Network


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

Hotspot


Dive into the research topics where Nicholas A. Delamere is active.

Publication


Featured researches published by Nicholas A. Delamere.


Glia | 2014

Mechanosensitive release of adenosine 5′-triphosphate through pannexin channels and mechanosensitive upregulation of pannexin channels in optic nerve head astrocytes: A mechanism for purinergic involvement in chronic strain

Jonathan M. Beckel; Arthur J. Argall; Jason C. Lim; Jingsheng Xia; Wennan Lu; Erin E. Coffey; Edward J. Macarak; Mohammed Shahidullah; Nicholas A. Delamere; Gulab S. Zode; Val C. Sheffield; Valery I. Shestopalov; Alan M. Laties; Claire H. Mitchell

As adenosine 5′‐triphosphate (ATP) released from astrocytes can modulate many neural signaling systems, the triggers and pathways for this ATP release are important. Here, the ability of mechanical strain to trigger ATP release through pannexin channels and the effects of sustained strain on pannexin expression were examined in rat optic nerve head astrocytes. Astrocytes released ATP when subjected to 5% of equibiaxial strain or to hypotonic swelling. Although astrocytes expressed mRNA for pannexins 1–3, connexin 43, and VNUT, pharmacological analysis suggested a predominant role for pannexins in mechanosensitive ATP release, with Rho kinase contribution. Astrocytes from panx1−/− mice had reduced baseline and stimulated levels of extracellular ATP, confirming the role for pannexins. Swelling astrocytes triggered a regulatory volume decrease that was inhibited by apyrase or probenecid. The swelling‐induced rise in calcium was inhibited by P2X7 receptor antagonists A438079 and AZ10606120, in addition to apyrase and carbenoxolone. Extended stretch of astrocytes in vitro upregulated expression of panx1 and panx2 mRNA. A similar upregulation was observed in vivo in optic nerve head tissue from the Tg‐MYOCY437H mouse model of chronic glaucoma; genes for panx1, panx2, and panx3 were increased, whereas immunohistochemistry confirmed increased expression of pannexin 1 protein. In summary, astrocytes released ATP in response to mechanical strain, with pannexin 1 the predominant efflux pathway. Sustained strain upregulated pannexins in vitro and in vivo. Together, these findings provide a mechanism by which extracellular ATP remains elevated under chronic mechanical strain, as found in the optic nerve head of patients with glaucoma. GLIA 2014;62:1486–1501


The Journal of Physiology | 1977

A comparison of ion concentrations, potentials and conductances of amphibian, bovine and cephalopod lenses

Nicholas A. Delamere; G. Duncan

1. The concentrations of sodium, potassium and chloride in frog and bovine lenses showed a normal intracellular ion distribution with the sum of the internal cations approximately equal to the external sum. In the cephalopod lens, however, the sum inside was much lower than that outside.


Endocrinology | 2010

New Therapeutic and Biomarker Discovery for Peripheral Diabetic Neuropathy: PARP Inhibitor, Nitrotyrosine, and Tumor Necrosis Factor-α

Viktor R. Drel; Sergey Lupachyk; Hanna Shevalye; Igor Vareniuk; Weizheng Xu; Jie Zhang; Nicholas A. Delamere; Mohammad Shahidullah; Barbara S. Slusher; Irina G. Obrosova

This study evaluated poly(ADP-ribose) polymerase (PARP) inhibition as a new therapeutic approach for peripheral diabetic neuropathy using clinically relevant animal model and endpoints, and nitrotyrosine (NT), TNF-alpha, and nitrite/nitrate as potential biomarkers of the disease. Control and streptozotocin-diabetic rats were maintained with or without treatment with orally active PARP inhibitor 10-(4-methyl-piperazin-1-ylmethyl)-2H-7-oxa-1,2-diaza-benzo[de]anthracen-3-one (GPI-15,427), 30 mg kg(-1) d(-1), for 10 wk after first 2 wk without treatment. Therapeutic efficacy was evaluated by poly(ADP-ribosyl)ated protein expression (Western blot analysis), motor and sensory nerve conduction velocities, and tibial nerve morphometry. Sciatic nerve and spinal cord NT, TNF-alpha, and nitrite/nitrate concentrations were measured by ELISA. NT localization in peripheral nervous system was evaluated by double-label fluorescent immunohistochemistry. A PARP inhibitor treatment counteracted diabetes-induced motor and sensory nerve conduction slowing, axonal atrophy of large myelinated fibers, and increase in sciatic nerve and spinal cord NT and TNF-alpha concentrations. Sciatic nerve NT and TNF-alpha concentrations inversely correlated with motor and sensory nerve conduction velocities and myelin thickness, whereas nitrite/nitrate concentrations were indistinguishable between control and diabetic groups. NT accumulation was identified in endothelial and Schwann cells of the peripheral nerve, neurons, astrocytes, and oligodendrocytes of the spinal cord, and neurons and glial cells of the dorsal root ganglia. The findings identify PARP as a compelling drug target for prevention and treatment of both functional and structural manifestations of peripheral diabetic neuropathy and provide rationale for detailed evaluation of NT and TNF-alpha as potential biomarkers of its presence, severity, and progression.


Experimental Eye Research | 2014

Purines in the eye: recent evidence for the physiological and pathological role of purines in the RPE, retinal neurons, astrocytes, Müller cells, lens, trabecular meshwork, cornea and lacrimal gland.

Julie Sanderson; Darlene A. Dartt; Vickery Trinkaus-Randall; Jesús Pintor; Mortimer M. Civan; Nicholas A. Delamere; Erica L. Fletcher; T.E. Salt; Antje Grosche; Claire H. Mitchell

This review highlights recent findings that describ how purines modulate the physiological and pathophysiological responses of ocular tissues. For example, in lacrimal glands the cross-talk between P2X7 receptors and both M3 muscarinic receptors and α1D-adrenergic receptors can influence tear secretion. In the cornea, purines lead to post-translational modification of EGFR and structural proteins that participate in wound repair in the epithelium and influence the expression of matrix proteins in the stroma. Purines act at receptors on both the trabecular meshwork and ciliary epithelium to modulate intraocular pressure (IOP); ATP-release pathways of inflow and outflow cells differ, possibly permitting differential modulation of adenosine delivery. Modulators of trabecular meshwork cell ATP release include cell volume, stretch, extracellular Ca(2+) concentration, oxidation state, actin remodeling and possibly endogenous cardiotonic steroids. In the lens, osmotic stress leads to ATP release following TRPV4 activation upstream of hemichannel opening. In the anterior eye, diadenosine polyphosphates such as Ap4A act at P2 receptors to modulate the rate and composition of tear secretion, impact corneal wound healing and lower IOP. The Gq11-coupled P2Y1-receptor contributes to volume control in Müller cells and thus the retina. P2X receptors are expressed in neurons in the inner and outer retina and contribute to visual processing as well as the demise of retinal ganglion cells. In RPE cells, the balance between extracellular ATP and adenosine may modulate lysosomal pH and the rate of lipofuscin formation. In optic nerve head astrocytes, mechanosensitive ATP release via pannexin hemichannels, coupled with stretch-dependent upregulation of pannexins, provides a mechanism for ATP signaling in chronic glaucoma. With so many receptors linked to divergent functions throughout the eye, ensuring the transmitters remain local and stimulation is restricted to the intended target may be a key issue in understanding how physiological signaling becomes pathological in ocular disease.


Progress in Retinal and Eye Research | 2004

Expression, regulation and function of Na, K-ATPase in the lens

Nicholas A. Delamere; Shigeo Tamiya

Na,K-ATPase is responsible for maintaining the correct concentrations of sodium and potassium in lens cells. Na,K-ATPase activity is different in the two cell types that make up the lens, epithelial cells and fibers; specific activity in the epithelium is higher than in fibers. In some parts of the fiber mass Na,K-ATPase activity is barely detectable. There is a large body of evidence that suggests Na,K-ATPase-mediated ion transport by the epithelium contributes significantly to the regulation of ionic composition in the entire lens. In some species different Na,K-ATPase isoforms are present in epithelium and fibers but in general, fibers and epithelium express a similar amount of Na,K-ATPase protein. Turnover of Na,K-ATPase by protein synthesis may contribute to preservation of high Na,K-ATPase activity in the epithelium. In ageing lens fibers, oxidation, and glycation may decrease Na,K-ATPase activity. Na,K-ATPase activity in lens fibers and epithelium also may be subject to regulation as the result of protein tyrosine phosphorylation. Moreover, activation of G protein-coupled receptors by agonists such as endothelin-1 elicits changes of Na,K-ATPase activity. The asymmetrical distribution of Na,K-ATPase activity in the epithelium and fibers may contribute to ionic currents that flow in and around the lens. Studies on human cataract and experimental cataract in animals reveal changes of Na,K-ATPase activity but no clear pattern is evident. However, there is a convincing link between abnormal elevation of lens sodium and the opacification of the lens cortex that occurs in age-related human cataract.


Current Eye Research | 1989

Ca2+-ATPase activity in the human lens

Douglas Borchman; Christopher A. Patenon; Nicholas A. Delamere

A membrane-rich preparation of paired human lenses was prepared in such a manner as to preserve ATPase activity. The lipid:protein ratio of these preparations was increased 12-fold with an 85% recovery of total phospholipid. The pattern of stimulation of ATPase activity by a range of calcium concentrations was found to be similar in membrane preparations of epithelium and cortex. The concentration of calcium necessary for half-maximal simulations of ATPase activity was approximately 10(-6) M. Ca2+-ATPase activity is undetectable in the lens nuclear region. A shift in the sensitivity of lens epithelial Ca2+-ATPase activity was observed with increasing age concomitant with a general increase in Ca2+-ATPase activity suggesting age related modifications of the membrane.A membrane-rich preparation of paired human lenses was prepared in such a manner as to preserve ATPase activity. The 1ipid:protein ratio of these preparations was increased 12-fold with an 85% recovery of total phospholipid. The pattern of stimulation of ATPase activity by a range of calcium concentrations was found to be similar in membrane preparations of epithelium and cortex. The concentration of calcium necessary for half-maximal simulations of ATPase activity was approximately 10-6 M. Ca2+-ATPase activity is undetectable in the lens nuclear region. A shift in the sensitivity of lens epithelial Ca2+-ATPase activity was observed with increasing age concomitant with a general increase in Ca2+-ATPase activity suggesting age related modifications of the membrane.


American Journal of Physiology-cell Physiology | 2012

TRPV4 in porcine lens epithelium regulates hemichannel-mediated ATP release and Na-K-ATPase activity

Mohammad Shahidullah; Amritlal Mandal; Nicholas A. Delamere

In several tissues, transient receptor potential vanilloid 4 (TRPV4) channels are involved in the response to hyposmotic challenge. Here we report TRPV4 protein in porcine lens epithelium and show that TRPV4 activation is an important step in the response of the lens to hyposmotic stress. Hyposmotic solution (200 mosM) elicited ATP release from intact lenses and TRPV4 antagonists HC 067047 and RN 1734 prevented the release. In isosmotic solution, the TRPV4 agonist GSK1016790A (GSK) elicited ATP release. When propidium iodide (PI) (MW 668) was present in the bathing medium, GSK and hyposmotic solution both increased PI entry into the epithelium of intact lenses. Increased PI uptake and ATP release in response to GSK and hyposmotic solution were abolished by a mixture of agents that block connexin and pannexin hemichannels, 18α-glycyrrhetinic acid and probenecid. Increased Na-K-ATPase activity occurred in the epithelium of lenses exposed to GSK and 18α-glycyrrhetinic acid + probenecid prevented the response. Hyposmotic solution caused activation of Src family kinase and increased Na-K-ATPase activity in the lens epithelium and TRPV4 antagonists prevented the response. Ionomycin, which is known to increase cytoplasmic calcium, elicited ATP release, the magnitude of which was no greater when lenses were exposed simultaneously to ionomycin and hyposmotic solution. Ionomycin-induced ATP release was significantly reduced in calcium-free medium. TRPV4-mediated calcium entry was examined in Fura-2-loaded cultured lens epithelium. Hyposmotic solution and GSK both increased cytoplasmic calcium that was prevented by TRPV4 antagonists. The cytoplasmic calcium rise in response to hyposmotic solution or GSK was abolished when calcium was removed from the bathing solution. The findings are consistent with hyposmotic shock-induced TRPV4 channel activation which triggers hemichannel-mediated ATP release. The results point to TRPV4-mediated calcium entry that causes a cytoplasmic calcium increase which is an essential early step in the mechanism used by the lens to sense and respond to hyposmotic stress.


Experimental Eye Research | 1983

Lens cation transport and permeability changes following exposure to hydrogen peroxide

Nicholas A. Delamere; Christoper A. Paterson; Theodor R. Cotton

Exposure of the rabbit lens, in vitro, to 10(-3) M-hydrogen peroxide resulted in an increase of the lens sodium, calcium and water content together with a decrease of the lens potassium content. No such changes were observed in the presence of 10(-4) or 10(-5) M-hydrogen peroxide. 10(-3) M-hydrogen peroxide was observed to diminish 86Rb uptake by 32% and to reduce the lens Na+-, K+-ATPase activity by 37%. The lens potential remained ouabain-sensitive after peroxide treatment. These findings suggest that only partial inhibition of the lens sodium pump takes place in the presence of 10(-3) M-hydrogen peroxide. Lens depolarization, increased lens conductance and an increase rate of 86Rb efflux were also observed upon exposure of the lens to 10(-3) M-hydrogen peroxide. Such alterations of 86Rb efflux and electrophysiological parameters indicate that hydrogen peroxide changes the passive permeability of lens membranes.


Experimental Eye Research | 1988

Characteristics of ascorbate transport in the rabbit iris-ciliary body

Robin R. Socci; Nicholas A. Delamere

The accumulation and efflux of [14C]ascorbic acid by the rabbit iris-ciliary body were examined in vitro. Accumulation of labelled ascorbate was observed to be reduced significantly by ouabain(10(-4) M), low sodium solutions, 0 degree C and dinitrophenol (10(-3) M). These findings suggest that the ascorbate uptake mechanism is linked to metabolism via the sodium gradient. In addition, labelled ascorbate uptake was diminished by both phloretin (10(-4) M) and phlorhizin-(10(-4) M), two glucose transport inhibitors. Clear differences were observed between the characteristics of [14C]ascorbic acid uptake and efflux. In contrast to the uptake, 14C-labelled, ascorbate efflux from the iris-ciliary body was not significantly altered by dinitrophenol, phloretin, or phlorhizin. The observation that the efflux of labelled ascorbate was not diminished by either dinitrophenol or ouabain supports the concept that the process of ascorbate efflux from the iris-ciliary body is passive and not sodium dependent. It was demonstrated that a vigorous rate of [14C]ascorbate efflux from the iris-ciliary body persisted even when the external ascorbate level was elevated to 1 mM, the concentration found in aqueous humor.


Gut | 2010

A novel mechanism of acid and bile acid-induced DNA damage involving Na+/H+ exchanger: implication for Barrett's oesophagus

Aaron Goldman; Mohammad Shahidullah; David S. Goldman; Ludmila Khailova; George S. Watts; Nicholas A. Delamere; Katerina Dvorak

Objective Barretts oesophagus is a premalignant disease associated with oesophageal adenocarcinoma. The major goal of this study was to determine the mechanism responsible for bile acid-induced alteration in intracellular pH (pHi) and its effect on DNA damage in cells derived from normal oesophagus (HET1A) or Barretts oesophagus (CP-A). Design Cells were exposed to bile acid cocktail (BA) and/or acid in the presence/absence of inhibitors of nitric oxide synthase (NOS) or sodium–hydrogen exchanger (NHE). Nitric oxide (NO), pHi and DNA damage were measured by fluorescent imaging and comet assay. Expression of NHE1 and NOS in cultured cells and biopsies from Barretts oesophagus or normal squamous epithelium was determined by RT-PCR, immunoblotting or immunohistochemistry. Results A dose dependent decrease in pHi was observed in CP-A cells exposed to BA. This effect of BA is the consequence of NOS activation and increased NO production, which leads to NHE inhibition. Exposure of oesophageal cells to acid in combination with BA synergistically decreased pHi. The decrease was more pronounced in CP-A cells and resulted in >2-fold increase in DNA damage compared to acid treatment alone. Examination of biopsies and cell lines revealed robust expression of NHE1 in Barretts oesophagus but an absence of NHE1 in normal epithelium. Conclusions The results of this study identify a new mechanism of bile acid-induced DNA damage. We found that bile acids present in the refluxate activate immediately all three isoforms of NOS, which leads to an increased NO production and NHE inhibition. This consequently results in increased intracellular acidification and DNA damage, which may lead to mutations and cancer progression. Therefore, we propose that in addition to gastric reflux, bile reflux should be controlled in patients with Barretts oesophagus.

Collaboration


Dive into the Nicholas A. Delamere's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shigeo Tamiya

University of Louisville

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy E. Moseley

University of Louisville

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard N. Williams

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge