Network


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

Hotspot


Dive into the research topics where James J. A. Contos is active.

Publication


Featured researches published by James J. A. Contos.


Nature | 2005

LPA3-mediated lysophosphatidic acid signalling in embryo implantation and spacing

Xiaoqin Ye; Kotaro Hama; James J. A. Contos; Brigitte Anliker; Asuka Inoue; Michael K. Skinner; Hiroshi Suzuki; Tomokazu Amano; Grace Kennedy; Hiroyuki Arai; Junken Aoki; Jerold Chun

Every successful pregnancy requires proper embryo implantation. Low implantation rate is a major problem during infertility treatments using assisted reproductive technologies. Here we report a newly discovered molecular influence on implantation through the lysophosphatidic acid (LPA) receptor LPA3 (refs 2–4). Targeted deletion of LPA3 in mice resulted in significantly reduced litter size, which could be attributed to delayed implantation and altered embryo spacing. These two events led to delayed embryonic development, hypertrophic placentas shared by multiple embryos and embryonic death. An enzyme demonstrated to influence implantation, cyclooxygenase 2 (COX2) (ref. 5), was downregulated in LPA3-deficient uteri during pre-implantation. Downregulation of COX2 led to reduced levels of prostaglandins E2 and I2 (PGE2 and PGI2), which are critical for implantation. Exogenous administration of PGE2 or carbaprostacyclin (a stable analogue of PGI2) into LPA3-deficient female mice rescued delayed implantation but did not rescue defects in embryo spacing. These data identify LPA3 receptor-mediated signalling as having an influence on implantation, and further indicate linkage between LPA signalling and prostaglandin biosynthesis.


Nature Medicine | 2004

Initiation of neuropathic pain requires lysophosphatidic acid receptor signaling.

Makoto Inoue; Harunor Rashid; Ryousuke Fujita; James J. A. Contos; Jerold Chun; Hiroshi Ueda

Lysophosphatidic acid (LPA) is a bioactive lipid with activity in the nervous system mediated by G-protein-coupled receptors. Here, we examined the role of LPA signaling in the development of neuropathic pain by pharmacological and genetic approaches, including the use of mice lacking the LPA1 receptor. Wild-type animals with nerve injury develop behavioral allodynia and hyperalgesia paralleled by demyelination in the dorsal root and increased expression of both the protein kinase C γ-isoform within the spinal cord dorsal horn and the α2δ1 calcium channel subunit in dorsal root ganglia. Intrathecal injection of LPA induced behavioral, morphological and biochemical changes similar to those observed after nerve ligation. In contrast, mice lacking a single LPA receptor (LPA1, also known as EDG2) that activates the Rho–Rho kinase pathway do not develop signs of neuropathic pain after peripheral nerve injury. Inhibitors of Rho and Rho kinase also prevented these signs of neuropathic pain. These results imply that receptor-mediated LPA signaling is crucial in the initiation of neuropathic pain.


Nature Neuroscience | 2003

Non-proliferative effects of lysophosphatidic acid enhance cortical growth and folding

Marcy A. Kingsbury; Stevens Kastrup Rehen; James J. A. Contos; Christine M Higgins; Jerold Chun

Lysophosphatidic acid (LPA) is a phospholipid that has extracellular signaling properties mediated by G protein–coupled receptors. Two LPA receptors, LPA1 and LPA2, are expressed in the embryonic cerebral cortex, suggesting roles for LPA signaling in cortical formation. Here we report that intact cerebral cortices exposed to extracellular LPA ex vivo rapidly increased in width and produced folds resembling gyri, which are not normally present in mouse brains and are absent in LPA1 LPA2 double-null mice. Mechanistically, growth was not due to increased proliferation but rather to receptor-dependent reduced cell death and increased terminal mitosis of neural progenitor cells (NPCs). Our results implicate extracellular lipid signals as new influences on brain formation during embryonic development.


Cell Biochemistry and Biophysics | 1999

A growing family of receptor genes for lysophosphatidic acid (LPA) and other lysophospholipids (LPs)

Jerold Chun; James J. A. Contos; Donald Munroe

A missing component in the experimental analysis of cell signaling by extracellular lysophospholipids such as lysophosphatidic acid (LPA) or sphingosine-1-phosphate (S1P) has been cloned receptors. Through studies on the developing brain, the first such receptor gene (referred to asvzg-1) was identified, representing a member of the G-protein coupled receptor (GPCR) super family(1). Here we review the neurobiological approach that led to both its cloning and identification as a receptor for LPA, along with related expression data. Summarized sequence and genomic structure analyses indicate that this first, functionally identified receptor is encoded by a member of a growing gene family that divides into at least two subgroups: genes most homologous to the high-affinity LPA receptor encoded byvzg-1, and those more homologous to an orphan receptor geneedg-1 that has recently been identified as a S1P receptor. A provisional nomenclature is proposed, based on published functional ligand actions, amino acid composition and genomic structure whereby the receptors encoded by these genes are referred to as lysophospholipid (LP) receptors, with subgroups distinguished by letter and number subscripts (e.g., LPA1 for Vzg-1, and LPB1 for Edg-1). Presented expression data support the recently published work indicating that members of the LPB1 subgroup are receptors for the structurally-related molecule, S1P. The availability of cloned LP receptors will enhance the analysis of the many documented LP effects, while their prominent expression in the nervous system indicates significant but as yet unknown roles in development, normal function, and neuropathology.


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

Formyl peptide receptors are candidate chemosensory receptors in the vomeronasal organ

Stephen D. Liberles; Lisa F. Horowitz; Donghui Kuang; James J. A. Contos; Kathleen L. Wilson; Jessica Siltberg-Liberles; David A. Liberles; Linda B. Buck

The identification of receptors that detect environmental stimuli lays a foundation for exploring the mechanisms and neural circuits underlying sensation. The mouse vomeronasal organ (VNO), which detects pheromones and other semiochemicals, has 2 known families of chemoreceptors, V1Rs and V2Rs. Here, we report a third family of mouse VNO receptors comprising 5 of 7 members of the formyl peptide receptor (FPR) family. Unlike other FPRs, which function in the immune system, these FPRs are selectively expressed in VNO neurons in patterns strikingly similar to those of V1Rs and V2Rs. Each FPR is expressed in a different small subset of neurons that are highly dispersed in the neuroepithelium, consistently coexpress either Gαi2 or Gαo, and lack other chemoreceptors examined. Given the presence of formylated peptides in bacteria and mitochondria, possible roles for VNO FPRs include the assessment of conspecifics or other species based on variations in normal bacterial flora or mitochondrial proteins.


Gene | 1999

Comparative analysis of three murine G-protein coupled receptors activated by sphingosine-1-phosphate.

Guangfa Zhang; James J. A. Contos; Joshua A. Weiner; Nobuyuki Fukushima; Jerold Chun

The cloning and analysis of the first identified lysophosphatidic acid (LPA) receptor gene, lpA1 (also referred to as vzg-1 or edg-2), led us to identify homologous murine genes that might also encode receptors for related lysophospholipid ligands. Three murine genomic clones (designated lpB1, lpB2, and lpB3) were isolated, corresponding to human/rat Edg-1, rat H218/AGR16, and human edg-3, respectively. Based on the amino acid similarities of their predicted proteins (44-52% identical), the three lpB genes could be grouped into a separate G-protein coupled receptor subfamily, distinct from that containing the LPA receptor genes lpA1 and lpA2. Unlike lpA1 and lpA2, which contain multiple coding exons, all lpB members contained a single coding exon. Heterologous expression of individual lpB members in a hepatoma cell line (RH7777), followed by 35S-GTPgammaS incorporation assays demonstrated that each of the three LPB receptors conferred sphingosine-1-phosphate-dependent, but not lysophosphatidic acid-dependent, G-protein activation. Northern blot and in situ hybridization analyses revealed overlapping as well as distinct expression patterns in both embryonic and adult tissues. This comparative characterization of multiple sphingosine-1-phosphate receptor genes and their spatiotemporal expression patterns will aid in understanding the biological roles of this enlarging lysophospholipid receptor family.


Molecular and Cellular Neuroscience | 2002

Lysophosphatidic Acid Influences the Morphology and Motility of Young, Postmitotic Cortical Neurons

Nobuyuki Fukushima; Joshua A. Weiner; Dhruv Kaushal; James J. A. Contos; Stevens Kastrup Rehen; Marcy A. Kingsbury; Kyung Yong Kim; Jerold Chun

Lysophosphatidic acid (LPA) is a bioactive lysophospholipid that produces process retraction and cell rounding through its cognate receptors in neuroblastoma cell lines. Although the expression profile of LPA receptors in developing brains suggests a role for LPA in central nervous system (CNS) development, how LPA influences the morphology of postmitotic CNS neurons remains to be determined. Here we have investigated the effects of exogenous LPA on the morphology of young, postmitotic neurons in primary culture. When treated with LPA, these neurons responded by not only retracting processes but also producing retraction fiber caps characterized by fine actin filaments emanating from a dense core. Retraction fiber caps gradually vanished due to the outward spread of regrowing membranes along the fibers, suggesting a role for caps as scaffolds for regrowth of retracted processes. Furthermore, LPA also affects neuronal migration in vitro and in vivo. Taken together, these results implicate LPA as an extracellular lipid signal affecting process outgrowth and migration of early postmitotic neurons during development.


FEBS Letters | 2002

Embryonic brain expression analysis of lysophospholipid receptor genes suggests roles for s1p1 in neurogenesis and s1p1–3 in angiogenesis

Christine McGiffert; James J. A. Contos; Beth Friedman; Jerold Chun

In a comparison of embryonic brain expression patterns of lysophosphatidic acid and sphingosine 1‐phosphate receptor genes (lpa1–3 and s1p1–5 , respectively), transcripts detected by Northern blot were subsequently localized using in situ hybridization. We found striking s1p1 expression adjacent to several ventricles. Near the lateral ventricle, s1p1 expression was temporally and spatially coincident with neurogenesis and overlapped with lpa1 in the neocortical area. We also observed a widespread diffuse pattern for lpa2–3 and a scattered punctate pattern for s1p1–3 . The punctate pattern colocalized with vascular endothelial markers. Together, these results suggest that s1p1 influences neurogenesis and s1p1–3 influence angiogenesis in the developing brain.


Gene | 2001

The mouse lpA3/Edg7 lysophosphatidic acid receptor gene: genomic structure, chromosomal localization, and expression pattern

James J. A. Contos; Jerold Chun

The extracellular signaling molecule, lysophosphatidic acid (LPA), mediates proliferative and morphological effects on cells and has been proposed to be involved in several biological processes including neuronal development, wound healing, and cancer progression. Three mammalian G protein-coupled receptors, encoded by genes designated lp (lysophospholipid) receptor or edg (endothelial differentiation gene), mediate the effects of LPA, activating similar (e.g. Ca(2+) release) as well as distinct (neurite retraction) responses. To understand the evolution and function of LPA receptor genes, we characterized lp(A3)/Edg7 in mouse and human and compared the expression pattern with the other two known LPA receptor genes (lp(A1)/Edg2 and lp(A2)/Edg4non-mutant). We found mouse and human lp(A3) to have nearly identical three-exon genomic structures, with introns upstream of the coding region for transmembrane domain (TMD) I and within the coding region for TMD VI. This structure is similar to lp(A1) and lp(A2), indicating a common ancestral gene with two introns. We localized mouse lp(A3) to distal Chromosome 3 near the varitint waddler (Va) gene, in a region syntenic with the human lp(A3) chromosomal location (1p22.3-31.1). We found highest expression levels of each of the three LPA receptor genes in adult mouse testes, relatively high expression levels of lp(A2) and lp(A3) in kidney, and moderate expression of lp(A2) and lp(A3) in lung. All lp(A) transcripts were expressed during brain development, with lp(A1) and lp(A2) transcripts expressed during the embryonic neurogenic period, and lp(A3) transcript during the early postnatal period. Our results indicate both overlapping as well as distinct functions of lp(A1), lp(A2), and lp(A3).


Annals of the New York Academy of Sciences | 2006

Neurobiology of receptor-mediated lysophospholipid signaling. From the first lysophospholipid receptor to roles in nervous system function and development

Jerold Chun; Joshua A. Weiner; Nobuyuki Fukushima; James J. A. Contos; Guangfa Zhang; Yuka Kimura; Adrienne E. Dubin; Isao Ishii; Jonathan H. Hecht; Carol Akita; Dhruv Kaushal

Abstract: Identification of the first lysophospholipid receptor, LPA1/Vzg‐1, cloned by way of neurobiological analyses on the embryonic cerebral cortex, has led to the realization and demonstration that there exist multiple, homologous LP receptors, including those encoded by a number of orphan receptor genes known as “Edg,” all of which are members of the G‐protein‐coupled receptor (GPCR) superfamily. These receptors interact with apparent high affinity for lysophosphatidic acid (LPA) or sphingosine‐1‐phosphate (S1P or SPP), and are referred to based upon their functional identity as lysophospholipid receptors: LPA and LPB receptors, respectively, with the expectation that additional subgroups will be identified (i.e., LPC, etc.). Here an update is provided on insights gained from analyses of these receptor genes as they relate to the nervous system, particularly the cerebral cortex, and myelinating cells (oligodendrocytes and Schwann cells).

Collaboration


Dive into the James J. A. Contos's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Isao Ishii

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marcy A. Kingsbury

Indiana University Bloomington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guangfa Zhang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Beth Friedman

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroshi Ueda

Tokyo Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge