Network


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

Hotspot


Dive into the research topics where Kafait U. Malik is active.

Publication


Featured researches published by Kafait U. Malik.


Journal of Biological Chemistry | 1996

Calcium/Calmodulin-dependent Protein Kinase IIα Mediates Activation of Mitogen-activated Protein Kinase and Cytosolic Phospholipase A2 in Norepinephrine-induced Arachidonic Acid Release in Rabbit Aortic Smooth Muscle Cells

Mubarack M. Muthalif; Ibrahim F. Benter; Mohammed R. Uddin; Kafait U. Malik

We have investigated the contribution of Ca2+/calmodulin-dependent protein kinase II (CaM kinase II) and mitogen-activated protein kinase (MAP kinase) in norepinephrine (NE)-induced arachidonic acid (AA) release in rabbit aortic vascular smooth muscle cells (VSMC). NE enhanced release of AA via activation of cytosolic phospholipase A2 (cPLA2) but not secretory PLA2 in VSMC prelabeled with [3H]AA. NE (10 μM) enhanced CaM kinase II and MAP kinase activity. In cells transiently transfected with antisense oligonucleotides complementary to the translation initiation sites of CaM kinase II and MAP kinase, NE-induced AA release was inhibited by 100 and 35% respectively. Treatment of cells with PD-098059, a MAP kinase kinase inhibitor, or with MAP kinase antisense oligonucleotide reduced NE-induced activation of MAP kinase and cPLA2. NE-induced MAP kinase and cPLA2 activation was also inhibited in cells treated with a CaM kinase II inhibitor, KN-93, or with CaM kinase II antisense oligonucleotide. On the other hand, inhibition of MAP kinase kinase with PD-098059 or of MAP kinase with antisense oligonucleotides did not alter the NE-induced increase in CaM kinase II activity. Phosphorylation of MAP kinase and CaM kinase II by NE, studied by 32P incorporation and immune complex kinase assays, was inhibited by KN-93. Collectively, these data suggest that CaM kinase II can activate MAP kinase, which in turn activates cPLA2 to release AA for prostacyclin synthesis in the rabbit VSMC. This novel pathway for activation of MAP kinase by CaM kinase II appears to be mediated through stimulation of MAP kinase kinase. Activation of adrenergic receptors with NE in VSMC caused translocation of CaM kinase II, MAP kinase, and cPLA2 to the nuclear envelope only in the presence of extracellular Ca2+. Okadaic acid, which increased phosphorylation and activity, did not translocate these enzymes. Therefore, it appears that in rabbit VSMC, NE, by promoting extracellular Ca2+ influx, increases CaM kinase II activity, leading to activation of MAP kinase and cPLA2 and translocation to the nuclear envelope, resulting in release of AA from the nuclear envelope for prostacyclin synthesis.


Biochemical Journal | 2001

Sphingosylphosphocholine is a naturally occurring lipid mediator in blood plasma: a possible role in regulating cardiac function via sphingolipid receptors.

Károly Liliom; Guoping Sun; Moritz Bünemann; Tamas Virag; Nóra Nusser; Daniel L. Baker; De-an Wang; Matthew J. Fabian; Bodo Brandts; Kirsten Bender; Andreas Eickel; Kafait U. Malik; Duane D. Miller; Dominic M. Desiderio; Gabor Tigyi; Lutz Pott

