Review

DOI:10.1002/ijch.201600023 Thioether Coordination Chemistry for Molecular Imaging of Copper in Biological Systems

Karla M. Ramos-Torres,[a] Safacan Kolemen,[a] and Christopher J. Chang*[a, b, c] We dedicate this submission to Harry Gray,who has beenateacher,mentor,friend, and inspiration to us all.

Abstract:Copper is an essentialelement in biological sys- ular imaging agents for this metal, drawing inspirationfrom tems. Its potent redox activity renders it necessary for life, both biological binding motifsand synthetic model com- but at the same time, misregulationofits cellular pools can plexes that reveal thioether coordination as ageneral design lead to oxidative stress implicated in aging and various dis- strategy for selective andsensitive copper recognition. In ease states. Copper is commonly thought of as astatic co- this review,wesummarize some contributions,primarily factor buried in protein active sites;however,evidence of from our own laboratory,onfluorescence- and magnetic res- amore loosely bound, labile pool of copper has emerged. onance-based molecular-imaging probes for studying To help identify and understand new roles for dynamic copper in living systemsusingthioether coordination copper pools in biology,wehave developed selective molec- chemistry. Keywords: copper · fluorescent probes · molecular imaging · MR probes · thioether coordination

1. Introduction Copper is an essential element for life.[1] Owing to its as lipolysis,[20] and the activation of the mitogen-activated potent redox activity,the participationofthis metalas protein (MAP) kinase pathway relevant in normal physi- acatalytic and structural cofactor in enzymes that func- ology and in oncogenic serine/threonine-protein kinase tion in biological processes spanning energy generation, B-Raf (BRAF) signalingand tumorogenesis.[21,22] oxygen transport, cellularmetabolism, and signal trans- Against this backdrop,molecularimaging provides duction renders it vital for the life of eukaryotic organ- aversatile approach that can be used to monitorlabile isms.[2] However, this same redox promiscuity, when mis- metal fluxes in real timewith spatial and temporal resolu- regulated, can also lead to aberrant generation of reactive tion.[23–25] In combination with complementary biochemi- oxygen species (ROS) that have been linked to aging and cal and genetic methods,aswell as direct readoutsof different disease states,including genetic disorders like total metal content,small-molecule probes with different Menkes[3–5] and Wilsons[6–8] diseases,neurodegenerative readoutsignals, including fluorescence,colorimetric,and diseases like Alzheimers,[9–12] Parkinsons,[13] and Hun- magnetic resonance (MR) relaxivitymodalities,can be tingtons[14] diseases,and metabolic disorders such as dia- used effectively as pilotscreening tools for quicklyassess- betes and obesity.[15–17] Forthis reason,copper and other ing metalstatus in different physiological states.Assuch, redox-active metals have been canonically thought of as the development of metal-selective small-molecule buried cofactors within enzymeactive sites and part of probesprovides apotentially powerful toolbox that atightly bound metal pool.However, amore loosely allows for the mappingoflabile metalpools and the bound pool, termed the labile pool, in whichcopper and study of the roles of these dynamic fluxes.Inthis review, other metals can dynamically exchange withligands on timescales commensurate with signaling, has been ob- [a] K. M. Ramos-Torres, S. Kolemen, C. J. Chang served. This metal pool, in which rapid ligandexchange Department of Chemistry and changes in metal concentration gradients can mediate University of California Berkeley CA 94704 (USA) signaling processes,has been extensively studied in e-mail:[email protected] regard to redox-inactive alkali, alkaline earth and transi- [b] C. J. Chang tion metals,such as calcium,sodium, potassium,and zinc. Department of Molecular and Cell Biology Nonetheless,recent work from our lab and others has re- University of California Berkeley vealed that despite its high reactivity,dynamic copper CA 94704 (USA) fluxes are observed in important physiologicalprocesses, [c] C. J. Chang [18] such as neuronal calcium signaling pathways, spontane- Howard Hughes Medical Institute ous activity in neural circuits,[19] metabolic processes such Tel.:(+1) 510-642-4704

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim 724 Review we focus on the use of thioether coordination chemistry 2. Bioinspired Strategies for Selective Copper as ageneral design strategy for achieving copper-selective Recognition probes for molecular imaging –optical and magnetic res- onance imaging, as an illustrative example of opportuni- Owing to environmentalheterogeneities in biological sys- ties at the interface between and tems,the development of small-moleculeprobes for bio- biology. logical use is inherently challenging. Because transition metals such ascopper are far less abundantthan their biologically relevant alkali/alkaline earth counterparts,se- lectivity poses one of the main challenges in probe design.Additionally for copper,redox specificity is re- Karla Ramos-Torres graduated from the quired,asitcan exist in two major oxidation states,Cu(I) University of Puerto Rico, RíoPiedras and Cu(II). In the cellular setting, most of the copper in 2011 with aB.S. in Chemistry.She is that is undergoing dynamicexchange is in the Cu(I)state, currently aPh.D. candidate at UC Ber- keley,where she works in the laboratory owing to the reducing intracellularenvironment (average [26] of Prof. Chris Chang. Her current re- potential of –0.25 V) buffered in partbythe thiol-con- search interests include the develop- taining low-molecular weight peptide glutathione (GSH). ment of molecular probes for the de- Preceding import across the plasma membrane, extracel- tection of biological copper.Outside of lular Cu(II) is reduced to the Cu(I)state by an insuffi- research, Karla enjoys playing volley- ciently understood mechanismthat presumably involves ball, hiking, and dancing. membrane reductases,[27] afterwhich intracellular accu- mulation of the metal is achieved primarily by the high-affinity Cu(I)-specific channel copper transporter Safacan Kolemen is apostdoctoral re- 1(CTR1). Theprevalence of the intracellular Cu(I) state searcher in the laboratory of Prof. Chris is further evidenced by the identification of Cu(I)-specific Chang at UC Berkeley.His work focus- metallochaperones such as Atox-1,[28] which delivers es on development of pH-sensitive and Cu(I) to the p-type ATPases ATP7A/B,the copper chap- copper-selective molecular probes. He erone for superoxide dismutase (CCS),[29,30] and the vari- received his Ph.D. in 2014 from Bilkent ous Sco[31] and Cox[32] copper chaperones for mitochondri- University,Turkey,where he worked al copper pools to metallate enzymes such as cytochrome under the supervision of Prof. Engin U. c oxidase (CcO). Thespecificity of this emerging class of Akkaya on designing photosensitizers for photodynamic therapy applications. proteins for copper binding is crucial for maintaining cel- lular homeostasisofthis metal.

2.1 Metal and Ligand Considerationsfor Copper Coordination Chris Chang is the Class of 1942 Chair Inspection of biological copper sites can give us insight Professor in the Departments of into its preferred modes of binding. Coordination in pro- Chemistry and Molecular and Cell Biol- ogy at UC Berkeley,Howard Hughes tein copper-binding sites is typically dominated by histi- Medical Institute Investigator,and Fac- dine,cysteine, and methionine residues.The makeupof ulty Scientist in the Chemical Sciences each ligand set depends on the specific role,copper trans- Division of Lawrence Berkeley National port or enzymatic activity,ofthe bindingsite.However, Laboratory.HeisaSenior Editor of the general use of these amino-acid side chains can be ACS Central Science.Chris received his viewed as also governed by the hard and soft acids and B.S. and M.S. from Caltech in 1997, bases (HSAB) principle.[33,34] In line with this theory,soft working with Harry Gray,studied as acid Cu(I) and borderline Cu(II) will preferentially bind aFulbright scholar in Strasbourg, ligandswith borderline to soft basicities,respectively, France with Jean-Pierre Sauvage, and leadingtoafavored use of amino-acid residues with ni- received his Ph.D. from MIT in 2002 with Dan Nocera. After post- doctoral studies with Steve Lippard, Chris joined the UC Berkeley trogen and sulfur donor .Additionally,the number Faculty in 2004. His laboratory focuses on chemical biology and in- of donors,aswell as the coordination geometry,can also organic chemistry with particular interests in molecular imaging impart adegree of selectivity between the possible metal and catalysis applied to neuroscience and sustainable energy.His oxidation states for these bindingsites.Even though group’s work has been recognized by honors from the Dreyfus, Cu(I) does not have ageometric preference based on Beckman, Sloan, and Packard Foundations, Amgen, Astra Zeneca, alack of ligand field stabilizationenergy (LFSE), it is and Novartis, Technology Review,ACS (Cope Scholar,Eli Lilly, often foundinlow-coordinate systemswith 2, 3, or 4li- Nobel Laureate Signature, Baekeland), RSC ( gands in linear, trigonal planar, or tetrahedralgeometries. Chemistry), SBIC, and the Blavatnik Foundation. Conversely,LFSE for Cu(II) results in higher coordinate

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 725 Review

(4, 5, or 6ligands) systemswith square planar and square 2.2 Biological Systems RevealMethionineasanIdeal Ligand pyramidal geometries. for Cu(I) Binding Moreover, the chemicalidentity of the ligating groups can also lead to favored oxidation states for copper in the Theuse of sulfur-based ligands enhances the selectivity binding site.Coordination environmentsincluding histi- for the cuprous over the cupric state,asshownbyhigher dine and cysteine residues are pH-dependent, as deproto- Cu(II) affinity when including nitrogen donorsinsynthet- [35] nation of the imidazole (pKa ~6and 14) and thiol(pKa ~ ic copper ligands. However,the use of the sulfur donor 8.5) side chains lead to anionic donors.While the chemi- cysteine over methionine (or vice versa)incuproprotein cal identity of the coordinating groups and their arrange- bindingsites can be associated with that specific proteins ment in aparticular three-dimensional geometry confer chemical role and surrounding biological environment. adegree of specificity for metal binding,itisimportant to Cysteine copper bindingispH-dependent and susceptible remember that proteins also control selectivity of their to redox activity,but thesesame residues achieve tighter binding sites by additional secondary sphere coordination bindingtothe metal than methionine,owing to their effects that can further alter the local protein environ- anioniccharge. In contrast, methionine binds with weaker ment upon metal binding. Forexample,inthe electron- affinities than the cysteine thiolate,but at the same time, transferenzymeCu,Zn-superoxide dismutase (SOD1), varyingthe number of methionine ligands can tune the the bridging histidineresidue between the Cu and Zn affinityofthe bindingsite.Additionally,methionine-rich sites has an altered pKa,causing the imidazolate to form sites provide aunique environment that stabilizes Cu(I) at physiological pH. (Type II center,Figure 1). coordination, as well as excellent selectivity over other Furthermore,the ability of cysteines to formthe thio- biologically-relevant cationslike Zn(II) and Fe(II).[36,37] late renders them prone to oxidation, which can be corre- As aresult, methionine has emerged as an important lated to aprevalence of cysteine-rich copper sites in re- ligand for copper in biological systems.Copper centers in- ducing cellular compartments in comparisonwith more volvingmethionine residues are prevalent in systems oxidizingintracellularlocales.Incontrast, the thioether rangingfrom electron-transfer proteins to copper traffick- side chain in methionine providesaneutral donor with ing proteins.Even though most S-donors in electron- pH-independent coordination, as well as less susceptibili- transferprotein coppersites are thiolate-based,there are ty to oxidation. This inability to be ionized, in combina- afew examples of methionine copper interactions.Cupre- tion with alonger sidechain, also gives methionine doxins,such as plastocyanin and azurin,show the typical aslight hydrophobic nature. 2-histidine 1-cysteine copper coordination in atrigonal

Figure 1. Examples of copper-containing sites in Nature.

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 726 Review planar geometry. Additionally,these blue copper proteins for the weaker Cu(I)-binding affinity of thioethers,in possess aweakly bound methionine thioether sulfur in comparisonwith thiolates. the axial position[38,39] (Figure 1). Otherelectron-transfer proteins with methionine binding within their active sites include Cytochrome c oxidase(CcO), which has amethio- 2.3 Synthetic Compounds have Exploited Thioether Donorsfor [40] Copper Coordination nine ligandinits mixed valence CuA center. As with electron-transfer proteins,the copper trafficking counter- Theability of thioethers to coordinate to avariety of parts also show prevalent cysteine-thiolate coordination metals,and the effect of their moderate p-acidity[68] on to copper instead of the thioether moiety.Bis-cysteinate the stabilization of low metal oxidation states have been Cu(I) coordination is ahallmark of cytosolic copper liga- historically important in motivating the study of thioeth- tion, with many transport proteins exhibiting a CXXC er-metal complexes.The need for homoleptic thioether binding motif (Figure 1). Among the copper sensing and complexes,inorder to evaluate the effect of thioether li- trafficking proteins that exhibit this motif are the eukary- gation on the electronic structure of metal ,was satis- [29,30,41,42] [43–47] otic copper chaperones CCS and Atox1, as fied by the development of crown thioethers.Inseminal [48–50] well as the Cu(I)-binding P-type ATPases ATP7A/B. work, Cooper and coworkers studied the synthesis,con- In comparison with cytosolic Cu(I) binding, which occurs formationalanalysis and metal coordination of macrocy- in avery reducing environment, transport of extracellular cles includingthe 9S3, 12S3, 18S6, and 24S6 variations[69] copper into the cell is highlyassociated withmethionine (Figure 2). Extensive efforts on the investigationofdiffer- thioether binding. Thehomotrimeric copper transporter ent metalcomplexes of crown thioethers (including CTR1 containsacell surface-located N-terminal region nickel, cobalt, copper,ruthenium, rhodium, and silver) in- with histidineand methionine-rich domains(Metsmotif) dicate that thioethers present amarked preference for that are implicated in copper transport across the mem- the lower, “softer”metal oxidationstates.This behavior [51–53] brane (Figure 1). Methionine motifs (MX3M) in the manifests itself in the redoxproperties of the complexes, transmembrane region are also important in shuttling as well as in their magnetic and EPR features.[70] copper down the pore of the protein.[51,54] Perhaps the best-characterized methionine-copper co- ordination chemistry is that of protein networksinvolved in copper resistance,predominantly found in less redox- balanced spaces of prokaryotic organisms.Torelieve the cytosol of excess copper,bacteria export the metal using the ATPase CopA, whichbinds copper in asimilar fash- ion as ATP7A/B.[55] Forgram-negativebacteria,this de- toxifying process is not sufficient, as there is still aneed to manage the copper that accumulatesinthe periplasm. Acanonicalexample of periplasmic,copper-handling ma- chinery is the Cus pathway, an export system that is com- posed of three proteins,CusA, CusB,and CusC.[56,57] The CusA and CusB proteins possess three-coordinate me- Figure 2. Metal-binding (a) crown thioethers;and (b) mixed-donor [57,58] macrocyclic ligands, with NxS4 x and NxS5 x donor sets. thionine-only binding sites (Figure 1), whereas CusC À À does not appear to have metal-bindingfeatures as ob- served by crystallographic experiments.[59] In contrast, CusF,acopper chaperone for CusB,[60] binds copper in Anotherimportant avenue for motivationofthe study a2-methionine, 1-histidine bindingsite with atryptophan of thioether-metal complexation arose from the previous residue that provides acation-p interaction with the speculation (since confirmed)[71,72] of methionine copper metal[61,62] (Figure1). In the relatively less studied Pco coordination in blue copper proteins.Inefforts to eluci- and Cop copper resistance pathways,where proteins are date the origin of the unexpected redox, optical, and encoded by plasmid DNAincomparison with chromoso- EPR properties of these copper centers,Rorabacher and mal DNA, methionine coordinationisobservedinthe coworkers carriedout an impressiveset of studies on the Cu(I) binding sites of PcoC and CopC,but not in their copper complexes of macrocyclic tetra- and pentadentate separate Cu(II) sites that mainly offer Nand Odonors as thioethers,inwhich they showed that coordination to thi- ligands.[63–67] Copper-handling systems like the Cus path- oethersshifts the Cu(II/I) potential to morepositive way,aswell as the Pco/Cop copper resistance pathways, values,enhances the rate of Cu(II/I) redox self-exchange are characterized by higher-coordinate Cu(I) complexes kinetics,and generates unusually intense optical (three or four ligands), whencomparedwith the cysteine- bands.[73,74] Furthermore,studies on mixed-donormacro- [35,75] dominated environments of the cytosoliccopper proteins. cyclic ligandswith NxS4 x and NxS5 x donor sets This situation is in part due to the need to compensate (Figure 2) established impoÀ rtant trendsiÀ ncopper coordi-

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 727 Review nation:1)stability constants for Cu(II) complexes in- state in which the highest occupied molecular orbital crease by 5–6 ordersofmagnitude for each replacement (HOMO) of the donorlies in an energetically favorable of athioether sulfur donor by an amine nitrogen donor state to transfer an electron to the excited-state fluoro- ;and 2) the more positive Cu(II/I)redox potential phore and hence quench the fluorescence relaxation path- of macrocyclic donors with sulfur donors,comparedwith way of this state.Upon analytebinding, the energy level aliphatic nitrogen atoms, is credited to adestabilization of the HOMO of the electron-richdonor is lowered, caus- of the Cu(II) oxidation state,rather than astabilization ing electron transfer to be less thermodynamically fa- of the Cu(I) state.Additional studies of tripodal ligands vored, alleviating PeT,and thus restoringfluorescencein with mixed nitrogen and sulfur donors to investigate the this state. effect of binding geometryonelectron-transfer kinetics of Combining whathas been learned from the studies of [76,77] Cu(II/I) systems provided further insight on copper- macrocyclic and aliphatic NxS4 x and NxS5 x copper com- thioether coordination, demonstrating anarrow distribu- plexes,aswell as the characteÀ rizationÀ of numerous tion of the stability constants of Cu(I) complexes,incom- copper-bindingsitesinvolvedincopper homeostasis,we parison with the Cu(II) counterparts,and anoticeable reason that thioether-richligandscan satisfy the require- effect of the geometry on the Cu(II) complexes.Interest- ment for amine coordinationneeded for PeT modulation ingly,the compatibility of these ligands for copper sensing by metal binding while maintaining selectivity for Cu(I), has been established with regard to their better stability and as aresult, can be used as the receptorsofchoice for towards oxidation, in comparison with thiolate ligands, synthetic copper sensors. and in that the reported Kd values of these complexes fall 11 18 within the window of 10À M(GSH)to10À M(SOD1) for biological Cu(I) ligands.

+ 3.1 Use of aThioether-rich Receptor (NS4)for Cu Sensing: Design of aFirst-generation Cu+ Sensor 3. Fluorescence-based Copper(I) Sensing Probes Use of aborondipyrromethene (BODIPY) fluorophore Inspired by Naturesuse of thioether ligation, as well as coupled to an azatetrathia receptor (termed NS4’)gave the aforementioned work on copper coordination chemis- rise to Coppersensor 1(CS1),[85] the first Cu+ -responsive try,weand others have extensively employed thioether probe with visible excitationand emissionprofiles receptormotifstodevelop aseries of recognition-based (Figure 3). TheNS4’ receptor,anacyclic analog to the fluorescent probes for the detection of Cu(I).[78–80] Using Cu(I)-binding tetrathiaza crown ether used in the impor- this strategy,afluorophore reporter is coupled to achela- tant first small-molecule fluorescent sensorfor Cu(I), tor (receptor) that is specific for the metal of interest. CTAP-1,[86] binds the metal ion to produceaselective re- Once bound to the analyte, achange in the optical prop- sponse.CS1 features a10-fold increase in probe fluores- 12 erties of the sensor is observed, which can be reversed cence upon metalbinding with Kd =4”10À M. upon analyte dissociation. Foramore in-depth discussion (Figure 4) Additionally,the dye responds to exogenous on the mechanisms and modes of small-molecule fluores- CuCl2 addition to HEK 293T cells in confocal imagingex- cence sensing of metalcations, we refer to the following periments,where the fluorescence enhancement can be review articles.[81,82] reversed by the addition of amembrane-permeable Cu(I) Selectivity of the receptor for the metalofinterest can chelator. Theability of CS1 to reversibly detect labile be achieved by carefuldesign of the ligand using funda- copper in this system was validatedbyanindependent mental principles of coordination chemistry.Considering study;[87] however, its use to detect copper changes upon some of the basics of copper homeostasis (e.g.,sulfur co- supplementation with CuCl2 or Cu(gtsm)(Cu(II)-glyoxal- ordination, low coordination number), one can envision bis(N4-methyl-3-thiosemicarbazonato)) in other mamma- afew possible strategies for building receptors thatcan lian cell lines,includingCHO,M17, U87Mg,and SH- chelate Cu(I) with good selectivity and competitivebind- SY5Y,provedunsuccessful. These observations point to ing constants.The commonuse of aphotoinduced elec- the fact that there is no one-size-fits-all chemical probe tron transfer (PeT)quenching mechanismasaturn-on/off for every biological model, and therefore characterization switch for metal sensing sets up aparticular stipulation of the sensorinits intended biological or environmental for the receptor moiety;asubstituted amine is customari- system of use is necessary.Indeed, when used in combina- ly used as the PeT switchdue to its lone-pair interaction tion with complementary techniques,such as direct metal with the cationicanalyte.[82–84] In PeT-based fluorescent detection via inductively coupled plasma massspectrome- sensors,two important entities,the electron donor (the try (ICP-MS) and biochemical assayslike DNAmicroar- substituted amine) and the electron acceptor (theexcited- rays and protein profiling,CS1 has been applied to study state fluorophore), interact to produce an opticalre- copper dynamics in awide range of bacterial,[88] plant,[89] sponse upon analyte binding. In the unbound state,pho- and yeast[90–92] models of copper accumulation and misre- toexcitation of the system leads to acharge-separated gulation.

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 728 Review

Figure 3. Cu(I)-selective fluorescent probes bearing thioether receptors.

3.2 DesignofMore Sensitive Next-generation Sensors for Additionally,work from others has exploited the use of DetectionofEndogenous Levels of Cu+ in Living Cells and CS3 as acomplementarytool to study copper in other Tissue

Application of the NS4 receptor to differentfluorophore platformshas yielded next-generation probes that allow the detectionofendogenous copper levelsindifferent biological models.For example,replacement of the fluo- rine atoms in the BODIPYcore with the more electron- rich methoxy substituents afforded Coppersensor 3 (CS3)[18] (Figure 3). Increasing the electron density at the fluorophore (PeTacceptor component) resulted in aturn- on response enhancement (75-fold for CS3vs. 10-fold for CS1), as well as higher quantum yield (F=0.40 for CS3 vs. F=0.13 for CS1). These improvements,incombina- tion with atighter bindingaffinity for Cu(I) (Kd =9” 14 10À M), allowed for the visualization of endogenous levels of labile copper in HEK 293T that could be deplet- ed by treatmentwith the cell-impermeable chelator bath- ocuproine disulfonate (BCS). Additionally,CS3 was able to report on neuronal copper translocation upon depolari- zation with KCl. Thebiological interpretation suggested by these pilot experiments was confirmed by X-ray fluo- rescencemicroscopy (XFM) of analogous fixedsamples, which directly showed that copper moves from somatic cell bodies to peripheral dendritic processesupon stimu- Figure 4. Selective Cu(I) recognition by CS1 and turn-on response lation. (Figure 5A). in probe fluorescence.

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 729 Review

and further use of Ctrl-CS3, amatched control dye to CS3 that is unable to bind copper due to the replacement of the thioether motifswith methylene units (Figure 3), allowed for the assessment of fluorescenthot spots that were observed with CS3 (but not with the control probe) in higher frequencyinzinc-deficient cells than in control cells (Figure5B). In addition,CS3 (but not Ctrl-CS3) fluorescence was diminished upon copper chelation. Direct metal-analysis techniques like Nano-SIMS and X- ray absorption spectroscopy,incombination with supple- mentary biochemicalassays,were usedtoconfirm the ex- istence of these copper trapsand suggestthat the appar- ent reversible sequestration of copper in these compart- ments may play arole in minimizingprotein mismetalla- tion during zinc deficiency while maintaining copper stores for future use. Despite the utility of the BODIPY-based probesinvar- ious biological models,the use of this fluorophore scaf- fold has been limited in other applications.Its relatively high hydrophobicity has led to restricted implementation of the probes to study more complexsystems,such as tissue and cell types with differential hydrophilicities due to uneven distribution and staining, as well as limited photostability,particularly in the methoxy-substituted BODIPYforms, has hindered theiruse for prolonged imagingexperiments.Inefforts to improve the properties of these reagents,werecently developedaseries of Cu(I)-responsive probes based on the rhodolscaffold. This hybridfluorescein-rhodamine fluorophoreshows high tunability in visible excitation/emission profiles and signal-to-noise responses,and high optical brightness,as well as improved hydrophilicity and photostability.[98] Indeed,the use of different amine substituents on the xanthone core gave rise to the Copper Rhodol (CR) family.[19] Of the five initial sensors,Copper Rhodol3 (CR3) was the best performing, with a13-fold turn-on re- sponse to Cu(I) in vitro (Figure 3). Further modification to the pendant aryl ring by substitution of the ortho methyl,with abulkier trifluoromethyl analog,afforded Figure 5. (a) Labile copper pool detection in rat hippocampal CopperFluor 3(CF3, Figure 3). This CH3 to CF3 modifi- neuron cells with CS3 and XFM. (b) Monitoring copper accumula- cation introduces two advantages:1)non-radiative decay tion in zinc-limited and zinc-replete wild-type Chlamydomonas rein- pathways by rotations about the aryl-aryl bond that de- hardtii (CC-4532) cells by using CS3 and Ctrl-CS3. crease fluorescence quantum yield are diminished;and 2) reducedelectron density in the aryl ring (PeT donor com- biological models. Among these studies are includedthe ponent) favors PeT quenching of the unbound probe. evaluation of CTR2functions,[93,94] the use of the probe to Consequently,CF3 exhibitsanimproved 40-fold fluores- track the efficiency of copper depletionorsupplementa- cence enhancement upon Cu(I) bindingwith adissocia- [95] 13 tion treatments, and the implementation of the dye in tion constant Kd =3”10À M. Themore hydrophilic screening assays to studythe endogenous ATP7A-trans- nature of CR3and CF3, compared withCS3, was con- port of copper intolysosomes asaresponse to elevation firmed by measurements of apparent octanol/water distri- in copper levels.[96] More recently,acomprehensive suite bution coefficients (logD values of 0.96, 1.15,and 3.46, re- of techniques,which werepursued after observations with spectively,where larger values indicate greater hydropho- the CS3 probe,revealed anew organelle termed the “cu- bicity). prosome” that acts as storage for reversible sequestration Theimproved properties of the CF3 probe allowed its of copper in response to zinc deficiencyinthe green use for assessing labile Cu(I) dynamicsindissociatedhip- algae Chlamydomonas reinhardtii.[97] Thedevelopment pocampal neuronalcultures and retinal tissue by one- and

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 730 Review

volved in spontaneous correlated activity,confirmed by calcium imaging. Taken together, the results identify afundamentalphysiologicalrole for copper in neural function. We have recently expanded this work to red- shifted versions that enable real-time monitoring of copper fluxes in other cell types,such as adipocytes, and these chemical probes have helped identify an essential role for copper in lipolysis,amajor metabolic process in the body for burning fat.[20] Finally,anadded challenge for fluorescentprobe design lies in the development of sensors with specific lo- calization to assess exclusive pools of metal in subcellular compartments.Toaddress this issue,further modifications to the BODIPY core afforded Mito-CS1,[99] amitochond- rially-targetable probe that makes use of atriphenyl phosphonium targeting group for specific subcellular lo- calization (Figure3).[100] Mito-CS1 features a10-fold turn- 12 Figure 6. Two-photon imaging of:(a) CF3;and (b) Ctrl-CF3 in hip- on response upon Cu(I)ligation with Kd =7”10À M, lo- pocampal neurons in the absence and presence of acopper chela- calizes to themitochondria in HEK 293T cells and pa- tor BCS. tient fibroblasts, and responds well in cellulo in copper supplementation and chelation experiments.Incombina- tion with geneticmodels of copper misregulation and two-photon microscopy.Molecularimaging of CF3 and direct bulk metalmeasurements,the use of Mito-CS1 the non-Cu(I)-responsive Ctrl-CF3 analog on dissociated helped establish the concept of aprioritization of mito- hippocampal neurons and mouseretinal tissue that had chondrial copper homeostasis over other cellular com- been acutely treated withthe copper chelator BCS partments,even in situations of systemic copper deficien- showed decreasedfluorescence for CF3, but an un- cy and mitochondrial metallochaperonemalfunction. Spe- changed signalfor the controldye (Figure 6). Afterob- cifically,molecular-imaging experiments with patient-de- serving the presence of alabile copper pool in both sys- rived fibroblasts with mutations in the copper exporter tems,wesought to probe the effects of pharmacological ATP7A showed arelativeincreaseinprobe fluorescence (chelation with BCS)and genetic (CTR1 knockout) alter- that was confirmed by inductivelycoupled plasma optical ation of these copper pools on spontaneous activity,afun- emission spectroscopy (ICP-OES). Patient fibroblasts damental property of developingneuralcircuits.Inboth with mutations in mitochondrialcopper metallochaper- cases,disruption of copper homeostasis resulted in in- ones SCO1 and SCO2 showednoappreciable difference, creases in event frequencyand the fraction of cells in- compared with control fibroblasts in Mito-CS1 signal or

Figure 7. Live-cell molecular imaging of mitochondrial copper homeostasis in:(a) control;(b) ATP7a;(c) SCO1;and (d) SCO2 patient fibro- blasts with Mito-CS1. (e) Mean fluorescence intensities of (a)–(d). Measured total copper level in:(f) patient fibroblasts;and (g) mitochon- dria.

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 731 Review

ICP-OES measurements on isolated mitochondria, but esters to aid in cell permeability and retention afforded showed decreased total copper levels vs.wild-type mea- CS790AM, which showed increases in fluorescence signal sured by ICP-OES (Figure 7). Combining these comple- in HEK 293T cells that were pretreatedwith CuCl2,com- mentary techniques,Mito-CS1 imaging and ICP-OES re- pared with the control, as well as areversal of this fluo- sults indicate that mitochondrialcopper homeostasis is rescence enhancement when the copper chelator tris[2- prioritized over other compartments,even in the course (ethylthio)ethyl]amine (NS3)was added. Theprobe was of overall copper deficiency,presumably to preserve CcO further capable of reporting changes in live hairless SKH- and SOD activity. 1mice upon copper supplementation with CuCl2 and che- lation with ATN-224,the choline salt of tetrathiomolyb- date currently in development as atreatment for Wilsons [103] 3.3 Use of an NS3 Receptor for SensingLabile Copper Pools in disease, marking the first exampleoflive-mouse Living Animals copper imaging with afluorescent sensor (Figure 8). Fur- thermore,CS790AM was used to monitor copperlevels / Owing to the poor tissue-penetrating ability of visible in Atp7bÀ À mice,amurinemodelthat is metabolically light and the desire to apply Cu(I)-responsiveprobesto and phenotypically similar to Wilsonsdisease due to the study thickerspecimens like tissue and whole animals,ef- inactivation of the ATP7B gene and subsequent anoma- forts to develop sensors with longer-wavelength/near-in- lous accumulation of copper in severaltissues.[6] Live- frared (NIR)excitation profiles have been applied.We animalimaging showed increased fluorescence in the combinedanNS3 modified thioether receptorplatform livers of mutant mice relative to those from wild-type first reported by Fahrni[101] to the NIR fluorophore scaf- mice,which could be reversed with copper chelator treat- fold cyanine 7toproduce Coppersensor 790 (CS790)[102] ment. Thecollective results point to apath forwardfor (Figure 3). CS790 has aselective 15-fold turn-on response the application of fluorescentcopper probes to study to Cu(I) with 760 nm excitationand a790 nm emission physiology,disease diagnosis,and monitor treatment of profile.Capping of the carboxylates with acetoxymethyl copper imbalance in whole animals.

4. Magnetic Resonance-based Copper-sensing Probes Whereas metal-responsivefluorescent sensors provide agreat tooltoassess relative metal levels in living biolog- ical systems from the cell to the tissue to the organism level, the translational application to medicine can be limited by the intrinsic properties of the use of visible to near-infraredlightasareadoutfor metalactivity.Inthis regard,magnetic resonance imaging (MRI)isapowerful, clinically-used molecular-imaging technique that allows for the capture of three-dimensional images of organisms with up to cellular resolution.[104] Even though the most common contrast observed in MR images results from water molecules in differentenvironments,the use of contrast agents with paramagnetic metals,such as Mn2+ , Mn3+ ,Fe3+,Cu2+ ,and Gd3+ ,can be implemented to en- hance image contrast. Of these paramagnetic metals,high spin Gd3 + is particularly well suited forthis application and is indeed used in 40–50%ofall clinical MRI applica- tions.[105] Theimage enhancing abilitiesofcoordination complexes of these metals relies on the ability to effi- ciently relax nearby nuclei and increase the relaxation rates of water protons.[106] Modulationofthe relaxivity, the efficiency of acontrast agent to enhance the relaxa- tion rate,byaspecificanalyte can therefore be applied to Figure 8. (a) Live SKH-1 mice imaging with fluorescent CS790AM asensingstrategy. probe. 1: vehicle only;2:vehicle and CS790AM;3:vehicle, CuCl , 2 Thedesign of metal-responsive MR probes can imple- and CS790AM;4:CuCl2,ATN-224, and CS790AM;and 5: vehicle, ATN-224, and CS790AM. (b) Calculated total photon flux from each ment the idea of relaxivitymodulation to afford atoolbox mouse, 5minutes after the injection of probe. that can be useful in studying complexcontributionsof

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 732 Review

Figure 9. Copper selective MR-contrast agents with thioether receptors. metals to physiology and disease.There are anumberof duced variousthioether-rich receptorstothe Gd3+ con- ways to affect the degreeofrelaxivity in acontrast agent trast agent scaffold through a2,6-dimethylpyridine linker sensor, includingthe numberofinner-sphere water mole- to afford Copper-Gad probes 2–6 (CG2–CG6, cules (q-modulation)aswell as rotational tumbling time Figure 9).[113] Thepyridyl linker acts as acoordination

(tR-modulation). Foramore in-depth discussion on the switch,where in the absenceofanalyte,the apo-receptor development and criteria for responsiveMRI contrast caps the Gd3+ center to minimize the interactionwith agents as chemical sensors,werefer the reader to the fol- inner-sphere water molecules,thus lowering the proton lowing review articles.[107–109] relaxivity.Upon analyte binding, the linker switchesto aconformation that lowers the steric bulk around the Gd3+,allowing for inner-sphere access of water and pro- 4.1 DevelopmentofaMR-contrast Agent for Selective ducing higher relaxivity.Introduction of thioether donors Copper(II) Sensing shifted the selectivity towards Cu+ binding in CG2–CG5, Inspired by the seminal work of Meadeand coworkers in with relaxivity increases ranging from 92%to360%, the development of EGad, a“smart” contrast agent that where higher turn-on responses correlate to higherS/N reports on b-galactosidase activity,[110,111] our laboratory donor ratios.CG6 had aunique and equal response to reported the first copper-responsive MR-sensor Copper- Cu+ and Cu2+ due to aquick redox equilibrationthat Gad 1(CG1)[112] (Figure 9). CG1 features aGd3+ contrast favors the highercopper oxidation state,possibly due to agent platformcoupled to an iminodiacetate ligandfor the N/O/S donor set provided by the receptor. Thecom- Cu2+ binding,the prevalent oxidationstate in extracellu- positionand number of potential donorsinthe ligand set lar fluids. In the absence of Cu2+ ,the anionic carboxylate influences the bindingaffinitiesaswell, where com- donors of the receptorhinder inner-sphere water access pounds with higher thioether coordination CG2 and CG3 3+ + 14 to the Gd thereby minimizingrelaxivity.Upon binding presented higher Cu bindingaffinities, Kd =3.7”10À M 2 + 3+ 13 to Cu ,the Gd is less sterically hindered, leading to and 2.6”10À M, respectively.Incomparison, sensors increasedinner-sphere water access and proton relaxivity with only three potential donors (CG4 and CG5) showed + 11 affordingaturn-on response.CG1 presents a41%in- lower affinities to Cu (Kd =1.4”10À Mand 3.2” 2+ 11 crease in relaxivity upon selective binding of Cu with 10À M, respectively). Thetetradentate N/S/O donor 2+ Kd =167 48 mM. We note that while binding was mostly system in CG6 afforded ahigher affinity for Cu (Kd = Æ 2+ 16 selective to Cu ,the turn-on response was partially 9.9”10À M) in comparison with the first-generation muted in the presenceof10-fold excess Zn2+.Neverthe- probe CG1. Additionally,tovalidate the potential use of less,the response to Cu2+ was stronger, owing to its these sensors for molecular-imaging applications,wees- higher affinity on the Irving-Williams series,and provided tablished that CG complexes are capable of visualizing aprototype candidate for prospectiveMR-based copper changes in biologically relevant copper levels in T1 phan- imaging in living systems. tom imagesatclinical fieldstrengths. We next evaluatedthe effects of anionsonrelaxivity changes for the CG probes.Relaxivity increases upon 4.2 DesignofMR-contrast Agents with Thioether-rich Receptors copper bindingseen in the presenceofphosphate anions for Copper (I) Sensing are unaffected;however, carboxylate-typeanions,includ- Following the initial results from the CG1 probe,we ing citrate,lactate,and carbonate,presumably bind to the sought to improve upon the relatively modest sensitivity Gd3+ contrast agent core and yield significantly lower in- and change in relaxivity,aswell as selectivity,versus creasesinrelaxivity upon analyte binding. To minimize Zn2+.Additionally,wedesigned anew family of copper- anion sensitivity,wedesigned Copper-Gad 7(CG7),[114] responsive MR probes to be activated by Cu + and/or an analog to CG2 where the acetate groups on the Gd3+ Cu2+ to potentially trackcopper in itstwo biologically contrast agent core aresubstituted by hexanedioate deriv- relevantoxidation states.Toachieve this goal, we intro- atives to shield the paramagnetic metal from biologically

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 733 Review abundantanionsbystericand electrostatic action (Figure 9). This modification led to acomparable relaxivi- ty turn-on ratiotoCG2 (340%for CG7 vs.360%for CG2), as well as an unperturbed selective binding of Cu+ over other biologically relevant cations in the N2S2 recep- tor.However, the Cu + response for CG7 is not signifi- cantly affected by carboxylate or phosphate anions,re- vealing that the installationofperipheral carboxylate functionalities improves anion compatibility for MR- based copper sensing.

4.3 DesignofaCopper-responsive MR Agent for Biological Imaging As the CG series of MR-based probes features excellent in vitro properties,wewere eager to exploretheir use in living systems, namelyinin cellulo applications.However, due to the large size of contrast agent probes (likeother typical Gd3+-containing MR contrast agents suchasGd- DTPAand Gd-DOTA), we reasoned and confirmed that the Cu+-responsiveCGprobes have poor cell membrane permeability and are largely confined to the extracellular space.Tocircumvent this issue,weintroduced apolyargi- nine tag on the CG2 scaffold to improve cellular uptake of the probe.Synthesis of Arg8CG2 resulted in acell-per- meable copper-responsiveMR-based probe with compa- rable spectroscopic properties and response to Cu+ that allowed for the visualization of biological perturbations of copper levels in amurine Menkes disease model cell [115] line. Indeed, Arg8CG2 showed greater cellular uptake compared with CG2 in HEK 293T cells as confirmedby ICP-MS studies of the lysedcells.Superior in cellulo re- laxivity of Arg8CG2 over CG2 was also established by copper supplementation and chelation experiments where

HEK 293T cells treated witheither CuCl2 or the copper chelatorBCS and later treated with probe (Arg8CG2 or CG2) showed more pronounced differences in relaxivity (increases or decreasesinthe case of supplementation or chelationrespectively) for Arg8CG2. Relaxivity measure- ments of the Menkes model fibroblast line WG1005, which bears amutation in the Atp7a gene,comparedwith control fibroblastsMCH58, showed clear differences be- tween the two cell lines,asreportedbyArg8CG2. Fur- thermore,phantom imaging experiments of the two cell lines treated with copper or copper chelator resultedin clear contrast variations,pointing to the applicability of the MR-based probefor in vivo imaging (Figure 10) and representingaunique application for copper-responsive MRI. Figure 10. (a) Structure of the Arg8CG2 construct. (b) Cellular uptake of CG2 (black) and Arg8CG2 (gray) in HEK 293T cells at dif- ferent dosing concentrations. Cells were lysed with RIPAbuffer after probe incubation and analyzed by ICP-MS. (c) Relaxivity meas- 5. Concluding Remarks urements of CG2 and Arg8CG2 in HEK 293T cells. Bars represent the incubation of cells with control vehicle (black), copper (light gray),

Theuse of thioether coordination chemistry for the selec- or BCS (dark grey). (d) T1-weighed phantom images of MCH58 and tive ligation of copper has been exploited by biology and WG1005 cells incubated with CG2 or Arg8CG2. syntheticchemists alike.Structural and spectroscopic

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 734 Review characterization of cuproprotein binding sites,incombi- [6] S. Lutsenko, Biochem. Soc. Trans. 2008, 36,1233–1238. nation with the study of synthetic small-molecule model [7] D. Huster, M. Hoppert, S. Lutsenko,J.Zinke,C.Lehmann, complexes,has revealedthe privileged natureofthe thio- J. Mossner, F. Berr, K. Caca, Gastroenterology 2003, 124, 335–345. ether moiety,via methionine coordination, as an impor- [8] D. Huster, Ann. N. Y. Acad. Sci. 2014, 1314,37–44. tant strategy for pH-independent and oxidation-resistant [9] S. Ayton, P. Lei, A. I. Bush, Free Radical Biol. Med. 2013, binding of the cuprous ion. Consequently,this type of 62,76–89. binding has been extensively implemented in the develop- [10] K. J. Barnham, C. L. Masters,A.I.Bush, Nat. Rev.Drug ment of copper-specific recognition moieties for molecu- Discovery 2004, 3,205–214. lar-imaging applications.Indeed, the diverse array of [11] M. G. Savelieff,S.Lee,Y.Liu, M. H. Lim, ACSChem. small-moleculefluorescent copper sensors with varying Biol. 2013, 8,856–865. [12] K. E. Matlack, D. F. Tardiff,P.Narayan, S. Hamamichi, properties (visible to NIR-excitationprofiles,signal-to- K. A. Caldwell, G. A. Caldwell, S. Lindquist, Proc. Natl. noise contrast, hydrophilicity,etc.) has led to the identifi- Acad. Sci. U.S.A. 2014, 111,4013–4018. cation of novel biological roles of copper asasignaling [13] W. I. Vonk, V. Kakkar, P. Bartuzi, D. Jaarsma, R. Berger, entity,inaddition to its canonical rolesasastatic cofactor M. H. Hofker, L. W. Klomp,C.Wijmenga, H. H. Kampin- within protein active sites.Furthermore,the use of thio- ga, B. van de Sluis, PLoS One 2014, 9,e92408. ether coordination of copper in different imaging modali- [14] G. Xiao,Q.Fan, X. Wang, B. Zhou, Proc. Natl. Acad. Sci. ties with more translatable potential, such as MRI,has U.S.A. 2013, 110,14995–15000. [15] J. L. Burkhead, S. Lutsenko,inLipid Metabolism (Ed.: pointed the way towards possible applications for disease R. V. Baez), InTech, 2013,DOI:10.5772/51819. diagnosis and treatment monitoring. [16] D. Huster, T. D. Purnat, J. L. Burkhead, M. Ralle,O. Despitethe success of thioether coordination of copper Fiehn, F. Stuckert, N. E. Olson, D. Teupser, S. Lutsenko, J. as astrategy for selective metal sensing,the structural va- Biol. Chem. 2007, 282,8343–8355. riety of the receptormoieties has been ratherlimited to [17] D. Huster, S. Lutsenko, Mol. BioSyst. 2007, 3,816–824. the use of macrocyclic or aliphatic amines containing thi- [18] S. C. Dodani, D. W. Domaille,C.I.Nam, E. W. Miller, oether donors. As such, apparent binding affinities of the L. A. Finney,S.Vogt, C. J. Chang, Proc. Natl. Acad. Sci. U.S.A. 2011, 108,5980–5985. resultingsensors,which can be narrowly alteredbyinclu- [19] S. C. Dodani, A. Firl, J. Chan, C. I. Nam, A. T. Aron, C. S. sion or modification of different reporter moieties (i.e., Onak, K. M. Ramos-Torres,J.Paek, C. M. Webster, M. B. fluorophores), leave roomfor future innovation. Indeed, Feller,C.J.Chang, Proc. Natl. Acad. Sci. U.S.A. 2014, 111, development of new ligands to explore the effects of 16280–16285. binding geometryand ligand topology/architecture on the [20] L. Krishnamoorthy,J.A.J.Cotruvo,J.Chan, H. Kaluar- affinity and selective bindingofcopper in biological sys- achchi, A. Muchenditsi, V. S. Pendyala, S. Jia, A. T. Aron, tems is aworthwhile goal. Theresulting expansionofthe M. N. Vander Wal, T. Guan, L. P. Smaga, S. S. Farhi, E. J. New,S.Lutsenko,C.J.Chang, Nat. Chem. Biol. 2016, chemicaltoolbox of molecular probes will undoubtedly DOI:10.1038/nchembio.2098. lead to new opportunities to discover and study new biol- [21] M. L. Turski, D. C. Brady,H.J.Kim, B. E. Kim, Y. Nose, ogy of copper and other metals. C. M. Counter, D. R. Winge,D.J.Thiele, Mol. Cell. Biol. 2012, 32,1284–1295. [22] D. C. Brady,M.S.Crowe,M.L.Turski, G. A. Hobbs,X. Acknowledgements Yao, A. Chaikuad, S. Knapp,K.Xiao,S.L.Campbell, D. J. Thiele,C.M.Counter, Nature 2014, 509,492–496. We thank the National Institutes of Health (GM 79465) [23] E. L. Que,D.W.Domaille,C.J.Chang, Chem. Rev. 2008, for supporting our work on metalimaging probes.K.M. 108,1517–1549. R.-T.was partially supported by aChemical Biology [24] D. W. Domaille,E.L.Que,C.J.Chang, Nat. Chem. Biol. Training Grantfrom the NIH (T32 GM066698). S. K. 2008, 4,168–175. [25] J. Chan, S. C. Dodani, C. J. Chang, Nat. Chem. 2012, 4, thanks TUBITAK for postdoctoralsupport. C. J. C. is an 973–984. Investigator of the Howard Hughes Medical Institute. [26] Y. M. Go,D.P.Jones, Biochim. Biophys.Acta 2008, 1780, 1273–1290. [27] S. Lutsenko, Curr.Opin. Chem. Biol. 2010, 14,211–217. References [28] Y. Hatori, S. Lutsenko, Antioxid. Redox Signaling 2013, 19, 945–957. [1] S. J. Lippard, J. M. Berg, Principles of Bioinorganic [29] V. C. Culotta, L. W. J. Klomp,J.Strain, R. L. B. Casareno, Chemistry,University Science Books,Mill Valley,CA, B. Krems,J.D.Gitlin, J. Biol. Chem. 1997, 272,23469– 1994. 23472. [2] H. Tapiero,D.M.Townsend, K. D. Tew, Biomed. Pharmac- [30] T. D. Rae,P.J.Schmidt, R. A. Pufahl, V. C. Culotta, T. V. other. 2003, 57,386–398. O’Halloran, Science 1999, 284,805–808. [3] C. Vulpe,B.Levinson, S. Whitney,S.Packman, J. Gitschi- [31] S. C. Leary,P.A.Cobine,B.A.Kaufman, G. H. Guercin, er, Nat. Genet. 1993, 3,7–13. A. Mattman, J. Palaty,G.Lockitch, D. R. Winge,P.Rustin, [4] S. G. Kaler, Nat. Rev.Neurol. 2011, 7,15–29. R. Horvath, E. A. Shoubridge, Cell Metab. 2007, 5,9–20. [5] S. La Fontaine,J.F.Mercer, Arch. Biochem. Biophys. 2007, [32] F. A. Punter, D. L. Adams,D.M.Glerum, Hum. Genet. 463,149–167. 2000, 107,69–74.

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 735 Review

[33] R. G. Pearson, J. Am. Chem. Soc. 1963, 85,3533–3539. [61] I. R. Loftin, S. Franke,N.J.Blackburn, M. M. McEvoy, [34] S. Ahrland, J. Chatt, N. R. Davies, Q. Rev.Chem. Soc. Protein Sci. 2007, 16,2287–2293. 1958, 12,265–276. [62] Y. Xue,A.V.Davis,G.Balakrishnan, J. P. Stasser, B. M. [35] B. C. Westerby,K.L.Juntunen, G. H. Leggett, V. B. Pett, Staehlin, P. Focia, T. G. Spiro,J.E.Penner-Hahn, T. V. M. J. Koenigbauer, M. D. Purgett, M. J. Taschner, L. A. O’Halloran, Nat. Chem. Biol. 2008, 4,107–109. Ochrymowycz, D. B.Rorabacher, Inorg. Chem. 1991, 30, [63] K. Peariso,D.L.Huffman, J. E. Penner-Hahn, T. V. O’Hal- 2109–2120. loran, J. Am. Chem. Soc. 2003, 125,342–343. [36] J. Reedijk, J. Inorg. Biochem. 2012, 115,182–185. [64] F. Arnesano,L.Banci, I. Bertini, S. Mangani, A. R. [37] A. V. Davis,T.V.O’Halloran, Nat. Chem. Biol. 2008, 4, Thompsett, Proc. Natl. Acad. Sci. U.S.A. 2003, 100,3814– 148–151. 3819. [38] P. M. Colman, H. C. Freeman, J. M. Guss,M.Murata, [65] F. Arnesano,L.Banci, I. Bertini, A. R. Thompsett, Struc- V. A. Norris,J.A.M.Ramshaw,M.P.Venkatappa, Nature ture 2002, 10,1337–1347. 1978, 272,319–324. [66] F. Arnesano,L.Banci, I. Bertini, I. C. Felli, C. Luchinat, [39] H. Nar, A. Messerschmidt, R. Huber, M. Vandekamp, A. R. Thompsett, J. Am. Chem. Soc. 2003, 125,7200–7208. G. W. Canters, J. Mol. Biol. 1991, 221,765–772. [67] L. Zhang, M. Koay,M.J.Maher, Z. Xiao,A.G.Wedd, J. [40] T. Tsukihara, H. Aoyama, E. Yamashita, T. Tomizaki, H. Am. Chem. Soc. 2006, 128,5834–5850. Yamaguchi, K. Shinzawaitoh, R. Nakashima, R. Yaono,S. [68] S. G. Murray,F.R.Hartley, Chem. Rev. 1981, 81,365–414. Yoshikawa, Science 1995, 269,1069–1074. [69] S. R. Cooper, Acc. Chem. Res. 1988, 21,141–146. [41] A. L. Lamb,A.K.Wernimont, R. A. Pufahl, T. V. O’Hal- [70] S. R. Cooper,S.C.Rawle, Struct. Bonding 1990, 72,1–72. loran, A. C. Rosenzweig, Biochemistry 2000, 39,1589– [71] J. M. Guss,H.C.Freeman, J. Mol. Biol. 1983, 169,521– 1595. 563. [42] A. L. Lamb,A.K.Wernimont, R. A. Pufahl, V. C. Culotta, [72] E. T. Adman, R. E. Stenkamp,L.C.Sieker, L. H. Jensen, J. Mol. Biol. 1978, 123,35–47. T. V. O’Halloran, A. C. Rosenzweig, Nat. Struct. Biol. 1999, 6,724–729. [73] D. B.Rorabacher, M. J. Martin, M. J. Koenigbauer,M. [43] A. K. Wernimont, D. L. Huffman, A. L. Lamb,T.V. Malik, R. R. Schroeder,J.F.Endicott, L. A. Ochrymowycz, in Copper Coordination Chemistry:Biochemical and Inor- O’Halloran, A. C. Rosenzweig, Nat. Struct. Biol. 2000, 7, ganic Perspectives 766–771. (Eds.: K. D. Karlin, J. Zubieta), Adenine Press,Guilderland, NY, 1983,p.167. [44] I. Anastassopoulou, L. Banci, I. Bertini, F. Cantini, E. Kat- [74] L. L. Diaddario,E.R.Dockal, M. D. Glick, L. A. Ochry- sari, A. Rosato, Biochemistry 2004, 43,13046–13053. mowycz, D. B. Rorabacher, Inorg. Chem. 1985, 24,356– [45] I. Hamza, M. Schaefer, L. W. J. Klomp,J.D.Gitlin, Proc. 363. Natl. Acad. Sci. U.S.A. 1999, 96,13363–13368. [75] M. M. Bernardo,M.J.Heeg,R.R.Schroeder,L.A. [46] D. Larin, C. Mekios,K.Das,B.Ross,A.S.Yang, T. C. Gil- Ochrymowycz, D. B.Rorabacher, Inorg. Chem. 1992, 31, liam, J. Biol. Chem. 1999, 274,28497–28504. 191–198. [47] J. M. Walker, R. Tsivkovskii, S. Lutsenko, J. Biol. Chem. [76] E. A. Ambundo,L.A.Ochrymowycz, D. B.Rorabacher, 2002, 277,27953–27959. Inorg. Chem. 2001, 40,5133–5138. [48] L. Banci, I. Bertini, S. Ciofi-Baffoni, D. L. Huffman, T. V. [77] E. A. Ambundo,Q.Y.Yu, L. A. Ochrymowycz, D. B. Ror- O Halloran, J. Biol. Chem. 2001, 276,8415–8426. ’ abacher, Inorg. Chem. 2003, 42,5267–5273. [49] L. Banci, I. Bertini, R. Del Conte,M.D’Onofrio,A. [78] J. A. Cotruvo,A.T.Aron, K. M. Ramos-Torres,C.J. Rosato, Biochemistry 2004, 43,3396–3403. Chang, Chem. Soc. Rev. 2015, 44,4400–4414. [50] T. M. DeSilva, G. Veglia, S. J. Opella, Proteins Struct. [79] A. T. Aron, K. M. Ramos-Torres,J.A.Cotruvo,C.J. Funct. Bioinf. 2005, 61,1038–1049. Chang, Acc.Chem. Res. 2015, 48,2434–2442. [51] S. Puig,J.Lee,M.Lau, D. J. Thiele, J. Biol. Chem. 2002, [80] C. J. Fahrni, Curr.Opin. Chem. Biol. 2013, 17,656–662. 277,26021–26030. [81] A. P. de Silva, H. Q. N. Gunaratne,T.Gunnlaugsson, [52] J. T. Rubino,P.Riggs-Gelasco,K.J.Franz, J. Biol. Inorg. A. J. M. Huxley,C.P.McCoy,J.T.Rademacher,T.E.Rice, Chem. 2010, 15,1033–1049. Chem. Rev. 1997, 97,1515–1566. [53] C. R. Pope,A.G.Flores,J.H.Kaplan, V. M. Unger, Curr. [82] A. P. de Silva, T. S. Moody,G.D.Wright, Analyst 2009, Top. Membr. 2012, 69,97–112. 134,2385–2393. [54] C. J. De Feo, S. G. Aller, G. S. Siluvai, N. J. Blackburn, [83] B. Valeur, I. Leray, Coord. Chem. Rev. 2000, 205,3–40. V. M. Unger, Proc. Natl. Acad. Sci. U.S.A. 2009, 106,4237– [84] E. A. Chandross,H.T.Thomas, Chem. Phys.Lett. 1971, 9, 4242. 393–396. [55] C. Rensing,B.Fan, R. Sharma, B. Mitra, B. P. Rosen, Proc. [85] L. Zeng, E. W. Miller, A. Pralle,E.Y.Isacoff,C.J.Chang, Natl. Acad. Sci. U.S.A. 2000, 97,652–656. J. Am. Chem. Soc. 2006, 128,10–11. [56] I. T. Paulsen, J. H. Park, P. S. Choi, M. H. Saier, Jr., FEMS [86] L. Yang, R. McRae,M.M.Henary,R.Patel, B. Lai, S. Microbiol. Lett. 1997, 156,1–8. Vogt, C. J. Fahrni, Proc. Natl. Acad. Sci. U.S.A. 2005, 102, [57] F. Long, C. C. Su, M. T. Zimmermann, S. E. Boyken, K. R. 11179–11184. Rajashankar, R. L. Jernigan, E. W. Yu, Nature 2010, 467, [87] K. A. Price,J.L.Hickey,Z.Xiao,A.G.Wedd, S. A. James, 484–488. J. R. Liddell, P. J. Crouch, A. R. White,P.S.Donnelly, [58] I. Bagai, W. Liu, C. Rensing,N.J.Blackburn, M. M. Chem. Sci. 2012, 3,2748–2759. McEvoy, J. Biol. Chem. 2007, 282,35695–35702. [88] C. Espirito Santo,E.W.Lam, C. G. Elowsky,D.Quaranta, [59] R. Kulathila, R. Kulathila, M. Indic, B. van den Berg, Plos D. W. Domaille,C.J.Chang,G.Grass, Appl. Environ. Mi- One 2011, 6,e15610. crobiol. 2011, 77,794–802. [60] S. Franke,G.Grass,C.Rensing, D. H. Nies, J. Bacteriol. [89] M. Bernal, D. Casero,V.Singh, G. T. Wilson, A. Grande, 2003, 185,3804–3812. H. Yang,S.C.Dodani, M. Pellegrini, P. Huijser, E. L. Con-

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 736 Review

nolly,S.S.Merchant, U. Kramer, Plant Cell 2012, 24,738– [100] M. P. Murphy,R.A.Smith, Annu. Rev.Pharmacol. Toxi- 761. col. 2007, 47,629–656. [90] J. Beaudoin, R. Ioannoni, L. López-Maury,J.Bähler, S. [101] A. F. Chaudhry,M.Verma, M. T. Morgan, M. M. Henary, Ait-Mohand, B. GuØrin, S. C. Dodani, C. J. Chang, S. N. Siegel, J. M. Hales,J.W.Perry,C.J.Fahrni, J. Am. LabbØ, J. Biol. Chem. 2011, 286,34356–34372. Chem. Soc. 2010, 132,737–747. [91] K. D. Cusick, S. C. Minkin, S. C. Dodani, C. J. Chang, S. W. [102] T. Hirayama, G. C. VandeBittner, L. W. Gray,S.Lutsen- Wilhelm, G. S. Sayler, Environ. Sci. Technol. 2012, 46, ko,C.J.Chang, Proc. Natl. Acad. Sci. U.S.A. 2012, 109, 2959–2966. 2228–2233. [92] D. Quaranta, T. Krans,C.Espirito Santo,C.G.Elowsky, [103] G. J. Brewer, Metallomics 2009, 1,199–206. D. W. Domaille,C.J.Chang,G.Grass, Appl. Environ. Mi- [104] W. Kuhn, Angew.Chem. Int. Ed. Engl. 1990, 29,1–19. crobiol. 2011, 77,416–426. [105] K. N. Raymond, V. C. Pierre, Bioconjugate Chem. 2005, 16, [93] H. Ohrvik, Y. Nose,L.K.Wood, B. E. Kim, S. C. Gleber, 3–8. M. Ralle,D.J.Thiele, Proc. Natl. Acad. Sci. U.S.A. 2013, [106] P. Caravan, J. J. Ellison, T. J. McMurry,R.B.Lauffer, 110,E4279–E4288. Chem. Rev. 1999, 99,2293–2352. [94] C. P. Huang, M. Fofana, J. Chan, C. J. Chang, S. B. Howell, [107] B. Yoo, M. D. Pagel, Front. Biosci. 2008, 13,1733–1752. Metallomics 2014, 6,654–661. [108] J. L. Major,T.J.Meade, Acc. Chem. Res. 2009, 42,893– [95] X. Ding, H. Xie,Y.J.Kang, J. Nutr.Biochem. 2011, 22, 903. 301–310. [109] E. L. Que,C.J.Chang, Chem. Soc.Rev. 2010, 39,51–60. [96] E. V. Polishchuk, M. Concilli, S. Iacobacci, G. Chesi, N. [110] R. A. Moats,S.E.Fraser,T.J.Meade, Angew.Chem. Int. Pastore,P.Piccolo,S.Paladino,D.Baldantoni, S. C. van Ij- Ed. Engl. 1997, 36,726–728. zendoorn, J. Chan, C. J. Chang, A. Amoresano,F.Pane,P. [111] A. Y. Louie,M.M.Huber, E. T. Ahrens,U.Rothbacher, Pucci, A. Tarallo,G.Parenti, N. Brunetti-Pierri, C. Settem- R. Moats,R.E.Jacobs,S.E.Fraser, T. J. Meade, Nat. Bio- bre,A.Ballabio,R.S.Polishchuk, Dev.Cell 2014,29, 686– technol. 2000, 18,321–325. 700. [112] E. L. Que,C.J.Chang, J. Am. Chem. Soc. 2006, 128, [97] A. Hong-Hermesdorf,M.Miethke,S.D.Gallaher, J. 15942–15943. Kropat, S. C. Dodani, J. Chan, D. Barupala, D. W. Do- [113] E. L. Que,E.Gianolio,S.L.Baker, A. P. Wong, S. Aime, maille,D.I.Shirasaki, J. A. Loo,P.K.Weber, J. Pett- C. J. Chang, J. Am. Chem. Soc. 2009, 131,8527–8536. Ridge,T.L.Stemmler, C. J. Chang, S. S. Merchant, Nat. [114] E. L. Que,E.Gianolio,S.L.Baker, S. Aime,C.J.Chang, Chem. Biol. 2014, 10,1034–1042. Dalton Trans. 2010, 39,469–476. [98] J. E. Whitaker,R.P.Haugland, D. Ryan, P. C. Hewitt, R. P. [115] E. L. Que,E.J.New,C.J.Chang, Chem. Sci. 2012, 3, Haugland, F. G. Prendergast, Anal. Biochem. 1992, 207, 1829–1834. 267–279. [99] S. C. Dodani, S. C. Leary,P.A.Cobine,D.R.Winge,C.J. Received:April 1, 2016 Chang, J. Am. Chem. Soc. 2011, 133,8606–8616. Published online:July 25, 2016

Isr.J.Chem. 2016,56, 724 –737  2016 Wiley-VCH Verlag GmbH &Co. KGaA, Weinheim www.ijc.wiley-vch.de 737