Network


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

Hotspot


Dive into the research topics where Mohamed Farrag is active.

Publication


Featured researches published by Mohamed Farrag.


Journal of Neurophysiology | 2014

Alteration of the mu opioid receptor: Ca2+ channel signaling pathway in a subset of rat sensory neurons following chronic femoral artery occlusion

Bassil Hassan; Joyce S. Kim; Mohamed Farrag; Marc P. Kaufman; Victor Ruiz-Velasco

The exercise pressor reflex, a crucial component of the cardiovascular response under physiological and pathophysiological states, is activated via metabolic and mechanical mediators that originate from contracting muscles and stimulate group III and IV afferents. We reported previously that stimulation of mu opioid receptors (MOR), expressed in both afferents, led to a significant attenuation of the reflex in rats whose femoral arteries had been occluded for 72 h. The present study examined the effect of arterial occlusion on the signaling components involved in the opioid-mediated modulation of Ca(2+) channels in rat dorsal root ganglion neurons innervating the triceps surae muscles. We focused on neurons that were transfected with cDNA coding for enhanced green fluorescent protein whose expression is driven by the voltage-gated Na(+) channel 1.8 (Na(V)1.8) promoter region, a channel expressed primarily in nociceptive neurons. With the use of a small interference RNA approach, our results show that the pertussis toxin-sensitive Gα(i3) subunit couples MOR with Ca(2+) channels. We observed a significant leftward shift of the MOR agonist [D-Ala2-N-Me-Phe4-Glycol5]-enkephalin concentration-response relationship in neurons isolated from rats with occluded arteries compared with those that were perfused freely. Femoral occlusion did not affect Ca(2+) channel density or the fraction of the main Ca(2+) channel subtype. Furthermore, Western blotting analysis indicated that the leftward shift did not result from either increased Gα(i3) or MOR expression. Finally, all neurons from both groups exhibited an inward current following exposure of the transient potential receptor vanilloid 1 (TRPV1) agonist, 8-methyl-N-vanillyl-6-nonenamide. These findings suggest that sensory neurons mediating the exercise pressor reflex express Na(V)1.8 and TRPV1 channels, and femoral occlusion alters the MOR pharmacological profile.


Journal of Neurophysiology | 2013

Identification of CaV channel types expressed in muscle afferent neurons

Renuka Ramachandra; Bassil Hassan; Stephanie G. McGrew; James Dompor; Mohamed Farrag; Victor Ruiz-Velasco; Keith S. Elmslie

Cardiovascular adjustments to exercise are partially mediated by group III/IV (small to medium) muscle afferents comprising the exercise pressor reflex (EPR). However, this reflex can be inappropriately activated in disease states (e.g., peripheral vascular disease), leading to increased risk of myocardial infarction. Here we investigate the voltage-dependent calcium (CaV) channels expressed in small to medium muscle afferent neurons as a first step toward determining their potential role in controlling the EPR. Using specific blockers and 5 mM Ba(2+) as the charge carrier, we found the major calcium channel types to be CaV2.2 (N-type) > CaV2.1 (P/Q-type) > CaV1.2 (L-type). Surprisingly, the CaV2.3 channel (R-type) blocker SNX482 was without effect. However, R-type currents are more prominent when recorded in Ca(2+) (Liang and Elmslie 2001). We reexamined the channel types using 10 mM Ca(2+) as the charge carrier, but results were similar to those in Ba(2+). SNX482 was without effect even though ∼27% of the current was blocker insensitive. Using multiple methods, we demonstrate that CaV2.3 channels are functionally expressed in muscle afferent neurons. Finally, ATP is an important modulator of the EPR, and we examined the effect on CaV currents. ATP reduced CaV current primarily via G protein βγ-mediated inhibition of CaV2.2 channels. We conclude that small to medium muscle afferent neurons primarily express CaV2.2 > CaV2.1 ≥ CaV2.3 > CaV1.2 channels. As with chronic pain, CaV2.2 channel blockers may be useful in controlling inappropriate activation of the EPR.


Neuroscience Letters | 2016

Gγ7 proteins contribute to coupling of nociceptin/orphanin FQ peptide (NOP) opioid receptors and voltage-gated Ca2+ channels in rat stellate ganglion neurons

Saifeldin Mahmoud; Mohamed Farrag; Victor Ruiz-Velasco

The nociceptin/orphanin FQ peptide (NOP) opioid receptors regulate neurotransmitter release via inhibition of voltage-gated Ca(2+) channels (CaV2.2) in sympathetic and sensory neurons. Stimulation of NOP receptors by its endogenous agonist, nociception (Noc), leads to membrane-delimited, voltage-dependent (VD) block of CaV2.2 channel currents mediated by Gβγ protein subunits. Previously we reported that the pertussis toxin-sensitive Gαi1 and Gβ2/β4 isoforms mediate the functional coupling of NOP opioid receptors with CaV channels in rat stellate ganglion (SG) sympathetic neurons. In the present report we extended our studies by identifying the Gγ subunit that forms the heterotrimer within this signaling pathway. Small interference RNA (or siRNA) was employed to silence the expression of the natively expressed Gγ subunits. Initial PCR assays indicated that SG neurons expressed seven Gγ subunits. Silencing Gγ3 subunits did not alter signaling between NOP receptors and Ca(2+) channels. However, after Gγ7 isoforms were silenced, the Noc-mediated inhibition of CaV channels was significantly decreased when compared to SG neurons transfected with scrambled siRNA. We observed that Gγ10 and Gγ11 mRNA levels increased 2.5- and 2.7-fold, respectively, after Gγ7 subunits were silenced. However, this compensatory increase in mRNA expression did not appear to fully rescue the NOP receptor coupling efficiency. Additionally, both Gγ2 and Gγ5 levels increased 50 and 75%, respectively, while Gγ3 and Gγ4 expression levels remained relatively unchanged. Taken together, our findings suggest that the Gαi1/Gβ2(β4)/Gγ7 heterotrimeric G protein complex determines the NOP receptor-mediated modulation of CaV channels in SG neurons.


PLOS ONE | 2015

Modulation of voltage-gated Ca2+ channels by G protein-coupled receptors in celiac-mesenteric ganglion neurons of septic rats.

Mohamed Farrag; Lacee J. Laufenberg; Jennifer L. Steiner; Gregory E. Weller; Charles H. Lang; Victor Ruiz-Velasco

Septic shock, the most severe complication associated with sepsis, is manifested by tissue hypoperfusion due, in part, to cardiovascular and autonomic dysfunction. In many cases, the splanchnic circulation becomes vasoplegic. The celiac-superior mesenteric ganglion (CSMG) sympathetic neurons provide the main autonomic input to these vessels. We used the cecal ligation puncture (CLP) model, which closely mimics the hemodynamic and metabolic disturbances observed in septic patients, to examine the properties and modulation of Ca2+ channels by G protein-coupled receptors in acutely dissociated rat CSMG neurons. Voltage-clamp studies 48 hr post-sepsis revealed that the Ca2+ current density in CMSG neurons from septic rats was significantly lower than those isolated from sham control rats. This reduction coincided with a significant increase in membrane surface area and a negligible increase in Ca2+ current amplitude. Possible explanations for these findings include either cell swelling or neurite outgrowth enhancement of CSMG neurons from septic rats. Additionally, a significant rightward shift of the concentration-response relationship for the norepinephrine (NE)-mediated Ca2+ current inhibition was observed in CSMG neurons from septic rats. Testing for the presence of opioid receptor subtypes in CSMG neurons, showed that mu opioid receptors were present in ~70% of CSMG, while NOP opioid receptors were found in all CSMG neurons tested. The pharmacological profile for both opioid receptor subtypes was not significantly affected by sepsis. Further, the Ca2+ current modulation by propionate, an agonist for the free fatty acid receptors GPR41 and GPR43, was not altered by sepsis. Overall, our findings suggest that CSMG function is affected by sepsis via changes in cell size and α2-adrenergic receptor-mediated Ca2+ channel modulation.


The Journal of Physiology | 2017

Endomorphins potentiate acid-sensing ion channel currents and enhance the lactic acid-mediated increase in arterial blood pressure: effects amplified in hindlimb ischaemia

Mohamed Farrag; Julie K. Drobish; Henry L. Puhl; Joyce S. Kim; Paul B. Herold; Marc P. Kaufman; Victor Ruiz-Velasco

Chronic limb ischaemia, characterized by inflammatory mediator release and a low extracellular pH, leads to acid‐sensing ion channel (ASIC) activation and reflexively increases mean arterial pressure; endomorphin release is also increased under inflammatory conditions. We examined the modulation of ASIC currents by endomorphins in sensory neurons from rats with freely perfused and ligated femoral arteries: peripheral artery disease (PAD) model. Endomorphins potentiated sustained ASIC currents in both groups of dorsal root ganglion neurons, independent of mu opioid receptor stimulation or G protein activation. Intra‐arterial administration of lactic acid (to simulate exercising muscle and evoke a pressor reflex), endomorphin‐2 and naloxone resulted in a significantly greater pressor response than lactic acid alone, while administration of APETx2 inhibited endomorphins enhancing effect in both groups. These results suggest a novel role for endomorphins in modulating ASIC function to effect lactic acid‐mediated reflex increase in arterial pressure in patients with PAD.


Journal of Neurophysiology | 2016

Gα14 subunit-mediated inhibition of voltage-gated Ca2+ and K+ channels via neurokinin-1 receptors in rat celiac-superior mesenteric ganglion neurons

Mohamed Farrag; Victor Ruiz-Velasco

The mechanisms by which G proteins modulate voltage-gated Ca(2+)channel currents (CaV), particularly CaV2.2 and CaV2.3, are voltage dependent (VD) or voltage independent (VI). VD pathways are typically mediated by Gαi/oand GαSsubfamilies. On the other hand, VI inhibition modulation is coupled to the Gαqsubfamily and signaling pathways downstream of phospholipase C stimulation. In most studies, this latter pathway has been shown to be linked to Gαqand/or Gα11protein subunits. However, there are no studies that have examined whether natively expressed Gα14subunits (Gαqsubfamily member) couple G protein-coupled receptors (GPCR) with CaV2.2 channels. We report that Gα14subunits functionally couple the substance P (SP)/neurokinin-1 (NK-1) receptor pathway to CaV2.2 channels in acutely dissociated rat celiac-superior mesenteric ganglion (CSMG) neurons. Exposure of CSMG neurons to SP blocked the CaV2.2 currents in a predominantly VD manner that was pertussis toxin and cholera toxin resistant, as well as Gαq/11independent. However, silencing Gα14subunits significantly attenuated the SP-mediated Ca(2+)current block. In another set of experiments, exposure of CSMG neurons to SP led to the inhibition of KCNQ K(+)M-currents. The SP-mediated M-current block was significantly reduced in neurons transfected with Gα14small-interference RNA. Finally, overexpression of the GTP-bound Gαq/11binding protein RGS2 did not alter the block of M-currents by SP but significantly abolished the oxotremorine methiodide-mediated M-current inhibition. Taken together, these results provide evidence of a new Gα14-coupled signaling pathway that modulates CaV2.2 and M-currents via SP-stimulated NK-1 receptors in CSMG neurons.


The Journal of Physiology | 2017

Endomorphins potentiate acid-sensing ion channel currents and enhance the lactic acid-mediated increase in arterial blood pressure: effects amplified in hindlimb ischaemia: Endomorphin-mediated modulation of ASIC3 channel subunits

Mohamed Farrag; Julie K. Drobish; Henry L. Puhl; Joyce S. Kim; Paul B. Herold; Marc P. Kaufman; Victor Ruiz-Velasco


PLOS ONE | 2015

Biophysical properties of CSMG neurons from sham control and septic rats.

Mohamed Farrag; Lacee J. Laufenberg; Jennifer L. Steiner; Gregory E. Weller; Charles H. Lang; Victor Ruiz-Velasco


PLOS ONE | 2015

Normalized current-voltage (I-V) relationships of acutely isolated CSMG neurons from sham control (A) and septic (B) rats 48 hr post-sepsis induction by CLP.

Mohamed Farrag; Lacee J. Laufenberg; Jennifer L. Steiner; Gregory E. Weller; Charles H. Lang; Victor Ruiz-Velasco


PLOS ONE | 2015

CSMG neurons express mu opioid (A) and free fatty acid (B) receptors.

Mohamed Farrag; Lacee J. Laufenberg; Jennifer L. Steiner; Gregory E. Weller; Charles H. Lang; Victor Ruiz-Velasco

Collaboration


Dive into the Mohamed Farrag's collaboration.

Top Co-Authors

Avatar

Victor Ruiz-Velasco

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Charles H. Lang

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Gregory E. Weller

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Jennifer L. Steiner

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Lacee J. Laufenberg

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Marc P. Kaufman

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Bassil Hassan

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Joyce S. Kim

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Henry L. Puhl

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Julie K. Drobish

Pennsylvania State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge