Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Equine Vet J ; 56(4): 735-741, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38268098

ABSTRACT

BACKGROUND: Management of twin pregnancy after conceptus vesicle fixation in the horse is challenging because the reduction techniques described are either invasive, difficult to perform or associated with disappointing success rates. OBJECTIVES: To evaluate the success of transrectal ultrasound-guided fetal thorax compression for reducing post-fixation twin pregnancy in mares. STUDY DESIGN: Retrospective clinical study. METHODS: Sixteen mares were presented for twin reduction between 51 and 79 days of gestation. History obtained from the owner and/or referring veterinarian detailed information regarding the mare (age, breed), pregnancy (day of gestation, dizygotic versus monozygotic twins, unilateral versus bilateral fixation), treatment and outcome (one live fetus at discharge; live singleton at foaling) after twin reduction. Transrectal fetal thorax compression was performed under ultrasound guidance by two experienced operators. RESULTS: Overall 9 of 16 twin pregnancies were successfully reduced and the likelihood of success was significantly higher in dizygotic than monozygotic twins. The procedure was successful in 9 of 10 dizygotic twins but unsuccessful in all six cases of monozygotic twins. Among the dizygotic twins, two mares lost the pregnancy after discharge from the clinic, seven mares delivered a healthy foal of normal size. MAIN LIMITATIONS: Small case number. CONCLUSIONS: Transrectal ultrasound-guided fetal thorax compression is a minimally-invasive and successful technique for reducing dizygotic twin pregnancies at approximately 2 months of gestation, but does not lead to any live births in cases of monozygotic twins.


Subject(s)
Pregnancy Reduction, Multifetal , Thorax , Horses , Animals , Female , Pregnancy , Retrospective Studies , Thorax/diagnostic imaging , Pregnancy Reduction, Multifetal/veterinary , Pregnancy Reduction, Multifetal/methods , Ultrasonography, Prenatal/veterinary
2.
Vet Clin North Am Equine Pract ; 32(3): 451-464, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27726984

ABSTRACT

Anti-Müllerian hormone (AMH) plays a major role in sexual differentiation, Leydig cell differentiation, and folliculogenesis. In addition, AMH has clinical value in equine practice. In stallions, AMH can serve as an endocrine marker for equine cryptorchidism and as an immunohistochemical marker for Sertoli cell tumors. Considering that AMH is also an ovarian specific product, intact mares can be differentiated from ovariectomized mares. Peripheral AMH concentrations reflect the follicular population in mares, and therefore, are useful in the assessment of ovarian reserve and reproductive life-span of aged mares. Last, AMH is particularly suitable as a diagnostic marker for equine granulosa cell tumors.


Subject(s)
Anti-Mullerian Hormone/metabolism , Horses/physiology , Animals , Anti-Mullerian Hormone/blood , Biomarkers/blood , Estrous Cycle/physiology , Female , Horse Diseases/blood , Male , Ovarian Follicle/physiology
3.
J Biol Chem ; 276(27): 24457-65, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11274181

ABSTRACT

We have identified and characterized potent and specific inhibitors of geranylgeranyl-protein transferase type I (GGPTase I), as well as dual inhibitors of GGPTase I and farnesyl-protein transferase. Many of these inhibitors require the presence of phosphate anions for maximum activity against GGPTase I in vitro. Inhibitors with a strong anion dependence were competitive with geranylgeranyl pyrophosphate (GGPP), rather than with the peptide substrate, which had served as the original template for inhibitor design. One of the most effective anions was ATP, which at low millimolar concentrations increased the potency of GGPTase I inhibitors up to several hundred-fold. In the case of clinical candidate l-778,123, this increase in potency was shown to result from two major interactions: competitive binding of inhibitor and GGPP, and competitive binding of ATP and GGPP. At 5 mm, ATP caused an increase in the apparent K(d) for the GGPP-GGPTase I interaction from 20 pm to 4 nm, resulting in correspondingly tighter inhibitor binding. A subset of very potent GGPP-competitive inhibitors displayed slow tight binding to GGPTase I with apparent on and off rates on the order of 10(6) m(-)1 s(-)1 and 10(-)3 s(-)1, respectively. Slow binding and the anion requirement suggest that these inhibitors may act as transition state analogs. After accounting for anion requirement, slow binding, and mechanism of competition, the structure-activity relationship determined in vitro correlated well with the inhibition of processing of GGPTase I substrate Rap1a in vivo.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Anions/metabolism , Enzyme Inhibitors/pharmacology , Adenosine Triphosphate/metabolism , Binding, Competitive , Humans , Imidazoles/pharmacology , Kinetics , Models, Chemical , Polyisoprenyl Phosphates/metabolism , Protein Binding , Structure-Activity Relationship
4.
J Med Chem ; 42(17): 3356-68, 1999 Aug 26.
Article in English | MEDLINE | ID: mdl-10464022

ABSTRACT

Inhibitors of farnesyl protein transferase (FPTase) based upon a pseudotripeptide template are described that comprise an imidazole group substituted with a hydrophobic substituent. (1, 5)-Disubstitution of the imidazole group is shown to be the optimal array that leads to potent and selective inhibitors of FPTase. A variety of aryl and isoprenyl substituents are shown to afford effective inhibitors, and the mechanism by which these compounds inhibit FPTase has been investigated. The biochemical behavior of these compounds suggests that they bind to FPTase at the site usually occupied by the protein substrate. In experiments in cell culture, the methyl ester prodrugs of these inhibitors are cell permeant and potently inhibit the posttranslational modification of H-Ras protein. Additionally, these molecules revert the phenotype of ras transformed cells as evidenced by their ability to slow the growth of ras transformed cell lines in soft agar. One of the inhibitors, as its methyl prodrug, was evaluated in two in vivo models of tumor growth. The compound selectively inhibited the growth of tumors derived from H-ras transformed cells, in nude mice, and caused the regression of preexisting tumors in an H-ras transgenic animal model.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Imidazoles/chemical synthesis , 3T3 Cells , Alkyl and Aryl Transferases/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Transformed , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Imidazoles/chemistry , Imidazoles/pharmacology , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Transplantation , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Structure-Activity Relationship
5.
Biofactors ; 6(3): 359-66, 1997.
Article in English | MEDLINE | ID: mdl-9288406

ABSTRACT

Ras, a signal-transducing protein involved in mediating growth factor-stimulated proliferation, is mutationally activated in over 30% of human tumors. To be functional Ras must bind to the inner surface of the plasma membrane, with post-translational lipid modifications being necessary for this localization. The essential, first modification of Ras is farnesylation catalyzed by the enzyme farnesyl: proteintransferase (FPTase). Inhibitors of FPTase (FTIs) are currently being tested to determine if they are capable of tumor growth inhibition. Here we describe our efforts, along with those of other groups, in testing the biological and biochemical effects of FTIs.


Subject(s)
Alkyl and Aryl Transferases , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Transferases/antagonists & inhibitors , Animals , Genes, ras , Humans , Neoplasms/pathology , Protein Prenylation , Transferases/chemistry , Transferases/metabolism , Tumor Cells, Cultured , ras Proteins/metabolism
7.
Breast Cancer Res Treat ; 38(1): 75-83, 1996.
Article in English | MEDLINE | ID: mdl-8825125

ABSTRACT

The oncoprotein encoded by mutant ras genes is initially synthesized as a cytoplasmic precursor which requires posttranslational processing to attain biological activity; farnesylation of the cysteine residue present in the CaaX motif located at the carboxy-terminus of all Ras proteins is the critical modification. Once farnesylated and further modified, the mature Ras protein is inserted into the cell's plasma membrane where it participates in the signal transduction pathways that control cell growth and differentiation. The farnesylation reaction that modifies Ras and other cellular proteins having an appropriate CaaX motif is catalyzed by a housekeeping enzyme termed farnesyl-protein transferase (FPTase). Inhibitors of this enzyme have been prepared by several laboratories in an effort to identify compounds that would block Ras-induced cell transformation and thereby function as Ras-specific anticancer agents. A variety of natural products and synthetic organic compounds were found to block farnesylation of Ras proteins in vitro. Some of these compounds exhibit antiproliferative activity in cell culture, block the morphological alterations associated with Ras-transformation, and can block the growth of Ras-transformed cell lines in tumor colony-forming assays. By contrast, these compounds do not affect the growth or morphology of cells transformed by the Raf or Mos oncoproteins, which do not require farnesylation to achieve biological activity. The efficacy and lack of toxicity observed with FPTase inhibitors in an animal tumor model suggest that specific FPTase inhibitors may be useful for the treatment of some types of cancer.


Subject(s)
Alkyl and Aryl Transferases , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Transferases/antagonists & inhibitors , ras Proteins/antagonists & inhibitors , Animals , Farnesyltranstransferase , Guanosine Triphosphate/metabolism , Humans
8.
Nat Med ; 1(8): 792-7, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7585182

ABSTRACT

For Ras oncoproteins to transform mammalian cells, they must be post-translationally modified with a farnesyl group in a reaction catalysed by the enzyme farnesyl-protein transferase (FPTase). Inhibitors of FPTase have therefore been proposed as anti-cancer agents. We show that L-744,832, which mimics the CaaX motif to which the farnesyl group is added, is a potent and selective inhibitor of FPTase. In MMTV-v-Ha-ras mice bearing palpable tumours, daily administration of L-744,832 caused tumour regression. Following cessation of treatment, tumours reappeared, the majority of which regressed upon retreatment. No systemic toxicity was found upon necropsy of L-744,832-treated mice. This first demonstration of anti-FPTase-mediated tumour regression suggests that FPTase inhibitors may be safe and effective anti-tumour agents in some cancers.


Subject(s)
Alkyl and Aryl Transferases , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Methionine/analogs & derivatives , Salivary Gland Neoplasms/drug therapy , Transferases/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Female , Genes, ras , Mammary Neoplasms, Experimental/pathology , Methionine/administration & dosage , Methionine/therapeutic use , Methionine/toxicity , Mice , Mice, Transgenic
9.
Proc Natl Acad Sci U S A ; 91(19): 9141-5, 1994 Sep 13.
Article in English | MEDLINE | ID: mdl-8090782

ABSTRACT

The posttranslational addition of a farnesyl moiety to the Ras oncoprotein is essential for its transforming activity. Cell-active inhibitors of the enzyme that catalyzes this reaction, protein farnesyltransferase, have been shown to selectively block ras-dependent transformation of cells in culture. Here we describe the protein farnesyltransferase inhibitor 2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl] pentyloxy-3-phenylpropionylmethioninesulfone methyl ester (L-739,749), which suppressed the anchorage-independent growth of Rat1 cells transformed with viral H-ras and the human pancreatic adenocarcinoma cell line PSN-1, which harbors altered K-ras, myc, and p53 genes. This compound also suppressed the growth of tumors arising from ras-transformed Rat1 cells in nude mice by 66%. Under the same conditions, doxorubicin inhibited tumor growth by 33%. Control tumors formed by v-raf- or v-mos-transformed Rat1 cells were unaffected by L-739,749. Furthermore, mice treated with L-739,749 exhibited no evidence of systemic toxicity. This is a demonstration of antitumor activity in vivo using a synthetic small molecule inhibitor of protein farnesyltransferase.


Subject(s)
Alkyl and Aryl Transferases , Cell Transformation, Viral , Genes, ras , Oligopeptides/pharmacology , Transferases/antagonists & inhibitors , Animals , Genes, mos , Mice , Mice, Nude
10.
Biochemistry ; 31(15): 3800-7, 1992 Apr 21.
Article in English | MEDLINE | ID: mdl-1567835

ABSTRACT

The steady-state kinetic mechanism of bovine brain farnesyl:protein transferase (FPTase) has been determined using a series of initial velocity studies, including both dead-end substrate and product inhibitor experiments. Reciprocal plots of the initial velocity data intersected on the 1/[s] axis, indicating that a ternary complex forms (sequential mechanism) and suggesting that the binding of one substrate does not affect the binding of the other. The order of substrate addition was probed by determining the patterns of dead-end substrate and product inhibition. Two nonhydrolyzable analogues of farnesyl diphosphate, (alpha-hydroxyfarnesyl)phosphonic acid (1) and [[(farnesylmethyl)hydroxyphosphinyl]methyl]phosphonic acid (2), were both shown to be competitive inhibitors of farnesyl diphosphate and noncompetitive inhibitors of Ras-CVLS. Four nonsubstrate tetrapeptides, CV[D-L]S, CVLS-NH2, N-acetyl-L-penicillamine-VIM, and CIFM, were all shown to be noncompetitive inhibitors of farnesyl diphosphate and competitive inhibitors of Ras-CVLS. These data are consistent with random order of substrate addition. Product inhibition patterns corroborated the results found with the dead-end substrate inhibitors. We conclude that bovine brain FPTase proceeds through a random order sequential mechanism. Determination of steady-state parameters for several physiological Ras-CaaX variants showed that amino acid changes affected the values of KM, but not those of kcat, suggesting that the catalytic efficiencies (kcat/KM) of Ras-CaaX substrates depend largely upon their relative binding affinity for FPTase.


Subject(s)
Alkyl and Aryl Transferases , Transferases/metabolism , Animals , Brain/enzymology , Catalysis , Cattle , Chromatography, Thin Layer , Electrophoresis, Polyacrylamide Gel , Kinetics , Oligopeptides/pharmacology , Polyisoprenyl Phosphates/antagonists & inhibitors , Polyisoprenyl Phosphates/pharmacology , Sesquiterpenes , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...