of various tissues, especially bone tissue. MSCs are now considered one of the most promising cell types in the field of tissue engineering. Traditional petri dish-based culture of MSCs generate heterogeneity, which leads to inconsistent efficacy of MSC applications. Biodegradable and biocompatible polymers, poly(3-hydroxyalkanoates) (PHAs), are actively used for the manufacture of scaffolds that serve as carriers for MSC growth. The growth and differentiation of MSCs grown on PHA scaffolds depend on the physicochemical properties of the polymers, the 3D and surface microstructure of the scaffolds, and the biological activity of PHAs, which was discovered in a series of investigations. The mechanisms of the biological activity of PHAs in relation to MSCs remain insufficiently studied. We suggest that this effect on MSCs could be associated with the natural properties of bacteria-derived PHAs, especially the most widespread representative poly(3-hydroxybutyrate) (PHB). This biopolymer is present in the bacteria of mammalian microbiota, whereas endogenous poly(3-hydroxybutyrate) is found in mammalian tissues. The possible association of PHA effects on MSCs with various biological functions of poly(3-hydroxybutyrate) in bacteria and eukaryotes, including in humans, is discussed in this paper.
Collapse
Affiliation(s)
- Vera Voinova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow 119234, Russia
| | - Garina Bonartseva
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow 119071, Russia
| | - Anton Bonartsev
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow 119234, Russia
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow 119071, Russia.
| |
Collapse
|
12
|
Bonartsev AP, Voinova VV, Bonartseva GA. Poly(3-hydroxybutyrate) and Human Microbiota (Review). APPL BIOCHEM MICRO+ 2018; 54:547-568. [DOI: 10.1134/s0003683818060066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Indexed: 01/11/2025]
|
13
|
Saska S, Pires LC, Cominotte MA, Mendes LS, de Oliveira MF, Maia IA, da Silva JVL, Ribeiro SJL, Cirelli JA. Three-dimensional printing and in vitro evaluation of poly(3-hydroxybutyrate) scaffolds functionalized with osteogenic growth peptide for tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 89:265-273. [DOI: 10.1016/j.msec.2018.04.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 02/01/2018] [Accepted: 04/10/2018] [Indexed: 01/29/2023]
|
14
|
New Insights into PhaM-PhaC-Mediated Localization of Polyhydroxybutyrate Granules in Ralstonia eutropha H16. Appl Environ Microbiol 2017; 83:AEM.00505-17. [PMID: 28389545 DOI: 10.1128/aem.00505-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/04/2017] [Indexed: 01/08/2023] Open
Abstract
The formation and localization of polyhydroxybutyrate (PHB) granules in Ralstonia eutropha are controlled by PhaM, which interacts both with the PHB synthase (PhaC) and with the bacterial nucleoid. Here, we studied the importance of proline and lysine residues of two C-terminal PAKKA motifs in PhaM for their importance in attaching PHB granules to DNA by in vitro and in vivo methods. Substitution of the lysine residues but not of the proline residues resulted in detachment of formed PHB granules from the nucleoid. Instead, formation of PHB granule clusters at polar regions of the rod-shaped cells and an unequal distribution of PHB granules to daughter cells were observed. The formation of PHB granules was studied by the expression of chromosomally anchored gene fusions of fluorescent proteins with PhaM and PhaC in different backgrounds. PhaM and PhaC fusions showed a distinct colocalization at formed PHB granules in the nucleoid region of the wild type. In a ΔphaC background, PhaM and the catalytically inactive PhaCC319A protein were not able to form fluorescent foci, indicating that correct positioning requires the formation of PHB. Furthermore, time-lapse experiments revealed that PhaC and PhaM proteins detach from formed PHB granules at later stages, resulting in a nonhomogeneous population of PHB granules. This could explain why growth of individual PHB granules stops under PHB-permissive conditions at a certain size.IMPORTANCE PHB granules are storage compounds for carbon and energy in many prokaryotes. Equal distribution of accumulated PHB granules during cell division is therefore important for optimal fitness of the daughter cells. In R. eutropha, PhaM is responsible for maximal activity of PHB synthase, for initiation of PHB granule formation at discrete regions in the cells, and for association of formed PHB granules with the nucleoid. Here we found that four lysine residues of C-terminal PhaM sequence motifs are essential for association of PHB granules with the nucleoid. Furthermore, we followed PHB granule formation by time-lapse microscopy and provide evidence for aging of PHB granules that is manifested by detachment of previously PHB granule-associated PhaM and PHB synthase.
Collapse
|
15
|
Elustondo PA, Nichols M, Negoda A, Thirumaran A, Zakharian E, Robertson GS, Pavlov EV. Mitochondrial permeability transition pore induction is linked to formation of the complex of ATPase C-subunit, polyhydroxybutyrate and inorganic polyphosphate. Cell Death Discov 2016; 2:16070. [PMID: 27924223 PMCID: PMC5137186 DOI: 10.1038/cddiscovery.2016.70] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/10/2016] [Accepted: 08/19/2016] [Indexed: 12/25/2022] Open
Abstract
Mitochondrial permeability transition pore (mPTP) opening allows free movement of ions and small molecules leading to mitochondrial membrane depolarization and ATP depletion that triggers cell death. A multi-protein complex of the mitochondrial ATP synthase has an essential role in mPTP. However, the molecular identity of the central 'pore' part of mPTP complex is not known. A highly purified fraction of mammalian mitochondria containing C-subunit of ATPase (C-subunit), calcium, inorganic polyphosphate (polyP) and polyhydroxybutyrate (PHB) forms ion channels with properties that resemble the native mPTP. We demonstrate here that amount of this channel-forming complex dramatically increases in intact mitochondria during mPTP activation. This increase is inhibited by both Cyclosporine A, an inhibitor of mPTP and Ruthenium Red, an inhibitor of the Mitochondrial Calcium Uniporter. Similar increases in the amount of complex formation occurs in areas of mouse brain damaged by ischemia-reperfusion injury. These findings suggest that calcium-induced mPTP is associated with de novo assembly of a channel comprising C-subunit, polyP and PHB.
Collapse
Affiliation(s)
- P A Elustondo
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University , Halifax, NS, B3H 4R2 Canada
| | - M Nichols
- Departments of Psychiatry and Pharmacology, Brain Repair Centre, Faculty of Medicine Dalhousie University , Halifax, NS, B3H 4R2f Canada
| | - A Negoda
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University , Halifax, NS, B3H 4R2 Canada
| | - A Thirumaran
- Departments of Psychiatry and Pharmacology, Brain Repair Centre, Faculty of Medicine Dalhousie University , Halifax, NS, B3H 4R2f Canada
| | - E Zakharian
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine , 1 Illini Drive, Peoria, IL 61605, USA
| | - G S Robertson
- Departments of Psychiatry and Pharmacology, Brain Repair Centre, Faculty of Medicine Dalhousie University , Halifax, NS, B3H 4R2f Canada
| | - E V Pavlov
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2 Canada; Department of Basic Sciences, New York University, College of Dentistry, 345 East 24th Street, New York, NY 10010, USA
| |
Collapse
|
16
|
Michalak M, Kurcok P, Hakkarainen M. Polyhydroxyalkanoate-based drug delivery systems. POLYM INT 2016. [DOI: 10.1002/pi.5282] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Michał Michalak
- Centre of Polymer and Carbon Materials; Polish Academy of Sciences; M Curie-Skłodowskiej 34 41-819 Zabrze Poland
| | - Piotr Kurcok
- Centre of Polymer and Carbon Materials; Polish Academy of Sciences; M Curie-Skłodowskiej 34 41-819 Zabrze Poland
| | - Minna Hakkarainen
- Department of Fibre and Polymer Technology; KTH Royal Institute of Technology; Stockholm Sweden
| |
Collapse
|
17
|
Elustondo PA, Nichols M, Robertson GS, Pavlov EV. Mitochondrial Ca2+ uptake pathways. J Bioenerg Biomembr 2016; 49:113-119. [DOI: 10.1007/s10863-016-9676-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/05/2016] [Indexed: 12/19/2022]
|
18
|
Affiliation(s)
- Dieter Seebach
- Laboratorium für Organische Chemie; ETH Zürich; Vladimir-Prelog-Weg 3 8093 Zürich Switzerland
| |
Collapse
|
19
|
Asuthkar S, Velpula KK, Elustondo PA, Demirkhanyan L, Zakharian E. TRPM8 channel as a novel molecular target in androgen-regulated prostate cancer cells. Oncotarget 2016; 6:17221-36. [PMID: 25980497 PMCID: PMC4627303 DOI: 10.18632/oncotarget.3948] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/07/2015] [Indexed: 12/12/2022] Open
Abstract
The cold and menthol receptor TRPM8 is highly expressed in prostate and prostate cancer (PC). Recently, we identified that TRPM8 is as an ionotropic testosterone receptor. The TRPM8 mRNA is expressed in early prostate tumors with high androgen levels, while anti-androgen therapy greatly reduces its expression. Here, from the chromatin-immunoprecipitation (ChIP) analysis, we found that an androgen response element (ARE) mediates androgen regulation of trpm8. Furthermore, using immunofluorescence, calcium-imaging and planar lipid bilayers, we identified that TRPM8 channel is functionally regulated by androgens in the prostate. Although TRPM8 mRNA is expressed at high levels, we found that the TRPM8 protein undergoes ubiquitination and degradation in PC cells. The mass-spectrometry analysis of TRPM8, immunoprecipitated from LNCaP cells identified ubiquitin-like modifier-activating enzyme 1 (UBA1). PYR-41, a potent inhibitor of initial enzyme in the ubiquitination cascade, UBA1, increased TRPM8 activity on the plasma membrane (PM) of LNCaP cells. Furthermore, PYR-41-mediated PMTRPM8 activity was accompanied by enhanced activation of p53 and Caspase-9. Interestingly, we found that the trpm8 promoter possesses putative binding sites for p53 and that the overexpression of p53 increased the TRPM8 mRNA levels. In addition to the genomic regulation of TRPM8 by AR and p53, our findings indicate that the testosterone-induced PMTRPM8 activity elicits Ca2+ uptake, subsequently causing apoptotic cell death. These findings support the strategy of rescuing PMTRPM8 expression as a new therapeutic application through the regulation of PC cell growth and proliferation.
Collapse
Affiliation(s)
- Swapna Asuthkar
- University of Illinois College of Medicine, Department of Cancer Biology and Pharmacology, Peoria, IL, USA
| | - Kiran Kumar Velpula
- University of Illinois College of Medicine, Department of Cancer Biology and Pharmacology, Peoria, IL, USA
| | | | - Lusine Demirkhanyan
- University of Illinois College of Medicine, Department of Cancer Biology and Pharmacology, Peoria, IL, USA
| | - Eleonora Zakharian
- University of Illinois College of Medicine, Department of Cancer Biology and Pharmacology, Peoria, IL, USA
| |
Collapse
|
20
|
Methyl-esterified 3-hydroxybutyrate oligomers protect bacteria from hydroxyl radicals. Nat Chem Biol 2016; 12:332-8. [PMID: 26974813 DOI: 10.1038/nchembio.2043] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 01/05/2016] [Indexed: 12/29/2022]
Abstract
Bacteria rely mainly on enzymes, glutathione and other low-molecular weight thiols to overcome oxidative stress. However, hydroxyl radicals are the most cytotoxic reactive oxygen species, and no known enzymatic system exists for their detoxification. We now show that methyl-esterified dimers and trimers of 3-hydroxybutyrate (ME-3HB), produced by bacteria capable of polyhydroxybutyrate biosynthesis, have 3-fold greater hydroxyl radical-scavenging activity than glutathione and 11-fold higher activity than vitamin C or the monomer 3-hydroxybutyric acid. We found that ME-3HB oligomers protect hypersensitive yeast deletion mutants lacking oxidative stress-response genes from hydroxyl radical stress. Our results show that phaC and phaZ, encoding polymerase and depolymerase, respectively, are activated and polyhydroxybutyrate reserves are degraded for production of ME-3HB oligomers in bacteria infecting plant cells and exposed to hydroxyl radical stress. We found that ME-3HB oligomer production is widespread, especially in bacteria adapted to stressful environments. We discuss how ME-3HB oligomers could provide opportunities for numerous applications in human health.
Collapse
|
21
|
Yee NS. TRPM8 Ion Channels as Potential Cancer Biomarker and Target in Pancreatic Cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016:127-155. [DOI: 10.1016/bs.apcsb.2016.01.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
22
|
Rath P, Hilton JK, Sisco NJ, Van Horn WD. Implications of Human Transient Receptor Potential Melastatin 8 (TRPM8) Channel Gating from Menthol Binding Studies of the Sensing Domain. Biochemistry 2015; 55:114-24. [PMID: 26653082 DOI: 10.1021/acs.biochem.5b00931] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The transient receptor potential melastatin 8 (TRPM8) ion channel is the primary cold sensor in humans. TRPM8 is gated by physiologically relevant cold temperatures and chemical ligands that induce cold sensations, such as the analgesic compound menthol. Characterization of TRPM8 ligand-gated channel activation will lead to a better understanding of the fundamental mechanisms that underlie TRPM8 function. Here, the direct binding of menthol to the isolated hTRPM8 sensing domain (transmembrane helices S1-S4) is investigated. These data are compared with two mutant sensing domain proteins, Y745H (S2 helix) and R842H (S4 helix), which have been previously identified in full length TRPM8 to be menthol insensitive. The data presented herein show that menthol specifically binds to the wild type, Y745H, and R842H TRPM8 sensing domain proteins. These results are the first to show that menthol directly binds to the TRPM8 sensing domain and indicates that Y745 and R842 residues, previously identified in functional studies as crucial to menthol sensitivity, do not affect menthol binding but instead alter coupling between the sensing domain and the pore domain.
Collapse
Affiliation(s)
- Parthasarathi Rath
- School of Molecular Sciences, Arizona State University , 551 E. University Drive, Tempe, Arizona 85287, United States.,The Biodesign Institute, Arizona State University , Tempe, Arizona 85281, United States.,The Virginia G. Piper Center for Personalized Diagnostics, Arizona State University , Tempe, Arizona 85281, United States.,The Magnetic Resonance Research Center, Arizona State University , Tempe, Arizona 85287, United States
| | - Jacob K Hilton
- School of Molecular Sciences, Arizona State University , 551 E. University Drive, Tempe, Arizona 85287, United States.,The Biodesign Institute, Arizona State University , Tempe, Arizona 85281, United States.,The Virginia G. Piper Center for Personalized Diagnostics, Arizona State University , Tempe, Arizona 85281, United States.,The Magnetic Resonance Research Center, Arizona State University , Tempe, Arizona 85287, United States
| | - Nicholas J Sisco
- School of Molecular Sciences, Arizona State University , 551 E. University Drive, Tempe, Arizona 85287, United States.,The Biodesign Institute, Arizona State University , Tempe, Arizona 85281, United States.,The Virginia G. Piper Center for Personalized Diagnostics, Arizona State University , Tempe, Arizona 85281, United States.,The Magnetic Resonance Research Center, Arizona State University , Tempe, Arizona 85287, United States
| | - Wade D Van Horn
- School of Molecular Sciences, Arizona State University , 551 E. University Drive, Tempe, Arizona 85287, United States.,The Biodesign Institute, Arizona State University , Tempe, Arizona 85281, United States.,The Virginia G. Piper Center for Personalized Diagnostics, Arizona State University , Tempe, Arizona 85281, United States.,The Magnetic Resonance Research Center, Arizona State University , Tempe, Arizona 85287, United States
| |
Collapse
|
23
|
Yee NS. Roles of TRPM8 Ion Channels in Cancer: Proliferation, Survival, and Invasion. Cancers (Basel) 2015; 7:2134-2146. [PMID: 26512697 PMCID: PMC4695882 DOI: 10.3390/cancers7040882] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 11/24/2022] Open
Abstract
The goal of this article is to provide a critical review of the transient receptor potential melastatin-subfamily member 8 (TRPM8) in cancers, with an emphasis on its roles in cellular proliferation, survival, and invasion. The TRPM8 ion channels regulate Ca²⁺ homeostasis and function as a cellular sensor and transducer of cold temperature. Accumulating evidence has demonstrated that TRPM8 is aberrantly expressed in a variety of malignant solid tumors. Clinicopathological analysis has shown that over-expression of TRPM8 correlates with tumor progression. Experimental data have revealed important roles of TRPM8 channels in cancer cells proliferation, survival, and invasion, which appear to be dependent on the cancer type. Recent reports have begun to reveal the signaling mechanisms that mediate the biological roles of TRPM8 in tumor growth and metastasis. Determining the mechanistic roles of TRPM8 in cancer is expected to elucidate the impact of thermal and chemical stimuli on the formation and progression of neoplasms. Translational research and clinical investigation of TRPM8 in malignant diseases will help exploit these ion channels as molecular biomarkers and therapeutic targets for developing precision cancer medicine.
Collapse
Affiliation(s)
- Nelson S Yee
- Division of Hematology-Oncology, Department of Medicine, Penn State College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA.
- Program of Experimental Therapeutics, Penn State Hershey Cancer Institute, Pennsylvania State University, Hershey, PA 17033, USA.
- Penn State Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, PA 17033, USA.
| |
Collapse
|
24
|
Sun X, Zakharian E. Regulation of the temperature-dependent activation of transient receptor potential vanilloid 1 (TRPV1) by phospholipids in planar lipid bilayers. J Biol Chem 2015; 290:4741-4747. [PMID: 25561742 DOI: 10.1074/jbc.m114.611459] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TRPV1 (transient receptor potential vanilloid 1) proteins are heat-activated nonselective cation channels. TRPV1 channels are polymodal in their function and exhibit multifaceted regulation with various molecular compounds. In this regard, phosphoinositides, particularly phosphatidylinositol 4,5-bisphosphate and phosphatidylinositol 4-phosphate, are important channel regulators. However, their effects on TRPV1 channel activity have not been conclusively determined. To characterize temperature-induced activation of TRPV1 in the presence of different phospholipids, we purified the TRPV1 protein from HEK-293 cells and incorporated it into planar lipid bilayers. In the presence of 2.5 μm phosphatidylinositol 4,5-bisphosphate, TRPV1 channels demonstrated rapid activation at 33-39 °C and achieved full channel opening at 42 °C. At this temperature range, TRPV1 heat activation exhibited steep temperature dependence (temperature coefficient (Q10) of 18), and the channel openings were accompanied by large changes in entropy and enthalpy, suggesting a substantial conformation change. At a similar temperature range, another phosphoinositide, phosphatidylinositol 4-phosphate, also potentiated heat activation of TRPV1, but with much lower efficiency. Negatively charged phosphatidylglycerol could also induce heat activation of TRPV1 channels, although with a small-conductance state. Our data demonstrate that phospholipids, specifically phosphoinositides, are important regulators of TRPV1 and are required for heat-induced channel activity.
Collapse
Affiliation(s)
- Xiaohui Sun
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | - Eleonora Zakharian
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605.
| |
Collapse
|
25
|
Itoh H, Oyama K, Suzuki M, Ishiwata S. Microscopic heat pulse-induced calcium dynamics in single WI-38 fibroblasts. Biophysics (Nagoya-shi) 2014; 10:109-19. [PMID: 27493505 PMCID: PMC4629654 DOI: 10.2142/biophysics.10.109] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/16/2014] [Indexed: 12/31/2022] Open
Abstract
Temperature-sensitive Ca2+ dynamics occur primarily through transient receptor potential channels, but also by means of Ca2+ channels and pumps on the endoplasmic reticulum membrane. As such, cytoplasmic Ca2+ concentration ([Ca2+]cyt) is re-equilibrated by changes in ambient temperature. The present study investigated the effects of heat pulses (heating duration: 2 s or 150 s) on [Ca2+]cyt in single WI-38 fibroblasts, which are considered as normal cells. We found that Ca2+ burst occurred immediately after short (2 s) heat pulse, which is similar to our previous report on HeLa cells, but with less thermosensitivity. The heat pulses originated from a focused 1455-nm infrared laser light were applied in the vicinity of cells under the optical microscope. Ca2+ bursts induced by the heat pulse were suppressed by treating cells with inhibitors for sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) or inositol trisphosphate receptor (IP3R). Long (150 s) heat pulses also induced Ca2+ bursts after the onset of heating and immediately after re-cooling. Cells were more thermosensitive at physiological (37°C) than at room (25°C) temperature; however, at 37°C, cells were responsive at a higher temperature (ambient temperature+heat pulse). These results strongly suggest that the heat pulse-induced Ca2+ burst is caused by a transient imbalance in Ca2+ flow between SERCA and IP3R, and offer a potential new method for thermally controlling Ca2+-regulated cellular functions.
Collapse
Affiliation(s)
- Hideki Itoh
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan; Institute of Medical Biology, Agency for Science Technology & Research (ASTAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Kotaro Oyama
- Department of Physics, Faculty of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Madoka Suzuki
- Waseda Bioscience Research Institute in Singapore (WABIOS), 11 Biopolis Way, #05-02 Helios, Singapore 138667, Singapore; Organization for University Research Initiatives, Waseda University, 513 Waseda Tsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Shin'ichi Ishiwata
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan; Department of Physics, Faculty of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan; Waseda Bioscience Research Institute in Singapore (WABIOS), 11 Biopolis Way, #05-02 Helios, Singapore 138667, Singapore; Organization for University Research Initiatives, Waseda University, 513 Waseda Tsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| |
Collapse
|
26
|
Asuthkar S, Demirkhanyan L, Sun X, Elustondo PA, Krishnan V, Baskaran P, Velpula KK, Thyagarajan B, Pavlov EV, Zakharian E. The TRPM8 protein is a testosterone receptor: II. Functional evidence for an ionotropic effect of testosterone on TRPM8. J Biol Chem 2014; 290:2670-88. [PMID: 25480785 DOI: 10.1074/jbc.m114.610873] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Testosterone is a key steroid hormone in the development of male reproductive tissues and the regulation of the central nervous system. The rapid signaling mechanism induced by testosterone affects numerous behavioral traits, including sexual drive, aggressiveness, and fear conditioning. However, the currently identified testosterone receptor(s) is not believed to underlie the fast signaling, suggesting an orphan pathway. Here we report that an ion channel from the transient receptor potential family, TRPM8, commonly known as the cold and menthol receptor is the major component of testosterone-induced rapid actions. Using cultured and primary cell lines along with the purified TRPM8 protein, we demonstrate that testosterone directly activates TRPM8 channel at low picomolar range. Specifically, testosterone induced TRPM8 responses in primary human prostate cells, PC3 prostate cancer cells, dorsal root ganglion neurons, and hippocampal neurons. Picomolar concentrations of testosterone resulted in full openings of the purified TRPM8 channel in planar lipid bilayers. Furthermore, acute applications of testosterone on human skin elicited a cooling sensation. Our data conclusively demonstrate that testosterone is an endogenous and highly potent agonist of TRPM8, suggesting a role of TRPM8 channels well beyond their well established function in somatosensory neurons. This discovery may further imply TRPM8 channel function in testosterone-dependent behavioral traits.
Collapse
Affiliation(s)
- Swapna Asuthkar
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | - Lusine Demirkhanyan
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | - Xiaohui Sun
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | | | - Vivek Krishnan
- the College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, Wyoming 82071, and
| | - Padmamalini Baskaran
- the College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, Wyoming 82071, and
| | - Kiran Kumar Velpula
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | - Baskaran Thyagarajan
- the College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, Wyoming 82071, and
| | - Evgeny V Pavlov
- Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada, the Department of Basic Sciences, College of Dentistry, New York University, New York, New York 10010
| | - Eleonora Zakharian
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605,
| |
Collapse
|
27
|
Asuthkar S, Elustondo PA, Demirkhanyan L, Sun X, Baskaran P, Velpula KK, Thyagarajan B, Pavlov EV, Zakharian E. The TRPM8 protein is a testosterone receptor: I. Biochemical evidence for direct TRPM8-testosterone interactions. J Biol Chem 2014; 290:2659-69. [PMID: 25480783 DOI: 10.1074/jbc.m114.610824] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The transient receptor potential ion channel of the melastatin subfamily, TRPM8, is a major cold receptor in the peripheral nervous system. Along with the sensory neurons, the TRPM8 protein is highly expressed in the prostate epithelial cells, and this expression is regulated by androgens. Here we investigated the expression and intracellular localization of the TRPM8 channel in relationship to androgens. We performed experiments using human prostate tissues obtained from healthy individuals and patients with prostate cancer at various stages of the disease as well as in cultured cells. Using an immunohistochemistry approach, we detected an intensive colocalization pattern of the TRPM8 protein with endogenous androgens in all tissues tested, suggesting possible interactions. Co-immunoprecipitation experiments performed using cultured prostate epithelial cells, prostate cancer cells, and HEK-293 cells stably expressing TRPM8 further confirmed direct binding of the steroid hormone, testosterone, to the TRPM8 protein. Applications of picomolar concentrations of testosterone to the primary human prostate cells, endogenously expressing TRPM8, elicited Ca(2+) responses and channel currents, and those were inhibited in the presence of TRPM8 antagonist, N-(2-aminoethyl)-N-(4-(benzyloxy)-3-methoxybenzyl)thiophene-2-carboxamide hydrochloride. These results indicate that the TRPM8 channel is physically associated with testosterone and suggest that, in addition to a genomic role, testosterone plays a role in direct regulation of the TRPM8 channel function.
Collapse
Affiliation(s)
- Swapna Asuthkar
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | | | - Lusine Demirkhanyan
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | - Xiaohui Sun
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | - Padmamalini Baskaran
- the College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, Wyoming 82071, and
| | - Kiran Kumar Velpula
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | - Baskaran Thyagarajan
- the College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, Wyoming 82071, and
| | - Evgeny V Pavlov
- Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada, the Department of Basic Sciences, College of Dentistry, New York University, New York, New York 10010
| | - Eleonora Zakharian
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605,
| |
Collapse
|
28
|
Norris V, Reusch RN, Igarashi K, Root-Bernstein R. Molecular complementarity between simple, universal molecules and ions limited phenotype space in the precursors of cells. Biol Direct 2014; 10:28. [PMID: 25470982 PMCID: PMC4264330 DOI: 10.1186/s13062-014-0028-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 11/24/2014] [Indexed: 01/29/2023] Open
Abstract
Background Fundamental problems faced by the protocells and their modern descendants include how to go from one phenotypic state to another; escape from a basin of attraction in the space of phenotypes; reconcile conflicting growth and survival strategies (and thereby live on ‘the scales of equilibria’); and create a coherent, reproducible phenotype from a multitude of constituents. Presentation of the hypothesis The solutions to these problems are likely to be found with the organic and inorganic molecules and inorganic ions that constituted protocells, which we term SUMIs for Simple Universal Molecules and Ions. These SUMIs probably included polyphosphate (PolyP) as a source of energy and of phosphate; poly-(R)-3-hydroxybutyrate (PHB) as a source of carbon and as a transporter in association with PolyP; polyamines as a source of nitrogen; lipids as precursors of membranes; as well as peptides, nucleic acids, and calcium. Here, we explore the hypothesis that the direct interactions between PHB, PolyP, polyamines and lipids – modulated by calcium – played a central role in solving the fundamental problems faced by early and modern cells. Testing the hypothesis We review evidence that SUMIs (1) were abundant and available to protocells; (2) are widespread in modern cells; (3) interact with one another and other cellular constituents to create structures with new functions surprisingly similar to those of proteins and RNA; (4) are essential to creating coherent phenotypes in modern bacteria. SUMIs are therefore natural candidates for reducing the immensity of phenotype space and making the transition from a “primordial soup” to living cells. Implications of the hypothesis We discuss the relevance of the SUMIs and their interactions to the ideas of molecular complementarity, composomes (molecular aggregates with hereditary properties based on molecular complementarity), and a prebiotic ecology of co-evolving populations of composomes. In particular, we propose that SUMIs might limit the initial phenotype space of composomes in a coherent way. As examples, we propose that acidocalcisomes arose from interactions and self-selection among SUMIs and that the phosphorylation of proteins in modern cells had its origin in the covalent modification of proteins by PHB. Reviewers This article was reviewed by Doron Lancet and Kepa Ruiz-Mirazo.
Collapse
|
29
|
Ferrandiz-Huertas C, Mathivanan S, Wolf CJ, Devesa I, Ferrer-Montiel A. Trafficking of ThermoTRP Channels. MEMBRANES 2014; 4:525-64. [PMID: 25257900 PMCID: PMC4194048 DOI: 10.3390/membranes4030525] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/11/2014] [Accepted: 08/08/2014] [Indexed: 12/19/2022]
Abstract
ThermoTRP channels (thermoTRPs) define a subfamily of the transient receptor potential (TRP) channels that are activated by changes in the environmental temperature, from noxious cold to injurious heat. Acting as integrators of several stimuli and signalling pathways, dysfunction of these channels contributes to several pathological states. The surface expression of thermoTRPs is controlled by both, the constitutive and regulated vesicular trafficking. Modulation of receptor surface density during pathological processes is nowadays considered as an interesting therapeutic approach for management of diseases, such as chronic pain, in which an increased trafficking is associated with the pathological state. This review will focus on the recent advances trafficking of the thermoTRP channels, TRPV1, TRPV2, TRPV4, TRPM3, TRPM8 and TRPA1, into/from the plasma membrane. Particularly, regulated membrane insertion of thermoTRPs channels contributes to a fine tuning of final channel activity, and indeed, it has resulted in the development of novel therapeutic approaches with successful clinical results such as disruption of SNARE-dependent exocytosis by botulinum toxin or botulinomimetic peptides.
Collapse
Affiliation(s)
| | - Sakthikumar Mathivanan
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| | - Christoph Jakob Wolf
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| | - Isabel Devesa
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| |
Collapse
|
30
|
Dedkova EN, Blatter LA. Role of β-hydroxybutyrate, its polymer poly-β-hydroxybutyrate and inorganic polyphosphate in mammalian health and disease. Front Physiol 2014; 5:260. [PMID: 25101001 PMCID: PMC4102118 DOI: 10.3389/fphys.2014.00260] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 06/19/2014] [Indexed: 12/14/2022] Open
Abstract
We provide a comprehensive review of the role of β-hydroxybutyrate (β-OHB), its linear polymer poly-β-hydroxybutyrate (PHB), and inorganic polyphosphate (polyP) in mammalian health and disease. β-OHB is a metabolic intermediate that constitutes 70% of ketone bodies produced during ketosis. Although ketosis has been generally considered as an unfavorable pathological state (e.g., diabetic ketoacidosis in type-1 diabetes mellitus), it has been suggested that induction of mild hyperketonemia may have certain therapeutic benefits. β-OHB is synthesized in the liver from acetyl-CoA by β-OHB dehydrogenase and can be used as alternative energy source. Elevated levels of PHB are associated with pathological states. In humans, short-chain, complexed PHB (cPHB) is found in a wide variety of tissues and in atherosclerotic plaques. Plasma cPHB concentrations correlate strongly with atherogenic lipid profiles, and PHB tissue levels are elevated in type-1 diabetic animals. However, little is known about mechanisms of PHB action especially in the heart. In contrast to β-OHB, PHB is a water-insoluble, amphiphilic polymer that has high intrinsic viscosity and salt-solvating properties. cPHB can form non-specific ion channels in planar lipid bilayers and liposomes. PHB can form complexes with polyP and Ca(2+) which increases membrane permeability. The biological roles played by polyP, a ubiquitous phosphate polymer with ATP-like bonds, have been most extensively studied in prokaryotes, however polyP has recently been linked to a variety of functions in mammalian cells, including blood coagulation, regulation of enzyme activity in cancer cells, cell proliferation, apoptosis and mitochondrial ion transport and energy metabolism. Recent evidence suggests that polyP is a potent activator of the mitochondrial permeability transition pore in cardiomyocytes and may represent a hitherto unrecognized key structural and functional component of the mitochondrial membrane system.
Collapse
Affiliation(s)
- Elena N Dedkova
- Department of Molecular Biophysics and Physiology, Rush University Medical Center Chicago, IL, USA
| | - Lothar A Blatter
- Department of Molecular Biophysics and Physiology, Rush University Medical Center Chicago, IL, USA
| |
Collapse
|
31
|
Pertusa M, González A, Hardy P, Madrid R, Viana F. Bidirectional modulation of thermal and chemical sensitivity of TRPM8 channels by the initial region of the N-terminal domain. J Biol Chem 2014; 289:21828-43. [PMID: 24917670 DOI: 10.1074/jbc.m114.565994] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
TRPM8, a nonselective cation channel activated by cold, voltage, and cooling compounds such as menthol, is the principal molecular detector of cold temperatures in primary sensory neurons of the somatosensory system. The N-terminal domain of TRPM8 consists of 693 amino acids, but little is known about its contribution to channel function. Here, we identified two distinct regions within the initial N terminus of TRPM8 that contribute differentially to channel activity and proper folding and assembly. Deletion or substitution of the first 40 residues yielded channels with augmented responses to cold and menthol. The thermal threshold of activation of these mutants was shifted 2 °C to higher temperatures, and the menthol dose-response curve was displaced to lower concentrations. Site-directed mutagenesis screening revealed that single point mutations at positions Ser-26 or Ser-27 by proline caused a comparable increase in the responses to cold and menthol. Electrophysiological analysis of the S27P mutant revealed that the enhanced sensitivity to agonists is related to a leftward shift in the voltage dependence of activation, increasing the probability of channel openings at physiological membrane potentials. In addition, we found that the region encompassing positions 40-60 is a key element in the proper folding and assembly of TRPM8. Different deletions and mutations within this region rendered channels with an impaired function that are retained within the endoplasmic reticulum. Our results suggest a critical contribution of the initial region of the N-terminal domain of TRPM8 to thermal and chemical sensitivity and the proper biogenesis of this polymodal ion channel.
Collapse
Affiliation(s)
- María Pertusa
- From the Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, 9160000 Santiago, Chile and the Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas, 03550 Alicante, Spain
| | - Alejandro González
- From the Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, 9160000 Santiago, Chile and
| | - Paulina Hardy
- From the Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, 9160000 Santiago, Chile and
| | - Rodolfo Madrid
- From the Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, 9160000 Santiago, Chile and
| | - Félix Viana
- the Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas, 03550 Alicante, Spain
| |
Collapse
|
32
|
To be or not to be a poly(3-hydroxybutyrate) (PHB) depolymerase: PhaZd1 (PhaZ6) and PhaZd2 (PhaZ7) of Ralstonia eutropha, highly active PHB depolymerases with no detectable role in mobilization of accumulated PHB. Appl Environ Microbiol 2014; 80:4936-46. [PMID: 24907326 DOI: 10.1128/aem.01056-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The putative physiological functions of two related intracellular poly(3-hydroxybutyrate) (PHB) depolymerases, PhaZd1 and PhaZd2, of Ralstonia eutropha H16 were investigated. Purified PhaZd1 and PhaZd2 were active with native PHB granules in vitro. Partial removal of the proteinaceous surface layer of native PHB granules by trypsin treatment or the use of PHB granules isolated from ΔphaP1 or ΔphaP1-phaP5 mutant strains resulted in increased specific PHB depolymerase activity, especially for PhaZd2. Constitutive expression of PhaZd1 or PhaZd2 reduced or even prevented the accumulation of PHB under PHB-permissive conditions in vivo. Expression of translational fusions of enhanced yellow fluorescent protein (EYFP) with PhaZd1 and PhaZd2 in which the active-site serines (S190 and Ser193) were replaced with alanine resulted in the colocalization of only PhaZd1 fusions with PHB granules. C-terminal fusions of inactive PhaZd2(S193A) with EYFP revealed the presence of spindle-like structures, and no colocalization with PHB granules was observed. Chromosomal deletion of phaZd1, phaZd2, or both depolymerase genes had no significant effect on PHB accumulation and mobilization during growth in nutrient broth (NB) or NB-gluconate medium. Moreover, neither proteome analysis of purified native PHB granules nor lacZ fusion studies gave any indication that PhaZd1 or PhaZd2 was detectably present in the PHB granule fraction or expressed at all during growth on NB-gluconate medium. In conclusion, PhaZd1 and PhaZd2 are two PHB depolymerases with a high capacity to degrade PHB when artificially expressed but are apparently not involved in PHB mobilization in the wild type. The true in vivo functions of PhaZd1 and PhaZd2 remain obscure.
Collapse
|
33
|
Stotz SC, Scott LO, Drummond-Main C, Avchalumov Y, Girotto F, Davidsen J, Gómez-Gárcia MR, Rho JM, Pavlov EV, Colicos MA. Inorganic polyphosphate regulates neuronal excitability through modulation of voltage-gated channels. Mol Brain 2014; 7:42. [PMID: 24886461 PMCID: PMC4061113 DOI: 10.1186/1756-6606-7-42] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/26/2014] [Indexed: 01/06/2023] Open
Abstract
Background Inorganic polyphosphate (polyP) is a highly charged polyanion capable of interacting with a number of molecular targets. This signaling molecule is released into the extracellular matrix by central astrocytes and by peripheral platelets during inflammation. While the release of polyP is associated with both induction of blood coagulation and astrocyte extracellular signaling, the role of secreted polyP in regulation of neuronal activity remains undefined. Here we test the hypothesis that polyP is an important participant in neuronal signaling. Specifically, we investigate the ability of neurons to release polyP and to induce neuronal firing, and clarify the underlying molecular mechanisms of this process by studying the action of polyP on voltage gated channels. Results Using patch clamp techniques, and primary hippocampal and dorsal root ganglion cell cultures, we demonstrate that polyP directly influences neuronal activity, inducing action potential generation in both PNS and CNS neurons. Mechanistically, this is accomplished by shifting the voltage sensitivity of NaV channel activation toward the neuronal resting membrane potential, the block KV channels, and the activation of CaV channels. Next, using calcium imaging we found that polyP stimulates an increase in neuronal network activity and induces calcium influx in glial cells. Using in situ DAPI localization and live imaging, we demonstrate that polyP is naturally present in synaptic regions and is released from the neurons upon depolarization. Finally, using a biochemical assay we demonstrate that polyP is present in synaptosomes and can be released upon their membrane depolarization by the addition of potassium chloride. Conclusions We conclude that polyP release leads to increased excitability of the neuronal membrane through the modulation of voltage gated ion channels. Together, our data establishes that polyP could function as excitatory neuromodulator in both the PNS and CNS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Michael A Colicos
- Department of Physiology & Pharmacology and the Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
34
|
Post-Translational Modifications of TRP Channels. Cells 2014; 3:258-87. [PMID: 24717323 PMCID: PMC4092855 DOI: 10.3390/cells3020258] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 03/25/2014] [Accepted: 03/27/2014] [Indexed: 01/07/2023] Open
Abstract
Transient receptor potential (TRP) channels constitute an ancient family of cation channels that have been found in many eukaryotic organisms from yeast to human. TRP channels exert a multitude of physiological functions ranging from Ca2+ homeostasis in the kidney to pain reception and vision. These channels are activated by a wide range of stimuli and undergo covalent post-translational modifications that affect and modulate their subcellular targeting, their biophysical properties, or channel gating. These modifications include N-linked glycosylation, protein phosphorylation, and covalent attachment of chemicals that reversibly bind to specific cysteine residues. The latter modification represents an unusual activation mechanism of ligand-gated ion channels that is in contrast to the lock-and-key paradigm of receptor activation by its agonists. In this review, we summarize the post-translational modifications identified on TRP channels and, when available, explain their physiological role.
Collapse
|
35
|
Jendrossek D, Pfeiffer D. New insights in the formation of polyhydroxyalkanoate granules (carbonosomes) and novel functions of poly(3-hydroxybutyrate). Environ Microbiol 2014; 16:2357-73. [PMID: 24329995 DOI: 10.1111/1462-2920.12356] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 11/27/2013] [Accepted: 12/05/2013] [Indexed: 12/18/2022]
Abstract
The metabolism of polyhydroxybutyrate (PHB) and related polyhydroxyalkanoates (PHAs) has been investigated by many groups for about three decades, and good progress was obtained in understanding the mechanisms of biosynthesis and biodegradation of this class of storage molecules. However, the molecular events that happen at the onset of PHB synthesis and the details of the initiation of PHB/PHA granule formation, as well as the complex composition of the proteinaceous surface layer of PHB/PHA granules, have only recently come into the focus of research and were not reviewed yet. In this contribution, we summarize the progress in understanding the initiation and formation of the PHA granule complex at the example of Ralstonia eutropha H16 (model organism of PHB-accumulating bacteria). Where appropriate, we include information on PHA granules of Pseudomonas putida as a representative species for medium-chain-length PHA-accumulating bacteria. We suggest to replace the previous micelle mode of PHB granule formation by the Scaffold Model in which the PHB synthase initiation complex is bound to the bacterial nucleoid. In the second part, we highlight data on other forms of PHB: oligo-PHB with ≈100 to 200 3-hydroxybutyrate (3HB) units and covalently bound PHB (cPHB) are unrelated in function to storage PHB but are presumably present in all living organisms, and therefore must be of fundamental importance.
Collapse
|
36
|
Samways DSK. Applications for mass spectrometry in the study of ion channel structure and function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 806:237-61. [PMID: 24952185 DOI: 10.1007/978-3-319-06068-2_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Ion channels are intrinsic membrane proteins that form gated ion-permeable pores across biological membranes. Depending on the type, ion channels exhibit sensitivities to a diverse range of stimuli including changes in membrane potential, binding by diffusible ligands, changes in temperature and direct mechanical force. The purpose of these proteins is to facilitate the passive diffusion of ions down their respective electrochemical gradients into and out of the cell, and between intracellular compartments. In doing so, ion channels can affect transmembrane potentials and regulate the intracellular homeostasis of the important second messenger, Ca(2+). The ion channels of the plasma membrane are of particular clinical interest due to their regulation of cell excitability and cytosolic Ca(2+) levels, and the fact that they are most amenable to manipulation by exogenously applied drugs and toxins. A critical step in improving the pharmacopeia of chemicals available that influence the activity of ion channels is understanding how their three-dimensional structure imparts function. Here, progress has been slow relative to that for soluble protein structures in large part due to the limitations of applying conventional structure determination methods, such as X-ray crystallography, nuclear magnetic resonance imaging, and mass spectrometry, to membrane proteins. Although still an underutilized technique in the assessment of membrane protein structure, recent advances have pushed mass spectrometry to the fore as an important complementary approach to studying the structure and function of ion channels. In addition to revealing the subtle conformational changes in ion channel structure that accompany gating and permeation, mass spectrometry is already being used effectively for identifying tissue-specific posttranslational modifications and mRNA splice variants. Furthermore, the use of mass spectrometry for high-throughput proteomics analysis, which has proven so successful for soluble proteins, is already providing valuable insight into the functional interactions of ion channels within the context of the macromolecular-signaling complexes that they inhabit in vivo. In this chapter, the potential for mass spectrometry as a complementary approach to the study of ion channel structure and function will be reviewed with examples of its application.
Collapse
Affiliation(s)
- Damien S K Samways
- Department of Biology, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699, USA,
| |
Collapse
|
37
|
Madrid R, Pertusa M. Intimacies and physiological role of the polymodal cold-sensitive ion channel TRPM8. CURRENT TOPICS IN MEMBRANES 2014; 74:293-324. [PMID: 25366241 DOI: 10.1016/b978-0-12-800181-3.00011-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The detection of environmental temperature is critical for the survival of the most diverse organisms. Thermosensitive transient receptor potential (thermoTRP) channels have evolved as a class of ion channels activated by a wide range of temperatures. These molecular thermal sensors are spread through the different TRP channel subfamilies. Among the Melastatin subfamily of TRP channels, the eighth member, TRPM8, is a calcium-permeable cationic ion channel activated by cold, by substances that evoke cold sensation such as menthol, and by voltage. This channel is considered the main molecular entity responsible for the sensitivity to cold of primary sensory neurons of the somatosensory system. Here we present to the readers a summary of some the most relevant biophysical properties, physiological role, and molecular intimacies of this polymodal thermoTRP channel.
Collapse
Affiliation(s)
- Rodolfo Madrid
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - María Pertusa
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
38
|
Abstract
Transient receptor potential melastatin 8 (TRPM8) was originally cloned from prostate tissue. Shortly thereafter, the protein was identified as a cold- and menthol-activated ion channel in peripheral sensory neurons, where it plays a critical role in cold temperature detection. In this chapter, we review our current understanding of the molecular and biophysical properties, the pharmacology, and the modulation by signaling molecules of this TRP channel. Finally, we examine the physiological role of TRPM8 and its emerging link to various human diseases, including pain, prostate cancer, dry eye disease, and metabolic disorders.
Collapse
Affiliation(s)
- Laura Almaraz
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Avenida S. Ramón y Cajal s.n., San Juan de Alicante, 03550, Spain
| | | | | | | |
Collapse
|
39
|
Török Z, Crul T, Maresca B, Schütz GJ, Viana F, Dindia L, Piotto S, Brameshuber M, Balogh G, Péter M, Porta A, Trapani A, Gombos I, Glatz A, Gungor B, Peksel B, Vigh L, Csoboz B, Horváth I, Vijayan MM, Hooper PL, Harwood JL, Vigh L. Plasma membranes as heat stress sensors: from lipid-controlled molecular switches to therapeutic applications. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:1594-618. [PMID: 24374314 DOI: 10.1016/j.bbamem.2013.12.015] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/09/2013] [Accepted: 12/18/2013] [Indexed: 12/31/2022]
Abstract
The classic heat shock (stress) response (HSR) was originally attributed to protein denaturation. However, heat shock protein (Hsp) induction occurs in many circumstances where no protein denaturation is observed. Recently considerable evidence has been accumulated to the favor of the "Membrane Sensor Hypothesis" which predicts that the level of Hsps can be changed as a result of alterations to the plasma membrane. This is especially pertinent to mild heat shock, such as occurs in fever. In this condition the sensitivity of many transient receptor potential (TRP) channels is particularly notable. Small temperature stresses can modulate TRP gating significantly and this is influenced by lipids. In addition, stress hormones often modify plasma membrane structure and function and thus initiate a cascade of events, which may affect HSR. The major transactivator heat shock factor-1 integrates the signals originating from the plasma membrane and orchestrates the expression of individual heat shock genes. We describe how these observations can be tested at the molecular level, for example, with the use of membrane perturbers and through computational calculations. An important fact which now starts to be addressed is that membranes are not homogeneous nor do all cells react identically. Lipidomics and cell profiling are beginning to address the above two points. Finally, we observe that a deregulated HSR is found in a large number of important diseases where more detailed knowledge of the molecular mechanisms involved may offer timely opportunities for clinical interventions and new, innovative drug treatments. This article is part of a Special Issue entitled: Membrane Structure and Function: Relevance in the Cell's Physiology, Pathology and Therapy.
Collapse
Affiliation(s)
- Zsolt Török
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary.
| | - Tim Crul
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Bruno Maresca
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy
| | - Gerhard J Schütz
- Institute of Applied Physics, Vienna University of Technology, Wiedner Hauptstrasse 8-10, 1040 Vienna, Austria
| | - Felix Viana
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 San Juan de Alicante, Spain
| | - Laura Dindia
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Stefano Piotto
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy
| | - Mario Brameshuber
- Institute of Applied Physics, Vienna University of Technology, Wiedner Hauptstrasse 8-10, 1040 Vienna, Austria
| | - Gábor Balogh
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Mária Péter
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Amalia Porta
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy
| | - Alfonso Trapani
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy
| | - Imre Gombos
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Attila Glatz
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Burcin Gungor
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Begüm Peksel
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - László Vigh
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Bálint Csoboz
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Ibolya Horváth
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Mathilakath M Vijayan
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada; Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Phillip L Hooper
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Medical School, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - John L Harwood
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK
| | - László Vigh
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary.
| |
Collapse
|
40
|
PhaM is the physiological activator of poly(3-hydroxybutyrate) (PHB) synthase (PhaC1) in Ralstonia eutropha. Appl Environ Microbiol 2013; 80:555-63. [PMID: 24212577 DOI: 10.1128/aem.02935-13] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Poly(3-hydroxybutyrate) (PHB) synthase (PhaC1) is the key enzyme of PHB synthesis in Ralstonia eutropha and other PHB-accumulating bacteria and catalyzes the polymerization of 3-hydroxybutyryl-CoA to PHB. Activity assays of R. eutropha PHB synthase are characterized by the presence of lag phases and by low specific activity. It is assumed that the lag phase is caused by the time necessary to convert the inactive PhaC1 monomer into the active dimeric form by an unknown priming process. The lag phase can be reduced by addition of nonionic detergents such as hecameg [6-O-(N-heptyl-carbamoyl)-methyl-α-D-glucopyranoside], which apparently accelerates the formation of PhaC1 dimers. We identified the PHB granule-associated protein (PGAP) PhaM as the natural primer (activator) of PHB synthase activity. PhaM was recently discovered as a novel type of PGAP with multiple functions in PHB metabolism. Addition of PhaM to PHB synthase assays resulted in immediate polymerization of 3HB coenzyme A with high specific activity and without a significant lag phase. The effect of PhaM on (i) PhaC1 activity, (ii) oligomerization of PhaC1, (iii) complex formation with PhaC1, and (iv) PHB granule formation in vitro and in vivo was shown by cross-linking experiments of purified proteins (PhaM, PhaC1) with glutardialdehyde, by size exclusion chromatography, and by fluorescence microscopic detection of de novo-synthesized PHB granules.
Collapse
|
41
|
Elustondo PA, Angelova PR, Kawalec M, Michalak M, Kurcok P, Abramov AY, Pavlov EV. Polyhydroxybutyrate targets mammalian mitochondria and increases permeability of plasmalemmal and mitochondrial membranes. PLoS One 2013; 8:e75812. [PMID: 24086638 PMCID: PMC3781052 DOI: 10.1371/journal.pone.0075812] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 08/21/2013] [Indexed: 01/21/2023] Open
Abstract
Poly(3-hydroxybutyrate) (PHB) is a polyester of 3-hydroxybutyric acid (HB) that is ubiquitously present in all organisms. In higher eukaryotes PHB is found in the length of 10 to 100 HB units and can be present in free form as well as in association with proteins and inorganic polyphosphate. It has been proposed that PHB can mediate ion transport across lipid bilayer membranes. We investigated the ability of PHB to interact with living cells and isolated mitochondria and the effects of these interactions on membrane ion transport. We performed experiments using a fluorescein derivative of PHB (fluo-PHB). We found that fluo-PHB preferentially accumulated inside the mitochondria of HeLa cells. Accumulation of fluo-PHB induced mitochondrial membrane depolarization. This membrane depolarization was significantly delayed by the inhibitor of the mitochondrial permeability transition pore - Cyclosporin A. Further experiments using intact cells as well as isolated mitochondria confirmed that the effects of PHB directly linked to its ability to facilitate ion transport, including calcium, across the membranes. We conclude that PHB demonstrates ionophoretic properties in biological membranes and this effect is most profound in mitochondria due to the selective accumulation of the polymer in this organelle.
Collapse
Affiliation(s)
- Pia A. Elustondo
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Novia Scotia, Canada
| | | | - Michał Kawalec
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Michał Michalak
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Piotr Kurcok
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | | | - Evgeny V. Pavlov
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Novia Scotia, Canada
- * E-mail:
| |
Collapse
|