Network


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

Hotspot


Dive into the research topics where Yazen Alnouti is active.

Publication


Featured researches published by Yazen Alnouti.


Journal of Chromatography B | 2008

Quantitative-Profiling of Bile Acids and their Conjugates in Mouse Liver, Bile, Plasma, and Urine Using LC-MS/MS

Yazen Alnouti; Iván L. Csanaky; Curtis D. Klaassen

The differences among individual bile acids (BAs) in eliciting different physiological and pathological responses are largely unknown because of the lack of valid and simple analytical methods for the quantification of individual BAs and their taurine and glycine conjugates. Therefore, a simple and sensitive LC-MS/MS method for the simultaneous quantification of 6 major BAs, their glycine, and taurine conjugates in mouse liver, bile, plasma, and urine was developed and validated. One-step sample preparation using solid-phase extraction (for bile and urine) or protein precipitation (for plasma and liver) was used to extract BAs. This method is valid and sensitive with a limit of quantification ranging from 10 to 40 ng/ml for the various analytes, has a large dynamic range (2500), and a short run time (20 min). Detailed BA profiles were obtained from mouse liver, plasma, bile, and urine using this method. Muricholic acid (MCA) and cholic acid (CA) taurine conjugates constituted more than 90% of BAs in liver and bile. BA concentrations in liver were about 300-fold higher than in plasma, and about 180-fold higher in bile than in liver. In summary, a reliable and simple LC-MS/MS method to quantify major BAs and their metabolites was developed and applied to quantify BAs in mouse tissues and fluids.


Toxicological Sciences | 2009

Bile Acid Sulfation: A Pathway of Bile Acid Elimination and Detoxification

Yazen Alnouti

Sulfotransferase-2A1 catalyzes the formation of bile acid-sulfates (BA-sulfates). Sulfation of BAs increases their solubility, decreases their intestinal absorption, and enhances their fecal and urinary excretion. BA-sulfates are also less toxic than their unsulfated counterparts. Therefore, sulfation is an important detoxification pathway of BAs. Major species differences in BA sulfation exist. In humans, only a small proportion of BAs in bile and serum are sulfated, whereas more than 70% of BAs in urine are sulfated, indicating their efficient elimination in urine. The formation of BA-sulfates increases during cholestatic diseases. Therefore, sulfation may play an important role in maintaining BA homeostasis under pathologic conditions. Farnesoid X receptor, pregnane X receptor, constitutive androstane receptor, and vitamin D receptor are potential nuclear receptors that may be involved in the regulation of BA sulfation. This review highlights current knowledge about the enzymes and transporters involved in the formation and elimination of BA-sulfates, the effect of sulfation on the pharmacologic and toxicologic properties of BAs, the role of BA sulfation in cholestatic diseases, and the regulation of BA sulfation.


Drug Metabolism and Disposition | 2005

TISSUE DISTRIBUTION AND ONTOGENY OF ORGANIC CATION TRANSPORTERS IN MICE

Yazen Alnouti; Jay S. Petrick; Curtis D. Klaassen

Organic cation transporters (Octs) play an important role in transporting cationic xeno- and endobiotics across biological membranes. Little is known about Octs in mice; therefore, the tissue distribution and developmental changes in the mRNA expression of Octs in mice were quantified. Oct1, Oct2, Oct3, Octn1, Octn2, and Octn3 mRNA expression was quantified in 14 tissues from male and female mice using the branched DNA signal amplification assay. Oct1 mRNA expression was highest in kidney, followed by liver. Oct2 mRNA was almost exclusively expressed in kidney, with male mice having twice that in female mice. The higher expression of Oct2 in male mice is due to testosterone. Oct3 mRNA was most highly expressed in placenta, ovary, and uterus, but was expressed at low levels in most tissues. Octn1 and Octn2 mRNA expression was similar, with the highest levels in kidney followed by small intestine. Octn3 mRNA was almost exclusively expressed in testes. The developmental expression of Oct1, Oct2, Octn1, and Octn2 mRNA in kidneys as well as Oct1 in liver was determined in young mice. Ontogenic expression data indicate that each of the Octs approached adult expression levels by about 3 weeks of age. The gender difference in Oct mRNA expression did not become apparent until day 30 after birth. The differences in tissue distribution of the Octs may play an important role in drug disposition to various tissues. Furthermore, low expression of the Octs in young animals may affect the pharmacokinetic behavior of drugs compared with that in adults.


AIDS | 2012

Long-acting nanoformulated antiretroviral therapy elicits potent antiretroviral and neuroprotective responses in HIV-1-infected humanized mice.

Prasanta K. Dash; Howard E. Gendelman; Upal Roy; Shantanu Balkundi; Yazen Alnouti; R.L. Mosley; Harris A. Gelbard; JoEllyn McMillan; Santhi Gorantla; Larisa Y. Poluektova

Objectives:Long-acting nanoformulated antiretroviral therapy (nanoART) with improved pharmacokinetics, biodistribution and limited systemic toxicities will likely improve drug adherence and access to viral reservoirs. Design:Atazanavir and ritonavir crystalline nanoART were formulated in a poloxamer-188 excipient by high-pressure homogenization. These formulations were evaluated for antiretroviral and neuroprotective activities in humanized NOD/scid-IL-2Rgcnull (NSG) mice. Methods:NanoART-treated NSG mice were evaluated for drug biodistribution, pharmacodynamics and toxicity. CD34+ human hematopoietic stem cells were transplanted at birth in replicate NSG mice. The mice were infected with HIV-1ADA at 5 months of age. Eight weeks later, the infected animals were treated with weekly subcutaneous injections of nanoformulated ATV and RTV. Peripheral viral load, CD4+ T-cell counts and lymphoid and brain histopathology and immunohistochemistry tests were performed. Results:NanoART treatments by once-a-week injections reduced viral loads more than 1000-fold and protected CD4+ T-cell populations. This paralleled high ART levels in liver, spleen and blood that were in or around the human minimal effective dose concentration without notable toxicities. Importantly, examination of infected brain subregions showed that nanoART elicited neuroprotective responses with detectable increases in microtubule-associated protein-2, synaptophysin and neurofilament expression when compared to untreated virus-infected animals. Therapeutic interruptions produced profound viral rebounds. Conclusion:Long-acting nanoART has translational potential with sustained and targeted efficacy and with limited systemic toxicities. Such success in drug delivery and distribution could improve drug adherence and reduce viral resistance in infected people.


Journal of Pharmacology and Experimental Therapeutics | 2007

Regulation of Sulfotransferase Enzymes by Prototypical Microsomal Enzyme Inducers in Mice

Yazen Alnouti; Curtis D. Klaassen

In the present study, the regulation of the mRNA of 11 sulfotransferases (Sults) and two 3′-phosphoadenosine 5′-phosphosulfate synthase (PAPSs) isozymes by 15 microsomal enzyme inducers (MEI) in livers of male mice and five MEIs in livers of female mice was examined. These MEIs represent the transcriptionally mediated pathways: aryl hydrocarbon receptor (AhR), pregnane X receptor (PXR), constitutive androstane receptor (CAR), peroxisomal proliferator-activated receptor α (PPARα), and NF-E2-related factor 2 (Nrf2). AhR ligands suppress the expression of Sults, especially the Sult1 isoenzymes in female mice. CAR activators up-regulate several Sults and PAPSs2 in female but not in male mice. PXR ligands cause marked induction of Sult1e1 in male, Sult2a1/2a2 in female, and PAPSs2 in both male and female mice. PPARα ligands do not have a marked effect on Sult expression in males, but they tend to suppress the expression of several Sult isoforms in female mice. Nrf2 activators appear to induce the mRNA expression of Sults in male and have mixed effects in female mice. In silico analysis indicated the presence of putative binding sites for all five transcription factors in the promoter region of many Sult and PAPSs isoforms. In conclusion, induction of Sults by typical MEIs is not as marked as the induction of P450 enzymes in mice. In addition to gender differences in basal expression of Sults, there is also a marked gender difference in the inducibility of various Sult isoenzymes in mice by MEIs.


Journal of Pharmaceutical and Biomedical Analysis | 2011

Simultaneous characterization of bile acids and their sulfate metabolites in mouse liver, plasma, bile, and urine using LC–MS/MS

Jiangeng Huang; Sai Praneeth R. Bathena; Iván L. Csanaky; Yazen Alnouti

Sulfation is a major metabolic pathway involved in the elimination and detoxification of bile acids (BAs). Several lines of evidence are available to support the role of sulfation as a defensive mechanism to attenuate the toxicity of accumulated BAs during hepatobiliary diseases. Individual BAs and their sulfate metabolites vary markedly in their physiological roles as well as their toxicities. Therefore, analytical techniques are required for the quantification of individual BAs and BA-sulfates in biological fluids and tissues. Here we report a simple, sensitive, and validated LC-MS/MS method for the simultaneous quantification of major BAs and BA-sulfates in mouse liver, plasma, bile, and urine. One-step sample preparation using solid-phase extraction (for bile and urine) or protein precipitation (for liver and plasma) was used to extract BAs and BA-sulfates. Base-line separation of all analytes (unsulfated- and sulfated BAs) was achieved in 25min with a limit of quantification of 1ng/ml. This LC-MS/MS method was applied to simultaneously quantify BAs and BA-sulfates in both male and female mouse tissues and fluids. Less than 3% of total BAs are present in the sulfate form in the mouse liver, plasma, and bile, which provides strong evidence that sulfation is a minor metabolic pathway of BA elimination and detoxification in mice. Furthermore, we report that the marked female-predominant expression of Sult2a1 is not reflected into a female-predominant pattern of BA-sulfation.


The Journal of Infectious Diseases | 2012

Pharmacodynamic and Antiretroviral Activities of Combination Nanoformulated Antiretrovirals in HIV-1–Infected Human Peripheral Blood Lymphocyte–Reconstituted Mice

Upal Roy; JoEllyn McMillan; Yazen Alnouti; Nagsen Gautum; Nathan Smith; Shantanu Balkundi; Prasanta K. Dash; Santhi Gorantla; Andrea Martinez-Skinner; Jane L. Meza; Georgette D. Kanmogne; Susan Swindells; Samuel M. Cohen; R. Lee Mosley; Larisa Y. Poluektova; Howard E. Gendelman

Lack of adherence, inaccessibility to viral reservoirs, long-term drug toxicities, and treatment failures are limitations of current antiretroviral therapy (ART). These limitations lead to increased viral loads, medicine resistance, immunocompromise, and comorbid conditions. To this end, we developed long-acting nanoformulated ART (nanoART) through modifications of existing atazanavir, ritonavir, and efavirenz suspensions in order to establish cell and tissue drug depots to achieve sustained antiretroviral responses. NanoARTs abilities to affect immune and antiviral responses, before or following human immunodeficiency virus type 1 infection were tested in nonobese severe combined immune-deficient mice reconstituted with human peripheral blood lymphocytes. Weekly subcutaneous injections of drug nanoformulations at doses from 80 mg/kg to 250 mg/kg, 1 day before and/or 1 and 7 days after viral exposure, elicited drug levels that paralleled the human median effective concentration, and with limited toxicities. NanoART treatment attenuated viral replication and preserved CD4(+) Tcell numbers beyond that seen with orally administered native drugs. These investigations bring us one step closer toward using long-acting antiretrovirals in humans.


Antimicrobial Agents and Chemotherapy | 2013

Preclinical Pharmacokinetics and Tissue Distribution of Long-Acting Nanoformulated Antiretroviral Therapy

Nagsen Gautam; Upal Roy; Shantanu Balkundi; Pavan Puligujja; Dongwei Guo; Nathan Smith; Xin Ming Liu; Benjamin G. Lamberty; Brenda Morsey; Howard S. Fox; JoEllyn McMillan; Howard E. Gendelman; Yazen Alnouti

ABSTRACT Long-acting injectable nanoformulated antiretroviral therapy (nanoART) was developed with the explicit goal of improving medicine compliance and for drug targeting of viral tissue reservoirs. Prior nanoART studies completed in humanized virus-infected mice demonstrated sustained antiretroviral responses. However, the pharmacokinetics (PK) and tissue distribution of nanoART were not characterized. To this end, the PK and tissue distribution of nanoformulated atazanavir (ATV) and ritonavir (RTV) injected subcutaneously or intramuscularly in mice and monkeys were evaluated. Fourteen days after injection, ATV and RTV levels were up to 13-, 41-, and 4,500-fold higher than those resulting from native-drug administration in plasma, tissues, and at the site of injection, respectively. At nanoART doses of 10, 50, 100, and 250 mg/kg of body weight, relationships of more- and less-than-proportional increases in plasma and tissue levels with dose increases were demonstrated with ATV and RTV. Multiple-dose regimens showed serum and tissue concentrations up to 270-fold higher than native-drug concentrations throughout 8 weeks of study. Importantly, nanoART was localized in nonlysosomal compartments in tissue macrophages, creating intracellular depot sites. Reflective data were obtained in representative rhesus macaque studies. We conclude that nanoART demonstrates blood and tissue antiretroviral drug levels that are enhanced compared to those of native drugs. The sustained and enhanced PK profile of nanoART is, at least in part, the result of the sustained release of ATV and RTV from tissue macrophases and at the site of injection.


International Journal of Nanomedicine | 2012

Mononuclear phagocyte intercellular crosstalk facilitates transmission of cell-targeted nanoformulated antiretroviral drugs to human brain endothelial cells.

Georgette D. Kanmogne; Sangya Singh; Upal Roy; Xinming Liu; JoEllyn McMillan; Santhi Gorantla; Shantanu Balkundi; Nathan Smith; Yazen Alnouti; Nagsen Gautam; You Zhou; Larisa Y. Poluektova; Alexander V. Kabanov; Tatiana K. Bronich; Howard E. Gendelman

Despite the successes of antiretroviral therapy (ART), HIV-associated neurocognitive disorders remain prevalent in infected people. This is due, in part, to incomplete ART penetration across the blood–brain barrier (BBB) and lymph nodes and to the establishment of viral sanctuaries within the central nervous system. In efforts to improve ART delivery, our laboratories developed a macrophage-carriage system for nanoformulated crystalline ART (nanoART) (atazanavir, ritonavir, indinavir, and efavirenz). We demonstrate that nanoART transfer from mononuclear phagocytes (MP) to human brain microvascular endothelial cells (HBMEC) can be realized through cell-to-cell contacts, which can facilitate drug passage across the BBB. Coculturing of donor MP containing nanoART with recipient HBMEC facilitates intercellular particle transfer. NanoART uptake was observed in up to 52% of HBMEC with limited cytotoxicity. Folate coating of nanoART increased MP to HBMEC particle transfer by up to 77%. To translate the cell assays into relevant animal models of disease, ritonavir and atazanavir nanoformulations were injected into HIV-1-infected NOD/scid-γcnull mice reconstituted with human peripheral blood lymphocytes. Atazanavir and ritonavir levels in brains of mice treated with folate-coated nanoART were three- to four-fold higher than in mice treated with noncoated particles. This was associated with decreased viral load in the spleen and brain, and diminished brain CD11b-associated glial activation. We postulate that monocyte-macrophage transfer of nanoART to brain endothelial cells could facilitate drug entry into the brain.


Bioorganic & Medicinal Chemistry Letters | 2010

Praziquantel analogs with activity against juvenile Schistosoma mansoni.

Yuxiang Dong; Jacques Chollet; Mireille Vargas; Nuha R. Mansour; Q. D. Bickle; Yazen Alnouti; Jiangeng Huang; Jennifer Keiser; Jonathan L. Vennerstrom

Six amide and four urea derivatives of praziquantel were synthesized and tested for antischistosomal activity against juvenile and adults stages of Schistosoma mansoni in infected mice. Only one of these had significant activity against adult worms, but, unlike praziquantel, six of these had low to modest activity against juvenile worms. A praziquantel ketone derivative had the best combination of activity against juveniles and adults, but it had no effect on the motility of adult S. mansoni in ex vivo culture. Cytochrome P450 metabolic stability data support the hypothesis that the major trans-cyclohexanol metabolite of praziquantel plays an important role in the antischistosomal activity of this drug.

Collaboration


Dive into the Yazen Alnouti's collaboration.

Top Co-Authors

Avatar

Nagsen Gautam

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Howard E. Gendelman

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

JoEllyn McMillan

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rhishikesh Thakare

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sai Praneeth R. Bathena

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Benson Edagwa

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jiangeng Huang

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Larisa Y. Poluektova

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Santhi Gorantla

University of Nebraska Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge