Network


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

Hotspot


Dive into the research topics where Ying-Xian Pan is active.

Publication


Featured researches published by Ying-Xian Pan.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Generation of the mu opioid receptor (MOR-1) protein by three new splice variants of the Oprm gene

Ying-Xian Pan; Jin Xu; Loriann Mahurter; Elizabeth Bolan; Mingming Xu; Gavril W. Pasternak

Using 5′ RACE, we have isolated four additional exons of the mu opioid receptor gene (Oprm), resulting in a gene spanning over 250 kb. The four new exons are contained within eight additional splice variants containing exon 11 at the 5′ terminus. Exon 11, which is under the control of a previously unknown upstream promoter, and exon 12 are located ≈10 kb and ≈8 kb upstream from exon 1, respectively. Exon 13 and 14 are located between exons 1 and 2. The regional distributions of the variants, as determined by reverse transcription-PCR, varied among themselves and were distinct from that of MOR-1, implying region-specific RNA processing. Three variants (MOR-1H, MOR-1I, and MOR-1J) contained two potential translational start points, with the translational start point in exon 1 producing proteins identical to the original MOR-1 protein. When expressed, the receptor binding of these three variants was indistinguishable from that of MOR-1. The remaining eight proteins using the translation start point in exon 11 were all truncated, with three (MOR-1G, MOR-1M, and MOR-1N) predicting proteins of only six transmembrane domains and the rest giving proteins under 10 kDa. Western blots with an exon 11-specific antiserum revealed bands consistent with the six transmembrane domain proteins within the brain, but the shorter proteins were not detected. Thus, the MOR-1 protein can be generated by four different splice variants of the Oprm gene under the control of two physically distinct promoters. Although the truncated proteins are expressed in brain with a unique regional distribution, their functional significance remains unknown.


Pharmacological Reviews | 2013

Mu Opioids and Their Receptors: Evolution of a Concept

Gavril W. Pasternak; Ying-Xian Pan

Opiates are among the oldest medications available to manage a number of medical problems. Although pain is the current focus, early use initially focused upon the treatment of dysentery. Opium contains high concentrations of both morphine and codeine, along with thebaine, which is used in the synthesis of a number of semisynthetic opioid analgesics. Thus, it is not surprising that new agents were initially based upon the morphine scaffold. The concept of multiple opioid receptors was first suggested almost 50 years ago (Martin, 1967), opening the possibility of new classes of drugs, but the morphine-like agents have remained the mainstay in the medical management of pain. Termed mu, our understanding of these morphine-like agents and their receptors has undergone an evolution in thinking over the past 35 years. Early pharmacological studies identified three major classes of receptors, helped by the discovery of endogenous opioid peptides and receptor subtypes—primarily through the synthesis of novel agents. These chemical biologic approaches were then eclipsed by the molecular biology revolution, which now reveals a complexity of the morphine-like agents and their receptors that had not been previously appreciated.


Journal of Neurochemistry | 2002

Cloning and Characterization of a Mouse σ1 Receptor

Ying-Xian Pan; Jianfeng Mei; Jin Xu; Ban-Lin Wan; Amy Zuckerman; Gavril W. Pasternak

Abstract: A cDNA clone (S2‐1a) isolated from a mouse brain cDNA library, using a guinea pig σ1 cDNA as probe, has high homology to the predicted protein sequence of the guinea pig (88%) and human (90%) σ1 receptors. Northern analysis revealed a major mRNA of ∼1.8 kb in a wide range of mouse tissues, with highest levels in brain, liver, kidney, and thymus. Southern analysis and chromosomal mapping in the mouse suggested a single‐copy gene in region A5‐B2 of chromosome 4. Expression of the clone in MCF‐7 and CHO cells led to a pronounced increase in (+)‐[3H]pentazocine binding with a selectivity profile consistent with σ1 receptors. In vitro translation yielded a protein of ∼28 kDa, as did transfection of a probe containing the hemagglutinin (HA) epitope (S2‐1a.HA) into CHO cells, as determined by western analysis using an antibody directed against HA. (+)‐[3H]‐Pentazocine binding to immunopurified HA‐tagged receptor demonstrated conclusively that S2‐1a.HA encodes a high‐affinity (+)‐[3H]pentazocine binding site with characteristics of a murine σ1 receptor. An antisense oligodeoxynucleotide designed from S2‐1a potentiated opioid analgesia in vivo.


The Journal of Comparative Neurology | 2000

Differential distribution in rat brain of mu opioid receptor carboxy terminal splice variants MOR‐1C‐like and MOR‐1‐like immunoreactivity: Evidence for region‐specific processing

Catherine Abbadie; Ying-Xian Pan; Gavril W. Pasternak

The present study examined immunohistochemically the regional distribution of the mu opioid receptor splice variant MOR‐1C by using a rabbit antisera generated against the C‐terminal peptide sequences and compared it with MOR‐1. Overall, the distribution of MOR‐1C–like immunoreactivity (–LI) differed from MOR‐1–LI. Both MOR‐1C–LI and MOR‐1–LI were prominent in a few central nervous system regions, including the lateral parabrachial nucleus, the periaqueductal gray, and laminae I‐II of the spinal trigeminal nuclei and the spinal cord. In the striatum, hippocampal formation, presubiculum and parasubiculum, amygdaloid nuclei, thalamic nuclei, locus coeruleus, and nucleus ambiguous MOR‐1–LI predominated, whereas MOR‐1C–LI was absent or sparse. Conversely, MOR‐1C–LI exceeded MOR‐1–LI in the lateral septum, the deep laminae of the spinal cord, and most hypothalamic nuclei such as the median eminence, periventricular, suprachiasmatic, supraoptic, arcuate, paraventricular, ventromedial, and dorsomedial nuclei. Double‐labeling studies showed colocalization of the two receptors in neurons of the lateral septum, but not in the median eminence or in the arcuate nucleus, even though both MOR‐1 isoforms were expressed. Because both MOR‐1 and MOR‐1C are derived from the same gene, these differences in regional distribution represent region‐specific mRNA processing. The regional distributions reported in this study involve the epitope seen by the combinations of exons 7, 8, and 9. However, if other MOR‐1 variants containing exons 7, 8, and 9 exist, the antisera would not distinguish between them and MOR‐1C. J. Comp. Neurol. 419:244–256, 2000.


FEBS Letters | 2000

Isolation and expression of a novel alternatively spliced mu opioid receptor isoform, MOR-1F

Ying-Xian Pan; Jin Xu; Elizabeth Bolan; Albert Chang; Loriann Mahurter; Grace C. Rossi; Gavril W. Pasternak

The MOR‐1 gene is large, with a recent study reporting nine exons spanning 250 kb which combine to yield six different mu opioid receptor splice variants. We now report the isolation of exon 10, which is contained within yet another splice variant, MOR‐1F, which is composed of exons 1, 2, 3, 10, 6, 7, 8 and 9. Exon 10 comprises 186 bp which predict a unique 58 amino acid sequence extending beyond exon 3. It has been mapped between exons 4 and 6 and has flanking consensus splice sequences. On Northern blot analysis, the MOR‐1F mRNA is smaller than the other MOR‐1 mRNAs. When expressed in CHO cells, MOR‐1F binds the mu opioid radioligand [3H]DAMGO with high affinity (K D=1.04±0.03 nM). Competition studies demonstrated the selectivity of the variant for mu opioid ligands, supporting its classification within the mu opioid receptor family.


Neuroscience | 2000

Comparative immunohistochemical distributions of carboxy terminus epitopes from the mu-opioid receptor splice variants MOR-1D, MOR-1 and MOR-1C in the mouse and rat CNS

Catherine Abbadie; Ying-Xian Pan; Carrie T. Drake; Gavril W. Pasternak

The present study examined immunohistochemically the CNS distributions of a splice variant of the mu-opioid receptor, MOR-1D, in both rats and mice. In MOR-1D, exon 4 of MOR-1 is replaced by two additional exons that code for seven amino acids. Using rabbit antisera, we compared immunohistochemically the regional distribution of a C-terminal epitope of MOR-1D to that of a C-terminal epitope from MOR-1 and a C-terminal epitope from another splice variant, MOR-1C. The general distribution of MOR-1D-like immunoreactivity was similar in both mouse and rat. MOR-1D-like immunoreactivity was seen in the dentate gyrus and in the mossy fibers of the hippocampal formation, the nucleus of the solitary tract and the area postrema, the inferior olivary nucleus, the nucleus ambiguous, the spinal trigeminal nucleus and the spinal cord. MOR-1D-like immunoreactivity was not observed in some regions containing dense MOR-1-like immunoreactivity, such as the striatum or the locus coeruleus. In regions containing MOR-1, MOR-1C and MOR-1D, the pattern of each variant was unique.MOR-1D and MOR-1C are splice variants of the cloned mu-opioid receptor MOR-1. Although they differ only at the tip of the carboxy terminus, they show marked differences in their regional distributions, as determined immunohistochemically by epitopes in their unique carboxy termini. Since the splice variants are derived from the same gene, these differences in regional distribution imply region-specific messenger RNA processing.


Neuroscience | 2005

Identification and characterization of six new alternatively spliced variants of the human μ opioid receptor gene, Oprm

Ling Pan; Jin Xu; R. Yu; Mingming Xu; Ying-Xian Pan; Gavril W. Pasternak

The mu opioid receptor plays an important role in mediating the actions of morphine and morphine-like drugs. Receptor binding and a wide range of pharmacological studies have proposed several mu receptor subtypes, but only one mu opioid receptor (Oprm) gene has been isolated. Like the mouse and rat, the human Oprm gene undergoes alternative splicing. In the present studies, we have identified and characterized six new splice variants from the human Oprm gene using a reverse transcription-polymerase chain reaction strategy, yielding a total of 10 human splice variants of the mu opioid receptor MOR-1. All the variants identified contained exons 1, 2 and 3, but differed from MOR-1 itself and each other by splicing downstream from exon 3, resulting in different amino acid sequences. Northern blot analysis demonstrated expression of the variant mRNAs. Receptor binding assays established that these variants belonged to the mu opioid receptor family with limited differences in mu opioid ligand affinities and selectivity. However, adenylyl cyclase and [35S]GTPgammaS binding assays revealed major differences in both potency and efficacy among these variants. The dissociation between binding affinity, potency and efficacy for the opioids among these variants may provide insights into the wide range of opioid responses among these agents observed clinically and opens new avenues in designing selective drugs based upon their efficacy and potency rather simple binding affinity.


Biochemical and Biophysical Research Communications | 2003

Identification and characterization of two new human mu opioid receptor splice variants, hMOR-1O and hMOR-1X

Ying-Xian Pan; Jin Xu; Loriann Mahurter; Mingming Xu; Annie-Kim Gilbert; Gavril W. Pasternak

The mouse gene encoding the mu opioid receptor, Oprm, undergoes extensive alternatively splicing, with 14 variants having been identified. However, only one variant of human mu opioid receptor gene (Oprm), MOR-1A, has been described. We now report two novel splice variants of the human Oprm gene, hMOR-1O and hMOR-1X. The full-length cDNAs of hMOR-1O and hMO-1X contained the same exons 1, 2, and 3 as the original hMOR-1, but with exon O or exon X as the alternative fourth exon, respectively. Northern blots revealed several bands with the exon O probe in both human neuroblastoma BE(2)C cells and human brain and a single band (5.5kb) with the exon X probe in selected human brain regions. When transfected into CHO cells, both variants showed high selectivity for mu opioids in binding assays. These two new human mu opioid receptors are the first human MOR-1 variants containing new exons and suggest that the complex splicing present in mice may extend to humans.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Truncated G protein-coupled mu opioid receptor MOR-1 splice variants are targets for highly potent opioid analgesics lacking side effects

Susruta Majumdar; Steven Grinnell; Valerie Le Rouzic; Maxim Burgman; Lisa Polikar; Michael Ansonoff; John E. Pintar; Ying-Xian Pan; Gavril W. Pasternak

Pain remains a pervasive problem throughout medicine, transcending all specialty boundaries. Despite the extraordinary insights into pain and its mechanisms over the past few decades, few advances have been made with analgesics. Most pain remains treated by opiates, which have significant side effects that limit their utility. We now describe a potent opiate analgesic lacking the traditional side effects associated with classical opiates, including respiratory depression, significant constipation, physical dependence, and, perhaps most important, reinforcing behavior, demonstrating that it is possible to dissociate side effects from analgesia. Evidence indicates that this agent acts through a truncated, six-transmembrane variant of the G protein-coupled mu opioid receptor MOR-1. Although truncated splice variants have been reported for a number of G protein-coupled receptors, their functional relevance has been unclear. Our evidence now suggests that truncated variants can be physiologically important through heterodimerization, even when inactive alone, and can comprise new therapeutic targets, as illustrated by our unique opioid analgesics with a vastly improved pharmacological profile.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Involvement of exon 11-associated variants of the mu opioid receptor MOR-1 in heroin, but not morphine, actions

Ying-Xian Pan; Jin Xu; Mingming Xu; Grace C. Rossi; Joshua E. Matulonis; Gavril W. Pasternak

Heroin remains a major drug of abuse and is preferred by addicts over morphine. Like morphine, heroin has high affinity and selectivity for μ-receptors, but its residual analgesia in exon 1 MOR-1 knockout mice that do not respond to morphine suggests a different mechanism of action. MOR-1 splice variants lacking exon 1 have been observed in mice, humans, and rats, raising the possibility that they might be responsible for the residual heroin and morphine-6β-glucuronide (M6G) analgesia in the exon 1 knockout mice. To test this possibility, we disrupted exon 11 of MOR-1, which eliminates all of the variants that do not contain exon 1. Morphine and methadone analgesia in the exon 11 knockout mouse was normal, but the analgesic actions of heroin, M6G, and fentanyl were markedly diminished in the radiant heat tail-flick and hot-plate assays. Similarly, the ability of M6G to inhibit gastrointestinal transit was greatly diminished in these exon 11 knockout mice, whereas the ability of morphine was unchanged. These findings identify receptors selectively involved with heroin and M6G actions and confirm the relevance of the exon 11-associated variants and raise important issues regarding the importance of atypical truncated G-protein-coupled receptors.

Collaboration


Dive into the Ying-Xian Pan's collaboration.

Top Co-Authors

Avatar

Gavril W. Pasternak

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jin Xu

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mingming Xu

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Susruta Majumdar

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Zhigang Lu

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Valerie Le Rouzic

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge