123 results on '"John Pluta"'
Search Results
2. Identification of 22 susceptibility loci associated with testicular germ cell tumors
- Author
-
John Pluta, Louise C. Pyle, Kevin T. Nead, Rona Wilf, Mingyao Li, Nandita Mitra, Benita Weathers, Kurt D’Andrea, Kristian Almstrup, Lynn Anson-Cartwright, Javier Benitez, Christopher D. Brown, Stephen Chanock, Chu Chen, Victoria K. Cortessis, Alberto Ferlin, Carlo Foresta, Marija Gamulin, Jourik A. Gietema, Chiara Grasso, Mark H. Greene, Tom Grotmol, Robert J. Hamilton, Trine B. Haugen, Russ Hauser, Michelle A. T. Hildebrandt, Matthew E. Johnson, Robert Karlsson, Lambertus A. Kiemeney, Davor Lessel, Ragnhild A. Lothe, Jennifer T. Loud, Chey Loveday, Paloma Martin-Gimeno, Coby Meijer, Jérémie Nsengimana, David I. Quinn, Thorunn Rafnar, Shweta Ramdas, Lorenzo Richiardi, Rolf I. Skotheim, Kari Stefansson, Clare Turnbull, David J. Vaughn, Fredrik Wiklund, Xifeng Wu, Daphne Yang, Tongzhang Zheng, Andrew D. Wells, Struan F. A. Grant, Ewa Rajpert-De Meyts, Stephen M. Schwartz, D. Timothy Bishop, Katherine A. McGlynn, Peter A. Kanetsky, Katherine L. Nathanson, and The Testicular Cancer Consortium
- Subjects
Science - Abstract
Testicular germ cell tumors are highly heritable, and the authors present the largest genome association study, identifying 22 novel loci, which account for a third of those identified to date. Implicated pathways include male germ cell development and differentiation, and chromosomal segregation.
- Published
- 2021
- Full Text
- View/download PDF
3. Progress update from the hippocampal subfields group
- Author
-
Rosanna K. Olsen, Valerie A. Carr, Ana M. Daugherty, Renaud La Joie, Robert S.C. Amaral, Katrin Amunts, Jean C. Augustinack, Arnold Bakker, Andrew R. Bender, David Berron, Marina Boccardi, Martina Bocchetta, Alison C. Burggren, M. Mallar Chakravarty, Gaël Chételat, Robin deFlores, Jordan DeKraker, Song‐Lin Ding, Mirjam I. Geerlings, Yushan Huang, Ricardo Insausti, Elliott G. Johnson, Prabesh Kanel, Olga Kedo, Kristen M. Kennedy, Attila Keresztes, Joshua K. Lee, Ulman Lindenberger, Susanne G. Mueller, Elizabeth M. Mulligan, Noa Ofen, Daniela J. Palombo, Lorenzo Pasquini, John Pluta, Naftali Raz, Karen M. Rodrigue, Margaret L. Schlichting, Yee Lee Shing, Craig E.L. Stark, Trevor A. Steve, Nanthia A. Suthana, Lei Wang, Markus Werkle‐Bergner, Paul A. Yushkevich, Qijing Yu, Laura E.M. Wisse, and Hippocampal Subfields Group
- Subjects
Hippocampus ,Volumetry ,Human ,Neuroimaging ,Structural imaging ,Neuroanatomy ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Introduction Heterogeneity of segmentation protocols for medial temporal lobe regions and hippocampal subfields on in vivo magnetic resonance imaging hinders the ability to integrate findings across studies. We aim to develop a harmonized protocol based on expert consensus and histological evidence. Methods Our international working group, funded by the EU Joint Programme–Neurodegenerative Disease Research (JPND), is working toward the production of a reliable, validated, harmonized protocol for segmentation of medial temporal lobe regions. The working group uses a novel postmortem data set and online consensus procedures to ensure validity and facilitate adoption. Results This progress report describes the initial results and milestones that we have achieved to date, including the development of a draft protocol and results from the initial reliability tests and consensus procedures. Discussion A harmonized protocol will enable the standardization of segmentation methods across laboratories interested in medial temporal lobe research worldwide.
- Published
- 2019
- Full Text
- View/download PDF
4. Integrated Molecular Characterization of Testicular Germ Cell Tumors
- Author
-
Hui Shen, Juliann Shih, Daniel P. Hollern, Linghua Wang, Reanne Bowlby, Satish K. Tickoo, Vésteinn Thorsson, Andrew J. Mungall, Yulia Newton, Apurva M. Hegde, Joshua Armenia, Francisco Sánchez-Vega, John Pluta, Louise C. Pyle, Rohit Mehra, Victor E. Reuter, Guilherme Godoy, Jeffrey Jones, Carl S. Shelley, Darren R. Feldman, Daniel O. Vidal, Davor Lessel, Tomislav Kulis, Flavio M. Cárcano, Kristen M. Leraas, Tara M. Lichtenberg, Denise Brooks, Andrew D. Cherniack, Juok Cho, David I. Heiman, Katayoon Kasaian, Minwei Liu, Michael S. Noble, Liu Xi, Hailei Zhang, Wanding Zhou, Jean C. ZenKlusen, Carolyn M. Hutter, Ina Felau, Jiashan Zhang, Nikolaus Schultz, Gad Getz, Matthew Meyerson, Joshua M. Stuart, Rehan Akbani, David A. Wheeler, Peter W. Laird, Katherine L. Nathanson, Victoria K. Cortessis, Katherine A. Hoadley, David Wheeler, Daniel Hughes, Kyle Covington, Joy C. Jayaseelan, Viktoriya Korchina, Lora Lewis, Jianhong Hu, HarshaVardhan Doddapaneni, Donna Muzny, Richard Gibbs, Daniel Hollern, Benjamin G. Vincent, Shengjie Chai, Christof C. Smith, J. Todd Auman, Yan Shi, Shaowu Meng, Tara Skelly, Donghui Tan, Umadevi Veluvolu, Piotr A. Mieczkowski, Corbin D. Jones, Matthew D. Wilkerson, Saianand Balu, Tom Bodenheimer, Alan P. Hoyle, Stuart R. Jefferys, Lisle E. Mose, Janae V. Simons, Matthew G. Soloway, Jeffrey Roach, Joel S. Parker, D. Neil Hayes, Charles M. Perou, Gordon Saksena, Carrie Cibulskis, Steven E. Schumacher, Rameen Beroukhim, Stacey B. Gabriel, Adrian Ally, Miruna Balasundaram, Rebecca Carlsen, Dorothy Cheung, Eric Chuah, Noreen Dhalla, Robert A. Holt, Steven J.M. Jones, Yussanne Ma, Michael Mayo, Richard A. Moore, A. Gordon Robertson, Jacqueline E. Schein, Payal Sipahimalani, Angela Tam, Nina Thiessen, Tina Wong, Marco A. Marra, Daniel J. Weisenberger, David J. Van Den Berg, Phillip H. Lai, Mario Berrios, Andrea Holbrook, Moiz S. Bootwalla, Dennis T. Maglinte, Debyani Chakravarty, Jianjiong Gao, Zachary Heins, Ritika Kundra, Angelica Ochoa, Chris Sander, Marc Ladanyi, Vesteinn Thorsson, Amie J. Radenbaugh, Nils Gehlenborg, Doug Voet, Pei Lin, Scott Frazer, Jaegil Kim, Michael S. Lawrence, Sam Meier, Timothy Defreitas, Lynda Chin, John N. Weinstein, Wenbin Liu, Gordon B. Mills, Yiling Lu, Leendert Looijenga, Alan H. Bryce, André L. Carvalho, Darren Feldman, Michael Ittmann, Seth Lerner, Jay Bowen, Julie M. Gastier-Foster, Mark Gerken, Carmen Helsel, Nilsa C. Ramirez, Lisa Wise, Erik Zmuda, Sandra Cottingham, David Chesla, Charles Saller, Katherine Tarvin, Luiz Fernando Lopes, Cristovam Scapulatempo-Neto, Natália D.A. Aredes, Wolter Oosterhuis, Ad Gillis, Hans Stoop, Wil Eijkenboom, George Sandusky, Sue Ellen Martin, Manju Aron, Siamak Daneshmand, Hooman Djaladat, David Quinn, Tanya Dorff, Jochen K. Lennerz, Leigh B. Thorne, Marija Gamulin, Zeljko Kastelan, Tvrtko Hudolin, Christian Kubisch, Lori Boice, Mei Huang, Amy H. Perou, W. Kimryn Rathmell, Todd Pihl, Yunhu Wan, Qiang Sun, Rashi Naresh, Sudha Chudamani, Jia Liu, Laxmi Lolla, Ye Wu, Martin L. Ferguson, Jean C. Zenklusen, Jiashan (Julia) Zhang, Margi Sheth, John A. Demchok, Liming Yang, Zhining Wang, Roy Tarnuzzer, Heidi J. Sofia, and Tanja M. Davidsen
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: We studied 137 primary testicular germ cell tumors (TGCTs) using high-dimensional assays of genomic, epigenomic, transcriptomic, and proteomic features. These tumors exhibited high aneuploidy and a paucity of somatic mutations. Somatic mutation of only three genes achieved significance—KIT, KRAS, and NRAS—exclusively in samples with seminoma components. Integrated analyses identified distinct molecular patterns that characterized the major recognized histologic subtypes of TGCT: seminoma, embryonal carcinoma, yolk sac tumor, and teratoma. Striking differences in global DNA methylation and microRNA expression between histology subtypes highlight a likely role of epigenomic processes in determining histologic fates in TGCTs. We also identified a subset of pure seminomas defined by KIT mutations, increased immune infiltration, globally demethylated DNA, and decreased KRAS copy number. We report potential biomarkers for risk stratification, such as miRNA specifically expressed in teratoma, and others with molecular diagnostic potential, such as CpH (CpA/CpC/CpT) methylation identifying embryonal carcinomas. : Shen et al. identify molecular characteristics that classify testicular germ cell tumor types, including a separate subset of seminomas defined by KIT mutations. This provides a set of candidate biomarkers for risk stratification and potential therapeutic targeting. Keywords: The Cancer Genome Atlas, testicular germ cell tumors, seminoma, nonseminoma, DNA methylation, exome sequencing, KIT, copy number, miR-375
- Published
- 2018
- Full Text
- View/download PDF
5. BRCA locus-specific loss of heterozygosity in germline BRCA1 and BRCA2 carriers
- Author
-
Kara N. Maxwell, Bradley Wubbenhorst, Brandon M. Wenz, Daniel De Sloover, John Pluta, Lyndsey Emery, Amanda Barrett, Adam A. Kraya, Ioannis N. Anastopoulos, Shun Yu, Yuchao Jiang, Hao Chen, Nancy R. Zhang, Nicole Hackman, Kurt D’Andrea, Robert Daber, Jennifer J. D. Morrissette, Nandita Mitra, Michael Feldman, Susan M. Domchek, and Katherine L. Nathanson
- Subjects
Science - Abstract
Most tumours associated with germline BRCA1/BRCA2 loss of function mutations respond to DNA damaging agents, however, some do not. Herein, the authors identify that a subset of breast/ovarian tumors retain a normal allele, which is associated with decreased overall survival after DNA damage-inducing platinum chemotherapy.
- Published
- 2017
- Full Text
- View/download PDF
6. Genomic profiling of human vascular cells identifies TWIST1 as a causal gene for common vascular diseases.
- Author
-
Sylvia T Nurnberg, Marie A Guerraty, Robert C Wirka, H Shanker Rao, Milos Pjanic, Scott Norton, Felipe Serrano, Ljubica Perisic, Susannah Elwyn, John Pluta, Wei Zhao, Stephanie Testa, YoSon Park, Trieu Nguyen, Yi-An Ko, Ting Wang, Ulf Hedin, Sanjay Sinha, Yoseph Barash, Christopher D Brown, Thomas Quertermous, and Daniel J Rader
- Subjects
Genetics ,QH426-470 - Abstract
Genome-wide association studies have identified multiple novel genomic loci associated with vascular diseases. Many of these loci are common non-coding variants that affect the expression of disease-relevant genes within coronary vascular cells. To identify such genes on a genome-wide level, we performed deep transcriptomic analysis of genotyped primary human coronary artery smooth muscle cells (HCASMCs) and coronary endothelial cells (HCAECs) from the same subjects, including splicing Quantitative Trait Loci (sQTL), allele-specific expression (ASE), and colocalization analyses. We identified sQTLs for TARS2, YAP1, CFDP1, and STAT6 in HCASMCs and HCAECs, and 233 ASE genes, a subset of which are also GTEx eGenes in arterial tissues. Colocalization of GWAS association signals for coronary artery disease (CAD), migraine, stroke and abdominal aortic aneurysm with GTEx eGenes in aorta, coronary artery and tibial artery discovered novel candidate risk genes for these diseases. At the CAD and stroke locus tagged by rs2107595 we demonstrate colocalization with expression of the proximal gene TWIST1. We show that disrupting the rs2107595 locus alters TWIST1 expression and that the risk allele has increased binding of the NOTCH signaling protein RBPJ. Finally, we provide data that TWIST1 expression influences vascular SMC phenotypes, including proliferation and calcification, as a potential mechanism supporting a role for TWIST1 in CAD.
- Published
- 2020
- Full Text
- View/download PDF
7. Neural Correlates of Verbal Episodic Memory and Lexical Retrieval in Logopenic Variant Primary Progressive Aphasia
- Author
-
Khaing T. Win, John Pluta, Paul Yushkevich, David J. Irwin, Corey T. McMillan, Katya Rascovsky, David Wolk, and Murray Grossman
- Subjects
logopenic primary progressive aphasia ,Alzheimer's disease ,verbal episodic memory ,lexical retrieval ,hippocampal subfields ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Objective: Logopenic variant primary progressive aphasia (lvPPA) is commonly associated with Alzheimer's disease (AD) pathology. But lvPPA patients display different cognitive and anatomical profile from the common clinical AD patients, whose verbal episodic memory is primarily affected. Reports of verbal episodic memory difficulty in lvPPA are inconsistent, and we hypothesized that their lexical retrieval impairment contributes to verbal episodic memory performance and is associated with left middle temporal gyrus atrophy.Methods: We evaluated patients with lvPPA (n = 12) displaying prominent word-finding and repetition difficulties, and a demographically-matched cohort of clinical Alzheimer's disease (AD, n = 26), and healthy seniors (n = 16). We assessed lexical retrieval with confrontation naming and verbal episodic memory with delayed free recall. Whole-brain regressions related naming and delayed free recall to gray matter atrophy. Medial temporal lobe (MTL) subfields were examined using high in-plane resolution imaging.Results: lvPPA patients had naming and delayed free recall impairments, but intact recognition memory. In lvPPA, delayed free recall was related to naming; both were associated with left middle temporal gyrus atrophy but not MTL atrophy. Despite cerebrospinal fluid evidence consistent with AD pathology, examination of MTL subfields revealed no atrophy in lvPPA. While AD patients displayed impaired delayed free recall, this deficit did not correlate with naming. Regression analyses related delayed free recall deficits in clinical AD patients to MTL subfield atrophy, and naming to left middle temporal gyrus atrophy.Conclusion: Unlike amnestic AD patients, MTL subfields were not affected in lvPPA patients. Verbal episodic memory deficit observed in lvPPA was unlikely to be due to a hippocampal-mediated mechanism but appeared to be due to poor lexical retrieval. Relative sparing of MTL volume and intact recognition memory are consistent with previous reports of hippocampal-sparing variant cases of AD pathology, where neurofibrillary tangles are disproportionately distributed in cortical areas with relative sparing of the hippocampus. This suggests that AD neuropathology in lvPPA may originate in neuronal networks outside of the MTL, which deviates from the typical Braak staging pattern of spreading pathology in clinical AD.
- Published
- 2017
- Full Text
- View/download PDF
8. Characterizing the human hippocampus in aging and Alzheimer's disease using a computational atlas derived from ex vivo MRI and histology.
- Author
-
Daniel H. Adler, Laura E. M. Wisse, Ranjit Ittyerah, John Pluta, Song-Lin Ding, Long Xie, Jiancong Wang, Salmon Kadivar, John L. Robinson, Theresa Schuck, John Q. Trojanowski, Murray Grossman, John A. Detre, Mark A. Elliott, Jon B. Toledo, Weixia Liu, Stephen Pickup, Michael I. Miller, Sandhitsu R. Das, David A. Wolk, and Paul A. Yushkevich
- Published
- 2018
- Full Text
- View/download PDF
9. A framework for informing segmentation of in vivo MRI with information derived from ex vivo imaging: Application in the medial temporal lobe.
- Author
-
Paul A. Yushkevich, Laura E. M. Wisse, Daniel H. Adler, Ranjit Ittyerah, John Pluta, John L. Robinson, Theresa Schuck, John Q. Trojanowski, Murray Grossman, John A. Detre, Mark A. Elliott, Jon B. Toledo, Weixia Liu, Stephen Pickup, Sandhitsu R. Das, and David A. Wolk
- Published
- 2016
- Full Text
- View/download PDF
10. Probabilistic Atlas of the Human Hippocampus Combining Ex Vivo MRI and Histology.
- Author
-
Daniel H. Adler, Ranjit Ittyerah, John Pluta, Stephen Pickup, Weixia Liu, David A. Wolk, and Paul A. Yushkevich
- Published
- 2016
- Full Text
- View/download PDF
11. Multi-template analysis of human perirhinal cortex in brain MRI: Explicitly accounting for anatomical variability.
- Author
-
Long Xie, John Pluta, Sandhitsu R. Das, Laura E. M. Wisse, Hongzhi Wang 0002, Lauren Mancuso, Dasha Kliot, Brian B. Avants, Song-Lin Ding, José V. Manjón, David A. Wolk, and Paul A. Yushkevich
- Published
- 2017
- Full Text
- View/download PDF
12. Data from Genomic Signatures Predict the Immunogenicity of BRCA-Deficient Breast Cancer
- Author
-
Katherine L. Nathanson, Robert H. Vonderheide, Susan M. Domchek, Anupma Nayak, Michael Feldman, Jennifer J.D. Morrissette, Nandita Mitra, Amanda Barrett, Nicole Lunceford, Liza M. Dorfman, Andrew J. Rech, John Pluta, Brandon M. Wenz, Bradley Wubbenhorst, Kara N. Maxwell, and Adam A. Kraya
- Abstract
Purpose:Breast cancers with BRCA1/2 alterations have a relatively high mutational load, suggesting that immune checkpoint blockade may be a potential treatment option. However, the degree of immune cell infiltration varies widely, and molecular features contributing to this variability remain unknown.Experimental Design:We hypothesized that genomic signatures might predict immunogenicity in BRCA1/2 breast cancers. Using The Cancer Genome Atlas (TCGA) genomic data, we compared breast cancers with (89) and without (770) either germline or somatic BRCA1/2 alterations. We also studied 35 breast cancers with germline BRCA1/2 mutations from Penn using WES and IHC.Results:We found that homologous recombination deficiency (HRD) scores were negatively associated with expression-based immune indices [cytolytic index (P = 0.04), immune ESTIMATE (P = 0.002), type II IFN signaling (P = 0.002)] despite being associated with a higher mutational/neoantigen burden, in BRCA1/2 mutant breast cancers. Further, absence of allele-specific loss of heterozygosity (LOH negative; P = 0.01) or subclonality (P = 0.003) of germline and somatic BRCA1/2 mutations, respectively, predicted for heightened cytolytic activity. Gene set analysis found that multiple innate and adaptive immune pathways that converge on NF-κB may contribute to this heightened immunogenicity. IHC of Penn breast cancers demonstrated increased CD45+ (P = 0.039) and CD8+ infiltrates (P = 0.037) and increased PDL1 expression (P = 0.012) in HRD-low or LOH-negative cancers. Triple-negative cancers with low HRD had far greater CD8+ T cells (P = 0.0011) and Perforin 1 expression (P = 0.014) compared with hormone receptor-positive HRD-high cancers.Conclusions:HRD scores and hormone receptor subtype are predictive of immunogenicity in BRCA1/2 breast cancers and may inform the design of optimal immune therapeutic strategies.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Data from Genomic Signatures Predict the Immunogenicity of BRCA-Deficient Breast Cancer
- Author
-
Katherine L. Nathanson, Robert H. Vonderheide, Susan M. Domchek, Anupma Nayak, Michael Feldman, Jennifer J.D. Morrissette, Nandita Mitra, Amanda Barrett, Nicole Lunceford, Liza M. Dorfman, Andrew J. Rech, John Pluta, Brandon M. Wenz, Bradley Wubbenhorst, Kara N. Maxwell, and Adam A. Kraya
- Abstract
Supplementary Figures S1-S4, Tables S4, S6.
- Published
- 2023
- Full Text
- View/download PDF
14. Table S2 from Genomic Signatures Predict the Immunogenicity of BRCA-Deficient Breast Cancer
- Author
-
Katherine L. Nathanson, Robert H. Vonderheide, Susan M. Domchek, Anupma Nayak, Michael Feldman, Jennifer J.D. Morrissette, Nandita Mitra, Amanda Barrett, Nicole Lunceford, Liza M. Dorfman, Andrew J. Rech, John Pluta, Brandon M. Wenz, Bradley Wubbenhorst, Kara N. Maxwell, and Adam A. Kraya
- Abstract
Characteristics of breast cancers ascertained from Penn Medicine hospitals
- Published
- 2023
- Full Text
- View/download PDF
15. Automatic Clustering and Thickness Measurement of Anatomical Variants of the Human Perirhinal Cortex.
- Author
-
Long Xie, John Pluta, Hongzhi Wang 0002, Sandhitsu R. Das, Lauren Mancuso, Dasha Kliot, Brian B. Avants, Song-Lin Ding, David A. Wolk, and Paul A. Yushkevich
- Published
- 2014
- Full Text
- View/download PDF
16. Reconstruction of the human hippocampus in 3D from histology and high-resolution ex-vivo MRI.
- Author
-
Daniel H. Adler, Alex Yang Liu, John Pluta, Salmon Kadivar, Sylvia Orozco, Hongzhi Wang 0002, James C. Gee, Brian B. Avants, and Paul A. Yushkevich
- Published
- 2012
- Full Text
- View/download PDF
17. From label fusion to correspondence fusion: A new approach to unbiased groupwise registration.
- Author
-
Paul A. Yushkevich, Hongzhi Wang 0002, John Pluta, and Brian B. Avants
- Published
- 2012
- Full Text
- View/download PDF
18. Hippocampus segmentation using a stable maximum likelihood classifier ensemble algorithm.
- Author
-
Hongzhi Wang 0002, Jung Wook Suh, Sandhitsu R. Das, Murat Altinay, John Pluta, and Paul A. Yushkevich
- Published
- 2011
- Full Text
- View/download PDF
19. Regression-based label fusion for multi-atlas segmentation.
- Author
-
Hongzhi Wang 0002, Jung Wook Suh, Sandhitsu R. Das, John Pluta, Murat Altinay, and Paul A. Yushkevich
- Published
- 2011
- Full Text
- View/download PDF
20. Optimal Weights for Multi-atlas Label Fusion.
- Author
-
Hongzhi Wang 0002, Jung Wook Suh, John Pluta, Murat Altinay, and Paul A. Yushkevich
- Published
- 2011
- Full Text
- View/download PDF
21. Shape-based semi-automatic hippocampal subfield segmentation with learning-based bias removal.
- Author
-
Hongzhi Wang 0002, Sandhitsu R. Das, John Pluta, Caryne Craige, Murat Altinay, Michael Weiner 0001, Susanne G. Mueller, and Paul A. Yushkevich
- Published
- 2010
- Full Text
- View/download PDF
22. Standing on the Shoulders of Giants: Improving Medical Image Segmentation via Bias Correction.
- Author
-
Hongzhi Wang 0002, Sandhitsu R. Das, John Pluta, Caryne Craige, Murat Altinay, Brian B. Avants, Michael Weiner 0001, Susanne G. Mueller, and Paul A. Yushkevich
- Published
- 2010
- Full Text
- View/download PDF
23. Building an atlas of hippocampal subfields using postmortem MRI.
- Author
-
Paul A. Yushkevich, Brian B. Avants, John Pluta, David Minkoff, Stephen Pickup, Weixia Liu, John A. Detre, Murray Grossman, and James C. Gee
- Published
- 2008
- Full Text
- View/download PDF
24. Shape-Based Alignment of Hippocampal Subfields: Evaluation in Postmortem MRI.
- Author
-
Paul A. Yushkevich, Brian B. Avants, John Pluta, David Minkoff, John A. Detre, Murray Grossman, and James C. Gee
- Published
- 2008
- Full Text
- View/download PDF
25. HRDex: a tool for deriving homologous recombination deficiency (HRD) scores from whole exome sequencing data
- Author
-
John Pluta, Ryan Hausler, Brad Wubbenhorst, Heena Desai, Susan M. Domchek, Katherine L. Nathanson, and Kara N. Maxwell
- Abstract
BackgroundBreast and ovarian tumors in patients with biallelic BRCA1 and BRCA2 mutations either by germline mutations accompanied by allele-specific loss of heterozygosity (LOH) or truncal somatic mutations respond to PARP inhibition. The repair of double stranded DNA breaks in tumors these tumors leads to homologous recombination deficiency (HRD), which can be measured using a variety of genomic and transcriptomic signatures. However, the optimal biomarker for BRCA deficiency is unknown.MethodsWe developed HRDex to determine HRD and its composite scores from allele specific copy number data analysis of whole exome sequencing (WES) data and examined the discriminatory ability of HRDex and other genomic and transcriptomic measures to identify BRCA deficiency in breast and ovarian tumors from The Cancer Genome Atlas (TCGA).ResultsHRDex scores have high correlation with SNP array based HRD scores in both breast and ovarian cancers. HRDex scores have high discriminatory accuracy to distinguish BRCA deficient breast tumors, similar to SNP array based scores (AUC 0.87 vs 0.90); however, discriminatory ability for ovarian tumors was lower (AUC 0.79 vs 0.90). HRD-LST had the best discriminatory ability of the three composite HRD scores. HRDex had higher discriminatory ability for identification of BRCA deficiency than RNA expression based scores (eCARD, tp53, RPS and PARPi7) in breast and ovarian tumors. Tumor mutational burden (TMB) was associated with BRCA deficiency in breast but not ovarian cancer. Combining HRDex score with mutational signature 3 modestly increased discriminatory ability for BRCA deficient breast and ovarian tumors (breast: AUC 0.90 vs 0.87; ovarian: AUC 0.83 vs 0.79).ConclusionsWES based HRD scores perform similarly to SNP array HRD scores, and better than other genomic or transcriptomic signatures, for identification of tumors with BRCA deficiency due to biallelic BRCA loss.
- Published
- 2022
- Full Text
- View/download PDF
26. Histology-derived volumetric annotation of the human hippocampal subfields in postmortem MRI.
- Author
-
Daniel H. Adler, John Pluta, Salmon Kadivar, Caryne Craige, James C. Gee, Brian B. Avants, and Paul A. Yushkevich
- Published
- 2014
- Full Text
- View/download PDF
27. Multi-Atlas Segmentation with Joint Label Fusion.
- Author
-
Hongzhi Wang 0002, Jung Wook Suh, Sandhitsu R. Das, John Pluta, Caryne Craige, and Paul A. Yushkevich
- Published
- 2013
- Full Text
- View/download PDF
28. Measuring longitudinal change in the hippocampal formation from in vivo high-resolution T2-weighted MRI.
- Author
-
Sandhitsu R. Das, Brian B. Avants, John Pluta, Hongzhi Wang 0002, Jung Wook Suh, Michael W. Weiner, Susanne G. Mueller, and Paul A. Yushkevich
- Published
- 2012
- Full Text
- View/download PDF
29. Improving fMRI activation detection sensitivity using intervoxel coherence mapping.
- Author
-
Ze Wang 0017, Zhengjun Li, John Pluta, and John A. Detre
- Published
- 2012
- Full Text
- View/download PDF
30. A learning-based wrapper method to correct systematic errors in automatic image segmentation: Consistently improved performance in hippocampus, cortex and brain segmentation.
- Author
-
Hongzhi Wang 0002, Sandhitsu R. Das, Jung Wook Suh, Murat Altinay, John Pluta, Caryne Craige, Brian B. Avants, and Paul A. Yushkevich
- Published
- 2011
- Full Text
- View/download PDF
31. Heterogeneity of functional activation during memory encoding across hippocampal subfields in temporal lobe epilepsy.
- Author
-
Sandhitsu R. Das, Dawn Mechanic-Hamilton, John Pluta, Marc Korczykowski, John A. Detre, and Paul A. Yushkevich
- Published
- 2011
- Full Text
- View/download PDF
32. Bias in estimation of hippocampal atrophy using deformation-based morphometry arises from asymmetric global normalization: An illustration in ADNI 3 T MRI data.
- Author
-
Paul A. Yushkevich, Brian B. Avants, Sandhitsu R. Das, John Pluta, Murat Altinay, and Caryne Craige
- Published
- 2010
- Full Text
- View/download PDF
33. The optimal template effect in hippocampus studies of diseased populations.
- Author
-
Brian B. Avants, Paul A. Yushkevich, John Pluta, David Minkoff, Marc Korczykowski, John A. Detre, and James C. Gee
- Published
- 2010
- Full Text
- View/download PDF
34. Nearly automatic segmentation of hippocampal subfields in in vivo focal T2-weighted MRI.
- Author
-
Paul A. Yushkevich, Hongzhi Wang 0002, John Pluta, Sandhitsu R. Das, Caryne Craige, Brian B. Avants, Michael W. Weiner, and Susanne G. Mueller
- Published
- 2010
- Full Text
- View/download PDF
35. Function lateralization via measuring coherence laterality.
- Author
-
Ze Wang 0017, Dawn Mechanic-Hamilton, John Pluta, Simon Glynn, and John A. Detre
- Published
- 2009
- Full Text
- View/download PDF
36. A high-resolution computational atlas of the human hippocampus from postmortem magnetic resonance imaging at 9.4 T.
- Author
-
Paul A. Yushkevich, Brian B. Avants, John Pluta, Sandhitsu R. Das, David Minkoff, Dawn Mechanic-Hamilton, Simon Glynn, Stephen Pickup, Weixia Liu, James C. Gee, Murray Grossman, and John A. Detre
- Published
- 2009
- Full Text
- View/download PDF
37. Abstract 4648: How inherited mutations affect single cells within the tumor microenvironment in breast tumors stratified by receptor status
- Author
-
Dana Pueschl, Derek A. Oldridge, Jonathan Belman, William Chandler, Anupma Nayak, Bradley Wubbenhorst, John Pluta, Michael Feldman, E. John Wherry, Heather Thorne, Georgia Chenevix-Trench, Kathleen Cuningham Foundation Consortium for Resea kConFab, Susan M. Domchek, and Katherine L. Nathanson
- Subjects
Cancer Research ,Oncology - Abstract
The tumor microenvironment (TME) plays important roles in tumor progression, therapy response and patient survival. In this comprehensive study, we performed a detailed multiomic single cell analysis of breast cancers with inherited mutations in different genes (BRCA1, BRCA2, PALB2, ATM, CHEK2), and wild type mutation negative (WT) patients, stratified by hormone receptor status, to understand the relationship between inherited mutation and immunogenicity. Using spatial tissue multiplexing (PhenoCycler), single cell RNAseq (10X Genomics) and bulk RNAseq, we evaluated cellular markers within the TME associated with DNA damage as well as phenotypes with tumor suppressive (M1, CD8 T cells, NK cells) or tumor promotive (M2, Treg, CAF) properties. We developed a 43-plex antibody panel and characterized eight tissue microarrays containing 443 patient samples from mutation carriers (BRCA1: n=142, BRCA2: n=118, CHEK2: n=2, TP53: n=3, PALB2: n=7) and WT tumors (n=171). We addressed how the TME differs in mutation carriers versus WT tumors stratified by receptor status. To further investigate immune cell function (cytolytic function, exhaustion, immune checkpoint) as well as pathways involved in immune suppression and/or promotion, we performed scRNA sequencing using isolated single cells from fresh collected BC samples (WT ER: n=3, BRCA2 ER: n=1, CHEK2 ER: n=1, VUS ER: n=1) and bulk RNA (BRCA1: n=17, BRCA2: n=9). To determine immune cell (IC) frequency, we analyzed CD45+ cells detectable in each cohort and their co-expression of markers to determine specific phenotypes. Our data suggest that within BRCA mutation-associated BC, there are two groups: 1) with increased tumor infiltrating lymphocytes (TILs) including CD8+ cytotoxic T cells with high cytolytic function; and 2) with decreased TILs and cytolytic function. The frequency of CD8+ T cells is significantly decreased in breast cancers with BRCA2 mutations compared to BRCA1. Characterizing ER+ CHEK2 BC reveals T cell exhaustion compared to WT ER+ BC. Furthermore, preliminary scRNA results reveal increased fibroblast markers in ER+ CHEK2 and ER+ WT compared to ER+ BRCA2 tumors. Interestingly, cancer associated fibroblast (CAFs) markers (ACTA2, COL1A1, FAP, PDGFRA, PDGFRB, PDPN, THY1) as well as TAMs (CCL2, CD163, CD206, CD68, IL10, LOX, PLOD2, SIGLEC1) are increased in ER+ CHEK2 tumors compared to ER+ WT. These data could provide insights into how the TME associated with ER+ CHEK2 mutations might be driven by CAFs and TAMs with a frequency much higher than in ER+ WT. The detection of immune suppressive (Treg, M2) and immune promotive (M1) phenotypes differs in BRCA1 compared to BRCA2 mutated and WT TNBC. Our findings could decipher the role of DNA repair gene mutations and their effect on cells within the TME that might influence the design of treatment options in patients with inherited mutations based on their receptor status. Citation Format: Dana Pueschl, Derek A. Oldridge, Jonathan Belman, William Chandler, Anupma Nayak, Bradley Wubbenhorst, John Pluta, Michael Feldman, E. John Wherry, Heather Thorne, Georgia Chenevix-Trench, Kathleen Cuningham Foundation Consortium for Resea kConFab, Susan M. Domchek, Katherine L. Nathanson. How inherited mutations affect single cells within the tumor microenvironment in breast tumors stratified by receptor status. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4648.
- Published
- 2023
- Full Text
- View/download PDF
38. Neural correlates of voluntary and involuntary risk taking in the human brain: An fMRI Study of the Balloon Analog Risk Task (BART).
- Author
-
Hengyi Rao, Marc Korczykowski, John Pluta, Angela Hoang, and John A. Detre
- Published
- 2008
- Full Text
- View/download PDF
39. Structural consequences of diffuse traumatic brain injury: A large deformation tensor-based morphometry study.
- Author
-
Junghoon Kim 0006, Brian B. Avants, Sunil Patel, John Whyte, H. Branch Coslett, John Pluta, John A. Detre, and James C. Gee
- Published
- 2008
- Full Text
- View/download PDF
40. PTEN Loss and BRCA1 Promoter Hypermethylation Negatively Predict for Immunogenicity in BRCA-Deficient Ovarian Cancer
- Author
-
Adam A. Kraya, Kara N. Maxwell, Monika A. Eiva, Bradley Wubbenhorst, John Pluta, Michael Feldman, Anupma Nayak, Daniel J. Powell, Susan M. Domchek, Robert H. Vonderheide, and Katherine L. Nathanson
- Subjects
Cancer Research ,Oncology ,ORIGINAL REPORTS - Abstract
PURPOSE Ovarian cancers can exhibit a prominent immune infiltrate, but clinical trials have not demonstrated substantive response rates to immune checkpoint blockade monotherapy. We aimed to understand genomic features associated with immunogenicity in BRCA1/2 mutation–associated cancers. MATERIALS AND METHODS Using the Cancer Genome Atlas whole-exome sequencing, methylation, and expression data, we analyzed 66 ovarian cancers with either germline or somatic loss of BRCA1/2 and whole-exome sequencing, immunohistochemistry, and CyTOF in 20 ovarian cancers with germline BRCA1/2 pathogenic variants from Penn. RESULTS We found two groups of BRCA1/2 ovarian cancers differing in their immunogenicity: (1) 37 tumors significantly enriched for PTEN loss (11, 30%) and BRCA1 promoter–hypermethylated (10, 27%; P = .0016) and (2) PTEN wild-type (28 of 29 tumors) cancers, with the latter group having longer overall survival (OS; P = .0186, median OS not reached v median OS = 66.1 months). BRCA1/2-mutant PTEN loss and BRCA1 promoter–hypermethylated cancers were characterized by the decreased composition of lymphocytes estimated by gene expression ( P = .0030), cytolytic index ( P = .034), and cytokine expression but higher homologous recombination deficiency scores ( P = .00013). Large-scale state transitions were the primary discriminating feature ( P = .001); neither mutational burden nor neoantigen burden could explain differences in immunogenicity. In Penn tumors, PTEN loss and high homologous recombination deficiency cancers exhibited fewer CD3+ ( P = .05), CD8+ ( P = .012), and FOXP3+ ( P = .0087) T cells; decreased PRF1 expression ( P = .041); and lower immune costimulatory and inhibitory molecule expression. CONCLUSION Our study suggests that within ovarian cancers with genetic loss of BRCA1/2 are two subsets exhibiting differential immunogenicity, with lower levels associated with PTEN loss and BRCA hypermethylation. These genomic features of BRCA1/2-associated ovarian cancers may inform considerations around how to optimally deploy immune checkpoint inhibitors in the clinic.
- Published
- 2022
41. Genomic Signatures Predict the Immunogenicity of BRCA-Deficient Breast Cancer
- Author
-
Adam A. Kraya, Nandita Mitra, Jennifer J.D. Morrissette, Amanda Barrett, Anupma Nayak, Nicole Lunceford, Michael Feldman, Kara N. Maxwell, Susan M. Domchek, Robert H. Vonderheide, Katherine L. Nathanson, John Pluta, Bradley Wubbenhorst, Brandon Wenz, Liza Dorfman, and Andrew J. Rech
- Subjects
0301 basic medicine ,Cancer Research ,Somatic cell ,Breast Neoplasms ,CD8-Positive T-Lymphocytes ,Biology ,B7-H1 Antigen ,Article ,Germline ,Loss of heterozygosity ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Breast cancer ,Immune system ,Biomarkers, Tumor ,medicine ,Humans ,Homologous Recombination ,skin and connective tissue diseases ,BRCA2 Protein ,BRCA1 Protein ,Immunogenicity ,Genomics ,medicine.disease ,Immune checkpoint ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,CD8 - Abstract
Purpose: Breast cancers with BRCA1/2 alterations have a relatively high mutational load, suggesting that immune checkpoint blockade may be a potential treatment option. However, the degree of immune cell infiltration varies widely, and molecular features contributing to this variability remain unknown. Experimental Design: We hypothesized that genomic signatures might predict immunogenicity in BRCA1/2 breast cancers. Using The Cancer Genome Atlas (TCGA) genomic data, we compared breast cancers with (89) and without (770) either germline or somatic BRCA1/2 alterations. We also studied 35 breast cancers with germline BRCA1/2 mutations from Penn using WES and IHC. Results: We found that homologous recombination deficiency (HRD) scores were negatively associated with expression-based immune indices [cytolytic index (P = 0.04), immune ESTIMATE (P = 0.002), type II IFN signaling (P = 0.002)] despite being associated with a higher mutational/neoantigen burden, in BRCA1/2 mutant breast cancers. Further, absence of allele-specific loss of heterozygosity (LOH negative; P = 0.01) or subclonality (P = 0.003) of germline and somatic BRCA1/2 mutations, respectively, predicted for heightened cytolytic activity. Gene set analysis found that multiple innate and adaptive immune pathways that converge on NF-κB may contribute to this heightened immunogenicity. IHC of Penn breast cancers demonstrated increased CD45+ (P = 0.039) and CD8+ infiltrates (P = 0.037) and increased PDL1 expression (P = 0.012) in HRD-low or LOH-negative cancers. Triple-negative cancers with low HRD had far greater CD8+ T cells (P = 0.0011) and Perforin 1 expression (P = 0.014) compared with hormone receptor-positive HRD-high cancers. Conclusions: HRD scores and hormone receptor subtype are predictive of immunogenicity in BRCA1/2 breast cancers and may inform the design of optimal immune therapeutic strategies.
- Published
- 2019
- Full Text
- View/download PDF
42. Analysis of matched primary and recurrent BRCA1/2 mutation-associated tumors identifies recurrence-specific drivers
- Author
-
Violeta Serra, Anna Hubert, Catherine Ruan, Judith Balmaña, Alba Llop-Guevara, Kara N. Maxwell, Susan M. Domchek, Anupma Nayak, Wenting Zhou, John Pluta, Dana Pueschl, Michael Feldman, Katherine L. Nathanson, Adam A. Kraya, Jennifer Shah, Bradley Wubbenhorst, Kurt D.Andrea, and Jake S. Shilan
- Subjects
Oncology ,medicine.medical_specialty ,Brca1 2 mutation ,Primary (chemistry) ,endocrine system diseases ,business.industry ,Internal medicine ,medicine ,skin and connective tissue diseases ,business ,female genital diseases and pregnancy complications - Abstract
Recurrence is a major cause of death among BRCA1/2 mutation carriers with breast (BrCa) and ovarian cancers (OvCa). We performed multi-omic sequencing on 67 paired primary and recurrent BrCa and OvCa from 27 BRCA1/2 mutation carriers to identify potential recurrence-specific drivers. PARP1 amplifications were identified in recurrences (FDR q = 0.05), and PARP1 was significantly overexpressed across primary BrCa and recurrent BrCa and OvCa, independent of amplification status. RNA-seq analysis found two BRCA2 isoforms, BRCA2-201/Long and BRCA2-001/Short, predicted sensitive and insensitive to nonsense-mediated decay, respectively. BRCA2-001/Short was expressed more frequently in recurrences and associated with reduced overall survival in breast cancer (87 vs. 121 months; HR = 2.5 [1.18–5.5]). Loss of heterozygosity (LOH) status was discordant in 25% of patient’s primary and recurrent tumors, with switching between both LOH and lack of LOH found. Our study revealed multiple potential drivers of recurrent disease in BRCA1/2 mutation-associated cancer, improving our understanding of tumor evolution and suggesting potential biomarkers.
- Published
- 2021
- Full Text
- View/download PDF
43. Genetically inferred telomere length and testicular germ cell tumor risk
- Author
-
Qing Lan, Nathaniel Rothman, Katherine L. Nathanson, Chey Loveday, Kristian Almstrup, Tom Grotmol, Derek Brown, Fredrik Wiklund, Marlene Danner Dalgaard, John Pluta, Mark H. Greene, Katherine A. McGlynn, Mitchell J. Machiela, Stephen M. Schwartz, Clare Turnbull, and Peter A. Kanetsky
- Subjects
0301 basic medicine ,Oncology ,Male ,medicine.medical_specialty ,Epidemiology ,Testicular Germ Cell Tumor ,Biology ,Risk Assessment ,Article ,03 medical and health sciences ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Testicular Neoplasms ,Risk Factors ,Internal medicine ,Mendelian randomization ,Genotype ,medicine ,Humans ,Genetic Predisposition to Disease ,Testicular cancer ,Telomere Homeostasis ,Mendelian Randomization Analysis ,Neoplasms, Germ Cell and Embryonal ,Telomere ,medicine.disease ,Peripheral blood ,Confidence interval ,030104 developmental biology ,030220 oncology & carcinogenesis ,Case-Control Studies ,Biomarker (medicine) - Abstract
Background: Studies evaluating the association between peripheral blood leukocyte telomere length (LTL) and testicular germ cell tumor (TGCT) risk have produced conflicting results. Methods: Using available genotype data from the Testicular Cancer Consortium (TECAC), polygenic risk score and Mendelian randomization analyses of genetic variants previously associated with LTL were used to assess potential etiologic associations between telomere length and TGCT risk. Results: Genetically inferred telomere length was not associated with TGCT risk among 2,049 cases and 6,921 controls with individual-level genotype data (OR, 1.02; 95% confidence interval, 0.97–1.07). Mendelian randomization analyses using summary statistic data further indicated no evidence for an association between telomere length and TGCT risk among all available TECAC participants (3,558 cases and 13,971 controls). Conclusions: Our analyses in the largest molecular genetic testicular cancer study to date provide no evidence for an association between genetically inferred peripheral blood LTL and TGCT risk. Impact: The lack of evidence for an overall association indicates that peripheral blood LTL is likely not a strong biomarker for TGCT risk.
- Published
- 2021
- Full Text
- View/download PDF
44. Identification of 22 susceptibility loci associated with testicular germ cell tumors
- Author
-
Mingyao Li, Chey Loveday, Tongzhang Zheng, Chu Chen, Paloma Martin-Gimeno, Mark H. Greene, Javier Benitez, Stephen J. Chanock, Coby Meijer, Daphne Yang, Christopher D. Brown, Thorunn Rafnar, Lorenzo Richiardi, Marija Gamulin, Kari Stefansson, Michelle A.T. Hildebrandt, Matthew E. Johnson, Shweta Ramdas, Jourik A. Gietema, David J. Vaughn, Kevin T. Nead, Robert Karlsson, Struan F.A. Grant, Peter A. Kanetsky, Kristian Almstrup, David I. Quinn, Katherine L. Nathanson, Kurt D'Andrea, D. Timothy Bishop, Andrew D. Wells, Nandita Mitra, Katherine A. McGlynn, Jennifer T. Loud, Carlo Foresta, Lynn Anson-Cartwright, Jérémie Nsengimana, Alberto Ferlin, Lambertus A. Kiemeney, Rolf Inge Skotheim, Victoria K. Cortessis, Xifeng Wu, Chiara Grasso, Ragnhild A. Lothe, Tom Grotmol, Russ Hauser, Trine B. Haugen, Fredrik Wiklund, Clare Turnbull, Benita Weathers, Louise C. Pyle, Robert J. Hamilton, Stephen M. Schwartz, Davor Lessel, John Pluta, Ewa Rajpert-De Meyts, Rona Wilf, Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Damage and Repair in Cancer Development and Cancer Treatment (DARE)
- Subjects
Oncology ,Male ,Linkage disequilibrium ,Somatic cell ,General Physics and Astronomy ,Genome-wide association study ,Disease ,VARIANTS ,DMRT1 ,Linkage Disequilibrium ,Neoplasms, Germ Cell and Embryonal / metabolism ,Cell Line, Tumor ,Chromosome Mapping ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Genome-Wide Association Study ,Genotype ,Humans ,Meta-Analysis as Topic ,Neoplasms, Germ Cell and Embryonal ,Protein Interaction Maps ,Testicular Neoplasms ,Polymorphism, Single Nucleotide ,Testicular Neoplasms / genetics ,0302 clinical medicine ,Gene Regulatory Networks / genetics ,Neoplasms ,Medicine ,FAMILIAL RISK ,Cancer genetics ,Genome-Wide Association Study / methods ,0303 health sciences ,Tumor ,Multidisciplinary ,testicular germ cell tumors (TGCT) ,Protein Interaction Maps / genetics ,Single Nucleotide ,CANCER ,medicine.anatomical_structure ,SINGLE ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,030220 oncology & carcinogenesis ,CENP-I ,Germ cell ,endocrine system ,medicine.medical_specialty ,Science ,GENOTYPE IMPUTATION ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,Testicular cancer ,Internal medicine ,Neoplasms, Germ Cell and Embryonal / genetics ,Polymorphism ,GENOME-WIDE ASSOCIATION ,METAANALYSIS ,030304 developmental biology ,Genetic association study ,business.industry ,Cancer ,General Chemistry ,medicine.disease ,Genetic Predisposition to Disease / genetics ,BAX ,Germ Cell and Embryonal ,Testicular Neoplasms / metabolism ,business - Abstract
Testicular germ cell tumors (TGCT) are the most common tumor in young white men and have a high heritability. In this study, the international Testicular Cancer Consortium assemble 10,156 and 179,683 men with and without TGCT, respectively, for a genome-wide association study. This meta-analysis identifies 22 TGCT susceptibility loci, bringing the total to 78, which account for 44% of disease heritability. Men with a polygenic risk score (PRS) in the 95th percentile have a 6.8-fold increased risk of TGCT compared to men with median scores. Among men with independent TGCT risk factors such as cryptorchidism, the PRS may guide screening decisions with the goal of reducing treatment-related complications causing long-term morbidity in survivors. These findings emphasize the interconnected nature of two known pathways that promote TGCT susceptibility: male germ cell development within its somatic niche and regulation of chromosomal division and structure, and implicate an additional biological pathway, mRNA translation., Testicular germ cell tumors are highly heritable, and the authors present the largest genome association study, identifying 22 novel loci, which account for a third of those identified to date. Implicated pathways include male germ cell development and differentiation, and chromosomal segregation.
- Published
- 2020
45. Mapping the structural and functional network architecture of the medial temporal lobe using 7T MRI
- Author
-
John Pluta, Preya Shah, Sandhitsu R. Das, Paul A. Yushkevich, Molly Daffner, Joel M. Stein, Russell T. Shinohara, Mark A. Elliott, Brian Litt, Kathryn A. Davis, David A. Wolk, Elijah Valenciano, John A. Detre, Danielle S. Bassett, and Laura E.M. Wisse
- Subjects
Adult ,Male ,0301 basic medicine ,Rest ,chemical and pharmacologic phenomena ,Biology ,Hippocampal formation ,Article ,Temporal lobe ,Functional networks ,Correlation ,03 medical and health sciences ,0302 clinical medicine ,Neural Pathways ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Brain Mapping ,Radiological and Ultrasound Technology ,Resting state fMRI ,Dentate gyrus ,Subiculum ,Organ Size ,Magnetic Resonance Imaging ,Temporal Lobe ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Neurology ,Female ,Neurology (clinical) ,Anatomy ,Neuroscience ,psychological phenomena and processes ,030217 neurology & neurosurgery ,Parahippocampal gyrus - Abstract
Medial temporal lobe (MTL) subregions play integral roles in memory function and are differentially affected in various neurological and psychiatric disorders. The ability to structurally and functionally characterize these subregions may be important to understanding MTL physiology and diagnosing diseases involving the MTL. In this study, we characterized network architecture of the MTL in healthy subjects (n = 31) using both resting state functional MRI and MTL-focused T2-weighted structural MRI at 7 tesla. Ten MTL subregions per hemisphere, including hippocampal subfields and cortical regions of the parahippocampal gyrus, were segmented for each subject using a multi-atlas algorithm. Both structural covariance matrices from correlations of subregion volumes across subjects, and functional connectivity matrices from correlations between subregion BOLD time series were generated. We found a moderate structural and strong functional inter-hemispheric symmetry. Several bilateral hippocampal subregions (CA1, dentate gyrus, and subiculum) emerged as functional network hubs. We also observed that the structural and functional networks naturally separated into two modules closely corresponding to (a) bilateral hippocampal formations, and (b) bilateral extra-hippocampal structures. Finally, we found a significant correlation in structural and functional connectivity (r = 0.25). Our findings represent a comprehensive analysis of network topology of the MTL at the subregion level. We share our data, methods, and findings as a reference for imaging methods and disease-based research.
- Published
- 2017
- Full Text
- View/download PDF
46. A protocol for manual segmentation of medial temporal lobe subregions in 7 Tesla MRI
- Author
-
Anica Luther, Anne Maass, Paul A. Yushkevich, Emrah Düzel, David Berron, Sandhitsu R. Das, Long Xie, Thomas Wolbers, Laura E.M. Wisse, John Pluta, Paula Vieweg, David A. Wolk, Songlin Ding, and A. Hochkeppler
- Subjects
0301 basic medicine ,Male ,physiology [Hippocampus] ,Hippocampus ,AG, Ambient Gyrus ,lcsh:RC346-429 ,methods [Brain Mapping] ,0302 clinical medicine ,methods [Magnetic Resonance Imaging] ,Cortex (anatomy) ,physiology [Dentate Gyrus] ,Perirhinal cortex ,diagnostic imaging [Dentate Gyrus] ,CSa, anterior ,diagnostic imaging [Hippocampus] ,Brain Mapping ,medicine.diagnostic_test ,HB, Hippocampal Body ,Subiculum ,OTS, Occipito-temporal Sulcus ,Regular Article ,Magnetic Resonance Imaging ,Temporal Lobe ,CSp, posterior ,medicine.anatomical_structure ,standards [Brain Mapping] ,Neurology ,PhG, Parahippocampal Gyrus ,lcsh:R858-859.7 ,Female ,physiology [Temporal Lobe] ,Psychology ,Adult ,DG, Dentate Gyrus ,PrC, Perirhinal Cortex ,Cognitive Neuroscience ,CA3, Cornu Ammonis 3 ,lcsh:Computer applications to medicine. Medical informatics ,Temporal lobe ,03 medical and health sciences ,Young Adult ,Neuroimaging ,CA1, Cornu Ammonis 1 ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,HH, Hippocampal Head ,ddc:610 ,CA2, Cornu Ammonis 2 ,lcsh:Neurology. Diseases of the nervous system ,SaS, Semiannular Sulcus ,standards [Magnetic Resonance Imaging] ,PhC, Parahippocampal Cortex ,HT, Hippocampal Tail ,Magnetic resonance imaging ,MTL, Medial Temporal Lobe ,Entorhinal cortex ,ErC, Entorhinal Cortex ,FG, Fusiform Gyrus ,030104 developmental biology ,diagnostic imaging [Temporal Lobe] ,Sub, Subiculum ,SRLM, Stratum radiatum lacunosum-moleculare ,Dentate Gyrus ,Neurology (clinical) ,CaS, Calcarine sulcus ,CS, Collateral Sulcus ,Neuroscience ,030217 neurology & neurosurgery ,CSF, Cerebrospinal Fluid - Abstract
Recent advances in MRI and increasing knowledge on the characterization and anatomical variability of medial temporal lobe (MTL) anatomy have paved the way for more specific subdivisions of the MTL in humans. In addition, recent studies suggest that early changes in many neurodegenerative and neuropsychiatric diseases are better detected in smaller subregions of the MTL rather than with whole structure analyses. Here, we developed a new protocol using 7 Tesla (T) MRI incorporating novel anatomical findings for the manual segmentation of entorhinal cortex (ErC), perirhinal cortex (PrC; divided into area 35 and 36), parahippocampal cortex (PhC), and hippocampus; which includes the subfields subiculum (Sub), CA1, CA2, as well as CA3 and dentate gyrus (DG) which are separated by the endfolial pathway covering most of the long axis of the hippocampus. We provide detailed instructions alongside slice-by-slice segmentations to ease learning for the untrained but also more experienced raters. Twenty-two subjects were scanned (19–32 yrs, mean age = 26 years, 12 females) with a turbo spin echo (TSE) T2-weighted MRI sequence with high-resolution oblique coronal slices oriented orthogonal to the long axis of the hippocampus (in-plane resolution 0.44 × 0.44 mm2) and 1.0 mm slice thickness. The scans were manually delineated by two experienced raters, to assess intra- and inter-rater reliability. The Dice Similarity Index (DSI) was above 0.78 for all regions and the Intraclass Correlation Coefficients (ICC) were between 0.76 to 0.99 both for intra- and inter-rater reliability. In conclusion, this study presents a fine-grained and comprehensive segmentation protocol for MTL structures at 7 T MRI that closely follows recent knowledge from anatomical studies. More specific subdivisions (e.g. area 35 and 36 in PrC, and the separation of DG and CA3) may pave the way for more precise delineations thereby enabling the detection of early volumetric changes in dementia and neuropsychiatric diseases., Highlights • We present a new segmentation protocol for medial temporal lobe subregions at 7 T MRI. • Novel neuroanatomical knowledge from the work of Ding and Van Hoesen is incorporated. • Individual anatomical variability is covered by digitation- and depth-specific rules. • We provide detailed descriptions and slice-by-slice plots to guide the novice rater. • Reliability analyses provide compelling evidence that the rules can be replicated.
- Published
- 2017
- Full Text
- View/download PDF
47. Genomic profiling of human vascular cells identifies TWIST1 as a causal gene for common vascular diseases
- Author
-
Sanjay Sinha, Felipe Serrano, Marie A Guerraty, Thomas Quertermous, Yi-An Ko, Milos Pjanic, Wei Zhao, Scott Norton, Yoseph Barash, Daniel J. Rader, Ulf Hedin, John Pluta, Robert C. Wirka, Christopher D. Brown, Trieu Nguyen, Sylvia T. Nurnberg, Ljubica Perisic, Stephanie Testa, Ting Wang, Susannah Elwyn, H. Shanker Rao, YoSon Park, Nurnberg, Sylvia T. [0000-0002-0869-484X], Guerraty, Marie A. [0000-0002-0766-1253], Wirka, Robert C. [0000-0001-9131-9508], Rao, H. Shanker [0000-0001-6827-1470], Norton, Scott [0000-0002-1366-0628], Pluta, John [0000-0002-8941-6242], Zhao, Wei [0000-0002-8301-9297], Park, YoSon [0000-0002-0465-4744], Nguyen, Trieu [0000-0001-5647-1301], Hedin, Ulf [0000-0001-9212-3945], Barash, Yoseph [0000-0003-3005-5048], Brown, Christopher D. [0000-0002-3785-5008], Quertermous, Thomas [0000-0002-7645-9067], Rader, Daniel J. [0000-0002-9245-9876], Apollo - University of Cambridge Repository, Nurnberg, Sylvia T [0000-0002-0869-484X], Guerraty, Marie A [0000-0002-0766-1253], Wirka, Robert C [0000-0001-9131-9508], Rao, H Shanker [0000-0001-6827-1470], Brown, Christopher D [0000-0002-3785-5008], and Rader, Daniel J [0000-0002-9245-9876]
- Subjects
Cancer Research ,Gene Expression ,Genome-wide association study ,QH426-470 ,Smooth Muscle Cells ,Vascular Medicine ,Coronary artery disease ,0302 clinical medicine ,Animal Cells ,Medicine and Health Sciences ,Coronary Heart Disease ,Genetics (clinical) ,Coronary Arteries ,Cells, Cultured ,0303 health sciences ,Nuclear Proteins ,Genomics ,Arteries ,Coronary Vessels ,medicine.anatomical_structure ,Immunoglobulin J Recombination Signal Sequence-Binding Protein ,Anatomy ,Cellular Types ,Research Article ,Protein Binding ,animal structures ,Myocytes, Smooth Muscle ,Cardiology ,Muscle Tissue ,Locus (genetics) ,Single-nucleotide polymorphism ,Biology ,Quantitative trait locus ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,medicine ,Genome-Wide Association Studies ,Genetics ,Humans ,Vascular Diseases ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Muscle Cells ,RBPJ ,Twist-Related Protein 1 ,Colocalization ,Biology and Life Sciences ,Computational Biology ,Endothelial Cells ,Human Genetics ,Cell Biology ,medicine.disease ,Genome Analysis ,Coronary arteries ,Biological Tissue ,Genetic Loci ,Cancer research ,Cardiovascular Anatomy ,Blood Vessels ,Endothelium, Vascular ,Transcriptome ,030217 neurology & neurosurgery - Abstract
Genome-wide association studies have identified multiple novel genomic loci associated with vascular diseases. Many of these loci are common non-coding variants that affect the expression of disease-relevant genes within coronary vascular cells. To identify such genes on a genome-wide level, we performed deep transcriptomic analysis of genotyped primary human coronary artery smooth muscle cells (HCASMCs) and coronary endothelial cells (HCAECs) from the same subjects, including splicing Quantitative Trait Loci (sQTL), allele-specific expression (ASE), and colocalization analyses. We identified sQTLs for TARS2, YAP1, CFDP1, and STAT6 in HCASMCs and HCAECs, and 233 ASE genes, a subset of which are also GTEx eGenes in arterial tissues. Colocalization of GWAS association signals for coronary artery disease (CAD), migraine, stroke and abdominal aortic aneurysm with GTEx eGenes in aorta, coronary artery and tibial artery discovered novel candidate risk genes for these diseases. At the CAD and stroke locus tagged by rs2107595 we demonstrate colocalization with expression of the proximal gene TWIST1. We show that disrupting the rs2107595 locus alters TWIST1 expression and that the risk allele has increased binding of the NOTCH signaling protein RBPJ. Finally, we provide data that TWIST1 expression influences vascular SMC phenotypes, including proliferation and calcification, as a potential mechanism supporting a role for TWIST1 in CAD., Author summary Genome-wide association studies (GWAS) have identified hundreds of genetic variants that are associated with human vascular disease including coronary artery disease. These are predominantly common single nucleotide polymorphisms (SNPs) in non-coding regions, which makes the identification of the causal genes and their underlying connection to pathophysiology challenging. Mapping of expression quantitative trait loci (eQTLs) has been performed to associate GWAS SNPs with risk genes in vascular cells and tissues. However, atherosclerotic vascular tissues contain multiple cell types. We perform deep transcriptomic profiling of genotyped human-derived vascular cells–endothelial cells and smooth muscle cells–and use splicing quantitative trait locus, allele-specific expression, and colocalization analyses to annotate genetic variants associated with vascular diseases and gain insight into their potential function in a cell-type specific manner. Based on these analyses, we identified computationally and then validate experimentally an association between the CAD risk locus rs2107595 and the gene TWIST1. We propose that the minor allele for this locus can affect transcription factor binding and provide data supporting a role for TWIST1 in modulating smooth muscle cell phenotype.
- Published
- 2020
48. Progress update from the hippocampal subfields group
- Author
-
Trevor A. Steve, Katrin Amunts, Olga Kedo, Rosanna K. Olsen, Elliott G. Johnson, Joshua K. Lee, Martina Bocchetta, Ulman Lindenberger, Craig E.L. Stark, Ricardo Insausti, Daniela J. Palombo, Naftali Raz, Margaret L. Schlichting, Gaël Chételat, Valerie A. Carr, Markus Werkle-Bergner, Nanthia Suthana, Paul A. Yushkevich, Jordan DeKraker, Attila Keresztes, Andrew R. Bender, Noa Ofen, John Pluta, Arnold Bakker, David Berron, Karen M. Rodrigue, Kristen M. Kennedy, Robert S. C. Amaral, Prabesh Kanel, Renaud La Joie, Mirjam I. Geerlings, Susanne G. Mueller, Qijing Yu, Songlin Ding, Jean C. Augustinack, Laura E.M. Wisse, Marina Boccardi, Lei Wang, Lorenzo Pasquini, Yee Lee Shing, Ana M. Daugherty, Elizabeth M. Mulligan, Robin de Flores, Yushan Huang, M. Mallar Chakravarty, Alison C. Burggren, Rotman Research Institute at the Baycrest Centre (RRI), University of Toronto, San Jose State University [San José] (SJSU), Wayne State University [Detroit], Memory and Aging Center [San Francisco, CA, États-Unis], University of California [San Francisco] (UCSF), University of California-University of California, Douglas Mental Health University Institute [Montréal], McGill University = Université McGill [Montréal, Canada], Institute of Neuroscience and Medicine [Jülich] (INM-1), Massachusetts General Hospital [Boston], Johns Hopkins University School of Medicine [Baltimore], Lund University [Lund], German Research Center for Neurodegenerative Diseases - Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Neuroimaging and Telemedicine (LENITEM), IRCCS Fatebenefratelli - Brescia, Dementia Research Centre [London] (DRC), University College of London [London] (UCL), University of Oregon [Eugene], Physiopathologie et imagerie des troubles neurologiques (PhIND), Université de Caen Normandie (UNICAEN), Normandie Université (NU)-Normandie Université (NU)-Institut National de la Santé et de la Recherche Médicale (INSERM), GIP Cyceron (Cyceron), Normandie Université (NU)-Normandie Université (NU)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-CHU Caen, Normandie Université (NU)-Tumorothèque de Caen Basse-Normandie (TCBN)-Tumorothèque de Caen Basse-Normandie (TCBN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Normandie Université (NU), University of Western Ontario (UWO), Allen Institute for Brain Science [Seattle, WA, USA], University Medical Center [Utrecht], University of Alberta, Human Neuroanatomy Laboratory [Albacete, Spain] (School of Medicine - CRIB), University of Castilla-La Mancha (UCLM)-Centro Regional de Investigaciones Biomédicas (CRIB), University of California, University of Michigan Medical School [Ann Arbor], University of Michigan [Ann Arbor], University of Michigan System-University of Michigan System, Heinrich Heine Universität Düsseldorf = Heinrich Heine University [Düsseldorf], University of Texas at Dallas [Richardson] (UT Dallas), Max Planck Institute for Human Development, Max-Planck-Gesellschaft, University of California [Davis] (UC Davis), Freie Universität Berlin, Florida State University [Tallahassee] (FSU), Weizmann Institutes of Science [Rehovot, Israel], University of British Columbia (UBC), University of Pennsylvania [Philadelphia], Goethe-University Frankfurt am Main, University of California [Irvine] (UCI), University of California [Los Angeles] (UCLA), Northwestern University Feinberg School of Medicine, Penn Image Computing & Science Lab [Philadelphia] (PICSL), The work on this article was also supported by NIH grant R01 AG-011230 to Naftali Raz, NIH grant R01 AG-055121 to Lei Wang, NIH grant R01 AG-056014 to Paul Yushkevich, and NIH grant R01 AG-034613 to Craig Stark. M.M.C. receives salary support from the Fonds du Recherches Santes Quebec and also acknowledges support from NSERC, Weston Brain Institute, and CIHR., Hippocampal Subfields Group, San Jose State University [San Jose] (SJSU), University of California [San Francisco] (UC San Francisco), University of California (UC)-University of California (UC), Universidad de Castilla-La Mancha = University of Castilla-La Mancha (UCLM)-Centro Regional de Investigaciones Biomédicas (CRIB), University of California (UC), University of Pennsylvania, University of California [Irvine] (UC Irvine), and CHETELAT, Gaëlle
- Subjects
Histology ,Standardization ,Computer science ,Clinical Neurology ,Structural imaging ,Neuroimaging ,lcsh:Geriatrics ,Hippocampal formation ,Hippocampus ,lcsh:RC346-429 ,Special Section: Working Group Summaries for the European Joint Programme for Neurodegenerative Disease Research (JPND). (Guest Editors: Jorge Jovicich & Giovanni B. Frisoni) ,Temporal lobe ,ddc:616.89 ,03 medical and health sciences ,0302 clinical medicine ,Volumetry ,Segmentation ,[SDV.NEU] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,ddc:610 ,lcsh:Neurology. Diseases of the nervous system ,Reliability (statistics) ,030304 developmental biology ,Protocol (science) ,0303 health sciences ,ex vivo ,International working group ,Data science ,Ex vivo ,3. Good health ,lcsh:RC952-954.6 ,Psychiatry and Mental health ,Neuroanatomy ,Cytoarchitec- ture ,Cytoarchitecture ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,Neurology (clinical) ,030217 neurology & neurosurgery ,Human - Abstract
Introduction Heterogeneity of segmentation protocols for medial temporal lobe regions and hippocampal subfields on in vivo magnetic resonance imaging hinders the ability to integrate findings across studies. We aim to develop a harmonized protocol based on expert consensus and histological evidence. Methods Our international working group, funded by the EU Joint Programme–Neurodegenerative Disease Research (JPND), is working toward the production of a reliable, validated, harmonized protocol for segmentation of medial temporal lobe regions. The working group uses a novel postmortem data set and online consensus procedures to ensure validity and facilitate adoption. Results This progress report describes the initial results and milestones that we have achieved to date, including the development of a draft protocol and results from the initial reliability tests and consensus procedures. Discussion A harmonized protocol will enable the standardization of segmentation methods across laboratories interested in medial temporal lobe research worldwide., Highlights • Harmonization of MRI-based segmentation of medial temporal regions is needed. • The Hippocampal Subfield Group includes >200 imaging and anatomy experts worldwide. • Reliable and valid protocol based on specialized histology data set in development. • A modified Delphi procedure is used to determine consensus on protocol rules. • Final protocol will provide subfield delineation in hippocampal body, head, and tail.
- Published
- 2019
- Full Text
- View/download PDF
49. Characterization of hippocampal subfields using ex vivo MRI and histology data: lessons for in vivo segmentation
- Author
-
Paul A. Yushkevich, Ranjit Ittyerah, David Berron, Murray Grossman, Robin de Flores, Daniel H. Adler, Sandhitsu R. Das, Long Xie, John Pluta, Theresa Schuck, David A. Wolk, Weixia Liu, Stephen Pickup, Songlin Ding, Laura E.M. Wisse, John L. Robinson, and John Q. Trojanowski
- Subjects
Male ,genetic structures ,Databases, Factual ,Cognitive Neuroscience ,Hippocampus ,Biology ,Hippocampal formation ,050105 experimental psychology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Humans ,0501 psychology and cognitive sciences ,Segmentation ,Aged ,Aged, 80 and over ,Orientation (computer vision) ,Dentate gyrus ,05 social sciences ,Subiculum ,Middle Aged ,Uncus ,Magnetic Resonance Imaging ,nervous system ,Coronal plane ,Female ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Hippocampal subfield segmentation on in vivo MRI is of great interest for cognition, aging, and disease research. Extant subfield segmentation protocols have been based on neuroanatomical references, but these references often give limited information on anatomical variability. Moreover, there is generally a mismatch between the orientation of the histological sections and the often anisotropic coronal sections on in vivo MRI. To address these issues, we provide a detailed description of hippocampal anatomy using a postmortem dataset containing nine specimens of subjects with and without dementia, which underwent a 9.4 T MRI and histological processing. Postmortem MRI matched the typical orientation of in vivo images and segmentations were generated in MRI space, based on the registered annotated histological sections. We focus on the following topics: the order of appearance of subfields, the location of subfields relative to macroanatomical features, the location of subfields in the uncus and tail and the composition of the dark band, a hypointense layer visible in T2-weighted MRI. Our main findings are that: (a) there is a consistent order of appearance of subfields in the hippocampal head, (b) the composition of subfields is not consistent in the anterior uncus, but more consistent in the posterior uncus, (c) the dark band consists only of the CA-stratum lacunosum moleculare, not the strata moleculare of the dentate gyrus, (d) the subiculum/CA1 border is located at the middle of the width of the hippocampus in the body in coronal plane, but moves in a medial direction from anterior to posterior, and (e) the variable location and composition of subfields in the hippocampal tail can be brought back to a body-like appearance when reslicing the MRI scan following the curvature of the tail. Our findings and this publicly available dataset will hopefully improve anatomical accuracy of future hippocampal subfield segmentation protocols.
- Published
- 2019
50. Automated segmentation of medial temporal lobe subregions on in vivo T1-weighted MRI in early stages of Alzheimer's disease
- Author
-
José V. Manjón, Robin de Flores, Songlin Ding, Hongzhi Wang, John Pluta, David A. Wolk, Virgine Piskin, Laura E.M. Wisse, Paul A. Yushkevich, Sandhitsu R. Das, and Long Xie
- Subjects
Male ,Anterior and posterior hippocampus ,Dura mater ,Hippocampus ,Hippocampal formation ,0302 clinical medicine ,Perirhinal cortex ,Segmentation ,Cortex (anatomy) ,Aged, 80 and over ,Radiological and Ultrasound Technology ,medicine.diagnostic_test ,05 social sciences ,T1-weighted magnetic resonance imaging ,Anatomy ,Alzheimer's disease ,Middle Aged ,Mental Status and Dementia Tests ,Magnetic Resonance Imaging ,Temporal Lobe ,3. Good health ,medicine.anatomical_structure ,Neurology ,Female ,050105 experimental psychology ,Article ,Temporal lobe ,03 medical and health sciences ,Alzheimer Disease ,medicine ,Humans ,0501 psychology and cognitive sciences ,Radiology, Nuclear Medicine and imaging ,Cognitive Dysfunction ,Transentorhinal cortex ,Entorhinal cortex ,Aged ,business.industry ,Mild cognitive impairment ,Magnetic resonance imaging ,Biomarker ,nervous system ,FISICA APLICADA ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
[EN] Medial temporal lobe (MTL) substructures are the earliest regions affected by neurofibrillary tangle pathology-and thus are promising biomarkers for Alzheimer's disease (AD). However, automatic segmentation of the MTL using only T1-weighted (T1w) magnetic resonance imaging (MRI) is challenging due to the large anatomical variability of the MTL cortex and the confound of the dura mater, which is commonly segmented as gray matter by state-of-the-art algorithms because they have similar intensity in T1w MRI. To address these challenges, we developed a novel atlas set, consisting of 15 cognitively normal older adults and 14 patients with mild cognitive impairment with a label explicitly assigned to the dura, that can be used by the multiatlas automated pipeline (Automatic Segmentation of Hippocampal Subfields [ASHS-T1]) for the segmentation of MTL subregions, including anterior/posterior hippocampus, entorhinal cortex (ERC), Brodmann areas (BA) 35 and 36, and parahippocampal cortex on T1w MRI. Cross-validation experiments indicated good segmentation accuracy of ASHS-T1 and that the dura can be reliably separated from the cortex (6.5% mislabeled as gray matter). Conversely, FreeSurfer segmented majority of the dura mater (62.4%) as gray matter and the degree of dura mislabeling decreased with increasing disease severity. To evaluate its clinical utility, we applied the pipeline to T1w images of 663 ADNI subjects and significant volume/thickness loss is observed in BA35, ERC, and posterior hippocampus in early prodromal AD and all subregions at later stages. As such, the publicly available new atlas and ASHS-T1 could have important utility in the early diagnosis and monitoring of AD and enhancing brain-behavior studies of these regions., Northern California Institute for Research and Education; Foundation for the National Institutes of Health; Canadian Institutes of Health Research; Transition Therapeutics; Takeda Pharmaceutical Company; Servier; Piramal Imaging; Pfizer Inc.; Novartis Pharmaceuticals Corporation; Neurotrack Technologies; NeuroRx Research; Meso Scale Diagnostics, LLC.; Lundbeck and Merck & Co., Inc.; Lumosity; Johnson & Johnson Pharmaceutical Research & Development LLC.; Janssen Alzheimer Immunotherapy Research & Development, LLC.; IXICO Ltd.; GE Healthcare; Fujirebio; Genentech, Inc.; F. Hoffmann-La Roche Ltd.; EuroImmun; Eli Lilly and Company; Elan Pharmaceuticals, Inc.; Cogstate and Eisai Inc.; CereSpir, Inc.; Bristol-Myers Squibb Company; Biogen; BioClinica, Inc.; Araclon Biotech; Alzheimer's Drug Discovery Foundation; Alzheimer's Association; AbbVie; National Institute of Biomedical Imaging and Bioengineering; National Institute on Aging; Department of Defense ADNI, Grant/Award Number: W81XWH-12-2-0012; Alzheimer's Disease Neuroimaging Initiative, Grant/Award Number: U01 AG024904; Spain Ministry of Economy, Industry and Competitiveness, Grant/Award Number: DPI2017-87743-R; Foundation Philippe Chatrier; BrightFocus Foundation; National Institutes of Health, Grant/Award Numbers: R01-AG055005, R01-EB017255, P30-AG010124, R01-AG040271, R01-AG056014
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.