Network


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

Hotspot


Dive into the research topics where Micah T. McClain is active.

Publication


Featured researches published by Micah T. McClain.


Nature Medicine | 2005

Early events in lupus humoral autoimmunity suggest initiation through molecular mimicry.

Micah T. McClain; Latisha Heinlen; Gregory J. Dennis; Jon Roebuck; John B. Harley; Judith A. James

The origins of autoimmunity in systemic lupus erythematosus (SLE) are thought to involve both genetic and environmental factors. To identify environmental agents that could potentially incite autoimmunity, we have traced the autoantibody response in human SLE back in time, prior to clinical disease onset, and identified the initial autoantigenic epitope for some lupus patients positive for antibodies to 60 kDa Ro. This initial epitope directly cross-reacts with a peptide from the latent viral protein Epstein-Barr virus nuclear antigen-1 (EBNA-1). Animals immunized with either the first epitope of 60 kDa Ro or the cross-reactive EBNA-1 epitope progressively develop autoantibodies binding multiple epitopes of Ro and spliceosomal autoantigens. They eventually acquire clinical symptoms of lupus such as leukopenia, thrombocytopenia and renal dysfunction. These data support the hypothesis that some humoral autoimmunity in human lupus arises through molecular mimicry between EBNA-1 and lupus autoantigens and provide further evidence to suspect an etiologic role for Epstein-Barr virus in SLE.


PLOS Genetics | 2011

Temporal Dynamics of Host Molecular Responses Differentiate Symptomatic and Asymptomatic Influenza A Infection

Yongsheng Huang; Aimee K. Zaas; Arvind Rao; Nicolas Dobigeon; Peter J. Woolf; Timothy Veldman; N. Christine Øien; Micah T. McClain; Jay B. Varkey; Bradley Nicholson; Lawrence Carin; Stephen F. Kingsmore; Christopher W. Woods; Geoffrey S. Ginsburg; Alfred O. Hero

Exposure to influenza viruses is necessary, but not sufficient, for healthy human hosts to develop symptomatic illness. The host response is an important determinant of disease progression. In order to delineate host molecular responses that differentiate symptomatic and asymptomatic Influenza A infection, we inoculated 17 healthy adults with live influenza (H3N2/Wisconsin) and examined changes in host peripheral blood gene expression at 16 timepoints over 132 hours. Here we present distinct transcriptional dynamics of host responses unique to asymptomatic and symptomatic infections. We show that symptomatic hosts invoke, simultaneously, multiple pattern recognition receptors-mediated antiviral and inflammatory responses that may relate to virus-induced oxidative stress. In contrast, asymptomatic subjects tightly regulate these responses and exhibit elevated expression of genes that function in antioxidant responses and cell-mediated responses. We reveal an ab initio molecular signature that strongly correlates to symptomatic clinical disease and biomarkers whose expression patterns best discriminate early from late phases of infection. Our results establish a temporal pattern of host molecular responses that differentiates symptomatic from asymptomatic infections and reveals an asymptomatic host-unique non-passive response signature, suggesting novel putative molecular targets for both prognostic assessment and ameliorative therapeutic intervention in seasonal and pandemic influenza.


PLOS ONE | 2011

H3N2 influenza infection elicits more cross-reactive and less clonally expanded anti-hemagglutinin antibodies than influenza vaccination.

M. Anthony Moody; Ruijun Zhang; Emmanuel B. Walter; Christopher W. Woods; Geoffrey S. Ginsburg; Micah T. McClain; Thomas N. Denny; Xi Chen; Supriya Munshaw; Dawn J. Marshall; John F. Whitesides; Mark Drinker; Joshua D. Amos; Thaddeus C. Gurley; Joshua Eudailey; Andrew Foulger; Katherine R. DeRosa; Robert Parks; R. Ryan Meyerhoff; Jae-Sung Yu; Daniel M. Kozink; Brice E. Barefoot; Elizabeth Ramsburg; Surender Khurana; Hana Golding; Nathan Vandergrift; S. Munir Alam; Georgia D. Tomaras; Thomas B. Kepler; Garnett Kelsoe

Background During the recent H1N1 influenza pandemic, excess morbidity and mortality was seen in young but not older adults suggesting that prior infection with influenza strains may have protected older subjects. In contrast, a history of recent seasonal trivalent vaccine in younger adults was not associated with protection. Methods and Findings To study hemagglutinin (HA) antibody responses in influenza immunization and infection, we have studied the day 7 plasma cell repertoires of subjects immunized with seasonal trivalent inactivated influenza vaccine (TIV) and compared them to the plasma cell repertoires of subjects experimentally infected (EI) with influenza H3N2 A/Wisconsin/67/2005. The majority of circulating plasma cells after TIV produced influenza-specific antibodies, while most plasma cells after EI produced antibodies that did not react with influenza HA. While anti-HA antibodies from TIV subjects were primarily reactive with single or few HA strains, anti-HA antibodies from EI subjects were isolated that reacted with multiple HA strains. Plasma cell-derived anti-HA antibodies from TIV subjects showed more evidence of clonal expansion compared with antibodies from EI subjects. From an H3N2-infected subject, we isolated a 4-member clonal lineage of broadly cross-reactive antibodies that bound to multiple HA subtypes and neutralized both H1N1 and H3N2 viruses. This broad reactivity was not detected in post-infection plasma suggesting this broadly reactive clonal lineage was not immunodominant in this subject. Conclusion The presence of broadly reactive subdominant antibody responses in some EI subjects suggests that improved vaccine designs that make broadly reactive antibody responses immunodominant could protect against novel influenza strains.


Lupus | 2007

Hydroxychloroquine sulfate treatment is associated with later onset of systemic lupus erythematosus.

Judith A. James; Xr Kim-Howard; Benjamin F. Bruner; Maria Karolina Jonsson; Micah T. McClain; Arbuckle; C. Walker; Gregory J. Dennis; Joan T. Merrill; John B. Harley

Systemic lupus erythematosus (SLE) is a clinically diverse, complex autoimmune disease which may present with coincident onset of many criteria or slow, gradual symptom accrual. Early intervention has been postulated to delay or prevent the development of more serious sequelae. One option for treatment in this setting is hydroxychloroquine. Using 130 US military personnel who later met ACR SLE criteria, a retrospective study of onset, development and progression of SLE with and without pre-classification hydroxychloroquine (n = 26) use was performed. Patients treated with hydroxychloroquine prior to diagnosis had a longer (Wilcoxon signed rank test, P = 0.018) time between the onset of the first clinical symptom and SLE classification (median: 1.08 versus 0.29 years). Patients treated with prednisone before diagnosis also more slowly satisfied the classification criteria (Wilcoxon signed rank test, P = 0.011). The difference in median times between patients who received NSAIDs before diagnosis, as opposed to those who did not, was not different (P = 0.19). Patients treated with hydroxychloroquine also had a lower rate of autoantibody accumulation and a decreased number of autoantibody specificities at and after diagnosis. These findings are consistent with early hydroxychloroquine use being associated with delayed SLE onset. A prospective, blinded trial testing the capacity of hydroxychloroquine to delay or prevent SLE in high risk populations is warranted. Lupus (2007) 16, 401—409.


PLOS ONE | 2013

A host transcriptional signature for presymptomatic detection of infection in humans exposed to influenza H1N1 or H3N2.

Christopher W. Woods; Micah T. McClain; Minhua Chen; Aimee K. Zaas; Bradly P. Nicholson; Jay B. Varkey; Timothy Veldman; Stephen F. Kingsmore; Yongsheng Huang; Robert Lambkin-Williams; Anthony G. Gilbert; Alfred O. Hero; Elizabeth Ramsburg; Seth W. Glickman; Joseph E. Lucas; Lawrence Carin; Geoffrey S. Ginsburg

There is great potential for host-based gene expression analysis to impact the early diagnosis of infectious diseases. In particular, the influenza pandemic of 2009 highlighted the challenges and limitations of traditional pathogen-based testing for suspected upper respiratory viral infection. We inoculated human volunteers with either influenza A (A/Brisbane/59/2007 (H1N1) or A/Wisconsin/67/2005 (H3N2)), and assayed the peripheral blood transcriptome every 8 hours for 7 days. Of 41 inoculated volunteers, 18 (44%) developed symptomatic infection. Using unbiased sparse latent factor regression analysis, we generated a gene signature (or factor) for symptomatic influenza capable of detecting 94% of infected cases. This gene signature is detectable as early as 29 hours post-exposure and achieves maximal accuracy on average 43 hours (p = 0.003, H1N1) and 38 hours (p-value = 0.005, H3N2) before peak clinical symptoms. In order to test the relevance of these findings in naturally acquired disease, a composite influenza A signature built from these challenge studies was applied to Emergency Department patients where it discriminates between swine-origin influenza A/H1N1 (2009) infected and non-infected individuals with 92% accuracy. The host genomic response to Influenza infection is robust and may provide the means for detection before typical clinical symptoms are apparent.


Science Translational Medicine | 2013

A Host-Based RT-PCR Gene Expression Signature to Identify Acute Respiratory Viral Infection

Aimee K. Zaas; Thomas Burke; Minhua Chen; Micah T. McClain; Bradly P. Nicholson; Timothy Veldman; Ephraim L. Tsalik; Vance G. Fowler; Emanuel P. Rivers; Ronny M. Otero; Stephen F. Kingsmore; Deepak Voora; Joseph Lucas; Alfred O. Hero; Lawrence Carin; Christopher W. Woods; Geoffrey S. Ginsburg

To improve the diagnosis of respiratory viral infection, a multiplex RT-PCR assay based on the host response was derived from experimentally infected subjects and validated in patients with febrile illness. Diagnosing the Cause of Coughs and Sneezes Diagnosis of viral respiratory infections remains a challenge. Early differentiation between a viral and bacterial etiology of respiratory symptoms would help to direct therapy more appropriately and prevent overuse of antibiotics. Measuring the host immune response to infection is an alternative to pathogen-based diagnostic testing and may improve diagnostic accuracy. Now, Zaas et al. have developed a reverse transcription polymerase chain reaction (RT-PCR) assay for blood RNA that can classify respiratory viral infections based on the host immune response. They developed their assay using two groups of individuals experimentally infected with either influenza A H3N2/Wisconsin or influenza A H1N1/Brisbane. They then validated their RT-PCR diagnostic in a sample of adults presenting to the emergency department with fever, who had microbiologically confirmed viral or bacterial illness. The sensitivity of the RT-PCR assay was 89% [95% confidence interval (CI), 72 to 98%], and the specificity was 94% (95% CI, 86 to 99%). These data establish an important “proof of concept” that host expression of a relatively small set of genes measured by RT-PCR can be used to classify viral respiratory illness in unselected individuals presenting at an emergency department for evaluation of fever. The development of this new assay and its validation in an independent “real-world” patient population is an important step on the translational pathway to establishing this platform for diagnostic testing in the clinic. Improved ways to diagnose acute respiratory viral infections could decrease inappropriate antibacterial use and serve as a vital triage mechanism in the event of a potential viral pandemic. Measurement of the host response to infection is an alternative to pathogen-based diagnostic testing and may improve diagnostic accuracy. We have developed a host-based assay with a reverse transcription polymerase chain reaction (RT-PCR) TaqMan low-density array (TLDA) platform for classifying respiratory viral infection. We developed the assay using two cohorts experimentally infected with influenza A H3N2/Wisconsin or influenza A H1N1/Brisbane, and validated the assay in a sample of adults presenting to the emergency department with fever (n = 102) and in healthy volunteers (n = 41). Peripheral blood RNA samples were obtained from individuals who underwent experimental viral challenge or who presented to the emergency department and had microbiologically proven viral respiratory infection or systemic bacterial infection. The selected gene set on the RT-PCR TLDA assay classified participants with experimentally induced influenza H3N2 and H1N1 infection with 100 and 87% accuracy, respectively. We validated this host gene expression signature in a cohort of 102 individuals arriving at the emergency department. The sensitivity of the RT-PCR test was 89% [95% confidence interval (CI), 72 to 98%], and the specificity was 94% (95% CI, 86 to 99%). These results show that RT-PCR–based detection of a host gene expression signature can classify individuals with respiratory viral infection and sets the stage for prospective evaluation of this diagnostic approach in a clinical setting.


Science Translational Medicine | 2016

Host gene expression classifiers diagnose acute respiratory illness etiology.

Ephraim L. Tsalik; Ricardo Henao; Marshall Nichols; Thomas Burke; Emily R. Ko; Micah T. McClain; Lori L. Hudson; Anna Mazur; D. Freeman; Tim Veldman; Raymond J. Langley; Eugenia Quackenbush; Seth W. Glickman; Charles B. Cairns; Anja Kathrin Jaehne; Emanuel P. Rivers; Ronny M. Otero; Aimee K. Zaas; Stephen F. Kingsmore; Joseph Lucas; Vance G. Fowler; Lawrence Carin; Geoffrey S. Ginsburg; Christopher W. Woods

Pathogen-specific host gene expression changes may combat inappropriate antibiotic use and emerging antibiotic resistance. Resisting antibiotics No matter the cause, acute respiratory infections can be miserable. Indeed, these infections are one of the most common reasons for seeking medical care. A clear diagnostic can help medical practitioners resist the patient-induced pressure to prescribe antibiotics as a catch-all therapy, which increases the risk of bacteria developing antibiotic resistance. Now, Tsalik et al. report clear differences in host gene expression induced by bacterial and viral infection as well as by noninfectious illness. These differences can be used to discriminate between these groups, and a host gene expression classifier may be a helpful diagnostic platform to curb unnecessary antibiotic use. Acute respiratory infections caused by bacterial or viral pathogens are among the most common reasons for seeking medical care. Despite improvements in pathogen-based diagnostics, most patients receive inappropriate antibiotics. Host response biomarkers offer an alternative diagnostic approach to direct antimicrobial use. This observational cohort study determined whether host gene expression patterns discriminate noninfectious from infectious illness and bacterial from viral causes of acute respiratory infection in the acute care setting. Peripheral whole blood gene expression from 273 subjects with community-onset acute respiratory infection (ARI) or noninfectious illness, as well as 44 healthy controls, was measured using microarrays. Sparse logistic regression was used to develop classifiers for bacterial ARI (71 probes), viral ARI (33 probes), or a noninfectious cause of illness (26 probes). Overall accuracy was 87% (238 of 273 concordant with clinical adjudication), which was more accurate than procalcitonin (78%, P < 0.03) and three published classifiers of bacterial versus viral infection (78 to 83%). The classifiers developed here externally validated in five publicly available data sets (AUC, 0.90 to 0.99). A sixth publicly available data set included 25 patients with co-identification of bacterial and viral pathogens. Applying the ARI classifiers defined four distinct groups: a host response to bacterial ARI, viral ARI, coinfection, and neither a bacterial nor a viral response. These findings create an opportunity to develop and use host gene expression classifiers as diagnostic platforms to combat inappropriate antibiotic use and emerging antibiotic resistance.


Lupus | 2003

Rapid clinical progression to diagnosis among African-American men with systemic lupus erythematosus.

Melissa R. Arbuckle; Judith A. James; Gregory J. Dennis; Mark V. Rubertone; Micah T. McClain; X R Kim; John B. Harley

The initial clinical course of systemic lupus erythematosus (SLE) is variable, ranging from relatively minor manifestations progressing over years to rapid onset of fulminate disease. We sought to identify factors associated with the rapid manifestation of SLE. Chart review of military medical records was used to identify 130 patients who met the American College of Rheumatology classification criteria for SLE. Demographics, clinical criteria date of occurrence, and the date of SLE classification (at least four clinical criteria) met were documented. Prospectively stored serum samples prior to the diagnosis were evaluated for SLE autoantibodies. Median time from the first recorded criteria to diagnosis was significantly shorter in African-American (AA) males compared with AA females and European American (EA) females and males combined. AA males were more likely to have nephritis as their first clinical symptom. Also, less time transpired between the first clinical criterion and SLE diagnosis in AA males with nephritis than in other groups presenting with nephritis. Even when cases presenting with nephritis were excluded, a diagnosis of SLE was made more rapidly in AA males. African-American men progress from initial clinical manifestations to SLE diagnosis more rapidly than other ethnic or gender groups.


Journal of Immunology | 2002

Anti-Sm Autoantibodies in Systemic Lupus Target Highly Basic Surface Structures of Complexed Spliceosomal Autoantigens

Micah T. McClain; Paul A. Ramsland; Kenneth M. Kaufman; Judith A. James

Autoantibodies directed against spliceosomal proteins are a common and specific feature of systemic lupus erythematosus. These autoantibodies target a collection of proteins, including Sm B, B′, D1, D2, and D3. We define the common antigenic targets of Sm D2 and D3 and examine their role in spliceosomal autoimmunity. Our results define nine major common epitopes, five on Sm D2 and four on Sm D3. These epitopes have significantly higher (more basic) isoelectric points than do nonantigenic regions. In fact, this association is of sufficient power to make isoelectric point an excellent predictor of spliceosomal antigenicity. The crystallographic structure of Sm D2 and D3 is now partially described. The anti-Sm D2 and D3 antigenic targets are located on the surface of the respective three-dimensional complexed proteins, thereby suggesting that these epitopes are accessible in the native configuration. All but one of these nine epitopes conspicuously avoid the specific regions involved in intermolecular interactions within the spliceosomal complex. One of the D3 epitopes (RGRGRGMGR) has significant sequence homology with a major antigenic region of Sm D1 (containing a carboxyl-terminal glycine-arginine repeat), and anti-D3 Abs cross-react with this epitope of Sm D1. These results demonstrate that spliceosomal targets of autoimmunity are accessible on native structure surfaces and that cross-reactive epitopes, as well as structural associations of various spliceosomal Ags, may be involved in the induction of autoimmunity in systemic lupus.


PLOS ONE | 2010

60 kD Ro and nRNP A Frequently Initiate Human Lupus Autoimmunity

Latisha Heinlen; Micah T. McClain; Lauren L. Ritterhouse; Benjamin F. Bruner; Colin C. Edgerton; Michael P. Keith; Judith A. James; John B. Harley

Systemic lupus erythematosus (SLE) is a clinically heterogeneous, humoral autoimmune disorder. The unifying feature among SLE patients is the production of large quantities of autoantibodies. Serum samples from 129 patients collected before the onset of SLE and while in the United States military were evaluated for early pre-clinical serologic events. The first available positive serum sample frequently already contained multiple autoantibody specificities (65%). However, in 34 SLE patients the earliest pre-clinical serum sample positive for any detectable common autoantibody bound only a single autoantigen, most commonly 60 kD Ro (29%), nRNP A (24%), anti-phospholipids (18%) or rheumatoid factor (15%). We identified several recurrent patterns of autoantibody onset using these pre-diagnostic samples. In the serum samples available, anti-nRNP A appeared before or simultaneously with anti-nRNP 70 K in 96% of the patients who had both autoantibodies at diagnosis. Anti-60 kD Ro antibodies appeared before or simultaneously with anti-La (98%) or anti-52 kD Ro (95%). The autoantibody response in SLE patients begins simply, often binding a single specific autoantigen years before disease onset, followed by epitope spreading to additional autoantigenic specificities that are accrued in recurring patterns.

Collaboration


Dive into the Micah T. McClain's collaboration.

Top Co-Authors

Avatar

Judith A. James

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John B. Harley

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Latisha Heinlen

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge