1
|
Salminen A. Aryl hydrocarbon receptor impairs circadian regulation in Alzheimer's disease: Potential impact on glymphatic system dysfunction. Eur J Neurosci 2024; 60:3901-3920. [PMID: 38924210 DOI: 10.1111/ejn.16450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
Circadian clocks maintain diurnal rhythms of sleep-wake cycle of 24 h that regulate not only the metabolism of an organism but also many other periodical processes. There is substantial evidence that circadian regulation is impaired in Alzheimer's disease. Circadian clocks regulate many properties known to be disturbed in Alzheimer's patients, such as the integrity of the blood-brain barrier (BBB) as well as the diurnal glymphatic flow that controls waste clearance from the brain. Interestingly, an evolutionarily conserved transcription factor, that is, aryl hydrocarbon receptor (AhR), impairs the function of the core clock proteins and thus could disturb diurnal rhythmicity in the BBB. There is abundant evidence that the activation of AhR signalling inhibits the expression of the major core clock proteins, such as the brain and muscle arnt-like 1 (BMAL1), clock circadian regulator (CLOCK) and period circadian regulator 1 (PER1) in different experimental models. The expression of AhR is robustly increased in the brains of Alzheimer's patients, and protein level is enriched in astrocytes of the BBB. It seems that AhR signalling inhibits glymphatic flow since it is known that (i) activation of AhR impairs the function of the BBB, which is cooperatively interconnected with the glymphatic system in the brain, and (ii) neuroinflammation and dysbiosis of gut microbiota generate potent activators of AhR, which are able to impair glymphatic flow. I will examine current evidence indicating that activation of AhR signalling could disturb circadian functions of the BBB and impair glymphatic flow and thus be involved in the development of Alzheimer's pathology.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
2
|
Fry HC, Liu Y, Taylor SK. Design and Function of α-Helix-Rich, Heme-Binding Peptide Materials. Biomacromolecules 2024; 25:3398-3408. [PMID: 38752597 DOI: 10.1021/acs.biomac.4c00049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Peptide materials often employ short peptides that self-assemble into unique nanoscale architectures and have been employed across many fields relevant to medicine and energy. A majority of peptide materials are high in β-sheet, secondary structure content, including heme-binding peptide materials. To broaden the structural diversity of heme-binding peptide materials, a small series of peptides were synthesized to explore the design criteria required for (1) folding into an α-helix structure, (2) assembling into a nanoscale material, (3) binding heme, and (4) demonstrating functions similar to that of heme proteins. One peptide was identified to meet all four criteria, including the heme protein function of CO binding and its microsecond-to-millisecond recombination rates, as measured by transient absorption spectroscopy. Implications of new design criteria and peptide material function through heme incorporation are discussed.
Collapse
Affiliation(s)
- H Christopher Fry
- Center for Nanoscale Materials, Argonne National Laboratory, 9700 S. Cass Ave., Lemont, Illinois 60439, United States
| | - Yuzi Liu
- Center for Nanoscale Materials, Argonne National Laboratory, 9700 S. Cass Ave., Lemont, Illinois 60439, United States
| | - Sunny K Taylor
- Pritzker School for Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| |
Collapse
|
3
|
Anbalagan S. Oxygen is an essential gasotransmitter directly sensed via protein gasoreceptors. Animal Model Exp Med 2024; 7:189-193. [PMID: 38529771 PMCID: PMC11079153 DOI: 10.1002/ame2.12400] [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: 12/26/2023] [Accepted: 02/04/2024] [Indexed: 03/27/2024] Open
Abstract
The current restrictive criteria for gasotransmitters exclude oxygen (O2) as a gasotransmitter in vertebrates. In this manuscript, I propose a revision of gasotransmitter criteria to include O2 per se as a signaling molecule and 'essential gasotransmitter' for vertebrates. This revision would enable us to search for protein-based O2-binding sensors (gasoreceptors) in all cells in the brain or other tissues rather than specialized tissues such as the carotid body or gills. If microorganisms have protein-based O2-binding sensors or gasoreceptors such as DosP or FixL or FNR with diverse signaling domains, then eukaryotic cells must also have O2-binding sensors or gasoreceptors. Just as there are protein-based receptor(s) for nitric oxide (GUCY1A, GUCY1B, CLOCK, NR1D2) in cells of diverse tissues, it is reasonable to consider that there are protein-based receptors for O2 in cells of diverse tissues as well. In mammals, O2 must be acting as a gasotransmitter or gaseous signaling molecule via protein-based gasoreceptors such as androglobin that very likely mediate acute sensing of O2. Accepting O2 as an essential gasotransmitter will enable us to search for gasoreceptors not only for O2 but also for other nonessential gasotransmitters such as hydrogen sulfide, ammonia, methane, and ethylene. It will also allow us to investigate the role of environment-derived metal ions in acute gas (or solute) sensing within and between organisms. Finally, accepting O2 per se as a signaling molecule acting via gasoreceptors will open up the field of gasocrinology.
Collapse
Affiliation(s)
- Savani Anbalagan
- Faculty of Biology, Institute of Molecular Biology and BiotechnologyAdam Mickiewicz UniversityPoznańPoland
| |
Collapse
|
4
|
Bauer N, Liu D, Nguyen T, Wang B. Unraveling the Interplay of Dopamine, Carbon Monoxide, and Heme Oxygenase in Neuromodulation and Cognition. ACS Chem Neurosci 2024; 15:400-407. [PMID: 38214656 PMCID: PMC10853931 DOI: 10.1021/acschemneuro.3c00742] [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: 11/15/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 01/13/2024] Open
Abstract
The dopaminergic system plays important roles in neuromodulation, including prominent roles in complex neurological functions such as cognition, reward, motivation, and memory. Understandably, the highly complex nature of such physiological functions means that their regulation is intertwined with other signaling pathways, as has been demonstrated by numerous studies. Contrary to its public perception of being poisonous at all concentrations, carbon monoxide (CO) is produced endogenously from heme degradation by heme oxygenase (HO) as part of the physiological process of red blood cell turnover. Physiological concentrations of CO can reach high micromolar ranges in the hemoglobin bound form. Low-dose CO has shown therapeutic effects in numerous animal models, including traumatic brain injury via engaging various hemoprotein targets. As such, the HO-CO axis has been shown to offer beneficial effects in organ protection, anti-inflammation, and neuroprotection, among many others. Further, a large number of publications have shown the interactions among CO, HO, and the dopaminergic system. In this review, we critically examine such experimental evidence in a holistic fashion and in the context of a possible dopamine-HO-CO signaling axis. We hope that this Perspective will stimulate additional investigations into the molecular connectivity related to this possible axis and open doors to the development of novel therapeutics that impact the dopaminergic system.
Collapse
Affiliation(s)
- Nicola Bauer
- Department of Chemistry and
Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - Dongning Liu
- Department of Chemistry and
Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - TanPhat Nguyen
- Department of Chemistry and
Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - Binghe Wang
- Department of Chemistry and
Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| |
Collapse
|
5
|
Kaiser S, Henrich L, Kiessling I, Loy B, Schallner N. Neuroprotection via Carbon Monoxide Depends on the Circadian Regulation of CD36-Mediated Microglial Erythrophagocytosis in Hemorrhagic Stroke. Int J Mol Sci 2024; 25:1680. [PMID: 38338958 PMCID: PMC10855856 DOI: 10.3390/ijms25031680] [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: 12/20/2023] [Revised: 01/19/2024] [Accepted: 01/27/2024] [Indexed: 02/12/2024] Open
Abstract
The molecular basis for circadian dependency in stroke due to subarachnoid hemorrhagic stroke (SAH) remains unclear. We reasoned that microglial erythrophagocytosis, crucial for SAH response, follows a circadian pattern involving carbon monoxide (CO) and CD36 surface expression. The microglial BV-2 cell line and primary microglia (PMG) under a clocked medium change were exposed to blood ± CO (250 ppm, 1 h) in vitro. Circadian dependency and the involvement of CD36 were analyzed in PMG isolated from control mice and CD36-/- mice and by RNA interference targeting Per-2. In vivo investigations, including phagocytosis, vasospasm, microglia activation and spatial memory, were conducted in an SAH model using control and CD36-/- mice at different zeitgeber times (ZT). In vitro, the surface expression of CD36 and its dependency on CO and phagocytosis occurred with changed circadian gene expression. CD36-/- PMG exhibited altered circadian gene expression, phagocytosis and impaired responsiveness to CO. In vivo, control mice with SAH demonstrated circadian dependency in microglia activation, erythrophagocytosis and CO-mediated protection at ZT2, in contrast to CD36-/- mice. Our study indicates that circadian rhythmicity modulates microglial activation and subsequent CD36-dependent phagocytosis. CO altered circadian-dependent neuroprotection and CD36 induction, determining the functional outcome in a hemorrhagic stroke model. This study emphasizes how circadian rhythmicity influences neuronal damage after neurovascular events.
Collapse
Affiliation(s)
- Sandra Kaiser
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany (N.S.)
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Luise Henrich
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany (N.S.)
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Iva Kiessling
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany (N.S.)
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Benedikt Loy
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany (N.S.)
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Nils Schallner
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany (N.S.)
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| |
Collapse
|
6
|
Henrich L, Kiessling I, Steimer M, Frase S, Kaiser S, Schallner N. Circadian dependency of microglial heme oxygenase-1 expression and inflammation determine neuronal injury in hemorrhagic stroke. J Inflamm (Lond) 2023; 20:43. [PMID: 38104143 PMCID: PMC10725034 DOI: 10.1186/s12950-023-00371-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND The heme oxygenase-1 (HO-1) enzyme pathway is of crucial importance in the removal of toxic blood components and regulation of neuroinflammation following hemorrhagic stroke. Although a circadian pattern dependency in the incidence and severity of hemorrhagic stroke exists, it is unknown whether the activity of the HO-1 system in the context of hemorrhagic injury also exhibits circadian dependency. We hypothesized that the circadian regulation of microglial HO-1 would determine the extent of neuroinflammation and neuronal injury in a murine model of subarachnoid hemorrhage (SAH). METHODS In vitro expression patterns of HO-1 and circadian rhythm genes were analyzed in the microglial BV-2 cell line and primary microglia (PMG) using Western blot and qPCR. PMG isolated from Hmox1fl/fl and LyzM-Cre-Hmox1fl/fl mice were used to evaluate the role of microglial HO-1. We further investigated the in vivo relevance in a murine subarachnoid hemorrhage (SAH) model using Hmox1fl/fl and LyzM-Cre-Hmox1fl/fl mice with myeloid cell HO-1 deficiency, inducing SAH at different zeitgeber (ZT) times and analyzing the expression of HO-1 and the circadian control gene Period-2 (Per-2), respectively. Furthermore, we measured the inflammatory cytokine Monocyte Chemoattractant Protein-1 (MCP-1) in the cerebrospinal fluid of SAH patients in correlation with clinical outcome. RESULTS HO-1 baseline expression and response to CO with blood exposure depended on ZT. In vitro expression of circadian control genes was de-synchronized in LyzM-Cre-Hmox1fl/fl PMG and did not respond to exogenous CO exposure. We found that circadian rhythm plays a crucial role in brain damage after SAH. At ZT2, we observed less phagocytic function, more vasospasm and increased microglial activation. CO reduced mortality at ZT12 in HO-1 deficient mice and reduced the difference between ZT2 and ZT12 in the inflammatory response. Induction of MCP-1 in the CSF from SAH patients was time-dependent and correlated with the expression of circadian control genes, SAH severity, functional impairment and delirium. CONCLUSIONS Our data point towards a crucial role for the HO-1 enzyme system and circadian control in neuronal injury after a hemorrhagic stroke.
Collapse
Affiliation(s)
- Luise Henrich
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Iva Kiessling
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matti Steimer
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sibylle Frase
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurology and Neuroscience, Medical Center, University of Freiburg, Freiburg, Germany
| | - Sandra Kaiser
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nils Schallner
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany.
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
7
|
Rathod DC, Vaidya SM, Hopp MT, Kühl T, Imhof D. Shapes and Patterns of Heme-Binding Motifs in Mammalian Heme-Binding Proteins. Biomolecules 2023; 13:1031. [PMID: 37509066 PMCID: PMC10377097 DOI: 10.3390/biom13071031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Heme is a double-edged sword. On the one hand, it has a pivotal role as a prosthetic group of hemoproteins in many biological processes ranging from oxygen transport and storage to miRNA processing. On the other hand, heme can transiently associate with proteins, thereby regulating biochemical pathways. During hemolysis, excess heme, which is released into the plasma, can bind to proteins and regulate their activity and function. The role of heme in these processes is under-investigated, with one problem being the lack of knowledge concerning recognition mechanisms for the initial association of heme with the target protein and the formation of the resulting complex. A specific heme-binding sequence motif is a prerequisite for such complex formation. Although numerous short signature sequences indicating a particular protein function are known, a comprehensive analysis of the heme-binding motifs (HBMs) which have been identified in proteins, concerning specific patterns and structural peculiarities, is missing. In this report, we focus on the evaluation of known mammalian heme-regulated proteins concerning specific recognition and structural patterns in their HBMs. The Cys-Pro dipeptide motifs are particularly emphasized because of their more frequent occurrence. This analysis presents a comparative insight into the sequence and structural anomalies observed during transient heme binding, and consequently, in the regulation of the relevant protein.
Collapse
Affiliation(s)
- Dhruv C Rathod
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany
| | - Sonali M Vaidya
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany
| | - Marie-T Hopp
- Department of Chemistry, Institute for Integrated Natural Sciences, University of Koblenz, D-56070 Koblenz, Germany
| | - Toni Kühl
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany
| |
Collapse
|
8
|
Rasmussen ES, Takahashi JS, Green CB. Time to target the circadian clock for drug discovery. Trends Biochem Sci 2022; 47:745-758. [PMID: 35577675 PMCID: PMC9378619 DOI: 10.1016/j.tibs.2022.04.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/01/2022] [Accepted: 04/18/2022] [Indexed: 11/30/2022]
Abstract
The circadian clock is an intracellular timekeeping device that drives daily rhythms in diverse and extensive processes throughout the body. The clock mechanism comprises a core transcription/translation negative feedback loop that is modulated by a complex set of additional interlocking feedback loops. Pharmacological manipulation of the clock may be valuable for treating many maladies including jet lag, shift work and related sleep disorders, various metabolic diseases, and cancer. We review recent identification of small-molecule clock modulators and discuss the biochemical features of the core clock that may be amenable to future drug discovery.
Collapse
Affiliation(s)
- Emil Sjulstok Rasmussen
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph S Takahashi
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carla B Green
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
9
|
Yuan Z, De La Cruz LK, Yang X, Wang B. Carbon Monoxide Signaling: Examining Its Engagement with Various Molecular Targets in the Context of Binding Affinity, Concentration, and Biologic Response. Pharmacol Rev 2022; 74:823-873. [PMID: 35738683 DOI: 10.1124/pharmrev.121.000564] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Carbon monoxide (CO) has been firmly established as an endogenous signaling molecule with a variety of pathophysiological and pharmacological functions, including immunomodulation, organ protection, and circadian clock regulation, among many others. In terms of its molecular mechanism(s) of action, CO is known to bind to a large number of hemoproteins with at least 25 identified targets, including hemoglobin, myoglobin, neuroglobin, cytochrome c oxidase, cytochrome P450, soluble guanylyl cyclase, myeloperoxidase, and some ion channels with dissociation constant values spanning the range of sub-nM to high μM. Although CO's binding affinity with a large number of targets has been extensively studied and firmly established, there is a pressing need to incorporate such binding information into the analysis of CO's biologic response in the context of affinity and dosage. Especially important is to understand the reservoir role of hemoglobin in CO storage, transport, distribution, and transfer. We critically review the literature and inject a sense of quantitative assessment into our analyses of the various relationships among binding affinity, CO concentration, target occupancy level, and anticipated pharmacological actions. We hope that this review presents a picture of the overall landscape of CO's engagement with various targets, stimulates additional research, and helps to move the CO field in the direction of examining individual targets in the context of all of the targets and the concentration of available CO. We believe that such work will help the further understanding of the relationship of CO concentration and its pathophysiological functions and the eventual development of CO-based therapeutics. SIGNIFICANCE STATEMENT: The further development of carbon monoxide (CO) as a therapeutic agent will significantly rely on the understanding of CO's engagement with therapeutically relevant targets of varying affinity. This review critically examines the literature by quantitatively analyzing the intricate relationships among targets, target affinity for CO, CO level, and the affinity state of carboxyhemoglobin and provide a holistic approach to examining the molecular mechanism(s) of action for CO.
Collapse
Affiliation(s)
- Zhengnan Yuan
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| | - Ladie Kimberly De La Cruz
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| | - Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| |
Collapse
|
10
|
Fleischhacker AS, Sarkar A, Liu L, Ragsdale SW. Regulation of protein function and degradation by heme, heme responsive motifs, and CO. Crit Rev Biochem Mol Biol 2022; 57:16-47. [PMID: 34517731 PMCID: PMC8966953 DOI: 10.1080/10409238.2021.1961674] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Heme is an essential biomolecule and cofactor involved in a myriad of biological processes. In this review, we focus on how heme binding to heme regulatory motifs (HRMs), catalytic sites, and gas signaling molecules as well as how changes in the heme redox state regulate protein structure, function, and degradation. We also relate these heme-dependent changes to the affected metabolic processes. We center our discussion on two HRM-containing proteins: human heme oxygenase-2, a protein that binds and degrades heme (releasing Fe2+ and CO) in its catalytic core and binds Fe3+-heme at HRMs located within an unstructured region of the enzyme, and the transcriptional regulator Rev-erbβ, a protein that binds Fe3+-heme at an HRM and is involved in CO sensing. We will discuss these and other proteins as they relate to cellular heme composition, homeostasis, and trafficking. In addition, we will discuss the HRM-containing family of proteins and how the stability and activity of these proteins are regulated in a dependent manner through the HRMs. Then, after reviewing CO-mediated protein regulation of heme proteins, we turn our attention to the involvement of heme, HRMs, and CO in circadian rhythms. In sum, we stress the importance of understanding the various roles of heme and the distribution of the different heme pools as they relate to the heme redox state, CO, and heme binding affinities.
Collapse
Affiliation(s)
- Angela S. Fleischhacker
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anindita Sarkar
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Liu Liu
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Stephen W. Ragsdale
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Multi-Modal Regulation of Circadian Physiology by Interactive Features of Biological Clocks. BIOLOGY 2021; 11:biology11010021. [PMID: 35053019 PMCID: PMC8772734 DOI: 10.3390/biology11010021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022]
Abstract
The circadian clock is a fundamental biological timing mechanism that generates nearly 24 h rhythms of physiology and behaviors, including sleep/wake cycles, hormone secretion, and metabolism. Evolutionarily, the endogenous clock is thought to confer living organisms, including humans, with survival benefits by adapting internal rhythms to the day and night cycles of the local environment. Mirroring the evolutionary fitness bestowed by the circadian clock, daily mismatches between the internal body clock and environmental cycles, such as irregular work (e.g., night shift work) and life schedules (e.g., jet lag, mistimed eating), have been recognized to increase the risk of cardiac, metabolic, and neurological diseases. Moreover, increasing numbers of studies with cellular and animal models have detected the presence of functional circadian oscillators at multiple levels, ranging from individual neurons and fibroblasts to brain and peripheral organs. These oscillators are tightly coupled to timely modulate cellular and bodily responses to physiological and metabolic cues. In this review, we will discuss the roles of central and peripheral clocks in physiology and diseases, highlighting the dynamic regulatory interactions between circadian timing systems and multiple metabolic factors.
Collapse
|
12
|
Kitagishi H, Mao Q. Capture of carbon monoxide using a heme protein model: from biomimetic chemistry of heme proteins to physiological and therapeutic applications. Polym J 2021. [DOI: 10.1038/s41428-021-00591-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
13
|
Ricci A, Di Pierro E, Marcacci M, Ventura P. Mechanisms of Neuronal Damage in Acute Hepatic Porphyrias. Diagnostics (Basel) 2021; 11:diagnostics11122205. [PMID: 34943446 PMCID: PMC8700611 DOI: 10.3390/diagnostics11122205] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 01/12/2023] Open
Abstract
Porphyrias are a group of congenital and acquired diseases caused by an enzymatic impairment in the biosynthesis of heme. Depending on the specific enzyme involved, different types of porphyrias (i.e., chronic vs. acute, cutaneous vs. neurovisceral, hepatic vs. erythropoietic) are described, with different clinical presentations. Acute hepatic porphyrias (AHPs) are characterized by life-threatening acute neuro-visceral crises (acute porphyric attacks, APAs), featuring a wide range of neuropathic (central, peripheral, autonomic) manifestations. APAs are usually unleashed by external "porphyrinogenic" triggers, which are thought to cause an increased metabolic demand for heme. During APAs, the heme precursors δ-aminolevulinic acid (ALA) and porphobilinogen (PBG) accumulate in the bloodstream and urine. Even though several hypotheses have been developed to explain the protean clinical picture of APAs, the exact mechanism of neuronal damage in AHPs is still a matter of debate. In recent decades, a role has been proposed for oxidative damage caused by ALA, mitochondrial and synaptic ALA toxicity, dysfunction induced by relative heme deficiency on cytochromes and other hemeproteins (i.e., nitric oxide synthases), pyridoxal phosphate functional deficiency, derangements in the metabolic pathways of tryptophan, and other factors. Since the pathway leading to the biosynthesis of heme is inscribed into a complex network of interactions, which also includes some fundamental processes of basal metabolism, a disruption in any of the steps of this pathway is likely to have multiple pathogenic effects. Here, we aim to provide a comprehensive review of the current evidence regarding the mechanisms of neuronal damage in AHPs.
Collapse
Affiliation(s)
- Andrea Ricci
- Internal Medicine Unit, Department of Medical and Surgical Science for Children and Adults, University of Modena e Reggio Emilia, 41124 Modena, Italy; (A.R.); (M.M.)
| | - Elena Di Pierro
- Dipartimento di Medicina Interna, Fondazione IRCSS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Matteo Marcacci
- Internal Medicine Unit, Department of Medical and Surgical Science for Children and Adults, University of Modena e Reggio Emilia, 41124 Modena, Italy; (A.R.); (M.M.)
| | - Paolo Ventura
- Internal Medicine Unit, Department of Medical and Surgical Science for Children and Adults, University of Modena e Reggio Emilia, 41124 Modena, Italy; (A.R.); (M.M.)
- Correspondence: ; Tel.: +39-059-4225-542
| |
Collapse
|
14
|
Gallio A, Fung SSP, Cammack-Najera A, Hudson AJ, Raven EL. Understanding the Logistics for the Distribution of Heme in Cells. JACS AU 2021; 1:1541-1555. [PMID: 34723258 PMCID: PMC8549057 DOI: 10.1021/jacsau.1c00288] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Indexed: 05/03/2023]
Abstract
Heme is essential for the survival of virtually all living systems-from bacteria, fungi, and yeast, through plants to animals. No eukaryote has been identified that can survive without heme. There are thousands of different proteins that require heme in order to function properly, and these are responsible for processes such as oxygen transport, electron transfer, oxidative stress response, respiration, and catalysis. Further to this, in the past few years, heme has been shown to have an important regulatory role in cells, in processes such as transcription, regulation of the circadian clock, and the gating of ion channels. To act in a regulatory capacity, heme needs to move from its place of synthesis (in mitochondria) to other locations in cells. But while there is detailed information on how the heme lifecycle begins (heme synthesis), and how it ends (heme degradation), what happens in between is largely a mystery. Here we summarize recent information on the quantification of heme in cells, and we present a discussion of a mechanistic framework that could meet the logistical challenge of heme distribution.
Collapse
Affiliation(s)
- Andrea
E. Gallio
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - Simon S.-P. Fung
- Department
of Chemistry and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester LE1 7RH, U.K.
| | - Ana Cammack-Najera
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - Andrew J. Hudson
- Department
of Chemistry and Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester LE1 7RH, U.K.
| | - Emma L. Raven
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| |
Collapse
|
15
|
Wang T, Ashrafi A, Modareszadeh P, Deese AR, Chacon Castro MDC, Alemi PS, Zhang L. An Analysis of the Multifaceted Roles of Heme in the Pathogenesis of Cancer and Related Diseases. Cancers (Basel) 2021; 13:4142. [PMID: 34439295 PMCID: PMC8393563 DOI: 10.3390/cancers13164142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/08/2021] [Accepted: 08/13/2021] [Indexed: 12/28/2022] Open
Abstract
Heme is an essential prosthetic group in proteins and enzymes involved in oxygen utilization and metabolism. Heme also plays versatile and fascinating roles in regulating fundamental biological processes, ranging from aerobic respiration to drug metabolism. Increasing experimental and epidemiological data have shown that altered heme homeostasis accelerates the development and progression of common diseases, including various cancers, diabetes, vascular diseases, and Alzheimer's disease. The effects of heme on the pathogenesis of these diseases may be mediated via its action on various cellular signaling and regulatory proteins, as well as its function in cellular bioenergetics, specifically, oxidative phosphorylation (OXPHOS). Elevated heme levels in cancer cells intensify OXPHOS, leading to higher ATP generation and fueling tumorigenic functions. In contrast, lowered heme levels in neurons may reduce OXPHOS, leading to defects in bioenergetics and causing neurological deficits. Further, heme has been shown to modulate the activities of diverse cellular proteins influencing disease pathogenesis. These include BTB and CNC homology 1 (BACH1), tumor suppressor P53 protein, progesterone receptor membrane component 1 protein (PGRMC1), cystathionine-β-synthase (CBS), soluble guanylate cyclase (sGC), and nitric oxide synthases (NOS). This review provides an in-depth analysis of heme function in influencing diverse molecular and cellular processes germane to disease pathogenesis and the modes by which heme modulates the activities of cellular proteins involved in the development of cancer and other common diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA; (T.W.); (A.A.); (P.M.); (A.R.D.); (M.D.C.C.C.); (P.S.A.)
| |
Collapse
|
16
|
Gilun P, Flisikowski K, Flisikowska T, Kwiatkowska J, Wąsowska B, Koziorowska-Gilun M. Role of Methylation in Period2 ( PER2) Transcription in the Context of the Presence or Absence of Light Signals: Natural and Chemical-Studies on the Pig Model. Int J Mol Sci 2021; 22:ijms22157796. [PMID: 34360562 PMCID: PMC8346033 DOI: 10.3390/ijms22157796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 12/16/2022] Open
Abstract
It has been proposed that carbon monoxide (CO) is a chemical light carrier that is transferred by the humoral pathway from the retina to the brain. Here, we aimed to study how deeply CO is involved in regulating the expression of Period2 gene (PER2), one of the genes maintaining the intrinsic biological clock. In our in vivo experiment, we studied whether CO may be a chemical signal and is also equivalent to natural light in three groups of pigs: Normal: housed in natural conditions without any procedures, Control: adapted and kept in constant darkness, infused with blank plasma, and CO treated: adapted and kept in constant darkness infused with CO-enriched plasma. After the experiment, the animals were slaughtered at two times of day: 12 p.m. and 12 a.m. Next, hypothalamus samples were collected. Quantitative PCR, the DNA methylation of the promoter sequence containing enhancers (E-box) and a functional analysis of the PER2 promoter was performed. qPCR showed a differential pattern of PER2 mRNA expression at daytime oscillation in the examined groups. Pyrosequencing revealed daytime changes in the methylation level of regulatory sites of the examined sequence. Luciferase reporter assay confirmed that E-boxes (CANNTG) drive the expression of the porcine PER2 in vitro. In conclusion, changes in methylation over 24 h may regulate the oscillatory manner of PER2 expression.
Collapse
Affiliation(s)
- Przemysław Gilun
- Department of Local Physiological Regulations, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland;
- Correspondence:
| | - Krzysztof Flisikowski
- School of Life Sciences, Chair of Livestock Biotechnology, Technical University of Munich, D-85354 Freising, Germany; (K.F.); (T.F.)
| | - Tatiana Flisikowska
- School of Life Sciences, Chair of Livestock Biotechnology, Technical University of Munich, D-85354 Freising, Germany; (K.F.); (T.F.)
| | - Joanna Kwiatkowska
- Department of Neurosurgery, School of Medicine, University of Warmia and Mazury, 10-719 Olsztyn, Poland;
| | - Barbara Wąsowska
- Department of Local Physiological Regulations, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland;
| | - Magdalena Koziorowska-Gilun
- Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bioengineering, University of Warmia and Mazury, 10-719 Olsztyn, Poland;
| |
Collapse
|
17
|
Frase S, Kaiser S, Steimer M, Selzner L, Foit NA, Niesen WD, Schallner N. Patients with Subarachnoid Hemorrhage Exhibit Disturbed Expression Patterns of the Circadian Rhythm Gene Period-2. Life (Basel) 2021; 11:life11020124. [PMID: 33562664 PMCID: PMC7915417 DOI: 10.3390/life11020124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Circadian rhythm gene expression in cerebral pacemaker regions is regulated by a transcriptional-translational feedback loop across the 24-h day-night cycle. In preclinical models of subarachnoid hemorrhage (SAH), cyclic gene expression is disrupted. Stabilization of circadian rhythm gene expression attenuates susceptibility to ischemic damage in both neuronal and myocardial tissues. In this clinical observational study, circadian rhythm gene Period-2 (Per2) mRNA expression levels were determined from blood leukocytes and cerebrospinal fluid (CSF) cells via real-time PCR on days 1, 7 and 14 after aneurysm rupture in 49 patients with spontaneous SAH. CSF Per2 expression was markedly suppressed immediately after SAH and remained suppressed over the course of two weeks of ICU treatment. Short-term mortality as well as occurrence of delirium was associated with greater extent of Per2 suppression on day 1 after SAH. Patients that developed delayed cerebral ischemia exhibited comparatively lower Per2 expression levels on day 7 after SAH, while presence of vasospasm remained unaffected. However, Per2 expression did not differ in patient groups with favourable or non-favourable functional neurological outcome (modified Rankin Scales 1–3 vs. 4–6). While our findings suggest a potential protective effect of stable circadian rhythm gene expression on the extent of ischemic damage, this effect was confined to the early disease course and was not reflected in patients’ functional neurological outcome.
Collapse
Affiliation(s)
- Sibylle Frase
- Department of Neurology and Neuroscience, Medical Center—University of Freiburg, 79106 Freiburg, Germany;
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.K.); (M.S.); (L.S.); (N.A.F.); (N.S.)
- Correspondence:
| | - Sandra Kaiser
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.K.); (M.S.); (L.S.); (N.A.F.); (N.S.)
- Department of Anesthesiology and Critical Care, Medical Center—University of Freiburg, 79106 Freiburg, Germany
| | - Matti Steimer
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.K.); (M.S.); (L.S.); (N.A.F.); (N.S.)
- Department of Anesthesiology and Critical Care, Medical Center—University of Freiburg, 79106 Freiburg, Germany
| | - Lisa Selzner
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.K.); (M.S.); (L.S.); (N.A.F.); (N.S.)
- Department of Anesthesiology and Critical Care, Medical Center—University of Freiburg, 79106 Freiburg, Germany
| | - Niels Alexander Foit
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.K.); (M.S.); (L.S.); (N.A.F.); (N.S.)
- Department of Neurosurgery, Medical Center—University of Freiburg, 79106 Freiburg, Germany
| | - Wolf-Dirk Niesen
- Department of Neurology and Neuroscience, Medical Center—University of Freiburg, 79106 Freiburg, Germany;
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.K.); (M.S.); (L.S.); (N.A.F.); (N.S.)
| | - Nils Schallner
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.K.); (M.S.); (L.S.); (N.A.F.); (N.S.)
- Department of Anesthesiology and Critical Care, Medical Center—University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
18
|
Kitagishi H, Kano K. Synthetic heme protein models that function in aqueous solution. Chem Commun (Camb) 2021; 57:148-173. [DOI: 10.1039/d0cc07044k] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Supramolecular porphyrin–cyclodextrin complexes act as biomimetic heme protein models in aqueous solution.
Collapse
Affiliation(s)
- Hiroaki Kitagishi
- Department of Molecular Chemistry and Biochemistry
- Faculty of Science and Engineering
- Doshisha University
- Kyoto 610-0321
- Japan
| | - Koji Kano
- Department of Molecular Chemistry and Biochemistry
- Faculty of Science and Engineering
- Doshisha University
- Kyoto 610-0321
- Japan
| |
Collapse
|
19
|
Altered dynamics in the circadian oscillation of clock genes in serum-shocked NIH-3T3 cells by the treatment of GYY4137 or AOAA. Arch Biochem Biophys 2020; 680:108237. [DOI: 10.1016/j.abb.2019.108237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/02/2019] [Accepted: 12/21/2019] [Indexed: 11/19/2022]
|
20
|
Wißbrock A, Goradia NB, Kumar A, Paul George AA, Kühl T, Bellstedt P, Ramachandran R, Hoffmann P, Galler K, Popp J, Neugebauer U, Hampel K, Zimmermann B, Adam S, Wiendl M, Krönke G, Hamza I, Heinemann SH, Frey S, Hueber AJ, Ohlenschläger O, Imhof D. Structural insights into heme binding to IL-36α proinflammatory cytokine. Sci Rep 2019; 9:16893. [PMID: 31729440 PMCID: PMC6858345 DOI: 10.1038/s41598-019-53231-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 10/29/2019] [Indexed: 12/14/2022] Open
Abstract
Cytokines of the interleukin (IL)-1 family regulate immune and inflammatory responses. The recently discovered IL-36 family members are involved in psoriasis, rheumatoid arthritis, and pulmonary diseases. Here, we show that IL-36α interacts with heme thereby contributing to its regulation. Based on in-depth spectroscopic analyses, we describe two heme-binding sites in IL-36α that associate with heme in a pentacoordinated fashion. Solution NMR analysis reveals structural features of IL-36α and its complex with heme. Structural investigation of a truncated IL-36α supports the notion that the N-terminus is necessary for association with its cognate receptor. Consistent with our structural studies, IL-36-mediated signal transduction was negatively regulated by heme in synovial fibroblast-like synoviocytes from rheumatoid arthritis patients. Taken together, our results provide a structural framework for heme-binding proteins and add IL-1 cytokines to the group of potentially heme-regulated proteins.
Collapse
Affiliation(s)
- Amelie Wißbrock
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, D-53121, Bonn, Germany
| | - Nishit B Goradia
- CS Protein Production, Leibniz Institute on Aging/Fritz Lipmann Institute, D-07745, Jena, Germany.,European Molecular Biology Laboratory, D-22607, Hamburg, Germany
| | - Amit Kumar
- CS Protein Production, Leibniz Institute on Aging/Fritz Lipmann Institute, D-07745, Jena, Germany
| | - Ajay Abisheck Paul George
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, D-53121, Bonn, Germany
| | - Toni Kühl
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, D-53121, Bonn, Germany
| | - Peter Bellstedt
- Institute of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, D-07743, Jena, Germany
| | - Ramadurai Ramachandran
- CS Protein Production, Leibniz Institute on Aging/Fritz Lipmann Institute, D-07745, Jena, Germany
| | - Patrick Hoffmann
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, D-07747, Jena, Germany.,Leibniz Institute of Photonic Technology (Leibniz IPHT), D-07745, Jena, Germany
| | - Kerstin Galler
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, D-07747, Jena, Germany.,Leibniz Institute of Photonic Technology (Leibniz IPHT), D-07745, Jena, Germany
| | - Jürgen Popp
- Leibniz Institute of Photonic Technology (Leibniz IPHT), D-07745, Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, D-07743, Jena, Germany
| | - Ute Neugebauer
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, D-07747, Jena, Germany.,Leibniz Institute of Photonic Technology (Leibniz IPHT), D-07745, Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, D-07743, Jena, Germany
| | | | | | - Susanne Adam
- Department of Internal Medicine 3 - Rheumatology and Immunology, University of Erlangen-Nürnberg (FAU) and University Hospital Erlangen, D-91054, Erlangen, Germany
| | - Maximilian Wiendl
- Department of Internal Medicine 3 - Rheumatology and Immunology, University of Erlangen-Nürnberg (FAU) and University Hospital Erlangen, D-91054, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 - Rheumatology and Immunology, University of Erlangen-Nürnberg (FAU) and University Hospital Erlangen, D-91054, Erlangen, Germany
| | - Iqbal Hamza
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD, 20742, USA.,Department of Cell Biology & Molecular Genetics, University of Maryland, College Park, MD, 20742, USA
| | - Stefan H Heinemann
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, D-07745, Jena, Germany
| | - Silke Frey
- Department of Internal Medicine 3 - Rheumatology and Immunology, University of Erlangen-Nürnberg (FAU) and University Hospital Erlangen, D-91054, Erlangen, Germany
| | - Axel J Hueber
- Department of Internal Medicine 3 - Rheumatology and Immunology, University of Erlangen-Nürnberg (FAU) and University Hospital Erlangen, D-91054, Erlangen, Germany
| | - Oliver Ohlenschläger
- CS Protein Production, Leibniz Institute on Aging/Fritz Lipmann Institute, D-07745, Jena, Germany.
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, D-53121, Bonn, Germany.
| |
Collapse
|
21
|
Freeman SL, Kwon H, Portolano N, Parkin G, Venkatraman Girija U, Basran J, Fielding AJ, Fairall L, Svistunenko DA, Moody PCE, Schwabe JWR, Kyriacou CP, Raven EL. Heme binding to human CLOCK affects interactions with the E-box. Proc Natl Acad Sci U S A 2019; 116:19911-19916. [PMID: 31527239 PMCID: PMC6778266 DOI: 10.1073/pnas.1905216116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The circadian clock is an endogenous time-keeping system that is ubiquitous in animals and plants as well as some bacteria. In mammals, the clock regulates the sleep-wake cycle via 2 basic helix-loop-helix PER-ARNT-SIM (bHLH-PAS) domain proteins-CLOCK and BMAL1. There is emerging evidence to suggest that heme affects circadian control, through binding of heme to various circadian proteins, but the mechanisms of regulation are largely unknown. In this work we examine the interaction of heme with human CLOCK (hCLOCK). We present a crystal structure for the PAS-A domain of hCLOCK, and we examine heme binding to the PAS-A and PAS-B domains. UV-visible and electron paramagnetic resonance spectroscopies are consistent with a bis-histidine ligated heme species in solution in the oxidized (ferric) PAS-A protein, and by mutagenesis we identify His144 as a ligand to the heme. There is evidence for flexibility in the heme pocket, which may give rise to an additional Cys axial ligand at 20K (His/Cys coordination). Using DNA binding assays, we demonstrate that heme disrupts binding of CLOCK to its E-box DNA target. Evidence is presented for a conformationally mobile protein framework, which is linked to changes in heme ligation and which has the capacity to affect binding to the E-box. Within the hCLOCK structural framework, this would provide a mechanism for heme-dependent transcriptional regulation.
Collapse
Affiliation(s)
- Samuel L Freeman
- School of Chemistry, University of Bristol, BS8 1TS Bristol, United Kingdom
| | - Hanna Kwon
- School of Chemistry, University of Bristol, BS8 1TS Bristol, United Kingdom
| | - Nicola Portolano
- Department of Chemistry, University of Leicester, LE1 7RH Leicester, United Kingdom
- Leicester Institute of Structural and Chemical Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
| | - Gary Parkin
- Department of Chemistry, University of Leicester, LE1 7RH Leicester, United Kingdom
- Leicester Institute of Structural and Chemical Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
| | - Umakhanth Venkatraman Girija
- Department of Chemistry, University of Leicester, LE1 7RH Leicester, United Kingdom
- Leicester Institute of Structural and Chemical Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
| | - Jaswir Basran
- Department of Chemistry, University of Leicester, LE1 7RH Leicester, United Kingdom
- Leicester Institute of Structural and Chemical Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
| | - Alistair J Fielding
- School of Pharmacy and Biomolecular Science, Liverpool John Moores University, Liverpool L3 3AF, United Kingdom
| | - Louise Fairall
- Leicester Institute of Structural and Chemical Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
- Department of Molecular and Cell Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
| | - Dimitri A Svistunenko
- School of Biological Sciences, University of Essex, Colchester, Essex CO4 3SQ, United Kingdom
| | - Peter C E Moody
- Leicester Institute of Structural and Chemical Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
- Department of Molecular and Cell Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
| | - John W R Schwabe
- Leicester Institute of Structural and Chemical Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
- Department of Molecular and Cell Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
| | - Charalambos P Kyriacou
- Department of Genetics and Genome Biology, University of Leicester, LE1 7RH Leicester, United Kingdom
| | - Emma L Raven
- School of Chemistry, University of Bristol, BS8 1TS Bristol, United Kingdom;
| |
Collapse
|
22
|
2,3,7,8-Tetrachlorodibenzo-p-dioxin abolishes circadian regulation of hepatic metabolic activity in mice. Sci Rep 2019; 9:6514. [PMID: 31015483 PMCID: PMC6478849 DOI: 10.1038/s41598-019-42760-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 04/05/2019] [Indexed: 12/19/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR) activation is reported to alter the hepatic expression of circadian clock regulators, however the impact on clock-controlled metabolism has not been thoroughly investigated. This study examines the effects of AhR activation on hepatic transcriptome and metabolome rhythmicity in male C57BL/6 mice orally gavaged with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) every 4 days for 28 days. TCDD diminished the rhythmicity of several core clock regulators (e.g. Arntl, Clock, Nr1d1, Per1, Cry1, Nfil3) in a dose-dependent manner, involving either a ≥ 3.3-fold suppression in amplitude or complete loss of oscillation. Accordingly, protein levels (ARNTL, REV-ERBα, NFIL3) and genomic binding (ARNTL) of select regulators were reduced and arrhythmic following treatment. As a result, the oscillating expression of 99.6% of 5,636 clock-controlled hepatic genes was abolished including genes associated with the metabolism of lipids, glucose/glycogen, and heme. For example, TCDD flattened expression of the rate-limiting enzymes in both gluconeogenesis (Pck1) and glycogenesis (Gys2), consistent with the depletion and loss of rhythmicity in hepatic glycogen levels. Examination of polar hepatic extracts by untargeted mass spectrometry revealed that virtually all oscillating metabolites lost rhythmicity following treatment. Collectively, these results suggest TCDD disrupted circadian regulation of hepatic metabolism, altering metabolic efficiency and energy storage.
Collapse
|
23
|
Leung GCH, Fung SSP, Dovey NRB, Raven EL, Hudson AJ. Precise determination of heme binding affinity in proteins. Anal Biochem 2019; 572:45-51. [PMID: 30807737 DOI: 10.1016/j.ab.2019.02.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 01/22/2023]
Abstract
Accumulating evidence suggests a new role for cellular heme as a signalling molecule, in which interactions with target proteins are more transient than found with traditionally-defined hemoproteins. To study this role, a precise method is needed for determining the heme-binding affinity (or dissociation constant, Kd). Estimates of Kd are commonly made following a spectrophotometric titration of an apo-protein with hemin. An impediment to precise determination is, however, the challenge in discriminating between the Soret absorbance for the product (holo-protein) and that for the titrant (hemin). An altogether different approach has been used in this paper to separate contributions made by these components to absorbance values. The pure component spectra and concentration profiles are estimated by a multivariate curve-resolution (MCR) algorithm. This approach has significant advantages over existing methods. First, a more precise determination of Kd can be made as concentration profiles for all three components (apo-protein/holo-protein/hemin) are determined and can be simultaneously fitted to a theoretical-binding model. Second, an absorption spectrum for the holo-protein is calculated. This is a unique advantage of MCR and attractive for investigating proteins in which the nature of heme binding has not, hitherto, been characterised because the holo-protein spectrum provides information on the interaction.
Collapse
Affiliation(s)
- Galvin C-H Leung
- Department of Chemistry and the Leicester Institute of Structural & Chemical Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Simon S-P Fung
- Department of Chemistry and the Leicester Institute of Structural & Chemical Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Nicholas R B Dovey
- Department of Chemistry and the Leicester Institute of Structural & Chemical Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Emma L Raven
- School of Chemistry, University of Bristol, Bristol, BS8 1TS, United Kingdom
| | - Andrew J Hudson
- Department of Chemistry and the Leicester Institute of Structural & Chemical Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom.
| |
Collapse
|
24
|
Shimizu T, Lengalova A, Martínek V, Martínková M. Heme: emergent roles of heme in signal transduction, functional regulation and as catalytic centres. Chem Soc Rev 2019; 48:5624-5657. [DOI: 10.1039/c9cs00268e] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Molecular mechanisms of unprecedented functions of exchangeable/labile heme and heme proteins including transcription, DNA binding, protein kinase activity, K+ channel functions, cis–trans isomerization, N–N bond formation, and other functions are described.
Collapse
Affiliation(s)
- Toru Shimizu
- Department of Biochemistry
- Faculty of Science
- Charles University
- Prague 2
- Czech Republic
| | - Alzbeta Lengalova
- Department of Biochemistry
- Faculty of Science
- Charles University
- Prague 2
- Czech Republic
| | - Václav Martínek
- Department of Biochemistry
- Faculty of Science
- Charles University
- Prague 2
- Czech Republic
| | - Markéta Martínková
- Department of Biochemistry
- Faculty of Science
- Charles University
- Prague 2
- Czech Republic
| |
Collapse
|
25
|
Circadian clock disruption by selective removal of endogenous carbon monoxide. Sci Rep 2018; 8:11996. [PMID: 30097595 PMCID: PMC6086871 DOI: 10.1038/s41598-018-30425-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/30/2018] [Indexed: 11/13/2022] Open
Abstract
Circadian rhythms are regulated by transcription-translation feedback loops (TTFL) of clock genes. Previous studies have demonstrated that core transcriptional factors, NPAS2 and CLOCK, in the TTFL can reversibly bind carbon monoxide (CO) in vitro. However, little is known about whether endogenous CO, which is continuously produced during a heme metabolic process, is involved in the circadian system. Here we show that selective removal of endogenous CO in mice considerably disrupts rhythmic expression of the clock genes. A highly selective CO scavenger, hemoCD1, which is a supramolecular complex of an iron(II)porphyrin with a per-O-methyl-β-cyclodextrin dimer, was used to remove endogenous CO in mice. Intraperitoneal administration of hemoCD1 to mice immediately reduced the amount of internal CO. The removal of CO promoted the bindings of NPAS2 and CLOCK to DNA (E-box) in the murine liver, resulting in up-regulation of the E-box-controlled clock genes (Per1, Per2, Cry1, Cry2, and Rev-erbα). Within 3 h after the administration, most hemoCD1 in mice was excreted in the urine, and heme oxygenase-1 (HO-1) was gradually induced in the liver. Increased endogenous CO production due to the overexpression of HO-1 caused dissociation of NPAS2 and CLOCK from E-box, which in turn induced down-regulation of the clock genes. The down-regulation continued over 12 h even after the internal CO level recovered to normal. The late down-regulation was ascribed to an inflammatory response caused by the endogenous CO reduction. The CO pseudo-knockdown experiments provided the clear evidence that endogenous CO contributes to regulation in the mammalian circadian clock.
Collapse
|
26
|
Sun H, Zhang Y, Shi Y, Li Y, Li W, Wang Z. Evolution of the CLOCK and BMAL1 genes in a subterranean rodent species (Lasiopodomys mandarinus). Int J Biol Macromol 2018; 109:932-940. [DOI: 10.1016/j.ijbiomac.2017.11.076] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/10/2017] [Accepted: 11/12/2017] [Indexed: 12/01/2022]
|
27
|
Scheidt WR, Li J, Sage JT. What Can Be Learned from Nuclear Resonance Vibrational Spectroscopy: Vibrational Dynamics and Hemes. Chem Rev 2017; 117:12532-12563. [PMID: 28921972 PMCID: PMC5639469 DOI: 10.1021/acs.chemrev.7b00295] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Nuclear resonance
vibrational spectroscopy (NRVS; also known as
nuclear inelastic scattering, NIS) is a synchrotron-based method that
reveals the full spectrum of vibrational dynamics for Mössbauer
nuclei. Another major advantage, in addition to its completeness (no
arbitrary optical selection rules), is the unique selectivity of NRVS.
The basics of this recently developed technique are first introduced
with descriptions of the experimental requirements and data analysis
including the details of mode assignments. We discuss the use of NRVS
to probe 57Fe at the center of heme and heme protein derivatives
yielding the vibrational density of states for the iron. The application
to derivatives with diatomic ligands (O2, NO, CO, CN–) shows the strong capabilities of identifying mode
character. The availability of the complete vibrational spectrum of
iron allows the identification of modes not available by other techniques.
This permits the correlation of frequency with other physical properties.
A significant example is the correlation we find between the Fe–Im
stretch in six-coordinate Fe(XO) hemes and the trans Fe–N(Im)
bond distance, not possible previously. NRVS also provides uniquely
quantitative insight into the dynamics of the iron. For example, it
provides a model-independent means of characterizing the strength
of iron coordination. Prediction of the temperature-dependent mean-squared
displacement from NRVS measurements yields a vibrational “baseline”
for Fe dynamics that can be compared with results from techniques
that probe longer time scales to yield quantitative insights into
additional dynamical processes.
Collapse
Affiliation(s)
- W Robert Scheidt
- Department of Chemistry and Biochemistry, University of Notre Dame , Notre Dame, Indiana 46556 United States
| | - Jianfeng Li
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences , YanQi Lake, HuaiRou District, Beijing 101408, China
| | - J Timothy Sage
- Department of Physics and Center for Interdisciplinary Research on Complex Systems, Northeastern University , 120 Forsyth Street, Boston, Massachusetts 02115, United States
| |
Collapse
|
28
|
Schallner N, Lieberum JL, Gallo D, LeBlanc RH, Fuller PM, Hanafy KA, Otterbein LE. Carbon Monoxide Preserves Circadian Rhythm to Reduce the Severity of Subarachnoid Hemorrhage in Mice. Stroke 2017; 48:2565-2573. [PMID: 28747460 PMCID: PMC5575974 DOI: 10.1161/strokeaha.116.016165] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 06/23/2017] [Accepted: 06/28/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Subarachnoid hemorrhage (SAH) is associated with a temporal pattern of stroke incidence. We hypothesized that natural oscillations in gene expression controlling circadian rhythm affect the severity of neuronal injury. We moreover predict that heme oxygenase-1 (HO-1/Hmox1) and its product carbon monoxide (CO) contribute to the restoration of rhythm and neuroprotection. METHODS Murine SAH model was used where blood was injected at various time points of the circadian cycle. Readouts included circadian clock gene expression, locomotor activity, vasospasm, neuroinflammatory markers, and apoptosis. In addition, cerebrospinal fluid and peripheral blood leukocytes from SAH patients and controls were analyzed for clock gene expression. RESULTS Significant elevations in the clock genes Per-1, Per-2, and NPAS-2 were observed in the hippocampus, cortex, and suprachiasmatic nucleus in mice subjected to SAH at zeitgeber time (ZT) 12 when compared with ZT2. Clock gene expression amplitude correlated with basal expression of HO-1, which was also significantly greater at ZT12. SAH animals showed a significant reduction in cerebral vasospasm, neuronal apoptosis, and microglial activation at ZT12 compared with ZT2. In animals with myeloid-specific HO-1 deletion (Lyz-Cre-Hmox1fl/fl ), Per-1, Per-2, and NPAS-2 expression was reduced in the suprachiasmatic nucleus, which correlated with increased injury. Treatment with low-dose CO rescued Lyz-Cre-Hmox1fl/fl mice, restored Per-1, Per-2, and NPAS-2 expression, and reduced neuronal apoptosis. CONCLUSIONS Clock gene expression regulates, in part, the severity of SAH and requires myeloid HO-1 activity to clear the erythrocyte burden and inhibit neuronal apoptosis. Exposure to CO rescues the loss of HO-1 and thus merits further investigation in patients with SAH.
Collapse
Affiliation(s)
- Nils Schallner
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - Judith-Lisa Lieberum
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - David Gallo
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - Robert H LeBlanc
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - Patrick M Fuller
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - Khalid A Hanafy
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - Leo E Otterbein
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.).
| |
Collapse
|
29
|
Schallner N, Lieberum JL, Gallo D, LeBlanc RH, Fuller PM, Hanafy KA, Otterbein LE. Carbon Monoxide Preserves Circadian Rhythm to Reduce the Severity of Subarachnoid Hemorrhage in Mice. Stroke 2017; 48:2565-2573. [PMID: 28747460 DOI: 10.1161/strokeaha.116.016165.carbon] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 06/23/2017] [Accepted: 06/28/2017] [Indexed: 05/28/2023]
Abstract
BACKGROUND AND PURPOSE Subarachnoid hemorrhage (SAH) is associated with a temporal pattern of stroke incidence. We hypothesized that natural oscillations in gene expression controlling circadian rhythm affect the severity of neuronal injury. We moreover predict that heme oxygenase-1 (HO-1/Hmox1) and its product carbon monoxide (CO) contribute to the restoration of rhythm and neuroprotection. METHODS Murine SAH model was used where blood was injected at various time points of the circadian cycle. Readouts included circadian clock gene expression, locomotor activity, vasospasm, neuroinflammatory markers, and apoptosis. In addition, cerebrospinal fluid and peripheral blood leukocytes from SAH patients and controls were analyzed for clock gene expression. RESULTS Significant elevations in the clock genes Per-1, Per-2, and NPAS-2 were observed in the hippocampus, cortex, and suprachiasmatic nucleus in mice subjected to SAH at zeitgeber time (ZT) 12 when compared with ZT2. Clock gene expression amplitude correlated with basal expression of HO-1, which was also significantly greater at ZT12. SAH animals showed a significant reduction in cerebral vasospasm, neuronal apoptosis, and microglial activation at ZT12 compared with ZT2. In animals with myeloid-specific HO-1 deletion (Lyz-Cre-Hmox1fl/fl ), Per-1, Per-2, and NPAS-2 expression was reduced in the suprachiasmatic nucleus, which correlated with increased injury. Treatment with low-dose CO rescued Lyz-Cre-Hmox1fl/fl mice, restored Per-1, Per-2, and NPAS-2 expression, and reduced neuronal apoptosis. CONCLUSIONS Clock gene expression regulates, in part, the severity of SAH and requires myeloid HO-1 activity to clear the erythrocyte burden and inhibit neuronal apoptosis. Exposure to CO rescues the loss of HO-1 and thus merits further investigation in patients with SAH.
Collapse
Affiliation(s)
- Nils Schallner
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - Judith-Lisa Lieberum
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - David Gallo
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - Robert H LeBlanc
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - Patrick M Fuller
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - Khalid A Hanafy
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.)
| | - Leo E Otterbein
- From the Department of Surgery (N.S., J.-L.L., D.G., L.E.O.) and Department of Neurology (R.H.L., P.M.F., K.A.H.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Anesthesiology and Critical Care, Medical Center-University Freiburg, Faculty of Medicine, Germany (N.S., J.-L.L.); and Aston University, Birmingham, United Kingdom (L.E.O.).
| |
Collapse
|
30
|
Klemz R, Reischl S, Wallach T, Witte N, Jürchott K, Klemz S, Lang V, Lorenzen S, Knauer M, Heidenreich S, Xu M, Ripperger JA, Schupp M, Stanewsky R, Kramer A. Reciprocal regulation of carbon monoxide metabolism and the circadian clock. Nat Struct Mol Biol 2017; 24:15-22. [PMID: 27892932 DOI: 10.1038/nsmb.3331] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 11/01/2016] [Indexed: 01/22/2023]
Abstract
Circadian clocks are cell-autonomous oscillators regulating daily rhythms in a wide range of physiological, metabolic and behavioral processes. Feedback of metabolic signals, such as redox state, NAD+/NADH and AMP/ADP ratios, or heme, modulate circadian rhythms and thereby optimize energy utilization across the 24-h cycle. We show that rhythmic heme degradation, which generates the signaling molecule carbon monoxide (CO), is required for normal circadian rhythms as well as circadian metabolic outputs. CO suppresses circadian transcription by attenuating CLOCK-BMAL1 binding to target promoters. Pharmacological inhibition or genetic depletion of CO-producing heme oxygenases abrogates normal daily cycles in mammalian cells and Drosophila. In mouse hepatocytes, suppression of CO production leads to a global upregulation of CLOCK-BMAL1-dependent circadian gene expression and dysregulated glucose metabolism. Together, our findings show that CO metabolism is an important link between the basic circadian-clock machinery, metabolism and behavior.
Collapse
Affiliation(s)
- Roman Klemz
- Laboratory of Chronobiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Silke Reischl
- Laboratory of Chronobiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Wallach
- Laboratory of Chronobiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nicole Witte
- Institute of Pharmacology, Center for Cardiovascular Research CCR, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Karsten Jürchott
- Laboratory of Chronobiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sabrina Klemz
- Laboratory of Chronobiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Veronika Lang
- Laboratory of Chronobiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Miriam Knauer
- Institute of Pharmacology, Center for Cardiovascular Research CCR, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Steffi Heidenreich
- Institute of Pharmacology, Center for Cardiovascular Research CCR, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Min Xu
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Jürgen A Ripperger
- Division of Biochemistry, Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Michael Schupp
- Institute of Pharmacology, Center for Cardiovascular Research CCR, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ralf Stanewsky
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Achim Kramer
- Laboratory of Chronobiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
31
|
Interactions Between the Circadian Clock and Heme Oxygenase in the Retina of Drosophila melanogaster. Mol Neurobiol 2016; 54:4953-4962. [PMID: 27520276 PMCID: PMC5533861 DOI: 10.1007/s12035-016-0026-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/01/2016] [Indexed: 01/14/2023]
Abstract
The Drosophila retina has an autonomous peripheral circadian clock in which the expression of the gene encoding heme oxygenase (HO) is under circadian control with the ho mRNA peaking at the beginning of the day and in the middle of the night. The function of HO in the retina is unknown, but we observed that it regulates the circadian clock and protects photoreceptors against DNA damage. The decline in HO level increases and decreases the expression of the canonical clock genes period (per) and Clock (Clk), respectively. The opposite result was observed after increasing HO expression. Among three products of HO activity—carbon monoxide (CO), ferrous ions, and biliverdin—the latter has no effect on per and Clk expressions, but CO exerts the same effect as the increase of ho expression. This suggests that HO action on the clock is mediated by CO, which may affect Clk expression during the day and the level of per expression. While ho expression is not stimulated by nitric oxide (NO), NO has the same effect on the clock as HO, increasing Clk expression and decreasing the expression of per.
Collapse
|
32
|
A heme-binding domain controls regulation of ATP-dependent potassium channels. Proc Natl Acad Sci U S A 2016; 113:3785-90. [PMID: 27006498 DOI: 10.1073/pnas.1600211113] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Heme iron has many and varied roles in biology. Most commonly it binds as a prosthetic group to proteins, and it has been widely supposed and amply demonstrated that subtle variations in the protein structure around the heme, including the heme ligands, are used to control the reactivity of the metal ion. However, the role of heme in biology now appears to also include a regulatory responsibility in the cell; this includes regulation of ion channel function. In this work, we show that cardiac KATP channels are regulated by heme. We identify a cytoplasmic heme-binding CXXHX16H motif on the sulphonylurea receptor subunit of the channel, and mutagenesis together with quantitative and spectroscopic analyses of heme-binding and single channel experiments identified Cys628 and His648 as important for heme binding. We discuss the wider implications of these findings and we use the information to present hypotheses for mechanisms of heme-dependent regulation across other ion channels.
Collapse
|
33
|
Carter EL, Gupta N, Ragsdale SW. High Affinity Heme Binding to a Heme Regulatory Motif on the Nuclear Receptor Rev-erbβ Leads to Its Degradation and Indirectly Regulates Its Interaction with Nuclear Receptor Corepressor. J Biol Chem 2015; 291:2196-222. [PMID: 26670607 DOI: 10.1074/jbc.m115.670281] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Indexed: 01/11/2023] Open
Abstract
Rev-erbα and Rev-erbβ are heme-binding nuclear receptors (NR) that repress the transcription of genes involved in regulating metabolism, inflammation, and the circadian clock. Previous gene expression and co-immunoprecipitation studies led to a model in which heme binding to Rev-erbα recruits nuclear receptor corepressor 1 (NCoR1) into an active repressor complex. However, in contradiction, biochemical and crystallographic studies have shown that heme decreases the affinity of the ligand-binding domain of Rev-erb NRs for NCoR1 peptides. One explanation for this discrepancy is that the ligand-binding domain and NCoR1 peptides used for in vitro studies cannot replicate the key features of the full-length proteins used in cellular studies. However, the combined in vitro and cellular results described here demonstrate that heme does not directly promote interactions between full-length Rev-erbβ (FLRev-erbβ) and an NCoR1 construct encompassing all three NR interaction domains. NCoR1 tightly binds both apo- and heme-replete FLRev-erbβ·DNA complexes; furthermore, heme, at high concentrations, destabilizes the FLRev-erbβ·NCoR1 complex. The interaction between FLRev-erbβ and NCoR1 as well as Rev-erbβ repression at the Bmal1 promoter appear to be modulated by another cellular factor(s), at least one of which is related to the ubiquitin-proteasome pathway. Our studies suggest that heme is involved in regulating the degradation of Rev-erbβ in a manner consistent with its role in circadian rhythm maintenance. Finally, the very slow rate constant (10(-6) s(-1)) of heme dissociation from Rev-erbβ rules out a prior proposal that Rev-erbβ acts as an intracellular heme sensor.
Collapse
Affiliation(s)
- Eric L Carter
- From the Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Nirupama Gupta
- From the Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Stephen W Ragsdale
- From the Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
34
|
Shimizu T, Huang D, Yan F, Stranava M, Bartosova M, Fojtíková V, Martínková M. Gaseous O2, NO, and CO in signal transduction: structure and function relationships of heme-based gas sensors and heme-redox sensors. Chem Rev 2015; 115:6491-533. [PMID: 26021768 DOI: 10.1021/acs.chemrev.5b00018] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Toru Shimizu
- †Department of Cell Biology and Genetics and Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, Guangdong 515041, China
- ‡Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague 2 128 43, Czech Republic
- §Research Center for Compact Chemical System, National Institute of Advanced Industrial Science and Technology (AIST), Sendai 983-8551, Japan
| | - Dongyang Huang
- †Department of Cell Biology and Genetics and Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Fang Yan
- †Department of Cell Biology and Genetics and Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Martin Stranava
- ‡Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague 2 128 43, Czech Republic
| | - Martina Bartosova
- ‡Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague 2 128 43, Czech Republic
| | - Veronika Fojtíková
- ‡Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague 2 128 43, Czech Republic
| | - Markéta Martínková
- ‡Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague 2 128 43, Czech Republic
| |
Collapse
|
35
|
Sun F, Cheng Y, Chen C. Regulation of heme biosynthesis and transport in metazoa. SCIENCE CHINA-LIFE SCIENCES 2015; 58:757-64. [PMID: 26100009 DOI: 10.1007/s11427-015-4885-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/22/2015] [Indexed: 02/08/2023]
Abstract
Heme is an iron-containing tetrapyrrole that plays a critical role in regulating a variety of biological processes including oxygen and electron transport, gas sensing, signal transduction, biological clock, and microRNA processing. Most metazoan cells synthesize heme via a conserved pathway comprised of eight enzyme-catalyzed reactions. Heme can also be acquired from food or extracellular environment. Cellular heme homeostasis is maintained through the coordinated regulation of synthesis, transport, and degradation. This review presents the current knowledge of the synthesis and transport of heme in metazoans and highlights recent advances in the regulation of these pathways.
Collapse
Affiliation(s)
- FengXiu Sun
- College of Life Sciences and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China
| | | | | |
Collapse
|
36
|
Simcox JA, Mitchell TC, Gao Y, Just SF, Cooksey R, Cox J, Ajioka R, Jones D, Lee SH, King D, Huang J, McClain DA. Dietary iron controls circadian hepatic glucose metabolism through heme synthesis. Diabetes 2015; 64:1108-19. [PMID: 25315005 PMCID: PMC4375081 DOI: 10.2337/db14-0646] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The circadian rhythm of the liver maintains glucose homeostasis, and disruption of this rhythm is associated with type 2 diabetes. Feeding is one factor that sets the circadian clock in peripheral tissues, but relatively little is known about the role of specific dietary components in that regard. We assessed the effects of dietary iron on circadian gluconeogenesis. Dietary iron affects circadian glucose metabolism through heme-mediated regulation of the interaction of nuclear receptor subfamily 1 group d member 1 (Rev-Erbα) with its cosuppressor nuclear receptor corepressor 1 (NCOR). Loss of regulated heme synthesis was achieved by aminolevulinic acid (ALA) treatment of mice or cultured cells to bypass the rate-limiting enzyme in hepatic heme synthesis, ALA synthase 1 (ALAS1). ALA treatment abolishes differences in hepatic glucose production and in the expression of gluconeogenic enzymes seen with variation of dietary iron. The differences among diets are also lost with inhibition of heme synthesis with isonicotinylhydrazine. Dietary iron modulates levels of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), a transcriptional activator of ALAS1, to affect hepatic heme. Treatment of mice with the antioxidant N-acetylcysteine diminishes PGC-1α variation observed among the iron diets, suggesting that iron is acting through reactive oxygen species signaling.
Collapse
Affiliation(s)
- Judith A Simcox
- Department of Biochemistry, University of Utah, Salt Lake City, UT
| | | | - Yan Gao
- Department of Biochemistry, University of Utah, Salt Lake City, UT
| | - Steven F Just
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Robert Cooksey
- Veterans Administration Research Service, VA Salt Lake City Health Care System, Salt Lake City, UT
| | - James Cox
- Department of Biochemistry, University of Utah, Salt Lake City, UT
| | - Richard Ajioka
- Department of Biochemistry, University of Utah, Salt Lake City, UT
| | - Deborah Jones
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Soh-Hyun Lee
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Daniel King
- Department of Biochemistry, University of Utah, Salt Lake City, UT
| | - Jingyu Huang
- Department of Biochemistry, University of Utah, Salt Lake City, UT
| | - Donald A McClain
- Department of Biochemistry, University of Utah, Salt Lake City, UT Department of Internal Medicine, University of Utah, Salt Lake City, UT Veterans Administration Research Service, VA Salt Lake City Health Care System, Salt Lake City, UT
| |
Collapse
|
37
|
Crane BR, Young MW. Interactive features of proteins composing eukaryotic circadian clocks. Annu Rev Biochem 2015; 83:191-219. [PMID: 24905781 DOI: 10.1146/annurev-biochem-060713-035644] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Research into the molecular mechanisms of eukaryotic circadian clocks has proceeded at an electrifying pace. In this review, we discuss advances in our understanding of the structures of central molecular players in the timing oscillators of fungi, insects, and mammals. A series of clock protein structures demonstrate that the PAS (Per/Arnt/Sim) domain has been used with great variation to formulate the transcriptional activators and repressors of the clock. We discuss how posttranslational modifications and external cues, such as light, affect the conformation and function of core clock components. Recent breakthroughs have also revealed novel interactions among clock proteins and new partners that couple the clock to metabolic and developmental pathways. Overall, a picture of clock function has emerged wherein conserved motifs and structural platforms have been elaborated into a highly dynamic collection of interacting molecules that undergo orchestrated changes in chemical structure, conformational state, and partners.
Collapse
Affiliation(s)
- Brian R Crane
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853;
| | | |
Collapse
|
38
|
Hough MA, Andrew CR. Cytochromes c': Structure, Reactivity and Relevance to Haem-Based Gas Sensing. Adv Microb Physiol 2015; 67:1-84. [PMID: 26616515 DOI: 10.1016/bs.ampbs.2015.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cytochromes c' are a group of class IIa cytochromes with pentacoordinate haem centres and are found in photosynthetic, denitrifying and methanotrophic bacteria. Their function remains unclear, although roles in nitric oxide (NO) trafficking during denitrification or in cellular defence against nitrosoative stress have been proposed. Cytochromes c' are typically dimeric with each c-type haem-containing monomer folding as a four-α-helix bundle. Their hydrophobic and crowded distal sites impose severe restrictions on the binding of distal ligands, including diatomic gases. By contrast, NO binds to the proximal haem face in a similar manner to that of the eukaryotic NO sensor, soluble guanylate cyclase and bacterial analogues. In this review, we focus on how structural features of cytochromes c' influence haem spectroscopy and reactivity with NO, CO and O2. We also discuss the relevance of cytochrome c' to understanding the mechanisms of gas binding to haem-based sensor proteins.
Collapse
|
39
|
Gustafson CL, Partch CL. Emerging models for the molecular basis of mammalian circadian timing. Biochemistry 2014; 54:134-49. [PMID: 25303119 PMCID: PMC4303291 DOI: 10.1021/bi500731f] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mammalian circadian timekeeping arises from a transcription-based feedback loop driven by a set of dedicated clock proteins. At its core, the heterodimeric transcription factor CLOCK:BMAL1 activates expression of Period, Cryptochrome, and Rev-Erb genes, which feed back to repress transcription and create oscillations in gene expression that confer circadian timing cues to cellular processes. The formation of different clock protein complexes throughout this transcriptional cycle helps to establish the intrinsic ∼24 h periodicity of the clock; however, current models of circadian timekeeping lack the explanatory power to fully describe this process. Recent studies confirm the presence of at least three distinct regulatory complexes: a transcriptionally active state comprising the CLOCK:BMAL1 heterodimer with its coactivator CBP/p300, an early repressive state containing PER:CRY complexes, and a late repressive state marked by a poised but inactive, DNA-bound CLOCK:BMAL1:CRY1 complex. In this review, we analyze high-resolution structures of core circadian transcriptional regulators and integrate biochemical data to suggest how remodeling of clock protein complexes may be achieved throughout the 24 h cycle. Defining these detailed mechanisms will provide a foundation for understanding the molecular basis of circadian timing and help to establish new platforms for the discovery of therapeutics to manipulate the clock.
Collapse
Affiliation(s)
- Chelsea L Gustafson
- Department of Chemistry and Biochemistry, University of California , Santa Cruz, California 95064, United States
| | | |
Collapse
|
40
|
Ishimori K, Watanabe Y. Unique Heme Environmental Structures in Heme-regulated Proteins Using Heme as the Signaling Molecule. CHEM LETT 2014. [DOI: 10.1246/cl.140787] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
| | - Yuta Watanabe
- Department of Chemistry, Faculty of Science, Hokkaido University
| |
Collapse
|
41
|
Linder D, Silvernail NJ, Barabanschikov A, Zhao J, Alp EE, Sturhahn W, Sage JT, Scheidt WR, Rodgers KR. The diagnostic vibrational signature of pentacoordination in heme carbonyls. J Am Chem Soc 2014; 136:9818-21. [PMID: 24950373 PMCID: PMC4120987 DOI: 10.1021/ja503191z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Indexed: 11/28/2022]
Abstract
Heme-carbonyl complexes are widely exploited for the insight they provide into the structural basis of function in heme-based proteins, by revealing the nature of their bonded and nonbonded interactions with the protein. This report presents two novel results which clearly establish a FeCO vibrational signature for crystallographically verified pentacoordination. First, anisotropy in the NRVS density of states for ν(Fe-C) and δ(FeCO) in oriented single crystals of [Fe(OEP)(CO)] clearly reveals that the Fe-C stretch occurs at higher frequency than the FeCO bend and considerably higher than any previously reported heme carbonyl. Second, DFT calculations on a series of heme carbonyls reveal that the frequency crossover occurs near the weak trans O atom donor, furan. As ν(Fe-C) occurs at lower frequencies than δ(FeCO) in all heme protein carbonyls reported to date, the results reported herein suggest that they are all hexacoordinate.
Collapse
Affiliation(s)
- Douglas
P. Linder
- Department
of Chemistry and Biochemistry, North Dakota
State University, Fargo, North Dakota 58105, United States
| | - Nathan J. Silvernail
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre Dame, Indiana 46556, United States
| | | | - Jiyong Zhao
- Advanced
Photon Source, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - E. Ercan Alp
- Advanced
Photon Source, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Wolfgang Sturhahn
- Advanced
Photon Source, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - J. Timothy Sage
- Department
of Physics, Northeastern University, Boston, Massachusetts 02115, United States
| | - W. Robert Scheidt
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Kenton R. Rodgers
- Department
of Chemistry and Biochemistry, North Dakota
State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
42
|
Efimov I, Parkin G, Millett ES, Glenday J, Chan CK, Weedon H, Randhawa H, Basran J, Raven EL. A simple method for the determination of reduction potentials in heme proteins. FEBS Lett 2014; 588:701-4. [PMID: 24440354 PMCID: PMC3999514 DOI: 10.1016/j.febslet.2013.12.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/20/2013] [Accepted: 12/24/2013] [Indexed: 11/28/2022]
Abstract
A simple method for determination of heme protein reduction potentials is described. We use the method to determine reduction potentials for human NPAS2 and human CLOCK. The method can be easily applied to other heme proteins.
We describe a simple method for the determination of heme protein reduction potentials. We use the method to determine the reduction potentials for the PAS-A domains of the regulatory heme proteins human NPAS2 (Em = −115 mV ± 2 mV, pH 7.0) and human CLOCK (Em = −111 mV ± 2 mV, pH 7.0). We suggest that the method can be easily and routinely applied to the determination of reduction potentials across the family of heme proteins.
Collapse
Affiliation(s)
- Igor Efimov
- Department of Chemistry, Henry Wellcome Building, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Gary Parkin
- Department of Chemistry, Henry Wellcome Building, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Elizabeth S Millett
- Department of Chemistry, Henry Wellcome Building, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Jennifer Glenday
- Department of Chemistry, Henry Wellcome Building, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Cheuk K Chan
- Department of Chemistry, Henry Wellcome Building, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Holly Weedon
- Department of Chemistry, Henry Wellcome Building, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Harpreet Randhawa
- Department of Chemistry, Henry Wellcome Building, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Jaswir Basran
- Department of Biochemistry, Henry Wellcome Building, University of Leicester, University Road, Leicester LE1 9HN, United Kingdom
| | - Emma L Raven
- Department of Chemistry, Henry Wellcome Building, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom.
| |
Collapse
|
43
|
Kühl T, Wißbrock A, Goradia N, Sahoo N, Galler K, Neugebauer U, Popp J, Heinemann SH, Ohlenschläger O, Imhof D. Analysis of Fe(III) heme binding to cysteine-containing heme-regulatory motifs in proteins. ACS Chem Biol 2013; 8:1785-93. [PMID: 23730736 DOI: 10.1021/cb400317x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Regulatory heme binds to specific motifs in proteins and controls a variety of biochemical processes. Several of these proteins were recently shown to form complexes with ferric and/or ferrous heme via a cysteine residue as axial ligand. The objective of this study was to examine the heme-binding properties of a series of cysteine-containing peptides with focus on CP motif sequences. The peptides displayed different binding behavior upon Fe(III) heme application with characteristic wavelength shifts of the Soret band to 370 nm or 420-430 nm and in some cases to both wavelengths. Whereas for most of the peptides containing a cysteine only a shift to 420-430 nm was observed, CP-containing peptides exhibited a preference for a shift to 370 nm. Detailed structural investigation using Raman and NMR spectroscopy on selected representatives revealed different binding modes with respect to iron ion coordination, which reflected the results of the UV-vis studies. A predicted short sequence stretch derived from dipeptidyl peptidase 8 was additionally examined with respect to CP motif binding to heme on the peptide as well as on the protein level. The heme association was confirmed with the first solution structure of a CP-peptide-heme complex and, moreover, an inhibitory effect of Fe(III) heme on the enzyme's activity. The relevance of both the use of model compounds to elucidate the molecular mechanism underlying regulatory heme binding and its potential for the investigation of regulatory heme control is discussed.
Collapse
Affiliation(s)
- Toni Kühl
- Pharmaceutical
Chemistry I, Institute of Pharmacy, University of Bonn, Brühler Str. 7, D-53119 Bonn, Germany
| | - Amelie Wißbrock
- Pharmaceutical
Chemistry I, Institute of Pharmacy, University of Bonn, Brühler Str. 7, D-53119 Bonn, Germany
| | - Nishit Goradia
- Biomolecular NMR Spectroscopy, Leibniz Institute for Age Research−Fritz Lipmann Institute, Beutenbergstr. 11, D-07745 Jena, Germany
| | - Nirakar Sahoo
- Center of Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, Hans-Knöll-Str. 2, D-07745 Jena, Germany
| | - Kerstin Galler
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Erlanger Allee 101, D-07747
Jena, Germany
- Institute of Photonic Technology, Albert-Einstein-Str.
9, D-07745 Jena, Germany
| | - Ute Neugebauer
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Erlanger Allee 101, D-07747
Jena, Germany
- Institute of Photonic Technology, Albert-Einstein-Str.
9, D-07745 Jena, Germany
| | - Jürgen Popp
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Erlanger Allee 101, D-07747
Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4,
D-07743 Jena, Germany
| | - Stefan H. Heinemann
- Center of Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, Hans-Knöll-Str. 2, D-07745 Jena, Germany
| | - Oliver Ohlenschläger
- Biomolecular NMR Spectroscopy, Leibniz Institute for Age Research−Fritz Lipmann Institute, Beutenbergstr. 11, D-07745 Jena, Germany
| | - Diana Imhof
- Pharmaceutical
Chemistry I, Institute of Pharmacy, University of Bonn, Brühler Str. 7, D-53119 Bonn, Germany
| |
Collapse
|
44
|
Itoh R, Fujita KI, Mu A, Kim DHT, Tai TT, Sagami I, Taketani S. Imaging of heme/hemeproteins in nucleus of the living cells expressing heme-binding nuclear receptors. FEBS Lett 2013; 587:2131-6. [PMID: 23735699 DOI: 10.1016/j.febslet.2013.05.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 04/30/2013] [Accepted: 05/09/2013] [Indexed: 01/10/2023]
Abstract
Several factors involved in the core circadian rhythm are PAS domain proteins, one of which, neuronal PAS2 (NPAS2), contains a heme-binding motif. It is thought that heme controls the transcriptional activity of core circadian factors BMAL1-NPAS2, and that the heme-binding nuclear receptor REV-erbα negatively regulates the expression of BMAL1. To examine the role of heme in the nucleus, we expressed nuclear hemeproteins including the nuclear localization signal-added cytoglobin, NPAS2 and REV-erbα. Then, the living cells expressing these proteins were treated with 2',7'-dichlorodihydrofluorescin diacetate (DCFH-DA). The fluorescent signal derived from DCFH-DA was observed in the nucleus. When the cells were cultured with hemin, the signal of heme in the nucleus increased. Considering that DCFH-DA reacted with heme, we propose that the use of DCFH-DA could be useful in detection of the heme moiety of hemeprotein in vivo.
Collapse
Affiliation(s)
- Ryuhei Itoh
- Department of Biotechnology, Kyoto Institute of Technology, Kyoto 606-8585, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Gamble KL, Young ME. Metabolism as an integral cog in the mammalian circadian clockwork. Crit Rev Biochem Mol Biol 2013; 48:317-31. [PMID: 23594144 DOI: 10.3109/10409238.2013.786672] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Circadian rhythms are an integral part of life. These rhythms are apparent in virtually all biological processes studies to date, ranging from the individual cell (e.g. DNA synthesis) to the whole organism (e.g. behaviors such as physical activity). Oscillations in metabolism have been characterized extensively in various organisms, including mammals. These metabolic rhythms often parallel behaviors such as sleep/wake and fasting/feeding cycles that occur on a daily basis. What has become increasingly clear over the past several decades is that many metabolic oscillations are driven by cell-autonomous circadian clocks, which orchestrate metabolic processes in a temporally appropriate manner. During the process of identifying the mechanisms by which clocks influence metabolism, molecular-based studies have revealed that metabolism should be considered an integral circadian clock component. The implications of such an interrelationship include the establishment of a vicious cycle during cardiometabolic disease states, wherein metabolism-induced perturbations in the circadian clock exacerbate metabolic dysfunction. The purpose of this review is therefore to highlight recent insights gained regarding links between cell-autonomous circadian clocks and metabolism and the implications of clock dysfunction in the pathogenesis of cardiometabolic diseases.
Collapse
Affiliation(s)
- Karen L Gamble
- Division of Behavioral Neurobiology, Department of Psychiatry, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
46
|
Spiro TG, Soldatova AV, Balakrishnan G. CO, NO and O 2 as Vibrational Probes of Heme Protein Interactions. Coord Chem Rev 2013; 257:511-527. [PMID: 23471138 PMCID: PMC3587108 DOI: 10.1016/j.ccr.2012.05.008] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The gaseous XO molecules (X = C, N or O) bind to the heme prosthetic group of heme proteins, and thereby activate or inhibit key biological processes. These events depend on interactions of the surrounding protein with the FeXO adduct, interactions that can be monitored via the frequencies of the Fe-X and X-O bond stretching modes, νFeX and νXO. The frequencies can be determined by vibrational spectroscopy, especially resonance Raman spectroscopy. Backbonding, the donation of Fe dπ electrons to the XO π* orbitals, is a major bonding feature in all the FeXO adducts. Variations in backbonding produce negative νFeX/νXO correlations, which can be used to gauge electrostatic and H-bonding effects in the protein binding pocket. Backbonding correlations have been established for all the FeXO adducts, using porphyrins with electron donating and withdrawing substituents. However the adducts differ in their response to variations in the nature of the axial ligand, and to specific distal interactions. These variations provide differing vantages for evaluating the nature of protein-heme interactions. We review experimental studies that explore these variations, and DFT computational studies that illuminate the underlying physical mechanisms.
Collapse
Affiliation(s)
- Thomas G. Spiro
- Department of Chemistry, University of Washington Box 351700, Seattle, Washington 98195
| | | | - Gurusamy Balakrishnan
- Department of Chemistry, University of Washington Box 351700, Seattle, Washington 98195
| |
Collapse
|
47
|
Pond MP, Majumdar A, Lecomte JTJ. Influence of heme post-translational modification and distal ligation on the backbone dynamics of a monomeric hemoglobin. Biochemistry 2012; 51:5733-47. [PMID: 22775272 DOI: 10.1021/bi300624a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The cyanobacterium Synechococcus sp. PCC 7002 uses a hemoglobin of the truncated lineage (GlbN) in the detoxification of reactive species generated in the assimilation of nitrate. In view of a sensing or enzymatic role, several states of GlbN are of interest with respect to its structure-activity relationship. Nuclear magnetic resonance spectroscopy was applied to compare the structure and backbone dynamics of six GlbN forms differing in their oxidation state [Fe(II) or Fe(III)], distal ligand to the iron (histidine, carbon monoxide, or cyanide), or heme post-translational modification (b heme or covalently attached heme). Structural properties were assessed with pseudocontact shift calculations. (15)N relaxation data were analyzed by reduced spectral density mapping (picosecond to nanosecond motions) and by inspection of elevated R(2) values (microsecond to millisecond motions). On the picosecond to nanosecond time scale, GlbN exhibited little flexibility and was unresponsive to the differences among the various forms. Regions of slightly higher mobility were the CE turn, the EF loop, and the H-H' kink. In contrast, fluctuations on the microsecond to millisecond time scale depended on the form. Cyanide binding to the ferric state did not enhance motions, whereas reduction to the ferrous bis-histidine state resulted in elevated R(2) values for several amides. This response was attributed, at least in part, to a weakening of the distal histidine coordination. Carbon monoxide binding quenched some of these fluctuations. The results emphasized the role of the distal ligand in dictating backbone flexibility and illustrated the multiple ways in which motions are controlled by the hemoglobin fold.
Collapse
Affiliation(s)
- Matthew P Pond
- T. C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | |
Collapse
|
48
|
Iwadate R, Satoh Y, Watanabe Y, Kawai H, Kudo N, Kawashima Y, Mashino T, Mitsumoto A. Impairment of heme biosynthesis induces short circadian period in body temperature rhythms in mice. Am J Physiol Regul Integr Comp Physiol 2012; 303:R8-18. [DOI: 10.1152/ajpregu.00019.2011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It has been demonstrated that the function of mammalian clock gene transcripts is controlled by the binding of heme in vitro. To examine the effects of heme on biological rhythms in vivo, we measured locomotor activity (LA) and core body temperature (Tb) in a mouse model of porphyria with impaired heme biosynthesis by feeding mice a griseofulvin (GF)-containing diet. Mice fed with a 2.0% GF-containing diet (GF2.0) transiently exhibited phase advance or phase advance-like phenomenon by 1–3 h in terms of the biological rhythms of Tbor LA, respectively (both, P < 0.05) while mice were kept under conditions of a light/dark cycle (12 h:12 h). We also observed a transient, ∼0.3 h shortening of the period of circadian Tbrhythms in mice kept under conditions of constant darkness ( P < 0.01). Interestingly, the observed duration of abnormal circadian rhythms in GF2.0 mice lasted between 1 and 3 wk after the onset of GF ingestion; this finding correlated well with the extent of impairment of heme biosynthesis. When we examined the effects of therapeutic agents for acute porphyria, heme, and hypertonic glucose on the pathological status of GF2.0 mice, it was found that the intraperitoneal administration of heme (10 mg·kg−1·day−1) or glucose (9 g·kg−1·day−1) for 7 days partially reversed (50%) increases in urinary δ-aminolevulinic acids levels associated with acute porphyria. Treatment with heme, but not with glucose, suppressed the phase advance (-like phenomenon) in the diurnal rhythms ( P < 0.05) and restored the decrease of heme ( P < 0.01) in GF2.0 mice. These results suggest that impairments of heme biosynthesis, in particular a decrease in heme, may affect phase and period of circadian rhythms in animals.
Collapse
Affiliation(s)
- Reiko Iwadate
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Keio University, Minato-ku, Tokyo, Japan
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Chiba, Japan; and
| | - Yoko Satoh
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Chiba, Japan; and
| | - Yukino Watanabe
- Faculty of Pharmaceutical Sciences, Josai University, Sakado, Saitama, Japan
| | - Hiroshi Kawai
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Chiba, Japan; and
| | - Naomi Kudo
- Faculty of Pharmaceutical Sciences, Josai University, Sakado, Saitama, Japan
| | - Yoichi Kawashima
- Faculty of Pharmaceutical Sciences, Josai University, Sakado, Saitama, Japan
| | - Tadahiko Mashino
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Keio University, Minato-ku, Tokyo, Japan
| | - Atsushi Mitsumoto
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Chiba, Japan; and
| |
Collapse
|
49
|
Bowman LAH, McLean S, Poole RK, Fukuto JM. The diversity of microbial responses to nitric oxide and agents of nitrosative stress close cousins but not identical twins. Adv Microb Physiol 2012; 59:135-219. [PMID: 22114842 DOI: 10.1016/b978-0-12-387661-4.00006-9] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nitric oxide and related nitrogen species (reactive nitrogen species) now occupy a central position in contemporary medicine, physiology, biochemistry, and microbiology. In particular, NO plays important antimicrobial defenses in innate immunity but microbes have evolved intricate NO-sensing and defense mechanisms that are the subjects of a vast literature. Unfortunately, the burgeoning NO literature has not always been accompanied by an understanding of the intricacies and complexities of this radical and other reactive nitrogen species so that there exists confusion and vagueness about which one or more species exert the reported biological effects. The biological chemistry of NO and derived/related molecules is complex, due to multiple species that can be generated from NO in biological milieu and numerous possible reaction targets. Moreover, the fate and disposition of NO is always a function of its biological environment, which can vary significantly even within a single cell. In this review, we consider newer aspects of the literature but, most importantly, consider the underlying chemistry and draw attention to the distinctiveness of NO and its chemical cousins, nitrosonium (NO(+)), nitroxyl (NO(-), HNO), peroxynitrite (ONOO(-)), nitrite (NO(2)(-)), and nitrogen dioxide (NO(2)). All these species are reported to be generated in biological systems from initial formation of NO (from nitrite, NO synthases, or other sources) or its provision in biological experiments (typically from NO gas, S-nitrosothiols, or NO donor compounds). The major targets of NO and nitrosative damage (metal centers, thiols, and others) are reviewed and emphasis is given to newer "-omic" methods of unraveling the complex repercussions of NO and nitrogen oxide assaults. Microbial defense mechanisms, many of which are critical for pathogenicity, include the activities of hemoglobins that enzymically detoxify NO (to nitrate) and NO reductases and repair mechanisms (e.g., those that reverse S-nitrosothiol formation). Microbial resistance to these stresses is generally inducible and many diverse transcriptional regulators are involved-some that are secondary sensors (such as Fnr) and those that are "dedicated" (such as NorR, NsrR, NssR) in that their physiological function appears to be detecting primarily NO and then regulating expression of genes that encode enzymes with NO as a substrate. Although generally harmful, evidence is accumulating that NO may have beneficial effects, as in the case of the squid-Vibrio light-organ symbiosis, where NO serves as a signal, antioxidant, and specificity determinant. Progress in this area will require a thorough understanding not only of the biology but also of the underlying chemical principles.
Collapse
Affiliation(s)
- Lesley A H Bowman
- Department of Molecular Biology and Biotechnology, The University of Sheffield, Sheffield, UK
| | | | | | | |
Collapse
|
50
|
Unwinding the differences of the mammalian PERIOD clock proteins from crystal structure to cellular function. Proc Natl Acad Sci U S A 2012; 109:3311-6. [PMID: 22331899 DOI: 10.1073/pnas.1113280109] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The three PERIOD homologues mPER1, mPER2, and mPER3 constitute central components of the mammalian circadian clock. They contain two PAS (PER-ARNT-SIM) domains (PAS-A and PAS-B), which mediate homo- and heterodimeric mPER-mPER interactions as well as interactions with transcription factors and kinases. Here we present crystal structures of PAS domain fragments of mPER1 and mPER3 and compare them with the previously reported mPER2 structure. The structures reveal homodimers, which are mediated by interactions of the PAS-B β-sheet surface including a highly conserved tryptophan (Trp448(mPER1), Trp419(mPER2), Trp359(mPER3)). mPER1 homodimers are additionally stabilized by interactions between the PAS-A domains and mPER3 homodimers by an N-terminal region including a predicted helix-loop-helix motive. We have verified the existence of these homodimer interfaces in solution and inside cells using analytical gel filtration and luciferase complementation assays and quantified their contributions to homodimer stability by analytical ultracentrifugation. We also show by fluorescence recovery after photobleaching analyses that destabilization of the PAS-B/tryptophan dimer interface leads to a faster mobility of mPER2 containing complexes in human U2OS cells. Our study reveals structural and quantitative differences between the homodimeric interactions of the three mouse PERIOD homologues, which are likely to contribute to their distinct clock functions.
Collapse
|