Network


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

Hotspot


Dive into the research topics where James Randazzo is active.

Publication


Featured researches published by James Randazzo.


Journal of Medicinal Chemistry | 2010

Multifunctional antioxidants for the treatment of age-related diseases.

Hongxia Jin; James Randazzo; Peng Zhang; Peter F. Kador

Analogues of N,N-dimethyl-4-(pyrimidin-2-yl)piperazine-1-sulfonamide possessing a free radical scavenger group (FRS), chelating groups (CHL), or both (FRS + CHL) have been synthesized. Electrospray ionization mass spectrometry studies indicate that select members of this series bind ions in the relative order of Cu(1+) = Cu(2+) > Fe(2+) = Fe(3+) > Zn(2+) with no binding of Ca(2+) or Mg(2+) observed. In vitro evaluation of these compounds in human lens epithelial, human retinal pigmented epithelial, and human hippocampal astrocyte cell lines indicates that all analogues possessing the FRS group as well as the water-soluble vitamin E analogue 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid protect these cells against decreased cell viability and glutathione levels induced by hydrogen peroxide. In addition, those compounds possessing CHL groups also protected these cells against hydroxyl radicals generated by the Fenton reaction. These compounds are good candidates for the preventive treatment of cataract, age-related macular degeneration (AMD), and Alzheimers dementia (AD).


Experimental Eye Research | 2012

Osmotic stress, not aldose reductase activity, directly induces growth factors and MAPK signaling changes during sugar cataract formation.

Peng Zhang; Kuiyi Xing; James Randazzo; Karen Blessing; Marjorie F. Lou; Peter F. Kador

In sugar cataract formation in rats, aldose reductase (AR) activity is not only linked to lenticular sorbitol (diabetic) or galactitol (galactosemic) formation but also to signal transduction changes, cytotoxic signals and activation of apoptosis. Using both in vitro and in vivo techniques, the interrelationship between AR activity, polyol (sorbitol and galactitol) formation, osmotic stress, growth factor induction, and cell signaling changes have been investigated. For in vitro studies, lenses from Sprague Dawley rats were cultured for up to 48 h in TC-199-bicarbonate media containing either 30 mM fructose (control), or 30 mM glucose or galactose with/without the aldose reductase inhibitors AL1576 or tolrestat, the sorbitol dehydrogenase inhibitor (SDI) CP-470,711, or 15 mM mannitol (osmotic-compensated media). For in vivo studies, lenses were obtained from streptozotocin-induced diabetic Sprague Dawley rats fed diet with/without the ARIs AL1576 or tolrestat for 10 weeks. As expected, lenses cultured in high glucose/galactose media or from untreated diabetic rats all showed a decrease in the GSH pool that was lessened by ARI treatment. Lenses either from diabetic rats or from glucose/galactose culture conditions showed increased expression of basic-FGF, TGF-β, and increased signaling through P-Akt, P-ERK1/2 and P-SAPK/JNK which were also normalized by ARIs to the expression levels observed in non-diabetic controls. Culturing rat lenses in osmotically compensated media containing 30 mM glucose or galactose did not lead to increased growth factor expression or altered signaling. These studies indicate that it is the biophysical response of the lens to osmotic stress that results in an increased intralenticular production of basic-FGF and TGF-β and the altered cytotoxic signaling that is observed during sugar cataract formation.


PLOS ONE | 2011

Orally Active Multi-Functional Antioxidants Delay Cataract Formation in Streptozotocin (Type 1) Diabetic and Gamma-Irradiated Rats

James Randazzo; Peng Zhang; Jun Makita; Karen Blessing; Peter F. Kador

Background Age-related cataract is a worldwide health care problem whose progression has been linked to oxidative stress and the accumulation of redox-active metals. Since there is no specific animal model for human age-related cataract, multiple animal models must be used to evaluate potential therapies that may delay and/or prevent cataract formation. Methods/Principal Findings Proof of concept studies were conducted to evaluate 4-(5-hydroxypyrimidin-2-yl)-N,N-dimethyl-3,5-dioxopiperazine-1-sulfonamide (compound 4) and 4-(5-hydroxy-4,6-dimethoxypyrimidin-2-yl)-N,N-dimethyl-3,5-dioxopiperazine-1-sulfonamide (compound 8), multi-functional antioxidants that can independently chelate redox metals and quench free radicals, on their ability to delay the progression of diabetic “sugar” cataracts and gamma radiation-induced cataracts. Prior to 15 Gy of whole head irradiation, select groups of Long Evans rats received either diet containing compound 4 or 8, or a single i.p. injection of panthethine, a radioprotective agent. Compared to untreated, irradiated rats, treatment with pantethine, 4 and 8 delayed initial lens changes by 4, 47, and 38 days, respectively, and the average formation of posterior subcapsular opacities by 23, 53 and 58 days, respectively. In the second study, select groups of diabetic Sprague Dawley rats were administered chow containing compounds 4, 8 or the aldose reductase inhibitor AL1576. As anticipated, treatment with AL1576 prevented cataract by inhibiting sorbitol formation in the lens. However, compared to untreated rats, compounds 4 and 8 delayed vacuole formation by 20 days and 12 days, respectively, and cortical cataract formation by 8 and 3 days, respectively, without reducing lenticular sorbitol. Using in vitro lens culture in 30 mM xylose to model diabetic “sugar” cataract formation, western blots confirmed that multi-functional antioxidants reduced endoplasmic reticulum stress. Conclusions/Significance Multi-functional antioxidants delayed cataract formation in two diverse rat models. These studies provide a proof of concept that a general cataract treatment focused on reducing oxidative stress instead of a specific mechanism of cataractogenesis can be developed.


Journal of Ocular Pharmacology and Therapeutics | 2009

Polyol Formation in Cell Lines of Rat Retinal Capillary Pericytes and Endothelial Cells (TR-rPCT and TR-iBRB)

Peter F. Kador; James Randazzo; Karen Blessing; Jun Makita; Peng Zhang; Kuang Yu; Ken-ichi Hosoya; Tetsuya Terasaki

PURPOSE The two most widely investigated animal models for diabetic retinopathy (DR) are the rat and dog. In dogs, aldose reductase (AR) is present only in retinal capillary pericytes and their destruction has been linked to polyol accumulation and resulting apoptosis. Since both rat capillary pericytes and endothelial cells have been reported to contain AR, the role of polyol pathway activity in capillary cell destruction has been investigated in rat retinal capillary pericyte (TR-rPCT) and endothelial (TR-iBRB) cells. METHODS TR-rPCT and TR-iBRB cell lines were recloned and their identities were reconfirmed by characteristic immunostaining. Cells were cultured up to 72 h in media containing 50 mM glucose or galactose with/without the AR inhibitors or a sorbitol dehydrogenase inhibitor (SDI) or with 30 mM 3-fluoro-3-deoxyglucose. Polyol levels were determined by HPLC or (19)F-NMR. Apoptosis was detected with TUNEL/DAPI staining. RESULTS Smooth muscle actin is present only in pericytes while only endothelial cells stain for von Willebrand factor and accumulate acetylated low-density lipoprotein. AR is present in both cells but AR levels are lower in endothelial cells. Aldehyde reductase is also present in both cells. Cells cultured in 50 mM glucose or galactose show significant polyol accumulation in pericytes but endothelial cells show little accumulation of galactitol and no accumulation of sorbitol. Sorbitol accumulation in pericytes resulted in increased cellular permeability and increased TUNEL staining, which was reduced by AR inhibition. CONCLUSIONS Although both rat retinal pericytes and endothelial cells contain AR, sorbitol accumulation and TUNEL staining primarily occur in pericytes and are inhibited by AR inhibitors.


Journal of Nucleic Acids | 2010

Elevated Levels of DNA Strand Breaks Induced by a Base Analog in the Human Cell Line with the P32T ITPA Variant.

Irina S.-R. Waisertreiger; Miriam R. Menezes; James Randazzo; Youri I. Pavlov

Base analogs are powerful antimetabolites and dangerous mutagens generated endogenously by oxidative stress, inflammation, and aberrant nucleotide biosynthesis. Human inosine triphosphate pyrophosphatase (ITPA) hydrolyzes triphosphates of noncanonical purine bases (i.e., ITP, dITP, XTP, dXTP, or their mimic: 6-hydroxyaminopurine (HAP) deoxynucleoside triphosphate) and thus regulates nucleotide pools and protects cells from DNA damage. We demonstrate that the model purine base analog HAP induces DNA breaks in human cells and leads to elevation of levels of ITPA. A human polymorphic allele of the ITPA, 94C->A encodes for the enzyme with a P32T amino-acid change and leads to accumulation of nonhydrolyzed ITP. The polymorphism has been associated with adverse reaction to purine base-analog drugs. The level of both spontaneous and HAP-induced DNA breaks is elevated in the cell line with the ITPA P32T variant. The results suggested that human ITPA plays a pivotal role in the protection of DNA from noncanonical purine base analogs.


PLOS ONE | 2011

Orally Active Multi-Functional Antioxidants Are Neuroprotective in a Rat Model of Light-Induced Retinal Damage

James Randazzo; Zifeng Zhang; Michael Hoff; Hiroyoshi Kawada; Andrew J. Sachs; Yang Yuan; Neena B. Haider; Peter F. Kador

Background Progression of age-related macular degeneration has been linked to iron dysregulation and oxidative stress that induce apoptosis of neural retinal cells. Since both antioxidants and chelating agents have been reported to reduce the progression of retinal lesions associated with AMD in experimental animals, the present study evaluates the ability of multi-functional antioxidants containing functional groups that can independently chelate redox metals and quench free radicals to protect the retina against light-induced retinal degeneration, a rat model of dry atrophic AMD. Methods/Results Proof of concept studies were conducted to evaluate the ability of 4-(5-hydroxypyrimidin-2-yl)-N,N-dimethyl-3,5-dioxopiperazine-1-sulfonamide (compound 4) and 4-(5-hydroxy-4,6-dimethoxypyrimidin-2-yl)-N,N-dimethyl-3,5-dioxopiperazine-1-sulfonamide (compound 8) to reduce retinal damage in 2-week dark adapted Wistar rats exposed to 1000 lx of light for 3 hours. Assessment of the oxidative stress markers 4- hydroxynonenal and nitrotyrosine modified proteins and Thioredoxin by ELISA and Western blots indicated that these compounds reduced the oxidative insult caused by light exposure. The beneficial antioxidant effects of these compounds in providing significant functional and structural protection were confirmed by electroretinography and quantitative histology of the retina. Conclusions/Significance The present study suggests that multi-functional compounds may be effective candidates for preventive therapy of AMD.


PLOS ONE | 2012

Novel Diabetic Mouse Models as Tools for Investigating Diabetic Retinopathy

Peter F. Kador; Peng Zhang; Jun Makita; Zifeng Zhang; Changmei Guo; James Randazzo; Hiroyoshi Kawada; Neena B. Haider; Karen Blessing

Objective Mouse models possessing green fluorescent protein (GFP) and/or human aldose reductase (hAR) in vascular tissues have been established and crossed with naturally diabetic Akita mice to produce new diabetic mouse models. Research Design and Methods Colonies of transgenic C57BL mice expressing GFP (SMAA-GFP), hAR (SMAA-hAR) or both (SMAA-GFP-hAR) in vascular tissues expressing smooth muscle actin were established and crossbred with C57BL/6-Ins2Akita/J (AK) mice to produce naturally diabetic offspring AK-SMAA-GFP and AK-SMAA-GFP-hAR. Aldose reductase inhibitor AL1576 (ARI) was administered in chow. Retinal and lenticular sorbitol levels were determined by HPLC. Retinal functions were evaluated by electroretinography (ERGs). Growth factor and signaling changes were determined by Western Blots using commercially available antibodies. Retinal vasculatures were isolated from the neural retina by enzymatic digestion. Flat mounts were stained with PAS-hematoxylin and analyzed. Results Akita transgenics developed DM by 8 weeks of age with blood glucose levels higher in males than females. Sorbitol levels were higher in neural retinas of AK-SMAA-GFP-hAR compared to AK-SMAA-GFP mice. AK-SMAA-GFP-hAR mice also had higher VEGF levels and reduced ERG scotopic b-wave function, both of which were normalized by AL1576. AK-SMAA-GFP-hAR mice showed induction of the retinal growth factors bFGF, IGF-1, and TGFβ, as well as signaling changes in P-Akt, P-SAPK/JNK and P-44/42 MAPK that were also reduced by ARI treatment. Quantitative analysis of flat mounts in 18 week AK-SMAA-GFP-hAR mice revealed increased loss of nuclei/capillary length and a significant increase in the percentage of acellular capillaries present which was not seen in AK-SMAA-GFP-hAR treated with ARI. Conclusions/Significance These new mouse models of early onset diabetes may be valuable tools for assessing both the role of hyperglycemia and AR in the development of retinal lesions associated with diabetic retinopathy.


Molecular Vision | 2007

Cellular osmolytes reduce lens epithelial cell death and alleviate cataract formation in galactosemic rats

Michael L. Mulhern; Christian J. Madson; Peter F. Kador; James Randazzo; Toshimichi Shinohara


American Journal of Veterinary Research | 2006

Effects of topical administration of an aldose reductase inhibitor on cataract formation in dogs fed a diet high in galactose

Peter F. Kador; Daniel M. Betts; Milton Wyman; Karen Blessing; James Randazzo


Journal of Ocular Pharmacology and Therapeutics | 2007

Topical Aldose Reductase Inhibitor Formulations for Effective Lens Drug Delivery in a Rat Model for Sugar Cataracts

Peter F. Kador; James Randazzo; Thomas Babb; Kavitha Koushik; Y. Takamura; Wenjun Zhu; Karen Blessing; Uday B. Kompella

Collaboration


Dive into the James Randazzo's collaboration.

Top Co-Authors

Avatar

Karen Blessing

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter F. Kador

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peng Zhang

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

P. F. Kador

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jun Makita

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Zifeng Zhang

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hiroyoshi Kawada

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marjorie F. Lou

University of Nebraska–Lincoln

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge