Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Postgrad Med ; 70(1): 36-42, 2024.
Article in English | MEDLINE | ID: mdl-38197593

ABSTRACT

INTRODUCTION: Sleep apnea (SA) is an important comorbidity in end-stage renal disease (ESRD) patients. The association between SA and cardiac and neurological disease is known. This study investigates the relationship between SA and cardiovascular and cerebrovascular outcomes in the ESRD population. METHODS: In a retrospective cohort study, the United States Renal Data System was queried to identify ESRD patients aged 18-100 years in whom hemodialysis had been initiated between 2005 and 2013. Diagnoses of SA and clinical comorbidities were determined from International Classification of Disease-9 codes. Demographic variables were obtained from Centers for Medicare and Medicaid Services Form-2728. Logistic regression was used to examine the association of SA with myocardial infarction (MI) or with stroke, controlling for demographic and clinical variables. RESULTS: Of 858,131 subjects meeting the inclusion criteria, 587 had central SA, and 22,724 had obstructive SA. The SA cohort was younger, more likely to be male and Caucasian compared to the non-SA cohort. Patients with SA also had more tobacco and alcohol use, hypertension, heart failure, and diabetes. Central SA (aRR = 1.69, 95% CI = 1.28-2.23) and obstructive SA (aRR = 1.15, 95% CI = 1.09-1.21) were associated with an increased risk of stroke but not MI. CONCLUSION: In the ESRD population, a diagnosis of central SA or obstructive SA increased the risk of stroke, but not MI. Early identification and treatment of SA in the ESRD population may help reduce the risk of stroke in these patients.


Subject(s)
Kidney Failure, Chronic , Myocardial Infarction , Sleep Apnea Syndromes , Sleep Apnea, Obstructive , Stroke , Humans , Male , Aged , United States/epidemiology , Female , Retrospective Studies , Risk Factors , Medicare , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/epidemiology , Sleep Apnea Syndromes/complications , Sleep Apnea Syndromes/epidemiology , Sleep Apnea Syndromes/diagnosis , Sleep Apnea, Obstructive/diagnosis , Stroke/complications , Stroke/epidemiology
2.
J Endocrinol Invest ; 46(10): 2125-2132, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37012521

ABSTRACT

PURPOSE: Hashimoto's thyroiditis (HT) is a common autoimmune thyroid disorder that can disrupt thyroid function and homeostasis. As HT results from a dysregulated immune system, we hypothesized that these patients might be more susceptible to transplant failure; however, literature on this association is limited. The purpose of this study is to examine the association of HT with the risk of renal transplant failure. METHODS: We utilized the United States Renal Database System dataset collected from 2005 to 2014 and compared the time from first renal transplant to transplant failure in end-stage renal disease (ESRD) patients with a HT diagnosis to ESRD patients without a HT diagnosis that underwent renal transplant. RESULTS: A total of 144 ESRD patients had International Classification of Disease-9 claim codes for HT prior to renal transplant, amongst a total cohort of 90,301 renal transplant patients aged 18-100 and meeting criteria. Patients with HT were significantly more likely to be female, white, and to have a diagnosis of cytomegalovirus compared to patients without. ESRD patients with a HT diagnosis that underwent renal transplant had a significantly increased risk of renal transplant failure compared to those ESRD renal transplant patients without an HT diagnosis. There was a significantly increased adjusted hazard ratio for graft failure in patients with a HT diagnosis compared to those without. CONCLUSION: Thyroid health and HT may play a significant role in the development of the increased risk of renal transplant failure observed in this study. Additional studies are needed to investigate the underlying mechanisms for this association.


Subject(s)
Hashimoto Disease , Kidney Diseases , Kidney Failure, Chronic , Kidney Transplantation , Humans , Female , Male , Kidney Transplantation/adverse effects , Hashimoto Disease/complications , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/surgery
3.
Phytother Res ; 28(2): 252-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23595692

ABSTRACT

A commercial resin-based pine oil (PO) derived from Pinus palustris and Pinus elliottii was the major focus of this investigation. Extracts of pine resins, needles, and bark are folk medicines commonly used to treat skin ailments, including burns. The American Burn Association estimates that 500,000 people with burn injuries receive medical treatment each year; one-half of US burn victims are children, most with scald burns. This systematic study was initiated as follow-up to personal anecdotal evidence acquired over more than 10 years by MH Bhattacharyya regarding PO's efficacy for treating burns. The results demonstrate that PO counteracted dermal inflammation in both a mouse ear model of contact irritant-induced dermal inflammation and a second degree scald burn to the mouse paw. Furthermore, PO significantly counteracted the tactile allodynia and soft tissue injury caused by the scald burn. In mouse dorsal root ganglion neuronal cultures, PO added to the medium blocked adenosine triphosphate-activated, but not capsaicin-activated, pain pathways, demonstrating specificity. These results together support the hypothesis that a pine-oil-based treatment can be developed to provide effective in-home care for second degree burns.


Subject(s)
Burns/drug therapy , Ganglia, Spinal/drug effects , Pinus/chemistry , Plant Oils/pharmacology , Adenosine Triphosphate , Animals , Capsaicin , Cells, Cultured , Dermatitis/drug therapy , Disease Models, Animal , Hyperalgesia/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neurons/drug effects , Neurons/metabolism , Pain/drug therapy , Resins, Plant/pharmacology , Skin/pathology
4.
Oncogene ; 30(13): 1586-96, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21132013

ABSTRACT

Our previous studies demonstrated that protein kinase D (PKD), a serine/threonine kinase implicated in various cell processes, is upregulated in basal cell carcinoma (BCC), supporting a possible tumorigenic role for PKD in skin. As the greatest risk factor for BCC is sun exposure, the ability of ultraviolet B (UVB) irradiation to activate PKD in primary mouse keratinocytes was investigated. Using western analysis with two autophosphorylation-specific antibodies, we show for the first time that UVB activated PKD in a time- and dose-dependent manner. UVB-induced PKD activation was verified using an in vitro kinase assay. Furthermore, activation was reduced by antioxidant pretreatment, suggesting a link with oxidative stress. UVB-induced PKD activation was mediated primarily by Src family tyrosine kinases rather than protein kinase C (PKC), and in fact, UVB did not alter PKC-mediated transphosphorylation. UVB induced apoptosis dose dependently, and this death could be prevented by overexpression of wild-type PKD, but not mutant PKD or the empty adenovirus. Indeed, a mutant that cannot be phosphorylated by Src kinases exacerbated UVB-elicited apoptosis. Thus, our data indicate that UVB irradiation of keratinocytes induces Src-mediated activation of PKD, which protects cells from UVB-stimulated apoptosis, providing a possible explanation for the observed upregulation of PKD in BCC.


Subject(s)
Epidermal Cells , Keratinocytes/radiation effects , Protein Kinase C/physiology , Ultraviolet Rays/adverse effects , Animals , Apoptosis/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Enzyme Activation , Keratinocytes/enzymology , Mice , Oxidation-Reduction , Phosphorylation , src-Family Kinases/antagonists & inhibitors
5.
Br J Dermatol ; 154(4): 586-93, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16536798

ABSTRACT

BACKGROUND: Keratinocytes undergo a defined programme of proliferation and differentiation during normal stratification of the epidermis. Anomalies in the signalling pathways controlling this process probably contribute to the pathogenesis of hyperproliferative dermatological diseases, including psoriasis and basal cell carcinoma (BCC). We have previously proposed that protein kinase D (PKD) is a proproliferative signalling enzyme in keratinocytes and have speculated that abnormalities in its levels or regulation may contribute to hyperproliferative disorders of the skin. OBJECTIVES: To determine if hyperproliferative human skin disorders are characterized by abnormal protein expression or distribution of PKD, normal human epidermis was compared with BCC and uninvolved and involved psoriatic epidermis. METHODS: To examine protein expression, immunohistochemical analysis of human samples and Western blotting of neoplastic mouse keratinocytes was performed. Western analysis of neoplastic mouse cells using a phosphospecific PKD antibody allowed estimation of PKD activation status. RESULTS: Normal human epidermis demonstrated predominant PKD protein expression in the stratum basalis, the proliferative epidermal compartment, with decreased relative expression throughout the suprabasal strata. Uninvolved psoriatic skin showed a similar pattern, but in contrast, psoriatic lesions demonstrated a diffuse distribution of PKD staining throughout all strata. The majority of BCCs examined showed significant PKD protein levels and, in those biopsies in which the levels could be compared, elevated PKD levels relative to normal epidermis. PKD levels and activation status were also increased in a neoplastic mouse keratinocyte cell line. CONCLUSIONS: PKD was elevated or misdistributed in the hyperproliferative human skin disorders, BCC and psoriasis, as well as neoplastic mouse keratinocytes. We speculate that PKD exerts proproliferative and/or antidifferentiative effects in the epidermis, and that anomalous distribution and/or activation of PKD may be involved in precipitating or sustaining the disease process in BCC and psoriasis.


Subject(s)
Carcinoma, Basal Cell/enzymology , Epidermis/enzymology , Protein Kinase C/metabolism , Psoriasis/enzymology , Skin Neoplasms/enzymology , Animals , Blotting, Western , Cells, Cultured , Humans , Immunoenzyme Techniques , Keratinocytes/enzymology , Mice , Mice, Inbred ICR , Neoplasm Proteins/metabolism
6.
Mol Cell Endocrinol ; 184(1-2): 65-76, 2001 Nov 26.
Article in English | MEDLINE | ID: mdl-11694342

ABSTRACT

Elevated extracellular potassium concentrations ([K(+)](e)) are known to stimulate aldosterone secretion from adrenal glomerulosa cells in vivo and in vitro. The mechanism is thought to involve depolarization-elicited activation of voltage-dependent calcium channels and an increase in calcium influx. Until now protein kinase C (PKC) was thought not to play a role in the steroidogenic response to elevated [K(+)](e). In this report, we provide evidence in bovine adrenal glomerulosa cells to suggest that elevated [K(+)](e) increases PKC activity, as shown by an enhancement in the phosphorylation of myristoylated alanine-rich C-kinase substrate (MARCKS). Elevated [K(+)](e)-induced MARCKS phosphorylation was delayed and transient and was not the result of a local production of angiotensin II (AngII). MARCKS phosphorylation in response to elevated [K(+)](e) was not accompanied by phosphoinositide hydrolysis but was inhibited by a selective PKC inhibitor. Elevated [K(+)](e) also activated phospholipase D (PLD) in a delayed but sustained manner. We propose that the observed PLD activation mediates the elevated [K(+)](e)-induced MARCKS phosphorylation via PKC, although other factors may modulate this phosphorylation event.


Subject(s)
Intracellular Signaling Peptides and Proteins , Membrane Proteins , Phospholipase D/metabolism , Phosphoproteins/metabolism , Potassium/pharmacology , Zona Glomerulosa/cytology , Angiotensin II/pharmacology , Animals , Cattle , Diglycerides/analysis , Enzyme Activation/drug effects , Myristoylated Alanine-Rich C Kinase Substrate , Phosphatidylinositols/analysis , Phospholipase D/drug effects , Phospholipase D/physiology , Phosphoproteins/drug effects , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein Kinase C/physiology , Second Messenger Systems
7.
Mol Cell Endocrinol ; 177(1-2): 71-9, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11377822

ABSTRACT

Atrial natriuretic peptide (ANP) is a cardiac hormone that inhibits aldosterone secretion induced by all physiologic agonists. The purpose of this study is to explore ANP-induced changes in the phosphorylation of myristoylated alanine-rich C-kinase substrate (MARCKS) and the steroidogenic acute regulatory protein (StAR), in AngII or K(+)-stimulated glomerulosa cells. The data show that ANP completely inhibits the phosphorylation of MARCKS and partially inhibits that of StAR in cells stimulated with K(+). ANP also partially inhibits MARCKS phosphorylation but does not affect StAR phosphorylation in cells stimulated with AngII. These effects appear to be cGMP-independent and at least partially dependent on inhibition of protein kinase C (PKC). To our knowledge, this is the first report of ANP modulating either MARCKS or StAR phosphorylation in [(32)P]-labeled cells. The data also support the hypothesis that ANP inhibits aldosterone secretion acting as a step involved in cholesterol transport to the mitochondria.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Phosphoproteins/metabolism , Zona Glomerulosa/cytology , Aldosterone/metabolism , Angiotensin II/pharmacology , Animals , Cattle , Cyclic GMP/pharmacology , Myristoylated Alanine-Rich C Kinase Substrate , Phosphoproteins/drug effects , Phosphorus Radioisotopes , Phosphorylation/drug effects , Potassium/pharmacology , Protein Kinase C/pharmacology , Zona Glomerulosa/drug effects
8.
Mol Cell Endocrinol ; 173(1-2): 87-94, 2001 Feb 28.
Article in English | MEDLINE | ID: mdl-11223180

ABSTRACT

The steroidogenic acute regulatory (StAR) protein mediates cholesterol transport within the mitochondria, and its phosphorylation is believed to be required for steroidogenesis. Increased extracellular potassium concentrations (K(+)), angiotensin II (AngII), and adrenocorticotropic hormone (ACTH) induce aldosterone secretion from bovine adrenal glomerulosa cells. We hypothesized that, although these agonists act via different signaling pathways, StAR phosphorylation should be common to their action. We studied the effects of K(+), AngII, and ACTH, at concentrations that yield comparable secretory responses, on StAR phosphorylation. All three agents induced significant increases in StAR phosphorylation although the response to ACTH was less than that of AngII and K(+). In cells stimulated with the protein kinase C (PKC) agonist 12-tetradecanoylphorbol 13-acetate (TPA), the Ca(2+) channel agonist BAY K8644, and the adenylate cyclase agonist forskolin, TPA caused a small but statistically significant increase in StAR phosphorylation while BAY K8644 and forskolin had no significant effect. Interestingly, the combination of TPA and BAY K8644 produced a larger increase in StAR phosphorylation than the agents alone. We conclude that in cultured bovine adrenal glomerulosa cells the PKC signaling pathway is most effective at inducing StAR phosphorylation but that there is no simple correlation between this event and aldosterone production.


Subject(s)
Aldosterone/agonists , Aldosterone/metabolism , Phosphoproteins/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Adrenocorticotropic Hormone/pharmacology , Angiotensin II/pharmacology , Animals , Blotting, Western , Calcium Channel Agonists/pharmacology , Cattle , Cells, Cultured , Colforsin/pharmacology , Electrophoresis, Gel, Two-Dimensional , Phosphorylation/drug effects , Potassium Chloride/pharmacology , Protein Kinase C/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Vasoconstrictor Agents/pharmacology , Zona Glomerulosa/cytology , Zona Glomerulosa/drug effects , Zona Glomerulosa/metabolism
9.
J Invest Dermatol ; 117(6): 1588-93, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11886527

ABSTRACT

In some cell systems, the antiproliferative effects of 8-Cl-cAMP, a site-selective cAMP analog specific for the type I cAMP-dependent protein kinase, are mediated by its metabolite, 8-Cl-adenosine. These effects were once thought to be specific to transformed cells. We investigated the ability of 8-Cl-adenosine to regulate growth and differentiation in primary cultures of mouse epidermal keratinocytes. A 24 h exposure of keratinocytes to 8-Cl-adenosine inhibited [3H]thymidine incorporation in a dose-dependent manner with an apparent IC(50) of 7.5 microM, and these effects were completely reversible. To determine the ability of 8-Cl-adenosine to induce differentiation of primary keratinocytes, we measured keratin-1 expression and transglutaminase activity, markers of early and later stages of keratinocyte differentiation, respectively. Interestingly, exposure of keratinocytes to 25 microM 8-Cl-adenosine for 24 h had no effect on keratin-1 expression or transglutaminase activity. The 8-Cl-adenosine-induced growth arrest of keratinocytes required uptake of the compound and was accompanied by an increase in protein expression of the cyclin-dependent protein kinase inhibitor p21(WAF1/Cip1). These results demonstrate that 8-Cl-adenosine inhibits growth in a non-transformed/non-immortalized cell system, possibly through an elevation in p21(WAF1/Cip1) protein levels, without inducing differentiation.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Antineoplastic Agents/pharmacology , Keratinocytes/cytology , Animals , Biomarkers , Cell Differentiation/drug effects , Cell Division/drug effects , Dose-Response Relationship, Drug , Epidermal Cells , G1 Phase/drug effects , Keratinocytes/drug effects , Keratinocytes/enzymology , Keratins/biosynthesis , Mice , Mice, Inbred ICR , Resting Phase, Cell Cycle/drug effects , Transglutaminases/metabolism
10.
J Pharmacol Exp Ther ; 294(3): 1219-24, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10945880

ABSTRACT

Inhibitors of sphingolipid metabolism are frequently used to investigate the role of ceramide and other sphingolipids as intracellular signaling molecules. For example, the inhibitor of glucosylceramide synthase D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP) is commonly used to deplete glycosphingolipids and increase ceramide levels. Ceramide is known to induce growth arrest and differentiation of keratinocytes, and we hypothesized that PDMP would increase ceramide levels and induce growth arrest of primary cultures of mouse epidermal keratinocytes. As expected, PDMP increased ceramide levels and decreased the incorporation of [(3)H]thymidine into DNA, but surprisingly, PDMP was found to rapidly inhibit the intracellular transport of [(3)H]thymidine. This is likely due to a direct effect on nucleoside transport by PDMP and not due to elevations in ceramide levels because increasing ceramide levels by the addition of exogenous sphingomyelinase had no effect on [(3)H]thymidine transport. Furthermore, it is unlikely that the decreased [(3)H]thymidine transport is in response to growth arrest because PDMP had no effect on the cell cycle profile of keratinocytes. These results reveal that PDMP inhibits nucleoside transport independent of effects on ceramide levels or cell growth but probably by a direct effect on the nucleoside transport apparatus. Thus, this compound may be unsuitable for investigating the role of ceramide or other sphingolipids in some cellular processes.


Subject(s)
Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Keratinocytes/metabolism , Morpholines/pharmacology , Thymidine/metabolism , Animals , Biological Transport , Cell Division/drug effects , Cells, Cultured , Ceramides/metabolism , Mice , Mice, Inbred ICR
11.
Mol Cell Endocrinol ; 154(1-2): 1-9, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10509794

ABSTRACT

Angiotensin II (AngII) is thought to stimulate aldosterone secretion from bovine adrenal glomerulosa cells in part via activation of protein kinase C (PKC), while adrenocorticotropic hormone (ACTH) functions through increases in intracellular cAMP levels and calcium influx. Rather than using invasive homogenization techniques as in previous studies, we chose to monitor PKC activity in intact glomerulosa cells in situ by measuring the phosphorylation of the endogenous PKC substrate, myristoylated alanine-rich C-kinase substrate (MARCKS). AngII enhanced MARCKS phosphorylation in a rapid, sustained manner; whereas ACTH induced a rapid and sustained inhibition of MARCKS phosphorylation. Studies using pharmacological agents to mimic various signals indicated that the AngII-induced MARCKS phosphorylation was due to PKC activation, and the ACTH-elicited decrease was mediated by increases in calcium influx rather than cAMP production. We propose that changes in the phosphorylation state of MARCKS, an actin-binding protein, may contribute to cytoskeletal rearrangements involved in steroidogenesis.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Angiotensin II/pharmacology , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Proteins/metabolism , Zona Glomerulosa/cytology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Aldosterone/metabolism , Animals , Calcium Channel Agonists/pharmacology , Cattle , Cells, Cultured , Colforsin/pharmacology , Humans , Myristoylated Alanine-Rich C Kinase Substrate , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein Kinase C/physiology , Zona Glomerulosa/metabolism
12.
J Biol Chem ; 274(8): 4663-70, 1999 Feb 19.
Article in English | MEDLINE | ID: mdl-9988703

ABSTRACT

The hormone 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) elicits the programmed pattern of differentiation in epidermal keratinocytes. Based on data indicating a potential role of phospholipase D (PLD) in mediating keratinocyte differentiation, we investigated the effect of 1,25(OH)2D3 on PLD expression. A 24-h exposure to 1, 25(OH)2D3 stimulated PLD-1, but not PLD-2, mRNA expression. This 1, 25(OH)2D3-enhanced expression was accompanied by increased total PLD and PLD-1 activity. Time course studies indicated that 1,25(OH)2D3 induced PLD-1 expression by 8 h, with a maximal increase at 20-24 h. Exposure to 1,25(OH)2D3 inhibited proliferation over the same time period with similar kinetics. Expression of the early (spinous) differentiation marker keratin 1 decreased in response to 1, 25(OH)2D3 over 12-24 h. Treatment with 1,25(OH)2D3 enhanced the activity of transglutaminase, a late (granular) differentiation marker, by 12 h with a maximal increase after 24 h. In situ hybridization studies demonstrated that the highest levels of PLD-1 expression are in the more differentiated (spinous and granular) layers of the epidermis, with little expression in basal keratinocytes. Our results suggest a role for PLD expression/activity during keratinocyte differentiation.


Subject(s)
Calcitriol/physiology , Epidermis/enzymology , Phospholipase D/biosynthesis , Animals , Animals, Newborn , Cell Differentiation , Cells, Cultured , Enzyme Induction , Epidermal Cells , Keratinocytes/cytology , Keratinocytes/enzymology , Mice , Mice, Inbred ICR , Phospholipase D/metabolism , Signal Transduction
13.
Biochem J ; 330 ( Pt 1): 445-51, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9461542

ABSTRACT

We have demonstrated previously that in bovine adrenal glomerulosa cells, phospholipase D (PLD) activity can indirectly result in the generation of sn-1,2-diacylglycerol (DAG) through its production of phosphatidic acid (PA) and the subsequent action of PA phosphohydrolase. Furthermore, the PLD-generated DAG can trigger aldosterone secretion. Therefore, we characterized PLD activation by two agonists, angiotensin II (Ang II) and carbachol, to determine if the activity of the enzyme might underlie sustained aldosterone secretion. We determined that Ang II-induced PLD activation occurred via the angiotensin-1 receptor (AT1), and that a specific AT1 antagonist, losartan, inhibited this activation, whereas the same concentration of the AT2-specific antagonist, PD 123319, had no effect. Ang II activated PLD with a dose dependence similar to that observed for aldosterone secretion, with slight increases in activity induced by 0.1 nM Ang II and maximal activation at 10 nM. We also found that Ang II induced a sustained activation of PLD, but that the effect of carbachol, a stable analogue of acetylcholine, was transient; PLD activity increased within 5 min of exposure to carbachol but then ceased by 15 min. Higher carbachol concentrations were also unable to sustain PLD activation. These results suggest that the Ang II-elicited activation of PLD is associated with a sustained increase in aldosterone secretion from glomerulosa cells and further provide the first evidence, to our knowledge, of differences in the kinetics of PLD activation in response to two physiologically relevant agonists. Finally, we speculate that this disparity correlates with different functional responses induced by the two agents.


Subject(s)
Angiotensin II/pharmacology , Carbachol/pharmacology , Phospholipase D/metabolism , Zona Glomerulosa/enzymology , Angiotensin Receptor Antagonists , Animals , Cattle , Cells, Cultured , Diglycerides/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Losartan/pharmacology , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/physiology
14.
J Invest Dermatol ; 110(4): 318-23, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9540968

ABSTRACT

We have previously determined that sustained phospholipase D (PLD) activation is associated with differentiation induction in primary mouse epidermal keratinocytes. We therefore investigated the effect of two bacterial PLD on keratinocyte proliferation and differentiation. We found that Streptomyces sp. PLD was much less potent at inhibiting proliferation than S. chromofuscus PLD, with a half-maximal inhibitory concentration of 0.05 versus less than 0.001 IU per ml for S. chromofuscus PLD. Similarly, S. chromofuscus PLD stimulated transglutaminase activity more effectively and potently than S. sp. PLD. When we examined the formation of products by the two PLD, we found that the S. sp. PLD showed higher activity at all concentrations. Whereas the PLD from S. sp. is relatively inactive on sphingomyelin, S. chromofuscus PLD is known to hydrolyze both glycerophospholipids and sphingomyelin. Based on recent data indicating a role for ceramide in regulating cell growth and differentiation, we hypothesized that the ability of S. chromofuscus PLD to hydrolyze sphingomyelin might underlie its greater potency. Therefore, we examined the effect of exogenous sphingomyelinase and synthetic ceramides on DNA synthesis. We found that sphingomyelinase exhibited a potent concentration-dependent effect on [3H]thymidine incorporation, much like S. chromofuscus PLD. Synthetic cell-permeable ceramides (C6- and C2-ceramide) also concentration dependently inhibited DNA synthesis, with a half-maximal inhibitory concentration of approximately 12 microM. Finally, we obtained evidence suggesting that ceramide is generated in response to a physiologically relevant agent, because tumor necrosis factor-alpha, a known effector of sphingomyelin turnover in other systems and a cytokine that is produced and released by keratinocytes, increased ceramide levels in primary epidermal keratinocytes.


Subject(s)
Ceramides/physiology , Epidermal Cells , Keratinocytes/cytology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Membrane Permeability/physiology , Cells, Cultured , Ceramides/pharmacokinetics , Ceramides/pharmacology , Hydrolysis , Mice , Mice, Inbred ICR , Phosphatidic Acids/metabolism , Phospholipase D/analysis , Phospholipase D/pharmacology , Sphingomyelin Phosphodiesterase/pharmacology , Sphingomyelins/metabolism , Streptomyces/chemistry , Tumor Necrosis Factor-alpha/pharmacology
15.
Methods Mol Biol ; 105: 89-98, 1998.
Article in English | MEDLINE | ID: mdl-10427553
16.
Methods Mol Biol ; 105: 151-60, 1998.
Article in English | MEDLINE | ID: mdl-10427558
17.
Biochem J ; 326 ( Pt 3): 745-53, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9307024

ABSTRACT

Activation of phosphatidylcholine-specific phospholipase D(PLD) occurs as part of the complex signal-transduction cascade initiated by agonist stimulation of tyrosine kinase and G-protein-coupled receptors. A variety of mammalian PLD activities have been described, and cDNAs for two PLDs recently reported (human PLD1 and murine PLD2). We describe here the cloning and chromosomal localization of murine PLD1. Northern-blot hybridization and RNase protection analyses were used to examine the expression of murine PLD1 and PLD2 ina variety of cell lines and tissues. PLD1 and PLD2 were expressed in all RNA samples examined, although the absolute expression of each isoform varied, as well as the ratio of PLD1 to PLD2. Moreover, in situ hybridization of adult brain and murine embryo sections revealed high levels of expression of individual PLDs in some cell types and no detectable expression in others. Thus the two PLDs probably carry out distinct roles in restricted subsets of cells rather than ubiquitous roles in all cells.


Subject(s)
Phospholipase D/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , Gene Expression , In Situ Hybridization , Mice , Molecular Sequence Data , Organ Specificity , Phospholipase D/metabolism , Sequence Alignment
18.
Br J Dermatol ; 133(5): 686-93, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8555017

ABSTRACT

In this study we have used a human hair follicle whole-organ culture system to examine the effects of 12-O-tetradecanoyl-phorbol-13-acetate (TPA), a potent activator of protein kinase C (PKC), on hair follicle growth and hair fibre production. Anagen hair follicles were isolated from human facial skin by microdissection and placed in suspension culture in supplemented Williams E medium. Hair follicle and hair fibre lengths were measured daily using an inverted microscope and cumulative growth values were calculated. Treatment with TPA resulted in a potent, dose-dependent inhibition of total cumulative hair follicle growth (IC50 = 1 nM). Hair follicles grew at a comparable rate for 4 days in the presence or absence of 10 nM TPA, after which growth of TPA-treated follicles ceased while control follicles grew by a further 0.8 mm over the subsequent 6 days. In contrast, 10 nM TPA treatment did not affect hair fibre elongation for a period of 8 days, after which TPA-treated fibre production ceased while control fibres grew by a further 0.79 mm over the subsequent 7 days. Incubation of hair follicles with TPA resulted in a 41% inhibition of hair fibre protein synthesis, as measured biochemically from the incorporation of 3H-leucine using a differential akali extraction method. The inhibitory effect of TPA on follicle growth was partially prevented by preincubation with the selective PKC inhibitor H-7, and almost completely prevented by preincubation with the more potent PKC inhibitor Ro 31-7549. Neither agent alone significantly affected follicle growth at concentrations that reversed the TPA response. These findings indicate that PKC is a negative regulator of hair follicle growth, and suggest that PKC may play a part in the transduction of follicular growth-inhibitory signals.


Subject(s)
Hair Follicle/growth & development , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , Depression, Chemical , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Hair/drug effects , Hair/growth & development , Hair/metabolism , Hair Follicle/drug effects , Hair Follicle/enzymology , Humans , Indoles/pharmacology , Isoquinolines/pharmacology , Maleimides/pharmacology , Organ Culture Techniques , Piperazines/pharmacology , Protein Biosynthesis , Protein Kinase C/antagonists & inhibitors , Time Factors
19.
J Cell Physiol ; 163(2): 248-56, 1995 May.
Article in English | MEDLINE | ID: mdl-7706369

ABSTRACT

1,25-Dihydroxyvitamin D3 [1,25(OH)2D3] has been proposed as a physiologic regulator of keratinocyte growth and differentiation. Utilizing a proliferative serum-free culture system, we have found that a physiologic (picomolar) concentrations this hormone stimulated proliferation of primary mouse epidermal keratinocytes; at higher (nanomolar to micromolar) doses, growth was inhibited by 1,25(OH)2D3. We investigated the nature of the signal transduction mechanism underlying the response to 1,25(OH)2D3 and observed little or no effect of either low or high concentrations of the hormone on cytosolic calcium levels or Fos expression. Furthermore, the protein kinase C inhibitor, Ro 31-7549, had very little effect on the growth inhibition induced by a high dose (1 microM) of 1,25(OH)2D3. This lack of rapid signal transduction events was consistent with the inability of a short (4-hour) exposure to 1,25(OH)2D3 to initiate a complete growth-inhibitory response as measured using [3H]thymidine incorporation. Our results indicate that physiologic concentrations of 1,25(OH)2D3 are required for optimal keratinocyte growth. Furthermore, we found no evidence of rapid effects of 1,25(OH)2D3 and suggest that in mouse epidermal keratinocytes, the response to this hormone is mediated by a slow transduction pathway, such as that activated by the intracellular 1,25(OH)2D3 receptor (VDR).


Subject(s)
Calcitriol/pharmacology , Epidermal Cells , Keratinocytes/cytology , Animals , Calcium/metabolism , Cell Division/drug effects , Cells, Cultured , Culture Media, Serum-Free , DNA/biosynthesis , Gene Expression/drug effects , Genes, fos , Indoles/pharmacology , Intracellular Membranes/metabolism , Keratinocytes/metabolism , Maleimides/pharmacology , Mice , Mice, Inbred Strains , Osmolar Concentration , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , beta-Galactosidase/metabolism
20.
Biochem J ; 300 ( Pt 1): 263-70, 1994 May 15.
Article in English | MEDLINE | ID: mdl-8198544

ABSTRACT

The expression of a fos-lacZ fusion gene was studied in primary mouse epidermal keratinocytes obtained from transgenic mice. This gene construct contains the entire upstream regulatory sequence of c-fos, and expression of the endogenous and fusion gene was shown by Northern analysis to correlate upon induction with the phorbol ester 12-O-tetradecanoylphorbol 13-acetate (TPA). Using a chromogenic substrate of beta-galactosidase, we also demonstrated that expression of the fusion gene product, like that of Fos, was localized to the cell nucleus. In addition, we showed that epidermal keratinocytes responded to dialysed fetal bovine serum (FBS), TPA and high-calcium medium with enhanced Fos-lacZ expression and an inhibition of proliferation. The time course of induction of Fos-lacZ expression was similar for dialysed FBS and TPA, with a peak approximately 2 h after exposure. Exposure for approximately 24 h to an elevated extracellular calcium concentration was required to elicit an increase in Fos-lacZ expression. The lack of an immediate effect of raising medium calcium levels on Fos-lacZ expression contrasted with the rapidity of its effect on DNA synthesis, which was significantly inhibited within 6-8 h. In addition, we found that the protein kinase C inhibitor Ro 31-7549 blocked Fos-lacZ expression induced by TPA but had little or no effect on that elicited by high calcium levels. Thus, although our results indicate that the fos gene product may be involved in mediating epidermal keratinocyte growth arrest in response to differentiative agents such as FBS, TPA and high medium calcium levels, the exact role of this gene product remains unclear.


Subject(s)
Gene Expression Regulation , Genes, fos , Keratinocytes/metabolism , beta-Galactosidase/genetics , Animals , Calcium/pharmacology , Cells, Cultured , Gene Expression Regulation/drug effects , Indoles/pharmacology , Male , Maleimides/pharmacology , Mice , Mice, Transgenic , Protein Kinase C/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...