Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Martin Petkovich is active.

Publication


Featured researches published by Martin Petkovich.


Journal of Biological Chemistry | 1997

cDNA Cloning of Human Retinoic Acid-metabolizing Enzyme (hP450RAI) Identifies a Novel Family of Cytochromes P450 (CYP26)

Jay A. White; Beckett-Jones B; Guo Yd; Dilworth Fj; Bonasoro J; Glenville Jones; Martin Petkovich

Retinoids, including all-trans-retinoic acid (RA) and its stereoisomer 9-cis-RA play important roles in regulating gene expression, through interactions with nuclear receptors, during embryonic development and in the maintenance of adult epithelial tissues (Chambon, P. (1995) Rec. Prog. Horm. Res. 50, 317–32; Mangelsdorf, D. J., and Evans, R. M. (1995)Cell 83, 841–850; Petkovich, M. (1992) Annu. Rev. Nutr. 12, 443–471). Evidence suggests that 4-hydroxylation of RA inside the target cell limits its biological activity and initiates a degradative process of RA leading to its eventual elimination. However, 18-hydroxylation and glucuronidation may also be important steps in this process. In this paper, we describe the cloning and characterization of the first mammalian retinoic acid-inducible retinoic acid-metabolizing cytochrome P450 (hP450RAI), which belongs to a novel class of cytochromes (CYP26). We demonstrate that hP450RAI is responsible for generation of several hydroxylated forms of RA, including 4-OH-RA, 4-oxo-RA, and 18-OH-RA. We also show that hP450RAI mRNA expression is highly induced by RA in certain human tumor cell lines and further show that RA-inducible RA metabolism may correlate with P450RAI expression. We conclude that this enzyme plays a key role in RA metabolism, functioning in a feedback loop where RA levels are controlled in an autoregulatory manner.


Journal of Biological Chemistry | 1996

Identification of the retinoic acid-inducible all-trans-retinoic acid 4-hydroxylase.

Jay A. White; Guo Yd; Baetz K; Beckett-Jones B; Bonasoro J; Hsu Ke; Dilworth Fj; Glenville Jones; Martin Petkovich

Retinoic acid (RA) metabolites of vitamin A are key regulators of gene expression involved in embryonic development and maintenance of epithelial tissues. The cellular effects of RA are dependent upon the complement of nuclear receptors expressed (RARs and RXRs), which transduce retinoid signals into transcriptional regulation, the presence of cellular retinoid-binding proteins (CRABP and CRBP), which may be involved in RA metabolism, and the activity of RA metabolizing enzymes. We have been using the zebrafish as a model to study these processes. To identify genes regulated by RA during exogenous RA exposure, we utilized mRNA differential display. We describe the isolation and characterization of a cDNA, P450RAI, encoding a novel member of the cytochrome P450 family. mRNA transcripts for P450RAI are expressed normally during gastrulation, and in a defined pattern in epithelial cells of the regenerating caudal fin in response to exogenous RA. In COS-1 cells transfected with the P450RAI cDNA, all-trans-RA is rapidly metabolized to more polar metabolites. We have identified 4-oxo-RA and 4-OH-RA as major metabolic products of this enzyme. P450RAI represents the first enzymatic component of RA metabolism to be isolated and characterized at the molecular level and provides key insight into regulation of retinoid homeostasis.


Nature Genetics | 2002

Genetic evidence that oxidative derivatives of retinoic acid are not involved in retinoid signaling during mouse development.

Karen Niederreither; Suzan Abu-Abed; Brigitte Schuhbaur; Martin Petkovich; Pierre Chambon; Pascal Dollé

Retinoic acid, the active derivative of vitamin A (retinol), is a hormonal signaling molecule that acts in developing and adult tissues. The Cyp26a1 (cytochrome p450, 26) protein metabolizes retinoic acid into more polar hydroxylated and oxidized derivatives. Whether some of these derivatives are biologically active metabolites has been debated. Cyp26a1−/− mouse fetuses have lethal morphogenetic phenotypes mimicking those generated by excess retinoic acid administration, indicating that human CYP26A1 may be essential in controlling retinoic acid levels during development. This hypothesis suggests that the Cyp26a1−/− phenotype could be rescued under conditions in which embryonic retinoic acid levels are decreased. We show that Cyp26a1−/− mice are phenotypically rescued by heterozygous disruption of Aldh1a2 (also known as Raldh2), which encodes a retinaldehyde dehydrogenase responsible for the synthesis of retinoic acid during early embryonic development. Aldh1a2 haploinsufficiency prevents the appearance of spina bifida and rescues the development of posterior structures (sacral/caudal vertebrae, hindgut, urogenital tract), while partly preventing cervical vertebral transformations and hindbrain pattern alterations in Cyp26a1−/− mice. Thus, some of these double-mutant mice can reach adulthood. This study is the first report of a mutation acting as a dominant suppressor of a lethal morphogenetic mutation in mammals. We provide genetic evidence that ALDH1A2 and CYP26A1 activities concurrently establish local embryonic retinoic acid levels that must be finely tuned to allow posterior organ development and to prevent spina bifida.


Mechanisms of Development | 2001

Cloning of a novel retinoic-acid metabolizing cytochrome P450, Cyp26B1, and comparative expression analysis with Cyp26A1 during early murine development

Glenn MacLean; Suzan Abu-Abed; Pascal Dollé; Ali Tahayato; Pierre Chambon; Martin Petkovich

Tight regulation of retinoic acid (RA) distribution in the embryo is critical for normal morphogenesis. The RA-metabolizing enzymes Cyp26A1 and Cyp26B1 are believed to play important roles in protecting certain embryonic tissues from inappropriate RA signaling. We have cloned the murine Cyp26B1 cDNA and compared its expression pattern to that of Cyp26A1 from embryonic day (E) E7-E11.5 using in situ hybridization. Northern blot analysis shows the presence of two Cyp26B1 transcripts of approximately 2.3 and 3.5 kb in embryonic limb bud. Whereas Cyp26A1 is expressed in gastrulating embryos by E7, Cyp26B1 is first expressed at E8.0 in prospective rhombomeres 3 and 5. Cyp26B1 expression expands to specific dorso-ventral locations in rhombomeres 2-6 between E8.5 and E9.5, whereas Cyp26A1 hindbrain expression is limited to rhombomere 2 at E8.5. No (or very weak) Cyp26B1 expression is observed in the tail bud, a major site of Cyp26A1 expression. Differential expression is seen in branchial arches, with Cyp26A1 being mainly expressed in neural crest-derived mesenchyme, and Cyp26B1 in specific ectodermal and endodermal areas. Cyp26B1 is markedly expressed in the ectoderm and distal mesoderm of the limb buds from the beginning of their outgrowth. Cyp26A1 transcripts are seen later and at lower levels in limb ectoderm, and both transcripts are excluded from the apical ectodermal ridge.


Journal of Biological Chemistry | 1998

Mouse P450RAI (CYP26) Expression and Retinoic Acid-inducible Retinoic Acid Metabolism in F9 Cells Are Regulated by Retinoic Acid Receptor γ and Retinoid X Receptor α

Suzan Abu-Abed; Barbara R. Beckett; Hideki Chiba; James V. Chithalen; Glenville Jones; Daniel Metzger; Pierre Chambon; Martin Petkovich

We have cloned a mouse cDNA homolog of P450RAI, a cytochrome P450 belonging to a new family (CYP26), which has previously been isolated from zebrafish and human cDNAs and found to encode a retinoic acid-inducible retinoic acid hydroxylase activity. The cross-species conservation of the amino acid sequence is high, particularly between the mouse and the human enzymes, in which it is over 90%. Like its human and zibrafish counterparts, the mouse P450RAI cDNA catalyzes metabolism of retinoic acid into 4-OH-retinoic acid, 4-oxo-retinoic acid, 18-OH-retinoic acid, and unidentified water-soluble metabolites when transfected into COS-1 cells. Retinoic acid-inducible retinoic acid metabolism has previously been observed in F9 murine embryonal carcinoma cells and some derivatives lacking retinoid receptors. We were interested in determining whether P450RAI could be responsible for retinoic acid metabolism in F9 cells and in studying the effect of retinoid receptor ablation on P450RAIexpression. In wild-type F9 cells and derivatives lacking RARγ, RARα, and/or RXRα, we observed a direct relationship between the level of retinoic acid metabolic activity and retinoic acid-induced P450RAI mRNA. These experiments, as well as others using synthetic receptor subtype-specific retinoids, suggest that the RARγ and RXRα receptors mediate the effects of retinoic acid on the expression of theP450RAI gene.


Mechanisms of Development | 2002

Differential expression of the retinoic acid-metabolizing enzymes CYP26A1 and CYP26B1 during murine organogenesis.

Suzan Abu-Abed; Glenn MacLean; Valérie Fraulob; Pierre Chambon; Martin Petkovich; Pascal Dollé

We recently cloned the murine homologue of Cyp26B1, a novel retinoic acid (RA)-metabolizing enzyme and showed that its gene expression pattern is unique from that of Cyp26A1 during early embryogenesis. Here, we complete this comparative expression analysis from embryonic day (E) 12 to postnatal stages. Cyp26B1 expression was found in developing tendons and precartilaginous elements and in perichondrium by E14.5, while Cyp26A1 expression was restricted to extremities of rib and vertebral cartilage. Cyp26A1 and Cyp26B1 were expressed, in the distal epithelium and mesenchyme of the limbs and genital tubercle, respectively. High Cyp26B1 expression was found in craniofacial areas undergoing morphogenetic growth, whereas Cyp26A1 message was restricted to the mouth and dental epithelium. Cyp26A1 alone was expressed in the developing neural retina, while both genes were co-expressed in the retinal pigment epithelium. Cyp26B1 was specifically expressed in the developing hindbrain (pons, cerebellum) and forebrain (striatum, hippocampus), with forebrain expression persisting postnatally. In addition, Cyp26B1 was expressed at specific levels of the differentiating upper and lower thoracic spinal cord, adjacent to the cervical and lumbar regions that express the RA-synthesizing enzyme RALDH-2. In viscera, Cyp26B1 transcripts were detected in the developing lung, kidney, spleen, thymus and testis, whereas Cyp26A1 transcripts were found in the diaphragm and outer stomach mesenchyme. Cyp26B1 was also specifically expressed in dermis surrounding the developing hair follicles. Regulated RA metabolism may therefore be required in many developing systems.


Mechanisms of Development | 1999

DORSAL AND VENTRAL RETINAL TERRITORIES DEFINED BY RETINOIC ACID SYNTHESIS,BREAK-DOWN AND NUCLEAR RECEPTOR EXPRESSION

Peter McCaffery; Elisabeth Wagner; Jennifer O'Neil; Martin Petkovich; Ursula C. Dräger

Determination of the dorso-ventral dimension of the vertebrate retina is known to involve retinoic acid (RA), in that high RA activates expression of a ventral retinaldehyde dehydrogenase and low RA of a dorsal dehydrogenase. Here we show that in the early eye vesicle of the mouse embryo, expression of the dorsal dehydrogenase is preceded by, and transiently overlaps with, the RA-degrading oxidase CYP26. Subsequently in the embryonic retina, CYP26 forms a narrow horizontal boundary between the dorsal and ventral dehydrogenases, creating a trough between very high ventral and moderately high dorsal RA levels. Most of the RA receptors are expressed uniformly throughout the retina except for the RA-sensitive RARbeta, which is down-regulated in the CYP26 stripe. The orphan receptor COUP-TFII, which modulates RA responses, colocalizes with the dorsal dehydrogenase. The organization of the embryonic vertebrate retina into dorsal and ventral territories divided by a horizontal boundary has parallels to the division of the Drosophila eye disc into dorsal, equatorial and ventral zones, indicating that the similarities in eye morphogenesis extend beyond single molecules to topographical patterns.


Gene Expression Patterns | 2003

Cyp26C1 encodes a novel retinoic acid-metabolizing enzyme expressed in the hindbrain, inner ear, first branchial arch and tooth buds during murine development.

Ali Tahayato; Pascal Dollé; Martin Petkovich

Retinoic acid (RA), an active metabolite of vitamin A, is a crucial signaling molecule involved in tissue morphogenesis during embryonic development. RA distribution and concentration is precisely regulated during embryogenesis by balanced complementary activities of RA synthesizing (RALDH) and metabolizing (CYP26) enzymes. Here, we describe the identification of a novel murine p450 cytochrome belonging to the CYP26 family, mCYP26C1. Sequence alignment show that mCYP26C1 is more closely related to mCYP26B1 than mCYP26A1. At early developmental stages (E8.0-E8.5), mCyp26C1 is expressed in prospective rhombomeres 2 and 4, in the first branchial arch and along the lateral surface mesenchyme adjacent to the rostral hindbrain. At E9.5, mCyp26C1 expression persists in rhombomere 2 and in the maxillary and mandibular components of the first branchial arch, and is strongly induced in the lateral cervical mesenchyme. By mid-gestation, mCyp26C1 is weakly expressed in the cervical mesenchyme and in the maxillary component of the first branchial arch. At E11.5, mCyp26C1 can only be seen in a narrow band in the lateral cervical mesenchyme. During late gestation, mCyp26C1 exhibits region-specific expression in the inner ear epithelium and a persistent expression in the inner dental epithelium of the developing teeth. This pattern of expression suggests that mCYP26C1 may play an important role in protecting the hindbrain, first branchial arch, otocyst and tooth buds against RA exposure during embryonic development.


Biochemical Journal | 2005

Transcriptional co-operativity between distant retinoic acid response elements in regulation of Cyp26A1 inducibility

Olivier Loudig; Glenn MacLean; Naomi L. Dore; Luong Luu; Martin Petkovich

Cyp26A1 encodes an RA (retinoic acid)-catabolizing CYP (cytochrome P450) protein that plays a critical role in regulating RA distribution in vivo. Cyp26A1 expression is inducible by RA, and the locus has previously been shown to contain a RARE (RA response element), R1, within the minimal promoter [Loudig, Babichuk, White, Abu-Abed, Mueller and Petkovich (2000) Mol. Endocrinol. 14, 1483-1497]. In the present study, we report the identification of a second functional RARE (R2) located 2.0 kb upstream of the Cyp26A1 transcriptional start site. Constructs containing murine sequences encompassing both R1 and R2 showed that these elements work together to generate higher transcriptional activity upon treatment with RA than those containing R1 alone. Inclusion of R2 also dramatically enhanced the sensitivity of reporter constructs to RA, as even treatment with 10(-8) M RA resulted in a 5-fold induction of reporter activity. Mutational analysis identified R2 as the functional element responsible for the increased RA inducibility of promoter constructs. The element was shown to bind RARgamma (RA receptor gamma)/RXRalpha (retinoid X receptor alpha) heterodimers in vitro, and inclusion of nuclear receptors in transfections boosted the transcriptional response. A construct containing both R1 and R2 was used to generate a stable luciferase reporter cell line that can be used as a tool to identify factors regulating Cyp26A1 expression. The analysis of R1 and R2 has led to the proposal that the two elements work synergistically to provide a maximal response to RA and that R2 is an upstream enhancer.


Kidney International | 2010

Dysregulation of renal vitamin D metabolism in the uremic rat

Christian Helvig; Dominic Cuerrier; Christopher M. Hosfield; Breanna Ireland; Aza Z. Kharebov; Jae W. Kim; Navindra J. Ramjit; Kara Ryder; Samir P. Tabash; Andrew M. Herzenberg; Tina Epps; Martin Petkovich

The progressive decline in kidney function and concomitant loss of renal 1alpha-hydroxylase (CYP27B1) in chronic kidney disease (CKD) are associated with a gradual loss of circulating 25-hydroxyvitamin D(3) (25(OH)D(3)) and 1alpha,25-dihydroxyvitamin D(3) (1alpha,25(OH)(2)D(3)). However, only the decrease in 1alpha,25(OH)(2)D(3) can be explained by the decline of CYP27B1, suggesting that insufficiency of both metabolites may reflect their accelerated degradation by the key catabolic enzyme 24-hydroxylase (CYP24). To determine whether CYP24 is involved in causing vitamin D insufficiency and/or resistance to vitamin D therapy in CKD, we determined the regulation of CYP24 and CYP27B1 in normal rats and rats treated with adenine to induce CKD. As expected, CYP24 decreased whereas CYP27B1 increased when normal animals were rendered vitamin D deficient. Unexpectedly, renal CYP24 mRNA and protein expression were markedly elevated, irrespective of the vitamin D status of the rats. A significant decrease in serum 1alpha,25(OH)(2)D(3) levels was found in uremic rats; however, we did not find a coincident decline in CYP27B1. Analysis in human kidney biopsies confirmed the association of elevated CYP24 with kidney disease. Thus, our findings suggest that dysregulation of CYP24 may be a significant mechanism contributing to vitamin D insufficiency and resistance to vitamin D therapy in CKD.

Collaboration


Dive into the Martin Petkovich's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pascal Dollé

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christian Helvig

Wisconsin Alumni Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erica Hellestad

Wisconsin Alumni Research Foundation

View shared research outputs
Researchain Logo
Decentralizing Knowledge