SCS | SHR | SME | SMR | UMSPC | SMST | SSR | Main Page

 

The Biotransformation of Endobiotics by Sulfonation

 

Charles A. Strott, MD, Head, Section on Steroid Regulation

Young C. Lee, PhD, Staff Scientist

Hirotoshi Fuda, PhD, Research Fellow

Osamu Hanyu, MD, PhD, Postdoctoral Fellow

Yuko Higashi, MD, PhD, Postdoctoral Fellow

Atsushi Kojitani, DDS, PhD, Postdoctoral Fellow

Norman B. Javitt, MD, PhD, Adjunct Investigator

Hidekatsu Yanai, MD, PhD, Adjunct Investigator

 


We investigate the molecular mechanisms and biologic implications of modifying substances by sulfonation, a fundamental process in the biotransformation of endobiotics as well as of drugs and xenobiotics. The transfer of an SO3-1 group from the universal donor molecule 3´-phosphoadenosine 5´-phosphosulfate (PAPS) to an acceptor molecule is essential for normal growth and development and for maintenance of the internal milieu. Sulfonated macromolecules such as glycosaminoglycans and proteoglycans are involved in cell-surface and connective tissue structures and bone formation. Tyrosine sulfonation is a widespread post-translational modification of many secretory and membrane proteins. Glycoprotein hormones are sulfonated on specific saccharide moieties, creating unique structural motifs with important functional consequences. Sulfolipids are concentrated in the brain, peripheral nerves, and reproductive tissues. Sulfonation also modifies low molecular weight compounds, such as catecholamines, iodothyronines, neuroendocrine peptides, and cholesterol along with its metabolites, oxysterols, bile acids, vitamin D, and steroid hormones. By modulating the availability of biologically active hormones, sulfonation can influence biologic activity regardless of whether the compounds act in their unconjugated or sulfoconjugated state or whether they act via a genomic or nongenomic mechanism. Thus, sulfoconjugating enzymes, i.e., sulfotransferases, can play an essential role in specific physiologic systems and associated disorders.

The hydroxysteroid sulfotransferases SULT2A1, SULT2B1a, and SULT2B1b

Fuda, Lee, Yanai, Strott; in collaboration with Javitt, Pedersen

Often referred to as hydroxysteroid sulfotransferases, the enzymes that sulfoconjugate neutral steroids constitute the SULT2 family, which, in turn, comprises two subfamilies, SULT2A1 and SULT2B1. Whereas the SULT2A1 subfamily consists of a single form, the SULT2B1 subfamily consists of the two isoforms SULT2B1a and SULT2B1b, which derive from an alternative exon 1. SULT2A1 has a broad substrate predilection, acting on alpha- and beta-hydroxylated compounds, whereas the SULT2B1 isoforms have narrower substrate preferences that are limited to selective 3-beta-hydroxylated steroids. For example, SULT2B1a avidly sulfonates pregnenolone, whereas it less efficiently sulfoconjugates cholesterol; on the other hand, SULT2B1b functions as the physiologic cholesterol sulfotransferase. The fact that the SULT2A1 and SULT2B1 isozymes are differentially expressed and display dissimilar substrate propensities strongly suggests that they play distinct biologic roles. Importantly, the SULT2B1 isoforms are structurally unique when compared with SULT2A1 and all other known cognate cytosolic sulfotransferases. For instance, the SULT2B1 isoforms have extended amino- and carboxy-termini. Moreover, the carboxy termini are enriched in prolines as well as in glutamate, serine, and threonine, suggesting polypeptide sequences that target proteins for rapid degradation. The crystal structure of human SULT2B1b reveals a catalytic binding orientation for the substrate that differs from the previously determined structure for the SULT2A1 substrate-binding pocket. In addition, an amino terminal helix, comprising residues D19 to K26, determines the specificity difference between the SULT2B1 isoforms, which becomes ordered upon substrate binding covering the binding pocket. Mutational analysis suggests that the specificity difference for cholesterol and pregnenolone between the SULT2B1a and SULT2B1b isoforms can be traced to the unique amino-terminal residues 19-DISEI-23. Alanine scanning of this region reveals that only the I20A and I23A mutants are associated with a complete loss of cholesterol sulfonating activity. Furthermore, the activity can be partially restored by replacement with conservative substitutions such as leucine and methionine. I20 and I23 lie on the same side of the helix facing on the inside of the hydrophobic pocket, whereas residues S21 and E22 are exposed to solvent.

The structure of the SULT2B1 gene is highly conserved in that the human and mouse genes are essentially identical. Differential expression of mouse SULT2B1a, SULT2B1b, and SULT2A1, along with their distinct substrate preferences, has physiologic relevance. For instance, the exclusive expression of SULT2B1a in the central nervous system, and its tendency for greater activity with pregnenolone as a substrate, would be in keeping with the importance of pregnenolone sulfate as a neurosteroid involved in learning and memory processes. The sole expression of SULT2B1b in skin, and the fact that it clearly has a predilection for cholesterol as a substrate, is consistent with the importance of cholesterol sulfate as a regulatory molecule in keratinocyte differentiation and development of the epidermal barrier. On the other hand, the overwhelming expression of SULT2A1 in the liver is in accord with its role in general metabolism involving both xenobiotics and endobiotics. Importantly, the mouse SULT2B1 and SULT2A1 genes are differentially expressed during embryonic development, with the former gene expressed at all stages from E8.5 to E19 and the latter gene not expressed until E19. The results suggest that expression of the mouse SULT2B1 gene is more critical during early development than that of the SULT2A1 gene, especially the expression of SULT2B1a during the early stages of brain development and the expression of SULT2B1b during growth of epithelia.

Lee KA, Fuda H, Lee YC, Negishi M, Strott CA, Pedersen LC. Crystal structure of human cholesterol sulfotransferase (SULT2B1b) in the presence of pregnenolone and PAP: rationale for specificity differences between prototypical SULT2A1 and the SULT2B1 isoforms. J Biol Chem 2003;278:44593-44599.

Shimizu C, Fuda H, Yanai H, Strott CA. Conservation of the hydroxysteroid sulfotransferase SULT2B1 gene structure in the mouse: pre- and postnatal expression, kinetic analysis of isoforms, and comparison with prototypical SULT2A1. Endocrinology 2003;144:1186-1193.

Strott CA. Hydroxysteroid sulfotransferases SULT2A1, SULT2B1a and SULT2B1b. In: Coughtrie MWH, Pacifici GM, eds. Human Cytosolic Sulfotransferases. London: Taylor & Francis, 2004, in press.

Cholesterol sulfate

Higashi, Yanai, Fuda, Lee, Strott; in collaboration with Javitt, Kanzaki

Cholesterol sulfate is widely distributed in human plasma and is quantitatively more significant than other known sterol sulfates. While our knowledge of the physiologic role(s) of cholesterol sulfate is still evolving, the steroid has been implicated in several biologic systems. For example, it is a component of cell membranes, where it has a stabilizing effect, such as protecting erythrocytes from osmotic lysis. Cholesterol sulfate can regulate serine proteases such as those involved in blood clotting, fibrinolysis, and epidermal cell adhesion. Cholesterol sulfate has also been implicated in adrenal and gonadal steroid synthesis, in the regulation of cholesterol and fatty acid synthesis, and in controlling sperm capacitation. Given its ability to regulate the activity of selective protein kinase C isoforms and modulate the specificity of phosphatidylinositol 3-kinase, cholesterol sulfate is involved in signal transduction. In keratinocyte differentiation, cholesterol sulfate can induce genes that encode key components involved in development of the barrier. In addition, cholesterol sulfate can induce platelet aggregation. That cholesterol sulfate circulates in blood and demonstrates an emerging role as a regulatory molecule suggests a similarity to compounds categorized as hormones.

Quantitative expression of the gene encoding SULT2B1b is consistent with the involvement of cholesterol sulfate in keratinocyte development, i.e., the progressive expression of SULT2B1b mRNA in primary cultures of normal human epidermal keratinocytes (NHEK) during Ca2+-induced differentiation. The rise in SULT2B1b mRNA expression parallels increases in protein and enzymatic activity. SULT2B1b is the only SULT2 enzyme expressed in normal human skin as well as in cultured NHEK, which would be the expected finding if the sole purpose of SULT2 expression in skin were the production of cholesterol sulfate. Nonetheless, the question remains as to the principal physiologic role of cholesterol sulfate in the epidermis. The answer can be found, in part, by knowing where SULT2B1b is expressed during keratinocyte development. In this regard, immunocytochemical analysis has revealed that expression of SULT2B1b is confined to the granular layer of the epidermis, suggesting that it is a late marker of keratinocyte differentiation, i.e., the expression pattern is the same as that of the barrier protein filaggrin, an acknowledged late marker of differentiation. On the other hand, involucrin, another barrier protein, is clearly expressed by cells in the immediate suprabasal region or spinous layer of the epidermis, the earliest-appearing cells in the pathway toward keratinocyte differentiation.

The confinement of cholesterol sulfotransferase to the granular layer of the living epidermis beneath the stratum corneum, coupled with the fact that this region of the epidermis contains the highest content of cholesterol sulfate, strongly suggests that the principal function of this sulfolipid is carried out primarily in the region of the granular-stratum corneum junction. The epidermis is a perpetually renewing tissue whereby keratinocytes arise from stem cells in the basal layer and move through a series of cellular differentiation events until, as dead squames, they are finally sloughed off from the outer stratum corneum. Cholesterol sulfate could play a significant role in the proper functioning of the stratum corneum and the normal sloughing of dead cells, a process termed desquamation. Cholesterol sulfate can retard desquamation by inhibiting serine proteases in the stratum corneum that are responsible for the degradation of cell adhesion structures known as desmosomes. In addition, cholesterol sulfate can influence cell cohesiveness by affecting the stability of corneocyte lipid bilayers. We therefore hypothesized that the inhibitory influence of cholesterol sulfate on normal desquamation would subsequently be removed by the presence of a cholesterol sulfate sulfohydrolase in the outer stratum corneum, thus allowing normal desquamation to occur. Our hypothesis is supported by the finding of markedly elevated cholesterol sulfate in the stratum corneum of patients with recessive X-linked ichthyosis, a scaling disorder that is a form of abnormal desquamation resulting from a deficiency in steroid sulfatase activity. Interestingly, about 90 percent of the cholesterol sulfate formed during Ca2+ induction of keratinocyte differentiation is found in the cellular membrane fraction, with only 10 percent present in the soluble cell fraction. The membrane localization of cholesterol sulfate suggests that such localization is an important determinant of cholesterol sulfate’s physiologic effect.

Cholesterol sulfate supports platelet aggregation: it potentiates arachidonic acid–, ADP-, and thrombin-induced platelet aggregation as well as serotonin secretion, effects specific for cholesterol sulfate that require both the sterol ring structure and the sulfate moiety. The content of cholesterol sulfate in platelets is 566 ± 62 pmol/109 platelets. The presence of SUL2B1b confirmed that platelet cholesterol sulfate can be produced locally and not just taken up from the circulation. In fact, as in skin, platelet SULT2B1b is the only SULT2 enzyme expressed by these discoid anucleate particles. Although platelets lack a nucleus, they do contain rough endoplasmic reticulum and polysomes and are known to engage in protein synthesis. It is recognized that human platelets and plasma lipoproteins interact and are intimately involved in the pathogenesis of atherosclerosis, thrombosis, and coronary artery disease. Based on real-time PCR, the level of SULT2B1b mRNA in platelets is stably maintained at 4°C but markedly diminished over a period of 4 h at 37°C. The presence of HDL but not of LDL, however, significantly attenuates the loss of SULT2B1b mRNA. Furthermore, the stabilizing influence of HDL is attributable specifically to its apolipoprotein A-I component, whereas apolipoprotein A-II and apolipoprotein E are without effect. Importantly, we observed a direct correlation between platelet SULT2B1b mRNA and protein levels in the presence or absence of lipoprotein as reflected in enzymatic activity and cholesterol sulfate production.

Higashi Y, Fuda H, Yanai H, Lee Y, Fukushige T, Kanzaki T, Strott CA. Expression of cholesterol sulfotransferase (SULT2B1b) in human skin and primary cultures of human epidermal keratinocytes. J Invest Dermatol 2004;122:1207-1213.

Strott CA, Higashi Y. Cholesterol sulfate in human physiology: what’s it all about? J Lipid Res 2003;44:1268-1278.

Yanai H, Javitt NB, Higashi Y, Fuda H, Strott CA. Expression of cholesterol sulfotransferase (SULT2B1b) in human platelets. Circulation 2004;109:92-96.

Transcriptional regulation of cholesterol sulfotransferase (SULT2B1b)

Lee, Higashi, Fuda, Strott

Studies involving RT-PCR reveal that human SULT2B1a and SULT2B1b are differentially expressed. Real-time PCR demonstrates that expression of SULT2B1b is high in the prostate, placenta, and skin; weak in the kidney, lung, small intestine, and colon; and with little or no expression in other tissues. Information is lacking as to how expression of the two human SULT2B1 isoforms is regulated. Given the importance of cholesterol sulfate in the epidermis of human skin, we have focused on the expression of SUT2B1b by using primary cultures of normal human epidermal keratinocytes (NHEK) as well as immortalized but highly differentiated human keratinocytes (HaCaT) cells.

The gene encoding SULT2B1 contains neither a canonical TATAAA nor a CCAAT motif in the upstream region flanking exon 1B, and there is no initiator motif. An RLM-RACE investigation of the start of transcription yielded multiple transcription start sites (TSS) that tended to cluster in two areas. Some TATA-less promoters retain the ability to direct transcription initiation from a specific nucleotide, whereas others appear to direct transcription initiation from several sites, ranging from a few tightly clustered start sites to dozens of sites spanning hundreds of nucleotides. Although inconsistency with the RLM-RACE procedure might be attributable to the existence of multiple TSS, we cannot exclude the possibility of premature termination of RT as a consequence of the high GC content in the 5´-UTR. In fact, experiments employing RT-PCR placed the TSS upstream of the sites determined by RLM-RACE, suggesting that premature termination of RT might have occurred.

Many TATA-less promoters are characterized by the presence of multiple GC boxes, which bind to the Sp1 transcription activator and thus play a central role in the assembly of the transcription complex of these promoters. Regulation of the gene encoding SULT2B1 appears to be similarly under the influence, at least part, of the Sp1 family of transcription factors; that is, the area upstream of the coding region of SULT2B1b contains several GC/GT boxes, and mutational analyses suggest involvement of specific motifs in transcriptional regulation. In addition, deletion analyses correctly confirmed the mutational analyses. Importantly, nuclear extracts from HaCaT cells contain proteins that bind to probes incorporating Sp1 motifs implicated in gene regulation by the mutational and deletion analyses; furthermore, supershift analyses confirmed the presence of Sp1 and Sp2 proteins in the HaCaT cell nuclear extracts. Co-transfection experiments that used NHEK and HaCaT cells provided additional support for the involvement of Sp1 and Sp2 in transcriptional regulation.

While the case for Sp1 and Sp2 involvement in regulating expression of SULT2B1b is appealing, the picture is undoubtedly more complicated. Activation of a given promoter requires several transcription factors that bind cooperatively to their cognate sites or possibly act synergistically through other mechanisms. In this regard, a unique feature of Sp1 as a transcription factor is its synergistic activation and interaction with other transcription factors. Interestingly, in the case of the SULT2B21b promoter, we observed, in addition to Sp1 and Sp2, two unidentified proteins that specifically bind to probes containing the Sp1 motifs; we are currently trying to identify these proteins. Notably, Sp1 and related family members are not the only proteins to recognize GC/GT boxes. We have found several other zinc-finger proteins that have a binding specificity similar to Sp1.

Assuming that the location of the TSS for SULT2B1b is correct, then a major Sp1 regulatory element is located in the 5´ UTR. Reports, however, support transcriptional regulation by the 5´ UTR. The presence of transcriptional regulatory activity within exon 1 represents a positive internal promoter regulatory sequence. Interestingly, promoter activity within the 5´ UTR is usually attributable to the presence of functional Sp1 binding elements. Furthermore, CpG dinucleotides, common within the promoter and 5´ UTR of SULT2B1b, may be subject to hypermethylation, a process that can be associated with loss of expression. Importantly, Sp1 has been implicated in the protection of CpG islands from methylation. Most CpG islands encompass the promoter and first exon of most housekeeping genes and more than 50 percent of tissue-restricted genes. We are currently investigating the role of CpG methylation in transcriptional regulation of human SULT2B1b expression.

Isolation, identification, and characterization of human platelet cholesterol sulfate-binding protein

Yanai, Fuda, Higashi, Lee, Strott

Based on preliminary experiments demonstrating saturable and specific cholesterol sulfate-binding activity in human platelet membrane extracts, we initiated experiments to identify the binding factor. We used anion-exchange chromatography to fractionate platelet membrane extracts. Analysis of the fractions demonstrated a protein of about 55,000 MW with an N-terminal sequence EPAVYFKEQFLDG, which is identical to the N-terminus (minus the signal sequence) of calreticulin, a calcium-dependent chaperone protein first detected in the endoplasmic reticulum but now also known to be present in the plasma membrane. As a confirmatory experiment, we examined the effect of anticalreticulin antibodies on cholesterol sulfate binding to human platelets. The results supported the notion that calreticulin is capable of binding to cholesterol sulfate and is responsible for the binding of cholesterol sulfate to human platelets.

As human calreticulin was not initially available, we used the bovine ortholog (93 percent identical to human calreticulin) for binding analyses, competition studies, and the effect of calcium on the binding activity. In summary, cholesterol sulfate binding to bovine calreticulin is saturable with a Kd of 193 pmol/mg; furthermore, the binding activity is significantly enhanced by calcium at low concentration, whereas binding is inhibited at high calcium concentration. Competition studies reveal that binding of cholesterol sulfate to bovine calreticulin is highly specific, i.e., other steroid sulfates as well as cholesterol and other cholesterol derivatives do not compete effectively. With the recent acquisition of the cDNA for human calreticulin, we will now proceed to overexpress and purify the human protein for further characterization and cholesterol sulfate-binding analyses.

Characterization of unknown cytosolic sulfotransferases

Fuda, Hanyu, Javitt, Strott

Recently, we cloned two structurally unique cytosolic sulfotransferases for which substrates have not yet been identified. Nonetheless, it is interesting that one sulfotransferase (SULT4A1) appears to be exclusively expressed in the central nervous system, whereas expression of the other sulfotransferase (SULT6B1) appears to be restricted to the testes. Although it is possible that the proteins are products of pseudogenes and are thus inactive enzymes, the specific tissue expression patterns would suggest otherwise; that is, the fact that the proteins are expressed in selective tissues suggests that they have a specific function in those tissues. To explore such possibility, we cloned, overexpressed, and purified both SULT4A1 and SULT6B1 and have initiated a search to identify active substrates for them.

COLLABORATORS

Norman B. Javitt, MD, PhD, New York University School of Medicine, New York, NY

Tamotsu Kanzaki, MD, PhD, Faculty of Medicine, Kagoshima University, Kagoshima, Japan

Lars C. Pedersen, PhD, Laboratory of Structural Biology, NIEHS, Research Triangle Park, NC

For further information, contact chastro@mail.nih.gov