1
|
Choi JH, Lee J, Kang U, Chang H, Cho KH. Network dynamics-based subtyping of Alzheimer's disease with microglial genetic risk factors. Alzheimers Res Ther 2024; 16:229. [PMID: 39415193 PMCID: PMC11481771 DOI: 10.1186/s13195-024-01583-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 09/29/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND The potential of microglia as a target for Alzheimer's disease (AD) treatment is promising, yet the clinical and pathological diversity within microglia, driven by genetic factors, poses a significant challenge. Subtyping AD is imperative to enable precise and effective treatment strategies. However, existing subtyping methods fail to comprehensively address the intricate complexities of AD pathogenesis, particularly concerning genetic risk factors. To address this gap, we have employed systems biology approaches for AD subtyping and identified potential therapeutic targets. METHODS We constructed patient-specific microglial molecular regulatory network models by utilizing existing literature and single-cell RNA sequencing data. The combination of large-scale computer simulations and dynamic network analysis enabled us to subtype AD patients according to their distinct molecular regulatory mechanisms. For each identified subtype, we suggested optimal targets for effective AD treatment. RESULTS To investigate heterogeneity in AD and identify potential therapeutic targets, we constructed a microglia molecular regulatory network model. The network model incorporated 20 known risk factors and crucial signaling pathways associated with microglial functionality, such as inflammation, anti-inflammation, phagocytosis, and autophagy. Probabilistic simulations with patient-specific genomic data and subsequent dynamics analysis revealed nine distinct AD subtypes characterized by core feedback mechanisms involving SPI1, CASS4, and MEF2C. Moreover, we identified PICALM, MEF2C, and LAT2 as common therapeutic targets among several subtypes. Furthermore, we clarified the reasons for the previous contradictory experimental results that suggested both the activation and inhibition of AKT or INPP5D could activate AD through dynamic analysis. This highlights the multifaceted nature of microglial network regulation. CONCLUSIONS These results offer a means to classify AD patients by their genetic risk factors, clarify inconsistent experimental findings, and advance the development of treatments tailored to individual genotypes for AD.
Collapse
Affiliation(s)
- Jae Hyuk Choi
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jonghoon Lee
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Uiryong Kang
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hongjun Chang
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Kwang-Hyun Cho
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
2
|
Sasner M, Preuss C, Pandey RS, Uyar A, Garceau D, Kotredes KP, Williams H, Oblak AL, Lin PB, Perkins B, Soni D, Ingraham C, Lee‐Gosselin A, Lamb BT, Howell GR, Carter GW. In vivo validation of late-onset Alzheimer's disease genetic risk factors. Alzheimers Dement 2024; 20:4970-4984. [PMID: 38687251 PMCID: PMC11247676 DOI: 10.1002/alz.13840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Genome-wide association studies have identified over 70 genetic loci associated with late-onset Alzheimer's disease (LOAD), but few candidate polymorphisms have been functionally assessed for disease relevance and mechanism of action. METHODS Candidate genetic risk variants were informatically prioritized and individually engineered into a LOAD-sensitized mouse model that carries the AD risk variants APOE ε4/ε4 and Trem2*R47H. The potential disease relevance of each model was assessed by comparing brain transcriptomes measured with the Nanostring Mouse AD Panel at 4 and 12 months of age with human study cohorts. RESULTS We created new models for 11 coding and loss-of-function risk variants. Transcriptomic effects from multiple genetic variants recapitulated a variety of human gene expression patterns observed in LOAD study cohorts. Specific models matched to emerging molecular LOAD subtypes. DISCUSSION These results provide an initial functionalization of 11 candidate risk variants and identify potential preclinical models for testing targeted therapeutics. HIGHLIGHTS A novel approach to validate genetic risk factors for late-onset AD (LOAD) is presented. LOAD risk variants were knocked in to conserved mouse loci. Variant effects were assayed by transcriptional analysis. Risk variants in Abca7, Mthfr, Plcg2, and Sorl1 loci modeled molecular signatures of clinical disease. This approach should generate more translationally relevant animal models.
Collapse
Affiliation(s)
| | | | - Ravi S. Pandey
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Asli Uyar
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | | | | | | | - Adrian L. Oblak
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Peter Bor‐Chian Lin
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Bridget Perkins
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Disha Soni
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Cindy Ingraham
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Audrey Lee‐Gosselin
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Bruce T. Lamb
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | | | - Gregory W. Carter
- The Jackson LaboratoryBar HarborMaineUSA
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| |
Collapse
|
3
|
Uzuner D, İlgün A, Düz E, Bozkurt FB, Çakır T. Multilayer Analysis of RNA Sequencing Data in Alzheimer's Disease to Unravel Molecular Mysteries. ADVANCES IN NEUROBIOLOGY 2024; 41:219-246. [PMID: 39589716 DOI: 10.1007/978-3-031-69188-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Alzheimer's disease (AD) is a complex disease, and numerous cellular events may be involved in etiology. RNAseq-based transcriptome data hold multilayer information content, which could be crucial in unraveling molecular mysteries of AD. It enables quantification of gene expression levels, identification of genomic variants, and elucidation of splicing anomalies such as exon skipping and intron retention. Additional integration of this information into protein-protein interaction networks and genome-scale metabolic models from the literature has potential to decipher functional modules and affected mechanisms for complex scenarios such as AD. In this chapter, we review the application areas of the multilayer content of RNAseq and associated integrative approaches available, with a special focus on AD.
Collapse
Affiliation(s)
- Dilara Uzuner
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Atılay İlgün
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Elif Düz
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Fatma Betül Bozkurt
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey.
| |
Collapse
|
4
|
Sasner M, Preuss C, Pandey RS, Uyar A, Garceau D, Kotredes KP, Williams H, Oblak AL, Lin PBC, Perkins B, Soni D, Ingraham C, Lee-Gosselin A, Lamb BT, Howell GR, Carter GW. In vivo validation of late-onset Alzheimer's disease genetic risk factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572849. [PMID: 38187758 PMCID: PMC10769393 DOI: 10.1101/2023.12.21.572849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Introduction Genome-wide association studies have identified over 70 genetic loci associated with late-onset Alzheimer's disease (LOAD), but few candidate polymorphisms have been functionally assessed for disease relevance and mechanism of action. Methods Candidate genetic risk variants were informatically prioritized and individually engineered into a LOAD-sensitized mouse model that carries the AD risk variants APOE4 and Trem2*R47H. Potential disease relevance of each model was assessed by comparing brain transcriptomes measured with the Nanostring Mouse AD Panel at 4 and 12 months of age with human study cohorts. Results We created new models for 11 coding and loss-of-function risk variants. Transcriptomic effects from multiple genetic variants recapitulated a variety of human gene expression patterns observed in LOAD study cohorts. Specific models matched to emerging molecular LOAD subtypes. Discussion These results provide an initial functionalization of 11 candidate risk variants and identify potential preclinical models for testing targeted therapeutics.
Collapse
Affiliation(s)
- Michael Sasner
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME, 04609 USA
| | | | - Ravi S Pandey
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032 USA
| | - Asli Uyar
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032 USA
| | - Dylan Garceau
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME, 04609 USA
| | | | | | - Adrian L Oblak
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Peter Bor-Chian Lin
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Bridget Perkins
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Disha Soni
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Cindy Ingraham
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Audrey Lee-Gosselin
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Bruce T Lamb
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Gareth R Howell
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME, 04609 USA
| | - Gregory W Carter
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME, 04609 USA
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032 USA
| |
Collapse
|
5
|
Winchester LM, Harshfield EL, Shi L, Badhwar A, Khleifat AA, Clarke N, Dehsarvi A, Lengyel I, Lourida I, Madan CR, Marzi SJ, Proitsi P, Rajkumar AP, Rittman T, Silajdžić E, Tamburin S, Ranson JM, Llewellyn DJ. Artificial intelligence for biomarker discovery in Alzheimer's disease and dementia. Alzheimers Dement 2023; 19:5860-5871. [PMID: 37654029 PMCID: PMC10840606 DOI: 10.1002/alz.13390] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 09/02/2023]
Abstract
With the increase in large multimodal cohorts and high-throughput technologies, the potential for discovering novel biomarkers is no longer limited by data set size. Artificial intelligence (AI) and machine learning approaches have been developed to detect novel biomarkers and interactions in complex data sets. We discuss exemplar uses and evaluate current applications and limitations of AI to discover novel biomarkers. Remaining challenges include a lack of diversity in the data sets available, the sheer complexity of investigating interactions, the invasiveness and cost of some biomarkers, and poor reporting in some studies. Overcoming these challenges will involve collecting data from underrepresented populations, developing more powerful AI approaches, validating the use of noninvasive biomarkers, and adhering to reporting guidelines. By harnessing rich multimodal data through AI approaches and international collaborative innovation, we are well positioned to identify clinically useful biomarkers that are accurate, generalizable, unbiased, and acceptable in clinical practice. HIGHLIGHTS: Artificial intelligence and machine learning approaches may accelerate dementia biomarker discovery. Remaining challenges include data set suitability due to size and bias in cohort selection. Multimodal data, diverse data sets, improved machine learning approaches, real-world validation, and interdisciplinary collaboration are required.
Collapse
Affiliation(s)
| | - Eric L Harshfield
- Department of Clinical Neurosciences, Stroke Research Group, University of Cambridge, Cambridge, UK
| | - Liu Shi
- Novo Nordisk Research Centre Oxford (NNRCO), Headington, UK
| | - AmanPreet Badhwar
- Département de Pharmacologie et Physiologie, Institut de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montreal, Canada
- Centre de recherche de l'Institut Universitaire de Gériatrie (CRIUGM), Montreal, Canada
| | - Ahmad Al Khleifat
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Natasha Clarke
- Centre de recherche de l'Institut Universitaire de Gériatrie (CRIUGM), Montreal, Canada
| | - Amir Dehsarvi
- School of Medicine, Medical Sciences, and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Imre Lengyel
- Wellcome-Wolfson Institute of Experimental Medicine, Queen's University, Belfast, UK
| | - Ilianna Lourida
- Health and Community Sciences, University of Exeter Medical School, Exeter, UK
| | | | - Sarah J Marzi
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Petroula Proitsi
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Anto P Rajkumar
- Institute of Mental Health, Mental Health and Clinical Neurosciences academic unit, University of Nottingham, Nottingham, UK, Mental health services of older people, Nottinghamshire healthcare NHS foundation trust, Nottingham, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Edina Silajdžić
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Janice M Ranson
- Health and Community Sciences, University of Exeter Medical School, Exeter, UK
| | - David J Llewellyn
- Health and Community Sciences, University of Exeter Medical School, Exeter, UK
- The Alan Turing Institute, London, UK
| |
Collapse
|
6
|
Nguyen TTT, Lee HH, Huang LK, Hu CJ, Yeh CY, Yang WCV, Lin MC. Heterogeneity of Alzheimer's disease identified by neuropsychological test profiling. PLoS One 2023; 18:e0292527. [PMID: 37797059 PMCID: PMC10553816 DOI: 10.1371/journal.pone.0292527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023] Open
Abstract
Alzheimer's disease (AD) is a highly heterogeneous disorder. Untangling this variability could lead to personalized treatments and improve participant recruitment for clinical trials. We investigated the cognitive subgroups by using a data-driven clustering technique in an AD cohort. People with mild-moderate probable AD from Taiwan was included. Neuropsychological test results from the Cognitive Abilities Screening Instrument were clustered using nonnegative matrix factorization. We identified two clusters in 112 patients with predominant deficits in memory (62.5%) and non-memory (37.5%) cognitive domains, respectively. The memory group performed worse in short-term memory and orientation and better in attention than the non-memory group. At baseline, patients in the memory group had worse global cognitive status and dementia severity. Linear mixed effect model did not reveal difference in disease trajectory within 3 years of follow-up between the two clusters. Our results provide insights into the cognitive heterogeneity in probable AD in an Asian population.
Collapse
Affiliation(s)
- Truc Tran Thanh Nguyen
- Graduate Institute of Biomedical Informatics, Division of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Memory and Dementia Center, Hospital 30–4, Ho Chi Minh City, Vietnam
| | - Hsun-Hua Lee
- Department of Neurology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Dizziness and Balance Disorder Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Li-Kai Huang
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, Dementia Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, Dementia Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Chih-Yang Yeh
- Graduate Institute of Biomedical Informatics, Division of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chung Vivian Yang
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ming-Chin Lin
- Graduate Institute of Biomedical Informatics, Division of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Biomedical Informatics, Division of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| |
Collapse
|
7
|
Higginbotham L, Carter EK, Dammer EB, Haque RU, Johnson ECB, Duong DM, Yin L, De Jager PL, Bennett DA, Felsky D, Tio ES, Lah JJ, Levey AI, Seyfried NT. Unbiased classification of the elderly human brain proteome resolves distinct clinical and pathophysiological subtypes of cognitive impairment. Neurobiol Dis 2023; 186:106286. [PMID: 37689213 PMCID: PMC10750427 DOI: 10.1016/j.nbd.2023.106286] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/24/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023] Open
Abstract
Cognitive impairment in the elderly features complex molecular pathophysiology extending beyond the hallmark pathologies of traditional disease classification. Molecular subtyping using large-scale -omic strategies can help resolve this biological heterogeneity. Using quantitative mass spectrometry, we measured ∼8000 proteins across >600 dorsolateral prefrontal cortex tissues with clinical diagnoses of no cognitive impairment (NCI), mild cognitive impairment (MCI), and Alzheimer's disease (AD) dementia. Unbiased classification of MCI and AD cases based on individual proteomic profiles resolved three classes with expression differences across numerous cell types and biological ontologies. Two classes displayed molecular signatures atypical of AD neurodegeneration, such as elevated synaptic and decreased inflammatory markers. In one class, these atypical proteomic features were associated with clinical and pathological hallmarks of cognitive resilience. We were able to replicate these classes and their clinicopathological phenotypes across two additional tissue cohorts. These results promise to better define the molecular heterogeneity of cognitive impairment and meaningfully impact its diagnostic and therapeutic precision.
Collapse
Affiliation(s)
- Lenora Higginbotham
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
| | - E Kathleen Carter
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B Dammer
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Rafi U Haque
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Erik C B Johnson
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc M Duong
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Luming Yin
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Taub Institute, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Daniel Felsky
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Earvin S Tio
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - James J Lah
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
8
|
Donato L, Mordà D, Scimone C, Alibrandi S, D'Angelo R, Sidoti A. How Many Alzheimer-Perusini's Atypical Forms Do We Still Have to Discover? Biomedicines 2023; 11:2035. [PMID: 37509674 PMCID: PMC10377159 DOI: 10.3390/biomedicines11072035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer-Perusini's (AD) disease represents the most spread dementia around the world and constitutes a serious problem for public health. It was first described by the two physicians from whom it took its name. Nowadays, we have extensively expanded our knowledge about this disease. Starting from a merely clinical and histopathologic description, we have now reached better molecular comprehension. For instance, we passed from an old conceptualization of the disease based on plaques and tangles to a more modern vision of mixed proteinopathy in a one-to-one relationship with an alteration of specific glial and neuronal phenotypes. However, no disease-modifying therapies are yet available. It is likely that the only way to find a few "magic bullets" is to deepen this aspect more and more until we are able to draw up specific molecular profiles for single AD cases. This review reports the most recent classifications of AD atypical variants in order to summarize all the clinical evidence using several discrimina (for example, post mortem neurofibrillary tangle density, cerebral atrophy, or FDG-PET studies). The better defined four atypical forms are posterior cortical atrophy (PCA), logopenic variant of primary progressive aphasia (LvPPA), behavioral/dysexecutive variant and AD with corticobasal degeneration (CBS). Moreover, we discuss the usefulness of such classifications before outlining the molecular-genetic aspects focusing on microglial activity or, more generally, immune system control of neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology, Via Michele Miraglia, 98139 Palermo, Italy
| | - Domenico Mordà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology, Via Michele Miraglia, 98139 Palermo, Italy
| | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology, Via Michele Miraglia, 98139 Palermo, Italy
| | - Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| | - Rosalia D'Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
| |
Collapse
|
9
|
Yang M, Matan-Lithwick S, Wang Y, De Jager PL, Bennett DA, Felsky D. Multi-omic integration via similarity network fusion to detect molecular subtypes of ageing. Brain Commun 2023; 5:fcad110. [PMID: 37082508 PMCID: PMC10110975 DOI: 10.1093/braincomms/fcad110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/17/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
Molecular subtyping of brain tissue provides insights into the heterogeneity of common neurodegenerative conditions, such as Alzheimer's disease. However, existing subtyping studies have mostly focused on single data modalities and only those individuals with severe cognitive impairment. To address these gaps, we applied similarity network fusion, a method capable of integrating multiple high-dimensional multi-omic data modalities simultaneously, to an elderly sample spanning the full spectrum of cognitive ageing trajectories. We analyzed human frontal cortex brain samples characterized by five omic modalities: bulk RNA sequencing (18 629 genes), DNA methylation (53 932 CpG sites), histone acetylation (26 384 peaks), proteomics (7737 proteins) and metabolomics (654 metabolites). Similarity network fusion followed by spectral clustering was used for subtype detection, and subtype numbers were determined by Eigen-gap and rotation cost statistics. Normalized mutual information determined the relative contribution of each modality to the fused network. Subtypes were characterized by associations with 13 age-related neuropathologies and cognitive decline. Fusion of all five data modalities (n = 111) yielded two subtypes (n S1 = 53, n S2 = 58), which were nominally associated with diffuse amyloid plaques; however, this effect was not significant after correction for multiple testing. Histone acetylation (normalized mutual information = 0.38), DNA methylation (normalized mutual information = 0.18) and RNA abundance (normalized mutual information = 0.15) contributed most strongly to this network. Secondary analysis integrating only these three modalities in a larger subsample (n = 513) indicated support for both three- and five-subtype solutions, which had significant overlap, but showed varying degrees of internal stability and external validity. One subtype showed marked cognitive decline, which remained significant even after correcting for tests across both three- and five-subtype solutions (p Bonf = 5.9 × 10-3). Comparison to single-modality subtypes demonstrated that the three-modal subtypes were able to uniquely capture cognitive variability. Comprehensive sensitivity analyses explored influences of sample size and cluster number parameters. We identified highly integrative molecular subtypes of ageing derived from multiple high dimensional, multi-omic data modalities simultaneously. Fusing RNA abundance, DNA methylation, and histone acetylation measures generated subtypes that were associated with cognitive decline. This work highlights the potential value and challenges of multi-omic integration in unsupervised subtyping of post-mortem brain.
Collapse
Affiliation(s)
- Mu Yang
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Stuart Matan-Lithwick
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Yanling Wang
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL 60612, USA
| | - Philip L De Jager
- The Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY 10033, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL 60612, USA
| | - Daniel Felsky
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
10
|
Katabathula S, Davis PB, Xu R. Comorbidity-driven multi-modal subtype analysis in mild cognitive impairment of Alzheimer's disease. Alzheimers Dement 2023; 19:1428-1439. [PMID: 36166485 PMCID: PMC10040466 DOI: 10.1002/alz.12792] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Mild cognitive impairment (MCI) is a heterogeneous condition with high individual variabilities in clinical outcomes driven by patient demographics, genetics, brain structure features, blood biomarkers, and comorbidities. Multi-modality data-driven approaches have been used to discover MCI subtypes; however, disease comorbidities have not been included as a modality though multiple diseases including hypertension are well-known risk factors for Alzheimer's disease (AD). The aim of this study was to examine MCI heterogeneity in the context of AD-related comorbidities along with other AD-relevant features and biomarkers. METHODS A total of 325 MCI subjects with 32 AD-relevant comorbidities and features were considered. Mixed-data clustering is applied to discover and compare MCI subtypes with and without including AD-related comorbidities. Finally, the relevance of each comorbidity-driven subtype was determined by examining their MCI to AD disease prognosis, descriptive statistics, and conversion rates. RESULTS We identified four (five) MCI subtypes: poor-, average-, good-, and best-AD prognosis by including comorbidities (without including comorbidities). We demonstrated that comorbidity-driven MCI subtypes differed from those identified without comorbidity information. We further demonstrated the clinical relevance of comorbidity-driven MCI subtypes. Among the four comorbidity-driven MCI subtypes there were substantial differences in the proportions of participants who reverted to normal function, remained stable, or converted to AD. The groups showed different behaviors, having significantly different MCI to AD prognosis, significantly different means for cognitive test-related and plasma features, and by the proportion of comorbidities. CONCLUSIONS Our study indicates that AD comorbidities should be considered along with other diverse AD-relevant characteristics to better understand MCI heterogeneity.
Collapse
Affiliation(s)
- Sreevani Katabathula
- Center for Artificial Intelligence in Drug Discovery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Pamela B Davis
- Center for Community Health Integration, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
11
|
Identification of the Subtypes of Renal Ischemia-Reperfusion Injury Based on Pyroptosis-Related Genes. Biomolecules 2023; 13:biom13020275. [PMID: 36830644 PMCID: PMC9952921 DOI: 10.3390/biom13020275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/29/2022] [Accepted: 01/19/2023] [Indexed: 02/05/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) often occurs in the process of kidney transplantation, which significantly impacts the subsequent treatment and prognosis of patients. The prognosis of patients with different subtypes of IRI is quite different. Therefore, in this paper, the gene expression data of multiple IRI samples were downloaded from the GEO database, and a double Laplacian orthogonal non-negative matrix factorization (DL-ONMF) algorithm was proposed to classify them. In this algorithm, various regularization constraints are added based on the non-negative matrix factorization algorithm, and the prior information is fused into the algorithm from different perspectives. The connectivity information between different samples and features is added to the algorithm by Laplacian regularization constraints on samples and features. In addition, orthogonality constraints on the basis matrix and coefficient matrix obtained by the algorithm decomposition are added to reduce the influence of redundant samples and redundant features on the results. Based on the DL-ONMF algorithm for clustering, two PRGs-related IRI isoforms were obtained in this paper. The results of immunoassays showed that the immune microenvironment was different among PRGS-related IRI types. Based on the differentially expressed PRGs between subtypes, we used LASSO and SVM-RFE algorithms to construct a diagnostic model related to renal transplantation. ROC analysis showed that the diagnostic model could predict the outcome of renal transplant patients with high accuracy. In conclusion, this paper presents an algorithm, DL-ONMF, which can identify subtypes with different disease characteristics. Comprehensive bioinformatic analysis showed that pyroptosis might affect the outcome of kidney transplantation by participating in the immune response of IRI.
Collapse
|
12
|
Bansal B, Sahoo A. Multi-omics data fusion using adaptive GTO guided Non-negative matrix factorization for cancer subtype discovery. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 228:107246. [PMID: 36434961 DOI: 10.1016/j.cmpb.2022.107246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND OBJECTIVE Cancer subtype discovery is essential for personalized clinical treatment. With the onset of progressive profile techniques for cancer, a large amount of heterogeneous and high-dimensional transcriptomic, proteomic and genomic datasets are easily accumulated. Integrative clustering of such multi-omics data is crucial to recognize their latent structure and to acknowledge the correlation within and across them. Although the integrative analysis of diversified multi-omics data is informative, it is challenging when multiplicity in data inflicts poor accordance w.r.t. clustering structure. The objective of this work is to develop an effective integrative analysis framework that encapsulates the heterogeneity of various biological mechanisms and predicts homogeneous subgroups of cancer patients. METHOD In this paper, improved sparse-joint non-negative matrix factorization (sparse-jNMF) has been devised for the problem of cancer-subtype discovery. The initial points of sparse-jNMF have improved using a novel meta-heuristic algorithm adaptive gorilla troops optimizer (Ada-GTO). Improving the initialization of sparse-jNMF enhances its convergence behavior and further strengthens the clustering performance. In addition, the consensus clustering approach has been adopted to construct a patient-patient similarity matrix for obtaining stable clusters of patient samples. RESULT The proposed framework has been applied to 4 different real-life multi-omics cancer datasets, namely colon adenocarcinoma, breast-invasive carcinoma, kidney-renal clear-cell carcinoma, and lung adenocarcinoma. The proposed method results in patient clusters with better silhouette scores and cluster purity than classical initialization and similar meta-heuristics for initial point estimation approaches. Survival probabilities estimated using Kaplan-Meier (KM) curve show statistically significant difference (p < 0.05) for the homogenous cancer patient clusters obtained using the proposed method as compared to iCluster. The presented approach further identified the somatic mutations for the classified subgroups, which is beneficial to provide targeted treatments. CONCLUSION This paper proposes Ada-GTO guided sparse-jNMF framework for cancer subtype discovery, considering the information from multiple omic features that provide comprehension. The proposed meta-guided framework outperforms all other state-of-the-art counterparts. It also has great potential for obtaining the homogeneous subgroups of other diseases.
Collapse
Affiliation(s)
- Bhavana Bansal
- Department of CSE & IT, Jaypee Institute of Information Technology, Noida, India.
| | - Anita Sahoo
- Department of CSE & IT, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
13
|
Rather AA, Chachoo MA. Manifold learning based robust clustering of gene expression data for cancer subtyping. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
14
|
Jellinger KA. Recent update on the heterogeneity of the Alzheimer’s disease spectrum. J Neural Transm (Vienna) 2021; 129:1-24. [DOI: 10.1007/s00702-021-02449-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/25/2021] [Indexed: 02/03/2023]
|