Network


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

Hotspot


Dive into the research topics where Monicah A. Otieno is active.

Publication


Featured researches published by Monicah A. Otieno.


Clinical Pharmacology & Therapeutics | 2018

Primary Human Lung Alveolus‐on‐a‐chip Model of Intravascular Thrombosis for Assessment of Therapeutics

Abhishek Jain; Riccardo Barrile; A. D. van der Meer; Akiko Mammoto; K. De Ceunynck; Omozuanvbo Aisiku; Monicah A. Otieno; Calvert Louden; Geraldine A. Hamilton; Robert Flaumenhaft; Donald E. Ingber

Pulmonary thrombosis is a significant cause of patient mortality; however, there are no effective in vitro models of thrombi formation in human lung microvessels that could also assess therapeutics and toxicology of antithrombotic drugs. Here, we show that a microfluidic lung alveolus‐on‐a‐chip lined by human primary alveolar epithelium interfaced with endothelium and cultured under flowing whole blood can be used to perform quantitative analysis of organ‐level contributions to inflammation‐induced thrombosis. This microfluidic chip recapitulates in vivo responses, including platelet‐endothelial dynamics and revealed that lipopolysaccharide (LPS) endotoxin indirectly stimulates intravascular thrombosis by activating the alveolar epithelium, rather than acting directly on endothelium. This model is also used to analyze inhibition of endothelial activation and thrombosis due to a protease activated receptor‐1 (PAR‐1) antagonist, demonstrating its ability to dissect complex responses and identify antithrombotic therapeutics. Thus, this methodology offers a new approach to study human pathophysiology of pulmonary thrombosis and advance drug development.


Toxicological Sciences | 2016

Beyond miR-122: Identification of microRNA alterations in blood during a time course of hepatobiliary injury and biliary hyperplasia in rats

Rachel J. Church; Monicah A. Otieno; James Eric McDuffie; Bhanu Singh; Manisha Sonee; Le Roy Hall; Paul B. Watkins; Heidrun Ellinger-Ziegelbauer; Alison H. Harrill

Identification of circulating microRNAs for the diagnosis of liver injury and as an indicator of underlying pathology has been the subject of recent investigations. While several studies have been conducted, with particular emphasis on miR-122, the timing of miRNA release into the circulation and anchoring to tissue pathology has not been systematically evaluated. In this study, miRNA profiling was conducted over a time course of hepatobiliary injury and repair using alpha-naphthylisothiocyanate (ANIT) and a proprietary compound, FP004BA. ANIT administration (50 mg/kg) to rats caused significant biliary epithelial cell and hepatocellular necrosis between 24 and 72 h, followed by resolution and progression to biliary hyperplasia by 120 h which was associated with miRNA release into the blood. FP004BA (100 mg/kg) was used to confirm associations of miRNA along a time course with similar hepatic pathology to ANIT. Treatment with ANIT or FP004BA resulted in significant alterations of overlapping miRNAs during the early and peak injury phases. In addition to well-characterized liver injury markers miR-122-5p and miR-192-5p, multiple members of the 200 family and the 101 family along with miR-802-5p and miR-30d-5p were consistently elevated during hepatobiliary injury caused by both toxicants, suggesting that these species may be potential biomarker candidates for hepatobiliary injury. After 14 days of dosing with 4BA, miR-182-5p remained elevated-while miR-122-5p and miR-192-5p had returned to baseline-suggesting that miR-182-5p may have added utility to monitor for hepatobiliary injury in the repair phases when there remains histological evidence of ongoing cellular injury.


Toxicology Research | 2014

MicroRNA-34c-3p is an early predictive biomarker for doxorubicin-induced glomerular injury progression in male Sprague-Dawley rats

Rachel J. Church; J. Eric McDuffie; Manisha Sonee; Monicah A. Otieno; Jing Ying Ma; Xuejun Liu; Paul B. Watkins; Alison H. Harrill

Recently, eight urinary protein biomarkers were qualified for renal toxicity prediction in drug development; however, there are no biomarkers unique to glomerular toxicity. Albuminuria is a hallmark biomarker for primary glomerular injury but lacks specificity. MicroRNA species associated with genes that regulate kidney injury could potentially be used as biomarkers of nephrotoxicity. In this study, microRNA and protein expression changes in urine, blood, and/or kidneys, in addition to histopathology were evaluated in male Sprague-Dawley rats following weekly intravenous injections of doxorubicin (5 mg kg−1 per dose). Following the first administration, urinary miRNA-34c-3p was significantly increased on day 2 and remained elevated on day 7. Urinary osteopontin was significantly increased on day 2 only. Significant urinary albumin was detected on day 7, in the absence of histopathological findings. Following a second doxorubicin administration on day 7, significantly increased urinary kidney injury molecule 1, cystatin C, β2-microglobulin, total protein, and neutrophil gelatinase-associated lipocalin concentrations were detected on day 14. These alterations were concurrent to significant and progressive albuminuria, urinary miR-34c-3p, remarkable microscopic primary glomerular injury and secondary tubular alterations. Urinary miR-34c-3p elevations were predictive of histopathologic injury progression and outperformed the traditional renal biomarkers, serum creatinine and blood urea nitrogen, which did not increase with treatment. MiR-34c-3p was also significantly enriched in damaged glomeruli compared to adjacent nonglomerular tissue. Taken together, miR-34c-3p was identified as a highly sensitive candidate renal safety biomarker with relative specificity, particularly for early prediction of doxorubicin-induced glomerular injury progression in male Sprague-Dawley rats.


Toxicological Sciences | 2018

Fasiglifam (TAK-875): Mechanistic Investigation and Retrospective Identification of Hazards for Drug Induced Liver Injury

Monicah A. Otieno; Jan Snoeys; Wing W. Lam; Avi Ghosh; Mark R. Player; Alessandro Pocai; Rhys Salter; Damir Simic; Hollie Skaggs; Bhanu Singh; Heng-Keang Lim

TAK-875, a GPR40 agonist, was withdrawn from Phase III clinical trials due to drug-induced liver injury (DILI). Mechanistic studies were conducted to identify potential DILI hazards (covalent binding burden (CVB), hepatic transporter inhibition, mitochondrial toxicity, and liver toxicity in rats) associated with TAK-875. Treatment of hepatocytes with radiolabeled TAK-875 resulted in a CVB of 2.0 mg/day, which is above the threshold of 1 mg/day considered to be a risk for DILI. Covalent binding to hepatocytes was due to formation of a reactive acyl glucuronide (AG) and, possibly, an acyl-CoA thioester intermediate. Formation of TAK-875AG in hepatocytes and/or in vivo was in the order of non-rodents > human (in vitro only) > rat. These data suggest that non-rodents, and presumably humans, form TAK-875AG more efficiently than rats, and that AG-mediated toxicities in rats may only occur at high doses. TAK-875 (1000 mg/kg/day) formed significant amounts of AG metabolite (≤32.7 μM) in rat liver that was associated with increases in ALT (×4), bilirubin (×9), and bile acids (×3.4), and microscopic findings of hepatocellular hypertrophy and single cell necrosis. TAK-875 and TAK-875AG had similar potencies (within 3-fold) for human multi-drug resistant associated protein 2/4 (MRP2/4) and bile salt export pump, but TAK-875AG was exceptionally potent against MRP3 (0.21 μM). Inhibition of MRPs may contribute to liver accumulation of TAK-875AG. TAK-875 also inhibited mitochondrial respiration in HepG2 cells, and mitochondrial Complex 1 and 2 activities in isolated rat mitochondria. In summary, formation of TAK-875AG, and possibly TAK-875CoA in hepatocytes, coupled with inhibition of hepatic transporters and mitochondrial respiration may be key contributors to TAK-875-mediated DILI.


Experimental Biology and Medicine | 2017

Navigating tissue chips from development to dissemination: A pharmaceutical industry perspective

Lorna Ewart; Kristin Fabre; Ananthsrinivas Chakilam; Yvonne P. Dragan; David B Duignan; Jeetu Eswaraka; Jinping Gan; Peggy Guzzie-Peck; Monicah A. Otieno; Claire Jeong; Douglas A. Keller; Sonia M de Morais; Jonathan A. Phillips; William Proctor; Radhakrishna Sura; Terry Van Vleet; David E. Watson; Yvonne Will; Danilo A. Tagle; Brian R. Berridge

