Bio


Dr. Kevin K. Kumar was born in New York City and grew up in Long Island, New York. He attended college at Cornell University where he majored in Biological Sciences with a concentration in Neurobiology and Behavior. After graduation, Dr. Kumar joined the Medical Scientist Training Program (MSTP) at Vanderbilt University to earn his combined MD/PhD. He completed his PhD in Neuroscience in the laboratory of Dr. Aaron Bowman, where he studied the regulation of manganese in both Parkinson’s and Huntington’s disease using high throughput screening, induced pluripotent stem cells, and untargeted metabolomics. He then began his neurosurgery residency at Stanford. As a resident, Dr. Kumar completed an NIH-funded postdoctoral fellowship in the laboratory of Dr. Marius Wernig in the Stanford Institute for Stem Cell Biology and Regenerative Medicine. His work focused on developing a platform to replace microglia in the brain as a novel therapy for neurodevelopmental disorders.

From 2023-2024, Dr. Kumar will serve as Clinical Instructor in Pediatric Neurosurgery at Lucile Packard Children’s Hospital at Stanford. In addition to his clinical duties, he will continue to advance cellular therapeutics for pediatric neurological diseases.

Clinical Focus


  • Pediatric Neurosurgery, Functional Neurosurgery, Stem Cell Biology, Neuroregeneration
  • Neurological Surgery

Academic Appointments


Professional Education


  • Residency: Stanford University Dept of Neurosurgery (2023) CA
  • Medical Education: Vanderbilt University School of Medicine (2016) TN
  • MD, Vanderbilt University, Medical Scientist Training Program (MSTP) (2016)
  • PhD, Vanderbilt University, Neuroscience (2014)
  • BS, Cornell University, Biological Sciences, Neurobiology and Behavior (2009)

All Publications


  • Treatment of Trigeminal Neuralgia Secondary to Petroclival Meningioma Using Microsurgical Resection, Microvascular Decompression, and Stereotactic Radiosurgery: 2-Dimensional Operative Video. Operative neurosurgery (Hagerstown, Md.) Park, D. J., Kumar, K. K., Marianayagam, N. J., Yener, U., Rahimy, E., Hancock, S., Meola, A., Chang, S. D. 2024; 26 (1): 107-108

    View details for DOI 10.1227/ons.0000000000000927

    View details for PubMedID 38099694

  • Vascular anomaly, lipoma, and polymicrogyria associated with schizencephaly: developmental and diagnostic insights. Illustrative case. Journal of neurosurgery. Case lessons Kumar, K. K., Toland, A., Fischbein, N., Morrell, M., Heit, J. J., Born, D. E., Steinberg, G. K. 2023; 5 (21)

    Abstract

    BACKGROUND: Schizencephaly is an uncommon central nervous system malformation. Intracranial lipomas are also rare, accounting for approximately 0.1% of brain "tumors." They are believed to be derived from a persistent meninx primitiva, a neural crest-derived mesenchyme that develops into the dura and leptomeninges.OBSERVATIONS: The authors present a case of heterotopic adipose tissue and a nonshunting arterial vascular malformation arising within a schizencephalic cleft in a 22-year-old male. Imaging showed right frontal gray matter abnormality and an associated suspected arteriovenous malformation with evidence of hemorrhage. Brain magnetic resonance imaging revealed right frontal polymicrogyria lining an open-lip schizencephaly, periventricular heterotopic gray matter, fat within the schizencephalic cleft, and gradient echo hypointensity concerning for prior hemorrhage. Histological assessment demonstrated mature adipose tissue with large-bore, thick-walled, irregular arteries. Mural calcifications and subendothelial cushions suggesting nonlaminar blood flow were observed. There were no arterialized veins or direct transitions from the arteries to veins. Hemosiderin deposition was scant, and hemorrhage was not present. The final diagnosis was consistent with ectopic mature adipose tissue and arteries with meningocerebral cicatrix.LESSONS: This example of a complex maldevelopment of derivatives of the meninx primitiva in association with cortical maldevelopment highlights the unique challenges from both a radiological and histological perspective during diagnostic workup.

    View details for DOI 10.3171/CASE2388

    View details for PubMedID 37218736

  • Collateral status, hyperglycemia, and functional outcome after acute ischemic stroke. BMC neurology Arteaga, D. F., Ulep, R., Kumar, K. K., Southerland, A. M., Conaway, M. R., Faber, J., Wintermark, M., Joyner, D., Sharashidze, V., Hirsch, K., Giurgiutiu, D., Hannawi, Y., Aziz, Y., Shutter, L., Visweswaran, A., Williams, A., Williams, K., Gunter, S., Haughey, H. M., Bruno, A., Johnston, K. C., Patel, V. N., SHINE Trial Investigators 2022; 22 (1): 408

    Abstract

    BACKGROUND: Mixed data exist regarding the association between hyperglycemia and functional outcome after acute ischemic stroke when accounting for the impact of leptomeningeal collateral flow. We sought to determine whether collateral status modifies the association between treatment group and functional outcome in a subset of patients with large vessel occlusion enrolled in the Stroke Hyperglycemia Insulin Network Effort (SHINE) trial.METHODS: In this post-hoc analysis, we analyzed patients enrolled into the SHINE trial with anterior circulation large vessel occlusion who underwent imaging with CT angiography prior to glucose control treatment group assignment. The primary analysis assessed the degree to which collateral status modified the effect between treatment group and functional outcome as defined by the 90-day modified Rankin Scale score. Logistic regression was used to model the data, with adjustments made for thrombectomy status, age, post-perfusion thrombolysis in cerebral infarction (TICI) score, tissue plasminogen activator (tPA) use, and baseline National Institutes of Health Stroke Scale (NIHSS) score. Five SHINE trial centers contributed data for this analysis. Statistical significance was defined as a p-value <0.05.RESULTS: Among the 1151 patients in the SHINE trial, 57 with angiographic data were included in this sub-analysis, of whom 19 had poor collaterals and 38 had good collaterals. While collateral status had no effect (p=0.855) on the association between glucose control treatment group and functional outcome, patients with good collaterals were more likely to have a favorable functional outcome (p=0.001, OR 5.02; 95% CI 1.37-16.0).CONCLUSIONS: In a post-hoc analysis using a subset of patients with angiographic data enrolled in the SHINE trial, collateral status did not modify the association between glucose control treatment group and functional outcome. However, consistent with prior studies, there was a significant association between good collateral status and favorable outcome in patients with large vessel occlusion stroke.TRIAL REGISTRATION: ClinicalTrials.gov Identifier is NCT01369069. Registration date is June 8, 2011.

    View details for DOI 10.1186/s12883-022-02943-4

    View details for PubMedID 36333676

  • Balamuthia mandrillaris brain infection: a rare cause of a ring-enhancing central nervous system lesion. Illustrative case. Journal of neurosurgery. Case lessons Levinson, S., Kumar, K. K., Wang, H., Tayyar, R., Dunning, M., Toland, A., Budvytiene, I., Vogel, H., Chang, A., Banaei, N., Shuer, L. 2022; 3 (15)

    Abstract

    BACKGROUND: An 80-year-old man presented with subacute mental status change, dizziness, and left-sided vision loss. Magnetic resonance imaging demonstrated a ring-enhancing right parietooccipital lesion.OBSERVATIONS: Biopsy and laboratory testing demonstrated an amoebic Balamuthia mandrillaris infection. Fewer than 200 cases of this infection have been recognized in the United States, and no standardized treatment regimen currently exists.LESSONS: Rapid antimicrobial therapy with miltefosine, azithromycin, fluconazole, flucytosine, sulfadiazine, and albendazole was initiated. The pathophysiology, diagnosis, and management of this infection and the patient's course were reviewed. The importance of biopsy for pathologic and laboratory diagnosis and rapid treatment initiation with a multidisciplinary team was reinforced.

    View details for DOI 10.3171/CASE2268

    View details for PubMedID 36303497

  • Management of Severe Traumatic Brain Injury in Pediatric Patients. Frontiers in toxicology Lui, A., Kumar, K. K., Grant, G. A. 2022; 4: 910972

    Abstract

    The optimal management of severe traumatic brain injury (TBI) in the pediatric population has not been well studied. There are a limited number of research articles studying the management of TBI in children. Given the prevalence of severe TBI in the pediatric population, it is crucial to develop a reference TBI management plan for this vulnerable population. In this review, we seek to delineate the differences between severe TBI management in adults and children. Additionally, we also discuss the known molecular pathogenesis of TBI. A better understanding of the pathophysiology of TBI will inform clinical management and development of therapeutics. Finally, we propose a clinical algorithm for the management and treatment of severe TBI in children using published data.

    View details for DOI 10.3389/ftox.2022.910972

    View details for PubMedID 35812167

  • Treatment of a genetic brain disease by CNS-wide microglia replacement. Science translational medicine Shibuya, Y., Kumar, K. K., Mader, M. M., Yoo, Y., Ayala, L. A., Zhou, M., Mohr, M. A., Neumayer, G., Kumar, I., Yamamoto, R., Marcoux, P., Liou, B., Bennett, F. C., Nakauchi, H., Sun, Y., Chen, X., Heppner, F. L., Wyss-Coray, T., Südhof, T. C., Wernig, M. 2022; 14 (636): eabl9945

    Abstract

    Hematopoietic cell transplantation after myeloablative conditioning has been used to treat various genetic metabolic syndromes but is largely ineffective in diseases affecting the brain presumably due to poor and variable myeloid cell incorporation into the central nervous system. Here, we developed and characterized a near-complete and homogeneous replacement of microglia with bone marrow cells in mice without the need for genetic manipulation of donor or host. The high chimerism resulted from a competitive advantage of scarce donor cells during microglia repopulation rather than enhanced recruitment from the periphery. Hematopoietic stem cells, but not immediate myeloid or monocyte progenitor cells, contained full microglia replacement potency equivalent to whole bone marrow. To explore its therapeutic potential, we applied microglia replacement to a mouse model for Prosaposin deficiency, which is characterized by a progressive neurodegeneration phenotype. We found a reduction of cerebellar neurodegeneration and gliosis in treated brains, improvement of motor and balance impairment, and life span extension even with treatment started in young adulthood. This proof-of-concept study suggests that efficient microglia replacement may have therapeutic efficacy for a variety of neurological diseases.

    View details for DOI 10.1126/scitranslmed.abl9945

    View details for PubMedID 35294256

  • Commentary: Genome-Wide Association Study Identifies Genetic Risk Factors for Spastic Cerebral Palsy. Neurosurgery Kumar, K. K., Grant, G. A. 2021

    View details for DOI 10.1093/neuros/nyab201

    View details for PubMedID 34133737

  • Fluoroscopic versus CT-guided cortical bone trajectory pedicle screw fixation: Comparing trajectory related complications. Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia Kumar, K. K., Parikh, B., Jabarkheel, R., Dirlikov, B., Singh, H. 2021

    Abstract

    Cortical bone trajectory (CBT) pedicle screw fixation is an emerging technique for treatment of degenerative spine disease which requires either intraoperative fluoroscopy or intraoperative CT guidance (iCT). To date, there has been no direct comparison of these two navigation modalities; here we compare fluoroscopic versus iCT navigation for CBT pedicle screw fixation. We retrospectively reviewed all patients who underwent CBT screw fixation with either fluoroscopic or iCT guidance for lumbar degenerative disease by the senior author. Trajectory-related complications such as medial or lateral breach were compared on postoperative CT, in addition to the incidence of trajectory-related dural tear. We also compared general surgical complications such as postoperative infection and decompression related durotomies. Thirty-eight patients (19 fluoroscopic, 19 CT-guided) who underwent placement of 182 cortical screws (88 fluoroscopic, 94 CT-guided) were identified. In terms of trajectory-related complications, the iCT cohort had fewer medial breaches (1/94) compared to the fluoroscopic cohort (6/88) (p=0.05). Each group had one lateral breach (p=0.73). There was one case of CSF leak from screw placement in the fluoroscopic cohort, but none in the iCT cohort (p=0.48). Overall, there were eight trajectory-related complications in the fluoroscopic cohort versus two in the iCT cohort (p=0.04). Our data suggests statistically significant decreased trajectory-related complications with iCT-guided CBT screw fixation as compared to fluoroscopically guided. In terms of general surgical complications, while we observed increased postoperative infections in our fluoroscopic cohort, there was no statistically significant difference.

    View details for DOI 10.1016/j.jocn.2021.05.048

    View details for PubMedID 34088578

  • Spontaneous regression of a vein of Galen aneurysmal malformation in a pediatric patient: illustrative case. Journal of neurosurgery. Case lessons Kumar, K. K., Fornoff, L. E., Dodd, R. L., Marks, M. P., Hong, D. S. 2021; 1 (8): CASE20171

    Abstract

    Vein of Galen aneurysmal malformations (VGAMs) are rare congenital intracranial vascular lesions that represent 30% of all pediatric vascular anomalies. These lesions are associated with severe manifestations, including congestive heart failure, hydrocephalus, and spontaneous hemorrhage. The mainstay of management is medical stabilization followed by endovascular embolization of the lesion. Although VGAM was first reported in 1937, there are few published cases demonstrating spontaneous regression of the lesion.The authors report the case of a 31-month-old female who presented with an incidentally found VGAM. After initial evaluation, including magnetic resonance imaging and angiography, the patient was lost to follow-up. Upon her return to the clinic at age 12 years, the previously identified VGAM was absent, indicative of involution of the lesion. The patient remained asymptomatic and met appropriate developmental milestones during this interval.This report adds a rare case of the spontaneous resolution of VGAM to the literature. This case may suggest the presence of VGAMs that are asymptomatic, undetected, and regress within the pediatric population. Future studies may benefit from identifying imaging and angiographic findings predictive of spontaneous regression. There may be a role for conservative management in particular cases of asymptomatic and medically stable children with VGAMs.

    View details for DOI 10.3171/CASE20171

    View details for PubMedID 35855311

    View details for PubMedCentralID PMC9241347

  • Commentary: Converting Pediatric Patients and Young Adults From a Shunt to a Third Ventriculostomy: A Multicenter Evaluation NEUROSURGERY Kumar, K. K., Grant, G. 2020; 87 (2): E105
  • Contemporaneous evaluation of patient experience, surgical strategy, and seizure outcomes in patients undergoing stereoelectroencephalography or subdural electrode monitoring. Epilepsia Kim, L. H., Parker, J. J., Ho, A. L., Feng, A. Y., Kumar, K. K., Chen, K. S., Ojukwu, D. I., Shuer, L. M., Grant, G. A., Graber, K. D., Halpern, C. H. 2020

    Abstract

    Intracranial electrographic localization of the seizure onset zone (SOZ) can guide surgical approaches for medically refractory epilepsy patients, especially when the presurgical workup is discordant or functional cortical mapping is required. Minimally invasive stereotactic placement of depth electrodes, stereoelectroencephalography (SEEG), has garnered increasing use, but limited data exist to evaluate its postoperative outcomes in the context of the contemporaneous availability of both SEEG and subdural electrode (SDE) monitoring. We aimed to assess the patient experience, surgical intervention, and seizure outcomes associated with these two epileptic focus mapping techniques during a period of rapid adoption of neuromodulatory and ablative epilepsy treatments.We retrospectively reviewed 66 consecutive adult intracranial electrode monitoring cases at our institution between 2014 and 2017. Monitoring was performed with either SEEG (n = 47) or SDEs (n = 19).Both groups had high rates of SOZ identification (SEEG 91.5%, SDE 88.2%, P = .69). The majority of patients achieved Engel class I (SEEG 29.3%, SDE 35.3%) or II outcomes (SEEG 31.7%, SDE 29.4%) after epilepsy surgery, with no significant difference between groups (P = .79). SEEG patients reported lower median pain scores (P = .03) and required less narcotic pain medication (median = 94.5 vs 594.6 milligram morphine equivalents, P = .0003). Both groups had low rates of symptomatic hemorrhage (SEEG 0%, SDE 5.3%, P = .11). On multivariate logistic regression, undergoing resection or ablation (vs responsive neurostimulation/vagus nerve stimulation) was the only significant independent predictor of a favorable outcome (adjusted odds ratio = 25.4, 95% confidence interval = 3.48-185.7, P = .001).Although both SEEG and SDE monitoring result in favorable seizure control, SEEG has the advantage of superior pain control, decreased narcotic usage, and lack of routine need for intensive care unit stay. Despite a heterogenous collection of epileptic semiologies, seizure outcome was associated with the therapeutic surgical modality and not the intracranial monitoring technique. The potential for an improved postoperative experience makes SEEG a promising method for intracranial electrode monitoring.

    View details for DOI 10.1111/epi.16762

    View details for PubMedID 33236777

  • Bilateral Deep Brain Stimulation is the Procedure to Beat for Advanced Parkinson Disease: A Meta-Analytic, Cost-Effective Threshold Analysis for Focused Ultrasound. Neurosurgery Mahajan, U. V., Ravikumar, V. K., Kumar, K. K., Ku, S. n., Ojukwu, D. I., Kilbane, C. n., Ghanouni, P. n., Rosenow, J. M., Stein, S. C., Halpern, C. H. 2020

    Abstract

    Parkinson disease (PD) impairs daily functioning for an increasing number of patients and has a growing national economic burden. Deep brain stimulation (DBS) may be the most broadly accepted procedural intervention for PD, but cost-effectiveness has not been established. Moreover, magnetic resonance image-guided focused ultrasound (FUS) is an emerging incisionless, ablative treatment that could potentially be safer and even more cost-effective.To (1) quantify the utility (functional disability metric) imparted by DBS and radiofrequency ablation (RF), (2) compare cost-effectiveness of DBS and RF, and (3) establish a preliminary success threshold at which FUS would be cost-effective compared to these procedures.We performed a meta-analysis of articles (1998-2018) of DBS and RF targeting the globus pallidus or subthalamic nucleus in PD patients and calculated utility using pooled Unified Parkinson Disease Rating Scale motor (UPDRS-3) scores and adverse events incidences. We calculated Medicare reimbursements for each treatment as a proxy for societal cost.Over a 22-mo mean follow-up period, bilateral DBS imparted the most utility (0.423 quality-adjusted life-years added) compared to (in order of best to worst) bilateral RF, unilateral DBS, and unilateral RF, and was the most cost-effective (expected cost: $32 095 ± $594) over a 22-mo mean follow-up. Based on this benchmark, FUS would need to impart UPDRS-3 reductions of ∼16% and ∼33% to be the most cost-effective treatment over 2- and 5-yr periods, respectively.Bilateral DBS imparts the most utility and cost-effectiveness for PD. If our established success threshold is met, FUS ablation could dominate bilateral DBS's cost-effectiveness from a societal cost perspective.

    View details for DOI 10.1093/neuros/nyaa485

    View details for PubMedID 33295629

  • Comparative effectiveness of neuroablation and deep brain stimulation for treatment-resistant obsessive-compulsive disorder: a meta-analytic study JOURNAL OF NEUROLOGY NEUROSURGERY AND PSYCHIATRY Kumar, K. K., Appelboom, G., Lamsam, L., Caplan, A. L., Williams, N. R., Bhati, M. T., Stein, S. C., Halpern, C. H. 2019; 90 (4): 469–73
  • MR-Guided Focused Ultrasound Versus Radiofrequency Capsulotomy for Treatment-Refractory Obsessive-Compulsive Disorder: A Cost-Effectiveness Threshold Analysis FRONTIERS IN NEUROSCIENCE Kumar, K. K., Bhati, M. T., Ravikumar, V. K., Ghanouni, P., Stein, S. C., Halpern, C. H. 2019; 13
  • Robot-assisted versus manual navigated stereoelectroencephalography in adult medically-refractory epilepsy patients. Epilepsy research Kim, L. H., Feng, A. Y., Ho, A. L., Parker, J. J., Kumar, K. K., Chen, K. S., Grant, G. A., Henderson, J. M., Halpern, C. H. 2019; 159: 106253

    Abstract

    Stereoelectroencephalography (SEEG) has experienced a recent growth in adoption for epileptogenic zone (EZ) localization. Advances in robotics have the potential to improve the efficiency and safety of this intracranial seizure monitoring method. We present our institutional experience employing robot-assisted SEEG and compare its operative efficiency, seizure reduction outcomes, and direct hospital costs with SEEG performed without robotic assistance using navigated stereotaxy.We retrospectively identified 50 consecutive adult SEEG cases at our institution in this IRB-approved study, of which 25 were navigated with image guidance (hereafter referred to as "navigated") (02/2014-10/2016) and 25 were robot-assisted (09/2016-12/2017). A thorough review of medical/surgical history and operative records with imaging and trajectory plans was done for each patient. Direct inpatient costs related to each technique were compared.Most common seizure etiologies for patients undergoing navigated and robot-assisted SEEG included non-lesional and benign temporal lesions. Despite having a higher mean number of leads-per-patient (10.2 ± 3.5 versus 7.2 ± 2.6, P = 0.002), robot-assisted cases had a significantly shorter mean operative time than navigated cases (125.5±48.5 versus 173.4±84.3 min, P = 0.02). Comparison of robot-assisted cases over the study interval revealed no significant difference in mean operative time (136.4±51.4 min for the first ten cases versus 109.9±75.8 min for the last ten cases, P = 0.25) and estimated operative time-per-lead (13.4±6.0 min for the first ten cases versus 12.9±7.7 min for the last ten cases, P = 0.86). The mean depth, radial, target, and entry point errors for robot-assisted cases were 2.12±1.89, 1.66±1.58, 3.05±2.02 mm, and 1.39 ± 0.75 mm, respectively. The two techniques resulted in equivalent EZ localization rate (navigated 88 %, robot-assisted 96 %, P = 0.30). Common types of epilepsy surgery performed consisted of implantation of responsive neurostimulation (RNS) device (56 %), resection (19.1 %), and laser ablation (23.8 %) for navigated SEEG. For robot-assisted SEEG, either RNS implantation (68.2 %) or laser ablation (22.7 %) were performed or offered. A majority of navigated and robot-assisted patients who underwent epilepsy surgery achieved either Engel Class I (navigated 36.8 %, robot-assisted 31.6 %) or II (navigated 36.8 %, robot-assisted 15.8 %) outcome with no significant difference between the groups (P = 0.14). Direct hospital cost for robot-assisted SEEG was 10 % higher than non-robotic cases.This single-institutional study suggests that robotic assistance can enhance efficiency of SEEG without compromising safety or precision when compared to image guidance only. Adoption of this technique with uniform safety and efficacy over a short period of time is feasible with favorable epilepsy outcomes.

    View details for DOI 10.1016/j.eplepsyres.2019.106253

    View details for PubMedID 31855826

  • Angiomyolipomas As A Cause of Abnormal Uterine Bleeding In A Patient with Tuberous Sclerosis Jacobs Journal of Gynecology and Obstetrics Harvey, L. F., Kumar, K. K., Desouki, M. M., Anderson, T. L. 2018; 5 (1): 035
  • Genomic Instability Associated with p53 Knockdown in the Generation of Huntington's Disease Human Induced Pluripotent Stem Cells PLOS ONE Tidball, A. M., Neely, M. D., Chamberlin, R., Aboud, A. A., Kumar, K. K., Han, B., Bryan, M. R., Aschner, M., Ess, K. C., Bowman, A. B. 2016; 11 (3)

    Abstract

    Alterations in DNA damage response and repair have been observed in Huntington's disease (HD). We generated induced pluripotent stem cells (iPSC) from primary dermal fibroblasts of 5 patients with HD and 5 control subjects. A significant fraction of the HD iPSC lines had genomic abnormalities as assessed by karyotype analysis, while none of our control lines had detectable genomic abnormalities. We demonstrate a statistically significant increase in genomic instability in HD cells during reprogramming. We also report a significant association with repeat length and severity of this instability. Our karyotypically normal HD iPSCs also have elevated ATM-p53 signaling as shown by elevated levels of phosphorylated p53 and H2AX, indicating either elevated DNA damage or hypersensitive DNA damage signaling in HD iPSCs. Thus, increased DNA damage responses in the HD genotype is coincidental with the observed chromosomal aberrations. We conclude that the disease causing mutation in HD increases the propensity of chromosomal instability relative to control fibroblasts specifically during reprogramming to a pluripotent state by a commonly used episomal-based method that includes p53 knockdown.

    View details for DOI 10.1371/journal.pone.0150372

    View details for Web of Science ID 000372574900038

    View details for PubMedID 26982737

    View details for PubMedCentralID PMC4794230

  • Age-related sperm DNA methylation changes are transmitted to offspring and associated with abnormal behavior and dysregulated gene expression MOLECULAR PSYCHIATRY Milekic, M. H., Xin, Y., O'Donnell, A., Kumar, K. K., Bradley-Moore, M., Malaspina, D., Moore, H., Brunner, D., Ge, Y., Edwards, J., Paul, S., Haghighi, F. G., Gingrich, J. A. 2015; 20 (8): 995-1001

    Abstract

    Advanced paternal age (APA) has been shown to be a significant risk factor in the offspring for neurodevelopmental psychiatric disorders, such as schizophrenia and autism spectrum disorders. During aging, de novo mutations accumulate in the male germline and are frequently transmitted to the offspring with deleterious effects. In addition, DNA methylation during spermatogenesis is an active process, which is susceptible to errors that can be propagated to subsequent generations. Here we test the hypothesis that the integrity of germline DNA methylation is compromised during the aging process. A genome-wide DNA methylation screen comparing sperm from young and old mice revealed a significant loss of methylation in the older mice in regions associated with transcriptional regulation. The offspring of older fathers had reduced exploratory and startle behaviors and exhibited similar brain DNA methylation abnormalities as observed in the paternal sperm. Offspring from old fathers also had transcriptional dysregulation of developmental genes implicated in autism and schizophrenia. Our findings demonstrate that DNA methylation abnormalities arising in the sperm of old fathers are a plausible mechanism to explain some of the risks that APA poses to resulting offspring.

    View details for DOI 10.1038/mp.2014.84

    View details for Web of Science ID 000358527100008

    View details for PubMedID 25092244

  • Deficits in neural responses to manganese exposure in Huntington's disease models Tidball, A. M., Kumar, K. K., Bryan, M. R., Bichell, T. J., Horning, K., Uhouse, M. A., Goodwin, C. R., Bornhorst, J., Schwerdtle, T., Neely, M. D., McClean, J. A., Aschner, M. A., Bowman, A. B. PERGAMON-ELSEVIER SCIENCE LTD. 2015: 105
  • A novel manganese-dependent ATM-p53 signaling pathway is selectively impaired in patient-based neuroprogenitor and murine striatal models of Huntington's disease HUMAN MOLECULAR GENETICS Tidball, A. M., Bryan, M. R., Uhouse, M. A., Kumar, K. K., Aboud, A. A., Feist, J. E., Ess, K. C., Neely, M. D., Aschner, M., Bowman, A. B. 2015; 24 (7): 1929-1944

    Abstract

    The essential micronutrient manganese is enriched in brain, especially in the basal ganglia. We sought to identify neuronal signaling pathways responsive to neurologically relevant manganese levels, as previous data suggested that alterations in striatal manganese handling occur in Huntington's disease (HD) models. We found that p53 phosphorylation at serine 15 is the most responsive cell signaling event to manganese exposure (of 18 tested) in human neuroprogenitors and a mouse striatal cell line. Manganese-dependent activation of p53 was severely diminished in HD cells. Inhibitors of ataxia telangiectasia mutated (ATM) kinase decreased manganese-dependent phosphorylation of p53. Likewise, analysis of ATM autophosphorylation and additional ATM kinase targets, H2AX and CHK2, support a role for ATM in the activation of p53 by manganese and that a defect in this process occurs in HD. Furthermore, the deficit in Mn-dependent activation of ATM kinase in HD neuroprogenitors was highly selective, as DNA damage and oxidative injury, canonical activators of ATM, did not show similar deficits. We assessed cellular manganese handling to test for correlations with the ATM-p53 pathway, and we observed reduced Mn accumulation in HD human neuroprogenitors and HD mouse striatal cells at manganese exposures associated with altered p53 activation. To determine if this phenotype contributes to the deficit in manganese-dependent ATM activation, we used pharmacological manipulation to equalize manganese levels between HD and control mouse striatal cells and rescued the ATM-p53 signaling deficit. Collectively, our data demonstrate selective alterations in manganese biology in cellular models of HD manifest in ATM-p53 signaling.

    View details for DOI 10.1093/hmg/ddu609

    View details for Web of Science ID 000353065300011

    View details for PubMedID 25489053

    View details for PubMedCentralID PMC4355025

  • PARK2 patient neuroprogenitors show increased mitochondrial sensitivity to copper NEUROBIOLOGY OF DISEASE Aboud, A. A., Tidball, A. M., Kumar, K. K., Neely, M. D., Han, B., Ess, K. C., Hong, C. C., Erikson, K. M., Hedera, P., Bowman, A. B. 2015; 73: 204-212

    Abstract

    Poorly-defined interactions between environmental and genetic risk factors underlie Parkinson's disease (PD) etiology. Here we tested the hypothesis that human stem cell derived forebrain neuroprogenitors from patients with known familial risk for early onset PD will exhibit enhanced sensitivity to PD environmental risk factors compared to healthy control subjects without a family history of PD. Two male siblings (SM and PM) with biallelic loss-of-function mutations in PARK2 were identified. Human induced pluripotent stem cells (hiPSCs) from SM, PM, and four control subjects with no known family histories of PD or related neurodegenerative diseases were utilized. We tested the hypothesis that hiPSC-derived neuroprogenitors from patients with PARK2 mutations would show heightened cell death, mitochondrial dysfunction, and reactive oxygen species generation compared to control cells as a result of exposure to heavy metals (PD environmental risk factors). We report that PARK2 mutant neuroprogenitors showed increased cytotoxicity with copper (Cu) and cadmium (Cd) exposure but not manganese (Mn) or methyl mercury (MeHg) relative to control neuroprogenitors. PARK2 mutant neuroprogenitors also showed a substantial increase in mitochondrial fragmentation, initial ROS generation, and loss of mitochondrial membrane potential following Cu exposure. Our data substantiate Cu exposure as an environmental risk factor for PD. Furthermore, we report a shift in the lowest observable effect level (LOEL) for greater sensitivity to Cu-dependent mitochondrial dysfunction in patients SM and PM relative to controls, correlating with their increased genetic risk for PD.

    View details for DOI 10.1016/j.nbd2014.10.002

    View details for Web of Science ID 000346328100018

    View details for PubMedID 25315681

    View details for PubMedCentralID PMC4394022

  • Untargeted metabolic profiling identifies interactions between Huntington's disease and neuronal manganese status METALLOMICS Kumar, K. K., Goodwin, C. R., Uhouse, M. A., Bornhorst, J., Schwerdtle, T., Aschner, M., McLean, J. A., Bowman, A. B. 2015; 7 (2): 363-370

    Abstract

    Manganese (Mn) is an essential micronutrient for development and function of the nervous system. Deficiencies in Mn transport have been implicated in the pathogenesis of Huntington's disease (HD), an autosomal dominant neurodegenerative disorder characterized by loss of medium spiny neurons of the striatum. Brain Mn levels are highest in striatum and other basal ganglia structures, the most sensitive brain regions to Mn neurotoxicity. Mouse models of HD exhibit decreased striatal Mn accumulation and HD striatal neuron models are resistant to Mn cytotoxicity. We hypothesized that the observed modulation of Mn cellular transport is associated with compensatory metabolic responses to HD pathology. Here we use an untargeted metabolomics approach by performing ultraperformance liquid chromatography-ion mobility-mass spectrometry (UPLC-IM-MS) on control and HD immortalized mouse striatal neurons to identify metabolic disruptions under three Mn exposure conditions, low (vehicle), moderate (non-cytotoxic) and high (cytotoxic). Our analysis revealed lower metabolite levels of pantothenic acid, and glutathione (GSH) in HD striatal cells relative to control cells. HD striatal cells also exhibited lower abundance and impaired induction of isobutyryl carnitine in response to increasing Mn exposure. In addition, we observed induction of metabolites in the pentose shunt pathway in HD striatal cells after high Mn exposure. These findings provide metabolic evidence of an interaction between the HD genotype and biologically relevant levels of Mn in a striatal cell model with known HD by Mn exposure interactions. The metabolic phenotypes detected support existing hypotheses that changes in energetic processes underlie the pathobiology of both HD and Mn neurotoxicity.

    View details for DOI 10.1039/c4mt00223g

    View details for Web of Science ID 000349470000018

    View details for PubMedID 25599126

    View details for PubMedCentralID PMC4326616

  • Cellular manganese content is developmentally regulated in human dopaminergic neurons SCIENTIFIC REPORTS Kumar, K. K., Lowe, E. W., Aboud, A. A., Neely, M. D., Redha, R., Bauer, J. A., Odak, M., Weaver, C. D., Meiler, J., Aschner, M., Bowman, A. B. 2014; 4

    Abstract

    Manganese (Mn) is both an essential biological cofactor and neurotoxicant. Disruption of Mn biology in the basal ganglia has been implicated in the pathogenesis of neurodegenerative disorders, such as parkinsonism and Huntington's disease. Handling of other essential metals (e.g. iron and zinc) occurs via complex intracellular signaling networks that link metal detection and transport systems. However, beyond several non-selective transporters, little is known about the intracellular processes regulating neuronal Mn homeostasis. We hypothesized that small molecules that modulate intracellular Mn could provide insight into cell-level Mn regulatory mechanisms. We performed a high throughput screen of 40,167 small molecules for modifiers of cellular Mn content in a mouse striatal neuron cell line. Following stringent validation assays and chemical informatics, we obtained a chemical 'toolbox' of 41 small molecules with diverse structure-activity relationships that can alter intracellular Mn levels under biologically relevant Mn exposures. We utilized this toolbox to test for differential regulation of Mn handling in human floor-plate lineage dopaminergic neurons, a lineage especially vulnerable to environmental Mn exposure. We report differential Mn accumulation between developmental stages and stage-specific differences in the Mn-altering activity of individual small molecules. This work demonstrates cell-level regulation of Mn content across neuronal differentiation.

    View details for DOI 10.1038/srep06801

    View details for Web of Science ID 000343980500009

    View details for PubMedID 25348053

    View details for PubMedCentralID PMC4210885

  • Optimization of Fluorescence Assay of Cellular Manganese Status for High Throughput Screening JOURNAL OF BIOCHEMICAL AND MOLECULAR TOXICOLOGY Kumar, K. K., Aboud, A. A., Patel, D. K., Aschner, M., Bowman, A. B. 2013; 27 (1): 42-49

    Abstract

    The advent of high throughput screening (HTS) technology permits identification of compounds that influence various cellular phenotypes. However, screening for small molecule chemical modifiers of neurotoxicants has been limited by the scalability of existing phenotyping assays. Furthermore, the adaptation of existing cellular assays to HTS format requires substantial modification of experimental parameters and analysis methodology to meet the necessary statistical requirements. Here we describe the successful optimization of the Cellular Fura-2 Manganese Extraction Assay (CFMEA) for HTS. By optimizing cellular density, manganese (Mn) exposure conditions, and extraction parameters, the sensitivity and dynamic range of the fura-2 Mn response was enhanced to permit detection of positive and negative modulators of cellular manganese status. Finally, we quantify and report strategies to control sources of intra- and interplate variability by batch level and plate-geometric level analysis. Our goal is to enable HTS with the CFMEA to identify novel modulators of Mn transport.

    View details for DOI 10.1002/jbt.21457

    View details for Web of Science ID 000313777200005

    View details for PubMedID 23169769

    View details for PubMedCentralID PMC3774111

  • Down Syndrome COMPREHENSIVE DEVELOPMENTAL NEUROSCIENCE: NEURAL CIRCUIT DEVELOPMENT AND FUNCTION IN THE HEALTHY AND DISEASED BRAIN Bowman, A. B., Ess, K. C., Kumar, K. K., Summar, K. L., Rubenstein, J. L., Rakic, P. 2013: 547–71
  • Genetic risk for Parkinson's disease correlates with alterations in neuronal manganese sensitivity between two human subjects NEUROTOXICOLOGY Aboud, A. A., Tidball, A. M., Kumar, K. K., Neely, M. D., Ess, K. C., Erikson, K. M., Bowman, A. B. 2012; 33 (6): 1443-1449

    Abstract

    Manganese (Mn) is an environmental risk factor for Parkinson's disease (PD). Recessive inheritance of PARK2 mutations is strongly associated with early onset PD (EOPD). It is widely assumed that the influence of PD environmental risk factors may be enhanced by the presence of PD genetic risk factors in the genetic background of individuals. However, such interactions may be difficult to predict owing to the complexities of genetic and environmental interactions. Here we examine the potential of human induced pluripotent stem (iPS) cell-derived early neural progenitor cells (NPCs) to model differences in Mn neurotoxicity between a control subject (CA) with no known PD genetic risk factors and a subject (SM) with biallelic loss-of-function mutations in PARK2 and family history of PD but no evidence of PD by neurological exam. Human iPS cells were generated from primary dermal fibroblasts of both subjects. We assessed several outcome measures associated with Mn toxicity and PD. No difference in sensitivity to Mn cytotoxicity or mitochondrial fragmentation was observed between SM and CA NPCs. However, we found that Mn exposure was associated with significantly higher reactive oxygen species (ROS) generation in SM compared to CA NPCs despite significantly less intracellular Mn accumulation. Thus, this report offers the first example of human subject-specific differences in PD-relevant environmental health related phenotypes that are consistent with pathogenic interactions between known genetic and environmental risk factors for PD.

    View details for DOI 10.1016/j.neuro.2012.10.009

    View details for Web of Science ID 000313027100007

    View details for PubMedID 23099318

    View details for PubMedCentralID PMC3518601

  • The potential of induced pluripotent stem cells as a translational model for neurotoxicological risk NEUROTOXICOLOGY Kumar, K. K., Aboud, A. A., Bowman, A. B. 2012; 33 (3): 518-529

    Abstract

    An important goal of neurotoxicological research is to provide relevant and accurate risk assessment of environmental and pharmacological agents for populations and individuals. Owing to the challenges of human subject research and the real possibility of species specific toxicological responses, neuronal lineages derived from human embryonic stem cells (hESCs) and human neuronal precursors have been offered as a potential solution for validation of neurotoxicological data from model organism systems in humans. More recently, with the advent of induced pluripotent stem cell (iPSC) technology, there is now the possibility of personalized toxicological risk assessment, the ability to predict individual susceptibility to specific environmental agents, by this approach. This critical advance is widely expected to facilitate analysis of cellular physiological pathways in the context of human neurons and the underlying genetic factors that lead to disease. Thus this technology opens the opportunity, for the first time, to characterize the physiological, toxicological, pharmacological and molecular properties of living human neurons with identical genetic determinants as human patients. Furthermore, armed with a complete clinical history of the patients, human iPSC (hiPSC) studies can theoretically compare patients and at risk groups with distinct sensitivities to particular environmental agents, divergent clinical outcomes, differing co-morbidities, and so forth. Thus iPSCs and neuronal lineages derived from them may reflect the unique genetic blueprint of the individuals from which they are generated. Indeed, iPSC technology has the potential to revolutionize scientific approaches to human health. However, before this overarching goal can be reached a number of technical and theoretical challenges must be overcome. This review seeks to provide a realistic assessment of hiPSC technology and its application to risk assessment and mechanistic studies in the area of neurotoxicology. We seek to identify, prioritize, and detail the primary hurdles that need to be overcome if personalized toxicological risk assessment using patient-derived iPSCs is to succeed.

    View details for DOI 10.1016/j.neuro.2012.02.005

    View details for Web of Science ID 000304730100031

    View details for PubMedID 22330734

    View details for PubMedCentralID PMC3358591

  • Bone loss in anorexia nervosa: leptin, serotonin, and the sympathetic nervous system MOLECULAR AND INTEGRATIVE PHYSIOLOGY OF THE MUSCULOSKELETAL SYSTEM Kumar, K. K., Tung, S., Lqbal, J. 2010; 1211: 51-65

    Abstract

    Anorexia nervosa (AN), a disorder characterized by the refusal to sustain a healthy weight, has the highest mortality of any psychiatric disorder. This review presents a model of AN that ties together advances in our understanding of how leptin, serotonin, and hypogonadism are brought about in AN and how they influence bone mass. Serotonin (5-hydroxytryptamine) is a key regulator of satiety and mood. The primary disturbance in AN results from alterations in serotonin signaling. AN patients suffer from serotonergic hyperactivity of Htr1a-dependent pathways that causes dysphoric mood and promotes restrictive behavior. By limiting carbohydrate ingestion, anorexics decrease their serotonin levels. Reduced serotonergic signaling in turn suppresses appetite through Htr1a/2b, decreases dysphoric mood through Htr1a/2a, and activates the sympathetic nervous system (SNS) through Htr2c receptors in the ventromedial hypothalamus. Activation of the SNS decreases bone mass through β2-adrenergic signaling in osteoblasts. Additional topics reviewed here include osteoblastic feedback of metabolism in anorexia, mechanisms whereby dietary changes exacerbate bone loss, the role of caloric restriction and Sirt1 in bone metabolism, hypothalamic hypogonadism's effects on bone mass, and potential treatments.

    View details for DOI 10.1111/j.1749-6632.2010.05810.x

    View details for Web of Science ID 000287463400006

    View details for PubMedID 21062295

  • Selective upregulation of the ADP-ribosyl cyclases CD38 and CD157 by TNF but not by RANK-L reveals differences in downstream signaling AMERICAN JOURNAL OF PHYSIOLOGY-RENAL PHYSIOLOGY Iqbal, J., Kumar, K., Sun, L., Zaidi, M. 2006; 291 (3): F557-F566

    Abstract

    In macrophages and osteoclast precursors, the cytokines TNF and RANK-L induce similar downstream pathways and share some of the same adaptor molecules. However, despite these similarities, no defined signaling schematic has emerged to show how each cytokine favors particular pathways. In this report, we investigate whether TNF and RANK-L differentially regulate ADP-ribosyl cyclases-enzymes that are unique in being crucial for immunological function yet detrimental to osteoclastogenesis. TNF but not RANK-L led to the sustained upregulation of both CD38 and CD157 as demonstrated by real-time PCR and flow cytometry. Further investigation demonstrated that this upregulation was a result of continuous, direct TNF signaling and involved JNK, and more critically PKC and NF-kappaB. Using this approach allowed us to highlight the relative importance of the PKC, NF-kappaB, and JNK pathways in actualizing proper outcomes of TNF signaling. Albeit speculative, we believe that differences between TNF- and RANK-l-induced activation of downstream signaling pathways, in particular PKC, are crucial for determining whether progenitor cells become geared for immunity or bone resorption.

    View details for DOI 10.1152/ajprenal.00066.2006

    View details for Web of Science ID 000239658900006

    View details for PubMedID 16705149