Network


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

Hotspot


Dive into the research topics where James C. Lamb is active.

Publication


Featured researches published by James C. Lamb.


ALTEX-Alternatives to Animal Experimentation | 2014

t4 workshop report: Lessons learned, challenges, and opportunities: The U.S. endocrine disruptor screening program

Daland R. Juberg; Susan J. Borghoff; Richard A. Becker; Warren Casey; Thomas Hartung; Michael P. Holsapple; M. Sue Marty; Ellen Mihaich; Glen Van Der Kraak; Michael G. Wade; Catherine Willett; Melvin E. Andersen; Christopher J. Borgert; Katherine Coady; Michael Dourson; John R. Fowle; L. Earl Gray; James C. Lamb; Lisa S. Ortego; Thaddeus T. Schug; Colleen Toole; Leah M. Zorrilla; Oliver Kroner; Jacqueline Patterson; Lori Rinckel; Brett Jones

In 1996, the U.S. Congress passed the Food Quality Protection Act and amended the Safe Drinking Water Act (SDWA) requiring the U.S. Environmental Protection Agency (EPA) to implement a screening program to investigate the potential of pesticide chemicals and drinking water contaminants to adversely affect endocrine pathways. Consequently, the EPA launched the Endocrine Disruptor Screening Program (EDSP) to develop and validate estrogen, androgen, and thyroid (EAT) pathway screening assays and to produce standardized and harmonized test guidelines for regulatory application. In 2009, the EPA issued the first set of test orders for EDSP screening and a total of 50 pesticide actives and 2 inert ingredients have been evaluated using the battery of EDSP Tier 1 screening assays (i.e., five in vitro assays and six in vivo assays). To provide a framework for retrospective analysis of the data generated and to collect the insight of multiple stakeholders involved in the testing, more than 240 scientists from government, industry, academia, and non-profit organizations recently participated in a workshop titled Lessons Learned, Challenges, and Opportunities: The U.S. Endocrine Disruptor Screening Program. The workshop focused on the science and experience to date and was organized into three focal sessions: (a) Performance of the EDSP Tier 1 Screening Assays for Estrogen, Androgen, and Thyroid Pathways; (b) Practical Applications of Tier 1 Data; and (c) Indications and Opportunities for Future Endocrine Testing. A number of key learnings and recommendations related to future EDSP evaluations emanated from the collective sessions.


Regulatory Toxicology and Pharmacology | 2014

Critical comments on the WHO-UNEP State of the Science of Endocrine Disrupting Chemicals - 2012.

James C. Lamb; Paolo Boffetta; Warren G. Foster; Julie E. Goodman; Karyn L. Hentz; Lorenz R. Rhomberg; Jane Staveley; Gerard Swaen; Glen Van Der Kraak; Amy Lavin Williams

Early in 2013, the World Health Organization (WHO) released a 2012 update to the 2002 State of the Science of Endocrine Disrupting Chemicals. Several significant concerns have been identified that raise questions about conclusions reached in this report regarding endocrine disruption. First, the report is not a state-of-the-science review and does not follow the 2002 WHO recommended weight-of-evidence approach. Second, endocrine disruption is often presumed to occur based on exposure or a potential mechanism despite a lack of evidence to show that chemicals are causally established as endocrine disruptors. Additionally, causation is often inferred by the presentation of a series of unrelated facts, which collectively do not demonstrate causation. Third, trends in disease incidence or prevalence are discussed without regard to known causes or risk factors; endocrine disruption is implicated as the reason for such trends in the absence of evidence. Fourth, dose and potency are ignored for most chemicals discussed. Finally, controversial topics (i.e., low dose effects, non-monotonic dose response) are presented in a one-sided manner and these topics are important to understanding endocrine disruption. Overall, the 2012 report does not provide a balanced perspective, nor does it accurately reflect the state of the science on endocrine disruption.


Toxicological Sciences | 2013

An F1-extended one-generation reproductive toxicity study in Crl:CD(SD) rats with 2,4-dichlorophenoxyacetic acid.

Mary Sue Marty; Barbara H. Neal; Carol L. Zablotny; Barry L. Yano; Amanda K. Andrus; Michael R. Woolhiser; Darrell R. Boverhof; Shakil A. Saghir; Adam W. Perala; Julie K. Passage; Marie Lawson; James S. Bus; James C. Lamb; Larry Hammond

2,4-Dichlorophenoxyacetic acid (2,4-D) was assessed for systemic toxicity, reproductive toxicity, developmental neurotoxicity (DNT), developmental immunotoxicity (DIT), and endocrine toxicity. CD rats (27/sex/dose) were exposed to 0, 100, 300, 600 (female), or 800 (male) ppm 2,4-D in diet. Nonlinear toxicokinetic behavior was shown at high doses; the renal clearance saturation threshold for 2,4-D was exceeded markedly in females and slightly exceeded in males. Exposure was 4 weeks premating, 7 weeks postmating for P1 males and through lactation for P1 females. F1 offspring were examined for survival and development, and at weaning, pups were divided in cohorts, by sex and dose, and by systemic toxicity (10), DNT (10), DIT (20), and reproductive toxicity (≥ 23). Remaining weanlings were evaluated for systemic toxicity and neuropathology (10–12). Body weight decreased during lactation in high-dose P1 females and in F1 pups. Kidney was the primary target organ, with slight degeneration of proximal convoluted tubules observed in high-dose P1 males and in high-dose F1 males and females. A slight intergenerational difference in kidney toxicity was attributed to increased intake of 2,4-D in F1 offspring. Decreased weanling testes weights and delayed preputial separation in F1 males were attributed to decreased body weights. Endocrine-related effects were limited to slight thyroid hormone changes and adaptive histopathology in high-dose GD 17 dams seen only at a nonlinear toxicokinetic dose. 2,4-D did not cause reproductive toxicity, DNT, or DIT. The “No Observed Adverse Effect Level” for systemic toxicity was 300 ppm in both males (16.6mg/kg/day) and females (20.6mg/kg/day), which is approximately 6700- to 93 000-fold higher than that reported for 2,4-D exposures in human biomonitoring studies.


Regulatory Toxicology and Pharmacology | 2015

Comments on the opinions published by Bergman et al. (2015) on Critical Comments on the WHO-UNEP State of the Science of Endocrine Disrupting Chemicals (Lamb et al., 2014)

James C. Lamb; Paolo Boffetta; Warren G. Foster; Julie E. Goodman; Karyn L. Hentz; Lorenz R. Rhomberg; Jane Staveley; Gerard M. H. Swaen; Glen Van Der Kraak; Amy Lavin Williams

Recently Bergman et al. (2015) took issue with our comments (Lamb et al., 2014) on the WHO-UNEP(1) report entitled the State of the Science of Endocrine Disrupting Chemicals - 2012 (WHO 2013a). We find several key differences between their view and ours regarding the selection of studies and presentation of data related to endocrine disrupting chemicals (EDCs) under the WHO-IPCS(2) definition (2002). In this response we address the factors that we think are most important: 1. the difference between hazard and risk; 2. the different approaches for hazard identification (weight of the evidence [WOE] vs. emphasizing positive findings over null results); and 3. the lack of a justification for conceptual or practical differences between EDCs and other groups of agents.


Regulatory Toxicology and Pharmacology | 2012

Acetylcholinesterase inhibition dose-response modeling for chlorpyrifos and chlorpyrifos-oxon.

Richard Reiss; Barbara H. Neal; James C. Lamb; Daland R. Juberg

This paper evaluates new data for cholinesterase inhibition with chlorpyrifos (CPF). Marty et al. (2012) recently conducted a CPF cholinesterase inhibition study in rats that included testing of males and females, dosing by gavage or diet, administration in corn oil or milk, and with pups and adults. Additionally, the study included cholinesterase inhibition testing for CPF-oxon, the active moiety that inhibits cholinesterase. The study included 5-6 dose groups with eight animals/sex/group for most of the tests. This paper provides a benchmark dose (BMD) analysis of the data from Marty et al. (2012), including a BMD meta-analysis that includes CPF cholinesterase inhibition data from different assays within the Marty et al. (2012) study and, in one case, from another study. From the meta-analysis, the recommended BMD(10)s, based on brain acetylcholinesterase inhibition, are 1.7 mg/kg/day (BMDL₁₀ = 1.3mg/kg/day) for acute doses to children and adults, and 0.67 mg/kg/day (BMDL₁₀ = 0.53 mg/kg/day) for repeat doses to children and adults. At the dose levels considered in this analysis, there was no evidence of a difference in responses between males and females, corn oil versus milk administration, or pups versus adults. The data on pups versus adults show that an extra safety factor to protect the young is not needed for CPF. CPF data from the literature suggest that brain cholinesterase inhibition is the most appropriate metric for cholinesterase inhibition risk assessment.


Toxicological Sciences | 2015

Mechanistic Investigation of Toxaphene Induced Mouse Liver Tumors

Zemin Wang; Barbara H. Neal; James C. Lamb; James E. Klaunig

Chronic exposure to toxaphene resulted in an increase in liver tumors in B6C3F1 mice. This study was performed to investigate the mode of action of toxaphene induced mouse liver tumors. Following an initial 14 day dietary dose range-finding study in male mice, a mechanistic study (0, 3, 32, and 320 ppm toxaphene in diet for 7, 14, and 28 days of treatment) was performed to examine the potential mechanisms of toxaphene induced mouse liver tumors. Toxaphene induced a significant increase in expression of constitutive androstane receptor (CAR) target genes (Cyp2b10, Cyp3a11) at 32 and 320 ppm toxaphene. aryl hydrocarbon receptor (AhR) target genes (Cyp1a1 and Cyp1a2) were slightly increased in expression at the highest toxaphene dose (320 ppm). No increase in peroxisome proliferator-activated receptor alpha activity or related genes was seen following toxaphene treatment. Lipid peroxidation was seen following treatment with 320 ppm toxaphene. These changes correlated with increases in hepatic DNA synthesis. To confirm the role of CAR in this mode of action, CAR knockout mice (CAR(-/-)) treated with toxaphene confirmed that the induction of CAR responsive genes seen in wild-type mice was abolished following treatment with toxaphene for 14 days. These findings, taken together with previously reported studies, support the mode of action of toxaphene induced mouse liver tumors is through a nongenotoxic mechanism involving primarily a CAR-mediated processes that results in an increase in cell proliferation in the liver, promotes the clonal expansion of preneoplastic lesions leading to adenoma formation.


Journal of Applied Toxicology | 2017

Toxaphene-induced mouse liver tumorigenesis is mediated by the constitutive androstane receptor: Nuclear receptor CAR-mediated toxaphene-induced mouse liver tumors

Zemin Wang; Xilin Li; Qiangen Wu; James C. Lamb; James E. Klaunig

Toxaphene was shown to increase liver tumor incidence in B6C3F1 mice following chronic dietary exposure. Preliminary evidence supported a role for the constitutive androstane receptor (CAR) in the mode of action of toxaphene‐induced mouse liver tumors. However, these results could not rule out a role for the pregnane X receptor (PXR) in liver tumor formation. To define further the nuclear receptors involved in this study, we utilized CAR, PXR and PXR/CAR knockout mice (CAR−/−, PXR−/− and PXR−/−/CAR−/−) along with the wild‐type C57BL/6. In this study CAR‐responsive genes Cyp3a11 and Cyp2b10 were induced in the liver of C57BL/6 (wild‐type) mice by toxaphene (30–570‐fold) (at the carcinogenic dose 320u2009ppm) and phenobarbital (positive control) (16–420‐fold) following 14u2009days dietary treatment. In contrast, in CAR−/− mice, no induction of these genes was seen following treatment with either chemical. Cyp3a11 and Cyp2b10 were also induced in PXR−/− mice with toxaphene and phenobarbital but were not changed in treated PXR−/−/CAR−/− mice. Similarly, induction of liver pentoxyresorufin‐O‐deethylase (CAR activation) activity by toxaphene and phenobarbital was absent in CAR−/− and PXR−/−/CAR−/− mice treated with phenobarbital or toxaphene. Ethoxyresorufin‐O‐deethylase (EROD, represents aryl hydrocarbon receptor activation) activity in CAR−/− mice treated with toxaphene or phenobarbital was increased compared with untreated control, but lower overall in activity in comparison to the wild‐type mouse. Liver EROD activity was also induced by both phenobarbital and toxaphene in the PXR−/− mice but not in the PXR−/−/CAR−/− mice. Toxaphene treatment increased 7‐benzyloxyquinoline activity (a marker for PXR activation) in a similar pattern to that seen with pentoxyresorufin‐O‐deethylase. These observations indicate that EROD and PXR activation are evidence, as expected, of secondary overlap to primary CAR receptor activation. Together, these results definitively show that activation of the CAR nuclear receptor is the mode of action of toxaphene‐induced mouse liver tumors. Copyright


Human and Ecological Risk Assessment | 2000

The federal government's Agricultural Health Study: a critical review with suggested improvements.

George M. Gray; Bernard D. Goldstein; John C. Bailar; Devra Lee Davis; Elizabeth Delzell; Frank N. Dost; Raymond S. Greenberg; Maureen Hatch; Ernest Hodgson; Michel A. Ibrahim; James C. Lamb; Terry L. Lavy; Jack S. Mandel; Richard R. Monson; Mark G. Robson; Roy E. Shore; John D. Graham

The Agricultural Health Study (AHS) has approximately 90,000 pesticide applicators and their spouses enrolled in a number of studies to determine whether exposures to specific pesticides are associated with various cancers and other adverse health outcomes. Although the AHS was intended to be an integrated program of studies, some significant difficulties have emerged. In this report, we examine the design of the AHS, identify important program strengths and flaws, suggest various improvements in the program, and recommend ancillary studies that could be undertaken to strengthen the AHS. Overall, the AHS is collecting a large amount of information on potential determinants of health status among farmers and farm families. A promising feature of the AHS is the prospective cohort study of cancers among farmers in which the research design determines exposures prior to the diagnosis of disease. More effort needs to be devoted to reducing selection bias and information bias. Success of the cohort study will depend in part on follow-up surveys of the cohort to determine how exposures and disease states change as the cohort ages. The cross-sectional and case-control studies planned in the AHS are less promising because they will be subject to some of the same criticisms, such as potentially biased and imprecise exposure assessment, that have characterized the existing literature in this field. Important limitations of the AHS include low and variable rates of subject response to administered surveys, concerns about the validity of some self-reported non-cancer health outcomes, limited understanding of the reliability and validity of self-reporting of chemical use, an insufficient program of biological monitoring to validate the exposure surrogates employed in the AHS questionnaires, possible confounding by unmeasured, nonchemical risk factors for disease, and the absence of detailed plans for data analysis and interpretation that include explicit, a priori hypotheses. Although the AHS is already well underway, most of these limitations can be addressed by the investigators if adequate resources are made available. If these limitations are not addressed, the large amounts of data generated in the AHS will be difficult to interpret. If the exposure and health data can be validated, the scientific value of the AHS should be substantial and enduring. A variety of research recommendations are made to strengthen the AHS. They include reliability and validity studies of farmer reporting of chemical use, biological monitoring studies of farmers and members of farm families, and validity studies of positive and negative self-reports of disease status. Both industry and government should consider expanded research programs to strengthen the AHS.


Critical Reviews in Toxicology | 2017

Weight-of-the-evidence evaluation of 2,4-D potential for interactions with the estrogen, androgen and thyroid pathways and steroidogenesis

Barbara H. Neal; Mary Sue Marty; Katherine Coady; Amy Lavin Williams; Jane Staveley; James C. Lamb

Abstract A comprehensive weight-of-the-evidence evaluation of 2,4-dichlorophenoxyacetic acid (2,4-D) was conducted for potential interactions with the estrogen, androgen and thyroid pathways and with steroidogenesis. This assessment was based on an extensive database of high quality in vitro, in vivo ecotoxicological and in vivo mammalian toxicological studies. Epidemiological studies were also considered. Toxicokinetic data provided the basis for determining rational cutoffs above which exposures were considered irrelevant to humans based on exceeding thresholds for saturation of renal clearance (TSRC); extensive human exposure and biomonitoring data support that these boundaries far exceed human exposures and provide ample margins of exposure. 2,4-D showed no evidence of interacting with the estrogen or androgen pathways. 2,4-D interacts with the thyroid axis in rats through displacement of thyroxine from plasma binding sites only at high doses exceeding the TSRC in mammals. 2,4-D effects on steroidogenesis parameters are likely related to high-dose specific systemic toxicity at doses exceeding the TSRC and are not likely to be endocrine mediated. No studies, including high quality studies in the published literature, predict significant endocrine-related toxicity or functional decrements in any species at environmentally relevant concentrations, or, in mammals, at doses below the TSRC that are relevant for human hazard and risk assessment. Overall, there is no basis for concern regarding potential interactions of 2,4-D with endocrine pathways or axes (estrogen, androgen, steroidogenesis or thyroid), and thus 2,4-D is unlikely to pose a threat from endocrine disruption to wildlife or humans under conditions of real-world exposures.


Critical Reviews in Toxicology | 2009

Weight-of-the-evidence review of acrylonitrile reproductive and developmental toxicity studies

Barbara H. Neal; James J. Collins; Dale E. Strother; James C. Lamb

Risk assessment of acrylonitrile (AN) toxicity to humans has focused on potential carcinogenicity and acute toxicity. Epidemiological studies from China reported reproductive and developmental effects in AN workers, including infertility, birth defects, and spontaneous abortions. A weight-of-the-evidence (WoE) evaluation of the AN database assessed study strength, characterized toxicity, and identified no-observed-adverse-effect levels (NOAELs). The epidemiological studies do not demonstrate causality and are not sufficiently robust to be used for risk assessment. Rodent developmental studies showed fetotoxicity and malformations at maternally toxic levels; there was no unique developmental susceptibility. NOAELs for oral and inhalation exposures were 10u2009mg/kg/day and 12u2009ppm (6u2009h/day), respectively. Drinking-water and inhalation reproductive toxicity studies showed no clear effects on reproductive performance or fertility. Maternally toxic concentrations caused decreased pup growth. The drinking-water reproductive NOAEL was 100u2009ppm (moderate confidence due to study limitations). The inhalation exposure reproductive and neonatal toxicity high confidence NOAEL was 45u2009ppm (first generation 90u2009ppm) (6u2009h/day). The inhalation reproductive toxicity study provides the most robust data for risk assessment. Based on the WoE evaluation, AN is not expected to be a developmental or reproductive toxicant in the absence of significant maternal toxicity.

Collaboration


Dive into the James C. Lamb's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James E. Klaunig

Indiana University Bloomington

View shared research outputs
Top Co-Authors

Avatar

Julie E. Goodman

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge