Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Neuropeptides ; 67: 45-55, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29129406

ABSTRACT

Osteoporosis, a disease characterized by progressive bone loss and increased risk of fracture, often results from menopausal loss of estrogen in women. Neuropeptide Y has been shown to negatively regulate bone formation, with amygdala specific deletion of the Y2 receptor resulting in increased bone mass in mice. In this study, ovariectomized (OVX) mice were injected once daily with JNJ-31020028, a brain penetrant Y2 receptor small molecule antagonist to determine the effects on bone formation. Antagonist treated mice had reduced weight and showed increased whole-body bone mineral density compared to vehicle-injected mice. Micro computerized tomography (micro-CT) demonstrated increased vertebral trabecular bone volume, connectivity density and trabecular thickness. Femoral micro-CT analysis revealed increased bone volume within trabecular regions and greater trabecular number, without significant difference in other parameters or within cortical regions. A decrease was seen in serum P1NP, a measure used to confirm positive treatment outcomes in bisphosphonate treated patients. C-terminal telopeptide 1 (CTX-1), a blood biomarker of bone resorption, was decreased in treated animals. The higher bone mineral density observed following Y2 antagonist treatment, as determined by whole-body DEXA scanning, is indicative of either enhanced mineralization or reduced bone loss. Additionally, our findings that ex vivo treatment of bone marrow cells with the Y2 antagonist did not affect osteoblast and osteoclast formation suggests the inhibitor is not affecting these cells directly, and suggests a central role for compound action in this system. Our results support the involvement of Y2R signalling in bone metabolism and give credence to the hypothesis that selective pharmacological manipulation of Y2R may provide anabolic benefits for treating osteoporosis.


Subject(s)
Benzamides/pharmacology , Bone Density/drug effects , Neuropeptide Y/metabolism , Osteogenesis/drug effects , Ovariectomy , Piperazines/pharmacology , Animals , Bone Density/physiology , Bone and Bones/drug effects , Bone and Bones/metabolism , Female , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/physiology , Ovariectomy/methods , Receptors, Neuropeptide Y/metabolism
2.
Curr Osteoporos Rep ; 13(5): 280-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26243670

ABSTRACT

Frailty is highly prevalent in the elderly, increasing the risk of poor outcomes that include falls, incident disability, hospitalization, and mortality. Thus, a great need exists to characterize the underlying mechanisms and ultimately identify strategies that prevent, delay, and even reverse frailty. Mouse models can provide insight into molecular mechanisms of frailty by reducing variability in lifestyle and genetic factors that can complicate interpretation of human clinical data. Frailty, generally recognized as a syndrome involving reduced homeostatic reserve in response to physiologic challenges and increasing susceptibility to poor health outcomes, is predominantly assessed using two independent strategies, integrated phenotype and deficit accumulation. The integrated phenotype defines frailty by the presentation of factors affecting functional capacity such as weight loss, exhaustion, low activity levels, slow gait, and grip strength. The deficit accumulation paradigm draws parameters from a greater range of physiological systems, such as the ability to perform daily activities, coordination and gait, mental components, physiological problems, and history and presence of medical morbidities. This strategic division also applies within the emerging field of mouse frailty models, with both methodologies showing usefulness in providing insight into physiologic mechanisms and testing interventions. Our review will explore the strategies used, caveats in methodology, and future directions in the application of animal models for the study of the frailty syndrome.


Subject(s)
Aging , Models, Animal , Aged , Animals , Frail Elderly , Humans , Mice , Muscle Strength , Physical Endurance
3.
Minerva Med ; 100(1): 79-94, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19277006

ABSTRACT

Osteoporosis is a major health issue worldwide, with significant economic consequences and adverse impacts on the quality of life. Hip fractures are the most devastating complication of osteoporosis, are likely to increase exponentially with an increasingly aged population, are associated with high recurrence rate, and lead to significant morbidity and mortality. This review discusses the prevalence and impact of hip fractures, the assessment of fracture risk, fall prevention, and treatment of osteoporosis with emphasis on evidence for hip fracture reduction among the various agents currently available. The aim is to provide recommendations to optimize hip fracture prevention and treatment. Ample evidence exists in the literature of many other risk factors independent from bone mineral density that increase fracture risk. These clinical risk factors have been validated in large cohorts and are incorporated into clinical tools that are invaluable in treatment decisions. In addition, strategies to prevent or reduce falls are integral to comprehensive osteoporosis management. Vitamin D combined with calcium has a role in primary prevention. Alendronate, residronate, strontium and zoledronic acid have proven efficacy in primary and secondary hip fracture prevention. An aggressive approach to investigate, assess and manage an individual's fracture risk and fall risk is paramount to reduce the high morbidity and mortality associated with hip fractures. The choice of therapy should be determined by the patient's calculated fracture risk and efficacy of the potential treatment, including long term compliance associated with the agent of choice.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Hip Fractures/prevention & control , Osteoporosis/prevention & control , Accidental Falls/prevention & control , Aged , Aged, 80 and over , Bone Density , Female , Hip Fractures/drug therapy , Humans , Middle Aged , Osteoporosis/drug therapy , Risk Assessment , Risk Factors
5.
J Endocrinol ; 173(3): 395-405, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12065229

ABSTRACT

The effects of the related cytokines interleukin-6 (IL-6), leukemia inhibitory factor (LIF) and oncostatin-M on bone resorption and cytosolic Ca(2+) signaling were compared in isolated rat osteoclasts. In the traditional disaggregated osteoclast (pit) assay, IL-6 and LIF, but not oncostatin-M, conserved the bone resorption otherwise inhibited by high extracellular [Ca(2+)] (15 mM). It produced a paradoxical, concentration-dependent stimulation of resorption by elevated extracellular Ca(2+). In the micro-isolated single osteoclast resorption assay, IL-6, high [Ca(2+)] or IL-6 plus high [Ca(2+)] all increased pit formation. In contrast, the IL-6 receptor (IL-6R)-specific agonist antibody MT-18 inhibited bone resorption in a concentration-dependent manner (1:500 to 1:500 000). MT-18 triggered cytosolic Ca(2+) signals in fura 2-loaded osteoclasts within approximately 10 min of application. Each cytosolic Ca(2+) transient began with a peak deflection that persisted in Ca(2+)-free, EGTA-containing extracellular medium, consistent with a release of intracellularly stored Ca(2+). This was followed by a sustained elevation of cytosolic [Ca(2+)] that was abolished in Ca(2+)-free medium, as expected from an entry of extracellular Ca(2+), and by the Ca(2+) channel antagonist Ni(2+). The inclusion of either IL-6 or soluble human (sh) IL-6R specifically reversed both the above effects of MT-18, confirming that both effects were specific for the IL-6R. The findings suggest that IL-6R activation by IL-6 stimulates osteoclastic bone resorption either by reversing the inhibitory effect of high extracellular Ca(2+) in stromal-containing systems or itself stimulating bone resorption along with Ca(2+) by micro-isolated osteoclasts. In contrast, activation of the IL-6R by an agonist antibody produces an inhibition of bone resorption and an associated triggering of the cytosolic Ca(2+) signals previously associated with regulation of bone resorptive function in other situations.


Subject(s)
Bone Resorption , Calcium Signaling/drug effects , Cytokines/pharmacology , Cytosol/metabolism , Osteoclasts/metabolism , Receptors, Interleukin-6/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Calcium/metabolism , Cells, Cultured , Cytokines/metabolism , Growth Inhibitors/pharmacology , Interleukin-6/metabolism , Interleukin-6/pharmacology , Leukemia Inhibitory Factor , Lymphokines/pharmacology , Oncostatin M , Osteoclasts/ultrastructure , Peptides/pharmacology , Rats , Rats, Wistar , Receptors, Interleukin-6/immunology
7.
Biochem Biophys Res Commun ; 285(2): 328-34, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11444846

ABSTRACT

We provide the first evidence that the bone marrow-derived cytokine, macrophage colony-stimulating factor (M-CSF), inhibits the formation of bone-forming osteoblasts. We examined both osteoclast and osteoblast formation in primary rat bone marrow cultures. As expected, M-CSF together with osteoprotegerin ligand (OPGL) markedly accelerated osteoclastogenesis. In contrast, treatment with M-CSF alone yielded no osteoclasts at any time. The most striking and novel observation was that M-CSF with or without OPGL dramatically suppressed osteoblast formation. In separate experiments, estradiol markedly suppressed osteoclast formation in the M-CSF/OPGL-treated cultures independently of osteoblasts. Consistent with this was the expression of estrogen receptor-alpha (ERalpha) and ERbeta mRNA in osteoclast precursors. We therefore conclude that in addition to the well-known action of M-CSF to modulate osteoclastogenesis, this cytokine may also regulate osteoblast formation.


Subject(s)
Bone Marrow Cells/cytology , Macrophage Colony-Stimulating Factor/pharmacology , Osteoblasts/cytology , Osteoclasts/cytology , Animals , Animals, Newborn , Bone Marrow Cells/drug effects , Carrier Proteins/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Estrogen Receptor alpha , Estrogen Receptor beta , Gene Expression Regulation/drug effects , Humans , Kinetics , Membrane Glycoproteins/pharmacology , Osteoblasts/drug effects , Osteoblasts/physiology , Osteoclasts/drug effects , Osteoclasts/physiology , RANK Ligand , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptor Activator of Nuclear Factor-kappa B , Receptors, Estrogen/genetics , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic/drug effects
8.
Biochem Biophys Res Commun ; 285(2): 335-9, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11444847

ABSTRACT

Cathepsin K plays a key role in bone resorption. We provide the first evidence that osteoprotegerin ligand (OPGL), a critical pro-resorptive cytokine, acutely stimulates the expression of cathepsin K in osteoclasts. We used in situ RT-PCR and real time quantitative RT-PCR to analyze cathepsin K gene expression. OPGL enhanced cathepsin K mRNA levels in mature osteoclasts isolated from rat neonatal long bones. OPGL together with macrophage colony-stimulating factor (M-CSF) also stimulated cathepsin K gene expression in monocytic cells and multinucleate osteoclasts in bone marrow cultures. Real time quantitative RT-PCR demonstrated high levels of cathepsin K mRNA in bone marrow cultures, paralleling the degree of osteoclastogenesis. We therefore suggest that OPGL enhances bone resorption, at least in part, by inducing cathepsin K gene expression.


Subject(s)
Carrier Proteins/pharmacology , Cathepsins/genetics , Gene Expression Regulation, Enzymologic/physiology , Membrane Glycoproteins/pharmacology , Osteoclasts/enzymology , Transcription, Genetic/drug effects , Animals , Animals, Newborn , Bone Marrow Cells/cytology , Calcitriol/pharmacology , Cathepsin K , Cell Culture Techniques/methods , Cell Nucleus/physiology , Cells, Cultured , Cytokines/pharmacology , Femur , Gene Expression Regulation, Enzymologic/drug effects , Kinetics , Macrophage Colony-Stimulating Factor/pharmacology , Osteoclasts/cytology , Osteoclasts/drug effects , RANK Ligand , RNA, Messenger/genetics , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tibia , Time Factors
9.
DNA Cell Biol ; 19(2): 79-91, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10701774

ABSTRACT

Cathepsin B expression is increased at both the mRNA and protein levels in a wide variety of tumors. The mechanisms responsible for this regulation are not well elucidated. We have isolated a 2.2-kb cathepsin B genomic fragment that contains the 5'-flanking region of the cathepsin B gene. Using reporter gene analysis in human glioblastoma U87MG cells, we have mapped a 228-bp fragment (-172 to +56) having high promoter activity. This promoter region has a high G+C content; contains potential Spl, Ets, and USF binding motifs; and lacks canonical TATA and CAAT boxes immediately upstream of the major transcriptional initiation site. Cotransfection experiments demonstrated that Spl and Ets1 could trans-activate cathepsin B transcription, whereas Ets2 could not. Electrophoretic mobility shift assays and supershift assays revealed that three of the four putative Sp1 sites in this promoter region form a specific complex containing the Sp1 transcription factor. Mutating all four of the Spl binding sites individually markedly reduced the promoter activity of transfected reporter genes in U87 cells. Cotransfection of this cathepsin B promoter construct with Spl family expression vectors in Schneider's Drosophila line 2 (SL2) cells demonstrated that Spl and Sp3, but not Sp4, activated cathepsin B transcription. Taken together, these results suggest that Sp1, Sp3, and Ets1 are important factors in cathepsin B transcription. The regulation of cathepsin B transcription by Sp1- and Sp1-related factors is mediated through multiple GC boxes.


Subject(s)
Cathepsin B/genetics , Glioma/genetics , Glioma/metabolism , Proto-Oncogene Proteins/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Base Sequence , Binding Sites/genetics , Cathepsin B/metabolism , DNA/genetics , DNA/metabolism , DNA Probes/genetics , DNA-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins c-ets , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured
10.
J Leukoc Biol ; 66(4): 609-16, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10534117

ABSTRACT

We utilized HL-60 cells as a model system to examine the regulation of ctsb gene expression by differentiating agents. Inducers of monocytic differentiation [phorbol ester (PMA), calcitriol (D3), and sodium butyrate (NaB)] and inducers of granulocytic differentiation [all-trans retinoic acid (RA) and 9-cis retinoic acid (9-cis RA)] increase ctsb mRNA levels in a dose-dependent manner as determined by Northern blot hybridization. D3 and retinoids exert additive effects, suggesting that these agents act in part through distinct pathways. Actinomycin D decay experiments indicate that D3, NaB, RA, and 9-cis RA do not alter mRNA stability. In contrast, PMA markedly increases the half-life of ctsb mRNA. In transient transfection assays, PMA and NaB both stimulate transcription of the luciferase reporter gene placed under the control of ctsb promoter fragments. Thus, inducers of HL-60 cell differentiation can regulate the expression of the ctsb gene at both transcriptional and posttranscriptional levels.


Subject(s)
Butyrates/pharmacology , Calcitriol/pharmacology , Cathepsin B/genetics , Gene Expression Regulation/drug effects , Mitogens/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology , Alitretinoin , Cell Differentiation/drug effects , Cycloheximide/pharmacology , Dactinomycin/pharmacology , HL-60 Cells , Humans , Nucleic Acid Synthesis Inhibitors/pharmacology , Promoter Regions, Genetic , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger , Transcription, Genetic
11.
J Cell Biol ; 146(5): 1161-72, 1999 Sep 06.
Article in English | MEDLINE | ID: mdl-10477767

ABSTRACT

The multifunctional ADP-ribosyl cyclase, CD38, catalyzes the cyclization of NAD(+) to cyclic ADP-ribose (cADPr). The latter gates Ca(2+) release through microsomal membrane-resident ryanodine receptors (RyRs). We first cloned and sequenced full-length CD38 cDNA from a rabbit osteoclast cDNA library. The predicted amino acid sequence displayed 59, 59, and 50% similarity, respectively, to the mouse, rat, and human CD38. In situ RT-PCR revealed intense cytoplasmic staining of osteoclasts, confirming CD38 mRNA expression. Both confocal microscopy and Western blotting confirmed the plasma membrane localization of the CD38 protein. The ADP-ribosyl cyclase activity of osteoclastic CD38 was next demonstrated by its ability to cyclize the NAD(+) surrogate, NGD(+), to its fluorescent derivative cGDP-ribose. We then examined the effects of CD38 on osteoclast function. CD38 activation by an agonist antibody (A10) in the presence of substrate (NAD(+)) triggered a cytosolic Ca(2+) signal. Both ryanodine receptor modulators, ryanodine, and caffeine, markedly attenuated this cytosolic Ca(2+) change. Furthermore, the anti-CD38 agonist antibody expectedly inhibited bone resorption in the pit assay and elevated interleukin-6 (IL-6) secretion. IL-6, in turn, enhanced CD38 mRNA expression. Taken together, the results provide compelling evidence for a new role for CD38/ADP-ribosyl cyclase in the control of bone resorption, most likely exerted via cADPr.


Subject(s)
Antigens, CD , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Bone Resorption , NAD+ Nucleosidase/genetics , NAD+ Nucleosidase/metabolism , Osteoclasts/metabolism , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Antigens, Differentiation/chemistry , Base Sequence , Calcium Signaling , Cell Membrane/enzymology , Cells, Cultured , Cloning, Molecular , Cyclic ADP-Ribose , Enzyme Activation , Gene Expression Regulation, Enzymologic/drug effects , Humans , Interleukin-6/metabolism , Interleukin-6/pharmacology , Membrane Glycoproteins , Molecular Sequence Data , NAD/analogs & derivatives , NAD/metabolism , NAD+ Nucleosidase/chemistry , Osteoclasts/cytology , Osteoclasts/enzymology , RNA, Messenger/analysis , RNA, Messenger/genetics , Rabbits , Rats , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/metabolism , Sequence Homology, Amino Acid
13.
Blood ; 86(7): 2475-80, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7670094

ABSTRACT

In HL-60 cells, retinoic acid (RA) and 9 cis-RA induce granulocytic differentiation, and calcitriol and sodium butyrate induce monocytic differentiation. To study the role of retinoid resistance on the response to these agents, we investigated their effects in HL-60 cells, retinoid-resistant HL-60R cells, and HL-60R+ cells in which retinoid sensitivity has been restored. In HL-60 cells, cathepsin D (ctsd) mRNA levels are increased by these agents and by cholera toxin after pretreatment with each agent. Calcitriol, 9 cis-RA, and sodium butyrate increase interleukin-8 (IL-8) mRNA expression, and pretreatment with these agents or RA potentiates the stimulation of IL-8 by phorbol ester (TPA). Pretreatment of HL-60 cells with all of the agents confers inducibility of cathepsin L (ctsl) mRNA by TPA in previously unresponsive cells. In HL-60R cells, none of the agents alone or in combination significantly enhances the expression of the ctsd, IL-8, or ctsl mRNAs. Retinoid stimulation (either alone or in combination with the other agents) of the three mRNAs is partially restored in the HL-60R+ cells. Calcitriol does not alter the expression of any of these mRNAs, and only the stimulation of IL-8 mRNA by sodium butyrate is recovered. Treatment with all of the agents inhibits proliferation and stimulates differentiation of the HL-60 cells. RA and calcitriol are unable to inhibit proliferation of the HL-60R cells, whereas only calcitriol fails to inhibit proliferation of the HL-60R+ cells. None of the agents induces differentiation in either the HL-60R or HL-60R+ cells. Therefore, the mutation of the RA receptor alpha is insufficient to account for the altered responses of the HL-60R cells, and there are likely defects in other signaling pathways in these cells. These cells may prove useful in examining the mechanism of cross-resistance between various differentiating agents.


Subject(s)
Butyrates/pharmacology , Calcitriol/pharmacology , Endopeptidases , Leukemia, Promyelocytic, Acute/pathology , Tretinoin/pharmacology , Butyric Acid , Cathepsin D/genetics , Cathepsin L , Cathepsins/genetics , Cell Differentiation/drug effects , Cysteine Endopeptidases , Drug Resistance , Gene Expression/drug effects , Humans , Interleukin-8/genetics , Leukemia, Promyelocytic, Acute/metabolism , RNA, Messenger/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
14.
Cell Growth Differ ; 6(7): 871-7, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7547509

ABSTRACT

Cathepsin D (ctsd) is a lysosomal acid protease found in neutrophils and monocytes. We investigated whether differentiating agents increase the expression of ctsd mRNA in HL-60 cells. Treatment with either retinoic acid or calcitriol enhances the steady-state levels of ctsd mRNA in a dose-dependent manner. The stimulation by retinoic acid requires new protein synthesis. Pretreatment with retinoic acid enhances the response of the ctsd gene to prostaglandin E2. To determine whether the effects of retinoic acid and calcitriol are associated with differentiation, we pretreated Hl-60 cells for 120 h with inducers of granulocytic differentiation (lithium chloride, DMSO, and retinoic acid) and monocytic differentiation (calcitriol, sodium butyrate, and phorbol ester). Lithium chloride and DMSO do not significantly affect ctsd mRNA expression, and none of the granulocytic inducers alters the subsequent response of the ctsd gene to calcitriol. All of the monocytic inducers stimulate ctsd mRNA, and both calcitriol and sodium butyrate significantly potentiate the subsequent response to retinoic acid. Transcription initiation of the ctsd gene occurs at one major and several minor sites and is unaffected by treatment with retinoic acid and calcitriol or pretreatment with other differentiating agents. Although differentiation appears to influence ctsd mRNA expression, calcitriol and retinoic acid stimulate ctsd gene expression via mechanisms that are independent of their role in differentiation.


Subject(s)
Calcitriol/pharmacology , Cathepsin D/genetics , HL-60 Cells/physiology , Tretinoin/pharmacology , Base Sequence , Butyrates/pharmacology , Butyric Acid , Cell Differentiation/drug effects , Gene Expression Regulation, Developmental/drug effects , Growth Substances/pharmacology , Humans , Molecular Sequence Data , RNA, Messenger/metabolism , Transcription, Genetic/physiology
15.
Cell Growth Differ ; 6(6): 713-8, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7669726

ABSTRACT

Cathepsin L (ctsl) is a lysosomal cysteine proteinase, the synthesis and secretion of which is induced by transformation, growth factors, and tumor promoters. We studied the effect and the mechanism of action of phorbol ester (TPA) on the expression of ctsl mRNA in U937 histiocytic leukemia cells. TPA treatment induces ctsl mRNA in a manner that is dose-dependent, occurs at the level of transcription, and is ablated by cotreatment with cycloheximide but is unaffected by dexamethasone. Treatment with TPA plus staurosporine, a potent protein kinase C inhibitor, results in greater expression of ctsl mRNA than does treatment with TPA alone. Similar to TPA, staurosporine alone increases ctsl transcription, an effect that is inhibited by cycloheximide. Another PKC inhibitor, H7, exerted no effect upon the induction of ctsl mRNA by either TPA or staurosporine. Staurosporine and H7, however, inhibit the increase in c-jun mRNA by TPA. In contrast, the tyrosine kinase inhibitors herbimycin A and genistein inhibit the effect of TPA and staurosporine upon ctsl mRNA with little or no effect on c-jun expression. Pretreatment with sodium orthovanadate enhances the induction of ctsl expression by TPA and staurosporine. These data suggest that, in U937 cells, TPA-stimulated ctsl gene transcription is apparently activated by a protein kinase C-independent signal transduction pathway involving tyrosine kinase activation.


Subject(s)
Cathepsins/biosynthesis , Endopeptidases , Lymphoma, Large B-Cell, Diffuse/pathology , Monocytes/drug effects , Tetradecanoylphorbol Acetate/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , Alkaloids/pharmacology , Cathepsin L , Cathepsins/genetics , Cell Differentiation/drug effects , Cycloheximide/pharmacology , Cysteine Endopeptidases , Drug Interactions , Enzyme Induction/drug effects , Humans , Isoquinolines/pharmacology , Monocytes/enzymology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Piperazines/pharmacology , Protein Kinase C/antagonists & inhibitors , Staurosporine , Tumor Cells, Cultured , Vanadates/pharmacology
16.
J Biol Chem ; 268(2): 1039-45, 1993 Jan 15.
Article in English | MEDLINE | ID: mdl-8419312

ABSTRACT

Cathepsin L is a lysosomal cysteine protease whose expression and secretion is induced by malignant transformation, growth factors, and tumor promoters. Many human tumors express high levels of cathepsin L, which is a broad spectrum protease with potent elastase and collagenase activities. Two published human cathepsin L cDNA sequences differ only in their 5'-untranslated regions. In this study, we demonstrate the concurrent expression of two distinct human cathepsin L mRNAs (hCATL-A and hCATL-B) in adenocarcinoma, hepatoma, and renal cancer cell lines. Cloning of the human cathepsin L gene by polymerase chain reaction amplification of genomic DNA and subsequent sequencing reveals that hCATL-A and hCATL-B mRNAs are encoded by a single gene. The 3' end of the first intron contains the 5' portion of hCATL-B and is contiguous to the second exon of the gene. These data suggest either the possibility of alternative splicing or the presence of a second promoter within the first intron of the hCATL gene. We mapped the hCATL gene to chromosome 9q21-22. Sequencing of both the mouse and human cathepsin L genes demonstrates almost complete conservation of exon and intron position, but significant divergence in intron structure, possibly reflecting differences in regulation of expression of the mouse and human cathepsin L genes.


Subject(s)
Cathepsins/genetics , Chromosomes, Human, Pair 9 , Endopeptidases , Isoenzymes/genetics , Amino Acid Sequence , Animals , Base Sequence , Cathepsin L , Chromosome Banding , Chromosome Mapping , Cloning, Molecular , Cysteine Endopeptidases , Exons , Gene Expression Regulation, Enzymologic , Genetic Vectors , Humans , Introns , KB Cells , Karyotyping , Lymphocytes/enzymology , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Restriction Mapping , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
17.
Cell Growth Differ ; 2(1): 23-31, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1848774

ABSTRACT

The major excreted protein (MEP) of mouse fibroblasts is the precursor to a lysosomal acid protease (cathepsin L) whose synthesis is induced by malignant transformation, growth factors, tumor promoters, and cyclic AMP. We have previously cloned a functional gene for MEP from NIH 3T3 cells. When subcloned into chloramphenicol acetyl transferase (CAT) expression vectors, both 4-kilobase and 300 base pair fragments in the 5'-flanking region of the MEP gene confer CAT activity that is stimulated by cyclic AMP treatment but is not stimulated by phorbol ester treatment of NIH 3T3 cells. These fragments confer constitutive promoter activity that is comparable to that of the SV40 promoter. Primer extension, using RNA from cells transiently transfected with MEP-CAT fusion plasmids, demonstrates that phorbol ester treatment increases the amount of transcript from constructs containing both the promoter and sequences downstream of the transcription initiation site, including the first three introns, but not from constructs containing only the 5'-flanking region of the MEP gene. Nuclear run-off experiments confirm that the increase in endogenous MEP mRNA is mediated by increased transcription and not via relief of transcriptional attenuation. Since both the MEP promoter, which contains three potential binding sites for the AP-2 transcription factor, and the SV40 promoter, which contains both AP-1 and AP-2 binding sites, fail to respond to 12-O-tetradecanoylphorbol-13-acetate in NIH 3T3 cells, these upstream motifs are not sufficient to confer phorbol ester responsiveness in NIH 3T3 cells. These results suggest that the MEP gene is regulated in a complex manner by sequences both upstream and downstream of the transcription initiation site.


Subject(s)
Cathepsins/genetics , Endopeptidases , Enzyme Precursors/genetics , Gene Expression Regulation, Enzymologic/drug effects , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid , Tetradecanoylphorbol Acetate/pharmacology , Animals , Base Sequence , Cathepsin L , Chloramphenicol O-Acetyltransferase/genetics , Cyclic AMP/pharmacology , Cysteine Endopeptidases , Mice , Molecular Sequence Data , Plasmids/genetics , RNA, Messenger/biosynthesis , Transfection/genetics
18.
Mol Cell Biol ; 8(8): 3316-21, 1988 Aug.
Article in English | MEDLINE | ID: mdl-2463474

ABSTRACT

Malignantly transformed mouse fibroblasts synthesize and secrete large amounts of major excreted protein (MEP), a 39,000-dalton precursor to an acid protease (cathepsin L). To evaluate the possible role of this protease in the transformed phenotype, we transfected cloned genes for mouse or human MEP into mouse NIH 3T3 cells with an expression vector for the dominant, selectable human multidrug resistance (MDR1) gene. The cotransfected MEP sequences were efficiently coamplified and transcribed during stepwise selection for multidrug resistance in colchicine. The transfected NIH 3T3 cell lines containing amplified MEP sequences synthesized as much MEP as did Kirsten sarcoma virus-transformed NIH 3T3 cells. The MEP synthesized by cells transfected with the cloned mouse and human MEP genes was also secreted. Elevated synthesis and secretion of MEP by NIH 3T3 cells did not change the nontransformed phenotype of these cells.


Subject(s)
Cell Transformation, Neoplastic , Cloning, Molecular , Drug Resistance/genetics , Endopeptidases/genetics , Gene Amplification , Genes , Neoplasm Proteins/genetics , Animals , Cathepsin L , Cathepsins , Cell Line , Cells, Cultured , Cysteine Endopeptidases , DNA/genetics , DNA/isolation & purification , Endopeptidases/biosynthesis , Mice , Plasmids , RNA/genetics , RNA/isolation & purification , Transfection
19.
J Biol Chem ; 263(1): 254-61, 1988 Jan 05.
Article in English | MEDLINE | ID: mdl-2826441

ABSTRACT

The major excreted protein (MEP) of mouse fibroblast cells is the 39,000 Mr precursor to a lysosomal acid protease (cathepsin L) induced by malignant transformation, growth factors, and tumor promoters. We have cloned and characterized the gene for MEP from NIH-3T3 cells. This cosmid clone (pcosMMEP), containing the unique 12,000-base pair mouse MEP gene, has been transfected into monkey kidney (CV-1) cells and human epidermoid carcinoma (A431) cells. The stable A4MEP transfectants produce mouse MEP that is an active cathepsin which is secreted, glycosylated, and processed intracellularly to lower molecular weight forms as in the wild-type NIH-3T3 cells. The CVMEP cells (nontransformed phenotype) produce quantities of mouse MEP similar to that found in NIH-3T3 cells, whereas the A4MEP cells (transformed phenotype) produce greater amounts of MEP similar to the levels seen in Kirsten virus-transformed NIH-3T3 cells. The MEP mRNAs from both mouse cells and stably transfected human cells are the same size and have the same single major site for initiation of transcription, indicating that the cloned mouse MEP promoter is active in transfected cells.


Subject(s)
Cathepsins/genetics , Cloning, Molecular , Endopeptidases , Enzyme Precursors/genetics , Genes , Lysosomes/enzymology , Transfection , Animals , Cathepsin L , Cell Line , Cells, Cultured , Cosmids , Cysteine Endopeptidases , DNA Restriction Enzymes , Fibroblasts/enzymology , Humans , Mice , Nucleic Acid Hybridization
20.
Biochem J ; 246(3): 731-5, 1987 Sep 15.
Article in English | MEDLINE | ID: mdl-3689328

ABSTRACT

The major excreted protein (MEP) of malignantly transformed mouse fibroblasts is a secreted thiol proteinase. Sequencing of the MEP cDNA shows the coding region for the protein to be identical with the sequence for a mouse cysteine proteinase isolated from macrophages, but the MEP cDNA is polyadenylated at a different site in the 3' non-coding region. Strong homology of MEP with human cathepsin L suggests that MEP is the mouse analogue of cathepsin L. Amino acid sequencing of the N-terminus of the secreted form of MEP indicates that, during secretion, the polypeptide is cleaved between amino acids 17 and 18. We have placed the MEP cDNA in a eukaryotic expression vector and demonstrated the production of the 39 kDa polypeptide form of mouse MEP in monkey CV-1 cells.


Subject(s)
Cathepsins , DNA/genetics , Endopeptidases , Amino Acid Sequence , Base Sequence , Cathepsin L , Cells, Cultured , Cysteine Endopeptidases , Molecular Sequence Data , Transfection , Transformation, Bacterial
SELECTION OF CITATIONS
SEARCH DETAIL
...