Tissue chips are poised to deliver a paradigm shift in drug discovery. By emulating human physiology, these chips have the potential to increase the predictive power of preclinical modeling, which in turn will move the pharmaceutical industry closer to its aspiration of clinically relevant and ultimately animal-free drug discovery. Despite the tremendous science and innovation invested in these tissue chips, significant challenges remain to be addressed to enable their routine adoption into the industrial laboratory. This article describes the main steps that need to be taken and highlights key considerations in order to transform tissue chip technology from the hands of the innovators into those of the industrial scientists. Written by scientists from 13 pharmaceutical companies and partners at the National Institutes of Health, this article uniquely captures a consensus view on the progression strategy to facilitate and accelerate the adoption of this valuable technology. It concludes that success will be delivered by a partnership approach as well as a deep understanding of the context within which these chips will actually be used. Impact statement The rapid pace of scientific innovation in the tissue chip (TC) field requires a cohesive partnership between innovators and end users. Near term uptake of these human-relevant platforms will fill gaps in current capabilities for assessing important properties of disposition, efficacy and safety liabilities. Similarly, these platforms could support mechanistic studies which aim to resolve challenges later in development (e.g. assessing the human relevance of a liability identified in animal studies). Building confidence that novel capabilities of TCs can address real world challenges while they themselves are being developed will accelerate their application in the discovery and development of innovative medicines. This article outlines a strategic roadmap to unite innovators and end users thus making implementation smooth and rapid. With the collective contributions from multiple international pharmaceutical companies and partners at National Institutes of Health, this article should serve as an invaluable resource to the multi-disciplinary field of TC development.


Clinical Pharmacology & Therapeutics | 2018

Organ‐on‐Chip Recapitulates Thrombosis Induced by an anti‐CD154 Monoclonal Antibody: Translational Potential of Advanced Microengineered Systems

Riccardo Barrile; Andries Dirk van der Meer; Hyoungshin Park; Jacob P. Fraser; Damir Simic; Fang Teng; David Conegliano; Justin Nguyen; Abhishek Jain; Mimi Zhou; Katia Karalis; Donald E. Ingber; Geraldine A. Hamilton; Monicah A. Otieno

Clinical development of Hu5c8, a monoclonal antibody against CD40L intended for treatment of autoimmune disorders, was terminated due to unexpected thrombotic complications. These life‐threatening side effects were not discovered during preclinical testing due to the lack of predictive models. In the present study, we describe the development of a microengineered system lined by human endothelium perfused with human whole blood, a “Vessel‐Chip.” The Vessel‐Chip allowed us to evaluate key parameters in thrombosis, such as endothelial activation, platelet adhesion, platelet aggregation, fibrin clot formation, and thrombin anti‐thrombin complexes in the Chip‐effluent in response to Hu5c8 in the presence of soluble CD40L. Importantly, the observed prothrombotic effects were not observed with Hu5c8‐IgG2σ designed with an Fc domain that does not bind the FcγRIIa receptor, suggesting that this approach may have a low potential risk for thrombosis. Our results demonstrate the translational potential of Organs‐on‐Chips, as advanced microengineered systems to better predict human response.


Clinical and Applied Thrombosis-Hemostasis | 2017

Blocking α5β1 Integrin Attenuates sCD40L-Mediated Platelet Activation:

Damir Simic; Nancy Bogdan; Fang Teng; Monicah A. Otieno

The soluble form of CD40L (sCD40L) is a platelet-derived mediator that links inflammation, hemostasis, and vascular dysfunction. Indeed, blockade of CD40L by neutralizing antibodies or genetic disruption in mice prevents atherosclerosis and atherothrombosis. Until recently, it was believed that CD40 and αIIbβ3 were the only receptors on platelets responsible for binding sCD40L, leading to platelet activation and initiation of thrombotic events. Recent findings showed α5β1 integrin as a novel platelet sCD40L receptor, with an unknown function. For the first time, using anti-α5β1 blocking antibodies, we show that sCD40L/α5β1 interaction leads to platelet activation as evaluated in the human whole blood. Establishing α5β1 integrin’s role in platelet activation, and therefore thrombosis will help further shed light on the etiology of thrombotic disease.


Toxicological Sciences | 2008

Mechanistic Investigation of N,N-Diethyl-4-(phenyl-piperidin-4-ylidenemethyl)-benzamide–Induced Insulin Depletion in the Rat and RINm5F Cells

Monicah A. Otieno; Nicole Bavuso; Joseph Milano; Linda Foster-Brown; Khanh-Hui Bui; Yan Li; Thomas J. Hudzik; Debra Wescott; Calvert Louden; Martin Dyroff; François Pognan