Blood plasma and serum contain factors that activate inwardly rectifying GIRK1/GIRK4 K+ channels in atrial myocytes via one or more non-atropine-sensitive receptors coupled to pertussis-toxin-sensitive G-proteins. This channel is also the target of muscarinic M(2) receptors activated by the physiological release of acetylcholine from parasympathetic nerve endings. By using a combination of HPLC and TLC techniques with matrix-assisted laser desorption ionization-time-of-flight MS, we purified and identified sphingosine 1-phosphate (SPP) and sphingosylphosphocholine (SPC) as the plasma and serum factors responsible for activating the inwardly rectifying K+ channel (I(K)). With the use of MS the concentration of SPC was estimated at 50 nM in plasma and 130 nM in serum; those concentrations exceeded the 1.5 nM EC(50) measured in guinea-pig atrial myocytes. With the use of reverse-transcriptase-mediated PCR and/or Western blot analysis, we detected Edg1, Edg3, Edg5 and Edg8 as well as OGR1 sphingolipid receptor transcripts and/or proteins. In perfused guinea-pig hearts, SPC exerted a negative chronotropic effect with a threshold concentration of 1 microM. SPC was completely removed after perfusion through the coronary circulation at a concentration of 10 microM. On the basis of their constitutive presence in plasma, the expression of specific receptors, and a mechanism of ligand inactivation, we propose that SPP and SPC might have a physiologically relevant role in the regulation of the heart.


Hypertension | 2000

Angiotensin II-Induced Hypertension Contribution of Ras GTPase/Mitogen-Activated Protein Kinase and Cytochrome P450 Metabolites

Mubarack M. Muthalif; Nour A. Karzoun; Lillian W. Gaber; Zinat Khandekar; Ibrahim F. Benter; Abdelwahab E. Saeed; Jean Hugues Parmentier; Anne M. Estes; Kafait U. Malik

We reported that norepinephrine and angiotensin II (Ang II) activate the Ras/mitogen-activated protein (MAP) kinase pathway primarily through the generation of cytochrome P450 (CYP450) metabolites. The purpose of the present study was to determine the contribution of Ras and CYP450 to Ang II–dependent hypertension in rats. Infusion of Ang II (350 ng/min for 6 days) elevated mean arterial blood pressure (MABP) (171±3 mm Hg for Ang II versus 94±5 for vehicle group, P <0.05). Ras is activated on farnesylation by farnesyl protein transferase (FPT). When Ang II was infused in combination with FPT inhibitor FPT III (232 ng/min) or BMS-191563 (578 ng/min), the development of hypertension was attenuated (171±3 mm Hg for Ang II plus vehicle versus 134±5 mm Hg for Ang II plus FPT III and 116±6 mm Hg for Ang II plus BMS-191563, P <0.05). Treatment with the MAP kinase kinase inhibitor PD-98059 (5 mg SC) reduced MABP. The CYP450 inhibitor aminobenzotriazole (50 mg/kg) also diminished the development of Ang II–induced hypertension to 113±8 mm Hg. The activities of Ras, MAP kinase, and CYP450 measured in the kidney were elevated in hypertensive animals. The infusion of FPT III, BMS-191563, or aminobenzotriazole reduced the elevation in Ras and MAP kinase activity. Morphological studies of the kidney showed that FPT III treatment ameliorated the arterial injury, vascular lesions, fibrinoid necrosis, focal hemorrhage, and hypertrophy of muscle walls observed in hypertensive animals. These data suggest that the activation of Ras and CYP450 contributes to the development of Ang II–dependent hypertension and associated vascular pathology.


Life Sciences | 1979

Relationships between the kallikrein-kinin and prostaglandin systems

A. Nasjletti; Kafait U. Malik

Abstract Kinins are polypeptides released from plasma protein precursor(s) by plasma and tissue enzymes termed kallikreins (1). Bradykinin and kallidin, the peptide products of plasma and glandular kallikreins respectively, exert varied actions on numerous biological systems (2). Two recent findings that suggest interrelation of the kallikrein-kinin and prostaglandin systems have greatly enlarged the potential sphere of influence of kinins. First, bradykinin was shown to stimulate release of prostaglandins (3,4). Second, aspirin-like drugs which are known to antagonize several pharmacological actions of kinins (2) were found to inhibit prostaglandin biosynthesis (5). This led to the proposal that prostaglandins that are released from tissues by kinins contribute to the actions of this class of peptides (4). Interactions of kinins and prostaglandins may be a feature of biological processes as diverse as those involved in inflammatory responses and in regulation of electrolyte and fluid balance (6,7). This review analyzes the relationships between the kallikrein-kinin and prostaglandin systems and discusses the functional significance of such relations.


