Você está na página 1de 18

Expressional control of the constitutive isoforms of nitric oxide synthase (NOS I and NOS III)

ULRICH FORSTERMANN1, JEAN-PAUL BOISSEL, AND HARTMUT KLEINERT Department of Pharmacology, Johannes Gutenberg University, D-55101 Mainz, Germany
ABSTRACT

Nitric oxide synthase (NOS) exists in three established isoforms. NOS I (NOS1, ncNOS) was originally discovered in neurons. This enzyme and splice variants thereof have since been found in many other cells and tissues. NOS II (NOS2, iNOS) was rst identied in murine macrophages, but can also be induced in many other cell types. NOS III (NOS3, ecNOS) is expressed mainly in endothelial cells. Whereas NOS II is a transcriptionally regulated enzyme, NOS I and NOS III are considered constitutively expressed proteins. However, evidence generated in recent years indicates that these two isoforms are also subject to expressional regulation. In view of the important biological functions of these isoforms, changes in their expression may have physiological and pathophysiological consequences. This review recapitulates compounds and conditions that modulate the expression of NOS I and NOS III, summarizes transcriptional and posttranscriptional effects that underlie these changes, andwhere knowndescribes the molecular mechanisms leading to changes in transcription, RNA stability, or translation of these enzymes.Forstermann, U., Boissel, J.-P., Kleinert, H. Expressional control of the constitutive isoforms of nitric oxide synthase (NOS I and NOS III). FASEB J. 12, 773790 (1998)

stitutively expressed and whose activity is regulated by Ca2/ and calmodulin. The prototypical enzyme is being found in endothelial cells. Whereas transcriptional regulation of NOS II has been established for about 10 years, no expressional regulation was originally known for the other two isoforms. More recent evidence suggests, however, that the expression of NOS I and NOS III can also be regulated under various conditions. NOS I Cellular expression of NOS I NOS I was rst characterized in, and puried from, rat and porcine cerebellum (13). Since then, NOS I was found to have a widespread distribution in specic neurons of the central and peripheral nervous systems. The list of NOS I-containing neurons continues to expand, and to date it is difcult to nd an organ that is not innervated by neurons saying NO (4). NOS I is likely to play an important role not only
1 Correspondence: Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, 55101 Mainz, Germany. E-mail ulrich.forstermann@uni-mainz.de 2 Abbreviations: AP-1, activator protein 1; ATF, activating transcription factor; BAEC, bovine aortic endothelial cells; cFOS, mammalian homologue to the FBJ murine osteosarcoma virus oncogene; CNS, central nervous system; CRE, cAMP-responsive element; ERE, estrogen-responsive element; ETS, human analogs to avian acute leukemia virus E26 oncogene; GATA, transcription factor GATA; GFP, green uorescent protein; GLGF motif, gly-leu-gly-phe motif found in diverse membrane-associated proteins; HIF-1, hypoxia-inducible factor-1; HUVEC, human umbilical vein endothelial cells; IFN-g, interferon-g; IL-1, interleukin-1; LPS, lipopolysaccharide; LTA, lipoteichoic acid; NF-1, nuclear factor 1; NF-kB, nuclear factor kB; NMDA, N-methyl-D-aspartate; nNOS, neuronal nitric oxide synthase; NO, nitric oxide; NOS, nitric oxide synthase; mNOS, muscle-specic isoform; Oct-2, octamer binding factor 2; PEA3, polyomavirus enhancer activator 3; PDZ domain, same as the gly-leu-gly-phe (GLGF) motif; POU, transcription factors containing protein sequences homologous to the mammalian Pit-1, Oct-1, Oct-2, and Caenorhabditis elegans Unc-86 gene products; oxLDL, oxidized low density lipoprotein; Pu box, enhancer DNA sequence of the lymphotropic papovavirus; Sp1, SV40 virus promoter specic transcription protein 1; TGF-b, transforming growth factor b; TNF-a, tumor necrosis factor a; UTR, untranslated region.

Key Words: cellular expression gene structure transcription factors transcriptional regulation RNA stability protein stability

CONSTITUTIVE AND INDUCIBLE ISOFORMS OF NITRIC OXIDE SYNTHASE IN MAMMALS, THREE ISOFORMS of nitric oxide synthase (NOS)2 have been identied. NOS I (NOS1, ncNOS) is a low-output NOS that is constitutively expressed and whose activity is regulated by Ca2/ and calmodulin. The prototypical enzyme is present in neurons. NOS II (NOS2, iNOS) is a high-output NOS whose expression is induced by cytokines (and other agents) and whose activity is largely or completely Ca2/ independent. The prototypical enzyme is expressed by activated murine macrophages. NOS III (NOS3, ecNOS) is also a low-output NOS that is con0892-6638/98/0012-0773/$02.25 FASEB

773

in physiologic neuronal functions such as neurotransmitter release, neural development, regeneration, synaptic plasticity, and regulation of gene expression, but also in a variety of neurological disorders in which excessive production of NO leads to neural injury (5). However, NOS I expression is not conned to neuronal cells. In various species, NOS I mRNA transcripts and/or protein have been detected in nonneuronal cell types. Table 1 gives an overview of the neuronal and nonneuronal expression of NOS I. The subcellular localization of NOS I protein varies greatly among the cell types studied. In neurons, both soluble and particulate protein is found. Depending on the individual study, the particulate enzyme represents between 30 and 60% of the total neuronal NOS I protein (3638). Differences reported may be explained by shifts of the protein from one fraction to the other during development (as demonstrated in rat brain; see ref 39), by species differences, and by the different assay methods used. In electron microscopy studies of kidney macula densa cells, the neuronal isoform has been seen associated mainly with small vesicles (28). In skeletal muscle, NOS I protein is mostly particulate (23, 40). The particulate localization of part of the NOS I protein is probably due to the PDZ/GLGF motif found in the NH2-terminal sequence of the NOS I protein. This
TABLE 1. NOS I expression in various tissues and cell types
Tissue Cell type

motif participates in proteinprotein interactions with several other membrane-associated proteins (41, 42). In neurons, synaptic association of NOS I is mediated by the binding of the PDZ/GLGF motif to the postsynaptic density protein PSD-95 (43) and/or to the related PSD-93 protein (44). N-methyl-D-aspartate (NMDA) receptors are also known to be associated with PSD-95 (45). Colocalization of neuronal NOS and the huntingtin-associated protein (HAP1) has also been reported (46). In fast-twitch skeletal muscle bers, the muscle-specic isoform (mNOS I) is attached to the sarcolemmadystrophin complex via the PDZ/GLGF motif and interacts mainly with a1-synthrophin (47). Recent studies demonstrated a selective loss of sarcolemmal NOS I in mdx mice lacking the endogenous dystrophin. A similar loss of membrane-associated NOS I was seen in Duchenne muscular dystrophy and Becker muscular dystrophy, diseases in which the dystrophin gene is mutated (4749). Expressional regulation of NOS I Increasing evidence indicates that NOS I expression can be dynamically regulated by various physiological or pathological conditions. NOS I mRNA up-regulation seems to represent a general response of neu-

Species

Method of detection

References

Brain (abundant in the accessory olfactory bulb, the molecular layer of the cerebellum, and the pedunculopontine tegmental nucleus of the brainstem Hypothalamus Pineal gland Spinal cord Innervation of various organs Skeletal muscle

Neurons

Mouse, rat, primates, human

Immunohistochemistry, in situ hybridization, Northern blot, Western blot

(611)

Mouse, rat Rat, sheep Rat, cat, sheep Mouse, rat, hamster, guinea pig, cat, human Rat, guinea pig, human Rat, human Rat Human Rat Mast cells Gonadotrophs and folliculostellate cells Neutrophils Human Rat Rat Human

Immunohistochemistry, Western blot Immunohistochemistry, Western blot Immunohistochemistry, activity Immunohistochemistry Immunohistochemistry, Northern blot, RNase protection assay, RT-PCR, Western blot Immunohistochemistry, RT-PCR Immunohistochemistry, RTPCR, RNase protection assay Northern blot, RT-PCR Northern blot, RT-PCR Immunohistochemistry Northern blot, immunohistochemistry Northern blot, Western blot Immunohistochemistry, RT-PCR

(12, 13) (14, 15) (1619) (20, 21) (2225)

Myocytes

Lung Kidney Testis Penile corpora cavernosa, urethra, prostate Skin Pituitary gland Adrenal gland

Epithelial cells Macula densa cells

(26, 27) (28, 29) (30) (31) (32) (33) (34) (35)

774

Vol. 12

July 1998

The FASEB Journal

FORSTERMANN ET AL.

ronal cells to stress induced by a large array of physical, chemical, and biological agents such as heat (50), electrical stimulation (51), light exposure (52, 53), colchicine (54), formalin (55), phenobarbital (56), and allergic substances (57). A similar response is observed in the rat paraventricilar nucleus and adrenal cortex during immobilization stress (58, 59) and after mechanical or pathological lesions, including spinal cord, axonal, or nerve injuries (6062), hypophysectomy (63), or arterial occlusion leading to local ischemia (64, 65). Cellular stress is known to trigger the expression of a number of genes resulting in cellular damage and apoptosis. The observed NOS I up-regulation may, in most cases, represent only one component of the normal cellular stress response. Enhanced NOS I expression is often associated with coinduction of transcription factors such as c-jun (60, 66) and c-fos (67, 68). A down-regulation of NOS I expression has been documented in guinea pig skeletal muscle and rat brain after in vivo treatment with bacterial lipopolysaccharide (LPS) (25, 69). Treatment of rats with LPS or interferon-g (IFN-g) also decreased the expression of NOS I in the brain, stomach, rectum, and spleen (70). However, one conicting report suggested a transient increase of NOS I mRNA level in the paraventricular nucleus of LPS-treated rats (71). Physical stimuli regulating NOS I expression Several in vivo studies in rat suggested a timedependent increase in NOS I mRNA after hypoxia(7274). This up-regulation may be due to two distinct mechanisms: a general cellular stress response or a direct activation of the NOS I gene transcription through binding of hypoxia-induced factors to specic cis-acting elements, as seen in erythropoietin and several other hypoxia-induced genes (75). Putative BACGTSSK binding sites for the hypoxiainducible factor-1 (HIF-1) can be detected along the NOS I genomic sequence, but the functionality of these motifs has not yet been determined. In neurons that constitute the osmoresponsive circuit of the rat hypothalamo-hypophysal system, expression of the NOS I gene was up-regulated by changes in plasma osmolarity occurring, for instance, during chronic salt loading (76) or water deprivation (77). In contrast, a low-salt diet resulted in an increase in NOS I mRNA in the rat kidney, with a coordinate up-regulation of renal expression of renin and angiotensinogen (78, 79). In addition, angiotensinogen gene knockout mice displayed high levels of NOS I in the macula densa of the kidney (80). Regulation of NOS I expression by neurotransmitters and hormones NOS I expression also appears to be regulated by changes in neuronal activity (81). In cerebellar granCONSTITUTIVE ISOFORMS OF NO SYNTHASE

ule cells, inhibition of the glutamatergic transmission drastically increased NOS I expression (82). In neurons of the central nervous system (CNS), NOS I is often colocalized with NMDA receptors (83, 84), which are known to effectively mediate Ca2/ inux. NOS I expression can also be triggered by sex hormones. It has been demonstrated that estradiol and pregnancy could induce NOS I expression in several tissues in the rat (8587). However, increase in NOS I mRNA was not apparent in the lamina terminalis and the hypothalamo-neurohypophysial system of the rat during pregnancy (88). In male rats, testosterone treatment has been described to stimulate expression of the neuronal isoform in the penis (89). Corticosterone treatment results in up-regulation of heme oxygenase-2 and a concomitant decrease of NOS I transcription in rat brain (90). Lithium and tacrine, a cholinesterase inhibitor used to treat symptoms of Alzheimers disease, increase the expression of NOS I synergistically in the hippocampus of the rat. This effect could be inhibited by corticosterone (91). Developmental regulation of NOS I expression Spatial and temporal NOS I expression patterns occurred during development of the nervous system (9294) and the lung (26, 95). These changes seem to correlate with differential susceptibility of the cells, at particular developmental stages, toward specic inductors such as nerve growth factor (96), estrogens (97), neurotransmitters, and neurotrophins (98). Ogura et al. (99) showed that the NOS I mRNA level was increased in a human neuroblastoma cell line after trans-retinoic acid-induced neuronal differentiation. In skeletal muscle, a developmental switch from NOS I to the alternative transcript mNOS I (see below) has been reported concomitant with myotube fusion (24). Moreover, an enhanced expression of NOS I is generally observed with postnatal development and aging (100, 101). Chromosomal localization, genomic structure, and cDNA of NOS I Kishimoto et al. (102) mapped the human NOS I gene to the q14-qter position of chromosome 12 using humanrodent hybrid cells and a human NOS I cDNA fragment in Southern blots. Fluorescence in situ hybridization studies allowed investigators to further dene the precise location of the gene to 12q24.2 (103) and to 12q24.224.3 (104). Homologous genes of mouse and rat have been assigned to chromosome 5 (105) and chromosome 12 (106), respectively. The genomic structure of NOS I is well documented for the enzyme from human brain (9, 107). Several mouse genomic clones have also been obtained for the targeted disruption of the neuronal
775

NOS, but no sequence data have been divulged (108). Sequence data of mouse rst exons are also available (109). A yeast articial chromosome clone containing part of the rat neuronal NOS I transcriptional cluster has recently been published (110). Brain-type NOS I cDNAs have been isolated from rat (111) and mouse (112). Recently, the rabbit NOS I cDNA sequence has been deposited in Genbank (accession number: U91584). For the coding sequence, the three mammalian sequences show 86 to 88% of identity with the human NOS I at the nucleotide level. The human NOS I gene is present as a single copy in the haploid human genome and reveals a highly complex structural organization (Fig. 1). The locus is scattered over a region greater than 200 kb. The nucleotide sequence corresponding to the major neuronal mRNA transcript is encoded by 29 exons. The full-length open reading frame codes for a protein of 1434 amino acids with a predicted molecular mass of 160.8 kDa, in good agreement with results obtained by protein purication (13). Comparison of the NOS I locus with the genomic organization of the two other NOS isoforms, NOS II (113) and NOS III (103, 114), demonstrates a striking similarity in the size of the exons and the location of the intronic

splice junctions, suggesting that the three isoforms derive from duplications of a common ancestor. Genotypic analyses of normal individuals demonstrated the existence of multiple alleles that vary in size (9, 10, 115). In particular, alleles of NOS I that differ in the number of CA/TG dinucleotide repeats in the 5 -anking region may show differences in basal promoter activity. CA repeats are known to favor formation of left-hand Z-DNA. Z-DNA sequences are often present in DNA regions critical for the regulation of transcription and replication (116). Diversity of NOS I transcripts Cloning and/or characterization of NOS I cDNAs from different human and rodent tissues have revealed structural and allelic mRNA diversity. Multiple broadly distributed, tissue-specic, or developmentally regulated mRNA transcripts have been reported. The different transcripts arise from three different mechanisms: 1) the initiation by different transcriptional units containing alternative promoters, 2) cassette exon deletions or insertions, and 3) the use of alternate polyadenylation signals. Combinations of two or three of these mechanisms account for the variety of NOS I transcripts described.

Figure 1. Genomic organization of the human neuronal nitric oxide synthase (NOS I) (adapted from refs 9, 10, 30). Exons are numbered and illustrated by black boxes. The nucleotide sequence corresponding to the major neuronal mRNA transcript is encoded by 29 exons. Many exons are of relatively small size, ranging from 59 to 266 bp. However, two large exons are present, exon 2 (1145 bp) and exon 29 (2150 bp), that include the translation initiation and termination sites, respectively. Two described transcriptional clusters (neuronal and testis specic) are schematized above and below the genomic map. Due to the presence of multiple promoters and transcription start sites, different rst exons (5 -exons) can be used. All rst exons of the neuronal cluster seem to splice to the common exon 2; therefore, one unique translation product corresponding to the full-length NOS I protein is likely to be assembled. Transcription from the testis-specic cluster, located between exon 3 and exon 4, gives rise to mRNA transcripts encoding for NH2-truncated enzymes. Possible cassette exon deletions of exon 10, or exons 9 and 10, are circled. Reported cassette exon insertions in the full-length NOS I are boxed: Tex 2 from the testis transcriptional cluster, and the mNOS I insert located between exons 16 and 17 of the human NOS I. Locations for repetitive elements are indicated; the human gene contains three sets of dinucleotide CA repeats and one (CG)n island. Members of the Alu repetitive sequences, indicated by triangles, are also found scattered at several locations along the human NOS I gene.
776 Vol. 12 July 1998 The FASEB Journal FORSTERMANN ET AL.

Transcriptional units and alternative promoters The usage of various alternative promoters seems to be one of the main characteristic of the NOS I gene (Fig. 2). To date, two major transcriptional clusters (neuronal and testis-specic) have been distinguished for the human gene. The neuronal cluster is located upstream from exon 2. At least eight different rst exons and the corresponding 5 -anking promoter sequences have been detected. They are distributed over a large region ( 200 kb) of genomic DNA. Major transcription sites were found located 25 kb upstream from exon 2 (9, 10, 22, 117, 118). The different rst exons appear to splice to the common exon 2 that contains the initiator ATG codon. Therefore, the protein encoded by the different mRNA species is the same and corresponds to the full-length, wild-type NOS I protein. Nevertheless, the use of alternative promoters is a versatile mechanism that can inuence gene expression by various means (119). The level of transcriptional initiation can be different: the processing, localization, translation efciency, and half-life of the mRNAs with various leader exons may differ. Alternative promoters could have distinct tissue specicities or developmental regulation and could respond differently to stimuli. Indeed, in situ hybridization with antisense cRNAs localized the different exon 1 variants to distinct cell populations, indicating that cell type-specic transcription/splicing factors may control NOS I expression (10). Multiple rst exons have also been reported for the mouse NOS I gene, and cDNA sequencing revealed at least ve distinct mRNA transcripts (109). Some transcripts are expressed in a tissue- or developmentally specic manner. A recent report demonstrated the existence of at least three transcripts of the rat NOS I gene that also arise from alternative splicing (110). A high degree of tissue and developmental specicity in expression is observed. One of these alternative transcripts is specically induced during neuronal differentiation of the rat PC12 cell line. Two closely linked human promoters and the corresponding rst exons (named 5 1 and 5 2 by the authors) have been particularly well studied (117, 118). Each has a distinct promoter activity and can independently drive the expression of a reporter gene in transfection studies. Exon 5 1 is embedded within a (CG)n island and its associated promoter lacks an identiable TATA box, resulting in the initiation of transcription at multiple sites within the exon. On the other hand, an initiator element located upstream from exon 5 2 leads to a limited number of transcription starts. Reverse transcription/ polymerase chain reaction demonstrated high levels of transcriptional expression for both exon 5 1 and exon 5 2 in the cerebellum and low expression in skeletal muscle. Yet a high expression level of a
CONSTITUTIVE ISOFORMS OF NO SYNTHASE

Figure 2. Model of alternative promoter usage. Initiation of transcription from different rst exons (E1a to E1c) driven by distinct promoters, associated with alternatively spliced exons, results in various mRNA transcripts with divergent 5 -UTRs. In the case of the neuronal NOS I transcriptional cluster, all the alternative exons (E1) appear to be nally spliced to the same exon 2 (E2) that contains the initiator codon. Therefore, only one protein is translated.

5 1/5 2 promoter complexluciferase fusion plasmid was obtained in transfected HeLa cells, suggesting that this region lacks the critical cis-acting element (or elements) responsible for CNS-restricted expression. The levels of NOS I mRNA and protein are greatly decreased in neuronal cell lines with reduced Oct-2 expression, a transcription factor member of the POU family. Cotransfection experiments with Oct-2 expression vectors and NOS I promoterreporter gene constructs resulted in specic activation of the 5 1 promoter (120). However, no canonical ATGCAAAT Oct-2 binding site can be detected in the sequence of the NOS I promoter used in that study (120). In normal human testis, transcription can initiate from a genomic region located within intron 3 (30) (see Fig. 1). A novel NOS I mRNA transcript, with a 5 terminus encoded for by two new exons (named Tex 1 and Tex 2) spliced to exon 4 (of the full-length NOS I), appears to be expressed in the testis at a level comparable to that of the full-length neuronal enzyme. A minor transcript containing a unique 95 bp exon Tex 1b 5 of exon Tex 2 was also detected. Analysis of the two mRNAs predicted the translation to occur at an ATG within exon 5, giving rise to a protein (TnNOS) of 1098 amino acids, with a calculated molecular mass of 125 kDa. Functional studies of the TnNOS stably transfected in CHO-K1 cells indicate that this protein is calcium dependent and exhibits a catalytic activity comparable to that of full-length NOS I. TnNOS lacks the NH2-terminal extension of the full-length NOS I, which contains the PDZ/GLGF proteinprotein interaction motif. In stably transfected CHO-K1 cells, the TnNOS and full-length
777

NOS I were distributed identically among cytosolic and particulate fractions. This suggests that either the PDZ/GLGF motif is not the only structure conveying membrane association of NOS I, or CHO-K1 cells lack the expression of some important partner proteins. The mouse neuronal NOS-g (nNOS-g) is an analog of the human TnNOS. Nevertheless, the molecular mechanism involved in its synthesis is not the same. In nNOS D/D mice with a targeted deletion of exon 2 (108), two alternative transcripts (b and g) are produced at very low levels. These use different rst exons (1a and 1b, respectively) spliced to a common exon 3 (43). Translation of nNOS-g is initiated at the same ATG codon within exon 5 as the human TnNOS, giving rise to a similar 125 kDa protein. Translation of nNOS-b is initiated at a CTG initiation codon within exon 1a, generating a 136 kDa NOS protein with six new NH2-terminal amino acids. A noticeable expression of nNOS-b transcript occurs in the striatum and cortex of nNOS D/D mice. The nNOS-b transcript has also been detected in many areas of the brain of wild-type mice (particularly in the ventral cochlear nuclei), where it produces a catalytically active protein (121). This protein, which lacks the PDZ/GLGF domain, has been found mainly in the cytosolic fraction (109). Recently, several human brain tumors were shown to express an alternatively spliced form of NOS I that comigrates with nNOS-b and also lacks exon 2 (109). Cassette exon deletions Cassette deletions of exon 10 and exons 9/10 were detected in human neuronal NO synthase mRNA transcripts (9, 10). The deletion of exon 10 introduces a stop codon 16 bp downstream of the splice junction, leading potentially to a novel truncated 560 amino acid protein. Excision of exons 9 and 10 results in a 315 bp in frame deletion, possibly leading to a protein lacking 105 amino acids. Low expression of this mRNA transcript was observed in various human and mouse brain tissues and in neuroblastoma cell lines (112, 122). It remains to be determined whether these cassette deletions are translated in vivo. Olgivie et al. (123) identied a subshortened inactive NOS I variant (termed NOS I144) that is expressed during synaptogenesis. However, even though the molecular weight was in agreement with an excision of exons 9 and 10, the structure of this deletion mutant has not been demonstrated at the nucleotide level. Using antisense probes targeting specically the full-length NOS I transcript or the truncated form lacking exons 9 and 10, Kolesnikov et al. (124) investigated the role of these two splice variants in morphine tolerance. Whereas antisense probes to the full-length NOS I prevented development of morphine tolerance, antisense probes
778 Vol. 12 July 1998

targeting the truncated form blocked morphine analgesia and shifted the morphine dose-response curve by more than twofold to the right (124). The NOS I-related dNOS cloned recently from Drosophila exhibits the same 315 bp deletion and is fully active (125). A small mRNA of 2.5 kb has also been detected in mouse adult testis (109). This transcript is thought to translate into a protein encoding only the reductase domain of NOS I. No further characterization of this protein has been reported. Cassette exon insertions A 102 bp insertion between exons 16 and 17 has been reported for the rat neuronal NOS gene, resulting in the translation of a fully active mNOS I (muscle NOS I) protein that contains a 34 amino acid addition (24, 31) (see Fig. 1). mNOS I is expressed in the rat skeletal muscle, heart, penis, urethra, and prostate (31). It coexists with the classical NOS I in rat pelvic plexus and bladder, and is detectable at low levels in the cerebellum. In culture, its expression coincides with myotube fusion. Most of the mNOS I in muscle is found complexed with dystrophin at the sarcolemma of intrafusal bers in muscle spindles (126). The 102 bp insert was also found in human penile RNA and its transcription from intron 16 was established (31). A cassette insertion of exon Tex 2 between exons 3 and 4 of the full-length NOS I was isolated when screening a human testis cDNA library and was subsequently found in a variety of other human tissues (30) (see Fig. 1). The insertion of the corresponding 56 bp leads to a frameshift and introduces a stop codon 371 bp downstream of the splice junction within exon 6. In vivo translation of the resulting COOHtruncated, 407 amino acid protein has not yet been demonstrated. Alternate polyadenylation signals The human NOS I exon 29 contains three potential polyadenylation signals. The site for cleavage and poly (A) addition uctuates over a 200 bp region, with a predominant site located 6449 bp downstream of the initiator methionine (9). Exon 29 also contains a (CA)n repeat sequence. Genotypic analysis of normal individuals demonstrated the existence of multiple alleles of various sizes, depending on the number of (CA)n repeats. The effect of these 3 -end variations on mRNA transcription, processing, stability, or subcellular targeting is currently unknown. Characterization of the 5 -anking sequences of NOS I A 1634 nucleotide sequence from the human neuronal transcriptional cluster has been obtained (9).
FORSTERMANN ET AL.

The FASEB Journal

TABLE 2. NOS III expression in various cell types


Cell type Species Method of detection Reference

Endothelial cells Neuronal cells Bone marrow cells, trabecular lining cells, osteoblasts, stromal cells, osteoclasts Dermal broblasts Epidermal keratinocytes Epithelial cells of endometrial glands Leydig cells and Sertoli cells, prematurely shed spermatocytes and spermatids Megakaroycytes NCI-H441 bronchiolar epihelial cells Placental syncytiotrophoblasts Platelets T-cells Thyroid follicular cells U937 monocytic cells Cardiac myocytes Diaphragm muscle cells Gastrointestinal mucosa Hepatocytes Smooth muscle cells Theca cell layer, ovarian stroma, mural granulosa cells on the surface of oocytes C6 glioma cells Resident astrocytes of the central nervous system Colon interstitial cells LLC-PK1 kidney tubular epithelial cells

Human/bovine Human/rat Human Human Human Human Human Human Human Human Human Human Human Human Rat Rat/murine Rat Rat Rat Rat Murine Murine Rabbit Porcine

Western blot, immunohistochemistry, NOS activity Immunohistochemistry Immunohistochemistry, RT-PCR Immunohistochemistry, RT-PCR Immunohistochemistry NADPH-diaphorase staining, Northern blot, RPA Immunohistochemistry Western blot, RT-PCR Western blot, RT-PCR NADPH-diaphorase staining, in situ hybridization Western blot, RT-PCR Immunocytochemistry, RT-PCR Immunohistochemistry, RT-PCR Immunocytochemistry, RT-PCR Immunohistochemistry, Northern blot Western blot Western blot, Northern blot Immunohistochemistry Western blot Western blot, immunohistochemistry Immunocytochemistry Immunohistochemistry Immunohistochemistry Western blot, RT-PCR

(127129) (130, 131) (132) (133) (32) (134) (135) (35) (136) (137) (35, 138) (139) (140) (141) (142) (143) (144) (145) (146) (147) (148) (148) (149) (150)

In its 3 part, this DNA fragment encompasses potential rst exons. This region contains a TATA box and two inverted CAAT boxes. Potential binding sites for transcription factors are found such as AP-2, transcriptional enhancer factor-1/M-CAT binding factor, CREB (cAMP-responsive element binding protein)/ ATF (activating transcription factor)/cFOS, Ets, NF-1 (nuclear factor 1), and nuclear factor kB (NF-kB) -like sequences. In the human testis transcriptional cluster, analyses 1800 bp upstream of the Tex 1 exon and 1705 bp upstream of the Tex 1b exon indicate that the two genomic regions lack a typical TATA box (30). A CAAT box is located 330 bp upstream from exon Tex 1b. Numerous potential cis-acting DNA elements have been detected: Sp-1, ATF/CRE-like sequences, NF-kB, activator protein 1 (AP-1), and AP-2. In addition, a variety of cis-regulatory elements implicated in testis-specic transcription are evident including transcription factor GATA and GATA-like sites, p53 half-element, an Ets binding site, Pu box, polyomavirus enhancer activator 3 (PEA3) sequences, myocyte-specic enhancer factor 2 motif, and an insulin response element site. However, no functional characterization of the neuronal or testis
CONSTITUTIVE ISOFORMS OF NO SYNTHASE

NOS I transcriptional regulatory regions has yet been published.

NOS III Cellular expression of NOS III NOS III was rst identied in endothelial cells (127, 128). Using a specic antibody to NOS III, immunohistochemical studies located the enzyme to various types of arterial and venous endothelial cells in many tissues, including human tissues (129). NOS III expression has also been demonstrated in several nonendothelial cell types, including neurons of the rat hippocampus and other rat brain regions, and in human motor neurons (130, 131) (Table 2). NOS III has been shown to be targeted to Golgi membranes and plasmalemmal caveolae, a complex process probably dependent on myristylation, palmitoylation, and tyrosine phosporylation of the enzyme as well as proteinprotein interactions with caveolins (151, 152). Experiments with NOS III green uorescent protein (GFP) chimeras revealed
779

that the rst 35 amino acids (and fatty acylation sites) of NOS III are sufcient to target GFP into the Golgi region of NIH 3T3 cells (153). NOS III has also been shown to interact with caveolins, a family of transmembrane proteins that form a key structural component of the caveolae (151, 152, 154). This interaction seems to negatively regulate NOS III activity (154). Mislocalization of NOS III caused by mutation of the N-myristoylation or cysteine palmitoylation sites reduces intracellular enzyme activity, suggesting that intracellular targeting of the NOS III is critical for endothelial NO production (155). Endothelium-derived NO is a physiologically signicant vasodilator and an inhibitor of platelet aggregation and adhesion. In addition, vascular NO can prevent leukocyte adhesion to the endothelium by down-regulating the leukocyte adhesion glycoprotein complex CD11/CD18. Finally, NO has also been shown to inhibit the proliferation of vascular smooth muscle cells (for a review, see refs 156, 157). Therefore, endothelial NO is likely to represent a protective anti-atherogenic principle, and up- or downregulation of endothelial NOS could have important consequences for vascular homeostasis. Several compounds and conditions implicated in vascular physiology and/or pathophysiology can modify the expression of NOS III. These compounds and conditions are discussed below. Expressional regulation of NOS III

of transcription and a destabilization of the NOS III mRNA (164). The down-regulation of NOS III may be implicated in pulmonary ventilation-perfusion coupling (with poorly ventilated areas of the lung being poorly perfused). In nonpulmonary endothelial cells, the ndings are more controversial. In BAEC, Arnet et al. (165) saw an up-regulation of NOS III mRNA and protein expression in cells incubated at low oxygen tension (1%). In this study, hypoxia did not change the stability of NOS III mRNA. On the other hand, the promoter activity of a 1.6 kb DNA fragment of the 5 -anking sequence of the human NOS III gene was enhanced by hypoxia (165). Therefore, enhanced NOS III expression in response to hypoxia is likely to result from enhanced promoter activity (165). However, the human NOS III promoter contains no homology to the published binding sequence of the hypoxia induced transcription factor HIF-1. Also, Zhang et al. (166) reported up-regulation of endothelial NOS III immunoreactivity in cerebral blood vessels during cerebral ischemia. Other reports, however, demonstrated reductions of NOS III expression in human umbilical vein endothelial cells (HUVEC) and bovine aortic endothelial cells exposed to low oxygen tension (167, 168) (Table 3). This reduction in NOS III expression was reported to result from decreased stability of NOS III mRNA and reduced NOS III promoter activity (167). Proliferation and growth status

Shear stress Exercise training and shear stress produced by the owing blood up-regulate NOS III expression (158 160) (Table 3). The enhancement of NOS III mRNA expression in bovine aortic endothelial cells (BAEC) by shear stress was not inhibited by dexamethasone, inhibitors of protein tyrosine kinases, or inhibition of G-protein signaling. In contrast, chelation of intracellular calcium in BAEC reduced shear stress induction of NOS III mRNA by almost 70% (160). A putative shear stress-responsive element (6 bp core sequence 5 -GAGACC-3 ) has been described in the promoter sequence of the human and bovine NOS III gene (see below) (103, 161). However, the functionality of this element has not been proven. Oxygen tension and hypoxia For pulmonary endothelial cells, there seems to be consensus that hypoxia down-regulates NOS III expression. In human primary pulmonary artery endothelial cells (162), cultured porcine pulmonary artery endothelial cells (163), and bovine pulmonary artery endothelial cells (164), hypoxia reduced NOS III mRNA and/or protein. At least in the bovine species, this was attributed to both a decreased rate
780 Vol. 12 July 1998

It has been shown that NOS III mRNA and protein is increased in growing vs. resting BAEC (169) (Table 3). This enhanced NOS III expression was found to be the result of a greater stability of the NOS III mRNA in proliferating compared with conuent BAEC (169). Growth-arrested BAEC showed enhanced expression of a protein that interacts with the 3 -UTR of the bovine NOS III mRNA and destabilized the mRNA (170). In contrast to these results, Flowers et al. (171) reported a reduction of NOS III mRNA and protein in asynchronously proliferating cultures and wounded endothelial cell monolayers compared with quiescent nonproliferating cells (Table 3). Because run-on experiments showed no changes in transcription rates, the reduction of NOS III mRNA in proliferating cells was attributed to NOS III mRNA destabilization (171). The reason for these discrepant ndings between the two groups is not clear at this time. Cytokines and bacterial LPS In BAEC and HUVEC, tumor necrosis factor-a (TNF-a) down-regulates NOS mRNA, protein, and activity (158, 172). The down-regulation of NOS III mRNA expression by TNF-a in HUVEC has been as FORSTERMANN ET AL.

The FASEB Journal

cribed to a destabilization of NOS III mRNA with no effect on transcription (173). In bovine endothelial cells, this destabilization seems to result from the specic interaction of a TNF-a-induced protein with the 3 -UTR (untranslated retion) sequence of the NOS III mRNA (174). In contrast, in resident astrocytes of the CNS, Barna et al. (148) described an enhancement of NOS III immunoreactivity when mice were treated intraperitoneally with IL-12, a potent activator of IFN-g and TNF-a production. These authors also found a cytokine-mediated activation of NOS III expression after exposure of C6 glioma cells to IL-12, IFN-g, and TNF-a (148). C6 glioma cells express NOS III constitutively (cf Table 2). Kaku et al. (175) described an up-regulation of NOS III mRNA expression and NOS III promoter activity from treatment of BAEC with INF-a/b and LPS. Similarly, Bucher et al. (176) found an up-regulation of NOS III mRNA expression in the liver of LPS- or lipoteichoic acid (LTA)-treated rats. They concluded that NOS III may be an even more important source of NO than NOS II in the liver after stimulation with LPS or LTA (176). In contrast, LPS injection into rats has been shown to reduce NOS III mRNA expression (along with NOS I mRNA expression) in aorta, heart, and lung (69). Another study demonstrated a downregulation of NOS III in gastrointestinal mucosa after LPS treatment of rats (144). Therefore, cytokines and LPS seem to regulate NOS III expression in different ways depending on the cytokine combination, species, and cell type analyzed. Estrogens and other sex steroids There are several reports that estrogens can up-regulate the expression of NOS III mRNA and protein. In guinea pigs, near-term pregnancy and treatment with estradiol (but not progesterone) increased calcium-dependent NOS activity in various tissues. Both pregnancy and estradiol enhanced NOS III mRNA (along with NOS I mRNA) in guinea pig skeletal muscle (177). An increase in NOS III mRNA has also been seen in the aortas of pregnant or estrogen-treated, but not progesterone- or testosterone-treated, rats (178). Kidneys from female rats contain more NOS III protein than those of male or oophorectomized female rats (179). Estrogen replacement therapy increased medullary NOS III levels in oophorectomized animals (179). On the other hand, in in vitro studies with ovine fetal pulmonary artery endothelial cells, estrogens enhanced NOS III activity, but no change in NOS III mRNA expression was detected (180). A study of bovine endothelial cells claimed that 17a-ethinyl estradiol did not enhance the expression of NOS III, but increased the release of bioactive NO by inhibiting superoxide anion production (181). Studies performed in our own laboratory (182) demonstrated
CONSTITUTIVE ISOFORMS OF NO SYNTHASE

that estrogens did enhance NOS III mRNA and protein expression in permanent human endothelial EA.hy 926 cells. The increased NOS III expression resulted from increased NOS III promoter activity with unchanged mRNA stability. In the absence of a bona de estrogen-responsive element in the human NOS III promoter (see below), increased NOS III promoter activity may result from an enhanced binding activity of transcription factor SV40 virus promoter specic transcription protein 1 (Sp1, which is essential for the human NOS III promoter; see below) (182). Growth factors Recent evidence indicates that an incubation of endothelial cells with certain growth factors may upregulate NOS III expression. Incubation or BAEC with transforming growth factor-b1 (TGF-b1) produced a modest up-regulation of NOS III mRNA and protein (183). This up-regulation of NOS III expression was reported to result from enhanced promoter activity (see below). Deletion of a putative NF-1 binding site in the bovine NOS III promoter abolished TGF-b1-enhanced promoter activity (183). Similarly, incubation of BAEC with basic broblast growth factor enhanced NOS III mRNA, protein, and activity (184). Oxidized low density lipoproteins and lysophosphatidylcholine The effects of oxidized low density lipoprotein (oxLDL) on NOS III expression are complex. In human saphenous vein endothelial cells, oxLDL (50 mg/ml) has been reported to reduce NOS III mRNA levels by decreasing the half-life of mRNA (185). The effect of oxLDL (50 mg/ml) on NOS III promoter activity was biphasic as measured in nuclear runon experiments. Treatment with oxLDL decreased NOS III promoter activity by about 25% in the rst 6 h, followed by a 1.8- and 2.2-fold increase at 12 and 24 h, respectively (185) (Table 3). Hirata et al. (186) found an up-regulation of NOS III mRNA in BAEC incubated with low concentrations of oxLDL (10 mg/ ml), whereas high concentrations (100 mg/ml) reduced mRNA levels after 24 h. Similar responses were seen with lysophosphatidylcholine, another component of atherogenic lipoproteins (186) (Table 3). In HUVEC, lysophosphatidylcholine up-regulated NOS III expression (187) (Table 3). Using nuclear run-on assays and reporter gene analyses, this enhanced NOS III expression was shown to result from lysophosphatidylcholine-stimulated NOS III promoter activity (188).
781

TABLE 3. Conditions or compounds that modify NOS III expression


Condition or compound Cell tissue Species Regulation Method of detection References

Shear stress Exercise training Hypoxiaa

Aortic endothelial cells Aortic endothelial cells, coronary arteries Aortic endothelial cells

Bovine Canine Bovine

Up-regulation Up-regulation Up-regulation

Northern blot Northern blot, nitrite assay Northern Blot, Western Blot, L-arginine-to-Lcitrulline conversion assay, reporter gene assay Northern blot, Western blot, L-arginine-to-Lcitrulline conversion assay RT-PCR, Ca2/-dependent NOS activity RNase protection assay Northern blot, RNase protection assay, Western blot, reporter gene assay Northern blot, NOS III promoter analysis, Western blot, NO bioassay RNase protection assay, Northern blot, Nuclear run-on analysis, Western blot, L-arginine-to-Lcitrulline conversion assay RNase protection assay, nuclear run-on analysis, Western blot Northern blot, RNase protection assay, Western blot Immunohistochemistry Immunohistochemistry, in situ hybridization Northern blot, Western blot Western blot, RT-PCR Immunohistochemistry, Western blot Northern blot, Western blot Immunohistochemistry, RT-PCR Western blot

(158, 160) (159) (165, 166)

Proliferationb

Aortic endothelial cells Aortic endothelial cells Liver Aorta, other tissues, EA.hy 926 endothelial cells Aortic endothelial cells Aortic endothelial cells, saphenous vein endothelial cells

Bovine

Up-regulation

(169)

Interferon-a/b and lipopolysaccharide Lipopolysaccharide, lipoteichoic acid Estrogensc

Bovine Rat Guinea pig, rat, human Bovine

Up-regulation Up-regulation Up-regulation

(175) (176) (177, 178, 182)

Transforming growth factor-b1, basic broblast growth factor Oxidized low density lipoprotein (low concentration)

Up-regulation

(183, 184)

Bovine, human

Up-regulation

(185, 186)

Lysophosphatidylcholine

Phorbolestersd Amyotrophic lateral sclerosis Atherosclerosis Increase in cGMP Glucose Chorion gonadotropin Angiotensin II Hyperthyroidism (Graves disease) Liver cirrhosis Hypoxia

Aortic endothelial cells, umbilical vein endothelial cells Aortic endothelial cells, EA.hy 926 endothelial cells Motor neurons Thoracic aorta Pulmonary endothelial cells Pancreatic islets, aortic endothelial cells Ovary Pulmonary artery endothelial cells Thyroid follicular cells and endothelial cells Aorta and mesenteric artery Pulmonary artery endothelial cells

Bovine, human Bovine, human Human Rabbit Bovine Rat, human Rat Bovine Human Rat Human, porcine, bovine

Up-regulation

(186, 187)

Up-regulation Up-regulation Up-regulation Up-regulation Up-regulation Up-regulation Up-regulation Up-regulation Up-regulation Downregulation

(189, 190) (131) (191) (192) (193, 194) (147) (195) (140) (196)

(162164) Northern blot, DNARNA dot blot, Western blot, NADPHdiaphorase cytochemistry, nitrite determination (continued on next page)
FORSTERMANN ET AL.

782

Vol. 12

July 1998

The FASEB Journal

TABLE 3. (continued )
Condition or compound Cell tissue Species Regulation Method of detection References

Hypoxiaa

Proliferationb Tumor necrosis factor-a

Umbilcal vein endothelial cells, aortic endothelial cells Aortic endothelial cells Umbilical vein endothelial cells, aortic endothelial cells Aortic endothelial cells, saphenous vein endothelial cells

Human, bovine Bovine Human, bovine

Downregulation Downregulation Downregulation

Northern blot, Western blot, NO bioassay Northern blot, Nuclear run-on analysis, Western blot Northern blot, Nuclear run-on analysis, analysis of the 3 -UTR of the bovine NOS III mRNA RNase protection assay, Northern blot, Nuclear run-on analysis, Western blot, L-arginine-to-Lcitrulline conversion assay Northern blot Immunohistochemistry, RT-PCR Immunohistochemistry Northern blot, in situ hybridization, Immunohistochemistry Immunohistochemistry Western blot, Northern blot

(167, 168)

(171) (158, 172174)

Oxidized low density lipoprotein (high concentration)

Bovine, human

Downregulation

(185, 186)

Lipopolysaccharide Hypothyreosis Hypertension Pulmonary hypertension Secondary bilary brosis Increase in cAMP

Aorta, heart, lung, gastrointestinal mucosa Thyroid follicular cells and endothelial cells Endothelial cells Lung Hepatocytes Cardiac myocytes

Rat Human Rat Human Rat Rat

Downregulation Downregulation Downregulation Downregulation Downregulation Downregulation

(69, 144) (140) (197) (198) (145) (199)

a Discrepant ndings in nonpulmonary endothelial cells; both up-regulation (165, 166) and down-regulation (167, 168) of NOS III have b c Discrepant ndings; both up-regulation (169) and down-regulation (171) of NOS III have been reported. The been reported. d Causing down-regulation (189) or stimulation (190) of protein NOS III up-regulation has not been seen in some other models (180, 181). kinase C (see text).

Modulation of protein kinase C activity Incubation of BAEC (189) or human EA.hy 926 endothelial cells (190) with phorbol esters enhanced NOS III expression. Ohara et al. (189) concluded from their results that down-regulation of PKC by long-term incubation with phorbol esters (or PKC inhibition with staurosporine) enhances NOS III expression. In contrast to these results, in our own experiments with human EA.hy 926 endothelial cells, the time course of phorbol ester-induced enhancement of NOS III expression paralleled PKC activation (190) (Table 3). Also, specic PKC inhibitors such as bisindolylmaleimide I, Go 6976, Ro-318220, and chelerythrine prevented the phorbol ester-induced enhancement of NOS III expression. Based on transfection experiments with a 3.5 kb human NOS III promoter fragment, the phorbol ester-stimulated enhancement of NOS III expression seems to be a transcriptional event (190).
CONSTITUTIVE ISOFORMS OF NO SYNTHASE

Additional conditions and compounds that have been described to up- or down-regulate NOS III expression are summarized in Table 3. Chromosomal localization, genomic structure, and cDNA of NOS III The human NOS III mRNA is encoded by 26 exons spanning 2122 kb of genomic DNA (103, 200). The gene is present as a single copy in the haploid human genome. The human NOS III gene has been assigned to the 7q357q36 region of chromosome 7 (103). The bovine NOS III gene spans approximately 20 kb of DNA and also contains 26 exons. Two transcription start sites have been determined that are located 170 and 240 base pairs upstream of the methionine translation initiation codon (161). Full-length NOS III cDNAs have been isolated from human (201, 202), murine (203), bovine (158, 172, 204), and porcine (205) endothelial cells. The de783

duced amino acid sequences predict proteins of 133 kDa for all species, which is in good agreement with the molecular mass determined by protein purication (128). The homology between the human coding cDNA sequence and those of the other species is around 90%; the identity of the deduced amino acid sequences is about 94%. Polymorphisms of the NOS III gene and cardiovascular disease Minisatellite sequences, tandem repeats, and dinucleotide (CA)n repeats have been identied in introns of the human NOS III gene (103, 114, 200). Some studies have attempted to link polymorphisms of these sequences with cardiovascular disease. Bonnardeaux et al. (206) reported that the highly polymorphic (CA)n repeats in intron 13 and two biallelic markers in intron 18 are not associated with essential hypertension. Another study explored the distribution of a 27 bp repeat in intron 4 of the human NOS III gene (207). They found a more common larger allele (with ve repeats) and a rarer smaller allele (with four repeats). The study demonstrated a signicant association between this polymorphism and coronary artery disease. Patients homozygous for the rare four-repeat allele were more likely to have one or more signicantly diseased vessels. The increased risk could be demonstrated only in current and former cigarette smokers; heterozygous individuals (smokers or nonsmokers) had no increased risk of coronary artery disease (207).

Characterization of the 5 -anking sequences of NOS III Characterization of the 5 -anking genomic region of human NOS III indicates that the endothelial NO synthase promoter is TATA-less and exhibits proximal promoter elements consistent with a constitutively expressed gene in endothelial cells such as Sp1 and GATA motifs (208). Furthermore, the human NOS III promoter contains consensus sequences for the binding of transcription factors AP-1, AP-2, NF-1, nuclear factor IL6, NF-kB, and PEA3, as well as CACCC-, CCAAT-, heavy metal-, acute-phase response-, shear stress-, cAMP-response-, retinoblastoma control-, INF-g-response-, and sterol-regulatory cis elements (Fig. 3). The promoter sequence also contains several half-sites of the estrogen-responsive element (ERE), but no bona de EREs are found (Fig. 3). A functional relevance has been demonstrated for only a few of these potential binding sites. Deletion and mutation analyses revealed an essential role of the Sp1 binding site at position 0103 (208). Stimulation of the human NOS III promoter by estrogens in human EA.hy 926 endothelial cells (182) and by lysophosphatidylcholine in HUVEC (188) may result from an enhanced binding activity of transcription factor Sp1. Mutation of the consensus GATA site at position 0230 reduced human NOS III promoter activity by about 30% (208). Mutation of the PEA3 binding site at position 026 reduced promoter activity by about 50% (188). Arnet et al. (165) demonstrated an enhanced activity of the human NOS III promoter in transfected bovine endothelial cells exposed to hypoxia. However, the human NOS III promoter contains no homology to an HIF-1 consensus sequence. None of the other consensus sequences de-

Figure 3. Comparison of the transcription factor binding sites in the bovine and human NOS III promoter (adapted from refs 200, 208). Putative binding sites for transcription factors AP-1 (1), GATA (g or G), NF-1 (n or N), NF-kB (k), PEA3 (p or P), and Sp1 (s or S), and of shear stress-induced transcription factors (blood ow, f) are shown. The location of half-sites for the estrogen receptor binding element (e) are also shown. Capital letters indicate binding sites whose function has been demonstrated in transfection (deletion or mutation) or gelshift experiments. 5 UTR, 5 -untranslated region of the mRNA; cds, protein coding sequence of the mRNA.

784

Vol. 12

July 1998

The FASEB Journal

FORSTERMANN ET AL.

scribed above are known to mediate induction of promoter activity by hypoxia. Phorbol ester incubation of human endothelial cells transfected with a human NOS III promoter-luciferase construct produced an enhancement of promoter activity (190). However, the transcription factors involved are not yet known. Similar to the 5 -anking sequence of the the human NOS III gene, the known 2.9 kb 5 -anking sequence of the bovine gene lacks a typical TATA box and contains numerous putative transcription factor binding sites (Fig. 3). Comparison of the rst 1.6 kb 5 -anking sequence showed 75% nucleotide identity with the corresponding human sequence (161). In reporter gene assays, a bovine promoter fragment (positions 01548 to /240) showed signicant basal activity in transfected BAEC. A deletion fragment (positions 01548 to / 192) lacking two putative Sp1 binding sites in the 5 -UTR of the NOS III cDNA lost almost all of its promoter activity, suggesting that transcription factor Sp1 is important for NOS III gene transcription in the bovine species as well. The modest up-regulation of NOS III mRNA by TGF-b1 in BAEC is likely to be the result of enhanced activity of the bovine NOS III promoter based on increased binding of transcription factor NF-1 to its respective response element (183). CONCLUSION

Research in the authors laboratory pertaining to this topic was supported by Grant Fo 144/32 and SFB 553, Project A1 (to U.F.) from the DeutscheForschungsgemeinschaft.

REFERENCES
1. Mayer , B., John, M., and Bohme, E. (1990) Purication of a calcium/calmodulin-dependent nitric oxide synthase from porcine cerebellum. Cofactor role of tetrahydrobiopterin. FEBS Lett. 277, 215219 Schmidt, H. H. H. W., Pollock, J. S., Nakane, M., Gorsky, L. D., Forstermann, U., and Murad, F. (1991) Purication of a soluble isoform of guanylyl cyclase-activating-factor synthase. Proc. Natl. Acad. Sci. USA 88, 365369 Bredt, D. S., and Snyder, S. H. (1990) Isolation of nitric oxide synthetase, a calmodulin-requiring enzyme. Proc. Natl. Acad. Sci. USA 87, 682685 Vincent, S. R., and Hope, B. T. (1992) Neurons that say NO. Trends Neurosci. 15, 108113 Yun, H. Y., Dawson, V. L., and Dawson, T. M. (1997) Nitric oxide in health and disease of the nervous system. Mol. Psychiatry 2, 300310 Bredt, D. S., Glatt, C. E., Hwang, P. M., Fotuhi, M., Dawson, T. M., and Snyder, S. H. (1991) Nitric oxide synthase protein and mRNA are discretely localized in neuronal populations of the mammalian CNS together with NADPH diaphorase. Neuron 7, 615624 Sugaya, K., and McKinney, M. (1994) Nitric oxide synthase gene expression in cholinergic neurons in the rat brain examined by combined immunocytochemistry and in situ hybridization histochemistry. Mol. Brain Res. 23, 111125 Giuili, G., Luzi, A., Poyard, M., and Guellaen, G. (1994) Expression of mouse brain soluble guanylyl cyclase and NO synthase during ontogeny. Dev. Brain Res. 81, 269283 Hall, A. V., Antoniou, H., Wang, Y., Cheung, A. H., Arbus, A. M., Olson, S. L., Lu, W. C., Kau, C. L., and Marsden, P. A. (1994) Structural organization of the human neuronal nitric oxide synthase gene (NOS1). J. Biol. Chem. 269, 33082 33090 Wang, Y., and Marsden, P. A. (1995) Nitric oxide synthases: gene structure and regulation. Adv. Pharmacol. 34, 7190 Chao, D. S., Hwang, P. M., Huang, F., and Bredt, D. S. (1996) Localization of neuronal nitric oxide synthase. Methods Enzymol. 268, 488496 Reuss, S., Decker, K., Rosseler, L., Layes, E., Schollmayer, A., and Spessert, R. (1995) Nitric oxide synthase in the hypothalamic suprachiasmatic nucleus of rat: evidence from histochemistry, immunohistochemistry and Western blot; and colocalization with VIP. Brain Res. 695, 257262 Wang, H., and Morris, J. F. (1996) Presence of neuronal nitric oxide synthase in the suprachiasmatic nuclei of mouse and rat. Neuroscience 74, 10591068 Layes, E., Schollmayer, A., Spessert, R., and Vollrath, L. (1996) Expression of brain nitric oxide synthase in the developing rat pineal gland coincides with the appearance of the adrenergic cyclic guanosine 3 ,5 -monophosphate response. Neurosci. Lett. 212, 7173 Lopez-Figueroa, M. O., Ravault, J. P., Cozzi, B., and Moller, M. (1996) Presence of nitric oxide synthase in the sheep pineal gland: an experimental immunohistochemical study. Neuroendocrinology 63, 384392 Dun, N. L., Dun, S. L., Forstermann, U., and Tseng, L. F. (1992) Nitric oxide synthase immunoreactivity in rat spinal cord. Neurosci. Lett. 147, 217220 Vizzard, M. A., Erdman, S. L., Roppolo, J. R., Forstermann, U., and de Groat, W. C. (1994) Differential localization of neuronal nitric oxide synthase immunoreactivity and NADPH-diaphorase activity in the cat spinal cord. Cell Tissue Res. 278, 299309 Vizzard, M. A., Erdman, S. L., Forstermann, U., and de Groat, W. C. (1994) Ontogeny of nitric oxide synthase in the lumbosacral spinal cord of the neonatal rat. Dev. Brain Res. 81, 201 217 Xu, Z., Li, P., Tong, C., Figueroa, J., Tobin, J. R., and Eisenach, J. C. (1996) Location and characteristics of nitric oxide synthase

2.

3. 4. 5. 6.

7.

8. 9.

Having realized that a pluripotent molecule such as NO is produced by many different cells and probably acts on an equally large number of target cells, it comes as little surprise to learn that nature has also invented a large array of regulatory mechanisms for NO production. The original paradigm that NO is either synthesized by constitutive NO synthases (NOS I and NOS III) or by the inducible NOS II is being challenged by an increasing number of reports that demonstrate expressional regulation for the constitutive enzymes (and constitutive expression of NOS II). Thus, not only the high-output NOS II is turned on transcriptionally to produce NO as a nonspecic weapon of antimicrobial defense, but expressional levels of the servoregulatory, low-output enzymes NOS I and NOS III can also be adjusted to meet local demand. Toward this goal, cells can modify the rate of transcription of these genes, the stability of the transcripts, and probably also their translation. In addition, at least NOS I is subject to cell-specic assembly of alternative exons and expressional control by different promoters. This can lead to NOS Iproteins with different cellular localizations, different catalytic activities, and perhaps different functions. Many of the regulations discussed above make physiological sense: some are still controversial; the molecular mechanism of most is still poorly understood. Together they add to the puzzling diversity of mechanisms controlling NO production.

10. 11. 12.

13. 14.

15.

16. 17.

18.

19.

CONSTITUTIVE ISOFORMS OF NO SYNTHASE

785

20. 21.

22.

23.

24. 25.

26.

27.

28.

29.

30.

31.

32.

33.

34. 35.

36.

37.

in sheep spinal cord and its interaction with alpha(2)-adrenergic and cholinergic antinociception. Anesthesiology 84, 890899 Ekblad, E., Alm, P., and Sundler, F. (1994) Distribution, origin and projections of nitric oxide synthase-containing neurons in gut and pancreas. Neuroscience 63, 233248 Esteban, F. J., Pedrosa, J. A., Jimenez, A., Fernandez, A. P., Bentura, M. L., Martinez-Murillo, R., Rodrigo, J., and Peinado, M. A. (1997) Distribution of neuronal nitric oxide synthase in the rat liver. Neurosci. Lett. 226, 99102 Nakane, M., Schmidt, H. H., Pollock, J. S., Forstermann, U., and Murad, F. (1993) Cloned human brain nitric oxide synthase is highly expressed in skeletal muscle. FEBS Lett. 316, 175 180 Gath, I., Closs, E. I., Godtel-Armbrust, U., Schmitt, S., Nakane, M., Wessler, I., and Forstermann, U. (1996) Inducible NO syn thase II and neuronal NO synthase I are constitutively expressed in different structures of guinea pig skeletal muscle: implications for contractile function. FASEB J. 10, 16141620 Silvagno, F., Xia, H., and Bredt, D. S. (1996) Neuronal nitricoxide synthase-m, an alternatively spliced isoform expressed in differentiated skeletal muscle. J. Biol. Chem. 271, 1120411208 Gath, I., Godtel-Armbrust, U., and Forstermann, U. (1997) Ex pressional downregulation of neuronal-type NO synthase I in guinea pig skeletal muscle in response to bacterial lipopolysaccharide. FEBS Lett. 410, 319323 North, A. J., Star, R. A., Brannon, T. S., Ujiie, K., Wells, L. B., Lowenstein, C. J., Snyder, S. H., and Shaul, P. W. (1994) Nitric oxide synthase type I and type III gene expression are developmentally regulated in rat lung. Am. J. Physiol. 266, L635L641 Asano, K., Chee, C. B., Gaston, B., Lilly, C. M., Gerard, C., Drazen, J. M., and Stamler, J. S. (1994) Constitutive and inducible nitric oxide synthase gene expression, regulation, and activity in human lung epithelial cells. Proc. Natl. Acad. Sci. USA 91, 1008910093 Tojo, A., Gross, S. S., Zhang, L., Tisher, C. C., Schmidt, H. H., Wilcox, C. S., and Madsen, K. M. (1994) Immunocytochemical localization of distinct isoforms of nitric oxide synthase in the juxtaglomerular apparatus of normal rat kidney. J. Am. Soc. Nephrol. 4, 14381447 Schricker, K., Potzl, B., Hamann, M., and Kurtz, A. (1996) Coordinate changes of renin and brain-type nitric-oxide-synthase (b-NOS) mRNA levels in rat kidneys. Puegers Arch. 432, 394 400 Wang, Y., Goligorsky, M. S., Lin, M., Wilcox, J. N., and Marsden, P. A. (1997) A novel, testis-specic mRNA transcript encoding an NH2-terminal truncated nitric-oxide synthase. J. Biol. Chem. 272, 1139211401 Magee, T., Fuentes, A. M., Garban, H., Rajavashisth, T., Marquez, D., Rodriguez, J. A., Rajfer, J., and Gonzalez Cadavid, N. F. (1996) Cloning of a novel neuronal nitric oxide synthase expressed in penis and lower urinary tract. Biochem. Biophys. Res. Commun. 226, 145151 Shimizu, Y., Sakai, M., Umemura, Y., and Ueda, H. (1997) Immunohistochemical localization of nitric oxide synthase in normal human skin: expression of endothelial-type and inducible-type nitric oxide synthase in keratinocytes. J. Dermatol. 24, 8087 Ceccatelli, S., Hulting, A. L., Zhang, X., Gustafsson, L., Villar, M., and Hokfelt, T. (1993) Nitric oxide synthase in the rat anterior pituitary gland and the role of nitric oxide in regulation of luteinizing hormone secretion. Proc. Natl. Acad. Sci. USA 90, 1129211296 Iwai, N., Hanai, K., Tooyama, I., Kitamura, Y., and Kinoshita, M. (1995) Regulation of neuronal nitric oxide synthase in rat adrenal medulla. Hypertension 25, 431436 Wallerath, T., Gath, I., Aulitzky, W. E., Pollock, J. S., Kleinert, H., and Forstermann, U. (1997) Identication of the NO syn thase isoforms expressed in human neutrophil granulocytes, megakaryocytes and platelets. Thromb. Haemost. 77, 163167 Arbones, M. L., Ribera, J., Agullo, L., Baltrons, M. A., Casanovas, A., Riveros Moreno, V., and Garcia, A. (1996) Characteristics of nitric oxide synthase type I of rat cerebellar astrocytes. Glia 18, 224232 Hecker, M., Mu lsch, A., and Busse, R. (1994) Subcellular localization and characterization of neuronal nitric oxide synthase. J. Neurochem. 62, 15241529

38.

39. 40.

41. 42. 43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

55.

Rodrigo, J., Riveros Moreno, V., Bentura, M. L., Uttenthal, L. O., Higgs, E. A., Fernandez, A. P., Polak, J. M., Moncada, S., and Martinez Murillo, R. (1997) Subcellular localization of nitric oxide synthase in the cerebral ventricular system, subfornical organ, area postrema, and blood vessels of the rat brain. J. Comp. Neurol. 378, 522534 Matsumoto, T., Pollock, J. S., Nakane, M., and Forstermann, U. (1993) Developmental changes of cytosolic and particulate nitric oxide synthase in rat brain. Dev. Brain Res. 73, 199203 Chang, W. J., Iannaccone, S. T., Lau, K. S., Masters, B. S., McCabe, T. J., McMillan, K., Padre, R. C., Spencer, M. J., Tidball, J. G., and Stull, J. T. (1996) Neuronal nitric oxide synthase and dystrophin-decient muscular dystrophy. Proc. Natl. Acad. Sci. USA 93, 91429147 Ponting, C. P., Phillips, C., Davies, K. E., and Blake, D. J. (1997) PDZ domains: targeting signalling molecules to sub membranous sites. Bioessays 19, 469479 Schepens, J., Cuppen, E., Wieringa, B., and Hendriks, W. (1997) The neuronal nitric oxide synthase PDZ motif binds to -G(D,E)XV* carboxyterminal sequences. FEBS Lett. 409, 5356 Brenman, J. E., Chao, D. S., Gee, S. H., McGee, A. W., Craven, S. E., Santillano, D. R., Wu, Z., Huang, F., Xia, H., Peters, M. F., Froehner, S. C., and Bredt, D. S. (1996) Interaction of nitric oxide synthase with the postsynaptic density protein PSD95 and alpha1-syntrophin mediated by PDZ domains. Cell 84, 757767 Brenman, J. E., Christopherson, K. S., Craven, S. E., McGee, A. W., and Bredt, D. S. (1996) Cloning and characterization of postsynaptic density 93, a nitric oxide synthase interacting protein. J. Neurosci. 16, 74077415 Kornau, H.-C., Schenker, L. T., Kennedy, M. B., and Seeburg, P. H. (1995) Domain interaction between NMDA receptor subunits and the postsynaptic density protein PSD-95. Science 269, 17371740 Li, X. J., Sharp, A. H., Li, S. H., Dawson, T. M., Snyder, S. H., and Ross, C. A. (1996) Huntingtin-associated protein (HAP1): discrete neuronal localizations in the brain resemble those of neuronal nitric oxide synthase. Proc. Natl. Acad. Sci. USA 93, 48394844 Brenman, J. E., Chao, D. S., Xia, H., Aldape, K., and Bredt, D. S. (1995) Nitric oxide synthase complexed with dystrophin and absent from skeletal muscle sarcolemma in Duchenne muscular dystrophy. Cell 82, 743752 Chao, D. S., Gorospe, J. R., Brenman, J. E., Rafael, J. A., Peters, M. F., Froehner, S. C., Hoffman, E. P., Chamberlain, J. S., and Bredt, D. S. (1996) Selective loss of sarcolemmal nitric oxide synthase in Becker muscular dystrophy. J. Exp. Med. 184, 609 618 Grozdanovic, Z., Gosztonyi, G., and Gossrau, R. (1996) Nitric oxide synthase I (NOS-I) is decient in the sarcolemma of striated muscle bers in patients with Duchenne muscular dystrophy, suggesting an association with dystrophin. Acta Histochem. 98, 6169 Sharma, H. S., Westman, J., Alm, P., Sjoquist, P. O., Cervos Navarro, J., and Nyberg, F. (1997) Involvement of nitric oxide in the pathophysiology of acute heat stress in the rat. Inuence of a new antioxidant compound H-290/51. Ann. N.Y. Acad. Sci. 813, 581590 Reiser, P. J., Kline, W. O., and Vaghy, P. L. (1997) Induction of neuronal type nitric oxide synthase in skeletal muscle by chronic electrical stimulation in vivo. J. Appl. Physiol. 82, 1250 1255 Goldstein, J., LopezCosta, J. J., and Saavedra, J. P. (1997) Changes in NADPH diaphorase reactivity and neuronal nitric oxide synthase in the rat retina following constant illumination. Neurosci. Lett. 231, 4548 Spessert, R., Layes, E., Schollmayer, A., Reuss, S., and Vollrath, L. (1995) In the rat pineal gland, but not in the suprachiasmatic nucleus, the amount of constitutive neuronal nitric oxide synthase is regulated by environmental lighting conditions. Biochem. Biophys. Res. Commun. 212, 7076 Lumme, A., Vanhatalo, S., Sadeniemi, M., and Soinila, S. (1997) Expression of nitric oxide synthase in hypothalamic nuclei following axonal injury or colchicine treatment. Exp. Neurol. 144, 248257 Lam, H. H., Hanley, D. F., Trapp, B. D., Saito, S., Raja, S., Dawson, T. M., and Yamaguchi, H. (1996) Induction of spinal cord

786

Vol. 12

July 1998

The FASEB Journal

FORSTERMANN ET AL.

56. 57.

58. 59.

60.

61. 62.

63.

64.

65. 66. 67. 68.

69.

70.

71.

72.

73.

neuronal nitric oxide synthase (NOS) after formalin injection in the rat hind paw. Neurosci. Lett. 210, 201204 Thompson, C. M., Bernhard, A. E., and Strobel, H. W. (1997) Barbiturate-induced expression of neuronal nitric oxide synthase in the rat cerebellum. Brain Res. 754, 142146 Calza, L., Giardino, L., Pozza, M., Micera, A., and Aloe, L. (1997) Time-course changes of nerve growth factor, corticotropin-releasing hormone, and nitric oxide synthase isoforms and their possible role in the development of inammatory response in experimental allergic encephalomyelitis. Proc. Natl. Acad. Sci. USA 94, 33683373 Calza, L., Giardino, L., and Ceccatelli, S. (1993) NOS mRNA in the paraventricular nucleus of young and old rats after immobilization stress. Neuroreport 4, 627630 Tsuchiya, T., Kishimoto, J., and Nakayama, Y. (1996) Marked increases in neuronal nitric oxide synthase (nNOS) mRNA and NADPH-diaphorase histostaining in adrenal cortex after immobilization stress in rats. Psychoneuroendocrinology 21, 287293 Herdegen, T., Brecht, S., Mayer, B., Leah, J., Kummer, W., Bravo, R., and Zimmermann, M. (1993) Long-lasting expression of JUN and KROX transcription factors and nitric oxide synthase in intrinsic neurons of the rat brain following axotomy. J. Neurosci. 13, 41304145 Vizzard, M. A. (1997) Increased expression of neuronal nitric oxide synthase in bladder afferent and spinal neurons following spinal cord injury. Dev. Neurosci. 19, 232246 Lin, L. H., Sandra, A., Boutelle, S., and Talman, W. T. (1997) Up-regulation of nitric oxide synthase and its mRNA in vagal motor nuclei following axotomy in rat. Neurosci. Lett. 221, 97 100 Villar, M. J., Ceccatelli, S., Bedecs, K., Bartfai, T., Bredt, D., Synder, S. H., and Hokfelt, T. (1994) Upregulation of nitric oxide synthase and galanin message-associated peptide in hypothalamic magnocellular neurons after hypophysectomy. Immunohistochemical and in situ hybridization studies. Brain Res. 650, 219228 Zhang, Z. G., Chopp, M., Gautam, S., Zaloga, C., Zhang, R. L., Schmidt, H. H., Pollock, J. S., and Forstermann, U. (1994) Up regulation of neuronal nitric oxide synthase and mRNA, and selective sparing of nitric oxide synthase-containing neurons after focal cerebral ischemia in rat. Brain Res. 654, 8595 Samdani, A. F., Dawson, T. M., and Dawson, V. L. (1997) Nitric oxide synthase in models of focal ischemia. Stroke 28, 1283 1288 Wu, W., Li, Y., Schinco, and F. P. (1994) Expression of c-jun and neuronal nitric oxide synthase in rat spinal motoneurons following axonal injury. Neurosci. Lett. 179, 157161 Torres, G., and Rivier, C. (1994) Induction of c-fos in rat brain by acute cocaine and fenuramine exposure: a comparison study. Brain Res. 647, 19 Hatakeyama, S., Kawai, Y., Ueyama, T., and Senba, E. (1996) Nitric oxide synthase-containing magnocellular neurons of the rat hypothalamus synthesize oxytocin and vasopressin and express Fos following stress stimuli. J. Chem. Neuroanat. 11, 243 256 Liu, S. F., Adcock, I. M., Old, R. W., Barnes, P. J., and Evans, T. W. (1996) Differential regulation of the constitutive and inducible nitric oxide synthase mRNA by lipopolysaccharide treatment in vivo in the rat. Crit. Care Med. 24, 12191225 Bandyopadhyay, A., Chakder, S., and Rattan, S. (1997) Regulation of inducible and neuronal nitric oxide synthase gene expression by interferon-gamma and VIP. Am. J. Physiol. 272, C1790C1797 Lee, S., Barbanel, G., and Rivier, C. (1995) Systemic endotoxin increases steady-state gene expression of hypothalamic nitric oxide synthase: comparison with corticotropin-releasing factor and vasopressin gene transcripts. Brain Res. 705, 136148 Shaul, P. W., North, A. J., Brannon, T. S., Ujiie, K., Wells, L. B., Nisen, P. A., Lowenstein, C. J., Snyder, S. H., and Star, R. A. (1995) Prolonged in vivo hypoxia enhances nitric oxide synthase type I and type III gene expression in adult rat lung. Am. J. Respir. Cell. Mol. Biol. 13, 167174 Prabhakar, N. R., Rao, S., Premkumar, D., Pieramici, S. F., Kumar, G. K., and Kalaria, R. K. (1996) Regulation of neuronal nitric oxide synthase gene expression by hypoxia. Role of nitric oxide in respiratory adaptation to low pO2. Adv. Exp. Med. Biol. 410, 345348

74. Guo, Y., Ward, M. E., Beasjours, S., Mori, M., and Hussain, S. N. A. (1997) Regulation of cerebellar nitric oxide production in response to prolonged in vivo hypoxia. J. Neurosci. Res. 49, 8997 75. Kvietikova, I., Wenger, R. H., Marti, H. H., and Gassmann, M. (1995) The transcription factors ATF-1 and CREB-1 bind constitutively to the hypoxia-inducible factor-1 (HIF-1) DNA recognition site. Nucleic Acids Res. 23, 45424550 76. Kadowaki, K., Kishimoto, J., Leng, G., and Emson, P. C. (1994) Up-regulation of nitric oxide synthase (NOS) gene expression together with NOS activity in the rat hypothalamo-hypophysial system after chronic salt loading: evidence of a neuromodulatory role of nitric oxide in arginine vasopressin and oxytocin secretion. Endocrinology 134, 10111017 77. OShea, R. D., and Gundlach, A. L. (1996) Food or water deprivation modulate nitric oxide synthase (NOS) activity and gene expression in rat hypothalamic neurones: correlation with neurosecretory activity? J. Neuroendocrinol. 8, 417425 78. Tojo, A., Madsen, K. M., and Wilcox, C. S. (1995) Expression of immunoreactive nitric oxide synthase isoforms in rat kidney. Effects of dietary salt and losartan. Jpn. Heart J. 36, 389398 79. Singh, I., Grams, M., Wang, W. H., Yang, T., Killen, P., Smart, A., Schnermann, J., and Briggs, J. P. (1996) Coordinate regulation of renal expression of nitric oxide synthase, renin, and angiotensinogen mRNA by dietary salt. Am. J. Physiol. 270, F1027F1037 80. Kihara, M., Umemura, S., Kadota, T., Yabana, M., Tamura, K., Nyuui, N., Ogawa, N., Murakami, K., Fukamizu, A., and Ishii, M. (1997) The neuronal isoform of constitutive nitric oxide synthase is up-regulated in the macula densa of angiotensinogen gene-knockout mice. Lab. Invest. 76, 285294 81. Tascedda, F., Molteni, R., Racagni, G., and Riva, M. A. (1996) Acute and chronic changes in K(/)-induced depolarization alter NMDA and nNOS gene expression in cultured cerebellar granule cells. Mol. Brain Res. 40, 171174 82. Baader, S. L., and Schilling, K. (1996) Glutamate receptors mediate dynamic regulation of nitric oxide synthase expression in cerebellar granule cells. J. Neurosci. 16, 14401449 83. Bhat, G. K., Mahesh, V. B., Lamar, C. A., Ping, L., Aguan, K., and Brann, D. W. (1995) Histochemical localization of nitric oxide neurons in the hypothalamus: association with gonadotropin-releasing hormone neurons and co-localization with Nmethyl-D-aspartate receptors. Neuroendocrinology 62, 187197 84. Aoki, C., Rhee, J., Lubin, M., and Dawson, T. M. (1997) NMDAR1 subunit of the cerebral cortex co-localizes with neuronal nitric oxide synthase at pre- and postsynaptic sites and in spines. Brain Res. 750, 2540 85. Ceccatelli, S., Grandison, L., Scott, R. E., Pfaff, D. W., and Kow, L. M. (1996) Estradiol regulation of nitric oxide synthase mRNAs in rat hypothalamus. Neuroendocrinology 64, 357363 86. Weiner, C. P., Knowles, R. G., and Moncada, S. (1994) Induction of nitric oxide synthases early in pregnancy. Am. J. Obstet. Gynecol. 171, 838843 87. Xu, D. L., Martin, P. Y., St. John, J., Tsai, P., Summer, S. N., Ohara, M., Kim, J. K., and Schrier, R. W. (1996) Upregulation of endothelial and neuronal constitutive nitric oxide synthase in pregnant rats. Am. J. Physiol. 271, R1739R1745 88. Luckman, S. M., Huckett, L., Bicknell, R. J., Voisin, D. L., and Herbison, A. E. (1997) Up-regulation of nitric oxide synthase messenger RNA in an integrated forebrain circuit involved in oxytocin secretion. Neuroscience 77, 3748 89. Reilly, C. M., Zamorano, P., Stopper, V. S., and Mills, T. M. (1997) Androgenic regulation of NO availability in rat penile erection. J. Androl. 18, 110115 90. Weber, C. M., Eke, B. C., and Maines, M. D. (1994) Corticosterone regulates heme oxygenase-2 and NO synthase transcription and protein expression in rat brain. J. Neurochem. 63, 953962 91. Bagetta, G., Corasaniti, M. T., Melino, G., Paoletti, A. M., Finazzi Agro, A., and Nistico, G. (1993) Lithium and tacrine increase the expression of nitric oxide synthase mRNA in the hippocampus of rat. Biochem. Biophys. Res. Commun. 197, 11321139 92. Bredt, D. S., and Snyder, S. H. (1994) Transient nitric oxide synthase neurons in embryonic cerebral cortical plate, sensory ganglia, and olfactory epithelium. Neuron 13, 301313 93. Keilhoff, G., Seidel, B., Noack, H., Tischmeyer, W., Stanek, D., and Wolf, G. (1996) Patterns of nitric oxide synthase at the messenger RNA and protein levels during early rat brain development. Neuroscience 75, 11931201

CONSTITUTIVE ISOFORMS OF NO SYNTHASE

787

94.

95.

96.

97. 98.

99. 100. 101. 102.

103.

104.

105. 106. 107.

108. 109. 110.

111.

112.

113.

Northington, F. J., Tobin, J. R., Harris, A. P., Traystman, R. J., and Koehler, R. C. (1997) Developmental and regional differences in nitric oxide synthase activity and blood ow in the sheep brain. J. Cereb. Blood Flow Metab. 17, 109115 Xue, C., Reynolds, P. R., and Johns, R. A. (1996) Developmental expression of NOS isoforms in fetal rat lung: implications for transitional circulation and pulmonary angiogenesis. Am. J. Physiol. 270, L88L100 Holtzman, D. M., Lee, S., Li, Y., Chua Couzens, J., Xia, H., Bredt, D. S., and Mobley, W. C. (1996) Expression of neuronalNOS in developing basal forebrain cholinergic neurons: regulation by NGF. Neurochem. Res. 21, 861868 Lizasoain, I., Weiner, C. P., Knowles, R. G., and Moncada, S. (1996) The ontogeny of cerebral and cerebellar nitric oxide synthase in the guinea pig and rat. Pediatr. Res. 39, 779783 Baader, S. L., Bucher, S., and Schilling, K. (1997) The developmental expression of neuronal nitric oxide synthase in cerebellar granule cells is sensitive to GABA and neurotrophins. Dev. Neurosci. 19, 283290 Ogura, T., Nakayama, K., Fujisawa, H., and Esumi, H. (1996) Neuronal nitric oxide synthase expression in neuronal cell differentiation. Neurosci. Lett. 204, 8992 Kanda, K. (1996) Expression of neuronal nitric oxide synthase in spinal motoneurons in aged rats. Neurosci. Lett. 219, 4144 Ma, S. X., Holley, A. T., Sandra, A., Cassell, M. D., and Abboud, F. M. (1997) Increased expression of nitric oxide synthase in the Gracile nucleus of aged rats. Neuroscience 76, 659663 Kishimoto, J., Spurr, N., Liao, M., Lizhi, L., Emson, P., and Xu, W. (1992) Localization of brain nitric oxide synthase (NOS) to human chromosome 12. Genomics 14, 802804 [erratum (1993)] Genomics 15, 465 Marsden, P. A., Heng, H. H., Scherer, S. W., Stewart, R. J., Hall, A. V., Shi, X. M., Tsui, L. C., and Schappert, K. T. (1993) Structure and chromosomal localization of the human constitutive endothelial nitric oxide synthase gene. J. Biol. Chem. 268, 1747817488 Xu, W., Gorman, P., Sheer, D., Bates, G., Kishimoto, J., Lizhi, L., and Emson, P. (1993) Regional localization of the gene coding for human brain nitric oxide synthase (NOS1) to 12q24.2r24.31 by uorescent in situ hybridization. Cytogenet. Cell Genet. 64, 6263 Lee, C. G., Gregg, A. R., and OBrien, W. E. (1995) Localization of the neuronal form of nitric oxide synthase to mouse chromosome 5. Mamm. Genome 6, 5657 Deng, A. Y., Rapp, J. P., Kato, H., and Bihoreau, M. T. (1995) Linkage mapping of the neuronal nitric oxide synthase gene (Nos1) to rat chromosome 12. Mamm. Genome 6, 824 Marsden, P. A., Antoniou, H., Cheung, A., Arbus, A., and Hall, A. V. (1994) Characterization of the human neuronal nitric oxide synthase (NOS) gene: novel allelic structural mRNA diversity. In Abstracts of the First International Conference on Biochemistry and Molecular Biology of Nitric Oxide (Ignarro L. J., and Murad F., eds) Vol. 31, University of California, Los Angeles Huang, P. L., Dawson, T. M., Bredt, D. S., Snyder, S. H., and Fishman, M. C. (1993) Targeted disruption of the neuronal nitric oxide synthase gene. Cell 75, 12731286 Brenman, J. E., Xia, H. H., Chao, D. S., Black, S. M., and Bredt, D. S. (1997) Regulation of neuronal nitric oxide synthase through alternative transcripts. Dev. Neurosci. 19, 224231 Lee, M. A., Cai, L., Hu bner, N., Lee, Y. A., and Lindpaintner, K. (1997) Tissue- and development-specic expression of multiple alternatively spliced transcripts of rat neuronal nitric oxide synthase. J. Clin. Invest. 100, 15071512 Bredt, D. S., Hwang, P. M., Glatt, C. E., Lowenstein, C., Reed, R. R., and Snyder, S. H. (1991) Cloned and expressed nitric oxide synthase structurally resembles cytochrome P-450 reductase. Nature (London) 351, 714718 Ogura, T., Yokoyama, T., Fujisawa, H., Kurashima, Y., and Esumi, H. (1993) Structural diversity of neuronal nitric oxide synthase mRNA in the nervous system. Biochem. Biophys. Res. Commun. 193, 10141022 Chartrain, N. A., Geller, D. A., Koty, P. P., Sitrin, N. F., Nussler, A. K., Hoffman, E. P., Billiar, T. R., Hutchinson, N. I., and Mudgett, J. S. (1994) Molecular cloning, structure, and chromosomal localization of the human inducible nitric oxide synthase gene. J. Biol. Chem. 269, 67656772

114.

115. 116. 117.

118.

119. 120.

121.

122.

123.

124.

125.

126.

127.

128.

129.

130.

131.

132.

Nadaud, S., Bonnardeaux, A., Lathrop, M., and Soubrier, F. (1994) Gene structure, polymorphism and mapping of the human endothelial nitric oxide synthase gene. Biochem. Biophys. Res. Commun. 198, 10271033 Takahashi, Y., Nakayama, T., Soma, M., Izumi, Y., and Kanmatsuse, K. (1997) CA repeat polymorphism of the neuronal nitric oxide synthase gene. Human Hered. 47, 5859 Rich, A., Nordheim, A., and Wang, A. H.-J. (1984) The chemistry and biology of left-handed Z-DNA. Annu. Rev. Biochem. 53, 791846 Xie, J., Roddy, P., Rife, T. K., Murad, F., and Young, A. P. (1995) Two closely linked but separable promoters for human neuronal nitric oxide synthase gene transcription. Proc. Natl. Acad. Sci. USA 92, 12421246 Young, A. P., Murad, F., Vaessin, H., Xie, J., and Rife, T. K. (1995) Transcription of the human neuronal nitric oxide synthase gene in the central nervous system is mediated by multiple promoters. Adv. Pharmacol. 34, 91112 Ayoubi, T. A. Y., and Van de Ven, W. J. M. (1996) Regulation of gene expression by alternative promoters. FASEB J. 10, 453 460 Deans, Z., Dawson, S. J., Xie, J., Young, A. P., Wallace, D., and Latchman, D. S. (1996) Differential regulation of the two neuronal nitric-oxide synthase gene promoters by the Oct-2 transcription factor. J. Biol. Chem. 271, 3215332158 Eliasson, M. J., Blackshaw, S., Schell, M. J., and Snyder, S. H. (1997) Neuronal nitric oxide synthase alternatively spliced forms: prominent functional localizations in the brain. Proc. Natl. Acad. Sci. USA 94, 33963401 Fujisawa, H., Ogura, T., Kurashima, Y., Yokoyama, T., Yamashita, J., and Esumi, H. (1994) Expression of two types of nitric oxide synthase mRNA in human neuroblastoma cell lines. J. Neurochem 63, 140145 Ogilvie, P., Schilling, K., Billingsley, M. L., and Schmidt, H. H. H. W. (1995) Induction and variants of neuronal nitric oxide synthase type I during synaptogenesis. FASEB J. 9, 799 806 Kolesnikov, Y. A., Pan, Y. X., Babey, A. M., Jain, S., Wilson, R., Pasternak, G. W. (1997) Functionally differentiating two neuronal nitric oxide synthase isoforms through antisense mapping: evidence for opposing NO actions on morphine analgesia and tolerance. Proc. Natl. Acad. Sci. USA 94, 82208225 Regulski, M., and Tully, T. (1995) Molecular and biochemical characterization of dNOS: a Drosophila Ca2//calmodulin-dependent nitric oxide synthase. Proc. Natl. Acad. Sci. USA 92, 90729076 Chao, D. S., Silvagno, F., Xia, H., Cornwell, T. L., Lincoln, T. M., and Bredt, D. S. (1997) Nitric oxide synthase and cyclic GMP dependent protein kinase concentrated at the neuromuscular endplate. Neuroscience 76, 665672 Forstermann, U., Pollock, J. S., Schmidt, H. H. H. W., Heller, M., and Murad, F. (1991) Calmodulin-dependent endotheliumderived relaxing factor/nitric oxide synthase activity is present in the particulate and cytosolic fractions of bovine aortic endothelial cells. Proc. Natl. Acad. Sci. USA 88, 17881792 Pollock, J. S., Forstermann, U., Mitchell, J. A., Warner, T. D., Schmidt, H. H. H. W., Nakane, M., and Murad, F. (1991) Purication and characterization of particulate endothelium-derived relaxing factor synthase from cultured and native bovine aortic endothelial cells. Proc. Natl. Acad. Sci. USA 88, 1048010484 Pollock, J. S., Nakane, M., Buttery, L. K., Martinez, A., Springall, D., Polak, J. M., Forstermann, U., and Murad, F. (1993) Char acterization and localization of endothelial nitric oxide synthase using specic monoclonal antibodies. Am. J. Physiol. 265, C1379C1387 Dinerman, J. L., Dawson, T. M., Schell, M. J., Snowman, A., and Snyder, S. H. (1994) Endothelial nitric oxide synthase localized to hippocampal pyramidal cells: Implications for synaptic plasticity. Proc. Natl. Acad. Sci. USA 91, 42144218 Abe, K., Pan, L. H., Watanabe, M., Konno, H., Kato, T., and Itoyama, Y. (1997) Upregulation of protein-tyrosine nitration in the anterior horn cells of amyotrophic lateral sclerosis. Neurol. Res. 19, 124128 Helfrich, M. H., Evans, D. E., Grabowski, P. S., Pollock, J. S., Ohshima, H., and Ralston, S. H. (1997) Expression of nitric oxide synthase isoforms in bone and bone cell cultures. J. Bone Miner. Res. 12, 11081115

788

Vol. 12

July 1998

The FASEB Journal

FORSTERMANN ET AL.

133.

134.

135.

136.

137. 138. 139.

140.

141.

142.

143. 144.

145.

146.

147.

148. 149.

150.

151.

152.

Wang, R., Ghahary, A., Shen, Y. J., Scott, P. G., and Tredget, E. E. (1996) Human dermal broblasts produce nitric oxide and express both constitutive and inducible nitric oxide synthase isoforms. J. Invest. Dermatol. 106, 419427 Tseng, L., Zhang, J., Peresleni, T., and Goligorsky, M. S. (1996) Cyclic expression of endothelial nitric oxide synthase mRNA in the epithelial glands of human endometrium. J. Soc. Gynecol. Invest. 3, 3338 Zini, A., OBryan, M. K., Magid, M. S., and Schlegel, P. N. (1996) Immunohistochemical localization of endothelial nitric oxide synthase in human testis, epididymis, and vas deferens suggests a possible role for nitric oxide in spermatogenesis, sperm maturation, and programmed cell death. Biol. Reprod. 55, 935941 Shaul, P. W., North, A. J., Wu, L. C., Wells, L. B., Brannon, T. S., Lau, K. S., Michel, T., Margraf, L. R., and Star, R. A. (1994) Endothelial nitric oxide synthase is expressed in cultured human bronchiolar epithelium. J. Clin. Invest. 94, 22312236 Conrad, K. P., Vill, M., McGuire, P. G., Dail, W. G., and Davis, A. K. (1993) Expression of nitric oxide synthase by syncytiotrophoblast in human placental villi. FASEB J. 7, 12691276 Sase, K., and Michel, T. (1995) Expression of constitutive endothelial nitric oxide synthase in human blood platelets. Life Sci. 57, 20492055 Reiling, N., Kroncke, R., Ulmer, A. J., Gerdes, J., Flad, H. D., and Hauschildt, S. (1996) Nitric oxide synthase: expression of the endothelial, Ca2//calmodulin-dependent isoform in human B and T lymphocytes. Eur. J. Immunol. 26, 511516 Colin, I. M., Kopp, P., Zbaren, J., Haberli, A., Grizzle, W. E., and Jameson, J. L. (1997) Expression of nitric oxide synthase III in human thyroid follicular cells: Evidence for increased expression in hyperthyroidism. Eur. J. Endocrinol. 136, 649655 Roman, V., Dugas, N., Abadie, A., Amirand, C., Zhao, H., Dugas, B., and Kolb, J. P. (1997) Characterization of a constitutive type III nitric oxide synthase in human U937 monocytic cells: stimulation by soluble CD23. Immunology 91, 643648 Balligand, J. L., Kobzik, L., Han, X., Kaye, D. M., Belhassen, L., OHara, D. S., Kelly, R. A., Smith, T. W., and Michel, T. (1995) Nitric oxide-dependent parasympathetic signaling is due to activation of constitutive endothelial (type III) nitric oxide synthase in cardiac myocytes. J. Biol. Chem. 270, 1458214586 Hussain, S. N., El-Dwairi, Q., Abdul-Hussain, M. N., and Sakkal, D. (1997) Expression of nitric oxide synthase isoforms in normal ventilatory and limb muscles. J. Appl. Physiol. 83, 348353 Chen, K., Inoue, M., Wasa, M., Fukuzawa, M., Kamata, S., and Okada, A. (1997) Expression of endothelial constitutive nitric oxide synthase mRNA in gastrointestinal mucosa and its downregulation by endotoxin. Life Sci. 61, 13231329 Zimmermann, H., Kurzen, P., Klossner, W., Renner, E. L., and Marti, U. (1996) Decreased constitutive hepatic nitric oxide synthase expression in secondary biliary brosis and its changes after Roux-en-Y choledocho-jejunostomy in the rat. J. Hepatol. 25, 567573 Comini, L., Bachetti, T., Gaia, G., Pasini, E., Agnoletti, L., Pepi, P., Ceconi, C., Curello, S., and Ferrari, R. (1996) Aorta and skeletal muscle NO synthase expression in experimental heart failure. J. Mol. Cell. Cardiol 28, 22412248 Jablonka Shariff, A., and Olson, L. M. (1997) Hormonal regulation of nitric oxide synthases and their cell-specic expression during follicular development in the rat ovary. Endocrinology 138, 460468 Barna, M., Komatsu, T., and Reiss, C. S. (1996) Activation of type III nitric oxide synthase in astrocytes following a neurotropic viral infection. Virology 223, 331343 Xue, C., Pollock, J., Schmidt, H. H. H. W., Ward, S. M., and Sanders, K. M. (1994) Expression of nitric oxide synthase immunoreactivity by interstitial cells of the canine proximal colon. J. Auton. Nerv. Syst. 49, 114 Tracey, W. R., Pollock, J. S., Murad, F., Nakane, M., and For stermann, U. (1994) Identication of a type III (endotheliallike) particulate nitric oxide synthase in LLC-PK1 kidney tubular epithelial cells. Am. J. Physiol. 266, C22C26 Garcia Cardena, G., Oh, P., Liu, J., Schnitzer, J. E., and Sessa, W. C. (1996) Targeting of nitric oxide synthase to endothelial cell caveolae via palmitoylation: implications for nitric oxide signaling. Proc. Natl. Acad. Sci. USA 93, 64486453 Feron, O., Belhassen, L., Kobzik, L., Smith, T. W., Kelly, R. A., and Michel, T. (1996) Endothelial nitric oxide synthase target-

153.

154. 155.

156.

157. 158.

159.

160.

161.

162.

163.

164.

165. 166.

167.

168. 169.

170. 171.

ing to caveolae. Specic interactions with caveolin isoforms in cardiac myocytes and endothelial cells. J. Biol. Chem. 271, 2281022814 Liu, J. W., Hughes, T. E., and Sessa, W. C. (1997) The rst 35 amino acids and fatty acylation sites determine the molecular targeting of endothelial nitric oxide synthase into the golgi region of cells: a green uorescent protein study. J. Cell Biol. 137, 15251535 Michel, J. B., Feron, O., Sacks, D., and Michel, T. (1997) Reciprocal regulation of endothelial nitric-oxide synthase by Ca2/calmodulin and caveolin. J. Biol. Chem. 272, 1558315586 Sessa, W. C., Garcia Cardena, G., Liu, J., Keh, A., Pollock, J. S., Bradley, J., Thiru, S., Braverman, I. M., and Desai, K. M. (1995) The Golgi association of endothelial nitric oxide synthase is necessary for the efcient synthesis of nitric oxide. J. Biol. Chem. 270, 1764117644 Forstermann, U., Closs, E. I., Pollock, J. S., Nakane, M., Schwarz, P., Gath, I., and Kleinert, H. (1994) Nitric oxide synthase isozymes: characterization, molecular cloning and functions. Hypertension 23, 11211131 Forstermann, U., Nakane, M., Tracey, W. R., and Pollock, J. S. (1993) Isoforms of nitric oxide synthase: functions in the cardiovascular system. Eur Heart J. 14 (Suppl. I), 1015 Nishida, K., Harrison, D. G., Navas, J. P., Fisher, A. A., Dockery, S. P., Uematsu, M., Nerem, R. M., Alexander, R. W., and Murphy, T. J. (1992) Molecular cloning and characterization of the constitutive bovine aortic endothelial cell nitric oxide synthase. J. Clin. Invest. 90, 20922096 Sessa, W. C., Pritchard, K., Seyedi, N., Wang, J., and Hintze, T. H. (1994) Chronic exercise in dogs increases coronary vascular nitric oxide production and endothelial cell nitric oxide synthase gene expression. Circ. Res. 74, 349353 Xiao, Z., Zhang, Z., and Diamond, S. L. (1997) Shear stress induction of the endothelial nitric oxide synthase gene is calcium-dependent but not calcium-activated. J. Cell. Physiol. 171, 205211 Venema, R. C., Nishida, K., Alexander, R. W., Harrison, D. G., and Murphy, T. J. (1994) Organization of the bovine gene encoding the endothelial nitric oxide synthase. Biochim. Biophys. Acta 1218, 413420 Ziesche, R., Petkov, V., Williams, J., Zakeri, S. M., Mosgoller, W., Knoer, M., and Block, L. H. (1996) Lipopolysaccharide and interleukin 1 augment the effects of hypoxia and inammation in human pulmonary arterial tissue. Proc. Natl. Acad. Sci. USA 93, 1247812483 Dai, A., Zhang, Z., and Niu, R. (1995) The effects of hypoxia on nitric oxide synthase activity and mRNA expression of pulmonary artery endothelial cells in pigs. Chung Hua Chieh Ho Ho Hu Hsi Tsa Chih 18, 164166 Liao, J. K., Zulueta, J. J., Yu, F. S., Peng, H. B., Cote, C. G., and Hassoun, P. M. (1995) Regulation of bovine endothelial constitutive nitric oxide synthase by oxygen. J. Clin. Invest. 96, 2661 2666 Arnet, U. A., McMillan, A., Dinerman, J. L., Ballermann, B., and Lowenstein, C. J. (1996) Regulation of endothelial nitric-oxide synthase during hypoxia. J. Biol. Chem. 271, 1506915073 Zhang, Z. G., Chopp, M., Zaloga, C., Pollock, J. S., and Forster mann, U. (1993) Cerebral endothelial nitric oxide synthase expression after focal cerebral ischemia in rats. Stroke 24, 2016 2021 Mcquillan, L. P., Leung, G. K., Marsden, P. A., Kostyk, S. K., and Kourembanas, S. (1994) Hypoxia inhibits expression of eNOS via transcriptional and posttranscriptional mechanisms. Am. J. Physiol. 36, H1921H1927 Phelan, M. W., and Faller, D. V. (1996) Hypoxia decreases constitutive nitric oxide synthase transcript and protein in cultured endothelial cells. J. Cell. Physiol. 167, 469476 Arnal, J. F., Yamin, J., Dockery, S., and Harrison, D. G. (1994) Regulation of endothelial nitric oxide synthase mRNA, protein, and activity during cell growth. Am. J. Physiol. 36, C1381C1388 Harrison, D. G. (1997) Post transcriptional regulation of nitric oxide synthase. Jpn. J. Pharmacol. 75, P399 Flowers, M. A., Wang, Y., Stewart, R. J., Patel, B., and Marsden, P. A. (1995) Reciprocal regulation of endothelin-1 and endothelial constitutive NOS in proliferating endothelial cells. Am. J. Physiol. 269, H1988H1997

CONSTITUTIVE ISOFORMS OF NO SYNTHASE

789

172.

173.

174.

175.

176.

177.

178.

179. 180.

181.

182.

183.

184.

185. 186.

187. 188. 189. 190.

Lamas, S., Marsden, P. A., Li, G. K., Tempst, P., and Michel, T. (1992) Endothelial nitric oxide synthase: molecular cloning and characterization of a distinct constitutive enzyme isoform. Proc. Natl. Acad. Sci. USA 89, 63486352 Yoshizumi, M., Perrella, M. A., Burnett, J. C., and Lee, M. E. (1993) Tumor necrosis factor downregulates an endothelial nitric oxide synthase messenger RNA by shortening its half-life. Circ. Res. 73, 205209 Alonso, J., Sanchez de Miguel, L., Monton, M., Casado, S., and Lopez-Farre, A. (1997) Endothelial cytosolic proteins bind to the 3 untranslated region of endothelial nitric oxide synthase mRNA: regulation by tumor necrosis factor alpha. Mol. Cell. Biol. 17, 57195726 Kaku, Y., Nanri, H., Sakimura, T., Ejima, K., Kuroiwa, A., and Ikeda, M. (1997) Differential induction of constitutive and inducible nitric oxide synthases by distinct inammatory stimuli in bovine aortic endothelial cells. Biochim. Biophys. Acta 1356, 4352 Bucher, M., Ittner, K. P., Zimmermann, M., Wolf, K., Hobbhahn, J., and Kurtz, A. (1997) Nitric oxide synthase isoform III gene expression in rat liver is up-regulated by lipopolysaccharide and lipoteichoic acid. FEBS Lett. 412, 511514 Weiner, C. P., Lizasoain, I., Baylis, S. A., Knowles, R. G., Charles, I. G., and Moncada, S. (1994) Induction of calcium-dependent nitric oxide synthases by sex hormones. Proc. Natl. Acad. Sci. USA 91, 52125216 Goetz, R. M., Morano, I., Calovini, T., Studer, R., and Holtz, J. (1994) Increased expression of endothelial constitutive nitric oxide synthase in rat aorta during pregnancy. Biochem. Biophys. Res. Commun. 205, 905910 Neugarten, J., Ding, Q., Friedman, A., Lei, J., and Silbiger, S. (1997) Sex hormones and renal nitric oxide synthases. J. Am. Soc. Nephrol. 8, 12401246 Lantin-Hermoso, R. L., Rosenfeld, C. R., Yuhanna, I. S., German, Z., Chen, Z., and Shaul, P. W. (1997) Estrogen acutely stimulates nitric oxide synthase activity in fetal pulmonary artery endothelium. Am. J. Physiol. 17, L119L126 Arnal, J. F., Clamens, S., Pechet, C., Negre Salvayre, A., Allera, C., Girolami, J. P., Salvayre, R., and Bayard, F. (1996) Ethinylestradiol does not enhance the expression of nitric oxide synthase in bovine endothelial cells but increases the release of bioactive nitric oxide by inhibiting superoxide anion production. Proc. Natl. Acad. Sci. USA 93, 41084113 Kleinert, H., Wallerath, T., Euchenhofer, C. E., Ihrig-Biedert, I., Li, H., and Forstermann, U. (1998) Estrogens increase tran scription of the human endothelial NO synthase gene: analysis of the transcription factors involved. Hypertension 31, 582588 Inoue, N., Venema, R. C., Sayegh, H. S., Ohara, Y., Murphy, T. J., and Harrison, D. G. (1995) Molecular regulation of the bovine endothelial cell nitric oxide synthase by transforming growth factor-beta 1. Arterioscler. Thromb. Vasc. Biol. 15, 12551261 Kostyk, S. K., Kourembanas, S., Wheeler, E. L., Medeiros, D., McQuillan, L. P., DAmore, P. A., and Braunhut, S. J. (1995) Basic broblast growth factor increases nitric oxide synthase production in bovine endothelial cells. Am. J. Physiol. 269, H1583H1589 Liao, J. K., Shin, W. S., Lee, W. Y., and Clark, S. L. (1995) Oxidized low-density lipoprotein decreases the expression of endothelial nitric oxide synthase. J. Biol. Chem. 270, 319324 Hirata, K., Miki, N., Kuroda, Y., Sakoda, T., Kawashima, S., and Yokoyama, M. (1995) Low concentration of oxidized low-density lipoprotein and lysophosphatidylcholine upregulate constitutive nitric oxide synthase mRNA expression in bovine aortic endothelial cells. Circ. Res. 76, 958962 Zembowicz, A., Tang, J. L., and Wu, K. K. (1995) Transcriptional induction of endothelial nitric oxide synthase type III by lysophosphatidylcholine. J. Biol. Chem. 270, 1700617010 Wu, K. K., Cieslik, K., Zembowicz, A., and Tang, J. L. (1997) Transcriptional activation of endothelial nitric oxide-synthase (eNOS) gene. Jpn. J. Pharmacol. 75, A8 (Abstr.) Ohara, Y., Sayegh, H. S., Yamin, J. J., and Harrison, D. G. (1995) Regulation of endothelial constitutive nitric oxide synthase by protein kinase C. Hypertension 25, 415420 Li, H., Oehrlein, S. A., Wallerath, T., Ihrig-Biedert, I., Wohlfart, P., Ulshofer, T., Jessen, T., Herget, T., Forstermann, U., and Kleinert, H. (1998) Activation of protein kinase Ca and/or e enhances transcription of the human endothelial NO synthase gene. Mol. Pharmacol. 53 In press

191.

192. 193.

194.

195.

196.

197.

198. 199.

200.

201.

202.

203.

204.

205.

206.

207.

208.

Kanazawa, K., Kawashima, S., Mikami, S., Miwa, Y., Hirata, K., Suematsu, M., Hayashi, Y., Itoh, H., and Yokoyama, M. (1996) Endothelial constitutive nitric oxide synthase protein and mRNA increased in rabbit atherosclerotic aorta despite impaired endothelium-dependent vascular relaxation. Am. J. Pathol. 148, 19491956 Ravichandran, L. V., and Johns, R. A. (1995) Up-regulation of endothelial nitric oxide synthase expression by cyclic guanosine 3 ,5 -monophosphate. FEBS Lett. 374, 295298 Cosentino, F., Hishikawa, K., Katusic, Z. S., and Lu scher, T. F. (1997) High glucose increases nitric oxide synthase expression and superoxide anion generation in human aortic endothelial cells. Circulation 96, 2528 Suschek, C., Fehsel, K., Kroncke, K. D., Sommer, A., and Kolb Bachofen, V. (1994) Primary cultures of rat islet capillary endothelial cells. Constitutive and cytokine-inducible macrophage-like nitric oxide synthases are expressed and activities regulated by glucose concentration. Am. J. Pathol. 145, 685695 Olson, S. C., Dowds, T. A., Pino, P. A., Barry, M. T., and BurkeWolin, T. (1997) ANG II stimulates endothelial nitric oxide synthase expression in bovine pulmonary artery endothelium. Am. J. Physiol. 17, L315L321 Martin, P. Y., Xu, D. L., Niederberger, M., Weigert, A., Tsai, P., John, J., Gines, P., and Schrier, R. W. (1996) Upregulation of endothelial constitutive NOS: a major role in the increased NO production in cirrhotic rats. Am. J. Physiol. 270, F494F499 Crabos, M., Coste, P., Paccalin, M., Tariosse, L., Daret, D., Besse, P., and Bonoron Adele, S. (1997) Reduced basal NO-mediated dilation and decreased endothelial NO-synthase expression in coronary vessels of spontaneously hypertensive rats. J. Mol. Cell. Cardiol. 29, 5565 Giaid, A., and Saleh, D. (1995) Reduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N. Engl. J. Med. 333, 214221 Belhassen, L., Kelly, R. A., Smith, T. W., and Balligand, J. L. (1996) Nitric oxide synthase (NOS3) and contractile responsiveness to adrenergic and cholinergic agonists in the heart. Regulation of NOS3 transcription in vitro and in vivo by cyclic adenosine monophosphate in rat cardiac myocytes. J. Clin. Invest. 97, 19081915 Miyahara, K., Kawamoto, T., Sase, K., Yui, Y., Toda, K., Yang, L. X., Hattori, R., Aoyama, T., Yamamoto, Y., Doi, Y., Ogoshi, S., Hashimoto, K., Kawai, C., Sasayama, S., and Shizuta, Y. (1994) Cloning and structural characterization of the human endothelial nitric-oxide-synthase gene. Eur. J. Biochem. 223, 719726 Janssens, S. P., Shimouchi, A., Quertermous, T., Bloch, D. B., and Bloch, K. D. (1992) Cloning and expression of a cDNA encoding human endothelium-derived relaxing factor/nitric oxide synthase. J. Biol. Chem. 267, 1451914522 [erratum (1992)] J. Biol. Chem. 267, 22694 Marsden, P. A., Schappert, K. T., Chen, H. S., Flowers, M., Sundell, C. L., Wilcox, J. N., Lamas, S., and Michel, T. (1992) Molecular cloning and characterization of human endothelial nitric oxide synthase. FEBS Lett. 307, 287293 Gnanapandithen, K., Chen, Z., Kau, C. L., Gorczynski, R. M., and Marsden, P. A. (1996) Cloning and characterization of murine endothelial constitutive nitric oxide synthase. Biochim. Biophys. Acta 1308, 103106 Sessa, W. C., Harrison, J. K., Barber, C. M., Zeng, D., Durieux, M. E., DAngelo, D. D., Lynch, K. R., and Peach, M. J. (1992) Molecular cloning and expression of a cDNA encoding endothelial cell nitric oxide synthase. J. Biol. Chem. 267, 1527415276 Zhang, J., Patel, J. M., and Block, E. R. (1997) Molecular cloning, characterization and expression of a nitric oxide synthase from porcine pulmonary artery endothelial cells. Comp. Biochem. Physiol. 116, 485491 Bonnardeaux, A., Nadaud, S., Charru, A., Jeunemaitre, X., Corvol, P., and Soubrier, F. (1995) Lack of evidence for linkage of the endothelial cell nitric oxide synthase gene to essential hypertension. Circulation 91, 96102 Wang, X. L., Sim, A. S., Badenhop, R. F., McCredie, R. M., and Wilcken, D. E. (1996) A smoking-dependent risk of coronary artery disease associated with a polymorphism of the endothelial nitric oxide synthase gene. Nature Med. 2, 4145 Zhang, R., Min, W., and Sessa, W. C. (1995) Functional analysis of the human endothelial nitric oxide synthase promoter. Sp1 and GATA factors are necessary for basal transcription in endothelial cells. J. Biol. Chem. 270, 1532015326

790

Vol. 12

July 1998

The FASEB Journal

FORSTERMANN ET AL.

Você também pode gostar