1
|
Alur V, Vastrad B, Raju V, Vastrad C, Kotturshetti S. The identification of key genes and pathways in polycystic ovary syndrome by bioinformatics analysis of next-generation sequencing data. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2024; 29:53. [DOI: 10.1186/s43043-024-00212-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/17/2024] [Indexed: 01/02/2025] Open
Abstract
Abstract
Background
Polycystic ovary syndrome (PCOS) is a reproductive endocrine disorder. The specific molecular mechanism of PCOS remains unclear. The aim of this study was to apply a bioinformatics approach to reveal related pathways or genes involved in the development of PCOS.
Methods
The next-generation sequencing (NGS) dataset GSE199225 was downloaded from the gene expression omnibus (GEO) database and NGS dataset analyzed is obtained from in vitro culture of PCOS patients’ muscle cells and muscle cells of healthy lean control women. Differentially expressed gene (DEG) analysis was performed using DESeq2. The g:Profiler was utilized to analyze the gene ontology (GO) and REACTOME pathways of the differentially expressed genes. A protein–protein interaction (PPI) network was constructed and module analysis was performed using HiPPIE and cytoscape. The miRNA-hub gene regulatory network and TF-hub gene regulatory network were constructed. The hub genes were validated by using receiver operating characteristic (ROC) curve analysis.
Results
We have identified 957 DEG in total, including 478 upregulated genes and 479 downregulated gene. GO terms and REACTOME pathways illustrated that DEG were significantly enriched in regulation of molecular function, developmental process, interferon signaling and platelet activation, signaling, and aggregation. The top 5 upregulated hub genes including HSPA5, PLK1, RIN3, DBN1, and CCDC85B and top 5 downregulated hub genes including DISC1, AR, MTUS2, LYN, and TCF4 might be associated with PCOS. The hub gens of HSPA5 and KMT2A, together with corresponding predicted miRNAs (e.g., hsa-mir-34b-5p and hsa-mir-378a-5p), and HSPA5 and TCF4 together with corresponding predicted TF (e.g., RCOR3 and TEAD4) were found to be significantly correlated with PCOS.
Conclusions
These study uses of bioinformatics analysis of NGS data to obtain hub genes and key signaling pathways related to PCOS and its associated complications. Also provides novel ideas for finding biomarkers and treatment methods for PCOS and its associated complications.
Collapse
|
2
|
Simmonds E, Leonenko G, Yaman U, Bellou E, Myers A, Morgan K, Brookes K, Hardy J, Salih D, Escott-Price V. Chromosome X-wide association study in case control studies of pathologically confirmed Alzheimer's disease in a European population. Transl Psychiatry 2024; 14:358. [PMID: 39231932 PMCID: PMC11375158 DOI: 10.1038/s41398-024-03058-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024] Open
Abstract
Although there are several genome-wide association studies available which highlight genetic variants associated with Alzheimer's disease (AD), often the X chromosome is excluded from the analysis. We conducted an X-chromosome-wide association study (XWAS) in three independent studies with a pathologically confirmed phenotype (total 1970 cases and 1113 controls). The XWAS was performed in males and females separately, and these results were then meta-analysed. Four suggestively associated genes were identified which may be of potential interest for further study in AD, these are DDX53 (rs12006935, OR = 0.52, p = 6.9e-05), IL1RAPL1 (rs6628450, OR = 0.36, p = 4.2e-05; rs137983810, OR = 0.52, p = 0.0003), TBX22 (rs5913102, OR = 0.74, p = 0.0003) and SH3BGRL (rs186553004, OR = 0.35, p = 0.0005; rs113157993, OR = 0.52, p = 0.0003), which replicate across at least two studies. The SNP rs5913102 in TBX22 achieves chromosome-wide significance in meta-analysed data. DDX53 shows highest expression in astrocytes, IL1RAPL1 is most highly expressed in oligodendrocytes and neurons and SH3BGRL is most highly expressed in microglia. We have also identified SNPs in the NXF5 gene at chromosome-wide significance in females (rs5944989, OR = 0.62, p = 1.1e-05) but not in males (p = 0.83). The discovery of relevant AD associated genes on the X chromosome may identify AD risk differences and similarities based on sex and lead to the development of sex-stratified therapeutics.
Collapse
Affiliation(s)
- Emily Simmonds
- Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Ganna Leonenko
- Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Umran Yaman
- Institute of Neurology, University College London, London, UK
| | - Eftychia Bellou
- Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Amanda Myers
- Department of Cell Biology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | | | - Keeley Brookes
- Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - John Hardy
- Institute of Neurology, University College London, London, UK
| | - Dervis Salih
- Institute of Neurology, University College London, London, UK
| | - Valentina Escott-Price
- Dementia Research Institute, Cardiff University, Cardiff, UK.
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
3
|
Evans AD, Pournoori N, Saksala E, Oommen OP. Glycosaminoglycans' for brain health: Harnessing glycosaminoglycan based biomaterials for treating central nervous system diseases and in-vitro modeling. Biomaterials 2024; 309:122629. [PMID: 38797120 DOI: 10.1016/j.biomaterials.2024.122629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/06/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
Dysfunction of the central nervous system (CNS) following traumatic brain injuries (TBI), spinal cord injuries (SCI), or strokes remains challenging to address using existing medications and cell-based therapies. Although therapeutic cell administration, such as stem cells and neuronal progenitor cells (NPCs), have shown promise in regenerative properties, they have failed to provide substantial benefits. However, the development of living cortical tissue engineered grafts, created by encapsulating these cells within an extracellular matrix (ECM) mimetic hydrogel scaffold, presents a promising functional replacement for damaged cortex in cases of stroke, SCI, and TBI. These grafts facilitate neural network repair and regeneration following CNS injuries. Given that natural glycosaminoglycans (GAGs) are a major constituent of the CNS, GAG-based hydrogels hold potential for the next generation of CNS healing therapies and in vitro modeling of CNS diseases. Brain-specific GAGs not only offer structural and biochemical signaling support to encapsulated neural cells but also modulate the inflammatory response in lesioned brain tissue, facilitating host integration and regeneration. This review briefly discusses different roles of GAGs and their related proteoglycan counterparts in healthy and diseases brain and explores current trends and advancements in GAG-based biomaterials for treating CNS injuries and modeling diseases. Additionally, it examines injectable, 3D bioprintable, and conductive GAG-based scaffolds, highlighting their clinical potential for in vitro modeling of patient-specific neural dysfunction and their ability to enhance CNS regeneration and repair following CNS injury in vivo.
Collapse
Affiliation(s)
- Austin D Evans
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland
| | - Negin Pournoori
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland
| | - Emmi Saksala
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland
| | - Oommen P Oommen
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland; School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK.
| |
Collapse
|
4
|
Nashiry MA, Sumi SS, Alyami SA, Moni MA. Systems biology approach discovers comorbidity interaction of Parkinson's disease with psychiatric disorders utilizing brain transcriptome. Front Mol Neurosci 2023; 16:1232805. [PMID: 37654790 PMCID: PMC10466791 DOI: 10.3389/fnmol.2023.1232805] [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: 06/01/2023] [Accepted: 07/12/2023] [Indexed: 09/02/2023] Open
Abstract
Several studies found that most patients with Parkinson's disorder (PD) appear to have psychiatric symptoms such as depression, anxiety, hallucination, delusion, and cognitive dysfunction. Therefore, recognizing these psychiatrically symptoms of PD patients is crucial for both symptomatic therapy and better knowledge of the pathophysiology of PD. In order to address this issue, we created a bioinformatics framework to determine the effects of PD mRNA expression on understanding its relationship with psychiatric symptoms in PD patients. We have discovered a significant overlap between the sets of differentially expressed genes from PD exposed tissue and psychiatric disordered tissues using RNA-seq datasets. We have chosen Bipolar disorder and Schizophrenia as psychiatric disorders in our study. A number of significant correlations between PD and the occurrence of psychiatric diseases were also found by gene set enrichment analysis, investigations of the protein-protein interaction network, gene regulatory network, and protein-chemical agent interaction network. We anticipate that the results of this pathogenetic study will provide crucial information for understanding the intricate relationship between PD and psychiatric diseases.
Collapse
Affiliation(s)
- Md Asif Nashiry
- Data Analytics, Northern Alberta Institute of Technology, Edmonton, AB, Canada
| | - Shauli Sarmin Sumi
- Computer Science and Engineering, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Salem A. Alyami
- Mathematics and Statistics, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Mohammad Ali Moni
- Artificial Intelligence and Data Science, Faculty of Health and Behavioural Sciences, School of Health and Rehabilitation Sciences, The University of Queensland, Saint Lucia, QLD, Australia
- Artificial Intelligence and Cyber Futures Institute, Charles Stuart University, Bathurst, NSW, Australia
| |
Collapse
|
5
|
Sarbu M, Ica R, Sharon E, Clemmer DE, Zamfir AD. Glycomics by ion mobility tandem mass spectrometry of chondroitin sulfate disaccharide domain in biglycan. JOURNAL OF MASS SPECTROMETRY : JMS 2023; 58:e4908. [PMID: 36799777 DOI: 10.1002/jms.4908] [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: 12/15/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Biglycan (BGN), a small leucine-rich repeat proteoglycan, is involved in a variety of pathological processes including malignant transformation, for which the upregulation of BGN was found related to cancer cell invasiveness. Because the functions of BGN are mediated by its chondroitin/dermatan sulfate (CS/DS) chains through the sulfates, the determination of CS/DS structure and sulfation pattern is of major importance. In this study, we have implemented an advanced glycomics method based on ion mobility separation (IMS) mass spectrometry (MS) and tandem MS (MS/MS) to characterize the CS disaccharide domains in BGN. The high separation efficiency and sensitivity of this technique allowed the discrimination of five distinct CS disaccharide motifs, of which four irregulated in their sulfation pattern. For the first time, trisulfated unsaturated and bisulfated saturated disaccharides were found in BGN, the latter species documenting the non-reducing end of the chains. The structural investigation by IMS MS/MS disclosed that in one or both of the CS/DS chains, the non-reducing end is 3-O-sulfated GlcA in a rather rare bisulfated motif having the structure 3-O-sulfated GlcA-4-O-sulfated GalNAc. Considering the role played by BGN in cancer cell spreading, the influence on this process of the newly identified sequences will be investigated in the future.
Collapse
Affiliation(s)
- Mirela Sarbu
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300569, Romania
| | - Raluca Ica
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300569, Romania
- Department of Physics, West University of Timisoara, Timisoara, 300223, Romania
| | - Edie Sharon
- Department of Chemistry, The College of Arts & Science, Indiana University, Bloomington, Indiana, USA
| | - David E Clemmer
- Department of Chemistry, The College of Arts & Science, Indiana University, Bloomington, Indiana, USA
| | - Alina D Zamfir
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, Timisoara, 300569, Romania
- Department of Technical and Natural Sciences, "Aurel Vlaicu" University of Arad, Arad, 310330, Romania
| |
Collapse
|
6
|
Wang Y, Ren Y, Hong T, Lu D, Zhang F, Cao Y, Wang X. Lipidomics Changes in a Murine Model of Neuropsychiatric Lupus. J Inflamm Res 2022; 15:6569-6580. [PMID: 36506783 PMCID: PMC9733566 DOI: 10.2147/jir.s391595] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/18/2022] [Indexed: 12/11/2022] Open
Abstract
Purpose Neuropsychiatric lupus (NPSLE) is one of the important manifestations of systemic lupus erythematosus. Previous studies mainly focused on the disruption of the blood-brain barrier and the production of brain-reactive autoantibodies, However, there is no comprehensive lipidomic analysis in NPSLE. Therefore, this research evaluated the lipidomic analysis in the hippocampus and liver of NPSLE mice with mood disorders, to explore the influence of the liver-brain axis on this disease. Methods MRL/lpr mice and MRL/mpj mice were respectively used as NPSLE and control groups. Behavioral tests and systemic disease characteristics of mice were assessed at the age of 18 weeks. Ultra-Performance Liquid Chromatography-Tandem Mass Spectrometry (UPLC-MS/MS) was used for lipid metabolite determination. Multivariate statistical analysis was used to identify lipid metabolites that were differentially expressed in two groups. Results Our results showed that 355 and 405 lipid metabolites were differentially expressed between the NPSLE and control groups in the hippocampus and liver. According to the pathway enrichment analysis, several pathways were affected, and the glycerophospholipid metabolism pathway was most relevant to the mouse's depressive behavior. Conclusion Based on UPLC-MS/MS, the results provide evidence for how the liver-brain axis affects NPSLE and improve the understanding of NPSLE pathogenesis.
Collapse
Affiliation(s)
- Yihan Wang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Yating Ren
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Tao Hong
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Dingqi Lu
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Fan Zhang
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Yi Cao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Xinchang Wang
- Department of Rheumatology, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China,Correspondence: Xinchang Wang, Department of Rheumatology, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China, Tel +86 0571-85288249, Email
| |
Collapse
|
7
|
Zhou L, Fan R, Luo Y, Zhang C, Jia D, Wang R, Zeng Y, Ren M, Du K, Pan W, Yang J, Tian F, Gu C. A Metabolism-Related Gene Landscape Predicts Prostate Cancer Recurrence and Treatment Response. Front Immunol 2022; 13:837991. [PMID: 35359973 PMCID: PMC8960425 DOI: 10.3389/fimmu.2022.837991] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/11/2022] [Indexed: 11/22/2022] Open
Abstract
Background Prostate cancer (PCa) is the most common malignant tumor in men. Although clinical treatments of PCa have made great progress in recent decades, once tolerance to treatments occurs, the disease progresses rapidly after recurrence. PCa exhibits a unique metabolic rewriting that changes from initial neoplasia to advanced neoplasia. However, systematic and comprehensive studies on the relationship of changes in the metabolic landscape of PCa with tumor recurrence and treatment response are lacking. We aimed to construct a metabolism-related gene landscape that predicts PCa recurrence and treatment response. Methods In the present study, we used differentially expressed gene analysis, protein–protein interaction (PPI) networks, univariate and multivariate Cox regression, and least absolute shrinkage and selection operator (LASSO) regression to construct and verify a metabolism-related risk model (MRM) to predict the disease-free survival (DFS) and response to treatment for PCa patients. Results The MRM predicted patient survival more accurately than the current clinical prognostic indicators. By using two independent PCa datasets (International Cancer Genome Consortium (ICGC) PCa and Taylor) and actual patients to test the model, we also confirmed that the metabolism-related risk score (MRS) was strongly related to PCa progression. Notably, patients in different MRS subgroups had significant differences in metabolic activity, mutant landscape, immune microenvironment, and drug sensitivity. Patients in the high-MRS group were more sensitive to immunotherapy and endocrine therapy, while patients in the low-MRS group were more sensitive to chemotherapy. Conclusions We developed an MRM, which might act as a clinical feature to more accurately assess prognosis and guide the selection of appropriate treatment for PCa patients. It is promising for further application in clinical practice.
Collapse
Affiliation(s)
- Lijie Zhou
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruixin Fan
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongbo Luo
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cai Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Donghui Jia
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rongli Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiao tong University, Xi'an, China
| | - Youmiao Zeng
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengda Ren
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaixuan Du
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenbang Pan
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinjian Yang
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fengyan Tian
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaohui Gu
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Urology, Henan Institute of Urology and Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Briffa JF, Bevens W, Gravina S, Said JM, Wlodek ME. Pregnant biglycan knockout mice have altered cardiorenal adaptations and a shorter gestational length, but do not develop a pre-eclamptic phenotype. Placenta 2022; 119:52-62. [PMID: 35150975 DOI: 10.1016/j.placenta.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Pre-eclampsia complicates 4.6% of pregnancies and is linked to impaired placentation; likely due to dysregulated vasculogenesis/angiogenesis. Proteoglycans, such as biglycan, are located on the endothelial surface of fetal capillaries. Biglycan is reduced in the placenta of pregnancies complicated by fetal growth restriction and pre-eclampsia. Importantly, biglycan stimulates angiogenesis in numerous tissues. Therefore, this study investigated whether biglycan knockdown in mice results in a pre-eclamptic phenotype. METHODS Wild-type (WT) and Bgn-/- mice underwent cardiorenal measurements prior to and during pregnancy. One cohort of mice underwent post-mortem on gestational day 18 (E18) and another cohort underwent post-mortem on postnatal day 1 (PN1), with maternal and offspring tissues of relevance collected. RESULTS Bgn-/- dams had increased heart rate (+9%, p < 0.037) and reduced systolic (-11%, p < 0.001), diastolic (-15%, p < 0.001), and mean arterial (-12%, p < 0.001) pressures at all ages investigated compared to WT. Additionally, Bgn-/- dams had reduced urine flow rate (-64%, p < 0.001) as well as reduced urinary excretions (-49%, p < 0.004) during late gestation compared to WT. Bgn-/- pups had higher body weight (+8%, p = 0.004; E18 only) and a higher liver-to-brain weight ratio (+43%, p < 0.001). Placental weight was unaltered with only minor changes in vasculogenic and angiogenic gene abundances detected, which did not correlate to changes in protein expression. DISCUSSION This study demonstrated that total knockdown of biglycan is not associated with features of pre-eclampsia.
Collapse
Affiliation(s)
- J F Briffa
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - W Bevens
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - S Gravina
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - J M Said
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, 3010, Australia; Maternal Fetal Medicine, Sunshine Hospital, Western Health, St Albans, VIC, 3021, Australia
| | - M E Wlodek
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
9
|
Appunni S, Rubens M, Ramamoorthy V, Anand V, Khandelwal M, Sharma A. Biglycan: an emerging small leucine-rich proteoglycan (SLRP) marker and its clinicopathological significance. Mol Cell Biochem 2021; 476:3935-3950. [PMID: 34181183 DOI: 10.1007/s11010-021-04216-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/23/2021] [Indexed: 12/22/2022]
Abstract
Extracellular matrix (ECM) plays an important role in the structural organization of tissue and delivery of external cues to the cell. Biglycan, a class I small leucine-rich proteoglycans (SLRP), is a key component of the ECM that participates in scaffolding the collagen fibrils and mediates cell signaling. Dysregulation of biglycan expression can result in wide range of clinical conditions such as metabolic disorder, inflammatory disorder, musculoskeletal defects and malignancies. In this review, we aim to update our current understanding regarding the link between altered expression of biglycan and different clinicopathological states. Biglycan interacts with toll like receptors (TLR)-2 and TLR-4 on the immune cells which initiates inflammation and aggravates inflammatory disorders. ECM unbound soluble biglycan acts as a DAMP (danger associated molecular pattern) resulting in sterile inflammation. Dysregulation of biglycan expression is also observed in inflammatory metabolic conditions such as atherosclerosis and obesity. In cancer, high-biglycan expression facilitates tumor growth, invasion and metastasis which is associated with poor clinical outcome. As a pivotal structural component of the ECM, biglycan strengthens the musculoskeletal system and its absence is associated with musculoskeletal defects. Thus, SLRP biglycan is a potential marker which is significantly altered in different clinicopathological states.
Collapse
Affiliation(s)
- Sandeep Appunni
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110 029, India
- Government Medical College, Kozhikode, Kerala, India
| | | | | | | | - Madhuram Khandelwal
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110 029, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110 029, India.
| |
Collapse
|
10
|
Zheng Y, Chen ZY, Ma WJ, Wang QZ, Liang H, Ma AG. B Vitamins Supplementation Can Improve Cognitive Functions and May Relate to the Enhancement of Transketolase Activity in A Rat Model of Cognitive Impairment Associated with High-fat Diets. Curr Med Sci 2021; 41:847-856. [PMID: 34652631 DOI: 10.1007/s11596-021-2456-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/05/2020] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To determine whether B vitamin treatment was sufficient to reduce cognitive impairment associated with high-fat diets in rats and to modulate transketolase (TK) expression and activity. METHODS To test this, we separated 50 rats into five groups that were either fed a standard chow diet (controls) or a high-fat diet (experimental groups H0, H1, H2, and H3). H0 group animals received no additional dietary supplementation, while H1 group animals were administered 100 mg/kg body weight (BW) thiamine, 100 mg/kg BW riboflavin, and 250 mg/kg BW niacin each day, and group H2 animals received daily doses of 100 mg/kg BW pyridoxine, 100 mg/kg BW cobalamin, and 5 mg/kg BW folate. Animals in the H3 group received the B vitamin regimens administered to both H1 and H2 each day. RESULTS Over time, group H0 exhibited greater increases in BW and fat mass relative to other groups. When spatial and memory capabilities in these animals were evaluated via conditioned taste aversion (CTA) and Morris Water Maze (MWM), we found B vitamin treatment was associated with significant improvements relative to untreated H0 controls. Similarly, B vitamin supplementation was associated with elevated TK expression in erythrocytes and hypothalamus of treated animals relative to those in H0 (P<0.05). CONCLUSION Together, these findings suggest B vitamin can modulate hypothalamic TK activity to reduce the severity of cognitive deficits in a rat model of obesity. As such, B vitamin supplementation may be a beneficial method for reducing cognitive dysfunction in clinical settings associated with high-fat diets.
Collapse
Affiliation(s)
- Ying Zheng
- Department of Nutrition, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Zhi-Yong Chen
- Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510080, China
| | - Wen-Jun Ma
- Department of Nutrition, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Qiu-Zhen Wang
- Institute of Nutrition and Health, Medical College of Qingdao University, Qingdao, 266021, China
| | - Hui Liang
- Institute of Nutrition and Health, Medical College of Qingdao University, Qingdao, 266021, China
| | - Ai-Guo Ma
- Institute of Nutrition and Health, Medical College of Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
11
|
Drulis-Fajdasz D, Gostomska-Pampuch K, Duda P, Wiśniewski JR, Rakus D. Quantitative Proteomics Reveals Significant Differences between Mouse Brain Formations in Expression of Proteins Involved in Neuronal Plasticity during Aging. Cells 2021; 10:2021. [PMID: 34440790 PMCID: PMC8393337 DOI: 10.3390/cells10082021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/05/2021] [Indexed: 12/22/2022] Open
Abstract
Aging is associated with a general decline in cognitive functions, which appears to be due to alterations in the amounts of proteins involved in the regulation of synaptic plasticity. Here, we present a quantitative analysis of proteins involved in neurotransmission in three brain regions, namely, the hippocampus, the cerebral cortex and the cerebellum, in mice aged 1 and 22 months, using the total protein approach technique. We demonstrate that although the titer of some proteins involved in neurotransmission and synaptic plasticity is affected by aging in a similar manner in all the studied brain formations, in fact, each of the formations represents its own mode of aging. Generally, the hippocampal and cortical proteomes are much more unstable during the lifetime than the cerebellar proteome. The data presented here provide a general picture of the effect of physiological aging on synaptic plasticity and might suggest potential drug targets for anti-aging therapies.
Collapse
Affiliation(s)
- Dominika Drulis-Fajdasz
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| | - Kinga Gostomska-Pampuch
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; (K.G.-P.); (J.R.W.)
- Department of Biochemistry and Immunochemistry, Wrocław Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland
| | - Przemysław Duda
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| | - Jacek Roman Wiśniewski
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; (K.G.-P.); (J.R.W.)
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| |
Collapse
|
12
|
Chung I, Kim SA, Kim S, Lee JO, Park CY, Lee J, Kang J, Lee JY, Seo I, Lee HJ, Han JA, Kang MJ, Lim E, Kim SJ, Wu SW, Oh JY, Chung JH, Kim EK, Kim HS, Shin MJ. Biglycan reduces body weight by regulating food intake in mice and improves glucose metabolism through AMPK/AKT dual pathways in skeletal muscle. FASEB J 2021; 35:e21794. [PMID: 34314059 DOI: 10.1096/fj.202002039rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 06/11/2021] [Accepted: 06/28/2021] [Indexed: 12/25/2022]
Abstract
While biglycan (BGN) is suggested to direct diverse signaling cascades, the effects of soluble BGN as a ligand on metabolic traits have not been studied. Herein, we tested the effects of BGN on obesity in high-fat diet (HFD)-induced obese animals and glucose metabolism, with the underlying mechanism responsible for observed effects in vitro. Our results showed that BGN administration (1 mg/kg body weight, intraperitoneally) significantly prevented HFD-induced obesity, and this was mainly attributed to reduced food intake. Also, intracerebroventricular injection of BGN reduced food intake and body weight. The underlying mechanism includes modulation of neuropeptides gene expression involved in appetite in the hypothalamus in vitro and in vivo. In addition, BGN regulates glucose metabolism as shown by improved glucose tolerance in mice as well as AMPK/AKT dual pathway-driven enhanced glucose uptake and GLUT4 translocation in L6 myoblast cells. In conclusion, our results suggest BGN as a potential therapeutic target to treat risk factors for metabolic diseases.
Collapse
Affiliation(s)
- InHyeok Chung
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Republic of Korea
| | - Shin Ae Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Seolsong Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Jung Ok Lee
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Clara Yongjoo Park
- Department of Food and Nutrition, Chonnam National University, Gwangju, Republic of Korea
| | - Juhee Lee
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Republic of Korea
| | - Jun Kang
- Department of Biotechnology, CHA University, Gyeonggi-do, Republic of Korea
| | - Jin Young Lee
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Republic of Korea
| | - Ilhyeok Seo
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hye Jeong Lee
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jeong Ah Han
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Min Ju Kang
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Eunice Lim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Su Jin Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Sang Woo Wu
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Joo Yeon Oh
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ji Hyung Chung
- Department of Biotechnology, CHA University, Gyeonggi-do, Republic of Korea
| | - Eun-Kyoung Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.,Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Hyeon Soo Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Min-Jeong Shin
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Republic of Korea.,School of Biosystems and Biomedical Sciences, College of Health Science, Korea University, Seoul, Republic of Korea
| |
Collapse
|
13
|
Hayes AJ, Melrose J. Neural Tissue Homeostasis and Repair Is Regulated via CS and DS Proteoglycan Motifs. Front Cell Dev Biol 2021; 9:696640. [PMID: 34409033 PMCID: PMC8365427 DOI: 10.3389/fcell.2021.696640] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/13/2021] [Indexed: 01/04/2023] Open
Abstract
Chondroitin sulfate (CS) is the most abundant and widely distributed glycosaminoglycan (GAG) in the human body. As a component of proteoglycans (PGs) it has numerous roles in matrix stabilization and cellular regulation. This chapter highlights the roles of CS and CS-PGs in the central and peripheral nervous systems (CNS/PNS). CS has specific cell regulatory roles that control tissue function and homeostasis. The CNS/PNS contains a diverse range of CS-PGs which direct the development of embryonic neural axonal networks, and the responses of neural cell populations in mature tissues to traumatic injury. Following brain trauma and spinal cord injury, a stabilizing CS-PG-rich scar tissue is laid down at the defect site to protect neural tissues, which are amongst the softest tissues of the human body. Unfortunately, the CS concentrated in gliotic scars also inhibits neural outgrowth and functional recovery. CS has well known inhibitory properties over neural behavior, and animal models of CNS/PNS injury have demonstrated that selective degradation of CS using chondroitinase improves neuronal functional recovery. CS-PGs are present diffusely in the CNS but also form denser regions of extracellular matrix termed perineuronal nets which surround neurons. Hyaluronan is immobilized in hyalectan CS-PG aggregates in these perineural structures, which provide neural protection, synapse, and neural plasticity, and have roles in memory and cognitive learning. Despite the generally inhibitory cues delivered by CS-A and CS-C, some CS-PGs containing highly charged CS disaccharides (CS-D, CS-E) or dermatan sulfate (DS) disaccharides that promote neural outgrowth and functional recovery. CS/DS thus has varied cell regulatory properties and structural ECM supportive roles in the CNS/PNS depending on the glycoform present and its location in tissue niches and specific cellular contexts. Studies on the fruit fly, Drosophila melanogaster and the nematode Caenorhabditis elegans have provided insightful information on neural interconnectivity and the role of the ECM and its PGs in neural development and in tissue morphogenesis in a whole organism environment.
Collapse
Affiliation(s)
- Anthony J. Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Wales, United Kingdom
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital and The Faculty of Medicine and Health, The University of Sydney, St. Leonard’s, NSW, Australia
| |
Collapse
|
14
|
Gomez-Pinilla F, Cipolat RP, Royes LFF. Dietary fructose as a model to explore the influence of peripheral metabolism on brain function and plasticity. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166036. [PMID: 33508421 DOI: 10.1016/j.bbadis.2020.166036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
Abstract
High consumption of fructose has paralleled an explosion in metabolic disorders including obesity and type 2 diabetes. Even more problematic, sustained consumption of fructose is perceived as a threat for brain function and development of neurological disorders. The action of fructose on peripheral organs is an excellent model to understand how systemic physiology impacts the brain. Given the recognized action of fructose on liver metabolism, here we discuss mechanisms by which fructose can impact the brain by interacting with liver and other organs. The interaction between peripheral and central mechanisms is a suitable target to reduce the pathophysiological consequences of neurological disorders.
Collapse
Affiliation(s)
- Fernando Gomez-Pinilla
- Department of Neurosurgery, UCLA Brain Injury Research Center, University of California Los Angeles, USA; Department of Integrative Biology and Physiology, UCLA Brain Injury Research Center, University of California Los Angeles, USA.
| | - Rafael Parcianello Cipolat
- Exercise Biochemistry Laboratory, Center of Physical Education and Sports, Federal University of Santa Maria - UFSM, Santa Maria, RS, Brazil
| | - Luiz Fernando Freire Royes
- Exercise Biochemistry Laboratory, Center of Physical Education and Sports, Federal University of Santa Maria - UFSM, Santa Maria, RS, Brazil
| |
Collapse
|
15
|
Chen S, Guo D, Lei B, Bi J, Yang H. Biglycan protects human neuroblastoma cells from nitric oxide-induced death by inhibiting AMPK-mTOR mediated autophagy and intracellular ROS level. Biotechnol Lett 2020; 42:657-668. [PMID: 31989342 DOI: 10.1007/s10529-020-02818-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 01/22/2020] [Indexed: 02/08/2023]
Abstract
The ubiquitous proteoglycan, biglycan (BGN) acts as an important modulator, regulating key molecular pathways of metabolism and brain function. Autophagy is documented as a defining feature of neurodegeneration in Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). In the present study, we found that BGN protected neuronal cells from nitric oxide (NO)-induced cell apoptosis. However, it is still unclear that whether the neuroprotective effect of BGN relates to autophagy. Here, we discovered that an NO donor, sodium nitroprusside (SNP) induced autophagy in human SH-SY5Y neuroblastoma cells, including activating LC3B and inhibiting p62. Inhibiting autophagy by 3MA aggravated NO-induced cell death, otherwise promoting autophagy by Rapamycin rescued NO-triggered cell death. Notably, BGN downregulated by NO, significantly protected SH-SY5Y cells against NO-induced neurotoxicity by inhibiting the activation of autophagy-dependent AMPK signaling pathway. Moreover, BGN overexpression also diminished NO-induced the elevation of intracellular reactive oxygen species (ROS) level, but not NO content. These findings suggest that BGN protects neuroblastoma cells from NO-induced death by suppressing autophagy-dependent AMPK-mTOR signaling and intracellular ROS level.
Collapse
Affiliation(s)
- Sujuan Chen
- Synthetic Biology Engineering Lab of Henan Province, School of Sciences and Technology, Xinxiang Medical University, Henan, China.
| | - Dandan Guo
- Synthetic Biology Engineering Lab of Henan Province, School of Sciences and Technology, Xinxiang Medical University, Henan, China.,Shangai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Bingbing Lei
- School of Sciences and Technology, Xinxiang Medical University, Henan, China
| | - Jiajia Bi
- Synthetic Biology Engineering Lab of Henan Province, School of Sciences and Technology, Xinxiang Medical University, Henan, China
| | - Haijie Yang
- School of Sciences and Technology, Xinxiang Medical University, Henan, China
| |
Collapse
|
16
|
Zhang G, Byun HR, Ying Z, Blencowe M, Zhao Y, Hong J, Shu L, Chella Krishnan K, Gomez-Pinilla F, Yang X. Differential metabolic and multi-tissue transcriptomic responses to fructose consumption among genetically diverse mice. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165569. [PMID: 31669422 PMCID: PMC6993985 DOI: 10.1016/j.bbadis.2019.165569] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/05/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
Understanding how individuals react differently to the same treatment is a major concern in precision medicine. Metabolic challenges such as the one posed by high fructose intake are important determinants of disease mechanisms. We embarked on studies to determine how fructose affects differential metabolic dysfunctions across genetically dissimilar mice, namely, C57BL/6 J (B6), DBA/2 J (DBA) and FVB/NJ (FVB), by integrating physiological and gene regulatory mechanisms. We report that fructose has strain-specific effects, involving tissue-specific gene regulatory cascades in hypothalamus, liver, and white adipose tissues. DBA mice showed the largest numbers of genes associated with adiposity, congruent with their highest susceptibility to adiposity gain and glucose intolerance across the three tissues. In contrast, B6 and FVB mainly exhibited cholesterol phenotypes, accompanying the largest number of adipose genes correlating with total cholesterol in B6, and liver genes correlating with LDL in FVB mice. Tissue-specific network modeling predicted strain-and tissue-specific regulators such as Fgf21 (DBA) and Lss (B6), which were subsequently validated in primary hepatocytes. Strain-specific fructose-responsive genes revealed susceptibility for human diseases such that genes in liver and adipose tissue in DBA showed strong enrichment for human type 2 diabetes and obesity traits. Liver and adipose genes in FVB were mostly related to lipid traits, and liver and adipose genes in B6 showed relevance to most cardiometabolic traits tested. Our results show that fructose induces gene regulatory pathways that are tissue specific and dependent on the genetic make-up, which may underlie interindividual variability in cardiometabolic responses to high fructose consumption.
Collapse
Affiliation(s)
- Guanglin Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Hyae Ran Byun
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Zhe Ying
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Montgomery Blencowe
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Yuqi Zhao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Jason Hong
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Le Shu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Karthick Chella Krishnan
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA; Department of Neurosurgery, UCLA Brain Injury Research Center, University of California, Los Angeles, Los Angeles, California 90095, USA.
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, California 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California 90095, USA.
| |
Collapse
|
17
|
Blencowe M, Karunanayake T, Wier J, Hsu N, Yang X. Network Modeling Approaches and Applications to Unravelling Non-Alcoholic Fatty Liver Disease. Genes (Basel) 2019; 10:E966. [PMID: 31771247 PMCID: PMC6947017 DOI: 10.3390/genes10120966] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/18/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive condition of the liver encompassing a range of pathologies including steatosis, non-alcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma. Research into this disease is imperative due to its rapid growth in prevalence, economic burden, and current lack of FDA approved therapies. NAFLD involves a highly complex etiology that calls for multi-tissue multi-omics network approaches to uncover the pathogenic genes and processes, diagnostic biomarkers, and potential therapeutic strategies. In this review, we first present a basic overview of disease pathogenesis, risk factors, and remaining knowledge gaps, followed by discussions of the need and concepts of multi-tissue multi-omics approaches, various network methodologies and application examples in NAFLD research. We highlight the findings that have been uncovered thus far including novel biomarkers, genes, and biological pathways involved in different stages of NAFLD, molecular connections between NAFLD and its comorbidities, mechanisms underpinning sex differences, and druggable targets. Lastly, we outline the future directions of implementing network approaches to further improve our understanding of NAFLD in order to guide diagnosis and therapeutics.
Collapse
Affiliation(s)
- Montgomery Blencowe
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA; (M.B.); (T.K.); (J.W.); (N.H.)
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - Tilan Karunanayake
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA; (M.B.); (T.K.); (J.W.); (N.H.)
| | - Julian Wier
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA; (M.B.); (T.K.); (J.W.); (N.H.)
| | - Neil Hsu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA; (M.B.); (T.K.); (J.W.); (N.H.)
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA; (M.B.); (T.K.); (J.W.); (N.H.)
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA
- Interdepartmental Program of Bioinformatics, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| |
Collapse
|