These studies describe the effect of N,N-diethyl-4-(phenyl-piperidin-4-ylidenemethyl)-benzamide (AR-M100390), a delta-opioid agonist, on the pancreas and its mechanisms for pancreatic toxicity. Rats were treated with 5, 100, and 600 micromol/kg of AR-M100390 for 3 and/or 7 days; another group of rats treated with 600 micromol/kg of compound were allowed to recover for 14 days. AR-M100390 (600 micromol/kg) caused vacuolation in the beta-cell of the rat pancreas that was associated with depletion of insulin and hyperglycemia after 7 days of dosing. The loss of insulin by AR-M100390 was due to specific inhibition of rat insulin2 mRNA transcription in vivo. Insulin depletion and hyperglycemia were reversible. The effects of AR-M100390 in rats were reproduced in the rat pancreatic beta-cell line RINm5F, where it inhibited intracellular insulin content and secretion without affecting cell survival. Loss of insulin in vitro was also a result of specific inhibition of insulin2 mRNA transcription and was reversible. Pretreatment of cells with the delta-opioid antagonist naltrindole or pertussis toxin did not reverse loss of insulin in AR-M100390-treated cells suggesting that the effects were not mediated by the delta-opioid receptor. AR-M100390 inhibited KCl-mediated calcium mobilization in RINm5F cells, suggesting that L-type calcium channels found in these cells and in pancreatic beta-cells may partially play a role in the inhibition of insulin secretion by this compound. In summary, the in vitro and in vivo studies suggest that inhibition of insulin by AR-M100390 is due to a combination of inhibition of insulin synthesis and/or release.


Toxicologic Pathology | 2017

Regulatory Forum Opinion Piece*: Use and Utility of Animal Models of Disease for Nonclinical Safety Assessment: A Pharmaceutical Industry Survey

Sherry J. Morgan; Jessica Couch; Peggy Guzzie-Peck; Douglas A. Keller; Ray Kemper; Monicah A. Otieno; Robert J. Schulingkamp; Thomas W. Jones

An Innovation and Quality (IQ) Consortium focus group conducted a cross-company survey to evaluate current practices and perceptions around the use of animal models of disease (AMDs) in nonclinical safety assessment of molecules in clinical development. The IQ Consortium group is an organization of pharmaceutical and biotechnology companies with the mission of advancing science and technology. The survey queried the utilization of AMDs during drug discovery in which drug candidates are evaluated in efficacy models and limited short-duration non-Good Laboratory Practices (GLP) toxicology testing and during drug development in which drug candidates are evaluated in GLP toxicology studies. The survey determined that the majority of companies used AMDs during drug discovery primarily as a means for proactively assessing potential nonclinical safety issues prior to the conduct of toxicology studies, followed closely by the use of AMDs to better understand toxicities associated with exaggerated pharmacology in traditional toxicology models or to derisk issues when the target is only expressed in the disease state. In contrast, the survey results indicated that the use of AMDs in development is infrequent, being used primarily to investigate nonclinical safety issues associated with targets expressed only in disease states and/or in response to requests from global regulatory authorities.


Toxicological Sciences | 2018

Optimized Methods to Explore the Mechanistic and Biomarker Potential of Hepatocyte-Derived Exosomes in Drug-Induced Liver Injury

Sarah E Thacker; Manisha Nautiyal; Monicah A. Otieno; Paul B. Watkins; Merrie Mosedale

Recent evidence supports that alterations in hepatocyte-derived exosomes (HDE) may play a role in the pathogenesis of drug-induced liver injury (DILI). HDE-based biomarkers also hold promise to improve the sensitivity of existing in vitro assays for predicting DILI liability. Primary human hepatocytes (PHH) provide a physiologically relevant in vitro model to explore the mechanistic and biomarker potential of HDE in DILI. However, optimal methods to study exosomes in this culture system have not been defined. Here we use HepG2 and HepaRG cells along with PHH to optimize methods for in vitro HDE research. We compared the quantity and purity of HDE enriched from HepG2 cell culture medium by 3 widely used methods: ultracentrifugation (UC), OptiPrep density gradient ultracentrifugation (ODG), and ExoQuick (EQ)-a commercially available exosome precipitation reagent. Although EQ resulted in the highest number of particles, UC resulted in more exosomes as indicated by the relative abundance of exosomal CD63 to cellular prohibitin-1 as well as the comparative absence of contaminating extravesicular material. To determine culture conditions that best supported exosome release, we also assessed the effect of Matrigel matrix overlay at concentrations ranging from 0 to 0.25 mg/ml in HepaRG cells and compared exosome release from fresh and cryopreserved PHH from same donor. Sandwich culture did not impair exosome release, and freshly prepared PHH yielded a higher number of HDE overall. Taken together, our data support the use of UC-based enrichment from fresh preparations of sandwich-cultured PHH for future studies of HDE in DILI.

Collaboration


Dive into the Monicah A. Otieno's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fang Teng

Janssen Pharmaceutica

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul B. Watkins

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge