Network


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

Hotspot


Dive into the research topics where Howard P. Hendrickson is active.

Publication


Featured researches published by Howard P. Hendrickson.


PLOS ONE | 2012

Ellagic acid derivatives from Rubus ulmifolius inhibit Staphylococcus aureus biofilm formation and improve response to antibiotics.

Cassandra L. Quave; Miriam Estévez-Carmona; Cesar M. Compadre; Gerren Hobby; Howard P. Hendrickson; Karen E. Beenken; Mark S. Smeltzer

Background Biofilms contribute to the pathogenesis of many forms of Staphylococcus aureus infection. Treatment of these infections is complicated by intrinsic resistance to conventional antibiotics, thus creating an urgent need for strategies that can be used for the prevention and treatment of biofilm-associated infections. Methodology/Principal Findings This study demonstrates that a botanical natural product composition (220D-F2) rich in ellagic acid and its derivatives can limit S. aureus biofilm formation to a degree that can be correlated with increased antibiotic susceptibility. The source of this composition is Rubus ulmifolius Schott. (Rosaceae), a plant used in complementary and alternative medicine in southern Italy for the treatment of skin and soft tissue infections. All S. aureus clonal lineages tested exhibited a reduced capacity to form a biofilm at 220D-F2 concentrations ranging from 50–200 µg/mL, which were well below the concentrations required to limit bacterial growth (530–1040 µg/mL). This limitation was therapeutically relevant in that inclusion of 220D-F2 resulted in enhanced susceptibility to the functionally-distinct antibiotics daptomycin, clindamycin and oxacillin. Testing with kidney and liver cell lines also demonstrated a lack of host cell cytotoxicity at concentrations of 220D-F2 required to achieve these effects. Conclusions/Significance These results demonstrate that extract 220D-F2 from the root of Rubus ulmifolius can be used to inhibit S. aureus biofilm formation to a degree that can be correlated with increased antibiotic susceptibility without toxic effects on normal mammalian cells. Hence, 220D-F2 is a strong candidate for development as a botanical drug for use in the prevention and treatment of S. aureus biofilm-associated infections.


Pharmacology, Biochemistry and Behavior | 2004

(+)-Methamphetamine-induced spontaneous behavior in rats depends on route of (+)METH administration

W. Brooks Gentry; Abid U. Ghafoor; William D. Wessinger; Elizabeth M. Laurenzana; Howard P. Hendrickson; S. Michael Owens

These studies examined the role of (+)-methamphetamine ((+)METH) administration route on spontaneous behavioral activity vs. time relationships, and pharmacokinetic mechanisms for differences in effects. Male Sprague-Dawley rats (n=6 per administration route) received saline and three doses (0.3, 1.0 and 3.0 mg/kg) of (+)METH in a mixed-sequence design by intravenous (iv), subcutaneous (sc) or intraperitoneal (ip) administration. Locomotion and stereotypy were quantified by video-tracking analysis. The effects of (+)METH on spontaneous behavior were dose- and route-dependent. In particular, total locomotor activity was greatest following 3.0 mg/kg intraperitoneally (P<0.05) and stereotypy ratings were greatest following 3.0 mg/kg subcutaneously (P<0.05). In addition, the duration of locomotor effects was greatest after 3.0 mg/kg subcutaneously (P<0.05). Serum pharmacokinetic parameters were determined in separate rats given 3.0 mg/kg by subcutaneous and intraperitoneal administration (n=4 per administration route). The (+)METH elimination half-life was not different between the routes, but the (+)METH AUC was greater (P<0.05), and the (+)METH and (+)-amphetamine (AMP) maximum concentrations occurred later following subcutaneous than after intraperitoneal dosing (P<0.05), increasing and prolonging drug exposure. In conclusion, the overall pattern of (+)METH effects on locomotor activity depend on dose and the route of administration, which affects serum concentration and the time course of behavioral effects.


Journal of Pharmacology and Experimental Therapeutics | 2008

Development and Preclinical Testing of a High-Affinity Single-Chain Antibody against (+)-Methamphetamine

Eric C. Peterson; Elizabeth M. Laurenzana; William T. Atchley; Howard P. Hendrickson; S M Owens

Chronic or excessive (+)-methamphetamine (METH) use often leads to addiction and toxicity to critical organs like the brain. With medical treatment as a goal, a novel single-chain variable fragment (scFv) against METH was engineered from anti-METH monoclonal antibody mAb6H4 (IgG, κ light chain, Kd = 11 nM) and found to have similar ligand affinity (Kd = 10 nM) and specificity as mAb6H4. The anti-METH scFv (scFv6H4) was cloned, expressed in yeast, purified, and formulated as a naturally occurring mixture of monomer (∼75%) and dimer (∼25%). To test the in vivo efficacy of the scFv6H4, male Sprague-Dawley rats (n = 5) were implanted with 3-day s.c. osmotic pumps delivering 3.2 mg/kg/day METH. After reaching steady-state METH concentrations, an i.v. dose of scFv6H4 (36.5 mg/kg, equimolar to the METH body burden) was administered along with a [3H]scFv6H4 tracer. Serum pharmacokinetic analysis of METH and [3H]scFv6H4 showed that the scFv6H4 caused an immediate 65-fold increase in the METH concentrations and a 12-fold increase in the serum METH area under the concentration-time curve from 0 to 480 min after scFv6H4 administration. The scFv6H4 monomer was quickly cleared or converted to multivalent forms with an apparent t1/2λz of 5.8 min. In contrast, the larger scFv6H4 multivalent forms (dimers, trimers, etc.) showed a much longer t1/2λz (228 min), and the significantly increased METH serum molar concentrations correlated directly with scFv6H4 serum molar concentrations. Considered together, these data suggested that the scFv6H4 multimers (and not the monomer) were responsible for the prolonged redistribution of METH into the serum.


Vaccine | 2009

Functional and biological determinants affecting the duration of action and efficacy of anti-(+)-methamphetamine monoclonal antibodies in rats

Elizabeth M. Laurenzana; Howard P. Hendrickson; Dylan Carpenter; Eric C. Peterson; W. Brooks Gentry; Michael West; Yingni Che; F. Ivy Carroll; S. Michael Owens

These studies examined the in vivo pharmacokinetics and efficacy of five anti-methamphetamine monoclonal antibodies (mAbs, K(D) values from 11 to 250 nM) in rats. While no substantive differences in mAb systemic clearance (t(1/2)=6.1-6.9 days) were found, in vivo function was significantly reduced within 1-3 days for four of the five mAbs. Only mAb4G9 was capable of prolonged efficacy, as judged by prolonged high methamphetamine serum concentrations. MAb4G9 also maintained high amphetamine serum concentrations, along with reductions in methamphetamine and amphetamine brain concentrations, indicating neuroprotection. The combination of broad specificity for methamphetamine-like drugs, high affinity, and prolonged action in vivo suggests mAb4G9 is a potentially efficacious medication for treating human methamphetamine-related medical diseases.


Journal of Chromatography B | 2003

Determination of dextromethorphan and its metabolites in rat serum by liquid-liquid extraction and liquid chromatography with fluorescence detection.

Howard P. Hendrickson; Bill J. Gurley; William D. Wessinger

Dextromethorphan is an effective and safe antitussive, but has liabilities with respect to its abuse potential at doses above the therapeutic dose. At these higher doses, people report phencyclidine-like effects from the drug. A number of animal models have suggested that dextrorphan, an active metabolite of dextromethorphan, is responsible for the abuse liability of the parent compound when dextromethorphan is taken at high doses. Full pharmacokinetic profiles in single animals have not been demonstrated in these studies due to a lack of analytical sensitivity and/or selectivity for dextromethorphan and its metabolites. We have developed a low-cost liquid chromatographic method capable of characterizing the concentration-time profile for dextromethorphan and dextrorphan for 8 h in rats following an 18 mg/kg i.p. dose of dextromethorphan. Limits of quantitation (S/N=10) in 100 microL of serum were 0.25, 0.19, 0.27, and 0.22 nmol/mL for 3-hydroxymorphinan, dextrorphan, 3-methoxymorphinan, and dextromethorphan, respectively. Inter-day precision was better than 11% across the dynamic range of the method.


Aaps Journal | 2006

Quantitative determination of total methamphetamine and active metabolites in rat tissue by liquid chromatography with tandem mass spectrometric detection

Howard P. Hendrickson; Elizabeth M. Laurenzana; S. Michael Owens

High-throughput liquid chromatography with tandem mass spectrometric detection (LC-MS/MS) methodology for the determination of methamphetamine (METH), amphetamine (AMP), 4-hydroxymethamphetamine (4-OH-METH), and 4-hydroxyamphetamine (4-OH-AMP) was developed and validated using simple trichloroacetic acid sample treatment. The method was validated in rat serum, brain, and testis. Lower limits-of-quantitation (LOQ) for METH and AMP were 1 ng·mL−1 using positive ion electrospray tandem mass spectrometry (MS/MS). The accuracy of the method was within 25% of the actual values over a wide range of analyte concentrations. The within-assay precision was better than 12% (coefficient of variation). The method was linear over a wide dynamic range (0.3–1000 ng·mL−1). Quantitation was possible in all 3 matrices using only serum standards because of minimal matrix-associated ion effects or the use of an internal standard. Finally, the LC-MS/MS method was used to determine serum, brain, and testis METH and AMP concentrations during a subcutaneous infusion (5.6 mg kg−1 day−1) of METH in rats. Concentrations of 4-OH-AMP and 4-OH-METH were below the LOQ in experimental samples. The bias introduced by using serum calibrators for the determination of METH and AMP concentrations in testis and brain was less than 8% and insignificant relative to the interanimal variability.


Journal of Pharmaceutical and Biomedical Analysis | 2009

Validation of a liquid chromatography-tandem mass spectrometric assay for the quantitative determination of hydrastine and berberine in human serum

Prem K. Gupta; Martha A. Hubbard; Bill J. Gurley; Howard P. Hendrickson

A high throughput liquid chromatography/tandem mass spectrometry (LC-MS/MS) method for the simultaneous determination of berberine and hydrastine in human serum, after oral administration of goldenseal (Hydrastis canadensis L.), was developed using simple acetonitrile treatment of serum samples. Noscapine served as the internal standard. Lower limit of quantification for both analytes was 0.1 ng mL(-1) using positive ion electrospray tandem mass spectrometry (MS/MS). The intra-day (n=5) accuracy and precision of the method for hydrastine was 82+/-8.8%, 97.9+/-2.4% and 96.2+/-3.3%, respectively. The inter-day (n=4) accuracy and precision for hydrastine was 90.0+/-15.17%, 99.9+/-7.1% and 98+/-6.54%, respectively. For berberine quantitation intra-day accuracy and precision was 96.0+/-8.4%, 92.5+/-4.7% and 94.4+/-3.7%, respectively. The respective values for inter-day quantitation were 91.0+/-8.4%, 94.3+/-4.7% and 94.4+/-3.7%. The analytical recovery for hydrastine was 82.4-96.2% and for berberine it was 94.4-96.0%. The analytes and noscapine were stable for 24h at room temperature (CV 5-10%). Matrix ion effects were studied by post-column infusion of hydrastine and berberine, calculation of calibration curve slope precision was obtained using serum from five different subjects, and by comparison of the response of methanol standards and extracted serum samples. The method was further validated by determination of serum pharmacokinetics of hydrastine and berberine after administration of a single oral dose of goldenseal extract containing 77 mg of hydrastine and 132 mg of berberine.


Analytical Methods | 2011

Development of high-throughput HILIC-MS/MS methodology for plasma citrulline determination in multiple species

Prem K. Gupta; Joshua Brown; Prabath G. Biju; John J. Thaden; Nicolaas E. P. Deutz; Sree Kumar; Martin Hauer-Jensen; Howard P. Hendrickson

Circulating citrulline originates almost exclusively from the small intestinal enterocytes in mammals and therefore is a potential biomarker of disease states affecting enterocyte mass including exposure to ionizing radiation. There is a need for a simple and rapid method for citrulline quantification in plasma. To achieve this goal, a high-throughput separation and tandem mass spectrometric detection strategy has been developed and validated in six different species. HILIC separation was achieved on a 1.7 μm fused-core Diol column using an acidic acetonitrile/water gradient. A surrogate analyte (citrulline stable isotope) was used to determine the lower-limit-of-quantitation, extraction recovery, and matrix ion effects. Mass spectrometric detection was achieved in the multiple reaction-monitoring mode using m/z 176 → 159, 177 → 160, and 181 → 164, for citrulline, citrulline+1, and citrulline+5, respectively. The retention time of citrulline and total chromatographic run time were 1.1 min and 2.5 min, respectively, while effectively eliminating matrix-ion effects and achieving baseline separation from the confounding amino acid arginine. Quantitation was precise (CV <4.3%), accurate (90–110%), and sensitive (lower-limit-of-quantitation; 0.125 μM) without interference from the confounding amino acid, arginine. The throughput of the method was enhanced by incorporation of a 96-well filter plate for final sample cleanup. The method was used to determine plasma citrulline in six different species and in mice treated with escalating doses of radiation. This simple, accurate and high-throughput (up to 200 samples/day) methodology provides the first quantitative assay to meet the growing demand for a rapid and simple citrulline assay with high sensitivity.


Radiation Research | 2016

Radioprotective Efficacy of Gamma-Tocotrienol in Nonhuman Primates

Vijay K. Singh; Shilpa Kulkarni; Oluseyi O. Fatanmi; Stephen Y. Wise; Victoria L. Newman; Patricia L.P. Romaine; Howard P. Hendrickson; Jatinder Gulani; Sanchita P. Ghosh; K. Sree Kumar; Martin Hauer-Jensen

The search for treatments to counter potentially lethal radiation-induced injury over the past several decades has led to the development of multiple classes of radiation countermeasures. However, to date only granulocyte colony-stimulating factor (G-CSF; filgrastim, Neupogen)and pegylated G-CSF (pegfilgrastim, Neulasta) have been approved by the United States Food and Drug Administration (FDA) for the treatment of hematopoietic acute radiation syndrome (ARS). Gamma-tocotrienol (GT3) has demonstrated strong radioprotective efficacy in the mouse model, indicating the need for further evaluation in a large animal model. In this study, we evaluated GT3 pharmacokinetics (PK) and efficacy at different doses of cobalt-60 gamma radiation (0.6 Gy/min) using the nonhuman primate (NHP) model. The PK results demonstrated increased area under the curve with increasing drug dose and half-life of GT3. GT3 treatment resulted in reduced group mean neutropenia by 3–5 days and thrombocytopenia by 1–5 days. At 5.8 and 6.5 Gy total-body irradiation, GT3 treatment completely prevented thrombocytopenia. The capability of GT3 to reduce severity and duration of neutropenia and thrombocytopenia was dose dependent; 75 mg/kg treatment was more effective than 37.5 mg/kg treatment after a 5.8 Gy dose. However, the higher GT3 dose (75 mg/kg) was associated with higher frequency of adverse skin effects (small abscess) at the injection site. GT3 treatment of irradiated NHPs caused no significant difference in animal survival at 60 days postirradiation, however, low mortality was observed in irradiated, vehicle-treated groups as well. The data from this pilot study further elucidate the role and pharmacokinetics of GT3 in hematopoietic recovery after irradiation in a NHP model, and demonstrate the potential of GT3 as a promising radioprotector.


Journal of Medicinal Chemistry | 2015

Combining Active Immunization with Monoclonal Antibody Therapy to Facilitate Early Initiation of a Long-acting Anti-methamphetamine Antibody Response

Michael D. Hambuchen; F. Ivy Carroll; Daniela Rüedi-Bettschen; Howard P. Hendrickson; Leah J. Hennings; Bruce E. Blough; Lawrence E. Brieaddy; Ramakrishna R. Pidaparthi; S. Michael Owens

We hypothesized that an anti-METH mAb could be used in combination with a METH-conjugate vaccine (MCV) to safely improve the overall quality and magnitude of the anti-METH immune response. The benefits would include immediate onset of action (from the mAb), timely increases in the immune responses (from the combined therapy) and duration of antibody response that could last for months (from the MCV). A novel METH-like hapten (METH-SSOO9) was synthesized and then conjugated to immunocyanin monomers of keyhole limpet hemocyanin (IC(KLH)) to create the MCV ICKLH-SOO9. The vaccine, in combination with previously discovered anti-METH mAb7F9, was then tested in rats for safety and potential efficacy. The combination antibody therapy allowed safe achievement of an early high anti-METH antibody response, which persisted throughout the study. Indeed, even after 4 months the METH vaccine antibodies still had the capacity to significantly reduce METH brain concentrations resulting from a 0.56 mg/kg METH dose.

Collaboration


Dive into the Howard P. Hendrickson's collaboration.

Top Co-Authors

Avatar

S. Michael Owens

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Elizabeth M. Laurenzana

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Martin Hauer-Jensen

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Michael D. Hambuchen

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Lin Song

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

S M Owens

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

W. Brooks Gentry

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Peter A. Crooks

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Prem K. Gupta

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Sarah J. White

University of Arkansas for Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge