Skip to main content

Bifunctional protein ArsRM contributes to arsenite methylation and resistance in Brevundimonas sp. M20

Abstract

Background

Arsenic (As) with various chemical forms, including inorganic arsenic and organic arsenic, is the most prevalent water and environmental toxin. This metalloid occurs worldwide and many of its forms, especially arsenite [As(III)], cause various diseases including cancer. Organification of arsenite is an effective way for organisms to cope with arsenic toxicity. Microbial communities are vital contributors to the global arsenic biocycle and represent a promising way to reduce arsenite toxicity.

Methods

Brevundimonas sp. M20 with arsenite and roxarsone resistance was isolated from aquaculture sewage. The arsHRNBC cluster and the metRFHH operon of M20 were identified by sequencing. The gene encoding ArsR/methyltransferase fusion protein, arsRM, was amplified and expressed in Escherichia coli BL21 (DE3), and this strain showed resistance to arsenic in the present of 0.25–6 mM As(III), aresenate, or pentavalent roxarsone. The methylation activity and regulatory action of ArsRM were analyzed using Discovery Studio 2.0, and its functions were confirmed by methyltransferase activity analysis and electrophoretic mobility shift assays.

Results

The minimum inhibitory concentration of the roxarsone resistant strain Brevundimonas sp. M20 to arsenite was 4.5 mM. A 3,011-bp arsenite resistance ars cluster arsHRNBC and a 5649-bp methionine biosynthesis met operon were found on the 3.315-Mb chromosome. Functional prediction analyses suggested that ArsRM is a difunctional protein with transcriptional regulation and methyltransferase activities. Expression of ArsRM in E. coli increased its arsenite resistance to 1.5 mM. The arsenite methylation activity of ArsRM and its ability to bind to its own gene promoter were confirmed. The As(III)-binding site (ABS) and S-adenosylmethionine-binding motif are responsible for the difunctional characteristic of ArsRM.

Conclusions

We conclude that ArsRM promotes arsenite methylation and is able to bind to its own promoter region to regulate transcription. This difunctional characteristic directly connects methionine and arsenic metabolism. Our findings contribute important new knowledge about microbial arsenic resistance and detoxification. Future work should further explore how ArsRM regulates the met operon and the ars cluster.

Peer Review reports

Introduction

Arsenic, classified as a Group 1 human carcinogen by the International Agency for Research on Cancer, is the most prevalent water and environmental toxin [1], and ranks top of the US Priority List of Hazardous Substances. Arsenic compounds enter the biosphere from geochemical sources and anthropogenic sources. Herbicides, growth promoters for farm animals, the semiconductor industry, and other industrial sources contribute to arsenic contamination [2]. Humans are exposed to arsenic daily [3], mostly from food and water supplies. This exposure lead to numerous diseases, including cardiovascular and peripheral vascular diseases, neurological disorders, diabetes mellitus, chronic kidney disease [4], and cancer [3, 4]. In addition, low birth rate, fetal death, and delayed infant development are closely associated with arsenic exposure during pregnancy [5].

In the environment, inorganic arsenic exists in various chemical forms, including arsenate [As(V)], arsenite [As(III)], elemental arsenic [As(0)], and arsine [As(− 3)]. The predominant forms in oxic and reducing environments are As(V) and As(III), respectively [2]. Organic arsenic compounds include pentavalent roxarsone [Rox(V)], methylarsenate [MAs(V)], trivalent roxarsone [Rox(III)] [6], and methylarsenite [MAs(III)] [7]. In the past, the organoarsenic compound roxarsone was extensively used as a organoarsenic feed additive for poultry. Although its use has been forbidden in many countries in the world, residues of roxarsone in the environment still pollute waterways and lead to the enrichment of arsenic-tolerant bacteria [8, 9]. These relatively benign organic arsenic compounds including roxarsone, however, are largely degraded into more toxic inorganic forms after they are introduced into the environment [2]. As an important part of the ecosystem, microbial communities are an vital contributors to the global arsenic biocycle [10].

Arsenic resistance (ars) genes that confer resistance to arsenic and various organic arsenic compounds, have been identified in plasmids and/or chromosomes of various prokaryotes and eukaryotes [11,12,13,14,15]. ars genes are usually present in clusters, and at least one arsenic resistance system seems to be a necessary component of the genome in prokaryotic species, because canonical arsRBC and its variants appear to be quite common in bacterial and archaeal species [16]. ArsR, is a SmtB/ArsR family of metalloregulatory proteins that controls the transcription of the ars operon [16, 17]. Acr3 and ArsB are responsible for pumping As(III) from the cytosol across the cytoplasmic membrane into the periplasm or extracellular medium [18]. ArsC, an As(V) reductase, reduces intracellular As(V) to As(III) [19]. In organisms, As(III) tolerance and detoxification can be achieved by efflux and methylation [3, 13, 14]. ArsM, an S-adenosylmethionine (SAM) methyltransferase, catalyzes the methylation of As(III) using SAM as the substrate in microbes [2]. ArsH is an organoarsenical oxidase that confers resistance to trivalent forms of monosodium methanearsonate and roxarsone [20, 21].

In this study, a roxarsone-resistant strain, M20, was isolated from aquaculture sewage and identified as Brevundimonas sp. We characterized an ars cluster, arsHRNBC, with a novel gene arrangement in strain M20. Meanwhile, we identified a novel fusion protein, ArsRM, was found to connect arsenite methylation and methionine metabolism. Thus, our results reveal the mechanism of a novel pathway of arsenite resistance and a potential method by which methylation can detoxify arsenite.

Materials and methods

Isolation of the roxarsone-resistant strain Brevundimonas sp. M20.

Aquaculture sewage was sampled from the influx of a wastewater treatment facility in Shandong Province, China. The sewage samples were diluted and spread onto Luria–Bertani (LB)-agar plates (0.5% w/v yeast extract, 1% w/v tryptone, 1% w/v sodium chloride, 2% w/v agar) containing 30 µM roxarsone (filtered through a 0.22 µM micro-filtration membrane) (Sigma Co., Shanghai, China), which were then incubated at 28 °C for 24 h. All colonies with different phenotypes on the plate were selected and cultivated three to five consecutive times on LB-agar medium containing 30 µM roxarsone to obtain pure cultures of single colonies. One colony, named M20 (Table S1), was selected and grown in pure culture for further study.

Whole-genome sequencing and genomic analysis

The genome of strain M20 was sequenced using the Nanopore and BGISEQ-500 platform (BGI, Wuhan, China) and assembled using Unicycler software [22]. Additional genome annotation was performed using tools at the RASTtk server [23] and the Pathosystems Resource Integration Center (PATRIC) server [24]. Multisequence comparison was carried out using Clustal Omega [25] and ESPript [26]. Phylogenetic affiliation analysis of strain M20 was performed based on its genome sequence, and a whole-genome phylogenetic tree was constructed using the PATRIC server [24].

Bioinformatic analysis of arsenic resistance genes and proteins

The genetic contexts of the arsHRNBC cluster and the metRFHH operon were compared using BLASTn. Comparison of ArsRM and ArsM orthologs was conducted using Blastp, and multisequence comparisons were conducted using Clustal Omega and ESPript. Homology models of ArsRM were constructed using Discovery Studio 2.0 [27].

Expression and purification of fusion protein ArsRM

The arsRM gene was amplified by PCR using arsRM-His-F/R as primers (Table S2) and the genomic DNA of M20 as the template. The purified arsRM fragment was cloned into pMD18-T and verified by sequencing. NdeI and XhoI were used to digest the recombinant plasmid. The arsRM gene fragment was then inserted into digested pET-15b. The resulting plasmid, pET-arsRM, was transformed into Escherichia coli BL21 (DE3), to produce E. coli strain ARM3 (Table S1). When the E. coli ARM3 culture reached an OD600 nm of 0.6, 1.0 mM isopropyl-β-D-1-thiogalactopyranoside (IPTG) was added to induce the expression of the His-tagged ArsRM fusion protein. After incubation at 28°C for 4 h, the His-tagged ArsRM was purified using a Ni-NTA-Sefinose column (Sangon Biotech Co., Shanghai, China) and then analyzed by 10% SDS-PAGE as previously described [28].

Determination of minimum inhibitory concentrations

The M20 strain was cultured on LB-agar plates to determine the minimum inhibitory concentrations (MICs) of Rox(V), NaH2AsO3 [As(III)], and NaH2AsO4 [As(V)]. First, strain M20 was activated on an LB-agar plate and single colonies were individually transferred into LB medium and cultured overnight at 28 °C with shaking at 180 rpm. OD600 nm of the culture was adjusted to 0.1, and then it was diluted 100-fold with LB medium. Diluted culture was streaked on LB-agar plates containing 0.25–6 mM (0.25-mM increment) of filtered As(III), As(V), or Rox(V). The plates were cultured at 28 °C for 3 days. MICs toward E. coli ARM3 were tested in the presence and absence of IPTG (0.5mM). E. coli BL21 (DE3) harboring vector pET-15b in the presence of IPTG was used as the negative control.

Molecular simulation and electromobility shift assays (EMSAs)

The interactions between ArsRM and its substrates As(III) and SAM were analyzed using the CDOCKER protocol of Discovery Studio 2.0 [27]. Molecular docking between ArsRM and its target gene was analyzed by using the ZDOCKER protocol of Discovery Studio 2.0 with the arsRM promoter region as the ligand. The interaction between ArsRM and its gene target was verified by electromobility shift assays (EMSAs). The promoter region of the metRFHH operon was divided into 80- to 120-bp fragments, which were used to design three short oligonucleotide probes (Table S2). Biotin-labeled oligonucleotide probes were synthesized by Sangon. The EMSAs were conducted as previously described [28, 29].

Methyltransferase activity analysis of ArsRM

The ability of ArsRM to methylate As(III) was determined in vitro using a Methyltransferase Activity Assay Kit (Abcam, Cambridge, UK). Briefly, ArsRM, SAM, and arsenious acid were combined; the mixture was adjusted to 50 µL with MT Assay Buffer. Sample Reaction Mix (50 µL) was prepared and added. The absorbance was measured at 570 nm every 30 s for at least 45 min at 37 °C. One unit of methyltransferase activity was defined as the amount of enzyme that generated 1.0 µmol of S-adenosyl homocysteine per min at 37 °C.

Results and discussion

Isolation and identification of the arsenic-resistant strain M20

Strain M20 was isolated from aquaculture sewage under the pressure of 30 µM roxarsone, an organic compound of arsenic. MIC analysis revealed that strain M20 was also resistant to inorganic arsenic compounds, such as NaH2AsO3 and NaH2AsO4 (Table S3). On the basis of phylogenetic affiliation analysis, strain M20 was identified as Brevundimonas sp. (Fig. 1A). Brevundimonas sp. is opportunistic pathogens [30]. Brevundimonas sp. M20 harbors a 3.315-Mb chromosome (67.56% G + C mol%) (Fig. 1B). The genome annotation included 1,208 hypothetical proteins and 2,052 proteins with functional assignments. The proteins with functional assignments included 758 proteins with Enzyme Commission (EC) numbers, 648 with Gene Ontology (GO) assignments, and 567 proteins that were mapped to Kyoto Encyclopedia of Genes and Genomes pathways (Table S4). Subsystem analysis of PATRIC annotation indicted that strain M20 contains 97 stress response, defense, and virulence genes, and 53 membrane transport-related genes (Fig. 1C).

Fig. 1
figure 1

Identification and genome characteristics of Brevundimonas sp. strain M20. (A) Phylogenetic tree including strain M20 based on full genome sequences. GenBank accession no. CP041243. (B) PATRIC annotation of strain M20 genome. (C) Subsystem analysis of strain M20 genome

Identification of ars cluster and met operon

Genome annotation of Brevundimonas sp. M20 revealed a novel ars cluster with novel gene arrangement, consisting of the putative arsenic resistance genes arsH, arsR, arsN, arsB, and arsC (Fig. 2). In this 3,011-bp cluster, arsH is an organoarsenical oxidase-encoding gene, which was considered to confer resistance to roxarsone and methylarsenite [20]. arsR, encoding a regulatory protein, is responsible for regulating ars cluster gene expression. arsB and arsC are arsenic detoxification genes; arsenate is reduced to arsenite by arsenate reductase (ArsC), followed by efflux of arsenite by the arsenite transporter ArsB [7]. ArsN detoxifies arsenate by acetylation of the α-amino group of arsinothricin [10]. Gene mining was then operated by BLAST analysis. The novel ars cluster revealed high similarity with the ars cluster in Caulobacterales spp. (JAFLCT010000083) and Brevundimonas sp. Bin7 (JACVCC01000001), which were not reported in previous researches. To our knowledge, the ars cluster in B. nasdae strain Au-Bre29 (CP080034)is the only similar one reported so far [13], but it does not contain arsN (Fig. 2A).

No arsenic methylation gene was found in the ars cluster of Brevundimonas sp. M20. However, a 5649-bp met operon related to methionine biosynthesis is located downstream of the ars cluster. The met operon is located 21.5 kb downstream of the ars cluster and contains four genes, arsRM (encoding a transcriptional regulator fused with a methyltransferase), metF (encoding methylenetetrahydrofolate reductase), HMT-1 (encoding 5-methyltetrahydrofolate-homocysteine methyltransferase), and metH (encoding methionine synthase) (Fig. 2B). The DNA sequence of the arsR fragment in arsRM showed very low similarity to the arsR gene in the ars cluster, although the similarity of the amino acid sequence was 21.78%. The met operon showed high similarity with that in B. nasdae Au-Bre29, which was isolated in Fujian Province, China, in 2022 [13]. However, the gap between the ars cluster and the met operon in strains M20 and Au-Bre29 was 21.5 kb and 2.1 Mb, respectively. The function of ArsRM has not yet been explained.

Fig. 2
figure 2

ars cluster and met operon in Brevundimonas sp. M20. Physical maps were generated using Easyfig. (A) Linear comparison of ars clusters in Brevundimonas sp. M20, Caulobacterales spp., Brevundimonas sp. bin7, and B. nasdae Au-Bre29. (B) Linear comparison of met operons in Brevundimonas sp. M20 and B. nasdae Au-Bre29. Block arrows indicate gene length and orientation, and numbers in each box represent nucleotide sequence identity (percent). Distance between ars gene cluster and met operon is indicated

ArsR M increased the arsenite resistance of recombinant E. coli

No arsenic methylation gene was found in the ars cluster of Brevundimonas sp. M20, while ArsRM, encoded by arsRM of the met operon, was identified as a transcriptional regulator fused with a methyltransferase. To analyze the functions of ArsRM, the complete arsRM gene (960 bp) was cloned and inserted into pET-15b (Fig. 3A). The resulting plasmid was transferred into E. coli BL21 (DE3), and the resulting strain E. coli ARM3 was verified by PCR (Fig. 3B).

After induction by IPTG, the MIC of NaH2AsO3 for E. coli ARM3 was increased to 1.5 mM compared with that in the uninduced control (Table S3). This enhancement indicated that ArsRM likely contributes to arsenite resistance in strain M20. On the basis of similarities with various arsenite methyltransferases in different species (Figure S1), we speculated that the arsenite resistance mediated by ArsRM may be due to arsenite methylation.

Fig. 3
figure 3

Construction of Brevundimonas sp. M20 ArsRM expression strain Escherichia coli ARM3. (A) Expression plasmid construction; lane M1: DL 15,000 DNA Ladder (Solarbio, M1700), 1: pET-arsRM digested by NdeI and XhoI, 2: arsR M fragment amplified by PCR, 3: pET-15b digested by NdeI and XhoI. Full-length gel is presented in Supplementary Figure S2. (B) PCR verification of E. coli strain ARM3; lane M2: D 15,000 + 2000 DNA Ladder (TIANGEN, MD116), 1–4: positive transformants, 5: blank control. Full-length gel is presented in Supplementary Figure S3

ArsRM is a bifunctional fusion protein

To further investigate the function of ArsRM, its protein structure was homology modeled. The results indicated that ArsRM from Brevundimonas sp. M20 forms a dimer (Fig. 4A) and the two monomers adopt different orientations (Fig. 4B). Each monomer contains a transcriptional regulator domain (TRD; α1–α5 and β1–β2), a methyltransferase domain (MTD; α6–α9 and β3–β7), and an 11-amino acid-residue loop linker (residues E111–A121) (Fig. 4C).

Unlike the typical ArsR [Protein Data Bank Code (PDB): 1R1T], which contains five α-helixes and two antiparallel β-sheets [31, 32], the TRD of ArsRM is composed of six short α-helixes and two antiparallel β-sheets. The DNA-binding domain (DBD) in the TRD, which has a shorter diameter of approximately 37.2 Å, compared to that of 51 Å in ArsR (PDB: 3F6O), is composed of six helices (α1–α6) that form the core of the TRD and a C-terminal β-hairpin (β1–β2) [33]. TRD of ArsRM contains a longer loop between α5 and α4 compared with 3F6O, and an additional α6 (Fig. 4D). Compared with the tight domain in ArsR, these shortened α-helices in the DBD of ArsRM produce a flexible structure that may lead to a different regulatory mechanism.

The MTD of ArsRM has a mixed structure consisting of α-helices (α6–α9) and β-strands (β3–β7). This domain (residues 122–320) was compared with arsenite methyltransferases in bacteria (including Thermosediminibacter oceani, Streptomyces barringtoniae, Chloroflexi spp., Alteripontixanthobacter maritimus, and Hymenobacter roseosalivarius); Homo sapiens (residues 1–279); and the unicellular red alga Cyanidioschyzon sp. (complete protein). A glycine-rich sequence, “DLGTGSG,” is conserved as the hallmark of the SAM-binding motif [31] (Figure S1). This glycine-rich motif has a circular, open shape and is located among α1, α2, β3, β4, and β5 (Fig. 4E). The As(III)-binding site (ABS) has three modular components in ArsR (PDB: 1R1T) [31], while arsenite methyltransferases in different species are variable (Figure S1). The ABS of ArsRM, adjacent to the SAM-binding motif, is composed of residue C134 (equivalent to C72 in PDB: 1R1T) and residues in the region of Q224-L229 (Fig. 4F).

Fig. 4
figure 4

Structure of ArsRfrom Brevundimonassp. M20. (A) Homologous model of the ArsRM dimer; (B) Different orientations of the two monomers; (C) Structure of ArsRM monomer; (D) Structures of the DNA-binding domain (DBD) of ArsRM and ArsR. ArsRM and ArsR were presented by blue and gray cartoon respectively; (E) S-adenosylmethionine-binding motif; (F) As(III)-binding site residues

ArsRM binds to its own promoter region

Molecular docking analysis showed that the TRD of the fusion protein ArsRM from Brevundimonas sp. M20 can bind to its own gene promoter region (ParsRM). Similar to the DNA binding mechanism of ArsR [32], the dimer forms wing regions and helix α5 (equivalent to α4 in ArsR, PDB: 1GXP) interact with DNA in helix-turn-helix (HTH)–DNA complex structures (Fig. 5A). The DNA-binding sequence of ArsRM in the ParsRM region includes a region that contains the palindromic sequence “CTTTATATAAAG” located upstream of the initiation codon of arsRM (Fig. 5A). The interacting residues include R20 and E42, and, in helix α5, N96, A97, A98, D99, D100, L103, and E104 (Fig. 5B).

We tested three biotin-labeled probes of the arsRM promoter region (215-bp long) in EMSAs: ParsRM-1 (containing the predicted target sequence), ParsRM-2, and ParsRM-3 (Fig. 5C). Sufficient quantities of recombinant full-length ArsRM–His6 protein were produced in E. coli BL21 (DE3). The soluble protein was eluted from a Ni-NTA column in buffer containing 150 mM imidazole (Fig. 5D). Probe ParsRM-1, containing the predicted binding sequence of ArsRM to ParsRM, was shifted in EMSAs by the addition of purified ArsRM (Fig. 5E). This result confirmed the binding of ArsRM to its own promoter region, which might regulate the transcription of the methionine biosynthesis gene cluster in Brevundimonas sp. M20.

Fig. 5
figure 5

Verification of the regulatory function of Brevundimonas sp. ArsR M. (A) Interactions between the ArsRM DBD and its potential DNA target. (B) Residues that interact with DNA; red and black labels represent residues of monomer 1 and monomer 2, respectively. (C) Oligonucleotide probes from upstream of arsRM. (D) ArsRM expression in E. coli BL21 (DE3). Lanes 1–2: unpurified total protein; 3–4: eluent from Ni-NTA column. Full-length gel is presented in Supplementary Figure S4. (E) Electromobility shift assays of ArsRM and ParsRM. Lanes ck1–ck3: probes ParsRM-1, ParsRM-2, and ParsRM-3, respectively, without ArsRM; lanes P1–P3: probes ParsRM-1, ParsRM-2, ParsRM-3, respectively, with ArsRM. The positions of the free (f) and shifted (s) probes are indicated by black and red arrows, respectively. Full-length gel is presented in Supplementary Figure S5

ArsRM contributes to the methylation of as(III)

The process of arsenic methylation was originally proposed to be a detoxification mechanism, by which trivalent inorganic arsenic is biotransformed to a less toxic pentavalent methylated form. The MTD of ArsRM was predicted to interact with SAM and trivalent arsenic (Fig. 6A). Amino acid residues C134, H223, Q224, H227, F254, R263, H268, and P296 were identified as the SAM-binding residues (Fig. 6A). Unlike in the arsenite methyltransferases shown in Figure S1 where Cys interacts with As(III), in ArsRM, Q224 and Y228 are the As(III) interacting residues, at a distance of 2.3–2.4 Å (Fig. 6B). These distances are similar to those for residues (C174 and C224) in C. merolae ArsM (2.2–2.3 Å) [31].

The methyltransferase activity of ArsRM was measured using a Methyltransferase Activity Assay Kit. Activity was detected in the presence of a mixture of SAM, arsenious acid, and ArsRM (Fig. 6C). The results confirmed that arsenious acid can be methylated to monomethylarsonic acid by ArsRM with an activity level of 0.87U/mg.

Fig. 6
figure 6

ArsRMcontributes to the methylation of As(III). (A) Structural overview of ArsRM and interactions between ArsRM and its substrates As(III) and SAM. (B) Interactions of ArsRM-bound As(III). Bound arsenic is shown as purple spheres. (C) Methyltransferase activity of ArsRM. Control: reaction mixture with ArsRM only; no SAM: reaction mixture without SAM; no AS: reaction mixture without As(III); no ArsRM: reaction mixture without ArsRM; ArsRM: complete reaction mixture including ArsRM, SAM, and As(III).

Discussion

Arsenic is a metalloid found worldwide [4]. Natural and anthropogenic activities both lead to arsenic accumulation and its transfer into the biosphere [34]. The higher affinity and ability to form metallic compounds make As(III) more toxic than As(V) [2]. In the environment, inorganic arsenic, such as As(V), As(III) [2], are thought to be more toxic than organic arsenic, such as Rox(V)] and Rox(III) [7]. Exposure to arsenic causes diseases, including diabetes, peripheral vascular disease, and cancer [35, 36]. The toxicity, mobility, and fate of arsenic in the environment are influenced by many factors, including its speciation, changes in its speciation, and biogeochemical cycle [2]. In this study, we isolated an arsenic-resistant bacterial strain, Brevundimonas sp. M20, which is tolerant to organic and inorganic arsenicals. Genomic annotation indicated the presence of various arsenic-tolerance genes, including those encoding proteins involved in the valence transition of inorganic arsenic and the transformation of organic and inorganic arsenicals, as well as an arsenic efflux pump.

The ars cluster is a genetic system to cope with arsenic toxicity in microorganisms. Genes of the ars cluster occur in most prokaryotic genomes and are widely distributed in bacterial and archaeal species [37]. arsRDABC and arsRBC variants are the usual operons found in bacteria [16]. In the present study, we identified a novel ars cluster variant in the chromosome of Brevundimonas sp. M20; this ars cluster contains five genes with the arrangement arsH–arsR–arsN–arsB–arsC. Homologous genes of these in arsHRNBC cluster are known to participate in organoarsenical oxidation [20, 38, 39], cluster transcriptional repression [40], organoarsenical acetyl transfer [10], formation of arsenite efflux pump[3] and arsenate reduction[16]. Gene functional prediction analyses suggest that this novel arsHRNBC cluster contributes to As(III) resistance.

Fig. 7
figure 7

Mechanisms of arsenic resistance in Brevundimonas sp. M20. As(III): arsenite; As(V): arsenate; MAs(III): methylarsenite; MAs(V): methylarsenate; AcAs(V): acetylarsenate; AST: arsinothricin; SAM: S-adenosylmethionine

The methylation and demethylation of arsenic by microbes are important processes in the arsenic biogeochemical cycle [10], and arsenic methylation results in the natural occurrence of organoarsenic compounds [41]. For methylation, methionine is a universal biological cofactor that transfers its methyl group to substrates such as lipids, proteins, DNA, and other small molecules [10] in the presents of SAM, which is a conjugate of the nucleotide adenosine and amino acid methionine and an essential substrate/cofactor in numerous enzyme-catalyzed reactions, including transmethylation [42]. We found that Brevundimonas sp. M20 harbors a methionine biosynthesis gene cluster, the met operon, downstream of the ars cluster. The first gene, arsRM, encodes a transcriptional regulator fused with a methyltransferase. Molecular simulation and laboratory experiments confirmed the dual function of ArsRM. The TRD of ArsRM binds to the palindromic sequence “CTTTATATAAAG” in its own promoter region, ParsRM, to regulate expression of the methionine biosynthesis gene cluster, while the MTD of ArsRM has a SAM-dependent arsenic methylation function. ArsRM thus connects methionine biosynthesis and arsenic methylation to provide a precise and efficient pathway for arsenic resistance related to methionine metabolism. Moreover, the organification of As(III), mediated by ArsH along with ArsRM, broadens the microbial resistance spectrum of ars operons from inorganic to organic arsenic compounds.

The data presented here indicate a contribution to arsenic resistance mediated by the methyltransferase–transcriptional regulator fusion protein ArsRM in Brevundimonas sp. M20 (Fig. 7). In this pathway, inorganic arsenic and organic arsenic as As(V) and As(III) are mutually transformed. As(III) can be extruded by the pump ArsB or methylated by ArsRM, which also controls the biosynthesis of methionine, a component of SAM. During arsenic methylation, the methyl group of SAM is transferred to As(III) by ArsRM to form methylarsenite [MAs(III)]. MAs(III) is oxidized to MAs(V) by the organoarsenical oxidase ArsH. Elucidation of this novel arsenic-resistance pathway contributes to our understanding of microbial arsenic resistance.

Data Availability

The datasets supporting the conclusions of this article are available from the lead author (Bing Wang:345,807,969@qq.com) upon reasonable request.

References

  1. Yang HC, Rosen BP. New mechanisms of bacterial arsenic resistance. Biomed J. 2016;39(1):5–13. https://0-doi-org.brum.beds.ac.uk/10.1016/j.bj.2015.08.003.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Zhu YG, Yoshinaga M, Zhao FJ, Rosen BP. Earth abides arsenic biotransformations. Annu Rev Earth Planet Sci. 2014;42:443–67. https://0-doi-org.brum.beds.ac.uk/10.1146/annurev-earth-060313-054942.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Yang HC, Fu HL, Lin YF, Rosen BP. Pathways of arsenic uptake and efflux. Curr Top Membr. 2012;69:325–58. https://0-doi-org.brum.beds.ac.uk/10.1016/B978-0-12-394390-3.00012-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Abernathy CO, Thomasy DJ, Calderon RL. Health effects and risk assessment of arsenic. J Nutr. 2003;133(5 Suppl 1):1536S–8S.

    Article  CAS  PubMed  Google Scholar 

  5. Naujokas MF, Anderson B, Ahsan H, et al. The broad scope of health effects from chronic arsenic exposure: update on a worldwide public health problem. Environ Health Perspect. 2013;121(3):295–302. https://0-doi-org.brum.beds.ac.uk/10.1289/ehp.1205875.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Marapakala K, Packianathan C, Ajees AA, et al. A disulfide-bond cascade mechanism for arsenic(III) S-adenosylmethionine methyltransferase. Acta Crystallogr D Biol Crystallogr. 2015;71(Pt 3):505–15. https://0-doi-org.brum.beds.ac.uk/10.1107/S1399004714027552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Stolz JF, Basu P, Santini JM, Oremland RS. Arsenic and selenium in microbial metabolism. Annu Rev Microbiol. 2006;60. https://0-doi-org.brum.beds.ac.uk/10.1146/annurev.micro.60.080805.142053. :107 – 30.

  8. Fu W, Lu DL, Yao H, et al. Simultaneous roxarsone photocatalytic degradation and arsenic adsorption removal by TiO2/FeOOH hybrid. Environ Sci Pollut Res Int. 2020;27(15):18434–42. https://0-doi-org.brum.beds.ac.uk/10.1007/s11356-020-08310-5.

    Article  CAS  PubMed  Google Scholar 

  9. Zhang AY, Zhou Y, Liu X, Huang NH, Niu HH. Photochemical pollutant degradation on facet junction-engineered TiO(2) promoted by organic arsenical: governing roles of arsenic-terminated surface chemistry and bulk-free radical speciation. J Hazard Mater. 2020;390:122159. https://0-doi-org.brum.beds.ac.uk/10.1016/j.jhazmat.2020.122159.

    Article  CAS  PubMed  Google Scholar 

  10. Chen J, Rosen BP. The arsenic methylation cycle: how microbial communities adapted methylarsenicals for use as weapons in the continuing war for dominance. Front Environ Sci. 2020;8. https://0-doi-org.brum.beds.ac.uk/10.3389/fenvs.2020.00043.

  11. Chauhan NS, Ranjan R, Purohit HJ, Kalia VC, Sharma R. Identification of genes conferring arsenic resistance to Escherichia coli from an effluent treatment plant sludge metagenomic library. FEMS Microbiol Ecol. 2009;67(1):130–9. https://0-doi-org.brum.beds.ac.uk/10.1111/j.1574-6941.2008.00613.x.

    Article  CAS  PubMed  Google Scholar 

  12. Chen J, Zhang J, Rosen BP. Role of ArsEFG in roxarsone and nitarsone detoxification and resistance. Environ Sci Technol. 2019;53(11):6182–91. https://0-doi-org.brum.beds.ac.uk/10.1021/acs.est.9b01187.

    Article  CAS  PubMed  Google Scholar 

  13. Yang X, Li Y, Feng R, et al. Characterization of two highly arsenic-resistant Caulobacteraceae strains of Brevundimonas nasdae: discovery of a new arsenic resistance determinant. Int J Mol Sci. 2022;23(10). https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23105619.

  14. Giovannoni SJ, Halsey KH, Saw J, et al. A parasitic arsenic cycle that shuttles energy from phytoplankton to heterotrophic bacterioplankton. mBio. 2019;10(2). https://0-doi-org.brum.beds.ac.uk/10.1128/mBio.00246-19.

  15. Galvan AE, Paul NP, Chen J, et al. Identification of the biosynthetic gene cluster for the organoarsenical antibiotic arsinothricin. Microbiol Spectr. 2021;9(1):e0050221. https://0-doi-org.brum.beds.ac.uk/10.1128/Spectrum.00502-21.

    Article  PubMed  Google Scholar 

  16. Ben Fekih I, Zhang C, Li YP, et al. Distribution of arsenic resistance genes in Prokaryotes. Front Microbiol. 2018;9:2473. https://0-doi-org.brum.beds.ac.uk/10.3389/fmicb.2018.02473.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Busenlehner LS, Pennella MA, Giedroc DP. The SmtB/ArsR family of metalloregulatory transcriptional repressors: structural insights into prokaryotic metal resistance. FEMS Microbiol Rev. 2003;27(2–3):131–43. https://0-doi-org.brum.beds.ac.uk/10.1016/s0168-6445(03)00054-8.

    Article  CAS  PubMed  Google Scholar 

  18. Garbinski LD, Rosen BP, Chen J. Pathways of arsenic uptake and efflux. Environ Int. 2019;126:585–97. https://0-doi-org.brum.beds.ac.uk/10.1016/j.envint.2019.02.058.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mukhopadhyay R, Rosen BP. Arsenate reductases in prokaryotes and eukaryotes. Environ Health Perspect. 2002;110(Suppl 5):745–8. https://0-doi-org.brum.beds.ac.uk/10.1289/ehp.02110s5745.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chen J, Bhattacharjee H, Rosen BP. ArsH is an organoarsenical oxidase that confers resistance to trivalent forms of the herbicide monosodium methylarsenate and the poultry growth promoter roxarsone. Mol Microbiol. 2015;96(5):1042–52. https://0-doi-org.brum.beds.ac.uk/10.1111/mmi.12988.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Paez-Espino AD, Nikel PI, Chavarria M, de Lorenzo V. ArsH protects Pseudomonas putida from oxidative damage caused by exposure to arsenic. Environ Microbiol. 2020;22(6):2230–42. https://0-doi-org.brum.beds.ac.uk/10.1111/1462-2920.14991.

    Article  CAS  PubMed  Google Scholar 

  22. Wick RR, Judd LM, Gorrie CL, Holt KE, Unicycler. Resolving bacterial genome assemblies from short and long sequencing reads. PLoS Comput Biol. 2017;13(6):e1005595. https://0-doi-org.brum.beds.ac.uk/10.1371/journal.pcbi.1005595.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Brettin T, Davis JJ, Disz T, et al. RASTtk: a modular and extensible implementation of the RAST algorithm for building custom annotation pipelines and annotating batches of genomes. Sci Rep. 2015;5:8365. https://0-doi-org.brum.beds.ac.uk/10.1038/srep08365.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wattam AR, Davis JJ, Assaf R, et al. Improvements to PATRIC, the all-bacterial Bioinformatics Database and Analysis Resource Center. Nucleic Acids Res. 2017;45(D1):D535–d42. https://0-doi-org.brum.beds.ac.uk/10.1093/nar/gkw1017.

    Article  CAS  PubMed  Google Scholar 

  25. Madeira F, Park Ym, Buso JLN, et al. The EMBL-EBI search and sequence analysis tools APIs in 2019. Nucleic Acids Res. 2019;47:636–41. https://0-doi-org.brum.beds.ac.uk/10.1093/nar/gkz268.

    Article  CAS  Google Scholar 

  26. Robert X, Gouet P. Deciphering key features in protein structures with the new ENDscript server. Nucleic Acids Res. 2014;42(1):320–4. https://0-doi-org.brum.beds.ac.uk/10.1093/nar/gku316.

    Article  CAS  Google Scholar 

  27. BIOVIA. Discovery Studio modeling Environment, Release 2017. San Diego, CA: Dassault Systèmes; 2017.

    Google Scholar 

  28. Fu JF, Qin R, Zong G, et al. The CagRS two-component system regulates clavulanic acid metabolism via multiple pathways in Streptomyces clavuligerus F613-1. Front Microbiol. 2019;10:244. https://0-doi-org.brum.beds.ac.uk/10.3389/fmicb.2019.00244.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Zhang PP, Zhao Z, Li H, et al. Production of the antibiotic FR-008/candicidin in Streptomyces sp. FR-008 is co-regulated by two regulators, FscRI and FscRIV, from different transcription factor families. Microbiology. 2015;161(Pt 3):539–52. https://0-doi-org.brum.beds.ac.uk/10.1099/mic.0.000033.

    Article  CAS  PubMed  Google Scholar 

  30. Ryan MP, Pembroke JT. Brevundimonas spp: emerging global opportunistic pathogens. Virulence. 2018;9(1):480–93. https://0-doi-org.brum.beds.ac.uk/10.1080/21505594.2017.1419116.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Ajees AA, Marapakala K, Packianathan C, Sankaran B, Rosen BP. Structure of an as(III) S-adenosylmethionine methyltransferase: insights into the mechanism of arsenic biotransformation. Biochemistry. 2012;51(27):5476–85. https://0-doi-org.brum.beds.ac.uk/10.1021/bi3004632.

    Article  CAS  PubMed  Google Scholar 

  32. Gupta SS, Borin BN, Cover TL, Krezel AM. Structural analysis of the DNA-binding domain of the Helicobacter pylori response regulator ArsR. J Biol Chem. 2009;284(10):6536–45. https://0-doi-org.brum.beds.ac.uk/10.1074/jbc.M804592200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Prabaharan C, Kandavelu P, Packianathan C, Rosen BP, Thiyagarajan S. Structures of two ArsR as(III)-responsive transcriptional repressors: implications for the mechanism of derepression. J Struct Biol. 2019;207(2):209–17. https://0-doi-org.brum.beds.ac.uk/10.1016/j.jsb.2019.05.009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Brammer H, Ravenscroft P. Arsenic in groundwater: a threat to sustainable agriculture in South and South-east Asia. Environ Int. 2009;35(3):647–54. https://0-doi-org.brum.beds.ac.uk/10.1016/j.envint.2008.10.004.

    Article  CAS  PubMed  Google Scholar 

  35. Tseng CH, Tseng CP, Chiou HY, et al. Epidemiologic evidence of diabetogenic effect of arsenic. Toxicol Lett. 2002;133(1):69–76. https://0-doi-org.brum.beds.ac.uk/10.1016/s0378-4274(02)00085-1.

    Article  CAS  PubMed  Google Scholar 

  36. Chen CJ, Chen CW, Wu MM, Kuo TL. Cancer potential in liver, lung, bladder and kidney due to ingested inorganic arsenic in drinking water. Br J Cancer. 1992;66(5):888–92. https://0-doi-org.brum.beds.ac.uk/10.1038/bjc.1992.380.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Silver S. Phung le T. A bacterial view of the periodic table: genes and proteins for toxic inorganic ions. J Ind Microbiol Biotechnol. 2005;32(11–12):587–605. https://0-doi-org.brum.beds.ac.uk/10.1007/s10295-005-0019-6.

    Article  CAS  PubMed  Google Scholar 

  38. Falgenhauer L, Ghosh H, Guerra B, et al. Comparative genome analysis of IncHI2 VIM-1 carbapenemase-encoding plasmids of Escherichia coli and Salmonella enterica isolated from a livestock farm in Germany. Vet Microbiol. 2017;200:114–7. https://0-doi-org.brum.beds.ac.uk/10.1016/j.vetmic.2015.09.001.

    Article  CAS  PubMed  Google Scholar 

  39. Hao X, Li X, Pal C, et al. Bacterial resistance to arsenic protects against protist killing. Biometals. 2017;30(2):307–11. https://0-doi-org.brum.beds.ac.uk/10.1007/s10534-017-0003-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Busenlehner LS, Pennella MA, Giedroc DP. The SmtB/ArsR family of metalloregulatory transcriptional repressors: structural insights into prokaryotic metal resistance. FEMS Microbiol Rev. 2003;27(2–3):131–43. https://0-doi-org.brum.beds.ac.uk/10.1016/s0168-6445(03)00054-8.

    Article  CAS  PubMed  Google Scholar 

  41. Hoffmann T, Warmbold B, Smits SHJ, et al. Arsenobetaine: an ecophysiologically important organoarsenical confers cytoprotection against osmotic stress and growth temperature extremes. Environ Microbiol. 2018;20(1):305–23. https://0-doi-org.brum.beds.ac.uk/10.1111/1462-2920.13999.

    Article  CAS  PubMed  Google Scholar 

  42. Roje S. S-Adenosyl-L-methionine: beyond the universal methyl group donor. Phytochemistry. 2006;67(15):1686–98. https://0-doi-org.brum.beds.ac.uk/10.1016/j.phytochem.2006.04.019.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Liwen Bianji (Edanz) (www.liwenbianji.cn) for editing the language of a draft of this manuscript.

Funding

This work was supported by the Key Research and Development Project of Shandong Province (No. 2021ZDSYS27), the Academic Promotion Program of Shandong First Medical University [grant number LJ001, 2019], the Innovation Project of the Shandong Academy of Medical Sciences [grant number 2022], and the National College Students’ Innovation and Entrepreneurship Training Program [grant number 2029, 2022].

Author information

Authors and Affiliations

Authors

Contributions

CCL prepared the aquaculture sewage, co-led the data analysis and wrote the manuscript. GLZ co-led the data analysis and edited the manuscript. XC co-led the performance of bioinformatics analysis. MXT helped to analyze data. WHG co-led the data analysis and co-led the performance of bioinformatics analysis. JFF helped revised the manuscript. PPZ helped to analyze data and revised the manuscript. BW co-led the project and revised the manuscript. GXC co-led led the project and revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Bing Wang or Guangxiang Cao.

Ethics declarations

Nucleotide sequence accession number

The complete sequence of the Brevundimonas sp. M20 genome has been deposited in GenBank under the accession no. CP041243.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

12866_2023_2876_MOESM1_ESM.doc

Additional file 1. Table S1. Bacterial strains and plasmids used in this study. Table S2. Primers used in this study. Table S3. MICs of strains for organic and inorganic arsenics. Table S4. General features of the Brevundimonas sp. M20 genome. Figure S1. Multisequence comparison of the MTD domain of ArsRM from Brevundimonas sp. M20 with methyltransferase proteins from other species.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, C., Zong, G., Chen, X. et al. Bifunctional protein ArsRM contributes to arsenite methylation and resistance in Brevundimonas sp. M20. BMC Microbiol 23, 134 (2023). https://0-doi-org.brum.beds.ac.uk/10.1186/s12866-023-02876-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12866-023-02876-z

Keywords