1
|
Cimino A, Pat F, Oyebamiji O, Pferdehirt L, Pham CTN, Herzog ED, Guilak F. Programmable chronogenetic gene circuits for self-regulated circadian delivery of biologic drugs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643274. [PMID: 40161636 PMCID: PMC11952517 DOI: 10.1101/2025.03.14.643274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Cells of the body rely on the circadian clock to orchestrate daily changes in physiology that impact both homeostatic and pathological conditions, such as the inflammatory autoimmune disease rheumatoid arthritis (RA). In RA, high levels of proinflammatory cytokines peak early in the morning hours, reflected by daily changes in joint stiffness. Chronotherapy (or circadian medicine) seeks to delivery drugs at optimal times to maximize their efficacy. However, chronotherapy remains a largely unexplored approach for disease modifying, antirheumatic treatment, particularly for cell-based therapies. In this study, we developed autonomous chronogenetic gene circuits that produce the biologic drug interleukin-1 receptor antagonist (IL-1Ra) with desired phase and amplitude. We compared expression of IL-1Ra from circuits that contained different circadian promoter elements (E'-boxes, D-boxes, or RREs) and their ability to respond to inflammatory challenges in murine pre-differentiated induced pluripotent stem cells (PDiPSC) or engineered cartilage pellets. We confirmed that each circuit reliably peaked at a distinct circadian time over multiple days. Engineered cells generated significant amounts of IL-1Ra on a circadian basis, which protected them from circadian dysregulation and inflammatory damage. These programmable chronogenetic circuits have the potential to align with an individual's circadian rhythm for optimized, self-regulated daily drug delivery.
Collapse
|
2
|
Yusuff AS. Exploring Potential Mechanisms of Sleep Disorders in Alzheimer's Dementia: A Scoping Review. Cureus 2025; 17:e76859. [PMID: 39902010 PMCID: PMC11788456 DOI: 10.7759/cureus.76859] [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] [Accepted: 01/02/2025] [Indexed: 02/05/2025] Open
Abstract
Alzheimer's dementia (AD) is characterized by a progressive decline in behavioral and cognitive functions, with sleep disorders (SDs) increasingly recognized as one of the noncognitive symptoms. Sleep plays a critical role in the brain, supporting learning and memory, regulating synaptic plasticity, and facilitating waste clearance. However, the mechanisms underlying sleep disturbances in AD remain poorly understood. This review aims to explore these mechanisms and their potential relevance for clinicians managing AD. A systematic search was conducted across multiple sources and databases, using keywords such as "Alzheimer AND sleep disorder", along with terms related to neurodegeneration and sleep disturbances. Of the 1,511 records identified, 18 were included in the final analysis. The findings highlight several mechanisms linking AD and SDs, suggesting a bidirectional relationship. These mechanisms include (i) shared genetic factors; (ii) disruption of the glymphatic system; (iii) circadian system dysregulation; (iv) neuroinflammation; (v) abnormal functional connectivity between related brain regions; and (vi) atrophy in brain regions involved in memory and sleep. In conclusion, the relationship between AD and SDs is complex and bidirectional. Sleep disturbances not only precede the onset of AD but also worsen as the disease progresses. Sleep may, therefore, serve as a promising biomarker for AD, with targeting sleep disturbances offering a potential early therapeutic strategy in managing AD.
Collapse
|
3
|
Zhou X, Du K, Mao T, Wang N, Zhang L, Tian Y, Liu T, Wang L, Wang X. BMAL1 upregulates STX17 levels to promote autophagosome-lysosome fusion in hippocampal neurons to ameliorate Alzheimer's disease. iScience 2024; 27:111413. [PMID: 39687016 PMCID: PMC11647228 DOI: 10.1016/j.isci.2024.111413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/02/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
We aim to investigate muscle ARNT-like protein 1 (BMAL1) regulation of syntaxin17 (STX17) in mouse hippocampal neurons, focusing on autophagy and amyloid-β (Aβ) deposition. Autophagosome-lysosome fusion in APP/PS1 hippocampal tissues was observed using transmission electron microscopy, while mRNA levels of LC3II and P62 were measured via reverse-transcription PCR (RT-PCR) after Amyloid precursor protein (APP) overexpression. STX17, linked to autophagy and differentially expressed in Alzheimer's disease (AD) brains, was knocked down or overexpressed to assess its effects. The results showed that reduced STX17 impairs autophagosome-lysosome fusion, leading to abnormal Aβ deposition. Coimmunoprecipitation (Co-IP) and immunofluorescence confirmed STX17 interaction with SNAP29 and VAMP8 to form SNARE complexes. Furthermore, BMAL1 binding to STX17 was examined using luciferase assays. Circadian rhythm disturbances and decreased BMAL1 expression in APP/PS1 mice were noted, while BMAL1 overexpression upregulated STX17 expression and promoted autophagy to reduce Aβ deposition. Thus, the BMAL1 protein can promote STX17 transcription to induce STX17-SNAP29-VAMP8 complex formation to clear intracellular Aβ through autophagy.
Collapse
Affiliation(s)
- Xiuya Zhou
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Kaili Du
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Tian Mao
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Ning Wang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Lifei Zhang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Yuan Tian
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Ting Liu
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Li Wang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Xiaohui Wang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
4
|
Liu CR, Yang CY, Sharma D, Chen TH, Huang XQ, Hung TM, Kuo TBJ, Jou JH. Associations between Sleep Duration and Autonomic Nervous System Regulation in Patients with Probable Alzheimer's Disease: A Cross-Sectional Pilot Study. Clocks Sleep 2024; 6:533-545. [PMID: 39449309 PMCID: PMC11503315 DOI: 10.3390/clockssleep6040035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024] Open
Abstract
In this study, we aimed to investigate the relationships between sleep duration and autonomic nervous system (ANS) regulation. This cross-sectional pilot study included 27 older patients with probable Alzheimer's disease who were hospitalized at a psychiatric center. We measured heart rate variability to assess ANS regulation at night, evaluated dementia severity via the Clinical Dementia Rating scale, and obtained sleep duration data from sleep diaries maintained by psychiatric nurses. The data were analyzed using repeated-measures generalized linear models with age, sex, dementia severity, hypertension status, and medication use (antipsychotics) as covariates. A sleep duration of 6-9 h per night compared to shorter than 6 h was associated with a greater increase in parasympathetic nervous system activity (p = 0.03), and a sleep duration longer than 9 h was associated with a decrease sympathovagal balance (p = 0.02). In addition, we observed an inverted U-shaped association between sleep duration and ANS regulation. In this pilot study, we demonstrated that a sleep duration of 6-9 h per night may be beneficial for ANS regulation; however, the present study involved only a few participants and had some limitations. Additional research with a larger cohort is needed to confirm these findings.
Collapse
Affiliation(s)
- Chuen-Ru Liu
- Taipei City Hospital Songde Branch, Taipei City 110204, Taiwan; (C.-R.L.); (T.-M.H.)
| | - Chih-Yuan Yang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei City 112304, Taiwan;
| | - Dipanshu Sharma
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan; (D.S.); (T.-H.C.); (X.-Q.H.)
| | - Tun-Hao Chen
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan; (D.S.); (T.-H.C.); (X.-Q.H.)
| | - Xian-Qing Huang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan; (D.S.); (T.-H.C.); (X.-Q.H.)
| | - Tsui-Mei Hung
- Taipei City Hospital Songde Branch, Taipei City 110204, Taiwan; (C.-R.L.); (T.-M.H.)
| | - Terry B. J. Kuo
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei City 112304, Taiwan;
| | - Jwo-Huei Jou
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan; (D.S.); (T.-H.C.); (X.-Q.H.)
| |
Collapse
|
5
|
Chen Y, Al-Nusaif M, Li S, Tan X, Yang H, Cai H, Le W. Progress on early diagnosing Alzheimer's disease. Front Med 2024; 18:446-464. [PMID: 38769282 PMCID: PMC11391414 DOI: 10.1007/s11684-023-1047-1] [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: 09/23/2023] [Accepted: 11/15/2023] [Indexed: 05/22/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects both cognition and non-cognition functions. The disease follows a continuum, starting with preclinical stages, progressing to mild cognitive and behavioral impairment, ultimately leading to dementia. Early detection of AD is crucial for better diagnosis and more effective treatment. However, the current AD diagnostic tests of biomarkers using cerebrospinal fluid and/or brain imaging are invasive or expensive, and mostly are still not able to detect early disease state. Consequently, there is an urgent need to develop new diagnostic techniques with higher sensitivity and specificity during the preclinical stages of AD. Various non-cognitive manifestations, including behavioral abnormalities, sleep disturbances, sensory dysfunctions, and physical changes, have been observed in the preclinical AD stage before occurrence of notable cognitive decline. Recent research advances have identified several biofluid biomarkers as early indicators of AD. This review focuses on these non-cognitive changes and newly discovered biomarkers in AD, specifically addressing the preclinical stages of the disease. Furthermore, it is of importance to explore the potential for developing a predictive system or network to forecast disease onset and progression at the early stage of AD.
Collapse
Affiliation(s)
- Yixin Chen
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Murad Al-Nusaif
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Xiang Tan
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huijia Yang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China.
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China.
| |
Collapse
|
6
|
Song Z, Ho CT, Zhang X. Gut Microbiota Mediate the Neuroprotective Effect of Oolong Tea Polyphenols in Cognitive Impairment Induced by Circadian Rhythm Disorder. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12184-12197. [PMID: 38745351 DOI: 10.1021/acs.jafc.4c01922] [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: 05/16/2024]
Abstract
Oolong tea polyphenols (OTP) have attracted wide attention due to their ability to reduce inflammatory response, regulate gut microbiota, and improve cognitive function. However, exactly how the gut microbiota modulates nervous system activity is still an open question. We previously expounded that supplementing with OTP alleviated neuroinflammation in circadian rhythm disorder (CRD) mice. Here, we showed that OTP can relieve microglia activation by reducing harmful microbial metabolites lipopolysaccharide (LPS) that alleviate CRD-induced cognitive decline. Mechanistically, OTP suppressed the inflammation response by regulating the gut microbiota composition, including upregulating the relative abundance of Muribaculaceae and Clostridia_UCG-014 and downregulating Desulfovibrio, promoting the production of short-chain fatty acids (SCFAs). Moreover, the use of OTP alleviated intestinal barrier damage and decreased the LPS transport to the serum. These results further inhibited the activation of microglia, thus alleviating cognitive impairment by inhibiting neuroinflammation, neuron damage, and neurotoxicity metabolite glutamate elevation. Meanwhile, OTP upregulated the expression of synaptic plasticity-related protein postsynaptic density protein 95 (PSD-95) and synaptophysin (SYN) by elevating the brain-derived neurotrophic factor (BDNF) level. Taken together, our findings suggest that the OTP has the potential to prevent CRD-induced cognition decline by modulating gut microbiota and microbial metabolites.
Collapse
Affiliation(s)
- Zheyi Song
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, P. R. China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, P. R. China
| |
Collapse
|
7
|
Long DM, Cravetchi O, Chow ES, Allen C, Kretzschmar D. The amyloid precursor protein intracellular domain induces sleep disruptions and its nuclear localization fluctuates in circadian pacemaker neurons in Drosophila and mice. Neurobiol Dis 2024; 192:106429. [PMID: 38309627 DOI: 10.1016/j.nbd.2024.106429] [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: 04/07/2023] [Revised: 11/17/2023] [Accepted: 02/01/2024] [Indexed: 02/05/2024] Open
Abstract
The most prominent symptom of Alzheimer's disease (AD) is cognitive decline; however, sleep and other circadian disruptions are also common in AD patients. Sleep disruptions have been connected with memory problems and therefore the changes in sleep patterns observed in AD patients may also actively contribute to cognitive decline. However, the underlying molecular mechanisms that connect sleep disruptions and AD are unclear. A characteristic feature of AD is the formation of plaques consisting of Amyloid-β (Aβ) peptides generated by cleavage of the Amyloid Precursor Protein (APP). Besides Aβ, APP cleavage generates several other fragments, including the APP intracellular domain (AICD) that has been linked to transcriptional regulation and neuronal homeostasis. Here we show that overexpression of the AICD reduces the early evening expression of two core clock genes and disrupts the sleep pattern in flies. Analyzing the subcellular localization of the AICD in pacemaker neurons, we found that the AICD levels in the nucleus are low during daytime but increase at night. While this pattern of nuclear AICD persisted with age, the nighttime levels were higher in aged flies. Increasing the cleavage of the fly APP protein also disrupted AICD nuclear localization. Lastly, we show that the day/nighttime nuclear pattern of the AICD is also detectable in neurons in the suprachiasmatic nucleus of mice and that it also changes with age. Together, these data suggest that AD-associated changes in APP processing and the subsequent changes in AICD levels may cause sleep disruptions in AD.
Collapse
Affiliation(s)
- Dani M Long
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR 97239, USA.
| | - Olga Cravetchi
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| | - Eileen S Chow
- Department of Integrative Biology, Oregon State University, Corvallis, OR 97331, USA
| | - Charles Allen
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
8
|
Fang W, Le S, Han W, Peng-Jiao X, Shuai Y, Rui-Ling Z, Lin L, Ya-Hui X. Association between napping and cognitive impairment: A systematic review and meta-analysis. Sleep Med 2023; 111:146-159. [PMID: 37776585 DOI: 10.1016/j.sleep.2023.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/12/2023] [Accepted: 09/23/2023] [Indexed: 10/02/2023]
Abstract
STUDY OBJECTIVES Increasing evidence suggests that napping is associated with cognitive impairment and dementia, but the conclusions are inconsistent. Moreover, the extent of the risk is uncertain. We therefore conducted a systematic review and meta-analysis to quantify the connection between napping and cognitive impairment. METHODS We performed a systematic search of PubMed, EMBASE, Web of Science, and Cochrane Library for studies that were published up to June 2023, and assessed associations between napping and cognitive impairment. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated as the effect sizes for all studies. Heterogeneity and potential publication biases were assessed. RESULTS A total of 4535 papers were retrieved, with 20 reports assessing the relationships between napping and cognitive impairment. Pooled analysis indicated that napping was associated with dementia (OR = 1.14; 95% CI: 1.07-1.21). Importantly, we found that those napping longer than 30, 45, and 60 min/day were 35%, 41%, and 40%, respectively, more likely to have an increased risk of cognitive impairment (30 min: OR = 1.35; 95% CI: 1.24-1.48; 45 min: OR = 1.41; 95% CI: 1.27-1.58; 60 min: OR = 1.40; 95% CI: 1.26-1.56). North America and Europe showed that associations existed between napping and cognitive impairment (North America: OR = 1.15; 95% CI: 1.04-1.27; Europe: OR = 1.13; 95% CI: 1.08-1.18). CONCLUSIONS This meta-analysis indicated associations between long napping durations and cognitive impairment or dementia, suggesting that longer napping might be a potential risk factor of adverse cognitive outcomes.
Collapse
Affiliation(s)
- Wu Fang
- Department of Sleep Medicine, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shi Le
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No. 2018RU006), Peking University, Beijing, China
| | - Wang Han
- Department of Sleep Medicine, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xu Peng-Jiao
- Department of Sleep Medicine, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yu Shuai
- Department of Sleep Medicine, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhang Rui-Ling
- Department of Sleep Medicine, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Lu Lin
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No. 2018RU006), Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Beijing, China.
| | - Xu Ya-Hui
- Department of Sleep Medicine, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
9
|
Wirian YB, Jiang Y, Cerel-Suhl S, Suhl J, Cheng Q. Exploring the Link Between Brain Waves and Sleep Patterns with Deep Learning Manifold Alignment. THE 4TH JOINT INTERNATIONAL CONFERENCE ON DEEP LEARNING, BIG DATA AND BLOCKCHAIN (DBB 2023). JOINT INTERNATIONAL CONFERENCE ON DEEP LEARNING, BIG DATA AND BLOCKCHAIN (4TH : 2023 : MARRAKECH, MOROCCO ; ONLINE) 2023; 768:81-90. [PMID: 38939181 PMCID: PMC11210370 DOI: 10.1007/978-3-031-42317-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Medical data are often multi-modal, which are collected from different sources with different formats, such as text, images, and audio. They have some intrinsic connections in meaning and semantics while manifesting disparate appearances. Polysomnography (PSG) datasets are multi-modal data that include hypnogram, electrocardiogram (ECG), and electroencephalogram (EEG). It is hard to measure the associations between different modalities. Previous studies have used PSG datasets to study the relationship between sleep disorders and quality and sleep architecture. We leveraged a new method of deep learning manifold alignment to explore the relationship between sleep architecture and EEG features. Our analysis results agreed with the results of previous studies that used PSG datasets to diagnose different sleep disorders and monitor sleep quality in different populations. The method could effectively find the associations between sleep architecture and EEG datasets, which are important for understanding the changes in sleep stages and brain activity. On the other hand, the Spearman correlation method, which is a common statistical technique, could not find the correlations between these datasets.
Collapse
Affiliation(s)
| | - Yang Jiang
- Behavioral Science Department, University of Kentucky, Lexington, KY 40536, USA
| | - Sylvia Cerel-Suhl
- Sleep Center, Lexington Veterans Affairs Medical Center, Lexington, KY 40511, USA
| | - Jeremiah Suhl
- Sleep Center, Lexington Veterans Affairs Medical Center, Lexington, KY 40511, USA
| | - Qiang Cheng
- Computer Science Department, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
10
|
Abstract
Despite monumental advances in molecular technology to generate genome sequence data at scale, there is still a considerable proportion of heritability in most complex diseases that remains unexplained. Because many of the discoveries have been single-nucleotide variants with small to moderate effects on disease, the functional implication of many of the variants is still unknown and, thus, we have limited new drug targets and therapeutics. We, and many others, posit that one primary factor that has limited our ability to identify novel drug targets from genome-wide association studies may be due to gene interactions (epistasis), gene-environment interactions, network/pathway effects, or multiomic relationships. We propose that many of these complex models explain much of the underlying genetic architecture of complex disease. In this review, we discuss the evidence from multiple research avenues, ranging from pairs of alleles to multiomic integration studies and pharmacogenomics, that supports the need for further investigation of gene interactions (or epistasis) in genetic and genomic studies of human disease. Our goal is to catalog the mounting evidence for epistasis in genetic studies and the connections between genetic interactions and human health and disease that could enable precision medicine of the future.
Collapse
Affiliation(s)
- Pankhuri Singhal
- Genetics and Epigenetics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shefali Setia Verma
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA;
- Penn Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Contreras EG, Klämbt C. The Drosophila blood-brain barrier emerges as a model for understanding human brain diseases. Neurobiol Dis 2023; 180:106071. [PMID: 36898613 DOI: 10.1016/j.nbd.2023.106071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
The accurate regulation of the microenvironment within the nervous system is one of the key features characterizing complex organisms. To this end, neural tissue has to be physically separated from circulation, but at the same time, mechanisms must be in place to allow controlled transport of nutrients and macromolecules into and out of the brain. These roles are executed by cells of the blood-brain barrier (BBB) found at the interface of circulation and neural tissue. BBB dysfunction is observed in several neurological diseases in human. Although this can be considered as a consequence of diseases, strong evidence supports the notion that BBB dysfunction can promote the progression of brain disorders. In this review, we compile the recent evidence describing the contribution of the Drosophila BBB to the further understanding of brain disease features in human patients. We discuss the function of the Drosophila BBB during infection and inflammation, drug clearance and addictions, sleep, chronic neurodegenerative disorders and epilepsy. In summary, this evidence suggests that the fruit fly, Drosophila melanogaster, can be successfully employed as a model to disentangle mechanisms underlying human diseases.
Collapse
Affiliation(s)
- Esteban G Contreras
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| | - Christian Klämbt
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| |
Collapse
|