Skip to main content
  • Review Article
  • Published:

Ferric Uptake Regulator (FUR) protein: properties and implications in cyanobacteria

Abstract

The Ferric uptake regulator (Fur) protein is a global iron regulator found in most prokaryotes. Although the Fur protein is involved in a variety of metabolic pathways, it is specifically known for the regulation of several iron responsive genes. It binds to the highly conserved sequences located in the upstream promoter region known as iron boxes, using ferrous ion as a co-repressor. Apart from that, the Fur protein is also directly/indirectly involved in a variety of other crucial physiological pathways. Hence, understanding the mechanism of action and the mechanistic pathways of iron regulation by Fur is necessary and important. The basic understanding of the functioning and properties of Fur protein along with its role, interaction and regulation at various levels in cyanobacteria has been discussed in detail.

Introduction

Cyanobacteria are Gram-negative photosynthetic prokaryotes capable of fixing carbon dioxide with water as the reductant and oxygen as byproduct. Cyanobacteria are a highly diverse, widely distributed and ecologically important group of microorganisms (Abed and Garcia-Pichel 2001; Ferris 2005). They have been considered as major players in regulating carbon and nitrogen metabolism of soil and marine ecosystems. Cyanobacteria have a tremendous potential as nitrogenous biofertilizer and also produce amino acids, biological compounds and pharmaceutically important products. However, apart from their beneficial roles, they also have some harmful implications. Cyanobacteria form blooms in water reservoirs that are the primary source of toxins, named cyanotoxins, that have deleterious effects on human as well as animal health (de Figueiredo et al. 2004; Martin-Luna et al. 2006; Ferrao-Filho and Kozlowsky-Suzuki 2011; Deore and Bansal 2013). Iron availability represents one of the important factors controlling different metabolisms, including cyanotoxin production in cyanobacteria (Utkilen and Gjolme 1995; Martin-Luna et al. 2006). Although it is the fourth most abundant element in nature, iron is poorly soluble at physiological pH. In fact, while iron is easily available to microorganisms under anaerobic conditions, it is immediately converted to insoluble hydroxides when exposed to oxygen, which dramatically reduces its availability (Wang et al. 2009; Hayat et al. 2010).

Iron is considered to be one of the essential elements required for the growth and maintenance of cellular metabolism in a wide diversity of prokaryotes (Meyer 2000; Singh et al. 2008, 2010), the exception being members of the genus Lactobacillus (Archibald 1983). Iron availability is crucial for several important metabolic pathways such as photosynthesis, pigment synthesis, nitrogen fixation and metabolism (Straus 1994; Ferreira and Straus 1994; Kaushik et al. 2015). This is primarily because of the intricate association of iron as a cofactor of key metabolic enzymes (Rueter and Petersen 1987; Paerl et al. 2001). Accordingly, iron deficiency causes severe stress and abnormal changes in the growth and metabolism of microorganisms. However, not only the deficiency, but also the excess of iron results into deleterious effects on microbial metabolism, with the outcome being toxicity through the generation of reactive oxygen species (ROS) via the Fenton reaction (Andrews et al. 2003; Wang et al. 2011) and hydroxyl free radicals through the Haber-Weiss process (Bullen et al. 1978; Cox 1980; Cox et al. 1982). As a consequence, microorganisms have evolved complex metabolic pathways to regulate intracellular levels of iron for their survival, by tightly regulating iron uptake and storage systems.

Amongst all the molecular switches, the Fur (Ferric uptake regulator) protein is one of the most important regulators of iron homeostasis in prokaryotes. The extent of iron availability significantly influences the growth of cyanobacteria along with being an effective factor to the environment and human health, by regulating the production of cyanotoxins and carcinogens in the trophic chain (Lukac and Aegerter 1993; Boyd et al. 2000). Hence, understanding the mode of regulation and the mechanistic pathways of iron regulation by Fur in cyanobacterial system represents an important research field.

Fur protein: general properties

Ernst et al. (1978) isolated a mutant in Salmonella typhimurium for the first time while studying the mechanism of control of siderophore regulation. Hantke (1981) isolated the corresponding mutant in Escherichia coli strain K12 and named this mutant fur (ferric uptake regulator). Later, Wee et al. (1988) purified the Fur protein from E. coli. To date, more than 350 fur gene homologues have been purified and sequenced, with at least one putative fur regulatory sequence in several prokaryotes. With the increasing number of genomes that have been sequenced, it has been shown that many microorganisms have more than one putative Fur-like protein (Table 1).

Table 1 Bacterial and cyanobacterial species with more than one Fur homologue

Gram-positive and Gram-negative bacteria have been studied in different perspectives to understand iron stress and regulation (Braun and Hantke 1991; Heidrich et al. 1996; Bsat et al. 1998). All Fur proteins from different microorganisms form a family of proteins with different functions. The Fur family encompasses several subclasses, among which the Fur protein forms the major class. Apart from Fur, this family also consists of other regulators that perceive different signals to control the expression of a variety of genes (Table 2).

Table 2 Different Fur subclasses of the Fur family and their respective signals

Fur proteins are global transcriptional regulators that were initially considered to be exclusively involved in iron homeostasis in most prokaryotes. They bind to unique sequences (Fur boxes) present in the upstream promoter region of the iron responsive genes, using ferrous iron as a co-regulator (Fillat 2014). However, it has now been well established that the mechanism of regulation by Fur involves a much higher level of complexity, as Fur controls several processes, such as nitrogen metabolism, photosynthesis and respiration, intermediary metabolism (Gonzalez et al. 2014), virulence factor production (Delany et al. 2004; Zhou et al. 2006; Gao et al. 2008), and is involved in the defense against different type of stresses (Touati 2000; Cornelis et al. 2011) (Fig. 1). Fur is also associated with the regulation of toxin-producing genes in prokaryotes ( Martin-Luna et al. 2005, 2006). Fur mechanism of action and Fur-mediated regulation appear to be different, depending on the microorganism. Fur is capable of both positive as well as negative regulation of a variety of genes (Bagg and Neilands 1987; Pressler et al. 1988; Salinas et al. 1989; Litwin et al. 1992; Barton et al. 1996; Hernandez et al. 2004a, 2006b) (Table 3).

Fig. 1
figure 1

Cellular processes and related target genes under Fur Regulation

Table 3 Mode of regulation of different Fur protein along with their targets

The Fur protein is in general a 17–21 kDa polypeptide (Bagg and Neilands 1987; Saito et al. 1991; Martin-Luna et al. 2005) that acts as an iron-dependent transcriptional regulator that regulates the expression of several iron responsive genes (Bagg and Neilands 1987; de Lorenzo et al. 1987; Escolar et al. 1997, 1998). When plenty of iron is available to microorganisms, Fur binds the Fe2+ ion and undergoes conformational changes. Fe-bound Fur dimerises and then binds to the iron boxes, inhibiting the transcription of all iron responsive genes and operons (Fig. 2). However, when iron availability is scarce, Fe2+ ion is released from Fur, and the complex dissociates from target promoters, allowing the RNA polymerase to bind and initiate transcription (Klebba et al. 1982; Griggs et al. 1987; Griggs and Konisky 1989). Moreover, Fur is also known to autoregulate itself in the presence and absence of iron (Delany et al. 2001, 2002). Apart from its repressive effect, Fur also activates gene transcription by directly binding to promoter regions, although the actual mechanism for this Fur-mediated direct activation is not well understood. Several mechanisms were proposed to explain the above-mentioned process. Firstly, an activator may remove the Fur repressor from the binding sites and initiate the transcription of target genes (Browning and Busby 2004; Isabella et al. 2008; Nandal et al. 2010; Yu and Genco 2012). Secondly, Fur may recruit the RNA polymerase at the promoter of target genes, thereby enhancing transcription (Browning and Busby 2004), and thirdly, it was proposed that the binding of an activator may alter DNA morphology, making the promoter sequence free for the binding of RNA polymerase (Browning and Busby 2004; Teixido et al. 2011). In Anabaena sp. PCC 7120, FurA differentially activates the transcription of psaK, amt4, hetC, alr1728, patA and asr1734, depending on the presence of the metal co-regulator and a reducing environment (Gonzalez et al. 2014). Gonzalez and coworkers also demonstrated the dual role of Fur protein, i.e., as both activator and repressor in the same metabolism, as observed in the regulation of tetrapyrrole biosynthesis (Gonzalez et al. 2012).

Fig. 2
figure 2

Schematic representation of mechanism of action of Fur protein in cyanobacterium Anabaena sp. PCC 7120 (adopted from old www.bifi.es/research/protein_dna_inter/protein_dna_inter.php)

In contrast to its iron-bound repression in several heterotrophic bacteria, Fur was also shown to repress the expression of iron responsive genes, even under iron deficient conditions (apo-regulation) (Bereswill et al. 2000; Ernst et al. 2005). Miles et al. (2010), through their plasmid complementation study, demonstrated an iron-bound and apo-Fur regulation in Helicobacter pylori. Fur also activates the fur gene itself (autoregulation) in apo form in Vibrio vulnificus, apart from its repressing activity (Lee et al. 2007). Iron may not have any role in either repression or activation by apo-Fur, but other metals are possibly involved in apo-Fur functions (Mills and Marletta 2005; Ducey et al. 2005; Sheikh and Taylor 2009). apo-Fur regulation in iron deficient conditions has not been detected in any of the cyanobacteria, to date.

Although apo-Fur regulation has not been identified in other species except H. pylori, certain genes in E. coli, Pseudomonas aeruginosa and Vibrio cholerae are known to be indirectly repressed (Litwin and Calderwood 1994; Wilderman et al. 2004; Masse et al. 2007). In these organisms, this regulation is mediated by Fur-regulated small RNAs (sRNA) (Masse and Gottesman 2002). Small RNAs generally pair with the ribosome binding site of the target mRNA at a specific 8–9 base pair sequence and decrease the stability of mRNA, which ultimately leads to a reduced translation. RyhB regulates the expression of sodB, sdhCDAB, acnA, fumA and ferritin gene in E. coli (Dubrac and Touati 2000; Masse and Gottesman 2002; Masse et al. 2003, 2005, 2007), just like apo-Fur regulation of sodB and pfr in the case of H. pylori (Ernst et al. 2005 ). Like RyhB in E. coli, several other sRNAs were also reported to be repressed by Fur, such as PrrF1 and PrrF2 in P. aeruginosa (Wilderman et al. 2004), RyhB in V. cholerae (Mey et al. 2005; Davis et al. 2005), FsrA in Bacillus subtilis (Gaballa et al. 2008) and NrrF in Neisseria meningitidis (Mellin et al. 2007, 2010; Yu and Genco 2012). Non-coding (antisense) RNA-based regulation of the fur gene has also been demonstrated in several cyanobacteria (Hernandez et al. 2006a; Martin-Luna et al. 2011). The mechanism of this regulation is described in detail in this review.

The Fur protein acts as a dimer (Coy and Neilands 1991; Neilands and Nakamura 1991; Michaud-Soret et al. 1997), and each monomer consists of two different domains (Coy and Neilands 1991; Stojiljkovic and Hantke 1995; Hernandez et al. 2002). The C terminus is involved in protein dimerization, while the N-terminus is responsible for the DNA binding ability of the Fur protein (Holm et al. 1994; Stojiljkovic and Hantke 1995; Hernandez et al. 2005). The Fur protein folds by two state mechanisms and 40 % of its structure is composed of α-helix (Hernandez et al. 2005). In contrast to the other Fur homologues, no zinc ion or zinc binding pockets have been experimentally detected in Anabaena sp. (Pohl et al. 2003). Fur binds to DNA on opposite faces of the helix as an overlapping dimer (“Overlapping dimer binding” model) (Lavrrar et al. 2003), as in the case of DtxR–DNA interaction in Corynebacterium diptheriae and other Gram-positive bacteria (White et al. 1998; Pohl et al. 1999). Isothermal titration calorimetry (ITC) experiments have also shown the presence of two metal binding sites in the FurA protein from Anabaena sp. PCC 7120, one for co-repressor binding and the other site having a structural role (Jacquamet et al. 2009; Althaus et al. 1999; Hernandez et al. 2002, 2004a).

Structure and properties of Fur binding sites (Fur Boxes)

Fur proteins were previously thought to recognize and bind to a highly conserved consensus of 19 bp inverted repeats (5΄- GATAATGATAATCATTATC-3΄) found in the upstream promoter region of iron responsive genes in prokaryotes (Calderwood and Mekalanos 1988; Stojiljkovic et al. 1994) (Fig. 3). According to the recent hexamer model, this conserved consensus contains tandem repeats of three forward-reverse hexamers (5΄-GATAAT-3΄) with a Fur recognition unit (5΄- NAT(A/T)AT-3΄) (Escolar et al. 1998) (Fig. 3). Later, Baichoo and Helmann (2002) demonstrated that the Fur binding site is actually a 15 bp core region formed by a 7-1-7 heptamer motif. In the Anabaena sp. PCC 7120, Fur binding was observed at consensus sequences containing 7-1-7 inverted repeats (Napolitano et al. 2012). Fur boxes are generally characterized by the presence of an AT rich region. Several numbers of AT rich Fur boxes were also identified in different bacteria sharing 50–80 % of sequence similarity with the canonical consensus identified in E. coli (Baichoo et al. 2002; Sebastian et al. 2002; Thompson et al. 2002; Fillat 2014). The variations among Fur boxes were due to addition of sequences in the basal recognition sequence (GATTAT), which ultimately alters the affinity and polymerization of Fur homologues to the Fur boxes (Fillat 2014).

Fig. 3
figure 3

19 bp consensus sequence containing palindromic sequence with two 9 bp inverted repeats as Fur binding site (FBS) (a), Fur recognition unit according to recent hexamer model (b) (Escolar et al. 1998)

FurA boxes were also located within bidirectional promoters of genes involved in several metabolisms, for example all0396-schT, all2624-alr2625, all2586-alr2587, alr2594-alr2595 involved in iron metabolism, all0949-coxB, ccmK-ndhF involved in photosynthesis and respiration, and znuA-oprB, hupL3-xisC, and nifD3-xisA involved in other important metabolisms in Anabaena sp. PCC 7120 (Gonzalez et al. 2014). These binding sites play a role in the simultaneous regulation of genes with overlapping promoters (Hunt et al. 1994; Christoffersen et al. 2001; Lavrrar et al. 2003). In Anabaena sp. PCC 7120, the all2641–all2649 gene cluster regulating siderophore production and iron-siderophore transport systems (Jeanjean et al. 2008), contains multiple Fur binding sites (with different Fur binding affinities) located in various intergenic regions (one upstream and the other three downstream of the promoter region, respectively). These sequences sequentially modulate gene expression, depending on the iron status of the cell (Gonzalez et al. 2012). In silico analyses have also demonstrated the presence of FurA-binding sites in the promoter regions of aphC (encoding a putative photoreceptor of a two-component system), cyaC (encoding adenylate cyclase carrying two component sensorand regulatory domain) (Okamoto et al. 2004), asr (encoding bacteriorhodopsin) (Irieda et al. 2012) and cyaD (encoding adenylate cyclase) (Katayama and Ohmori 1997) in Anabaena sp. PCC 7120. These data suggested that Fur controls the expression of genes involved in the signal transduction cascade (Gonzalez et al. 2014). FurA binding site was also predicted in the upstream region of the promoter of the transposase-encoding gene all4465 in Anabaena sp. PCC 7120 and FurA also regulates the transcription of all4465 in metal and reducing condition dependent manners (Gonzalez et al. 2014). The transcription of znuA, cyaD, aphC, alr0240 and pbpH is co-modulated by other transcriptional regulators along with FurA under iron limitation in Anabaena sp. PCC 7120 (Gonzalez et al. 2014). The znuAB operon, which encodes for the components of a high affinity zinc uptake system, is co-regulated by FurB/Zur in Anabaena sp. PCC 7120 (Napolitano et al. 2012). A Fur/YbtA regulatory network was also postulated in Yersinia pestis, where Fur and YbtA coregulate the functioning of the ybtA locus encoding a virulence dependent iron uptake system (Gao et al. 2008). In Microcystis aeruginosa PCC 7806, two putative Fur boxes were identified in the mcyDA bidirectional promoter (P mcyDA ), a 732 bp region between the mcyA and mcyD genes ( Martin-Luna et al. 2006). mcyE, mcyH, mcyG and mcyJ genes also showed the presence of AT-rich DNA stretches in their promoter regions ( Martin-Luna et al. 2006) having varied matching scores with the previously determined Fur consensus of other cyanobacteria (Hernandez et al. 2006b).

Fur homologues in cyanobacteria

Until now, Fur homologues in cyanobacteria have mostly been studied in Synechococcus, Synechocystis, Microcystis and Anabaena (Ghassemian and Straus 1996; Kaneko et al. 1996; Bes et al. 2001; Martin-Luna et al. 2005). Ghassemian and Straus (1996) isolated a Fur homologue from the cyanobacterium Synechococcus sp. PCC 7942 using an E. coli-based in vivo repression assay. DNA sequence analysis of the clones depicted the presence of an open reading frame (ORF) coding for a Fur protein that showed 36 % homology to E. coli Fur (Ghassemian and Straus 1996). The amino acid sequence of the Fur homologue from Synechococcus sp. PCC 7942 showed no convincing homology to the DtxR homologue, but showed significant similarity to all known Fur sequences (Ghassemian and Straus 1996). This Fur protein contained (147 amino acids) a conserved and a putative iron-binding domain of Fur repressors (HHXHXXCXXC). Inactivation of the Fur homologues in Synechococcus sp. PCC 7942 resulted in a disturbed iron homeostasis with severe iron stress symptoms, i.e., continuous flavodoxin expression and siderophore production, which suggested that the inactivation of the fur gene had lethal implications.

The whole genome sequencing of the nitrogen fixing filamentous cyanobacterium Anabaena sp. PCC 7120, allowed the identification of three open reading frames (all1691, all2473 and alr0957) encoding proteins with histidine rich regions sharing properties with the Fur protein family from other prokaryotes (Gonzalez et al. 2014; Hernandez et al. 2004a). Amongst these three ORFs, all1691, located between the sigC (group 2 sigma factor that responds to carbon: nitrogen imbalance) (Brahamsha and Haselkorn 1992) and alr1690 gene (encoding cell wall binding protein with peptidoglycan-binding domain), encodes a putative FurA protein (17 kDa) (Fig. 4). The all2473 and alr0957 genes code for FurB (15.1 kDa) and FurC (17.3 kDa) paralogues, respectively ( Lopez-Gomollon et al. 2009). The FurB protein has 132 amino acids, five cysteine residues and seven histidine moieties while the FurC protein has 149 amino acids, three cysteine residues and six histidine moieties (Hernandez et al. 2004a). The FurA and FurB proteins bind to the promoter region and regulate the expression of the three fur-homologue genes, whereas FurC regulates the DNA binding ability of FurA and FurB (Hernandez et al. 2004a) (Fig. 4). Apart from regulating the expression of the FurA protein at a transcriptional level, FurB also prevents DNA damage against reactive oxygen species (ROS) and increases the survival rate of the cyanobacterium Anabaena sp. PCC 7120 ( Lopez-Gomollon et al. 2009). FurB expression increases drastically under the influence of ROS and protects DNA against the damage caused by ROS, by binding to unspecific sequences. As observed in several other cyanobacteria (Barnett et al. 2012) and heterotrophic bacteria (Patzer 2000; Lindsay and Foster 2001; Fuangthong and Helmann 2003; Maciag et al. 2007), FurB (or Zur) controls zinc homeostasis by binding to upstream promoter regions of target genes, such as those encoding putative metallochaperones (All4722, All1751), zinc metalloproteins (All4725/HemE, All4723/ThrS), components of plasma membrane ABC transport systems (ZnuABC) and several outer membrane proteins (Alr3242, Alr4028) in a zinc-dependent manner in the cyanobacterium Anabaena sp. PCC 7120 (Napolitano et al. 2012). Therefore, FurB has a dual role in Anabaena sp. PCC 7120, which depends on its concentration in the cell. When FurB concentration is low, it acts as a transcriptional regulator, while it provides protection against oxidative stress by binding unspecifically to the DNA when its concentration is high ( Lopez-Gomollon et al. 2009; Sein-Echaluce et al. 2014). FurC has not been reported to regulate FurA paralogue directly, but it is known to affect the FurB binding to the upstream sequence of P fur (Fur box) (Hernandez et al. 2004a). Unlike FurB, FurC is not involved in DNA protection against ROS, but it is thought to be involved in the regulation of photosynthetic proteins that act against oxidative stress ( Lopez-Gomollon et al. 2009). Recently, microarray and qRT-PCR-based analyses depicted the increase in transcription of the alr0957 (furC) gene in the presence of H2O2, and suggested that FurC is a PerR-like protein involved in regulating peroxide stress response, by sensing peroxide by metal catalyzed oxidation in cyanobacterium Anabaena sp. PCC 7120 (Yingping et al. 2014).

Fig. 4
figure 4

Schematic representation of binding of three Fur homologues at Fur binding site (FBS) in Anabaena sp. PCC 7120 (adopted from old www.bifi.es/research/protein_dna_inter/protein_dna_inter.php)

The complete sequence of the fur gene homologue from M. aeruginosa PCC 7806 has been identified using inverse PCR ( Martin-Luna et al. 2005). The 296 bp sequence obtained showed a high level of identity to the fur gene homologues from other cyanobacteria. Analysis of the upstream sequence of the Fur homologue showed it to have iron boxes sharing 40–50 % similarity to those determined in the other cyanobacteria (Hernandez et al. 2005). The mcy operon contains several iron boxes in the promoter regions of various mcy genes that allow for control at different levels. In M. aeruginosa PCC 7806, Fur proteins are involved in the regulation of the expression of the microcystin gene cluster, which is ultimately influenced by iron availability ( Martin-Luna et al. 2006). The Fur homologue from M. aeruginosa PCC 7806 is considered to be the sensor of iron availability and oxidative stress (Thompson et al. 2002; Martin-Luna et al. 2006). M. aeruginosa PCC 7806 Fur contains183 amino acids (21 kDa) and shares 81 % identity with Fur from Synechocystis PCC 6803 (sll0567) and 70 % with FurA from Anabaena sp. PCC 7120 (all1691) ( Martin-Luna et al. 2005). The only difference lies in the C-terminal domain, which is larger when compared to other Fur homologues; however, the DNA and metal binding domains are highly conserved ( Martin-Luna et al. 2005).

In the Synechocystis PCC 6803 genome, the sll0567 gene (Kaneko et al. 1996) codes for a putative FurA protein, which revealed the highest degree of similarity to a Synechococcus sp. PCC 7942 Fur, and was involved in the repression of the isiAB operon under iron-repleted conditions (Ghassemian and Straus 1996). Mutations in sll0567 resulted in iron-stress-like symptoms, and hence is considered essential for cellular function (Kunert et al. 2003). The protein encoded by the sll0567 gene also possesses a specific motif (HHXHXXCXXC) involved in metal binding (Hennecke 1990), and is similar to the ones found in other heterotrophic bacteria and cyanobacteria (Ghassemian and Straus 1996; Bes et al. 2001). Kunert et al. (2003) identified a consensus Fur box in the promoter region of the isiAB operon of Synechocystis sp. PCC 6803, by using the on-line MEME motif discovery tool (Bailey and Elkan 1994). The proposed location of the Fur box in Synechocystis sp. PCC 6803 was found in the 3'-untranslated region of the isiAB promoter fragment fused to gfp in the reporter strain. Although Fur represses isiAB transcription under iron depleted conditions, its effect was only partial. A binding site for an unidentified activator was found to be located in the 5'-untranslated region of the isiAB operon, suggestive of a dual regulation of the isiAB operon in Synechocystis sp. PCC 6803. Similar regulation was also found in other organisms (Kammler et al. 1993; Michel et al. 1999).

Fur–DNA interaction

The affinity, stability and quality of Fur–DNA interaction in cyanobacteria can be influenced by several factors, such as the architecture of Fur boxes, the presence of divalent metal ions, the redox status of cysteine and histidine residues within the Fur protein, ionic interactions (Hernandez et al. 2002, 2006b) and the involvement of effectors like heme (Hernandez et al. 2004b; Pellicer et al. 2012). It was previously considered that the A and T bases at position 5 and 6 of the consensus hexamer played a major role in the Fur–DNA interaction (Escolar et al. 1998). Later, it was confirmed that it is in fact the architecture of the Fur binding site, which is essential for Fur binding (Lavrrar et al. 2003; Gonzalez et al. 2011). Moreover, it has been proposed that Fur can bind DNA only in the presence of Zn2+ ions (Bagg and Neilands 1987; Althaus et al. 1999; Bsat and Helmann 1999; Xiong et al. 2000; Zheleznova et al. 2000), but this has been further contradicted (Saito and Williams 1991; Bsat and Helmann 1999) since the presence of Zn2+ ions were not detected in an active recombinant FurA protein overexpressed in Anabaena sp. PCC 7119 (Hernandez et al. 2002) and in the Fur protein from P. aeruginosa (Lewin et al. 2002). Fur–DNA interaction in the Anabaena Fur is also influenced by the presence of Mn2+ ion and the redox status of the Fur protein (Hernandez et al. 2006b). Fur–DNA interaction attained maximum stability in the presence of Mn2+ ion. The presence of Mn2+ also influences the oligomerization of Fur monomers, suggesting the involvement of hydrophobic interactions in this process (Hernandez et al. 2002). Similarly, the redox status of cysteine residues present in the Fur protein also influences the Fur–DNA interaction and the oligomerization of Fur monomers (Ortiz de Orue Lucana and Schrempf 2000; Hernandez et al. 2002). In several prokaryotes, cysteine residues present in Fur proteins are known to be involved in metal binding and DNA recognition activities (Wee et al. 1988; Coy 1995; Althaus et al. 1999; Zou et al. 1999; Zheleznova et al. 2000). Reduced cysteine residues allow Fur monomers (inert) to reorganize into dimers or oligomers, which are the active forms of the Fur protein and interact with DNA sequences to regulate different functions. Similar effects of Mn2+ (metalloregulator) and redox status of cysteines on Fur–DNA interaction were also observed in the cyanobacterium M. aeruginosa PCC 7806 ( Martin-Luna et al. 2006). In M. aeruginosa PCC 7806, Fur proteins differ at their C-terminal domains when compared to other Fur homologues. Seven redox active cysteine residues were detected in M. aeruginosa PCC 7806. Although the arrangement of the cysteine residues was the same as for the one identified in other cyanobacteria, only five of them were highly conserved ( Martin-Luna et al. 2006). Cysteine residues identified in M. aeruginosa PCC 7806 are considered as a redox sensor and are also involved in metal binding and protein oligomerization (Ortiz de Orue Lucana and Schrempf 2000).

Fur from Anabaena has five cysteine residues, and three of them are located at the C-terminus domain. However, four of the five cysteine residues are arranged in two CXXC motifs, but only one cysteine is involved in DNA recognition and metal binding activities. FurA is the master regulator of iron homeostasis, and for its optimal binding to target DNA in vitro, it requires a reducing environment. Recently, a CXXC motif (C101XXC104) with a novel disulfide reductase activity was identified in the cyanobacterium Anabaena sp. PCC 7120 ( Botello-Morte et al. 2014). Although FurA lacks Zn2+ ions responsible for the maintenance of the redox status of cell (Hernandez et al. 2002), the CXXC motif in Anabaena sp. PCC 7120 behaves as a redox rheostat ( Botello-Morte et al. 2014). FurA senses the redox status of the cell by forming an intermolecular disulfide linkage when it is partially oxidized. The CXXC motif (C101XXC104) resembles sequences previously reported in the active sites of thioredoxins and thioredoxins-like proteins, which act as reducing agents, thereby reducing structurally important disulfide bonds in target proteins and maintaining redox homeostasis (Quan et al. 2007). This canonical sequence motif acting as a redox rheostat has also been reported in other proteins such as glutaredoxins (Florencio et al. 2006), peroxiredoxins (Latifi et al. 2009), Dsb proteins (Heras et al. 2007) and in the eukaryotic protein-disulfide isomerases (Sevier and Kaiser 2002). Cross-linking studies on FurA in Anabaena spp. suggest a role of ionic interactions in protein–protein interactions and protein oligomerization (Hernandez et al. 2006b), in line with the study of Pohl et al. (2003). The Fur dimer from P. aeruginosa consists of a large dimerization interphase state and salt bridges between arginine and aspartic acid residues (Pohl et al. 2003). This possibly indicates that, apart from the redox status, ionic strength also influences the affinity of Fur for its target DNA.

Fur–DNA interactions have also been influenced by the presence of heme (Hernandez et al. 2004b). The involvement of heme in regulating important bacterial metabolic pathways such as respiration and energy transfer is already well documented (Smith et al. 1996). In addition to its function as a gas-sensing compound (i.e., O2, NO or CO), through its interaction with different heme-sensing proteins (Igarashi et al. 2011), heme also acts as a redox cofactor. Differential spectroscopy-based analyses have identified the presence of a Fur-heme complex in the cyanobacterium Anabaena sp. PCC 7120 (Hernandez et al. 2004b). Heme binds and inhibits the DNA binding ability of the Fur protein in vitro in a concentration dependent manner, which represses the expression of Fur-regulated genes (Hernandez et al. 2004b). Histidine residues present in Fur binding sites are responsible for the formation of Fur-heme complexes in vitro (Smith et al. 1996; Paoli et al. 2002), and this binding is critically affected by pH (Saito et al. 1991). Cysteine residues are also considered to be involved in Fur-heme complex formation. Based on site-directed mutagenesis and different spectral studies, it was observed that Cys141 located in the Cys-Pro (CP) motif (dipeptide Cys141-Pro142) at the C-terminal domain is an axial ligand of the Fe (III) heme (Pellicer et al. 2012). Except E. coli, which does not have any CP motif, the CP motif is also known for its heme sensing ability in other prokaryotes (Zhang and Guarente 1995; Ogawa et al. 2001). In Anabaena sp. PCC 7120, heme binds the Fur protein in an intricate, concentration-dependent and saturable manner at pH 8. In the case of the ferrous heme-FurA binding, Cys141 was not the ligand for heme binding, indicating that a redox-dependent ligand switch likely occurs in which the His residue becomes the ligand upon reduction of Cys141, by removal of thiol from heme iron (Pellicer et al. 2012). The binding of heme to FurA specifically affects the affinity of Fur protein (repressor) binding to the target sequence in vitro, even in the presence of Mn2+ ions (effector increases the affinity of Fur–DNA interaction) (Hernandez et al. 2004b). In E. coli, it was considered that the heme and Mn2+ moieties have similar or closely placed binding sites; hence, heme binding to the Fur protein blocks Mn2+ binding in vitro (Smith et al. 1996). Similar results were also observed in Anabaena sp. PCC 7120, where heme binding causes a conformational change in the metal binding site, which inhibits the affinity of Mn2+ to Fur protein in vitro (Hernandez et al. 2004b). The ability of the heme co-factor to impair the binding ability of other Fur homologues (FurB and FurC) was also studied in Anabaena sp. PCC 7120 ( Lopez-Gomollon et al. 2009). Heme binding severely affects the Fur–DNA interaction in vitro, while a heme-protein complex was not detected in the case of the FurC homologue.

Fur and oxidative stress

The association of iron metabolism and oxidative stress is a well-established fact in prokaryotes (Andrews et al. 2003; Latifi et al. 2009; Wang et al. 2011). Fur is mainly associated with regulation of iron metabolism, but in several studies, it has been observed that the expression of the fur gene was drastically upregulated under oxidative stress environmental conditions ( Lopez-Gomollon et al. 2009). Hence, an essential role of Fur in the defense against oxidative stress has also been hypothesized (Zheng et al. 1999; Thompson et al. 2002; Lopez-Gomollon et al. 2009). In Anabaena sp. PCC 7120, the redox status of the Fur cysteine residues seem to play a major role in combating oxidative stress (Gonzalez et al. 2011; Botello-Morte et al. 2014). In oxidative stress condition, FurA and FurB regulate the transcription at P furA and P furB in Anabaena sp. PCC 7120 (Hernandez et al. 2004a; Sein-Echaluce et al. 2014), suggesting a potential overlapping of FurA and FurB (Zur) promoters. In vivo and in vitro assays have demonstrated prxA and dpsA genes as the direct targets of FurB (Zur), and hence suggest that FurB plays an important role in connecting zinc homeostasis with oxidative stress protection in Anabaena sp. PCC 7120 ( Sein-Echaluce et al. 2014). Semi-quantitative RT-PCR and EMSA analyses identified genes involved in oxidative stress response, such as sodA (all0070), prxA (alr4641), gct1 (alr3183), gct3 (all2375), and dpsA (alr3808), as putative targets for FurB (Zur) ( Sein-Echaluce et al. 2014). In Anabaena sp. PCC 7120, prxA and dpsA contain multiple AT-rich regions that partially match with the FurB (Zur) consensus sequence described previously (Napolitano et al. 2012), and are modulated by additional regulatory factors (Hernandez et al. 2007; Yingping et al. 2014), as also reported in the case of heterotrophic bacteria (Kallifidas et al. 2009).

In M. aeruginosa PCC 7806, Fur could confer protection against oxidative stress by regulating the expression of the mcy genes, by binding to their bidirectional promoter (P mcyDA) and further by modulating the production of microcystin ( Martin-Luna et al. 2006). In iron starved conditions, Fur detaches from the bidirectional promoter of the mcyDA genes, which causes the production of microcystin. Microcystin acts as an intracellular iron chelator and increases the rate of iron uptake (Utkilen and Gjolme 1995) in the toxin-producing cyanobacterium M. aeruginosa PCC 7806, and thus it may manage survival in the oxidative stress ( Martin-Luna et al. 2006).

Generally, the PerR protein is known for its action in combating oxidative stress conditions in prokaryotes; however, no PerR orthologue was found in Anabaena sp. PCC 7120. In this species, FurA plays a dual role by regulating the transcription of PerR-regulated genes as well as the one involved in iron homeostasis (Hernandez et al. 2004a, 2006a). The correlation between Fur and PerR regulators has already been established (Hahn et al. 2000; Horsburgh et al. 2001; Singh et al. 2003; Li et al. 2004). Recently, elevated transcription of the alr0957 (furC) gene in the presence of H2O2 suggested that FurC is a PerR-like protein involved in regulating the peroxide stress response by sensing peroxide by metal catalyzed oxidation in Anabaena sp. PCC 7120 (Yingping et al. 2014).

Regulation of Fur in cyanobacteria

The complex regulation of fur has been extensively studied in several microorganisms. In cyanobacteria, the regulation of Fur occurs at three different levels i.e., transcriptional, post-transcriptional and post-translational ( Botello-Morte et al. 2013). In Anabaena sp. PCC 7120, the furA gene autoregulates its expression and has several putative Fur binding sites in its promoter region (Hernandez et al. 2004a). Under varying iron status, expression of furA is also influenced by other Fur paralogues, i.e., FurB and FurC (Hernandez et al. 2004a). Binding assays demonstrated the presence of binding sites for the FurB/Zur paralogue in P furA , and hence suggested the direct regulation of the expression of furA at the transcriptional level (Hernandez et al. 2004a; Sein-Echaluce et al. 2014). The promoter region of furA does not contain binding sites for the FurC paralogue, but FurC has been shown to influence the binding affinity of both FurA and FurB/Zur paralogues on P furA , by enhancing and inhibiting the FurA and FurB binding, respectively (Hernandez et al. 2004a). In the Anabaena sp. PCC 7120, northern blot and western blot analyses showed an increase in furA expression and a corresponding decrease in alr1690-α-furA expression in nitrogen fixing conditions. Nitrogen deprivation upregulates the level of furA mRNA and the corresponding proteins, specifically in pro-heterocysts and mature heterocysts ( Lopez-Gomollon et al. 2006). Nitrogen status has no effect on the transcription level or on the expression of FurB and FurC paralogues. Lopez-Gomollón et al. (2007) also demonstrated an NtcA-based regulation of furA in heterocysts in the Anabaena sp. PCC 7120. Footprinting and EMSA assays depicted the presence of several putative NtcA binding sites on the upstream region of furA and on the dicistron alr1690-α-furA promoters. NtcA can either act as an activator or as a repressor, depending on the metabolic status of cells (Kolb et al. 1993; Herrero et al. 2001). Under nitrogen fixing conditions (Nitrogen starvation), NtcA binds to the upstream sequence of the furA promoter and enhances its expression in heterocysts. In contrast to in nitrogen limiting conditions, NtcA acts as a repressor and causes the down regulation of furA in vegetative cells in the presence of combined nitrogen. The presence of several NtcA binding sites in the upstream sequence of furA and alr1690-α-furA is decisive for this differential regulation depending upon the nitrogen status of cells. The identification of common elements overlapping the ntcA and furA regulon unravelled the presence of a possible transcriptional regulatory network required for regulating iron homeostasis, redox control and nitrogen metabolism ( Lopez-Gomollón et al. 2007).

The non-coding (antisense) RNA-based regulation of Fur was studied and is considered as an important regulatory system in several bacteria (Chen and Crosa 1996; Gottesman 2002; Wilderman et al. 2004; Davis et al. 2005; Masse et al. 2005; Duhring et al. 2006; Hernandez et al. 2006a; Jacques et al. 2006) (Table 4). Regulation of fur expression by α-fur antisense mRNA has also been observed in Anabaena sp. PCC 7120 and M. aeruginosa PCC 7806 (Hernandez et al. 2006a; Martin-Luna et al. 2011). Generally, these antisense RNAs are trans-encoded on the accessory elements such as plasmids or transposable elements and regulate the expression of several regulatory genes by forming a partial and imperfect RNA-RNA duplex (Hernandez et al. 2006a). The filamentous cyanobacterium Anabaena sp. PCC 7120 has a large dicistronic transcript containing the alr1690 gene that possibly encodes a putative membrane protein Alr1690, and an antisense α-furA RNA, which is involved in the regulation of furA expression. An α-furA RNA covers the complete coding sequence of furA (Hernandez et al. 2006a) (Fig. 5). It has been hypothesized that α-furA RNA masks the ribosome binding site in the furA mRNA, and thus interferes with furA expression, which in turn alters the level of proteins belonging to the furA regulon. Mutants impaired in α-furA-alr1690 dicistron resulted in an increased expression of furA and exhibited an iron-deficient phenotype (Hernandez et al. 2010). In M. aeruginosa PCC 7806, implication of α-fur RNA has been mainly observed as a consequence of oxidative stress and change in light conditions ( Martin-Luna et al. 2011). The requirement of light and reduced environment has been essential for the expression of the Fur protein in the cyanobacterium M. aeruginosa PCC 7806. In the absence of either light or reduced environment, expression of the fur gene is inhibited due to expression of antisense α-fur RNA which binds to fur mRNA.

Table 4 Regulatory RNAs from different bacterial and cyanobacterial species
Fig. 5
figure 5

Schematic representation of post-transcriptional regulation of furA by α-furA antisense mRNA in cyanobacterium Anabaena sp. PCC 7120 (adopted from old www.bifi.es/research/protein_dna_inter/protein_dna_inter.php)

Regulation of the fur regulon has also been studied extensively at the post-translational level in prokaryotes. Binding of the heme complex to Fur proteins influences the affinity of Fur (repressor) for the Fur box in vitro (Hernandez et al. 2004b). In the Anabaena sp. PCC 7120, heme binding causes the conformational change in the ligand binding site, which inhibits the binding of Fur to the target sequences in vitro (Hernandez et al. 2004b) (Fig. 6). Krynicka and coworkers demonstrated the proteolysis-dependent regulation of abundance of Fur protein in Synechocystis sp. PCC 6803 (Krynicka et al. 2014). Two FtsH protease homologues, FtsH1 (encoded by slr1390 gene) and FtsH3 (encoded by slr1604 gene), together form a FtsH1/FtsH3 heterocomplex that is involved in the acclimation of cells to the iron deficiency (Krynicka et al. 2014). In Synechocystis sp. PCC 6803, it has recently been observed that the transcription of isiA/isiB operons and several other genes involved in iron homeostasis was also dependent on the level of the FtsH1/FtsH3 heterocomplex (Boehm et al. 2012). Under iron deficiency, the FtsH1/FtsH3 protease disturbed the equilibrium between DNA-bound and free Fur, by proteolytic degradation of detached Fur, and this would further induce the detachment of Fur from DNA (Krynicka et al. 2014).

Fig. 6
figure 6

Schematic representation of post-translational modifications of FurA in Anabaena sp. PCC 7120 (adopted from old www.bifi.es/research/protein_dna_inter/protein_dna_inter.php)

Conclusion

In contrast to previous findings, the Fur protein in cyanobacteria controls a plethora of genes regulating different metabolisms other than iron homeostasis. To date, a large number of Fur paralogues have been identified and characterized in a wide range of prokaryotes. However, concerning cyanobacteria, many of the present studies associated with Fur protein were mainly focused on Synechococcus PCC 7942, Synechocystis sp. PCC 6803, Anabaena sp. PCC 7120 and M. aeruginosa PCC 7806. The Fur protein acts by binding to the fur boxes located upstream of promoter sequences using ferrous ion as a co-regulator. Fur–DNA interaction is influenced by several factors, such as the architecture of Fur boxes, the presence of divalent metal ions, the redox status of cysteine and histidine residues, ionic interactions and the involvement of effectors such as heme. In contrast to other prokaryotes, zinc showed no effect on Fur–DNA interaction in Anabaena sp. PCC 7120. The Fur protein acts as a redox sensor and protects cyanobacteria against oxidative stress, using the reductive property of its cysteine residues. Fur has also been demonstrated to be actively involved in the regulation of nitrogen fixation and cellular metabolism, by controlling the acquisition of iron. Regulation of the fur gene expression has been studied at different levels, i.e., transcriptional, posttranscriptional and post-translational levels, in cyanobacteria. In the Anabaena sp. PCC 7120, the increase in FurA expression and the corresponding decrease in alr1690-α-furA expression, specifically in heterocysts in nitrogen fixing conditions were related to the NtcA regulator. NtcA can either act as an activator or as a repressor, depending on the metabolic status of cells. Regulation of fur expression by α-fur antisense mRNA has also been observed in the Anabaena sp. PCC 7120 and M. aeruginosa PCC 7806. Thus, considering the importance of cyanobacteria as biofertilizers in the aquatic environment, deciphering the role and mechanism of action of the Fur protein in a wide variety of cyanobacteria is necessary.

References

  • Abed RMM, Garcia-Pichel F (2001) Long-term compositional changes after transplant in a microbial mat cyanobacterial community revealed using a polyphasic approach. Environ Microbiol 3:53–62

    Article  CAS  PubMed  Google Scholar 

  • Ahn BE, Cha J, Lee EJ, Han AR, Thompson CJ, Roe JH (2006) Nur, a nickel-responsive regulator of the Fur family, regulates superoxide dismutases and nickel transport in Streptomyces coelicolor. Mol Microbiol 59:1848–1858

    Article  CAS  PubMed  Google Scholar 

  • Althaus EW, Outten CE, Olson KE, Cao H, O’Halloran TV (1999) The ferric uptake regulation (Fur) repressor is a zinc metalloprotein. Biochemistry 38:6559–6569

    Article  CAS  PubMed  Google Scholar 

  • Andrews SC, Robinson AK, Rodriguez-Quinones F (2003) Bacterial iron homeostasis. FEMS Microbiol Rev 27:215–237

    Article  CAS  PubMed  Google Scholar 

  • Archibald F (1983) Lactobacillus plantarum, an organism not requiring iron. FEMS Microbiol Lett 19:29–32

    Article  CAS  Google Scholar 

  • Bagg A, Neilands JB (1987) Ferric uptake regulation protein acts as a repressor, employing iron (II) as a cofactor to bind the operator of an iron transport operon in Escherichia coli. Biochemistry 26:5471–5477

    Article  CAS  PubMed  Google Scholar 

  • Baichoo N, Helmann JD (2002) Recognition of DNA by Fur: a reinterpretation of the Fur box consensus sequence. J Bacteriol 184:5826–5832

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Baichoo N, Wang T, Ye R, Helmann JD (2002) Global analysis of the Bacillus subtilis Fur regulon and the iron starvation stimulon. Mol Microbiol 45:1613–1629

    Article  CAS  PubMed  Google Scholar 

  • Bailey TL, Elkan C (1994) Fitting a mixture model by expectation maximization to discover motifs in biopolymers. In: Second International Conference on Intelligent Systems for Molecular Biology. AAAI Press, Menlo Park, pp 28–36

    Google Scholar 

  • Barnett JP, Millard A, Ksibe AZ, Scanlan DJ, Schmid R, Blindauer CA (2012) Mining genomes of marine cyanobacteria for elements of zinc homeostasis. Front Microbiol 3:1–21

    Article  Google Scholar 

  • Barton HA, Johnson Z, Cox CD, Vasil AI, Vasil ML (1996) Ferric uptake regulator mutants of Pseudomonas aeruginosa with distinct alterations in the iron-dependent repression of exotoxin A and siderophores in aerobic and microaerobic environments. Mol Microbiol 21:1001–1017

    Article  CAS  PubMed  Google Scholar 

  • Bereswill S, Greiner S, van Vliet AH, Waidner B, Fassbinder F, Schiltz E, Kusters JG, Kist M (2000) Regulation of ferritin-mediated cytoplasmic iron storage by the ferric uptake regulator homolog (Fur) of Helicobacter pylori. J Bacteriol 182(21):5948–5953

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Bes MT, Hernandez JA, Peleato M, Fillat MF (2001) Cloning, overexpression and interaction of recombinant Fur from the cyanobacterium Anabaena PCC 7119 with isiB and its own promoter. FEMS Microbiol Lett 194:187–192

    Article  CAS  PubMed  Google Scholar 

  • Boehm M, Yu J, Krynicka V, Barker M, Tichy M, Komenda J (2012) Subunit organization of a Synechocystis hetero-oligomeric thylakoid FtsH complex involved in Photosystem II repair. Plant Cell 24:3669–3683

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Botello-Morte L, Gonzalez A, Bes MT, Peleato ML, Fillat MF (2013) Functional genomics of metalloregulators in cyanobacteria. In: Chauvat F, Cassier-Chauvat C (eds) Genomics of Cyanobacteria. Academic, New York, pp 107–156

    Chapter  Google Scholar 

  • Botello-Morte L, Bes TM, Heras B, Fernandez-Otal A, Peleato ML, Fillat MF (2014) Unraveling the redox properties of the global regulator FurA from Anabaena sp. PCC 7120: Disulfide reductase activity based on its CXXC Motifs. Antiox Redox Signal 20:1396–1406

    Article  CAS  Google Scholar 

  • Boyd PW, Watson AJ, Law CS, Abraham ER, Trull T, Murdoch R, Bakker DCE, Bowie AR, Buesseler KO, Chang H, Charette M, Croot P, Downing K, Frew R, Gall M, Hadfield M, Hall J, Harvey M, Jameson G, LaRoche J, Liddicoat M, Ling R, Maldonado MT, McKay RM, Nodder S, Pickmere S, Pridmore R, Rintoul S, Safi K, Sutton P, Strzepek R, Tanneberger K, Turner S, Waite A, Zeldis J (2000) A mesoscale phytoplankton bloom in the polar Southern Ocean stimulated by iron fertilization. Nature 407:695–702

    Article  CAS  PubMed  Google Scholar 

  • Brahamsha B, Haselkorn R (1992) Identification of multiple RNA polymerase sigma factor homologs in the cyanobacterium Anabaena sp. strain PCC 7120: cloning, expression, and inactivation of the sigB and sigC genes. J Bacteriol 174:7273–7282

    PubMed Central  CAS  PubMed  Google Scholar 

  • Braun V, Hantke K (1991) Genetics of bacterial iron transport. In: Winkelmann G (ed) CRC handbook of microbial iron chelates. CRC Press, Boca Raton, pp 107–138

    Google Scholar 

  • Browning DF, Busby SJ (2004) The regulation of bacterial transcription initiation. Nat Rev Microbiol 2:57–65

    Article  CAS  PubMed  Google Scholar 

  • Bsat N, Helmann JD (1999) Interaction of Bacillus subtilis Fur (ferric uptake repressor) with the dhb operator in vitro and in vivo. J Bacteriol 181:4299–4307

    PubMed Central  CAS  PubMed  Google Scholar 

  • Bsat N, Herbig A, Casillas-Martinez L, Setlow P, Helmann JD (1998) Bacillus subtilis contains multiple Fur homologues: identification of the iron uptake (Fur) and peroxide regulon (PerR) repressors. Mol Microbiol 29:189–198

    Article  CAS  PubMed  Google Scholar 

  • Bullen JJ, Rogers HJ, Griffiths E (1978) Role of iron in bacterial infections. In: Arber W, Henle W, Hofschneider PH, Humphrey JH, Klein J, Koldovský P, Koprowski H, Maaløe O, Melchers F, Rott R, Schweiger HG, Syruek, L, Vogt PK (eds) Curr Top Microbiol Immunol. Springer-Verlag Berlin Heidelberg, Vol 80, pp 1–35

  • Calderwood S, Mekalanos JJ (1988) Conformation of the Fur operator site by insertion of a synthetic oligonucleotide into an operon fusion plasmid. J Bacteriol 170:1015–1017

    PubMed Central  CAS  PubMed  Google Scholar 

  • Chen Q, Crosa JH (1996) Antisense RNA, Fur, iron, and the regulation of iron transport genes in Vibrio anguillarum. J Biol Chem 271:18885–18891

    Article  CAS  PubMed  Google Scholar 

  • Christoffersen CA, Brickman TJ, Hook-Barnard I, McIntosh MA (2001) Regulatory architecture of the iron regulated fepD-ybdA bidirectional promoter region in Escherichia coli. J Bacteriol 183:2059–2070

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Cornelis P, Wie Q, Andrews SC, Vinckx T (2011) Iron homeostasis and management of oxidative stress response in bacteria. Metallomics 3:540–549

    Article  PubMed  CAS  Google Scholar 

  • Cox CD (1980) Iron uptake with ferric pyochelin and ferric citrate by Pseudomonas aeruginosa. J Bacteriol 142:581–587

    PubMed Central  CAS  PubMed  Google Scholar 

  • Cox CD, Rinehart KL, Moore ML, Cook JC (1982) Pyochelin: novel structure of an iron chelating growth promoter for Pseudomonas aeruginosa. Proc Natl Acad Sci 78:4256–4260

    Article  Google Scholar 

  • Coy M (1995) The interaction of the ferric uptake regulation protein with DNA. Biochem Biophys Res Commu 212:784–792

    Article  CAS  Google Scholar 

  • Coy M, Neilands JB (1991) Structural dynamics and functional domains of the Fur protein. Biochemistry 30:8201–8210

    Article  CAS  PubMed  Google Scholar 

  • Davis BM, Quinones M, Pratt J, Ding Y, Waldor MK (2005) Characterization of the small untranslated RNA RyhB and its regulon in Vibrio cholerae. J Bacteriol 187:4005–4014

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • de Figueiredo DR, Azeiteiro UM, Esteves SM, Goncalves FJM, Pereira MJ (2004) Microcystin- producing blooms- a serious global public health issue. Exotoxicol Environ Saf 59:151–163

    Article  CAS  Google Scholar 

  • de Lorenzo V, Wee S, Herrero M, Neilands JB (1987) Operator sequences of the aerobactin operon of plasmid ColV-K30 binding the ferric uptake regulation (Fur) repressor. J Bacteriol 169:2624–2630

    PubMed Central  PubMed  Google Scholar 

  • Delany I, Spohn G, Rappuoli R, Scarlato V (2001) The Fur repressor controls transcription of iron-activated and -repressed genes in Helicobacter pylori. Mol Microbiol 42:1297–1309

    Article  CAS  PubMed  Google Scholar 

  • Delany I, Spohn G, Pacheco A-B F, Ieva R, Alaimo C, Rappuoli R, Scarlato V (2002) Autoregulation of Helicobacter pylori Fur revealed by functional analysis of the iron-binding site. Mol Microbiol 46(4):1107–1122

    Article  CAS  PubMed  Google Scholar 

  • Delany I, Rappuoli R, Scarlato V (2004) Fur functions as an activator and as a repressor of putative virulence gene in Neisseria meningitidis. Mol Microbiol 54:1081–1090

    Article  CAS  Google Scholar 

  • Deore SR, Bansal GK (2013) A study on health hazards caused by microcystins to the animal life. Int J Chem Sci Appl 4:24–28

    Article  CAS  Google Scholar 

  • Dubrac S, Touati D (2000) Fur positive regulation of iron superoxide dismutase in Escherichia coli: functional analysis of the sodB promoter. J Bacteriol 182:3802–3808

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Ducey TF, Carson MB, Orvis J, Stintzi AP, Dyer DW (2005) Identification of the iron-responsive genes of Neisseria gonorrhoeae by microarray analysis in defined medium. J Bacteriol 187:4865–4874

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Duhring U, Axmann IM, Hess WR, Wilde A (2006) An internal antisense RNA regulates expression of the photosynthesis gene isiA. Proc Natl Acad Sci 103:7054–7058

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  • Ernst JF, Bennett RL, Rothfield LI (1978) Constitutive expression of the iron enterochelin and ferrichrome uptake systems in a mutant strain of Salmonella typhimurium. J Bacteriol 135:928–934

    PubMed Central  CAS  PubMed  Google Scholar 

  • Ernst FD, Homuth G, Stoof J, Mader U, Waidner B, Kuipers EJ, Kist M, Kusters JG, Bereswill S, van Vliet AH (2005) Iron-responsive regulation of the Helicobacter pylori iron-cofactored superoxide dismutase SodB is mediated by Fur. J Bacteriol 187(11):3687–3692

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Escolar L, de Lorenzo V, Pearez-Martoan J (1997) Metalloregulation in vitro of the aerobactin promoter of Escherichia coli by the Fur (ferric uptake regulation) protein. Mol Microbiol 26:799–808

    Article  CAS  PubMed  Google Scholar 

  • Escolar L, Perez-Martın J, de Lorenzo V (1998) Binding of the Fur (ferric uptake regulator) repressor of Escherichia coli to arrays of the GATAAT sequence. J Mol Biol 283:537–547

    Article  CAS  PubMed  Google Scholar 

  • Ferrao-Filho AS, Kozlowsky-Suzuki B (2011) Cyanotoxin: bioaccumulation and effects on aquatic animals. Mar Drug 9:2729–2772

    Article  CAS  Google Scholar 

  • Ferreira F, Straus NA (1994) Iron deprivation in cyanobacteria. J Appl Phycol 6:199–210

    Article  CAS  Google Scholar 

  • Ferris JP (2005) Mineral catalysis and prebiotic synthesis: montmorillonite-catalyzed formation of RNA. Elements 1:145–149

    Article  CAS  Google Scholar 

  • Fillat MF (2014) The FUR (ferric uptake regulator) superfamily: diversity and versatility of key transcriptional regulators. Arch Biochem Biophys 546:41–52

    Article  CAS  PubMed  Google Scholar 

  • Florencio FJ, Perez-Perez ME, Lopez-Maury L, Mata-Cabana A, Lindahl M (2006) The diversity and complexity of the cyanobacterial thioredoxin systems. Photosynth Res 89:157–171

    Article  CAS  PubMed  Google Scholar 

  • Fuangthong M, Helmann JD (2003) Recognition of DNA by three ferric uptake regulator (Fur) homologs in Bacillus subtilis. J Bacteriol 185:6348–6357

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Gaballa A, Antelmann H, Aguilar C, Khakh SK, Song KB, Smaldone GT, Helmann JD (2008) The Bacillus subtilis iron-sparing response is mediated by a Fur-regulated small RNA and three small, basic proteins. Proc Natl Acad Sci U S A 105:11927–11932

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Gao H, Zhou D, Li Y, Guo Z, Han Y, Song Y, Zhai J, Du Z, Wang X, Lu J, Yang R (2008) The iron-responsive Fur regulon in Yersinia pestis. J Bacteriol 190:3063–3075

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Ghassemian M, Straus NA (1996) Fur regulates the expression of iron-stress genes in the cyanobacterium Synechococcus sp. strain PCC 7942. Microbiology 142:1469–1476

    Article  CAS  PubMed  Google Scholar 

  • Gonzalez A, Bes MT, Peleato ML, Fillat MF (2011) Unraveling the regulatory function of FurA in Anabaena sp. PCC 7120 through 2-D DIGE proteomic analysis. J Proteomics 74:660–671

    Article  CAS  PubMed  Google Scholar 

  • Gonzalez A, Bes MT, Valladares A, Peleato ML, Fillat MF (2012) FurA is the master regulator of iron homeostasis and modulates the expression of tetrapyrrole biosynthesis genes in Anabaena sp. PCC 7120. Environ Microbiol 14:3175–3187

    Article  CAS  PubMed  Google Scholar 

  • Gonzalez A, Angarica AE, Sancho J, Fillat MF (2014) The FurA regulon in Anabaena sp. PCC 7120: in silico prediction and experimental validation of novel target genes. Nucleic Acids Res 42:4833–4846

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Gottesman S (2002) Stealth regulation: biological circuits with small RNA switches. Genes Dev 16:2829–2842

    Article  CAS  PubMed  Google Scholar 

  • Griggs DW, Konisky J (1989) Mechanism for iron-regulated transcription of the Escherichia coli cir gene: metal-dependent binding of Fur protein to the promoters. J Bacteriol 171:1048–1054

    PubMed Central  CAS  PubMed  Google Scholar 

  • Griggs DG, Tharp BB, Konisky J (1987) Cloning and promoter identification of the iron-regulated cir gene of Escherichia coli. J Bacteriol 169:5343–5352

    PubMed Central  CAS  PubMed  Google Scholar 

  • Hahn JS, Oh SY, Roe JH (2000) Regulation of the furA and catC Operon, encoding a Ferric Uptake Regulator homologue and Catalase-Peroxidase, respectively, in Streptomyces coelicolor A3. J Bacteriol 182:3767–3774

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Hall HK, Foster JW (1996) The role of Fur in the acid tolerance response of Salmonella typhimurium is physiologically and genetically separable from its role in iron acquisition. J Bacteriol 178:5683–5691

    PubMed Central  CAS  PubMed  Google Scholar 

  • Hamza I, Chauhan VS, Hassett R, O’ Brian MR (1998) The bacterial Irr protein is required for coordination of heme biosynthesis with iron availability. J Biol Chem 273:21669–22167

    Article  CAS  PubMed  Google Scholar 

  • Hantke K (1981) Regulation of ferric iron transport in Escherichia coli K12: isolation of a constitutive mutant. Mol Gen Genet 182:288–292

    Article  CAS  PubMed  Google Scholar 

  • Hayat R, Ali S, Amara U, Khalid R, Ahmed I (2010) Soil beneficial bacteria and their role in plant growth promotion: a review. Ann Microbiol 60:579–598

    Article  Google Scholar 

  • Heidrich C, Hantke K, Bierbaum G, Sahl HG (1996) Identification and analysis of a gene encoding a Fur-like protein of Staphylococcus epidermidis. FEMS Microbiol Lett 14:253–259

    Article  Google Scholar 

  • Hennecke H (1990) Regulation of bacterial gene expression by metal- protein complexes. Mol Microbiol 4:1621–1628

    Article  CAS  PubMed  Google Scholar 

  • Heras B, Kurz M, Shouldice SR, Martin JL (2007) The name’s bond…disulfide bond. Curr Opin Struct Biol 17:691–698

    Article  CAS  PubMed  Google Scholar 

  • Hernandez JA, Bes MT, Fillat MF, Neira JL, Peleato ML (2002) Biochemical analysis of the recombinant Fur (ferric uptake regulator) protein from Anabaena PCC 7119: factors affecting its oligomerization state. Biochem J 366:315–322

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Hernandez JA, Lopez-Gomollon S, Bes MT, Fillat MF, Peleato ML (2004a) Three Fur homologues from Anabaena sp. PCC 7120: exploring reciprocal protein-promoter recognition. FEMS Microbiol Lett 236:275–282

    Article  CAS  PubMed  Google Scholar 

  • Hernandez JA, Peleato ML, Fillat MF, Bes MT (2004b) Heme binds to and inhibits the DNA-binding activity of the global regulator FurA from Anabaena sp. PCC 7120. FEBS Lett 577:35–41

    Article  CAS  PubMed  Google Scholar 

  • Hernandez JA, Meier J, Barrera FN, de los Panos OR, Hurtado-Gomez E, Bes MT, Fillat MF, Peleato ML, Cavasotto CN, Neira JL (2005) The conformational stability and thermodyanamics of FurA (Ferric Uptake Regulator) from Anabaena sp. PCC 7119. Biophys J 89:4188–4200

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Hernandez JA, Muro-Pastor AM, Flores E, Bes MT, Peleato ML, Fillat MF (2006a) Identification of a furA cis antisense RNA in the cyanobacterium Anabaena sp. PCC 7120. J Mol Biol 355:325–334

    Article  CAS  PubMed  Google Scholar 

  • Hernandez JA, Lopez-Gomollon S, Muro-Pastor A, Valladares A, Bes MT, Peleato ML, Fillat MF (2006b) Interaction of FurA from Anabaena sp. PCC 7120 with DNA: A reducing environment and the presence of Mn2+ are positive effectors in the binding to isiB and furA promoters. BioMethods 19:259–268

    Article  CAS  Google Scholar 

  • Hernandez JA, Pellicer S, Huang L, Peleato ML, Fillat MF (2007) FurA modulates gene expression of alr3808, a DpsA homologue in Nostoc (Anabaena) sp. PCC 7120. FEBS Lett 581:1351–1356

    Article  CAS  PubMed  Google Scholar 

  • Hernandez JA, Alonso I, Pellicer S, Luisa Peleato M, Cases R, Strasser RJ, Barja F, Fillat MF (2010) Mutants of Anabaena sp. PCC 7120 lacking alr1690 and α-furA antisense RNA show a pleiotropic phenotype and altered photosynthetic machinery. J Plant Physiol 167:430–437

    Article  CAS  PubMed  Google Scholar 

  • Herrero A, Muro-Pastor AM, Flores E (2001) Nitrogen control in cyanobacteria. J Bacteriol 183:411–425

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Holm L, Syer C, Ruterjans H, Schnarr M, Fogh R, Boelens R, Kaptein R (1994) LexA repressor and iron uptake regulator from Escherichia coli: new members of the CAP-like DNA binding domain superfamily. Protein Eng 7:14449–14453

    Google Scholar 

  • Horsburgh MJ, Ingham E, Foster SJ (2001) In Staphylococcus aureus, Fur is an interactive regulator with PerR, contributes to virulence, and is necessary for oxidative stress resistance through positive regulation of catalase and iron homeostasis. J Bacteriol 183:468–475

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Hunt MD, Pettis GS, McIntosh MA (1994) Promoter and operator determinants for Fur-mediated iron regulation in the bidirectional fepA-fes control region of the Escherichia coli enterobactin gene system. J Bacteriol 176:3944–3955

    PubMed Central  CAS  PubMed  Google Scholar 

  • Igarashi J, Kitanishi K, Shimizu T (2011) Emerging roles of heme as a signal and a gas-sensing site: heme sensing and gas-sensing proteins. In: Kadish KM, Smith KM, Guilard R (eds) Handbook of porphyrin science, vol 15. World Scientific Publishing Co, Singapore, pp 399–461

    Google Scholar 

  • Irieda H, Morita T, Maki K, Homma M, Aiba H, Sudo Y (2012) Photo-induced regulation of the chromatic adaptive gene expression by Anabaena sensory rhodopsin. J Biol Chem 287:32485–32493

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Isabella V, Wright LF, Barth K, Spence JM, Grogan S, Genco CA, Clark VL (2008) cis- and trans-acting elements involved in regulation of norB (norZ), the gene encoding nitric oxide reductase in Neisseria gonorrhoeae. Microbiology 154:226–239

    Article  CAS  PubMed  Google Scholar 

  • Jacquamet L, Traoré DAK, Ferrer J-L, Proux O, Testemale D, Hazemann J-L, Nazarenko E, Ghazouani AE, Caux Thang C, Duarte V, Latour J-M (2009) Structural characterization of the active form of PerR: insights into the metal-induced activation of PerR and Fur proteins for DNA binding. Mol Microbiol 73:20–31

    Article  CAS  PubMed  Google Scholar 

  • Jacques JF, Jang S, Prevost K, Desnoyers G, Desmarais M, Imlay J, Masse E (2006) RyhB small RNA modulates the free intracellular iron pool and is essential for normal growth during iron limitation in Escherichia coli. Mol Microbiol 62:1181–1190

    Article  CAS  PubMed  Google Scholar 

  • Jeanjean R, Talla E, Latifi A, Havaux M, Janicki A, Zhang CC (2008) A large gene cluster encoding peptide synthetases and polyketide synthases is involved in production of siderophores and oxidative stress response in the cyanobacterium Anabaena sp. strain PCC 7120. Environ Microbiol 10:2574–2585

    Article  CAS  PubMed  Google Scholar 

  • Kallifidas D, Pascoe B, Owen GA, Strain-Damerell CM, Hong HJ, Paget MSB (2009) The zinc-responsive regulator zur controls expression of the coelibactin gene cluster in Streptomyces coelicolor. J Bacteriol 192:608–611

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  • Kammler M, Schon C, Hantke K (1993) Characterization of the ferrous iron uptake system of Escherichia coli. J Bacteriol 175:6212–6219

    PubMed Central  CAS  PubMed  Google Scholar 

  • Kaneko T, Sato S, Kotani H, Tanaka A, Asamizu E, Nakamura Y, Miyajima N, Hirosawa M, Sugiura M, Sasamoto S (1996) Sequence analysis of the genome of the unicellular cyanobacterium Synechocystis sp. strain PCC 6803. II. Sequence determination of the entire genome and assignment of potential protein-coding regions. DNA Res 3:109–136

    Article  CAS  PubMed  Google Scholar 

  • Katayama M, Ohmori M (1997) Isolation and characterization of multiple adenylate cyclase genes from the cyanobacterium Anabaena sp. strain PCC 7120. J Bacteriol 179:3588–3593

    PubMed Central  CAS  PubMed  Google Scholar 

  • Kaushik MS, Srivastava M, Verma E, Mishra AK (2015) Role of manganese in protection against oxidative stress under iron starvation in cyanobacterium Anabaena 7120. J Basic Microbiol 55:729–740

    Article  CAS  PubMed  Google Scholar 

  • Klebba PE, McIntosh MA, Neilands JB (1982) Kinetics of biosynthesis of iron-regulated membrane proteins in Escherichia coli. J Bacteriol 149:880–888

    PubMed Central  CAS  PubMed  Google Scholar 

  • Kolb A, Busby S, Buc H, Garges S, Adhya S (1993) Transcriptional regulation by cAMP and its receptor protein. Annu Rev Biochem 62:749–795

    Article  CAS  PubMed  Google Scholar 

  • Krynicka V, Tichý M, Krafl J, Yu J, Kanˇa R, Boehm M, Nixon PJ, Komenda J (2014) Two essential FtsH proteases control the level of the Fur repressor during iron deficiency in the cyanobacterium Synechocystis sp. PCC 6803. Mol Microbiol 94:609–624

    Article  CAS  PubMed  Google Scholar 

  • Kunert A, Vinnemeier J, Erdmann N, Hagemann M (2003) Repression by Fur is not the main mechanism controlling the iron-inducible isiAB operon in the cyanobacterium Synechocystis sp. PCC 6803. FEMS Microbiol Lett 227:255–262

    Article  CAS  PubMed  Google Scholar 

  • Latifi A, Ruiz M, Zhang CC (2009) Oxidative stress in cyanobacteria. FEMS Microbiol Rev 33:258–278

    Article  CAS  PubMed  Google Scholar 

  • Lavrrar JL, Christoffersen CA, McIntosh MA (2003) Fur–DNA interactions at the bidirectional fepDGC-entS promoter region in Escherichia coli. J Mol Biol 322:983–995

    Article  CAS  Google Scholar 

  • Lee HJ, Bang SH, Lee KH, Park SJ (2007) Positive regulation of fur gene expression via direct interaction of fur in a pathogenic bacterium, Vibrio vulnificus. J Bacteriol 189:2629–2636

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Lewin AC, Doughty PA, Flegg L, Moore GR, Spiro S (2002) The ferric uptake regulator of Pseudomonas aeruginosa has no essential cysteine residues and does not contain a structural zinc ion. Microbiology 148:2449–2456

    Article  CAS  PubMed  Google Scholar 

  • Li H, Singh H, McIntyre LM, Sherman LA (2004) Differential gene expression in response to hydrogen peroxide and the putative perR regulon of Synechocystis sp. strain PCC 6803. J Bacteriol 11:3331–3345

    Article  CAS  Google Scholar 

  • Lindsay JA, Foster SJ (2001) zur: a Zn2+-responsive regulatory element of Staphylococcus aureus. Microbiology 147:1259–1266

    Article  CAS  PubMed  Google Scholar 

  • Litwin CM, Calderwood SB (1994) Analysis of the complexity of gene regulation by fur in Vibrio cholerae. J Bacteriol 176(1):240–248

    PubMed Central  CAS  PubMed  Google Scholar 

  • Litwin M, Boyko SA, Calderwood SB (1992) Cloning, sequencing and transcriptional regulation of the Vibrio cholerae fur gene. J Bacteriol 174:1897–1903

    PubMed Central  CAS  PubMed  Google Scholar 

  • Lopez-Gomollon S, Hernandez JA, Wolk CP, Peleato ML, Fillat MF (2006) Expression of FurA is modulated by NtcA and strongly enhanced in heterocysts of Anabaena sp. PCC 7120. Microbiology 153:42–50

    Article  CAS  Google Scholar 

  • Lopez-Gomollón S, Hernández JA, Pellicer S, Angarica VE, Peleato ML, Fillat MF (2007) Cross-talk between iron and nitrogen regulatory networks in Anabaena (Nostoc) sp. PCC 7120: identification of overlapping genes in furA and ntcA regulons. J Mol Biol 374:267–281

    Article  PubMed  CAS  Google Scholar 

  • Lopez-Gomollon S, Sevilla S, Bes MT, Peleato ML, Fillat MF (2009) New insights into the role of Fur proteins: Fur (All2473) from Anabaena protects DNA and increases cell survival under oxidative stress. Biochem J 418:201–207

    Article  CAS  PubMed  Google Scholar 

  • Lukac M, Aegerter R (1993) Influence of trace metals on growth and toxin production of Microcystis aeruginosa. Toxicon 31:293–305

    Article  CAS  PubMed  Google Scholar 

  • Maciag A, Dainese E, Rodriguez GM, Milano A, Provvedi R, Pasca MR, Smith I, Palu G, Riccardi G, Manganelli R (2007) Global analysis of the Mycobacterium tuberculosis Zur (FurB) regulon. J Bacteriol 189:730–740

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Martin-Luna B, Hernandez JA, Bes MT, Fillat MF, Peleato ML (2005) Identification of a Ferric uptake regulator (Fur) from Microcystis aeruginosa PCC 7806. FEMS Microbiol Lett 254:63–70

    Article  CAS  Google Scholar 

  • Martin-Luna B, Sevilla E, Hernandez JA, Bes MT, Fillat MF, Peleato ML (2006) Fur from Microcystis aeruginosa binds in vitro promoter regions of the microcystin biosynthesis gene cluster. Phytochemistry 67:876–881

    Article  CAS  PubMed  Google Scholar 

  • Martin-Luna B, Sevilla E, Gonzalez A, Bes MT, Fillat MF, Peleato ML (2011) Expression of fur and its antisense fur from Microcystis aeruginosa PCC 7806 as response to light and oxidative stress. J Plant Physiol 168:2244–2250

    Article  CAS  PubMed  Google Scholar 

  • Masse E, Gottesman S (2002) A small RNA regulates the expression of genes involved in iron metabolism in Escherichia coli. Proc Natl Acad Sci 99:4620–4625

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Masse E, Majdalani N, Gottesman S (2003) Regulatory roles for small RNAs in bacteria. Curr Opin Microbiol 6:120–124

    Article  CAS  PubMed  Google Scholar 

  • Masse E, Vanderpool CK, Gottesman S (2005) Effect of RyhB small RNA on global iron use in Escherichia coli. J Bacteriol 187:6962–6971

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Masse E, Salvail H, Desnoyers G, Arguin M (2007) Small RNAs controlling iron metabolism. Curr Opin Microbiol 10(2):140–145

    Article  CAS  PubMed  Google Scholar 

  • Mellin JR, Goswami S, Grogan S, Tjaden B, Genco CA (2007) A novel Fur- and iron-regulated small RNA, NrrF, is required for indirect Fur mediated regulation of the sdhA and sdhC genes in Neisseria meningitidis. J Bacteriol 189:3686–3694

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Mellin JR, McClure R, Lopez D, Green O, Reinhard B, Genco C (2010) Role of Hfq in iron-dependent and –independent gene regulation in Neisseria meningitidis. Microbiology 156:2316–2326

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Mey AR, Craig SA, Payne SM (2005) Characterization of Vibrio cholerae RyhB: the RyhB regulon and role of ryhB in biofilm formation. Infect Immun 73:5706–5719

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Meyer JM (2000) Pyoverdins: pigments, siderophores and potential taxonomic markers of fluorescent Pseudomonas species. Arch Microbiol 174:135–142

    Article  CAS  PubMed  Google Scholar 

  • Michaud-Soret I, Adrait A, Jaquinod M, Forest E, Touati D, Latour JM (1997) Electrospray ionization mass spectroscopy analysis of theapo- and metal-substituted forms of the Fur protein. FEBS Lett 413:473–476

    Article  CAS  PubMed  Google Scholar 

  • Michel KP, Kruger F, Puhler A, Pistorius EK (1999) Molecular characterization of idiA and adjacent genes in the cyanobacteria Synechococcus sp. strains PCC 6301 and PCC 7942. Microbiology 145:1473–1484

    Article  CAS  PubMed  Google Scholar 

  • Miles S, Carpenter BM, Gancz H, Merrell DS (2010) Helicobacter pylori apo-Fur regulation appears unconserved across species. J Microbiol 48:378–386

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Mills SA, Marletta MA (2005) Metal binding characteristics and role of iron oxidation in the ferric uptake regulator from Escherichia coli. Biochemistry 44:13553–13559

    Article  CAS  PubMed  Google Scholar 

  • Nandal A, Huggins CC, Woodhall MR, McHugh J, Rodriguez-Quinones F, Quail MA (2010) Induction of the ferritin gene (ftnA) of Escherichia coli by Fe2+-Fur is mediated by reversal of H-NS silencing and is RyhB independent. Mol Microbiol 75:637–657

    Article  CAS  PubMed  Google Scholar 

  • Napolitano M, Rubio MÁ, Santamaría-Gómez J, Olmedo-Verd E, Robinson NJ, Luque I (2012) Characterization of the response to zinc deficiency in the cyanobacterium Anabaena sp. Strain PCC 7120. J Bacteriol 194:2426–2436

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Neilands JB, Nakamura K (1991) Detection, determination, isolation, characterization and regulation of microbial iron chelates. In: Winkelmann G (ed) Handbook of microbial iron chelates. CRC Press, Boca Raton, pp 1–14

    Google Scholar 

  • Ogawa K, Sun J, Taketani S, Nakajima O, Nishitani C, Sassa S, Hayashi N, Yamamoto M, Shibahara S, Fujita H (2001) Heme mediates derepression of Maf recognition element through direct binding to transcription repressor Bach1. EMBO J 20:2835–2843

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Okamoto S, Kasahara M, Kamiya A, Nakahira Y, Ohmori M (2004) A phytochrome-like protein AphC triggers the cAMP signaling induced by far-red light in the cyanobacterium Anabaena sp. strain PCC7120. Photochem Photobiol 80:429–433

    Article  CAS  PubMed  Google Scholar 

  • Ortiz de Orue Lucana D, Schrempf H (2000) The DNA-binding characteristics of the Streptomyces reticuli regulator FurS depend on the redox state of its cysteine residues. Mol Gen Genet 264:341–353

    Article  CAS  PubMed  Google Scholar 

  • Paerl HW, Fulton RS, Moisander PH (2001) Harmful freshwater algal blooms, with an emphasis on cyanobacteria. Sci World 1:76–113

    Article  CAS  Google Scholar 

  • Paoli M, Marles-Wright J, Smith A (2002) Structure–function relationships in heme-proteins. DNA Cell Biol 21:271–280

    Article  CAS  PubMed  Google Scholar 

  • Patzer SI (2000) The zinc-responsive regulator Zur and its control of the znu gene cluster encoding the ZnuABC zinc uptake system in Escherichia coli. J Biol Chem 275:24321–24332

    Article  CAS  PubMed  Google Scholar 

  • Pellicer S, Gonzalez A, Peleato ML, Martinez JI, Fillat MF, Bes MT (2012) Site-directed mutagenesis and spectral studies suggest a putative role of FurA from Anabaena sp. PCC 7120 as a heme sensor protein. FEBS J 279:2231–2246

    Article  CAS  PubMed  Google Scholar 

  • Pohl E, Holmes RK, Hol WGJ (1999) Crystal structure of a cobalt-activated diphtheria toxin repressor–DNA complex reveals a metal-binding SH3-like domain. J Mol Biol 292:653–667

    Article  CAS  PubMed  Google Scholar 

  • Pohl E, Haller JC, Mijovilovich A, Meyer-Klaucke W, Garman E, Vasil ML (2003) Architecture of a protein central to iron homeostasis: crystal structure and spectroscopic analysis of the ferric uptake regulator. Mol Microbiol 47:903–915

    Article  CAS  PubMed  Google Scholar 

  • Pressler U, Staudenmaier H, Zimmermann L, Braun V (1988) Genetics of the iron dicitrate transport system of Escherichia coli. J Bacteriol 170:2716–2724

    PubMed Central  CAS  PubMed  Google Scholar 

  • Quan S, Schneider I, Pan J, Von Hacht A, Bardwell JC (2007) The CXXC motif is more than a redox rheostat. J Biol Chem 282:28823–28833

    Article  CAS  PubMed  Google Scholar 

  • Rueter JG, Petersen RR (1987) Micronutrient effects on cyanobacterial growth and physiology. N Z J Mar Freshw Res 21:435–445

    Article  Google Scholar 

  • Saito T, Williams RJP (1991) The binding of the ferric uptake regulation protein to a DNA fragment. Eur J Biochem 197:43–47

    Article  CAS  PubMed  Google Scholar 

  • Saito I, Wormald MR, Williams RJP (1991) Some structural features of the iron-uptake regulation protein. Eur J Biochem 197:29–38

    Article  CAS  PubMed  Google Scholar 

  • Salinas PC, Tolmasky ME, Crosa JH (1989) Regulation of the iron uptake system in Vibrio anguillarum: evidence for a cooperative effect between two transcriptional activators. Proc Natl Acad Sci U S A 86:3529–3533

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Sebastian S, Agarwal S, Murphy JR, Genco CA (2002) The gonococcal fur regulon: identification of additional genes involved in major catabolic, recombination, and secretory pathways. J Bacteriol 184:3965–3974

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Sein-Echaluce VC, González A, Napolitano M, Luque I, Barja F, Peleato ML, Fillat MF (2014) Zur (FurB) is a key factor in the control of the oxidative stress response in Anabaena sp. PCC 7120. Environ Microbiol. doi:10.1111/1462-2920.12628

    PubMed  Google Scholar 

  • Sevier CS, Kaiser CA (2002) Formation and transfer of disulphide bonds in living cells. Nat Rev Mol Cell Biol 3:836–847

    Article  CAS  PubMed  Google Scholar 

  • Sheikh MA, Taylor GL (2009) Crystal structure of the Vibrio cholerae ferric uptake regulator (Fur) reveals insights into metal co-ordination. Mol Microbiol 72:1208–1220

    Article  CAS  PubMed  Google Scholar 

  • Singh AK, McIntyre LM, Sherman LA (2003) Microarray analysis of the genome-wide response to iron deficiency and iron reconstitution in the cyanobacterium Synechocystis sp. PCC 6803. Plant Physiol 132:1825–1839

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Singh A, Mishra AK, Singh SS, Shukla E (2008) Influence of iron and chelator on siderophore production in Frankia strains nodulating Hippopheae salicifolia D. Don. J Basic Microbiol 48:104–111

    Article  CAS  PubMed  Google Scholar 

  • Singh A, Singh SS, Pandey PC, Mishra AK (2010) Attenuation of metal toxicity by Frankial siderophores. Toxicol Environ Chem 92:1339–1346

    Article  CAS  Google Scholar 

  • Smith A, Hooper NI, Shipulina N, Morgan WT (1996) Heme binding by a bacterial repressor protein, the gene product of the ferric uptake regulation (Fur) gene of Escherichia coli. J Protein Chem 15:575–583

    Article  CAS  PubMed  Google Scholar 

  • Somerville G, Mikoryak CA, Reitzer L (1999) Physiological characterization of Pseudomonas aeruginosa during exotoxin A synthesis: glutamate, iron limitation, and aconitase activity. J Bacteriol 181:1072–1078

    PubMed Central  CAS  PubMed  Google Scholar 

  • Stojiljkovic I, Hantke K (1995) Functional domains of the Escherichia coli ferric uptake regulator protein (Fur). Mol Gen Genet 247:199–205

    Article  CAS  PubMed  Google Scholar 

  • Stojiljkovic I, Baumler AJ, Hantke K (1994) Fur regulon in Gram negative bacteria. Identification and characterization of new iron-regulated Escherichia coli genes by a Fur titration assay. J Mol Biol 236:531–545

    Article  CAS  PubMed  Google Scholar 

  • Straus NA (1994) Iron deprivation: physiology and gene regulation. In: Bryant DA (ed) The molecular biology of cyanobacteria. Kluwer academic publisher, Netherlands, pp 731–750

    Chapter  Google Scholar 

  • Teixido L, Carrasco B, Alonso JC, Barbe J, Campoy S (2011) Fur activates the expression of Salmonella enterica pathogenicity island 1 by directly interacting with the hilD operator in vivo and in vitro. PLoS One 6, e19711

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Thompson DK, Beliaev AS, Giometti CS, Tollaksen SL, Khare T, Lies DP, Nealson KH, Lim H, Yates J, Brandt CC, Tiedje JM, Zhou J (2002) Transcriptional and proteomic analysis of a Ferric Uptake Regulator (Fur) mutant of Shewanella oneidensis: possible involvement of Fur in energy metabolism, transcriptional regulation, and oxidative stress. Appl Environ Microbiol 68:881–892

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Touati D (2000) Iron and oxidative stress in bacteria. Arch Biochem Biophys 373:1–6

    Article  CAS  PubMed  Google Scholar 

  • Utkilen H, Gjolme N (1995) Iron-stimulated toxin production in Microcystis aeruginosa. Appl Environ Microbiol 61(2):797–800

    PubMed Central  CAS  PubMed  Google Scholar 

  • Wang Y, Zhang X, Feng S, Niu Z, Chen C (2009) Study on inactivation of iron bacteria isolated from real drinking water distribution systems by free chlorine and chloramines. Ann Microbiol 59(2):353–358

    Article  CAS  Google Scholar 

  • Wang Y, Mo X, Zhang L, Wang Q (2011) Four superoxide dismutase (isozymes) genes of Bacillus cereus. Ann Microbiol 61:355–360

    Article  CAS  Google Scholar 

  • Wee S, Neilands JB, Bittner ML, Hemming BC, Haymore BL, Seetharam R (1988) Expression, isolation and properties of Fur (ferric uptake regulation) protein of Escherichia coli K-12. Biol Metals 1:62–68

    Article  CAS  Google Scholar 

  • White A, Ding X, vander Spek JC, Murphy JR, Ringe D (1998) Structure of the metal-ion-activated diphtheria toxin repressor/tox operator complex. Nature 394:502–506

    Article  CAS  PubMed  Google Scholar 

  • Wilderman PJ, Sowa NA, FitzGerald DJ, FitzGerald PC, Gottesman S, Ochsner UA, Vasil ML (2004) Identification of tandem duplicate regulatory small RNAs in Pseudomonas aeruginosa involved in iron homeostasis. Proc Natl Acad Sci 101:9792–9797

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Xiong A, Singh VK, Cabrera G, Jayaswal RK (2000) Molecular characterization of the ferric-uptake regulator, Fur, from Staphylococcus aureus. Microbiology 146:659–668

    Article  CAS  PubMed  Google Scholar 

  • Yingping F, Lemeille S, Talla E, Janicki A, Denis Y, Zhang C-C, Latifi A (2014) Unravelling the cross-talk between iron starvation and oxidative stress responses highlights the key role of PerR (alr0957) in peroxide signalling in the cyanobacterium Nostoc PCC 7120. Environ Microbiol Rep 6:468–475

    Article  CAS  PubMed  Google Scholar 

  • Yu C, Genco CA (2012) Fur mediated global regulatory circuit in pathogenic Neisseria species. J Bacteriol 194:6372–6381

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Zhang L, Guarente L (1995) Heam binds to a short sequence that serves a regulatory function in diverse proteins. EMBO J 14:313–320

    PubMed Central  CAS  PubMed  Google Scholar 

  • Zheleznova EE, Crosa JH, Brennan RG (2000) Characterization of the DNA- and metal-binding properties of Vibrio anguillarum Fur reveals conservation of a structural Zn2+ ion. J Bacteriol 182:6264–6267

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Zheng M, Doan B, Schneider TD, Storz G (1999) OxyR and SoxRS regulation of Fur. J Bacteriol 181:4639–4643

    PubMed Central  CAS  PubMed  Google Scholar 

  • Zhou D, Qin L, Han Y, Qiu J, Chen Z, Li B, Song Y, Wang J, Guo Z, Zhai J, Du Z, Wang X, Yang R (2006) Global analysis of iron assimilation and Fur regulation in Yersinia pestis. FEMS Microbiol Lett 258:9–17

    Article  CAS  PubMed  Google Scholar 

  • Zou P, Borovok I, Ortiz de Orué Lucana D, Müller D, Schrempf H (1999) The mycelium-associated Streptomyces reticuli catalase-peroxidase, its gene and regulation by FurS. Microbiology 145:549–559

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We are thankful to the Council of Scientific and Industrial Research, New Delhi for providing financial support. The Head of the Department of Botany, Banaras Hindu University, Varanasi, India is gratefully acknowledged for providing the laboratory facility.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Arun Kumar Mishra.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kaushik, M.S., Singh, P., Tiwari, B. et al. Ferric Uptake Regulator (FUR) protein: properties and implications in cyanobacteria. Ann Microbiol 66, 61–75 (2016). https://doi.org/10.1007/s13213-015-1134-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13213-015-1134-x

Keywords