Biochemical and Biophysical Research Communications | 1980

Hepatic metabolism of prostacyclin (PGI2) in the rabbit: formation of a potent novel inhibitor of platelet aggregation.

Patrick Y-K Wong; Kafait U. Malik; Dominic M. Desiderio; John C. McGiff; Frank F. Sun

Abstract Metabolism of [9-3H]-PGI2 was studied in the isolated Tyrodes perfused rabbit liver. Five products, four radioactive and one non-radioactive, were identified in the perfusate: 19-hydroxy-6-keto-PGF1α, 6-keto-PGF1α, dinor-6-keto-PGF1α, pentanor PGF1α and a 6-keto-PGE1-like substance. The first two, 19-hydroxy-6-keto-PGF1α and 6-keto-PGF1α, represented 5% and 45% respectively, of the total radioactivity; the last two accounted for 39%. The presence of dinor and pentanor derivatives of 6-keto-PGF1α indicated that β -oxidation and oxidative-decarboxylation occurs in the liver as the major metabolic pathway of PGI2. One non-radioactive metabolite which co-migrated with authentic 6-keto-PGE1 was found to inhibit platelet aggregation, having a potency similar to authentic 6-keto-PGE1, and its effect can be eliminated by boiling and by alkali treatment. This metabolite, having similar Rf value on TLC and biological behavior as 6-keto-PGE1, may arise from oxidation of 6-keto-PGF1α via the 9-hydroxyprostaglandin dehydrogenase pathway, as suggested by recovery of tritiated water in the aqueous phase of the perfusate. This material, a potent inhibitor of platelet aggregation, may arise from PGI2 or its hydrolysis product, 6-keto-PGF1α.


Circulation Research | 2007

Mechanism of High Glucose–Induced Angiotensin II Production in Rat Vascular Smooth Muscle Cells

Eduard N. Lavrentyev; Anne M. Estes; Kafait U. Malik

Angiotensin II (Ang II), a circulating hormone that can be synthesized locally in the vasculature, has been implicated in diabetes-associated vascular complications. This study was conducted to determine whether high glucose (HG) (≈23.1 mmol/L), a diabetic-like condition, stimulates Ang II generation and the underlying mechanism of its production in rat vascular smooth muscle cells. The contribution of various enzymes involved in Ang II generation was investigated by silencing their expression with small interfering RNA in cells exposed to normal glucose (≈4.1 mmol/L) and HG. Angiotensin I (Ang I) was generated from angiotensinogen by cathepsin D in the presence of normal glucose or HG. Although HG did not affect the rate of angiotensinogen conversion, it decreased expression of angiotensin-converting enzyme (ACE), downregulated ACE-dependent Ang II generation, and upregulated rat vascular chymase–dependent Ang II generation. The ACE inhibitor captopril reduced Ang II levels in the media by 90% in the presence of normal glucose and 19% in HG, whereas rat vascular chymase silencing reduced Ang II production in cells exposed to HG but not normal glucose. The glucose transporter inhibitor cytochalasin B, the aldose reductase inhibitor alrestatin, and the advanced glycation end product formation inhibitor aminoguanidine attenuated HG-induced Ang II generation. HG caused a transient increase in extracellular signal-regulated kinase (ERK)1/2 phosphorylation, and ERK1/2 inhibitors reduced Ang II accumulation by HG. These data suggest that polyol pathway metabolites and AGE can stimulate rat vascular chymase activity via ERK1/2 activation and increase Ang II production. In addition, decreased Ang II degradation, which, in part, could be attributable to a decrease in angiotensin-converting enzyme 2 expression observed in HG, contributes to increased accumulation of Ang II in vascular smooth muscle cells by HG.


Hypertension | 1998

Cytochrome P-450 Metabolites Mediate Norepinephrine-Induced Mitogenic Signaling

Mohammed R. Uddin; Mubarack M. Muthalif; Nour A. Karzoun; Ibrahim F. Benter; Kafait U. Malik

Norepinephrine (NE) stimulates release of arachidonic acid (AA) from tissue lipids in blood vessels, which is metabolized via cyclooxygenase, lipoxygenase (LO), and cytochrome P-450 (CYP-450) pathways to biologically active products. Moreover, NE and AA have been shown to stimulate proliferation of vascular smooth muscle cells (VSMCs) of rat aorta. The purpose of this study was to determine the possible contribution of AA and its metabolites to NE-induced mitogenesis in VSMCs of rat aorta and the underlying mechanism of their actions. NE (0.1 to 10 micromol/L) increased DNA synthesis as measured by [3H]thymidine incorporation in VSMCs, and this effect was attenuated by inhibitors of CYP-450 (17-octadecynoic acid, 5 micromol/L; 12-diabromododec-11-enoic acid, 10 micromol/L; and dibromo-dodecenyl-methylsulfimide, 10 micromol/L) and by the LO inhibitor (baicalein, 20 micromol/L), but not by the cyclooxygenase inhibitor (indomethacin, 5 micromol/L). CYP-450 and LO metabolites of AA, 20-hydroxyeicosatetraenoic acid (HETE) (0.1 to 0.5 micromol/L) and 12(S)-HETE, respectively, increased [3H]thymidine incorporation in VSMCs. Both NE and 20-HETE increased mitogen activated protein (MAP) kinase activity as measured by the in-gel kinase assay. The inhibitor of MAP kinase kinase, PD-98059 (50 micromol/L), attenuated NE as well as 20-HETE induced [3H]thymidine incorporation and MAP kinase activation in VSMCs. These data suggest that products of AA formed via CYP-450, most likely 20-HETE, and via LO mediate NE induced mitogenesis in VSMCs.


Hypertension | 2000

Contribution of Ras GTPase/MAP Kinase and Cytochrome P450 Metabolites to Deoxycorticosterone-Salt–Induced Hypertension

Mubarack M. Muthalif; Ibrahim F. Benter; Zinat Khandekar; Lillian W. Gaber; Anne M. Estes; Suzanna Malik; Jean Hugues Parmentier; Veeraswamy Manne; Kafait U. Malik

We recently reported that norepinephrine and angiotensin II activate the Ras/mitogen-activated protein (MAP) kinase pathway through generation of a cytochrome P450 (CYP450) and lipoxygenase metabolites. The purpose of this study was to determine the contribution of Ras/MAP kinase to deoxycorticosterone acetate (DOCA)-salt-induced hypertension in rats. Administration of DOCA and 1% saline drinking water to uninephrectomized rats for 6 weeks significantly elevated mean arterial blood pressure (MABP) (166+/-5 mm Hg, n=19) compared with that of normotensive controls (95+/-5 mm Hg, n=7) (P<0.05). The activity of Ras and MAP kinase measured in the heart was increased in DOCA-salt hypertensive rats. Infusion of the Ras farnesyl transferase inhibitors FPT III (138 ng/min) and BMS-191563 (694 ng/min) significantly (P<0.05) attenuated MABP to 139+/-4 mm Hg (n=14) and 126+/-1 mm Hg (n=4), respectively. Moreover, infusion of MAP kinase kinase inhibitor PD-98059 (694 ng/min) also reduced MABP in hypertensive rats. Morphological studies of the kidney showed that treatment of rats with FPT III, which reduced Ras activity, minimized the hyperplastic occlusive arteriosclerosis and fibrinoid vasculitis observed in untreated hypertensive rats. In addition, the rise in CYP450 activity and MABP in hypertensive rats was prevented by the CYP450 inhibitor aminobenzotriazole (50 mg/kg) and was associated with a decrease in Ras and MAP kinase activity in the heart. These data suggest that the Ras/MAP kinase pathway contributes to DOCA-salt-induced hypertension and associated vascular pathology consequent to activation of CYP450.


Journal of Lipid Research | 2008

cPLA2 phosphorylation at serine-515 and serine-505 is required for arachidonic acid release in vascular smooth muscle cells

Zoran Pavicevic; Christina C. Leslie; Kafait U. Malik

Cytosolic phospholipase A2 (cPLA2) is activated by phosphorylation at serine-505 (S505) by extracellular regulated kinase 1/2 (ERK1/2). However, rat brain calcium/calmodulin-dependent kinase II (CaMKII) phosphorylates recombinant cPLA2 at serine-515 (S515) and increases its activity in vitro. We have studied the sites of cPLA2 phosphorylation and their significance in arachidonic acid (AA) release in response to norepinephrine (NE) in vivo in rabbit vascular smooth muscle cells (VSMCs) using specific anti-phospho-S515- and -S505 cPLA2 antibodies and by mutagenesis of S515 and S505 to alanine. NE increased the phosphorylation of cPLA2 at S515, followed by phosphorylation of ERK1/2 and consequently phosphorylation of cPLA2 at S505. The CaMKII inhibitor 2-[N-(2-hydroxyethyl)]-N-(4-methoxybenzene-sulfonyl)]amino-N-(4-chlorocinnamyl)-methylbenzylamine attenuated cPLA2 at S515 and S505, whereas the ERK1/2 inhibitor U0126 reduced phosphorylation at S505 but not at S515. NE in cells transduced with adenovirus carrying enhanced cyan fluorescent protein cPLA2 wild type caused phosphorylation at S515 and S505 and increased AA release. Expression of the S515A mutant in VSMCs reduced the phosphorylation of S505, ERK1/2, and AA release in response to NE. Transduction with a double mutant (S515A/S505A) blocked the phosphorylation of cPLA2 and AA release. These data suggest that the NE-stimulated phosphorylation of cPLA2 at S515 is required for the phosphorylation of S505 by ERK1/2 and that both sites of phosphorylation are important for AA release in VSMCs.


Annals of the New York Academy of Sciences | 1990

Prostaglandins and the Release of the Adrenergic Transmitter

Kafait U. Malik; Elmir Sehic

Prostaglandins (PG) are synthesized from arachidonic acid, which is deesterified from tissue lipids in response to various stimuli including adrenergic transmitter, consequent to activation of one or more lipase(s). The profile of arachidonic acid metabolites generated in response to sympathetic nerve stimulation or administration of norepinephrine (NE) may vary in different tissues. For example, in the kidney and spleen, PGE2, is the major and PGI2 and PGF2 alpha the minor products; whereas in the heart and blood vessels, PGI2 is the principal product of arachidonic acid generated in response to sympathetic nerve stimulation. PGE2 and PGI2 inhibit release of NE and/or the postjunctional actions of this neurotransmitter in several tissues. These observations and the findings that inhibitors of cyclooxygenase enhance NE release and the response of effector organs to nerve stimulation suggest that PGs act as physiological modulators of adrenergic transmission. The mechanism by which PGs modulate release of the adrenergic transmitter has not yet been established. NE appears to be released from sympathetic fibers during depolarization by influx of Na+, which is associated with entry of Ca++ through omega-conotoxin-sensitive Ca++ channels that are distinct from those in the vascular smooth muscle, which are sensitive to nifedipine. Ouabain in low external K+ activates the former, whereas external Na+ depletion activates the latter type of Ca++ channels in the nerve fiber and promotes release of NE. PGs (PGE2) may inhibit release of NE from nerve fibers by interfering with the availability of Ca++ through these Ca++ channels or promoting efflux of Ca++ from the nerve terminal.

Collaboration


Dive into the Kafait U. Malik's collaboration.

Top Co-Authors

Avatar

Anne M. Estes

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Fariborz A. Yaghini

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Chi Young Song

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nayaab S. Khan

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Xiao R. Fang

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mubarack M. Muthalif

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Jean-Hugues Parmentier

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge