Network


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

Hotspot


Dive into the research topics where Manohar Ratnam is active.

Publication


Featured researches published by Manohar Ratnam.


Cancer | 1994

Differential regulation of folate receptor isoforms in normal and malignant tissues in vivo and in established cell lines: Physiologic and clinical implications

F B S John Ross; Prabir K. Chaudhuri; Manohar Ratnam

Background. Despite significant differences in ligand binding between the two known isoforms of the human membrane folate receptor (FR), designated herein as FR β (placenta) and FR‐α (placenta, KB cells), little is known about their tissue specificities, and there is no report on the relative expression of FR‐β in any tissue other than in placenta.


Arthritis & Rheumatism | 1999

Selective expression of folate receptor β and its possible role in methotrexate transport in synovial macrophages from patients with rheumatoid arthritis

Noriko Nakashima-Matsushita; Toshio Homma; Su Yu; Takemasa Matsuda; Nobuhiko Sunahara; Tadashi Nakamura; Michishi Tsukano; Manohar Ratnam; Takami Matsuyama

OBJECTIVEnTo investigate the expression of folate receptors (FR) and reduced folate carrier (RFC) and determine their relevance to methotrexate (MTX) transport in synovial mononuclear cells (SMC) from patients with rheumatoid arthritis (RA).nnnMETHODSnLevels of FR and RFC messenger RNA (mRNA) were examined by reverse transcriptase-polymerase chain reaction (RT-PCR) in SMC from RA patients and peripheral blood mononuclear cells from healthy donors. Expression of FR-beta mRNA and protein was determined by Northern blot and Western blot analyses in RA SMC and monocyte/macrophage-lineage cells. FR-beta expression and folic acid binding capacity on the cell surface were examined by flow cytometric analysis and 3H-folic acid binding analysis. Studies of the inhibition of 3H-MTX uptake in the presence of unlabeled folic acid were performed to investigate the uptake of MTX through FR in RA SMC.nnnRESULTSnRT-PCR, Northern blot, and Western blot analyses showed that FR-beta mRNA and protein were expressed selectively in activated monocytes and CD14+ RA SMC. These cells exhibited folic acid binding capacity. Furthermore, the FR-beta protein was shown to have folic acid binding capacity. Uptake of 3H-MTX through RA SMC was significantly inhibited in the presence of unlabeled folic acid.nnnCONCLUSIONnThese results demonstrate that FR-beta expression is selectively elevated in RA synovial macrophages and suggest that MTX is transported through FR-beta in RA synovial macrophages. The findings suggest that folate antagonists with higher affinity for FR-beta would be useful in the treatment of RA.


Biochemical Pharmacology | 1992

Differential stereospecificities and affinities of folate receptor isoforms for folate compounds and antifolates

Wang Xin; Shen Feng; James H. Freisheim; Larry E. Gentry; Manohar Ratnam

Two membrane folate receptor (MFR) isoforms are present in human tissues i.e. MFR-1 (e.g. placenta) and MFR-2 (e.g. placenta, KB cells, CaCo-2 cells). MFR-1 was expressed in COS-1 cells and the resulting protein had the same polypeptide molecular weight as the native protein. The affinities of (6S) and (6R) diastereoisomers of N5-methyltetrahydrofolate, N5-formyltetrahydrofolate, and 5,10-dideazatetrahydrofolate as well as folic acid and methotrexate to MFR-1, MFR-2 and placental MFR (MFR-1 plus MFR-2) were determined in terms of the Ki values for their competitive inhibition of the binding of [3H]folic acid to these proteins. The results indicated a striking difference in the stereospecificity of MFR-1 and MFR-2 for reduced folate coenzymes; MFR-2 preferentially bound to the physiological (6S) diastereoisomers and MFR-1 bound preferentially to the unphysiological (6R) diastereoisomers, while dideazatetrahydrofolate did not show significant stereospecificity for MFR-1. Furthermore, MFR-2 displayed significantly (2- to 100-fold) greater affinities for all the compounds tested compared to MFR-1. Purified placental MFR, a natural source of MFR-1 which contains variable amounts of MFR-2, showed intermediate Ki values for the compounds tested compared with MFR-1 and MFR-2 and stereospecificities similar to MFR-1. These observations demonstrate striking differences in the ligand binding sites of MFR-1 and MFR-2 which could potentially be exploited in the design of MFR isoform specific antifolates.


Journal of Biological Chemistry | 1999

Complete mapping of divergent amino acids responsible for differential ligand binding of folate receptors alpha and beta.

Karen Maziarz; Hugo L. Monaco; Feng Shen; Manohar Ratnam

The folate receptor (FR) type α may be distinguished from FR-β by its higher affinity for the circulating folate coenzyme, (6S)-5-methyltetrahydrofolate (5-CH3H4folate), and its opposite stereospecificity for reduced folate coenzymes. Previous studies showed that a single leucine to alanine substitution at position 49 of the mature protein sequence is responsible for the functional divergence of FR-β (Shen, F., Zheng, X., Wang, H., and Ratnam, M. (1997)Biochemistry 36, 6157–6163); however, the results also indicated that the minimum requirement for conversion of FR-β to the functional equivalent of FR-α should include amino acid substitution(s) downstream of residue 92 in addition to mutation of L49A. To pinpoint those residues, chimeric FR-βL49A/FR-α constructs including progressively shorter segments of FR-α downstream of position 92 as well as selected point mutants were studied. Simultaneous substitution of Leu-49, Phe-104, and Gly-166 in FR-β with the corresponding FR-α residues Ala, Val, and Glu, respectively, reconstituted the ligand binding characteristics of FR-α. The results also exclude a role for other residues in FR-α in determining its functional divergence. A homology model of FR-α based on the three-dimensional structure of the chicken riboflavin-binding protein is used to show the position of residues 49, 104, and 166 in relation to the hydrophobic cleft corresponding to the riboflavin-binding pocket.


The Journal of Membrane Biology | 2002

Evidence for segregation of heterologous GPI-anchored proteins into separate lipid rafts within the plasma membrane.

J. Wang; William T. Gunning; K.M.M. Kelley; Manohar Ratnam

Cholesterol and glycosphingolipid-rich membrane rafts, which are rich in GPI-anchored proteins and are distinct from caveolae, are believed to serve as platforms for signal transduction events and protein recycling. GPI-anchored proteins with diverse functions as well as caveolin may be recovered in a membrane fraction insoluble in cold non-ionic detergent. This study tests for possible heterogeneity in the protein composition of the lipid rafts and detergent-insoluble membrane complexes by examining the two GPI-anchored homologous human folate receptors (FR)-a and -b, the GPI-anchored human placental alkaline phosphatase (PLAP), and caveolin (control) in transfected CHO cells. Both FR and PLAP showed the equal distribution of cell-surface vs. sequestered (recycling) protein typical of GPI-proteins. Quantitative affinity purification of detergent-insoluble complexes using biotinylated folate or specific antibodies demonstrated a strong association of the homologous FR-a and FR-b in the same detergent-insoluble complex and separate complexes containing either PLAP or caveolin. Immunogold localization experiments using antibody crosslinking to produce larger aggregates of GPI-anchored proteins for visualization by electron microscopy also showed a clear separation between FR- and PLAP-rich membrane microdomains. Thus, even though functionally diverse and heterologous GPI-anchored proteins are known to share endocytic and recycling vesicles, they may be segregated in distinct lipid rafts on the basis of their ecto(protein) domains facilitating clustering, compartmentalization and homotypic protein interactions.n


Advances in Enzyme Regulation | 1992

Molecular events in the membrane transport of methotrexate in human CCRF-CEM leukemia cell lines.

James H. Freisheim; Manohar Ratnam; Terence P. McAlinden; K.M.R. Prasad; Frederick E. Williams; G. Robbin Westerhof; Gerrit Jansen

A variant line (CEM-7A) overproducing the reduced folate/MTX carrier system was isolated from human CCRF-CEM leukemia cells grown under selective conditions in medium containing 0.25 nM 5-formyl-THF as the sole folate source. This line exhibits a 95-fold increased Vmax for [3H]-MTX influx as compared to parental cells. The values for [3H]-MTX influx Km, efflux t1/2 and structural specificity for other (anti)folate compounds were unchanged. The amount of carrier protein, estimated by NHS-[3H]-MTX affinity labeling, was approximately 30-fold higher in CEM-7A cells than in parental cells. Influx of [3H]-MTX in CEM-7A cells was found to be down-regulated 6-7-fold after preincubation of cells with adenosine, 5-formyl-THF or 5-methyl-THF, but could be prevented exclusively by inhibitors of dihydrofolate reductase. The underlying mechanism(s) of these effects have not as yet been elucidated. A radioiodinated photoaffinity analog of MTX was used to prove the molecular events in carrier-mediated MTX uptake in parental CCRF-CEM cells, CEM-7A cells, and a line exhibiting a MTX-transport defect (CEM-MTX). Specific labeling of an 80-85 kDa membrane protein was observed in parental cells, but not in CEM/MTX cells. Uptake of photoprobe and levels of the 80-85 kDa membrane protein were significantly increased in CEM-7A cells. Due to extensive glycosylation the MW of the carrier protein in human cells seems to be substantially higher than that of its counterpart in murine L1210 leukemia cells (46-48 kDa). Pulse-labeling experiments at 37 degrees C demonstrated that in CEM-7A cells photoprobe uptake proceeds via a specific pathway. The 80-85 kDa membrane protein is involved in the initial binding and translocation of photoprobe, after which a 38 kDa cytosolic protein is responsible for further intracellular distribution. At this time, the combination of photoaffinity labeling techniques and the availability of variant cell lines overexpressing the reduced folate/MTX carrier protein has provided new insights into the MTX transport process in human leukemia cell lines. In the near future this approach should also allow a further elucidation of the regulatory aspects of carrier function.


Cancer Chemotherapy and Pharmacology | 1996

Functional aspects of membrane folate receptors in human breast cancer cells with transport-related resistance to methotrexate.

Marie-France Pinard; Jacques Jolivet; Manohar Ratnam; Ietje Kathmann; Carla F. M. Molthoff; Robbin Westerhof; Gerrit Jansen

Abstractu2002Two methotrexate (MTX)-resistant human breast-cancer cell lines with impaired transport via the reduced folate carrier (RFC), one established in vitro (MTXR-ZR-75-1) and another inherently resistant (MDA-231), were adapted to grow in medium containing 2u2005nM folic acid. This induced the expression of previously undetectable membrane folate receptors (MFR) to levels of 8.2 and 2.3u2005pmol/107 cells, respectively. Polymerase chain reaction (PCR) quantitation revealed that MFR messenger-RNA levels of the isoform first described in human nasopharyngeal carcinoma KB cells (MFR-α) were increased in low-folate-adapted MTXR-ZR-75-1 cells, whereas placental transcripts (MFR-β) coincided with MFR-α expression in low-folate (LF)-adapted MDA-231 cells. These cell lines were used to study the role of MFR in the uptake and growth-inhibitory effects of five different antifolates with varying affinities for MFR: N10-propargyl-5, 8-dideazafolic acid (CB3717) >u20095,10-dideazatetra-hydrofolic acid (DDATHF) >u2009N-{5- [N-(3,4-di-hydro-2-methyl-4-oxoquinazolin-6-methyl)-N-methyl-amino] -2-theonyl}-glutamic acid (ZD1694) ≫ MTX > edatrexate (EDX). Expression of MFR only slightly decreased the resistant phenotype for MTX, EDX, and ZD1694, suggesting that these drugs are not transported intracellularly to cytotoxic concentrations at these levels of MFR expression. On the other hand, both cell lines became from at least 180- to 400-fold more sensitive to growth inhibition by CB3717 and DDATHF, which may be correlated with their high affinity for MFR. These sensitivity/resistance profiles were largely similar following cell culture in medium containing 1u2005nML-leucovorin, a folate with an affinity for MFR 10-fold lower than that of folic acid, the one exception being the increased sensitivity for ZD1694 seen in the LF-adapted cells with the highest level of MFR expression (MTXR-ZR-75-1). These results illustrate that the efficacy of MFR in mediating antifolate transport and cytotoxicity depends on their affinity for the folate antagonist, their degree of expression, and the levels of competing folates.


Advances in Enzyme Regulation | 1989

Folate coenzyme and antifolate transport proteins in normal and neoplastic cells

James H. Freisheim; Elmer M. Price; Manohar Ratnam

The transport systems for folate coenzymes and antifolate compounds into various types of normal and neoplastic cells display considerable diversity in their pharmacokinetics and also in terms of the apparent molecular weights of the proteins involved. Further, several uptake routes may exist in a given cell type. A variety of neoplastic tissues have been reported to rely upon a single major transport system that has a relatively high affinity for the reduced form of folate compounds and for antifolates such as methotrexate. Using a photoaffinity analogue of methotrexate, we have identified the involvement of a 48 kDa membrane protein and a 38 kDa cytosolic or peripheral membrane protein in the transport of this compound into murine L1210 leukemia cells. Such an uptake is absent in mutant L1210 cells that are defective in methotrexate transport. We propose a model for the uptake of reduced folate coenzymes in L1210 cells in which the compound is initially transported across the cell membrane by the 48 kDa protein and delivered on the cytoplasmic surface to the 38 kDa protein; the 38 kDa protein then carries the folate compound to a specific enzyme of folate metabolism. Antibodies to the membrane folate binding protein from human placenta cross-react with the 48 kDa protein in L1210 cell membranes indicating an immunological relationship between these two proteins. Comparison of the amino acid sequences of peptides of the placental receptor obtained by digestion with S. aureus V8 protease indicate the presence of two homologous forms of the folate binding protein in placenta; one of these forms appears to have an identical sequence to the soluble and membrane associated folate binding proteins in human epidermoid carcinoma (KB) cells, which in turn share the primary structure of the soluble and membrane associated folate binders in human milk in the regions that have been sequenced. These results indicate that the folate coenzyme transport proteins in various tissues may be structurally related and in several instances may even be identical. In the latter cases the observed differences in apparent molecular weights may be due to differences in glycosylation and/or proteolysis. In support of this view is our observation that the deglycosylated and/or partially proteolyzed placental receptor retains the ability to bind folate.


Gene | 2003

Dual regulation of ets-activated gene expression by SP1.

Karen M.M. Kelley; Huiquan Wang; Manohar Ratnam

The human folate receptor (hFR) type gamma gene is driven by a TATA-less promoter that uses a canonical Sp1 element for basal transcription. Using nuclear extract from 293 (human embryonic) cells, we mapped a second (non-canonical) Sp1 element to which Sp1 bound with a comparable affinity and which overlaps a functional ets binding site (EBS). Mutagenesis experiments revealed that the binding of ets to the EBS activates the promoter synergistically with Sp1 bound at the downstream site; however, binding of Sp1 to the EBS does not contribute to promoter activity. A further increase in Sp1 by inducible expression in recombinant 293 cells resulted in a small but significant decrease in the hFR-gamma promoter activity, but the decrease was abolished when the EBS was deleted from the promoter. In 293 cells, which do not express hFR-gamma, the Sp1 level was relatively high whereas in the hFR-gamma-positive HL60 leukemia cells, the Sp1 level was low and the EBS predominantly bound an ets protein. To account for the above observations, we propose a model in which when the Sp1 level is low, ets out competes Sp1 for binding to the EBS and synergistically enhances the hFR-gamma promoter activity by interacting with Sp1 bound at the canonical site whereas at higher levels, Sp1 represses the promoter by competitively inhibiting the binding of ets. As a partial extension of this model to the regulation of other ets activated genes, we show that Sp1 can predictably bind to a variety of ets elements including those responsive to Ets1 and Spi.1/Pu.1. A dual concentration-dependent action of Sp1 as an activator or a repressor offers a potential mechanism contributing to tissue-specific regulation of ets-dependent genes by Sp1.


Bioorganic & Medicinal Chemistry Letters | 1996

Synthesis and biological evaluation of a new series of dihydrofolate reductase inhibitors based on the 4-(2,6-diamino-5-pyrimidinyl)alkyl-L-glutamic acid structure

Lynn S. Gossett; Lillian L. Habeck; Susan B. Gates; Sherri L. Andis; John F. Worzalla; Richard M. Schultz; Laurane G. Mendelsonn; William Kohler; Manohar Ratnam; Gerald B. Grindey; Chuan Shih

Abstract A novel series of dihydrofolate reductase inhibitors was uncovered during an expansion of the SAR of 5,10-dideazatetrahydrofolic acid, and their biological activity was evaluated. These new analogs do not possess an oxygen at the 4 position and contain a monocyclic pyrimidine ring.

Collaboration


Dive into the Manohar Ratnam's collaboration.

Top Co-Authors

Avatar

James H. Freisheim

University of Toledo Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gerrit Jansen

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Feng Shen

University of Toledo Medical Center

View shared research outputs
Top Co-Authors

Avatar

Frederick E. Williams

University of Toledo Medical Center

View shared research outputs
Top Co-Authors

Avatar

Huiquan Wang

University of Toledo Medical Center

View shared research outputs
Top Co-Authors

Avatar

Karen Maziarz

University of Toledo Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jianyong Wang

University of Toledo Medical Center

View shared research outputs
Top Co-Authors

Avatar

K.M.R. Prasad

University of Toledo Medical Center

View shared research outputs
Top Co-Authors

Avatar

Terence P. McAlinden

University of Toledo Medical Center

View shared research outputs
Top Co-Authors

Avatar

Bridget Dillon

University of Toledo Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge