1
|
Lin Y, Lin Y, Zhou X, Ren A, Li X. Progress in the genetics and epigenetics of pelvic floor disorder. Gene 2025; 943:149277. [PMID: 39880343 DOI: 10.1016/j.gene.2025.149277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/14/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Pelvic floor disorder (PFD) is a common gynecological disorder, and with the ageing of the population, PFD has a serious impact on the physical and mental health of patients and their quality of life. The most prominent of these are pelvic organ prolapse (POP) and urinary incontinence (UI), about which the etiology is still unclear, and it is urgent to explore their pathogenesis. Advances in genetics and epigenetics have provided new insights into the pathophysiology of PFD. Candidate genes and genome-wide association studies have identified susceptibility genes for POP and UI. These susceptibility genes typically promote POP by affecting pelvic floor connective tissue. The role of susceptibility genes in UI is multifactorial and includes promoting inflammation, damaging pelvic floor connective tissue, and modulating neurogenic effects. The association of epigenetic changes with POP and UI has also been investigated. DNA methylation studies have identified several important pathways associated with POP. miRNAs play an important role in the development of POP and UI, and this may be an important therapeutic direction for the future. The studies conducted so far have shown that genetic and epigenetic techniques are of great importance in exploring the etiology of PFD and that more in-depth studies are needed in the future.
Collapse
Affiliation(s)
- Ya Lin
- Fourth Clinical Medical College Of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China.
| | - Yaoxiang Lin
- Hangzhou Normal University, Hangzhou, Zhejiang, China.
| | - Xiaojing Zhou
- Fourth Clinical Medical College Of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China.
| | - An Ren
- Hangzhou Normal University, Hangzhou, Zhejiang, China.
| | - Xiangjuan Li
- Hangzhou Women's Hospital, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Johnston CD, Pang APS, Siegler EL, Thomas C, Burchett CO, Crowley M, O'Brien R, Ndhlovu LC, Glesby MJ, Corley MJ. Sex differences in epigenetic ageing for older people living with HIV. EBioMedicine 2025; 113:105588. [PMID: 39923742 PMCID: PMC11849644 DOI: 10.1016/j.ebiom.2025.105588] [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: 04/19/2024] [Revised: 12/05/2024] [Accepted: 01/21/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND HIV-1 infection impacts biological ageing, and epigenetic clocks highlight epigenetic age acceleration in people with HIV. Despite evidence indicating sex differences in clinical, immunological, and virological measures, females have been underrepresented in most HIV epigenetic studies. Hence, we generated a more representative epigenetic dataset to examine sex differences in epigenetic ageing and relationships to clinical phenotypes and proteomics. METHODS We calculated first, second, and third-generation epigenetic ages using DNA methylation data in an observational cohort of 52 females and 106 males with HIV age 50 and over. We profiled plasma biomarkers with Olink high-throughput proteomics to test associations with epigenetic age acceleration. Survival was ascertained over 5 years. FINDINGS Epigenetic age acceleration measured by three principal-component based chronological epigenetic age clocks (p = 0.0029, 0.021, 0.010) and one epigenetic mortality risk clock was significantly lower in females living with HIV compared to males (p = 0.0011). Additionally, sex was significantly associated with epigenetic biomarker scores for proportion of naïve CD4+ T cells (p = 0.0006), physical fitness including DNAmGait (p = 0.0010), DNAmGrip (p < 0.0001), and DNAmV02 max (p < 0.0001). We found epigenetic age acceleration associated with plasma proteomic markers involved in inflammation, senescence, immune regulation, kidney function, and tissue homoeostasis (p < 0.0001). Higher epigenetic frailty risk scores were associated with lower CD4 T cell counts (p = 0.0072) and lower CD4/CD8 ratio (p = 0.0017). Slower gait (p = 0.0017), greater frailty (p = 0.0074), and history of smoking (p = 0.042) were associated with increased DNAmFitAge. Risk of death was increased in females with PCPhenoAge acceleration over a 5-year timespan compared to men with PCPhenoAge acceleration (p = 0.03). INTERPRETATION These results highlight the importance of studying sex-specific differences in epigenetic ageing biomarkers for HIV-related geroscience research. FUNDING National Institute on Aging (K23AG072960), National Center for Advancing Translational Sciences (UL1TR000457), National Institute of Mental Health (R21 MH115821).
Collapse
Affiliation(s)
- Carrie D Johnston
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York City, New York, USA
| | - Alina P S Pang
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York City, New York, USA
| | - Eugenia L Siegler
- Department of Medicine, Division of Geriatrics and Palliative Medicine, Weill Cornell Medicine, New York City, New York, USA
| | - Charlene Thomas
- Department of Population Health Sciences, Weill Cornell Medicine, New York City, New York, USA
| | - Chelsie O Burchett
- Department of Medicine, Division of Geriatrics and Palliative Medicine, Weill Cornell Medicine, New York City, New York, USA
| | - Mia Crowley
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York City, New York, USA
| | - Rochelle O'Brien
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York City, New York, USA
| | - Lishomwa C Ndhlovu
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York City, New York, USA
| | - Marshall J Glesby
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York City, New York, USA
| | - Michael J Corley
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York City, New York, USA.
| |
Collapse
|
3
|
Von Ruff ZD, Miller MJ, Moro T, Reidy PT, Ebert SM, Volpi E, Adams CM, Rasmussen BB. Resistance exercise training in older men reduces ATF4-activated and senescence-associated mRNAs in skeletal muscle. GeroScience 2025:10.1007/s11357-025-01564-2. [PMID: 40011348 DOI: 10.1007/s11357-025-01564-2] [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: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
Sarcopenia increases the risk of frailty, morbidity, and mortality in older adults. Resistance exercise training improves muscle size and function; however, the response to exercise training is variable in older adults. The objective of our study was to determine both the age-independent and age-dependent changes to the transcriptome following progressive resistance exercise training. Skeletal muscle biopsies were obtained before and after 12 weeks of resistance exercise training in 8 young (24 ± 3.3 years) and 10 older (72 ± 4.9 years) men. RNA was extracted from each biopsy and prepared for analysis via RNA sequencing. We performed differential mRNA expression, gene ontology, and gene set enrichment analyses. We report that when comparing post-training vs pre-training 226 mRNAs and 959 mRNAs were differentially expressed in the skeletal muscle of young and older men, respectively. Additionally, 94 mRNAs increased, and 17 mRNAs decreased in both young and old, indicating limited overlap in response to resistance exercise training. Furthermore, the differential gene expression was larger in older skeletal muscle. Finally, we report three novel findings: 1) resistance exercise training decreased the abundance of ATF4-activated and senescence-associated skeletal muscle mRNAs in older men; 2) resistance exercise-induced increases in lean mass correlate with increased mRNAs encoding mitochondrial proteins; and 3) increases in muscle strength following resistance exercise positively correlate with increased mRNAs involved in translation, rRNA processing, and polyamine metabolism. We conclude that resistance exercise training elicits a differential gene expression response in young and old skeletal muscle, including reduced ATF-4 activated and senescence-associated gene expression.
Collapse
Affiliation(s)
| | - Matthew J Miller
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- University of Iowa, Iowa City, IA, USA
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Paul T Reidy
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, OH, USA
| | - Scott M Ebert
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Elena Volpi
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA
| | - Christopher M Adams
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Blake B Rasmussen
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA.
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA.
| |
Collapse
|
4
|
Potluri T, You T, Yin P, Coon J, Stulberg JJ, Dai Y, Escobar DJ, Lieber RL, Zhao H, Bulun SE. Estrogen receptor-α ablation reverses muscle fibrosis and inguinal hernias. J Clin Invest 2025; 135:e179137. [PMID: 39903526 PMCID: PMC11910215 DOI: 10.1172/jci179137] [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: 01/05/2024] [Accepted: 01/24/2025] [Indexed: 02/06/2025] Open
Abstract
Fibrosis of the lower abdominal muscle (LAM) contributes to muscle weakening and inguinal hernia formation, an ailment that affects a noteworthy 50% of men by age 75 and necessitates surgical correction as the singular therapy. Despite its prevalence, the mechanisms driving LAM fibrosis and hernia development remain poorly understood. Using a humanized mouse model that replicates the elevated skeletal muscle tissue estrogen concentrations seen in aging men, we identified estrogen receptor-α (ESR1) as a key driver of LAM fibroblast proliferation, extracellular matrix deposition, and hernia formation. Fibroblast-specific ESR1 ablation effectively prevented muscle fibrosis and herniation, while pharmacological ESR1 inhibition with fulvestrant reversed hernias and restored normal muscle architecture. Multiomics analyses of in vitro LAM fibroblasts from humanized mice unveiled an estrogen/ESR1-mediated activation of a distinct profibrotic cistrome and gene expression signature, concordant with observations in inguinal hernia tissues in human males. Our findings hold significant promise for prospective medical interventions targeting fibrotic conditions and present non-surgical avenues for addressing inguinal hernias.
Collapse
Affiliation(s)
- Tanvi Potluri
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Tianming You
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ping Yin
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - John Coon
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jonah J. Stulberg
- Department of Surgery, McGovern Medical School at the University of Texas Health Sciences Center, Houston, Texas, USA
| | - Yang Dai
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois, USA
| | | | - Richard L. Lieber
- Departments of Physical Medicine and Rehabilitation and Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
- Research Service, Hines VA Medical Center, Maywood, Illinois, USA
- Shirley Ryan AbilityLab, Chicago, Illinois, USA
| | - Hong Zhao
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Serdar E. Bulun
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
5
|
Gharpure M, Vyavahare S, Asante DM, Chen J, Zhong R, Cooley MA, Deak F, Lu XY, Isales CM, Fulzele S. Sex-specific transcriptomic profiling reveals key players in bone loss associated with Alzheimer's disease. GeroScience 2025:10.1007/s11357-025-01535-7. [PMID: 39875754 DOI: 10.1007/s11357-025-01535-7] [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: 11/06/2024] [Accepted: 01/17/2025] [Indexed: 01/30/2025] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is frequently associated with musculoskeletal complications, including sarcopenia and osteoporosis, which substantially impair patient quality of life. Despite these clinical observations, the molecular mechanisms linking AD to bone loss remain insufficiently explored. In this study, we examined the femoral bone microarchitecture and transcriptomic profiles of APP/PS1 transgenic mouse models of AD to elucidate the disease's impact on bone pathology and identify potential gene candidates associated with bone deterioration. We performed micro-computed tomography (microCT) and RNA transcriptome analysis on the femoral bone of these mice. We observed a significant reduction in bone microstructure in both male and female APP/PS1 mice compared to their wild-type counterparts. Transcriptomic analysis of femoral bone tissue revealed substantial differential gene expression between AD mice and controls. Specifically, APP/PS1 mice exhibited differential expression in 289 protein-coding genes across both sexes. Notably, in female APP/PS1 mice, 664 genes were differentially expressed, with key genes such as Shh, Efemp1, Arg1, EphA2, Irx1, and PORCN potentially implicated in bone loss. In male APP/PS1 mice, 787 genes were differentially expressed, with Sel1l, Ffar4, Hspa1a, AMH, WFS1, and CLIC1 emerging as notable candidates in the context of bone deterioration. Gene Ontology (GO) enrichment analysis further revealed distinct sex-specific gene pathways between male and female APP/PS1 mice, underscoring the differential molecular underpinnings of bone pathology in AD. This study identifies novel sex-specific genes in the APP/PS1 mouse model and proposes potential therapeutic targets to mitigate bone loss in AD patients.
Collapse
Affiliation(s)
- Mohini Gharpure
- Division of Endocrinology, Department of Medicine, Augusta University, Augusta, GA, USA
| | - Sagar Vyavahare
- Division of Endocrinology, Department of Medicine, Augusta University, Augusta, GA, USA
| | - Diana M Asante
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Jie Chen
- Division of Biostatistics, Data Science, Augusta University, Augusta, GA, USA
| | - Roger Zhong
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA
| | - Marion A Cooley
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA
| | - Ferenc Deak
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA
| | - Xin-Yun Lu
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA
| | - Carlos M Isales
- Division of Endocrinology, Department of Medicine, Augusta University, Augusta, GA, USA
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA
- Department of Neuroscience & Regenerative Medicine, Augusta, GA, 30912, USA
| | - Sadanand Fulzele
- Division of Endocrinology, Department of Medicine, Augusta University, Augusta, GA, USA.
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA.
- Department of Neuroscience & Regenerative Medicine, Augusta, GA, 30912, USA.
- Center for Healthy Aging, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
6
|
Gupta K, Llewellyn J, Roberts E, Liu C, Naji A, Assoian RK, Wells RG. Biliary atresia susceptibility gene EFEMP1 regulates extrahepatic bile duct elastic fiber formation and mechanics. JHEP Rep 2025; 7:101215. [PMID: 39717503 PMCID: PMC11663959 DOI: 10.1016/j.jhepr.2024.101215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 12/25/2024] Open
Abstract
Background & Aims EGF-containing fibulin extracellular matrix protein 1 (EFEMP1, also called fibulin-3) is an extracellular matrix protein linked in a genome-wide association study to biliary atresia, a fibrotic disease of the neonatal extrahepatic bile duct. Fibulin-3 is deposited in most tissues and Efemp1 null mice have decreased elastic fibers in visceral fascia; however, fibulin-3 does not have a role in the development of large elastic fibers and its overall function in the extrahepatic bile ducts remains unclear. Methods We used staining and histology to define the amount and organization of key extracellular matrix components in the extrahepatic bile ducts. We also repurposed pressure myography, a technique heretofore applied to the vasculature, to determine the contribution of elastin and fibulin-3 to extrahepatic bile duct mechanics. We examined extrahepatic bile duct structure and mechanics in three models: neonatal vs. adult rat ducts (n = 6 each), elastase-treated adult rat ducts (n = 6-7 each), and Efemp1 +/- vs. wild-type mouse ducts (n = 6 each). Results We demonstrated that fibulin-3 is expressed in the submucosa of both neonatal and adult mouse, rat and human extrahepatic bile ducts and that, in adult Efemp1 +/- mouse ducts, elastin organization into fibers is decreased by approximately half. Pressure myography showed that Efemp1 +/- ducts have altered mechanics compared to control ducts, with Efemp1 +/- ducts displaying significant stretch compared to controls (p = 0.0376); these changes in stretch are similar to those observed in elastase-treated vs. normal ducts (p <0.0001) and in neonatal ducts vs. adult ducts (p <0.0001). Conclusion Fibulin-3 has an important role in the formation of elastic fibers and the mechanical properties of the extrahepatic bile duct. This provides functional relevance for the biliary atresia susceptibility gene EFEMP1. Impact and implications The gene EFEMP1 was found via a genome-wide association study to be a susceptibility gene for the neonatal disease biliary atresia. EFEMP1 encodes the protein fibulin-3, which regulates elastic fiber organization in the extrahepatic bile duct (EHBD), the major site of disease in biliary atresia. We showed that neonatal EHBDs as well as mice heterozygous for Efemp1 have decreased numbers of elastic fibers, and that this alters EHBD mechanics. This work is important for understanding the mechanism of biliary atresia, in particular susceptibility to obstruction.
Collapse
Affiliation(s)
- Kapish Gupta
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica Llewellyn
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Emilia Roberts
- Program in Translational Biomechanics, Institute for Translational Medicine and Therapeutics, University of Pennsylvania, PA, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania, PA, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania, PA, USA
| | - Richard K. Assoian
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
- Program in Translational Biomechanics, Institute for Translational Medicine and Therapeutics, University of Pennsylvania, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, PA, USA
| | - Rebecca G. Wells
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Chervinskaya I, Kuprina NI, Kruglikov I. A Retrospective Pragmatic Longitudinal Case-Series Clinical Study to Evaluate the Clinical Outcome of Triple-Frequency Ultrasound in Treatment of Cellulite. Clin Cosmet Investig Dermatol 2024; 17:2779-2794. [PMID: 39660032 PMCID: PMC11630702 DOI: 10.2147/ccid.s488977] [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: 10/15/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024]
Abstract
Objective In this single-centre, retrospective, pragmatic, longitudinal case-series clinical study triple-frequency LDM (TF-LDM) technology with frequencies of 1/3/10 MHz and 3/10/19 MHz was applied for treatment of cellulite to reveal the effect of these waves on the cellulite skin and assess the sustainability of treatment outcomes during the long-term follow-up controls. Methods Twenty Caucasian females with mild-to-severe gynoid lipodystrophy aged 27-53 years who received cellulite monotherapy with TF-LDM were included in this study. All participants were evaluated at three time points: baseline (T1), on the day of the last treatment (T2), and during the last follow-up (T3). Cellulite severity was assessed by six independent clinicians using the five-grade Clinician-Reported Photonumeric Cellulite Severity Scale (CR-PCSS). Patient satisfaction was evaluated using a 10-grade GAIS scale, ranging from 0 to 10 (0 - dissatisfied; 10 - fully satisfied). To objectify the treatment outcomes, 17 subjects were investigated using B-mode ultrasonography and real-time compression elastography at baseline and during follow-up. Results The average values of CR-PCSS (T1), CR-PCSS (T2) and CR-PCSS (T3) over all participants were 2.22±0.82, 1.18±0.77, and 0.84±0.77, respectively, which corresponded to the skin improvement between T1 and T2 of 0.93±0.27 (p < 0.0001) as well as between T1 and T3 of 1.38±0.47 (p < 0.0001). Assessment of elasticity of the dermis and adipose tissue on the basis of the 5-grade coloration scale revealed significant reinforcement of both tissues as well as of the superficial fascia at follow-up as compared to their baseline values. The obtained treatment outcomes were long-lasting and could be clearly observed even in individuals with a long-term follow-ups. Assessment of the satisfaction of participants with the treatment results revealed a high satisfaction of 8.95 ± 1.49. The method demonstrated no side effects, was pain-free, well-tolerated, and highly accepted by patients.
Collapse
Affiliation(s)
| | - Nadezhda I Kuprina
- North-West Public Health Research Center, St. Petersburg, Russian Federation
| | | |
Collapse
|
8
|
Mol MO, van Ham TJ, Bannink N, Bruggenwirth HT, Escher JC, Kros JM, Renkens JJM, van Unen L, Verdijk RM, Vlot J, Verhoeven VJM, Demirdas S. Biallelic and monoallelic variants in EFEMP1 can cause a severe and distinct subtype of heritable connective tissue disorder. Eur J Hum Genet 2024; 32:1567-1573. [PMID: 39367272 DOI: 10.1038/s41431-024-01692-x] [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: 05/30/2024] [Revised: 07/15/2024] [Accepted: 09/10/2024] [Indexed: 10/06/2024] Open
Abstract
Variants in EFEMP1, encoding Fibulin-3, were previously reported as a rare cause of heritable connective tissue disorder (HCTD) with recurrent hernias and joint hypermobility. We report three new cases with biallelic or monoallelic EFEMP1 variants and severe hernia phenotypes. Two male siblings of 10 and 13 years old presented with marfanoid habitus, recurrent inguinal and umbilical hernias, generalized joint hypermobility, and scoliosis. Parents and halfsiblings reported joint hypermobility and umbilical hernias. The eldest boy died at age 16 from incarcerated gastrointestinal herniation complicated by gastric and bowel necrosis with perforation. Autopsy revealed widespread intestinal diverticula. Immunohistochemistry of skin and fascia tissue did not reveal any abnormalities, including normal staining of elastic fibers. Both siblings harbored compound heterozygous likely pathogenic EFEMP1 variants (c.1320 + 2T > A, p.? and c.698G > A, p.Gly233Asp). An unrelated 58-year-old male had marfanoid features, high myopia, recurrent diaphragmatic and inguinal hernias, and chronic gastrointestinal dilatation with severe malabsorption. Both his dizygotic twin-brother and mother had recurrent hernias and high myopia. This man died at 59 years of age, and autopsy showed extensive diaphragmatic herniation, bowel diverticula, and pulmonary emphysema. A heterozygous EFEMP1 splice-variant (c.81 + 1G > A, p.?) was identified, causing exon skipping leading to a start-loss. Targeted genome reanalysis nor RNA-sequencing revealed a second variant at the other allele. The reported individuals expand the clinical and pathological phenotypes of EFEMP1-related disease, a distinct entity within the spectrum of HCTD. The severe and recurrent hernias, gastrointestinal dilatation, and diverticulosis result in an increased risk for life-threatening complications, demanding early recognition and close monitoring.
Collapse
Affiliation(s)
- M O Mol
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - T J van Ham
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - N Bannink
- Department of Pediatrics, Franciscus Gasthuis and Vlietland, Rotterdam and Schiedam, The Netherlands
| | - H T Bruggenwirth
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J C Escher
- Department of Pediatric Gastroenterology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J M Kros
- Department of Pathology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J J M Renkens
- Department of Orthopedics and Sportsmedicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - L van Unen
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - R M Verdijk
- Department of Pathology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J Vlot
- Department of Pediatric Surgery, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - V J M Verhoeven
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - S Demirdas
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
9
|
Ortega AJ, Daniel S, Renwick M, Kambhampati P, Thompson KN, Collier GE, Baker EL, Zaki H, Hulleman JD. Genetic removal of Nlrp3 protects against sporadic and R345W Efemp1-induced basal laminar deposit formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618289. [PMID: 39464028 PMCID: PMC11507711 DOI: 10.1101/2024.10.14.618289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Chronic, unresolved inflammation has long been speculated to serve as an initiating and propagating factor in numerous neurodegenerative diseases, including a leading cause of irreversible blindness in the elderly, age-related macular degeneration (AMD). Intracellular multiprotein complexes called inflammasomes in combination with activated caspases facilitate production of pro-inflammatory cytokines such as interleukin 1 beta. Specifically, the nucleotide-binding oligomerization (NOD)-like receptor protein 3 (NLRP3) has received heightened attention due to the wide range of stimuli to which it can respond and its potential involvement in AMD. In this study, we directly tested the role of Nlrp3 and its downstream effector, caspase 1 (Casp1) in mediating early AMD-like pathology (i.e., basal laminar deposits [BLamDs]) in wild-type (WT) mice and the Malattia Leventinese/Doyne honeycomb retinal dystrophy (ML/DHRD) mouse model (p.R345W mutation in Efemp1). Compared to aged-matched controls, R345W+/+ knockin mice demonstrated increased Muller cell gliosis, subretinal Iba-1+ microglial cells, higher Nlrp3 immunoreactivity in the retina, as well as significant transcriptional upregulation of complement component 3, Nlrp3, pro-Il1b, pro-caspase-1, and tissue inhibitor of matrix metalloproteinase 3 in the retinal pigmented epithelium (RPE)/choroid. These findings were accompanied by an age-related increase in BLamD formation in the R345W+/+ mice. Genetic elimination of either Nlrp3 or Casp1 significantly reduced both the size and coverage of BLamDs in the R345W+/+ background, highlighting an important and underappreciated pathway that could affect ML/DHRD onset and progression. Moreover, Nlrp3 knockout reduced spontaneous, idiopathic BLamDs in WT mice, suggesting translatability of our findings not only to rare inherited retinal dystrophies, but also potentially to AMD itself.
Collapse
Affiliation(s)
- Antonio J. Ortega
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Steffi Daniel
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Marian Renwick
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Pravallika Kambhampati
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Krista N. Thompson
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Gracen E. Collier
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Emily L. Baker
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Hasan Zaki
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - John D. Hulleman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| |
Collapse
|
10
|
Enzer NA, Chiles J, Mason S, Shirahata T, Castro V, Regan E, Choi B, Yuan NF, Diaz AA, Washko GR, McDonald ML, Estépar RSJ, Ash SY. Proteomics and machine learning in the prediction and explanation of low pectoralis muscle area. Sci Rep 2024; 14:17981. [PMID: 39097658 PMCID: PMC11297919 DOI: 10.1038/s41598-024-68447-y] [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: 02/14/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024] Open
Abstract
Low muscle mass is associated with numerous adverse outcomes independent of other associated comorbid diseases. We aimed to predict and understand an individual's risk for developing low muscle mass using proteomics and machine learning. We identified eight biomarkers associated with low pectoralis muscle area (PMA). We built three random forest classification models that used either clinical measures, feature selected biomarkers, or both to predict development of low PMA. The area under the receiver operating characteristic curve for each model was: clinical-only = 0.646, biomarker-only = 0.740, and combined = 0.744. We displayed the heterogenetic nature of an individual's risk for developing low PMA and identified two distinct subtypes of participants who developed low PMA. While additional validation is required, our methods for identifying and understanding individual and group risk for low muscle mass could be used to enable developments in the personalized prevention of low muscle mass.
Collapse
Affiliation(s)
- Nicholas A Enzer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Joe Chiles
- Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Stefanie Mason
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Toru Shirahata
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan
| | - Victor Castro
- Boston University School of Medicine, Boston, MA, USA
| | - Elizabeth Regan
- COPDGene Study Consortium, Denver, CO, USA
- Division of Rheumatology, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Bina Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Nancy F Yuan
- Department of Biomedical Informatics, University of California at San Diego, San Diego, CA, USA
| | - Alejandro A Diaz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Merry-Lynn McDonald
- Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Raúl San José Estépar
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Samuel Y Ash
- Department of Critical Care Medicine, South Shore Hospital, 55 Fogg Road, South Weymouth, MA, 02190, USA.
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
11
|
Daniel S, Hulleman JD. Exploring ocular fibulin-3 (EFEMP1): Anatomical, age-related, and species perspectives. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167239. [PMID: 38750770 PMCID: PMC11238277 DOI: 10.1016/j.bbadis.2024.167239] [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: 11/21/2023] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/24/2024]
Abstract
Fibulin-3 (FBLN3, aka EFEMP1) is a secreted extracellular matrix (ECM) glycoprotein implicated in ocular diseases including glaucoma and age-related macular degeneration. Yet surprisingly, little is known about its native biology, expression patterns, and localization in the eye. To overcome these shortcomings, we conducted gene expression analysis and immunohistochemistry for FBLN3 in ocular tissues from mice, pigs, non-human primates, and humans. Moreover, we evaluated age-related changes in FBLN3 and FBLN3-related ECM remodeling enzymes/inhibitors in aging mice. We found that FBLN3 displayed distinct staining patterns consistent across the mouse retina, particularly in the ganglion cell layer and inner nuclear layer (INL). In contrast, human retinas exhibited a unique staining pattern, with enrichment of FBLN3 in the retinal pigment epithelium (RPE), INL, and outer nuclear layer (ONL) in the peripheral retina. This staining transitioned to the outer plexiform layer (OPL) in the central retina/macula, and was accompanied by reduced RPE immunoreactivity approaching the fovea. Surprisingly, we found significant age-related increases in FBLN3 expression and protein abundance in the mouse retina which was paralleled by reduced transcript levels of FBLN3-degrading enzymes (i.e., Mmp2 and Htra1). Our findings highlight important species-dependent, retinal region-specific, and age-related expression and localization patterns of FBLN3 which favor its accumulation during aging. These findings contribute to a better understanding of FBLN3's role in ocular pathology and provide valuable insights for future FBLN3 research.
Collapse
Affiliation(s)
- Steffi Daniel
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th St. SE, Minneapolis, MN 55455, United States
| | - John D Hulleman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th St. SE, Minneapolis, MN 55455, United States.
| |
Collapse
|
12
|
Ichimata S, Hata Y, Yoshida K, Nishida N. Clinicopathological Appearance of Epidermal Growth-Factor-Containing Fibulin-like Extracellular Matrix Protein 1 Deposition in the Lower Gastrointestinal Tract: An Autopsy-Based Study. Int J Mol Sci 2024; 25:7581. [PMID: 39062824 PMCID: PMC11277079 DOI: 10.3390/ijms25147581] [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: 06/13/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
This study examined the patterns of epidermal growth-factor-containing fibulin-like extracellular matrix protein 1 (EFEMP1) deposition in the small intestine and colon to evaluate the association between the histopathological severity of EFEMP1 deposition and constipation and determine the colocalization of amyloid transthyretin (ATTR) and EFEMP1 deposits. In 40 older cases (≥80 years of age), EFEMP1 deposition in the small intestine initiated in the submucosal and subserous vessels, subserous interstitium, and serosa (early stage), progressing to the muscularis propria and peri-Auerbach plexus area (intermediate stage), and finally spreading diffusely to other areas, excluding the mucosa and muscularis mucosa (advanced stage). The colon had a similar pattern of progression. During the middle-to-advanced stages, amyloid formation was observed in some vascular and serous deposits. A subgroup of cases was identified in which EFEMP1 deposition was the only presumed cause of constipation. Additionally, we demonstrated the colocalization of ATTR and EFEMP1 deposition. Apple-green birefringence was detected under polarized light only in approximately one-half of the cases in the small intestine and one-third of the cases in the colon. These findings strongly suggest that EFEMP1 deposits are correlated with pathological conditions of the lower gastrointestinal tract. As the histopathological diagnosis using Congo red-stained specimens is challenging, the combined use of elastic fiber staining and EFEMP1 immunohistochemistry is recommended to identify EFEMP1 deposition.
Collapse
Affiliation(s)
- Shojiro Ichimata
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan (N.N.)
| | | | | | | |
Collapse
|
13
|
Forghani I, Lang SH, Rodier MJ, Bivona SA, Undiagnosed Diseases Network, Morales AA, Zuchner S, Bademci G, Tekin M. EFEMP1 haploinsufficiency causes a Marfan-like hereditary connective tissue disorder. Am J Med Genet A 2024; 194:e63556. [PMID: 38348595 PMCID: PMC11060917 DOI: 10.1002/ajmg.a.63556] [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: 09/10/2023] [Revised: 11/25/2023] [Accepted: 01/20/2024] [Indexed: 02/23/2024]
Abstract
Phenotypic features of a hereditary connective tissue disorder, including craniofacial characteristics, hyperextensible skin, joint laxity, kyphoscoliosis, arachnodactyly, inguinal hernia, and diverticulosis associated with biallelic pathogenic variants in EFEMP1 have been previously described in four patients. Genome sequencing on a proband and her mother with comparable phenotypic features revealed that both patients were heterozygous for a stop-gain variant c.1084C>T (p.Arg362*). Complementary RNA-seq on fibroblasts revealed significantly reduced levels of mutant EFEMP1 transcript. Considering the absence of other molecular explanations, we extrapolated that EFEMP1 could be the cause of the patient's phenotypes. Furthermore, nonsense-mediated decay was demonstrated for the mutant allele as the principal mechanism for decreased levels of EFEMP1 mRNA. We provide strong clinical and genetic evidence for the haploinsufficiency of EFEMP1 due to nonsense-medicated decay to cause severe kyphoscoliosis, generalized hypermobility of joints, high and narrow arched palate, and potentially severe diverticulosis. To the best of our knowledge, this is the first report of an autosomal dominant EFEMP1-associated hereditary connective tissue disorder and therefore expands the phenotypic spectrum of EFEMP1 related disorders.
Collapse
Affiliation(s)
- Irman Forghani
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL
| | - Steven H. Lang
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL
- Baylor College of Medicine, Houston, TX
| | - Matthew J. Rodier
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL
| | - Stephanie A. Bivona
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL
| | | | - Alejo A. Morales
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL
| | - Stephan Zuchner
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL
| | - Guney Bademci
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL
| | - Mustafa Tekin
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
14
|
Enzer NA, Chiles J, Mason S, Shirahata T, Castro V, Regan E, Choi B, Yuan NF, Diaz AA, Washko GR, McDonald ML, Estépar RSJ, Ash SY. Proteomics and Machine Learning in the Prediction and Explanation of Low Pectoralis Muscle Area. RESEARCH SQUARE 2024:rs.3.rs-3957125. [PMID: 38496412 PMCID: PMC10942559 DOI: 10.21203/rs.3.rs-3957125/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Low muscle mass is associated with numerous adverse outcomes independent of other associated comorbid diseases. We aimed to predict and understand an individual's risk for developing low muscle mass using proteomics and machine learning. We identified 8 biomarkers associated with low pectoralis muscle area (PMA). We built 3 random forest classification models that used either clinical measures, feature selected biomarkers, or both to predict development of low PMA. The area under the receiver operating characteristic curve for each model was: clinical-only = 0.646, biomarker-only = 0.740, and combined = 0.744. We displayed the heterogenetic nature of an individual's risk for developing low PMA and identified 2 distinct subtypes of participants who developed low PMA. While additional validation is required, our methods for identifying and understanding individual and group risk for low muscle mass could be used to enable developments in the personalized prevention of low muscle mass.
Collapse
|
15
|
Raman R, Bahri MA, Degueldre C, Caetano da Silva C, Sanchez C, Ostertag A, Collet C, Cohen-Solal M, Plenevaux A, Henrotin Y, Muller M. A Zebrafish Mutant in the Extracellular Matrix Protein Gene efemp1 as a Model for Spinal Osteoarthritis. Animals (Basel) 2023; 14:74. [PMID: 38200805 PMCID: PMC10778253 DOI: 10.3390/ani14010074] [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: 10/26/2023] [Revised: 12/17/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Osteoarthritis is a degenerative articular disease affecting mainly aging animals and people. The extracellular matrix protein Efemp1 was previously shown to have higher turn-over and increased secretion in the blood serum, urine, and subchondral bone of knee joints in osteoarthritic patients. Here, we use the zebrafish as a model system to investigate the function of Efemp1 in vertebrate skeletal development and homeostasis. Using in situ hybridization, we show that the efemp1 gene is expressed in the brain, the pharyngeal arches, and in the chordoblasts surrounding the notochord at 48 hours post-fertilization. We generated an efemp1 mutant line, using the CRISPR/Cas9 method, that produces a severely truncated Efemp1 protein. These mutant larvae presented a medially narrower chondrocranium at 5 days, which normalized later at day 10. At age 1.5 years, µCT analysis revealed an increased tissue mineral density and thickness of the vertebral bodies, as well as a decreased distance between individual vertebrae and ruffled borders of the vertebral centra. This novel defect, which has, to our knowledge, never been described before, suggests that the efemp1 mutant represents the first zebrafish model for spinal osteoarthritis.
Collapse
Affiliation(s)
- Ratish Raman
- Laboratory for Organogenesis and Regeneration (LOR), GIGA Institute, University of Liège, 4000 Liège, Belgium;
| | - Mohamed Ali Bahri
- GIGA CRC In Vivo Imaging, University of Liege, Sart Tilman, 4000 Liège, Belgium; (M.A.B.); (C.D.); (A.P.)
| | - Christian Degueldre
- GIGA CRC In Vivo Imaging, University of Liege, Sart Tilman, 4000 Liège, Belgium; (M.A.B.); (C.D.); (A.P.)
| | - Caroline Caetano da Silva
- Hospital Lariboisière, Reference Centre for Rare Bone Diseases, INSERM U1132, Université de Paris-Cité, F-75010 Paris, France; (C.C.d.S.); (A.O.); (C.C.); (M.C.-S.)
| | - Christelle Sanchez
- MusculoSKeletal Innovative Research Lab, Center for Interdisciplinary Research on Medicines, University of Liège, 4000 Liège, Belgium; (C.S.); (Y.H.)
| | - Agnes Ostertag
- Hospital Lariboisière, Reference Centre for Rare Bone Diseases, INSERM U1132, Université de Paris-Cité, F-75010 Paris, France; (C.C.d.S.); (A.O.); (C.C.); (M.C.-S.)
| | - Corinne Collet
- Hospital Lariboisière, Reference Centre for Rare Bone Diseases, INSERM U1132, Université de Paris-Cité, F-75010 Paris, France; (C.C.d.S.); (A.O.); (C.C.); (M.C.-S.)
- UF de Génétique Moléculaire, Hôpital Robert Debré, APHP, F-75019 Paris, France
| | - Martine Cohen-Solal
- Hospital Lariboisière, Reference Centre for Rare Bone Diseases, INSERM U1132, Université de Paris-Cité, F-75010 Paris, France; (C.C.d.S.); (A.O.); (C.C.); (M.C.-S.)
| | - Alain Plenevaux
- GIGA CRC In Vivo Imaging, University of Liege, Sart Tilman, 4000 Liège, Belgium; (M.A.B.); (C.D.); (A.P.)
| | - Yves Henrotin
- MusculoSKeletal Innovative Research Lab, Center for Interdisciplinary Research on Medicines, University of Liège, 4000 Liège, Belgium; (C.S.); (Y.H.)
| | - Marc Muller
- Laboratory for Organogenesis and Regeneration (LOR), GIGA Institute, University of Liège, 4000 Liège, Belgium;
| |
Collapse
|
16
|
Raja E, Clarin MTRDC, Yanagisawa H. Matricellular Proteins in the Homeostasis, Regeneration, and Aging of Skin. Int J Mol Sci 2023; 24:14274. [PMID: 37762584 PMCID: PMC10531864 DOI: 10.3390/ijms241814274] [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: 08/31/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Matricellular proteins are secreted extracellular proteins that bear no primary structural functions but play crucial roles in tissue remodeling during development, homeostasis, and aging. Despite their low expression after birth, matricellular proteins within skin compartments support the structural function of many extracellular matrix proteins, such as collagens. In this review, we summarize the function of matricellular proteins in skin stem cell niches that influence stem cells' fate and self-renewal ability. In the epidermal stem cell niche, fibulin 7 promotes epidermal stem cells' heterogeneity and fitness into old age, and the transforming growth factor-β-induced protein ig-h3 (TGFBI)-enhances epidermal stem cell growth and wound healing. In the hair follicle stem cell niche, matricellular proteins such as periostin, tenascin C, SPARC, fibulin 1, CCN2, and R-Spondin 2 and 3 modulate stem cell activity during the hair cycle and may stabilize arrector pili muscle attachment to the hair follicle during piloerections (goosebumps). In skin wound healing, matricellular proteins are upregulated, and their functions have been examined in various gain-and-loss-of-function studies. However, much remains unknown concerning whether these proteins modulate skin stem cell behavior, plasticity, or cell-cell communications during wound healing and aging, leaving a new avenue for future studies.
Collapse
Affiliation(s)
- Erna Raja
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; (E.R.); (M.T.R.D.C.C.)
| | - Maria Thea Rane Dela Cruz Clarin
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; (E.R.); (M.T.R.D.C.C.)
- Ph.D. Program in Humanics, School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba 305-8577, Japan
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; (E.R.); (M.T.R.D.C.C.)
| |
Collapse
|
17
|
Murtha LA, Hardy SA, Mabotuwana NS, Bigland MJ, Bailey T, Raguram K, Liu S, Ngo DT, Sverdlov AL, Tomin T, Birner-Gruenberger R, Hume RD, Iismaa SE, Humphreys DT, Patrick R, Chong JJH, Lee RJ, Harvey RP, Graham RM, Rainer PP, Boyle AJ. Fibulin-3 is necessary to prevent cardiac rupture following myocardial infarction. Sci Rep 2023; 13:14995. [PMID: 37696945 PMCID: PMC10495317 DOI: 10.1038/s41598-023-41894-9] [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: 06/13/2023] [Accepted: 09/01/2023] [Indexed: 09/13/2023] Open
Abstract
Despite the high prevalence of heart failure in the western world, there are few effective treatments. Fibulin-3 is a protein involved in extracellular matrix (ECM) structural integrity, however its role in the heart is unknown. We have demonstrated, using single cell RNA-seq, that fibulin-3 was highly expressed in quiescent murine cardiac fibroblasts, with expression highest prior to injury and late post-infarct (from ~ day-28 to week-8). In humans, fibulin-3 was upregulated in left ventricular tissue and plasma of heart failure patients. Fibulin-3 knockout (Efemp1-/-) and wildtype mice were subjected to experimental myocardial infarction. Fibulin-3 deletion resulted in significantly higher rate of cardiac rupture days 3-6 post-infarct, indicating a weak and poorly formed scar, with severe ventricular remodelling in surviving mice at day-28 post-infarct. Fibulin-3 knockout mice demonstrated less collagen deposition at day-3 post-infarct, with abnormal collagen fibre-alignment. RNA-seq on day-3 infarct tissue revealed upregulation of ECM degradation and inflammatory genes, but downregulation of ECM assembly/structure/organisation genes in fibulin-3 knockout mice. GSEA pathway analysis showed enrichment of inflammatory pathways and a depletion of ECM organisation pathways. Fibulin-3 originates from cardiac fibroblasts, is upregulated in human heart failure, and is necessary for correct ECM organisation/structural integrity of fibrotic tissue to prevent cardiac rupture post-infarct.
Collapse
Affiliation(s)
- Lucy A Murtha
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Sean A Hardy
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Nishani S Mabotuwana
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Mark J Bigland
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Taleah Bailey
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Kalyan Raguram
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Saifei Liu
- Department of Cardiology and Clinical Pharmacology, Basil Hetzel Institute, The University of Adelaide, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Doan T Ngo
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
- Department of Cardiology and Clinical Pharmacology, Basil Hetzel Institute, The University of Adelaide, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Aaron L Sverdlov
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
- Department of Cardiology and Clinical Pharmacology, Basil Hetzel Institute, The University of Adelaide, The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Tamara Tomin
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Ruth Birner-Gruenberger
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Robert D Hume
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Siiri E Iismaa
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- St Vincent's Clinical School, UNSW, Sydney, Kensington, NSW, Australia
| | - David T Humphreys
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- St Vincent's Clinical School, UNSW, Sydney, Kensington, NSW, Australia
| | - Ralph Patrick
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- St Vincent's Clinical School, UNSW, Sydney, Kensington, NSW, Australia
| | - James J H Chong
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
- Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Randall J Lee
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
- Edyth and Eli Broad Center for Regenerative Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- St Vincent's Clinical School, UNSW, Sydney, Kensington, NSW, Australia
- School of Biotechnology and Molecular Bioscience, UNSW, Sydney, Kensington, NSW, Australia
| | - Robert M Graham
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- St Vincent's Clinical School, UNSW, Sydney, Kensington, NSW, Australia
| | - Peter P Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Andrew J Boyle
- Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia.
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia.
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, NSW, Australia.
| |
Collapse
|
18
|
Liang Y, Liang Z, Huang J, Jia M, Liu D, Zhang P, Fang Z, Hu X, Li H. Identification and validation of aging-related gene signatures and their immune landscape in diabetic nephropathy. Front Med (Lausanne) 2023; 10:1158166. [PMID: 37404805 PMCID: PMC10316791 DOI: 10.3389/fmed.2023.1158166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/24/2023] [Indexed: 07/06/2023] Open
Abstract
Background Aging and immune infiltration have essential role in the physiopathological mechanisms of diabetic nephropathy (DN), but their relationship has not been systematically elucidated. We identified aging-related characteristic genes in DN and explored their immune landscape. Methods Four datasets from the Gene Expression Omnibus (GEO) database were screened for exploration and validation. Functional and pathway analysis was performed using Gene Set Enrichment Analysis (GSEA). Characteristic genes were obtained using a combination of Random Forest (RF) and Support Vector Machine Recursive Feature Elimination (SVM-RFE) algorithm. We evaluated and validated the diagnostic performance of the characteristic genes using receiver operating characteristic (ROC) curve, and the expression pattern of the characteristic genes was evaluated and validated. Single-Sample Gene Set Enrichment Analysis (ssGSEA) was adopted to assess immune cell infiltration in samples. Based on the TarBase database and the JASPAR repository, potential microRNAs and transcription factors were predicted to further elucidate the molecular regulatory mechanisms of the characteristic genes. Results A total of 14 differentially expressed genes related to aging were obtained, of which 10 were up-regulated and 4 were down-regulated. Models were constructed by the RF and SVM-RFE algorithms, contracted to three signature genes: EGF-containing fibulin-like extracellular matrix (EFEMP1), Growth hormone receptor (GHR), and Vascular endothelial growth factor A (VEGFA). The three genes showed good efficacy in three tested cohorts and consistent expression patterns in the glomerular test cohorts. Most immune cells were more infiltrated in the DN samples compared to the controls, and there was a negative correlation between the characteristic genes and most immune cell infiltration. 24 microRNAs were involved in the transcriptional regulation of multiple genes simultaneously, and Endothelial transcription factor GATA-2 (GATA2) had a potential regulatory effect on both GHR and VEGFA. Conclusion We identified a novel aging-related signature allowing assessment of diagnosis for DN patients, and further can be used to predict immune infiltration sensitivity.
Collapse
Affiliation(s)
- Yingchao Liang
- Graduate School of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhiyi Liang
- Graduate School of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Foshan, China
| | - Jinxian Huang
- Graduate School of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Mingjie Jia
- Graduate School of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Deliang Liu
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Pengxiang Zhang
- Graduate School of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zebin Fang
- Graduate School of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xinyu Hu
- Graduate School of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Huilin Li
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
19
|
Halsey G, Sinha D, Dhital S, Wang X, Vyavahare N. Role of elastic fiber degradation in disease pathogenesis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166706. [PMID: 37001705 PMCID: PMC11659964 DOI: 10.1016/j.bbadis.2023.166706] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
Elastin is a crucial extracellular matrix protein that provides structural integrity to tissues. Crosslinked elastin and associated microfibrils, named elastic fiber, contribute to biomechanics by providing the elasticity required for proper function. During aging and disease, elastic fiber can be progressively degraded and since there is little elastin synthesis in adults, degraded elastic fiber is not regenerated. There is substantial evidence linking loss or damage of elastic fibers to the clinical manifestation and pathogenesis of a variety of diseases. Disruption of elastic fiber networks by hereditary mutations, aging, or pathogenic stimuli results in systemic ailments associated with the production of elastin degradation products, inflammatory responses, and abnormal physiology. Due to its longevity, unique mechanical properties, and widespread distribution in the body, elastic fiber plays a central role in homeostasis of various physiological systems. While pathogenesis related to elastic fiber degradation has been more thoroughly studied in elastic fiber rich tissues such as the vasculature and the lungs, even tissues containing relatively small quantities of elastic fibers such as the eyes or joints may be severely impacted by elastin degradation. Elastic fiber degradation is a common observation in certain hereditary, age, and specific risk factor exposure induced diseases representing a converging point of pathological clinical phenotypes which may also help explain the appearance of co-morbidities. In this review, we will first cover the role of elastic fiber degradation in the manifestation of hereditary diseases then individually explore the structural role and degradation effects of elastic fibers in various tissues and organ systems. Overall, stabilizing elastic fiber structures and repairing lost elastin may be effective strategies to reverse the effects of these diseases.
Collapse
Affiliation(s)
- Gregory Halsey
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Dipasha Sinha
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Saphala Dhital
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Xiaoying Wang
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Naren Vyavahare
- Department of Bioengineering, Clemson University, SC 29634, United States of America.
| |
Collapse
|
20
|
Identification of Prognostic Aging-Related Genes Associated with Immune Cell Infiltration in Glioblastoma. JOURNAL OF ONCOLOGY 2023. [DOI: 10.1155/2023/9220547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Background. Aging is recognized as a main tumor risk factor, and thus aging has become a field of interest in the tumor research field. Glioblastoma multiforme represents the most typical primary malignant intracranial tumor, particularly in the elderly. However, the association between aging-related genes (AGs) and GBM prognosis remains unknown. As a result, the primary goal of this study was to determine the association among AGs and the prognosis of GBM. Methods. A total of 307 human AGs were downloaded from the HAGR database, while the expression profiles of GSE4290 and GSE4412 were obtained from the GEO database. Furthermore, data on GBM expression profiles were obtained from the Chinese Glioma Genome Atlas (CGGA) database. The DEAGs that were differentially expressed among the AG and GBM gene expression profiles derived from GSE4290 were then identified, followed by functional analysis of the DEAGs. The survival-related AGs were then screened using univariate Cox regression analysis , which was used to build and validate a prognostic risk model. Furthermore, the ESTIMATE and CIBERSORT algorithms were utilized to explore the association between the survival-related AGs and the tumor immune microenvironment. Results. In entire, 29 DEAGs were identified in the GSE4290. This was monitored by the construction of the prognosis risk model using four DEAGs from the CGGA training set, including C1QA, CDK1, EFEMP1, and IGFBP2. Next, the risk model was confirmed in the CGGA experiment set and the GSE 4412 dataset. Results showed that C1QA, CDK1, EFEMP1, and IGFBP2 levels were remarkably higher in the high-risk score groups, and they had a good association with immune and stromal scores. Conclusion. A robust prognostic risk model was constructed and validated using four AGs, including C1QA, CDK1, EFEMP1, and IGFBP2, which had a close relationship with the immune microenvironment of GBM. This study offers a new reference to further explore the pathogenesis of GBM and recognize new and more effective GBM treatments.
Collapse
|
21
|
Kruglikov IL, Scherer PE. Pathophysiology of cellulite: Possible involvement of selective endotoxemia. Obes Rev 2023; 24:e13517. [PMID: 36285892 PMCID: PMC9772045 DOI: 10.1111/obr.13517] [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: 06/10/2022] [Revised: 09/13/2022] [Accepted: 10/02/2022] [Indexed: 12/27/2022]
Abstract
The most relevant hallmarks of cellulite include a massive protrusion of superficial adipose tissue into the dermis, reduced expression of the extracellular glycoprotein fibulin-3, and an unusually high presence of MUSE cells in gluteofemoral white adipose tissue (gfWAT) that displays cellulite. Also typical for this condition is the hypertrophic nature of the underlying adipose tissue, the interaction of adipocytes with sweat glands, and dysfunctional lymph and blood circulation as well as a low-grade inflammation in the areas of gfWAT affected by cellulite. Here, we propose a new pathophysiology of cellulite, which connects this skin condition with selective accumulation of endogenous lipopolysaccharides (LPS) in gfWAT. The accumulation of LPS within a specific WAT depot has so far not been considered as a possible pathophysiological mechanism triggering localized WAT modifications, but may very well be involved in conditions such as cellulite and, secondary to that, lipedema.
Collapse
Affiliation(s)
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, 75390-8549, USA
| |
Collapse
|
22
|
Ahmed WUR, Patel MIA, Ng M, McVeigh J, Zondervan K, Wiberg A, Furniss D. Shared genetic architecture of hernias: A genome-wide association study with multivariable meta-analysis of multiple hernia phenotypes. PLoS One 2022; 17:e0272261. [PMID: 36584111 PMCID: PMC9803250 DOI: 10.1371/journal.pone.0272261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 07/14/2022] [Indexed: 12/31/2022] Open
Abstract
Abdominal hernias are common and characterised by the abnormal protrusion of a viscus through the wall of the abdominal cavity. The global incidence is 18.5 million annually and there are limited non-surgical treatments. To improve understanding of common hernia aetiopathology, we performed a six-stage genome-wide association study (GWAS) of 62,637 UK Biobank participants with either single or multiple hernia phenotypes including inguinal, femoral, umbilical and hiatus hernia. Additionally, we performed multivariable meta-analysis with metaUSAT, to allow integration of summary data across traits to generate combined effect estimates. On individual hernia analysis, we identified 3404 variants across 38 genome-wide significant (p < 5×10-8) loci of which 11 are previously unreported. Robust evidence for five shared susceptibility loci was discovered: ZC3H11B, EFEMP1, MHC region, WT1 and CALD1. Combined hernia phenotype analyses with additional multivariable meta-analysis of summary statistics in metaUSAT revealed 28 independent (seven previously unreported) shared susceptibility loci. These clustered in functional categories related to connective tissue and elastic fibre homeostasis. Weighted genetic risk scores also correlated with disease severity suggesting a phenotypic-genotypic severity correlation, an important finding to inform future personalised therapeutic approaches to hernia.
Collapse
Affiliation(s)
- Waheed Ul-Rahman Ahmed
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Manal I. A. Patel
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Michael Ng
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - James McVeigh
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Krina Zondervan
- Nuffield Department of Women’s & Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Akira Wiberg
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
- Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Dominic Furniss
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
- Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Gomes FE, Casanova MI, Mouttham L, Bannasch DL, Park S, Kim S, Young LJ, Daley NL, Thomasy SM, Castelhano MG, Ledbetter EC, Holmberg B, Boyd R, Van Der Woerdt A, McDonald J, Hayward JJ. A genome-wide association study to investigate genetic loci associated with primary glaucoma in American Cocker Spaniels. Am J Vet Res 2022; 83:1-8. [PMID: 36170212 DOI: 10.2460/ajvr.22.07.0106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To identify genetic associations with primary glaucoma (PG) in American Cocker Spaniels using a genome-wide association study (GWAS). ANIMALS A nationwide ambidirectional case-control cohort study was performed in American Cocker Spaniels that had an ophthalmic examination performed by a veterinarian. Ninety-four dogs with PG (cases) and 111 dogs without glaucoma (controls) met phenotypic criteria and had a blood sample collected after receiving informed owner consent. PROCEDURES Genomic DNA was extracted from whole blood samples and genotyped (CanineHD BeadChip, Illumina Inc). A case-control GWAS using a linear mixed model was performed, and 3 significance thresholds were calculated (1) using a Bonferroni correction on all single nucleotide polymorphisms (SNPs) included in the GWAS, (2) using a Bonferroni correction on only the unlinked SNPs from a pruned data set, and (3) using 10,000 random phenotype permutations. RESULTS Following genotype data quality control, 89 cases and 93 controls were included in the GWAS. We identified an association on canine chromosome (CFA10); however, it did not reach statistical significance. Potential candidate genes within the surrounding linkage disequilibrium interval include coiled-coil domain containing 85A (CCDC85A) and extracellular growth factor containing fibulin extracellular matrix protein 1 (EFEMP1). CLINICAL RELEVANCE Primary glaucoma in the American Cocker Spaniel is a complex heterogeneous disease that may be influenced by a locus on CFA10. The candidate genes CCDC85A and EFEMP1 within the identified linkage disequilibrium interval have been shown to be involved in human open-angle glaucoma.
Collapse
Affiliation(s)
- Filipe Espinheira Gomes
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY.,Small Animal Specialist Hospital, North Ryde, Australia
| | - Maria Isabel Casanova
- Department of Surgical & Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA
| | - Lara Mouttham
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY.,Cornell Veterinary Biobank, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Danika L Bannasch
- Department of Population Health & Reproduction, School of Veterinary Medicine, University of California-Davis, Davis, CA
| | - Sangwan Park
- Department of Surgical & Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA
| | - Soohyun Kim
- Department of Surgical & Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA
| | - Laura J Young
- Department of Surgical & Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA
| | - Nicole L Daley
- Department of Surgical & Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA
| | - Sara M Thomasy
- Department of Surgical & Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA.,Department of Ophthalmology & Vision Science, School of Medicine, University of California-Davis, Davis, CA
| | - Marta G Castelhano
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY.,Cornell Veterinary Biobank, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Eric C Ledbetter
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | | | - Ryan Boyd
- South Texas Veterinary Ophthalmology, San Antonio, TX
| | | | | | - Jessica J Hayward
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| |
Collapse
|
24
|
Molins B, Mesquida M, Adan A. Bioengineering approaches for modelling retinal pathologies of the outer blood-retinal barrier. Prog Retin Eye Res 2022:101097. [PMID: 35840488 DOI: 10.1016/j.preteyeres.2022.101097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 05/31/2022] [Accepted: 06/29/2022] [Indexed: 11/18/2022]
Abstract
Alterations of the junctional complex of the outer blood-retinal barrier (oBRB), which is integrated by the close interaction of the retinal pigment epithelium, the Bruch's membrane, and the choriocapillaris, contribute to the loss of neuronal signalling and subsequent vision impairment in several retinal inflammatory disorders such as age-related macular degeneration and diabetic retinopathy. Reductionist approaches into the mechanisms that underlie such diseases have been hindered by the absence of adequate in vitro models using human cells to provide the 3D dynamic architecture that enables expression of the in vivo phenotype of the oBRB. Conventional in vitro cell models are based on 2D monolayer cellular cultures, unable to properly recapitulate the complexity of living systems. The main drawbacks of conventional oBRB models also emerge from the cell sourcing, the lack of an appropriate Bruch's membrane analogue, and the lack of choroidal microvasculature with flow. In the last years, the advent of organ-on-a-chip, bioengineering, and stem cell technologies is providing more advanced 3D models with flow, multicellularity, and external control over microenvironmental properties. By incorporating additional biological complexity, organ-on-a-chip devices can mirror physiologically relevant properties of the native tissue while offering additional set ups to model and study disease. In this review we first examine the current understanding of oBRB biology as a functional unit, highlighting the coordinated contribution of the different components to barrier function in health and disease. Then we describe recent advances in the use of pluripotent stem cells-derived retinal cells, Bruch's membrane analogues, and co-culture techniques to recapitulate the oBRB. We finally discuss current advances and challenges of oBRB-on-a-chip technologies for disease modelling.
Collapse
Affiliation(s)
- Blanca Molins
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain.
| | - Marina Mesquida
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain; Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Alfredo Adan
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain; Instituto Clínic de Oftalmología, Hospital Clínic Barcelona, C/ Sabino de Arana 1, 08028, Barcelona, Spain
| |
Collapse
|
25
|
Mishra A, Duplaà C, Vojinovic D, Suzuki H, Sargurupremraj M, Zilhão NR, Li S, Bartz TM, Jian X, Zhao W, Hofer E, Wittfeld K, Harris SE, van der Auwera-Palitschka S, Luciano M, Bis JC, Adams HHH, Satizabal CL, Gottesman RF, Gampawar PG, Bülow R, Weiss S, Yu M, Bastin ME, Lopez OL, Vernooij MW, Beiser AS, Völker U, Kacprowski T, Soumare A, Smith JA, Knopman DS, Morris Z, Zhu Y, Rotter JI, Dufouil C, Valdés Hernández M, Muñoz Maniega S, Lathrop M, Boerwinkle E, Schmidt R, Ihara M, Mazoyer B, Yang Q, Joutel A, Tournier-Lasserve E, Launer LJ, Deary IJ, Mosley TH, Amouyel P, DeCarli CS, Psaty BM, Tzourio C, Kardia SLR, Grabe HJ, Teumer A, van Duijn CM, Schmidt H, Wardlaw JM, Ikram MA, Fornage M, Gudnason V, Seshadri S, Matthews PM, Longstreth WT, Couffinhal T, Debette S. Gene-mapping study of extremes of cerebral small vessel disease reveals TRIM47 as a strong candidate. Brain 2022; 145:1992-2007. [PMID: 35511193 PMCID: PMC9255380 DOI: 10.1093/brain/awab432] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/11/2021] [Accepted: 10/28/2021] [Indexed: 12/11/2022] Open
Abstract
Cerebral small vessel disease is a leading cause of stroke and a major contributor to cognitive decline and dementia, but our understanding of specific genes underlying the cause of sporadic cerebral small vessel disease is limited. We report a genome-wide association study and a whole-exome association study on a composite extreme phenotype of cerebral small vessel disease derived from its most common MRI features: white matter hyperintensities and lacunes. Seventeen population-based cohorts of older persons with MRI measurements and genome-wide genotyping (n = 41 326), whole-exome sequencing (n = 15 965), or exome chip (n = 5249) data contributed 13 776 and 7079 extreme small vessel disease samples for the genome-wide association study and whole-exome association study, respectively. The genome-wide association study identified significant association of common variants in 11 loci with extreme small vessel disease, of which the chr12q24.11 locus was not previously reported to be associated with any MRI marker of cerebral small vessel disease. The whole-exome association study identified significant associations of extreme small vessel disease with common variants in the 5' UTR region of EFEMP1 (chr2p16.1) and one probably damaging common missense variant in TRIM47 (chr17q25.1). Mendelian randomization supports the causal association of extensive small vessel disease severity with increased risk of stroke and Alzheimer's disease. Combined evidence from summary-based Mendelian randomization studies and profiling of human loss-of-function allele carriers showed an inverse relation between TRIM47 expression in the brain and blood vessels and extensive small vessel disease severity. We observed significant enrichment of Trim47 in isolated brain vessel preparations compared to total brain fraction in mice, in line with the literature showing Trim47 enrichment in brain endothelial cells at single cell level. Functional evaluation of TRIM47 by small interfering RNAs-mediated knockdown in human brain endothelial cells showed increased endothelial permeability, an important hallmark of cerebral small vessel disease pathology. Overall, our comprehensive gene-mapping study and preliminary functional evaluation suggests a putative role of TRIM47 in the pathophysiology of cerebral small vessel disease, making it an important candidate for extensive in vivo explorations and future translational work.
Collapse
Affiliation(s)
- Aniket Mishra
- University of Bordeaux, INSERM, Bordeaux Population Health Research Centre, Team ELEANOR, UMR 1219, F-33000 Bordeaux, France
| | - Cécile Duplaà
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, F-33600 Pessac, France
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus MC, University Medical Centre, 3015 GD Rotterdam, The Netherlands
- Department of Biomedical Data Sciences, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Hideaki Suzuki
- Department of Cardiovascular Medicine, Tohoku University Hospital, 1-1, Seiryo, Aoba, Sendai 980-8574, Japan
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo, Aoba, Sendai 980-8573, Japan
- Department of Brain Sciences and UK Dementia Research Institute Centre, Imperial College, London, W12 0NN, UK
| | - Muralidharan Sargurupremraj
- University of Bordeaux, INSERM, Bordeaux Population Health Research Centre, Team ELEANOR, UMR 1219, F-33000 Bordeaux, France
| | | | - Shuo Li
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02115, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Xueqiu Jian
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Michigan, MI 48104, USA
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, 8036 Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, 8036 Graz, Austria
| | - Katharina Wittfeld
- German Centre for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, 17489 Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Sarah E Harris
- Department of Psychology, University of Edinburgh, EH8 9JZ Edinburgh, UK
| | - Sandra van der Auwera-Palitschka
- German Centre for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, 17489 Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Michelle Luciano
- Department of Psychology, University of Edinburgh, EH8 9JZ Edinburgh, UK
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus MC, University Medical Centre, 3015 GD Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Centre, 3015 GD Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
- The Framingham Heart Study, Framingham, MA 01701, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 2115, USA
| | - Rebecca F Gottesman
- National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD 20814, USA
| | - Piyush G Gampawar
- Institute of Molecular Biology & Biochemistry, Gottfried Schatz Research Centre (for Cell Signalling, Metabolism and Aging), Medical University of Graz, 8036 Graz, Austria
| | - Robin Bülow
- Department of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Miao Yu
- Department of Epidemiology, School of Public Health, University of Michigan, Michigan, MI 48104, USA
| | - Mark E Bastin
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH8 9AB, UK
- Division of Neuroimaging Sciences, Brain Research Imaging Centre, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC, University Medical Centre, 3015 GD Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Alexa S Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02115, USA
- The Framingham Heart Study, Framingham, MA 01701, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 2115, USA
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Tim Kacprowski
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17489 Greifswald, Germany
- TUM School of Life Sciences Weihenstephan (WZW), Technical University of Munich (TUM), 85354 Freising-Weihenstephan, Germany
| | - Aicha Soumare
- University of Bordeaux, INSERM, Bordeaux Population Health Research Centre, Team ELEANOR, UMR 1219, F-33000 Bordeaux, France
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Michigan, MI 48104, USA
| | | | - Zoe Morris
- Neuroradiology Department, Department of Clinical Neurosciences, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Yicheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Carole Dufouil
- University of Bordeaux, INSERM, Bordeaux Population Health Research Centre, Team ELEANOR, UMR 1219, F-33000 Bordeaux, France
| | - Maria Valdés Hernández
- Division of Neuroimaging Sciences, Brain Research Imaging Centre, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Susana Muñoz Maniega
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH8 9AB, UK
| | - Mark Lathrop
- University of McGill Genome Center, Montreal, Quebec H3A 0G1, Canada
| | - Erik Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, 8036 Graz, Austria
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka 564-8565, Japan
| | - Bernard Mazoyer
- University of Bordeaux, Institut des Maladies Neurodégénératives, CNRS-CEA UMR 5293, 33000 Bordeaux, France
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02115, USA
- The Framingham Heart Study, Framingham, MA 01701, USA
| | - Anne Joutel
- Institute of Psychiatry and Neurosciences of Paris, INSERM UMR1266, Université de Paris, France
| | | | - Lenore J Launer
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ian J Deary
- Department of Psychology, University of Edinburgh, EH8 9JZ Edinburgh, UK
| | - Thomas H Mosley
- Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Philippe Amouyel
- University of Lille, INSERM, Institut Pasteur de Lille, UMR1167-RID-AGE—Risk Factors and Molecular Determinants of Aging-Related Diseases, F-59000 Lille, France
- LabEx DISTALZ, Institut Pasteur de Lille, 59000 Lille, France
- Department of Epidemiology and Public Health, Centre Hospital University of Lille, F-59000 Lille, France
| | - Charles S DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, CA 95816, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA
| | - Christophe Tzourio
- University of Bordeaux, INSERM, Bordeaux Population Health Research Centre, Team ELEANOR, UMR 1219, F-33000 Bordeaux, France
- CHU de Bordeaux, Pole de santé publique, Service d’information médicale, F-33000 Bordeaux, France
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Michigan, MI 48104, USA
| | - Hans J Grabe
- German Centre for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, 17489 Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Cornelia M van Duijn
- Department of Biomedical Data Sciences, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Helena Schmidt
- Institute of Molecular Biology & Biochemistry, Gottfried Schatz Research Centre (for Cell Signalling, Metabolism and Aging), Medical University of Graz, 8036 Graz, Austria
| | - Joanna M Wardlaw
- Division of Neuroimaging Sciences, Brain Research Imaging Centre, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - M Arfan Ikram
- Department of Biomedical Data Sciences, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, 200 Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Sudha Seshadri
- Department of Epidemiology, School of Public Health, University of Michigan, Michigan, MI 48104, USA
- The Framingham Heart Study, Framingham, MA 01701, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 2115, USA
| | - Paul M Matthews
- Department of Brain Sciences and UK Dementia Research Institute Centre, Imperial College, London, W12 0NN, UK
| | - William T Longstreth
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Neurology, University of Washington, Seattle, WA 98104-2420, USA
| | - Thierry Couffinhal
- University of Bordeaux, INSERM, Biologie des Maladies Cardiovasculaires, U1034, F-33600 Pessac, France
| | - Stephanie Debette
- University of Bordeaux, INSERM, Bordeaux Population Health Research Centre, Team ELEANOR, UMR 1219, F-33000 Bordeaux, France
- Department of Neurology, Boston University School of Medicine, Boston, MA 2115, USA
- CHU de Bordeaux, Department of Neurology, F-33000 Bordeaux, France
| |
Collapse
|
26
|
Sharma P, Beck D, Murtha LA, Figtree G, Boyle A, Gentile C. Fibulin-3 Deficiency Protects Against Myocardial Injury Following Ischaemia/ Reperfusion in in vitro Cardiac Spheroids. Front Cardiovasc Med 2022; 9:913156. [PMID: 35795376 PMCID: PMC9251181 DOI: 10.3389/fcvm.2022.913156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/30/2022] [Indexed: 12/02/2022] Open
Abstract
Myocardial infarction (MI, or heart attack) is a leading cause of death worldwide. Myocardial ischaemia reperfusion (I/R) injury typical of MI events is also associated with the development of cardiac fibrosis and heart failure in patients. Fibulin-3 is an extracellular matrix component that plays a role in regulating MI response in the heart. In this study, we generated and compared in vitro cardiac spheroids (CSs) from wild type (WT) and fibulin-3 knockout (Fib-3 KO) mice. These were then exposed to pathophysiological changes in oxygen (O2) concentrations to mimic an MI event. We finally measured changes in contractile function, cell death, and mRNA expression levels of cardiovascular disease genes between WT and Fib-3 KO CSs. Our results demonstrated that there are significant differences in growth kinetics and endothelial network formation between WT and Fib-3 KO CSs, however, they respond similarly to changes in O2 concentrations. Fib-3 deficiency resulted in an increase in viability of cells and improvement in contraction frequency and fractional shortening compared to WT I/R CSs. Gene expression analyses demonstrated that Fib-3 deficiency inhibits I/R injury and cardiac fibrosis and promotes angiogenesis in CSs. Altogether, our findings suggest that Fib-3 deficiency makes CSs resistant to I/R injury and associated cardiac fibrosis and helps to improve the vascular network in CSs.
Collapse
Affiliation(s)
- Poonam Sharma
- College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney, NSW, Australia
- Faculty of Engineering and IT, School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Dominik Beck
- Faculty of Engineering and IT, School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Lucy A. Murtha
- College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
| | - Gemma Figtree
- Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney, NSW, Australia
| | - Andrew Boyle
- College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
| | - Carmine Gentile
- Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney, NSW, Australia
- Faculty of Engineering and IT, School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
- *Correspondence: Carmine Gentile
| |
Collapse
|
27
|
Fadista J, Skotte L, Karjalainen J, Abner E, Sørensen E, Ullum H, Werge T, Esko T, Milani L, Palotie A, Daly M, Melbye M, Feenstra B, Geller F. Comprehensive genome-wide association study of different forms of hernia identifies more than 80 associated loci. Nat Commun 2022; 13:3200. [PMID: 35680855 PMCID: PMC9184475 DOI: 10.1038/s41467-022-30921-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 05/18/2022] [Indexed: 11/08/2022] Open
Abstract
Hernias are characterized by protrusion of an organ or tissue through its surrounding cavity and often require surgical repair. In this study we identify 65,492 cases for five hernia types in the UK Biobank and perform genome-wide association study scans for these five types and two combined groups. Our results show associated variants in all scans. Inguinal hernia has the most associations and we conduct a follow-up study with 23,803 additional cases from four study groups giving 84 independently associated variants. Identified variants from all scans are collapsed into 81 independent loci. Further testing shows that 26 loci are associated with more than one hernia type, suggesting substantial overlap between the underlying genetic mechanisms. Pathway analyses identify several genes with a strong link to collagen and/or elastin (ADAMTS6, ADAMTS16, ADAMTSL3, LOX, ELN) in the vicinity of associated loci for inguinal hernia, which substantiates an essential role of connective tissue morphology.
Collapse
Affiliation(s)
- João Fadista
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Sciences, Lund University Diabetes Centre, Malmö, Sweden
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Line Skotte
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Juha Karjalainen
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Henrik Ullum
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Statens Serum Institut, Copenhagen, Denmark
| | - Thomas Werge
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Lundbeck Foundation Center for GeoGenetics, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Copenhagen and Aarhus, Aarhus, Denmark
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
| | - Lili Milani
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric & Neurodevelopmental Genetics Unit, Department of Psychiatry, Analytic and Translational Genetics Unit, Department of Medicine, and the Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Mark Daly
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mads Melbye
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
- Norwegian Institute of Public Health, Oslo, Norway
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark.
| |
Collapse
|
28
|
Ichimata S, Hata Y, Katoh N, Kametani F, Yazaki M, Sekijima Y, Nishida N. Novel histopathological deposition patterns of EGF-containing fibulin-like extracellular matrix protein 1 amyloidosis: an autopsy case exhibiting a possible association between AEFEMP1 amyloidosis and elastic fibres. Amyloid 2022; 29:139-140. [PMID: 35023419 DOI: 10.1080/13506129.2021.2020754] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Shojiro Ichimata
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Yukiko Hata
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Nagaaki Katoh
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Fuyuki Kametani
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masahide Yazaki
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, Japan
| | - Yoshiki Sekijima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoki Nishida
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
29
|
Gardella B, Scatigno AL, Belli G, Gritti A, Visoná SD, Dominoni M. Aging of Pelvic Floor in Animal Models: A Sistematic Review of Literature on the Role of the Extracellular Matrix in the Development of Pelvic Floor Prolapse. Front Med (Lausanne) 2022; 9:863945. [PMID: 35492348 PMCID: PMC9051040 DOI: 10.3389/fmed.2022.863945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Pelvic organ prolapse (POP) affects many women and contributes significantly to a decrease in their quality of life causing urinary and/or fecal incontinence, sexual dysfunction and dyspareunia. To better understand POP pathophysiology, prevention and treatment, many researchers resorted to evaluating animal models. Regarding this example and because POP affects principally older women, our aim was to provide an overview of literature on the possible biomechanical changes that occur in the vaginas of animal models and their supportive structures as a consequence of aging. Papers published online from 2000 until May 2021 were considered and particular attention was given to articles reporting the effects of aging on the microscopic structure of the vagina and pelvic ligaments in animal models. Most research has been conducted on rodents because their vagina structure is well characterized and similar to those of humans; furthermore, they are cost effective. The main findings concern protein structures of the connective tissue, known as elastin and collagen. We have noticed a significant discordance regarding the quantitative changes in elastin and collagen related to aging, especially because it is difficult to detect them in animal specimens. However, it seems to be clear that aging affects the qualitative properties of elastin and collagen leading to aberrant forms which may affect the elasticity and the resilience of tissues leading to pelvic floor disease. The analysis of histological changes of pelvic floor tissues related to aging underlines how these topics appear to be not fully understood so far and that more research is necessary.
Collapse
Affiliation(s)
- Barbara Gardella
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Annachiara Licia Scatigno
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Giacomo Belli
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Andrea Gritti
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Silvia Damiana Visoná
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Mattia Dominoni
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| |
Collapse
|
30
|
MacDonald BT, Keshishian H, Mundorff CC, Arduini A, Lai D, Bendinelli K, Popp NR, Bhandary B, Clauser KR, Specht H, Elowe NH, Laprise D, Xing Y, Kaushik VK, Carr SA, Ellinor PT. TAILS Identifies Candidate Substrates and Biomarkers of ADAMTS7, a Therapeutic Protease Target in Coronary Artery Disease. Mol Cell Proteomics 2022; 21:100223. [PMID: 35283288 PMCID: PMC9035411 DOI: 10.1016/j.mcpro.2022.100223] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/05/2022] [Accepted: 03/02/2022] [Indexed: 12/22/2022] Open
Abstract
Loss-of-function mutations in the secreted enzyme ADAMTS7 (a disintegrin and metalloproteinase with thrombospondin motifs 7) are associated with protection for coronary artery disease. ADAMTS7 catalytic inhibition has been proposed as a therapeutic strategy for treating coronary artery disease; however, the lack of an endogenous substrate has hindered the development of activity-based biomarkers. To identify ADAMTS7 extracellular substrates and their cleavage sites relevant to vascular disease, we used TAILS (terminal amine isotopic labeling of substrates), a method for identifying protease-generated neo-N termini. We compared the secreted proteome of vascular smooth muscle and endothelial cells expressing either full-length mouse ADAMTS7 WT, catalytic mutant ADAMTS7 E373Q, or a control luciferase adenovirus. Significantly enriched N-terminal cleavage sites in ADAMTS7 WT samples were compared to the negative control conditions and filtered for stringency, resulting in catalogs of high confidence candidate ADAMTS7 cleavage sites from our three independent TAILS experiments. Within the overlap of these discovery sets, we identified 24 unique cleavage sites from 16 protein substrates, including cleavage sites in EFEMP1 (EGF-containing fibulin-like extracellular matrix protein 1/Fibulin-3). The ADAMTS7 TAILS preference for EFEMP1 cleavage at the amino acids 123.124 over the adjacent 124.125 site was validated using both endogenous EFEMP1 and purified EFEMP1 in a binary in vitro cleavage assay. Collectively, our TAILS discovery experiments have uncovered hundreds of potential substrates and cleavage sites to explore disease-related biological substrates and facilitate activity-based ADAMTS7 biomarker development.
Collapse
Affiliation(s)
- Bryan T MacDonald
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.
| | - Hasmik Keshishian
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Charles C Mundorff
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Alessandro Arduini
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Daniel Lai
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kayla Bendinelli
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Nicholas R Popp
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Bidur Bhandary
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Karl R Clauser
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Harrison Specht
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Nadine H Elowe
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Dylan Laprise
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yi Xing
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Virendar K Kaushik
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Steven A Carr
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
31
|
Collantes ERA, Delfin MS, Fan B, Torregosa JMR, Siguan-Bell C, de Guzman Florcruz NV, Martinez JMD, Masna-Hidalgo BJ, Guzman VPT, Anotado-Flores JF, Levina FD, Hernandez SRC, Collantes AA, Sibulo MC, Rong S, Wiggs JL. EFEMP1 rare variants cause familial juvenile-onset open-angle glaucoma. Hum Mutat 2022; 43:240-252. [PMID: 34923728 PMCID: PMC8972201 DOI: 10.1002/humu.24320] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 12/02/2021] [Accepted: 12/15/2021] [Indexed: 02/03/2023]
Abstract
Juvenile open-angle glaucoma (JOAG) is a severe type of glaucoma with onset before age 40 and dominant inheritance. Using exome sequencing we identified 3 independent families from the Philippines with novel EFEMP1 variants (c.238A>T, p.Asn80Tyr; c.1480T>C, p.Ter494Glnext*29; and c.1429C>T, p.Arg477Cys) co-segregating with disease. Affected variant carriers (N = 34) exhibited severe disease with average age of onset of 16 years and with 76% developing blindness. To investigate functional effects, we transfected COS7 cells with vectors expressing the three novel EFEMP1 variants and showed that all three variants found in JOAG patients caused significant intracellular protein aggregation and retention compared to wild type and also compared to EFEMP1 variants associated with other ocular phenotypes including an early-onset form of macular degeneration, Malattia Leventinese/Doyne's Honeycomb retinal dystrophy. These results suggest that rare EFEMP1 coding variants can cause JOAG through a mechanism involving protein aggregation and retention, and that the extent of intracellular retention correlates with disease phenotype. This is the first report of EFEMP1 variants causing JOAG, expanding the EFEMP1 disease spectrum. Our results suggest that EFEMP1 mutations appear to be a relatively common cause of JOAG in Filipino families, an ethnically diverse population.
Collapse
Affiliation(s)
- Edward Ryan A. Collantes
- Harvard Medical School Department of Ophthalmology, Massachusetts Eye and Ear, Boston, USA
- Department of Ophthalmology, Manila Doctors Hospital, Manila, Philippines
| | - Manuel S. Delfin
- Department of Ophthalmology, Manila Doctors Hospital, Manila, Philippines
| | - Baojian Fan
- Harvard Medical School Department of Ophthalmology, Massachusetts Eye and Ear, Boston, USA
| | - Justine May R. Torregosa
- Department of Ophthalmology, Cebu Institute of Medicine, Cebu Velez General Hospital, Cebu City, Philippines
| | - Christine Siguan-Bell
- Department of Ophthalmology, Cebu Institute of Medicine, Cebu Velez General Hospital, Cebu City, Philippines
| | - Nilo Vincent de Guzman Florcruz
- Department of Ophthalmology and Visual Sciences, University of the Philippines-Philippine General Hospital, Manila, Philippines
- Glaucoma Service, Department of Ophthalmology, East Avenue Medical Center, Quezon City, Philippines
| | - Jose Maria D. Martinez
- Glaucoma Service, Department of Ophthalmology, East Avenue Medical Center, Quezon City, Philippines
| | | | | | | | - Faye D. Levina
- Department of Ophthalmology, Jose R. Reyes Memorial Medical Center, Manila, Philippines
| | | | | | | | - Shisong Rong
- Harvard Medical School Department of Ophthalmology, Massachusetts Eye and Ear, Boston, USA
| | - Janey L. Wiggs
- Harvard Medical School Department of Ophthalmology, Massachusetts Eye and Ear, Boston, USA
| |
Collapse
|
32
|
Allen-Brady K, Bortolini MAT, Damaser MS. Mouse Knockout Models for Pelvic Organ Prolapse: a Systematic Review. Int Urogynecol J 2022; 33:1765-1788. [PMID: 35088092 DOI: 10.1007/s00192-021-05066-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/13/2021] [Indexed: 02/03/2023]
Abstract
INTRODUCTION AND HYPOTHESIS Mouse knockout (KO) models of pelvic organ prolapse (POP) have contributed mechanistic evidence for the role of connective tissue defects, specifically impaired elastic matrix remodeling. Our objective was to summarize what mouse KO models for POP are available and what have we learned from these mouse models about the pathophysiological mechanisms of POP development. METHODS We conducted a systematic review and reported narrative findings according to PRISMA guidelines. Two independent reviewers searched PubMed, Scopus and Embase for relevant manuscripts and conference abstracts for the time frame of January 1, 2000, to March 31, 2021. Conference abstracts were limited to the past 5 years. RESULTS The search strategy resulted in 294 total titles. We ultimately included 25 articles and an additional 11 conference abstracts. Five KO models have been studied: Loxl1, Fbln5, Fbln3, Hoxa11 and Upii-sv40t. Loxl1 and Fbln5 KO models have provided the most reliable and predictable POP phenotype. Loxl1 KO mice develop POP primarily from failure to heal after giving birth, whereas Fbln5 KO mice develop POP with aging. These mouse KO models have been used for a wide variety of investigations including genetic pathways involved in development of POP, biomechanical properties of the pelvic floor, elastic fiber deposition, POP therapies and the pathophysiology associated with mesh complications. CONCLUSIONS Mouse KO models have proved to be a valuable tool in the study of specific genes and their role in the development and progression of POP. They may be useful to study POP treatments and POP complications.
Collapse
Affiliation(s)
- Kristina Allen-Brady
- Department of Internal Medicine, University of Utah, Williams Building 295 Chipeta Way, Salt Lake City, UT, USA.
| | - Maria A T Bortolini
- Department of Gynecology, Sector of Urogynecology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Margot S Damaser
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| |
Collapse
|
33
|
Li Y, Nie N, Gong L, Bao F, An C, Cai H, Yao X, Liu Y, Yang C, Wu B, Zou X. Structural, functional and molecular pathogenesis of pelvic organ prolapse in patient and Loxl1 deficient mice. Aging (Albany NY) 2021; 13:25886-25902. [PMID: 34923484 PMCID: PMC8751609 DOI: 10.18632/aging.203777] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/11/2021] [Indexed: 12/02/2022]
Abstract
Pelvic organ prolapse is a worldwide health problem to elderly women. Understanding its pathogenesis and an ideal animal model are crucial to developing promising treatments. The present study aimed to investigate new clinical significance and detailed mechanism of pelvic organ prolapse by comparing the structural, functional and molecular dysfunctions of pelvic organ prolapse in patient and Loxl1 deficient mice. Our results showed that human vagina tissues from prolapsed site showed disarranged collagen and elastic fibers compared with the non-prolapse tissue. A gene ontology (GO) analysis of differentially expressed genes revealed molecular changes mainly related to inflammatory response and extracellular matrix (ECM) organization. While the mice lacking Loxl1 developed stable POP phenotype and disordered ECM structure in histology. Such Loxl1 knockout mice exhibited a significantly urinary dysfunction and decreased mechanical properties of the pelvic floor tissues, implying that POP in human condition might be induced by progressively decreased mechanics of pelvic tissues following ECM catabolism. Similarly, we not only identified significant up-regulated ECM catabolism processes and down-regulated ECM synthesis processes, but also characterized high level of inflammatory response in vagina tissue of the Loxl1 deficient mice. Thus, all these pathological changes in the POP mice model was consistent with those of the clinical elderly patients. These findings provide new insight into remodeling of POP by LOXL1 regulation and be of great importance to develop combination treatments of ECM metabolism and inflammation regulation strategy.
Collapse
Affiliation(s)
- Yu Li
- Clinical Research Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| | - Nanfang Nie
- Clinical Research Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| | - Lin Gong
- Clinical Research Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| | - Fangyuan Bao
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| | - Chengrui An
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| | - Hongxia Cai
- Clinical Research Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| | - Xudong Yao
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Department of Gynaecology, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| | - Yanshan Liu
- Clinical Research Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| | - Chunbo Yang
- Department of Gynaecology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| | - Bingbing Wu
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Department of Gynaecology, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| | - XiaoHui Zou
- Clinical Research Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang 310003, PR China
| |
Collapse
|
34
|
Fu KA, Paul KC, Lu AT, Horvath S, Keener AM, Bordelon Y, Bronstein JM, Ritz B. DNA methylation-based surrogates of plasma proteins are associated with Parkinson's disease risk. J Neurol Sci 2021; 431:120046. [PMID: 34768133 DOI: 10.1016/j.jns.2021.120046] [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: 07/23/2021] [Revised: 09/24/2021] [Accepted: 11/01/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND The epigenome may reflect Parkinson's disease (PD) risk, which serves as a point of convergence of genetic and environmental risk factors. Here, we investigate whether blood DNA methylation (DNAm) markers are associated with PD risk. METHODS We selected 12 plasma proteins known as predictors of cardiovascular conditions and mortality to evaluate their effects on PD risk in a case-control study. In lieu of protein level measures, however, we assessed the influence of their DNAm surrogates. Primary analysis was restricted to 569 PD patients and 238 controls with DNAm data available. Using univariate logistic regression, we evaluated associations between the DNAm markers and PD. RESULTS Of the 12 DNAm surrogates, the most robustly associated were DNAm EFEMP-1 and DNAm CD56, which were associated with PD with and without controlling for blood cell composition. DNAm EFEMP-1 was associated with a decreased risk of PD (OR = 0.83 per SD, 95% CI = 0.70, 0.98) whereas DNAm CD56 was associated with an increased risk of PD (OR = 1.41, 95% CI = 1.11, 1.79). CONCLUSIONS Several DNAm markers, selected as part of a panel to track cardiovascular outcomes and mortality, were associated with PD risk. DNAm markers may inform of factors that are affected differentially in early PD patients compared with controls.
Collapse
Affiliation(s)
- Katherine A Fu
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA.
| | - Kimberly C Paul
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA
| | - Ake T Lu
- Department of Human Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Steve Horvath
- Department of Human Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA; Department of Biostatistics, UCLA Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Adrienne M Keener
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA; Department of Neurology, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Yvette Bordelon
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Jeff M Bronstein
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Beate Ritz
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA; Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA.
| |
Collapse
|
35
|
Yue B, Wang Y, Zhang C, Ding Y, Liu Z. Efficacy of Shaobei injection in the treatment of grade II-III hemorrhoids and the effect on fibulin protein expression: A study protocol of a randomized controlled trial. Medicine (Baltimore) 2021; 100:e27706. [PMID: 34797296 PMCID: PMC8601367 DOI: 10.1097/md.0000000000027706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 10/21/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Hemorrhoids are a common and seriously disruptive condition that seriously affects people's lives in terms of treatment. Injection therapy is an effective minimally invasive scheme for the treatment of grade II-III hemorrhoids, but its clinical application is limited by the adverse reactions caused by injection drugs. Some clinical studies have confirmed the efficacy and safety of Shaobei injection as a traditional Chinese medicine extract. However, there is no standard randomized controlled study to verify its efficacy and explore its potential mechanism. METHODS This is a prospective, randomized, single blind, parallel controlled trial to study the efficacy of Shaobei injection in the treatment of grade II-III hemorrhoids and its effect on the expression of fibulin-3 and fibulin-5 in fibulin protein family. The patients will be randomly divided into a treatment group and control group. The treatment group will be treated with Shaobei injection, and the control group will be treated with rubber band ligation. The observation indexes include: visual analysis scale, postoperative hospital stay, total use of painkillers, fibulin-3 and fibulin-5, hemorrhoids recurrence, and adverse events. Finally, the data will be statistically analyzed by SPASS 18.0 software. DISCUSSION This study will compare the efficacy of Shaobei injection with the rubber band ligation method in the treatment of grade II-III haemorrhoids and investigate its effect on the expression of fibulin-3 and fibulin-5 in the fibulin protein family. The results of this study will provide a basis for the clinical use of Paeoniflora injection as an alternative to traditional sclerosing agent in the treatment of grade II-III haemorrhoids.Trial registration: OSF Registration number:DOI 10.17605/OSF.IO/MKVDB.
Collapse
Affiliation(s)
- Bin Yue
- Shenyang Coloproctology Hospital, Shenyang City, Liaoning Province, China
| | - Yangang Wang
- Weifang People's Hospital, Weifang, Shandong Province, China
| | - Chunxia Zhang
- Shenyang Coloproctology Hospital, Shenyang City, Liaoning Province, China
| | - Yunlong Ding
- Weifang People's Hospital, Weifang, Shandong Province, China
| | - Zhipeng Liu
- Weifang People's Hospital, Weifang, Shandong Province, China
| |
Collapse
|
36
|
Pietzner M, Wheeler E, Carrasco-Zanini J, Cortes A, Koprulu M, Wörheide MA, Oerton E, Cook J, Stewart ID, Kerrison ND, Luan J, Raffler J, Arnold M, Arlt W, O’Rahilly S, Kastenmüller G, Gamazon ER, Hingorani AD, Scott RA, Wareham NJ, Langenberg C. Mapping the proteo-genomic convergence of human diseases. Science 2021; 374:eabj1541. [PMID: 34648354 PMCID: PMC9904207 DOI: 10.1126/science.abj1541] [Citation(s) in RCA: 264] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Characterization of the genetic regulation of proteins is essential for understanding disease etiology and developing therapies. We identified 10,674 genetic associations for 3892 plasma proteins to create a cis-anchored gene-protein-disease map of 1859 connections that highlights strong cross-disease biological convergence. This proteo-genomic map provides a framework to connect etiologically related diseases, to provide biological context for new or emerging disorders, and to integrate different biological domains to establish mechanisms for known gene-disease links. Our results identify proteo-genomic connections within and between diseases and establish the value of cis-protein variants for annotation of likely causal disease genes at loci identified in genome-wide association studies, thereby addressing a major barrier to experimental validation and clinical translation of genetic discoveries.
Collapse
Affiliation(s)
- Maik Pietzner
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK,Computational Medicine, Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Eleanor Wheeler
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Julia Carrasco-Zanini
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | | | - Mine Koprulu
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Maria A. Wörheide
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Erin Oerton
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - James Cook
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Isobel D. Stewart
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Nicola D. Kerrison
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Jian’an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Johannes Raffler
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany,Institut für Digitale Medizin, Universitätsklinikum Augsburg, 86156 Augsburg, Germany
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany,Department of Psychiatry and Behavioural Sciences, Duke University, Durham, NC 27710, USA
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Stephen O’Rahilly
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany,German Centre for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Eric R. Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA,Clare Hall, University of Cambridge, Cambridge CB3 9AL, United Kingdom
| | - Aroon D. Hingorani
- UCL British Heart Foundation Research Accelerator, Institute of Cardiovascular Science, University College London, WC1E 6BT, UK.,Health Data Research UK, Gibbs Building, 215 Euston Road, London NW1 2BE, UK,Institute of Health Informatics, University College London, 222 Euston Road, London NW1 2DA, UK
| | | | - Nicholas J. Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK,Health Data Research UK, Gibbs Building, 215 Euston Road, London NW1 2BE, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK,Computational Medicine, Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, 10117 Berlin, Germany,Health Data Research UK, Gibbs Building, 215 Euston Road, London NW1 2BE, UK,Correspondence to Dr. Claudia Langenberg ()
| |
Collapse
|
37
|
Garland DL, Pierce EA, Fernandez-Godino R. Complement C5 is not critical for the formation of sub-RPE deposits in Efemp1 mutant mice. Sci Rep 2021; 11:10416. [PMID: 34001980 PMCID: PMC8128922 DOI: 10.1038/s41598-021-89978-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/05/2021] [Indexed: 12/27/2022] Open
Abstract
The complement system plays a role in the formation of sub-retinal pigment epithelial (RPE) deposits in early stages of age-related macular degeneration (AMD). But the specific mechanisms that connect complement activation and deposit formation in AMD patients are unknown, which limits the development of efficient therapies to reduce or stop disease progression. We have previously demonstrated that C3 blockage prevents the formation of sub-RPE deposits in a mouse model of EFEMP1-associated macular degeneration. In this study, we have used double mutant Efemp1R345W/R345W:C5-/- mice to investigate the role of C5 in the formation of sub-RPE deposits in vivo and in vitro. The data revealed that the genetic ablation of C5 does not eliminate the formation of sub-RPE deposits. Contrarily, the absence of C5 in RPE cultures promotes complement dysregulation that results in increased activation of C3, which likely contributes to deposit formation even in the absence of EFEMP1-R345W mutant protein. The results also suggest that genetic ablation of C5 alters the extracellular matrix turnover through an effect on matrix metalloproteinases in RPE cell cultures. These results confirm that C3 rather than C5 could be an effective therapeutic target to treat early AMD.
Collapse
Affiliation(s)
- Donita L Garland
- Ocular Genomics Institute at Massachusetts Eye and Ear, Harvard Medical School, Boston, USA
| | - Eric A Pierce
- Ocular Genomics Institute at Massachusetts Eye and Ear, Harvard Medical School, Boston, USA
| | | |
Collapse
|
38
|
Tsai YT, Li Y, Ryu J, Su PY, Cheng CH, Wu WH, Li YS, Quinn PMJ, Leong KW, Tsang SH. Impaired cholesterol efflux in retinal pigment epithelium of individuals with juvenile macular degeneration. Am J Hum Genet 2021; 108:903-918. [PMID: 33909993 PMCID: PMC8206198 DOI: 10.1016/j.ajhg.2021.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Macular degeneration (MD) is characterized by the progressive deterioration of the macula and represents one of the most prevalent causes of blindness worldwide. Abnormal intracellular accumulation of lipid droplets and pericellular deposits of lipid-rich material in the retinal pigment epithelium (RPE) called drusen are clinical hallmarks of different forms of MD including Doyne honeycomb retinal dystrophy (DHRD) and age-related MD (AMD). However, the appropriate molecular therapeutic target underlying these disorder phenotypes remains elusive. Here, we address this knowledge gap by comparing the proteomic profiles of induced pluripotent stem cell (iPSC)-derived RPEs (iRPE) from individuals with DHRD and their isogenic controls. Our analysis and follow-up studies elucidated the mechanism of lipid accumulation in DHRD iRPE cells. Specifically, we detected significant downregulation of carboxylesterase 1 (CES1), an enzyme that converts cholesteryl ester to free cholesterol, an indispensable process in cholesterol export. CES1 knockdown or overexpression of EFEMP1R345W, a variant of EGF-containing fibulin extracellular matrix protein 1 that is associated with DHRD and attenuated cholesterol efflux and led to lipid droplet accumulation. In iRPE cells, we also found that EFEMP1R345W has a hyper-inhibitory effect on epidermal growth factor receptor (EGFR) signaling when compared to EFEMP1WT and may suppress CES1 expression via the downregulation of transcription factor SP1. Taken together, these results highlight the homeostatic role of cholesterol efflux in iRPE cells and identify CES1 as a mediator of cholesterol efflux in MD.
Collapse
Key Words
- age-related macular degeneration, Doyne honeycomb destrophy, DHRD, cholesterol efflux, drusen, RPE, CRISPR, isogenic, EGFR signaling, unfolded protein response, lipid accumulation
Collapse
Affiliation(s)
- Yi-Ting Tsai
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Yao Li
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Joseph Ryu
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Pei-Yin Su
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Chia-Hua Cheng
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Wen-Hsuan Wu
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Yong-Shi Li
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Peter M J Quinn
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA; Department of Pathology & Cell Biology, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
39
|
Role of Fibulins in Embryonic Stage Development and Their Involvement in Various Diseases. Biomolecules 2021; 11:biom11050685. [PMID: 34063320 PMCID: PMC8147605 DOI: 10.3390/biom11050685] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/24/2022] Open
Abstract
The extracellular matrix (ECM) plays an important role in the evolution of early metazoans, as it provides structural and biochemical support to the surrounding cells through the cell–cell and cell–matrix interactions. In multi-cellular organisms, ECM plays a pivotal role in the differentiation of tissues and in the development of organs. Fibulins are ECM glycoproteins, found in a variety of tissues associated with basement membranes, elastic fibers, proteoglycan aggregates, and fibronectin microfibrils. The expression profile of fibulins reveals their role in various developmental processes such as elastogenesis, development of organs during the embryonic stage, tissue remodeling, maintenance of the structural integrity of basement membrane, and elastic fibers, as well as other cellular processes. Apart from this, fibulins are also involved in the progression of human diseases such as cancer, cardiac diseases, congenital disorders, and chronic fibrotic disorders. Different isoforms of fibulins show a dual role of tumor-suppressive and tumor-promoting activities, depending on the cell type and cellular microenvironment in the body. Knockout animal models have provided deep insight into their role in development and diseases. The present review covers details of the structural and expression patterns, along with the role of fibulins in embryonic development and disease progression, with more emphasis on their involvement in the modulation of cancer diseases.
Collapse
|
40
|
Perez NP, Chang DC, Goldstone RN, Bordeianou L, Ricciardi R, Cavallaro PM. Relationship Between Diverticular Disease and Incisional Hernia After Elective Colectomy: a Population-Based Study. J Gastrointest Surg 2021; 25:1297-1306. [PMID: 32748338 PMCID: PMC7854815 DOI: 10.1007/s11605-020-04762-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Recent genetic studies identified common mutations between diverticular disease and connective tissue disorders, some of which are associated with abdominal wall hernias. Scarce data exists, however, shedding light on the potential clinical implications of this shared etiology, particularly in the era of laparoscopic surgery. METHODS The New York Statewide Planning and Research Cooperative System database was used to identify adult patients undergoing elective sigmoid and left hemicolectomy (open or laparoscopic) from January 1, 2010, to December 31, 2016, for diverticulitis or descending/sigmoid colon cancer. The incidences of incisional hernia diagnosis and repair were compared using competing risks regression models, clustered by surgeon and adjusted for a host of demographic/clinical variables. Subsequent abdominal surgery and death were considered competing risks. RESULTS Among 8279 patients included in the study cohort, 6811 (82.2%) underwent colectomy for diverticulitis and 1468 (17.8%) for colon cancer. The overall 5-year risk of incisional hernia was 3.5% among patients with colon cancer, regardless of colectomy route, which was significantly lower than that among diverticulitis patients after both open (10.7%; p < 0.001) and laparoscopic (7.2%; p = 0.007) colectomies. Multivariable analyses demonstrated that patients with diverticulitis experienced a two-fold increase in the risk for hernia diagnosis (aHR 1.8; p < 0.001) and repair (aHR 2.1; p < 0.001), and these findings persisted after stratification by colectomy route. CONCLUSIONS Patients undergoing elective colectomy for diverticulitis, including via laparoscopic approach, experience higher rates of incisional hernia compared with patients undergoing similar resections for colon cancer. When performing resections for diverticulitis, surgeons should strongly consider adherence to evidence-based guidelines for fascial closure to prevent this important complication.
Collapse
Affiliation(s)
- Numa P Perez
- Department of Surgery, Massachusetts General Hospital, 55 Fruit St, GRB-425, Boston, MA, 02114, USA.
- Healthcare Transformation Lab, Massachusetts General Hospital, Boston, MA, USA.
| | - David C Chang
- Codman Center for Clinical Effectiveness in Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Robert N Goldstone
- Department of Surgery, Massachusetts General Hospital, 55 Fruit St, GRB-425, Boston, MA, 02114, USA
| | - Liliana Bordeianou
- Department of Surgery, Massachusetts General Hospital, 55 Fruit St, GRB-425, Boston, MA, 02114, USA
| | - Rocco Ricciardi
- Department of Surgery, Massachusetts General Hospital, 55 Fruit St, GRB-425, Boston, MA, 02114, USA
| | - Paul M Cavallaro
- Department of Surgery, Massachusetts General Hospital, 55 Fruit St, GRB-425, Boston, MA, 02114, USA
| |
Collapse
|
41
|
Abstract
Age-associated changes in gene expression in skeletal muscle of healthy individuals reflect accumulation of damage and compensatory adaptations to preserve tissue integrity. To characterize these changes, RNA was extracted and sequenced from muscle biopsies collected from 53 healthy individuals (22-83 years old) of the GESTALT study of the National Institute on Aging-NIH. Expression levels of 57,205 protein-coding and non-coding RNAs were studied as a function of aging by linear and negative binomial regression models. From both models, 1134 RNAs changed significantly with age. The most differentially abundant mRNAs encoded proteins implicated in several age-related processes, including cellular senescence, insulin signaling, and myogenesis. Specific mRNA isoforms that changed significantly with age in skeletal muscle were enriched for proteins involved in oxidative phosphorylation and adipogenesis. Our study establishes a detailed framework of the global transcriptome and mRNA isoforms that govern muscle damage and homeostasis with age.
Collapse
|
42
|
Verlee M, Beyens A, Gezdirici A, Gulec EY, Pottie L, De Feyter S, Vanhooydonck M, Tapaneeyaphan P, Symoens S, Callewaert B. Loss-of-Function Variants in EFEMP1 Cause a Recognizable Connective Tissue Disorder Characterized by Cutis Laxa and Multiple Herniations. Genes (Basel) 2021; 12:genes12040510. [PMID: 33807164 PMCID: PMC8066907 DOI: 10.3390/genes12040510] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/27/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022] Open
Abstract
Hereditary disorders of connective tissue (HDCT) compromise a heterogeneous group of diseases caused by pathogenic variants in genes encoding different components of the extracellular matrix and characterized by pleiotropic manifestations, mainly affecting the cutaneous, cardiovascular, and musculoskeletal systems. We report the case of a 9-year-old boy with a discernible connective tissue disorder characterized by cutis laxa (CL) and multiple herniations and caused by biallelic loss-of-function variants in EFEMP1. Hence, we identified EFEMP1 as a novel disease-causing gene in the CL spectrum, differentiating it from other HDCT.
Collapse
Affiliation(s)
- Maxim Verlee
- Center for Medical Genetics Ghent, Ghent University Hospital, 9000 Ghent, Belgium; (M.V.); (A.B.); (L.P.); (S.D.F.); (M.V.); (P.T.); (S.S.)
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Aude Beyens
- Center for Medical Genetics Ghent, Ghent University Hospital, 9000 Ghent, Belgium; (M.V.); (A.B.); (L.P.); (S.D.F.); (M.V.); (P.T.); (S.S.)
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Department of Dermatology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Alper Gezdirici
- Department of Medical Genetics, Basaksehir Cam and Sakura City Hospital, 34480 Istanbul, Turkey;
| | - Elif Yilmaz Gulec
- Department of Medical Genetics, Kanuni Sultan Suleyman Training and Research Hospital, Health Sciences University, 34303 Istanbul, Turkey;
| | - Lore Pottie
- Center for Medical Genetics Ghent, Ghent University Hospital, 9000 Ghent, Belgium; (M.V.); (A.B.); (L.P.); (S.D.F.); (M.V.); (P.T.); (S.S.)
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Silke De Feyter
- Center for Medical Genetics Ghent, Ghent University Hospital, 9000 Ghent, Belgium; (M.V.); (A.B.); (L.P.); (S.D.F.); (M.V.); (P.T.); (S.S.)
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Michiel Vanhooydonck
- Center for Medical Genetics Ghent, Ghent University Hospital, 9000 Ghent, Belgium; (M.V.); (A.B.); (L.P.); (S.D.F.); (M.V.); (P.T.); (S.S.)
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Piyanoot Tapaneeyaphan
- Center for Medical Genetics Ghent, Ghent University Hospital, 9000 Ghent, Belgium; (M.V.); (A.B.); (L.P.); (S.D.F.); (M.V.); (P.T.); (S.S.)
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Sofie Symoens
- Center for Medical Genetics Ghent, Ghent University Hospital, 9000 Ghent, Belgium; (M.V.); (A.B.); (L.P.); (S.D.F.); (M.V.); (P.T.); (S.S.)
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bert Callewaert
- Center for Medical Genetics Ghent, Ghent University Hospital, 9000 Ghent, Belgium; (M.V.); (A.B.); (L.P.); (S.D.F.); (M.V.); (P.T.); (S.S.)
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Correspondence:
| |
Collapse
|
43
|
Koba T, Takeda Y, Narumi R, Shiromizu T, Nojima Y, Ito M, Kuroyama M, Futami Y, Takimoto T, Matsuki T, Edahiro R, Nojima S, Hayama Y, Fukushima K, Hirata H, Koyama S, Iwahori K, Nagatomo I, Suzuki M, Shirai Y, Murakami T, Nakanishi K, Nakatani T, Suga Y, Miyake K, Shiroyama T, Kida H, Sasaki T, Ueda K, Mizuguchi K, Adachi J, Tomonaga T, Kumanogoh A. Proteomics of serum extracellular vesicles identifies a novel COPD biomarker, fibulin-3 from elastic fibres. ERJ Open Res 2021; 7:00658-2020. [PMID: 33778046 PMCID: PMC7983195 DOI: 10.1183/23120541.00658-2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/18/2020] [Indexed: 12/28/2022] Open
Abstract
There is an unmet need for novel biomarkers in the diagnosis of multifactorial COPD. We applied next-generation proteomics to serum extracellular vesicles (EVs) to discover novel COPD biomarkers. EVs from 10 patients with COPD and six healthy controls were analysed by tandem mass tag-based non-targeted proteomics, and those from elastase-treated mouse models of emphysema were also analysed by non-targeted proteomics. For validation, EVs from 23 patients with COPD and 20 healthy controls were validated by targeted proteomics. Using non-targeted proteomics, we identified 406 proteins, 34 of which were significantly upregulated in patients with COPD. Of note, the EV protein signature from patients with COPD reflected inflammation and remodelling. We also identified 63 upregulated candidates from 1956 proteins by analysing EVs isolated from mouse models. Combining human and mouse biomarker candidates, we validated 45 proteins by targeted proteomics, selected reaction monitoring. Notably, levels of fibulin-3, tripeptidyl-peptidase 2, fibulin-1, and soluble scavenger receptor cysteine-rich domain-containing protein were significantly higher in patients with COPD. Moreover, six proteins; fibulin-3, tripeptidyl-peptidase 2, UTP-glucose-1-phosphate uridylyl transferase, CD81, CD177, and oncoprotein-induced transcript 3, were correlated with emphysema. Upregulation of fibulin-3 was confirmed by immunoblotting of EVs and immunohistochemistry in lungs. Strikingly, fibulin-3 knockout mice spontaneously developed emphysema with age, as evidenced by alveolar enlargement and elastin destruction. We discovered potential pathogenic biomarkers for COPD using next-generation proteomics of EVs. This is a novel strategy for biomarker discovery and precision medicine. This study identified novel biomarkers for COPD using next-generation proteomics of serum extracellular vesicles. Notably, the expression of fibulin-3 is correlated with lung function and emphysema. This could be useful for personalised medicine.https://bit.ly/2JfRCgk
Collapse
Affiliation(s)
- Taro Koba
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshito Takeda
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryohei Narumi
- Laboratory of Proteome Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Takashi Shiromizu
- Laboratory of Proteome Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Yosui Nojima
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Mari Ito
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Muneyoshi Kuroyama
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yu Futami
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takayuki Takimoto
- Dept of Respiratory Internal Medicine, National Hospital Organization Kinki-Chuo Chest Medical Center, Kita-Ku, Sakai, Osaka, Japan
| | - Takanori Matsuki
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryuya Edahiro
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Satoshi Nojima
- Dept of Pathology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshitomo Hayama
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kiyoharu Fukushima
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Haruhiko Hirata
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shohei Koyama
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kota Iwahori
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Izumi Nagatomo
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Mayumi Suzuki
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuya Shirai
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Teruaki Murakami
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kaori Nakanishi
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takeshi Nakatani
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yasuhiko Suga
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kotaro Miyake
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takayuki Shiroyama
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroshi Kida
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takako Sasaki
- Dept of Biochemistry II, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Koji Ueda
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Koto, Tokyo, Japan
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Jun Adachi
- Laboratory of Proteome Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Atsushi Kumanogoh
- Dept of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
44
|
Sheyanth IN, Lolas IB, Okkels H, Kiruparajan LP, Abildgaard SK, Petersen MB. First reported case of Doyne honeycomb retinal dystrophy (Malattia Leventinese/autosomal dominant drusen) in Scandinavia. Mol Genet Genomic Med 2021; 9:e1652. [PMID: 33689237 PMCID: PMC8123724 DOI: 10.1002/mgg3.1652] [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: 08/25/2020] [Revised: 11/11/2020] [Accepted: 02/19/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Doyne honeycomb retinal dystrophy (DHRD)/malattia leventinese (ML) is an autosomal dominant, progressive retinal disorder characterized by massive central retinal drusen often partly coalescent forming a characteristic honeycomb-like pattern. Debut of vision loss often occurs in early to mid-adulthood, and the degree varies. A single variant in EFEMP1: c.1033C>T (R345W) has been identified as the cause in all cases. METHODS Following DNA isolation, exome sequencing was performed in seven genes associated with flecked retina. Direct sequencing was used for variant verification. RESULTS We report the first Scandinavian case of molecular genetically verified DHRD/ML: a 57-year-old woman debuting with vision loss and metamorphopsia. On both eyes, ophthalmological findings included massive hard drusen in the macular region and nasal to the optic disc as well as macular hyperpigmentation. Secondary choroidal neovascularizations were identified on both eyes, and anti-vascular endothelial growth factor was administered, without effect. CONCLUSION Molecular genetic investigation revealed heterozygosity for the known pathogenic missense variant in EFEMP1: c.1033C>T (R345W) previously reported in relation to DHRD/ML. Family history revealed no other cases of similar visual impairment suggesting a de novo mutation. Furthermore, there was no correlation between the unique DHRD/ML haplotypes reported in the literature and our patient.
Collapse
Affiliation(s)
- Inger Norlyk Sheyanth
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Ihab Bishara Lolas
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Molecular Diagnostics, Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Henrik Okkels
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Molecular Diagnostics, Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Ligor Pradeep Kiruparajan
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Ophthalmology, Aalborg University Hospital, Aalborg, Denmark
| | - Søren Kromann Abildgaard
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Ophthalmology, Aalborg University Hospital, Aalborg, Denmark
| | - Michael Bjørn Petersen
- Research and Knowledge Center in Sensory Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Genetics, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
45
|
Palmer D, Fabris F, Doherty A, Freitas AA, de Magalhães JP. Ageing transcriptome meta-analysis reveals similarities and differences between key mammalian tissues. Aging (Albany NY) 2021; 13:3313-3341. [PMID: 33611312 PMCID: PMC7906136 DOI: 10.18632/aging.202648] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022]
Abstract
By combining transcriptomic data with other data sources, inferences can be made about functional changes during ageing. Thus, we conducted a meta-analysis on 127 publicly available microarray and RNA-Seq datasets from mice, rats and humans, identifying a transcriptomic signature of ageing across species and tissues. Analyses on subsets of these datasets produced transcriptomic signatures of ageing for brain, heart and muscle. We then applied enrichment analysis and machine learning to functionally describe these signatures, revealing overexpression of immune and stress response genes and underexpression of metabolic and developmental genes. Further analyses revealed little overlap between genes differentially expressed with age in different tissues, despite ageing differentially expressed genes typically being widely expressed across tissues. Additionally we show that the ageing gene expression signatures (particularly the overexpressed signatures) of the whole meta-analysis, brain and muscle tend to include genes that are central in protein-protein interaction networks. We also show that genes underexpressed with age in the brain are highly central in a co-expression network, suggesting that underexpression of these genes may have broad phenotypic consequences. In sum, we show numerous functional similarities between the ageing transcriptomes of these important tissues, along with unique network properties of genes differentially expressed with age in both a protein-protein interaction and co-expression networks.
Collapse
Affiliation(s)
- Daniel Palmer
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK.,Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| | - Fabio Fabris
- School of Computing, University of Kent, Canterbury, Kent, UK
| | - Aoife Doherty
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Alex A Freitas
- School of Computing, University of Kent, Canterbury, Kent, UK
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| |
Collapse
|
46
|
Peng X, Guo Z, Zhang Y, Sun B, Zhang Q. EFEMP1 in Direct Inguinal Hernia: correlation with TIMP3 and Regulation Toward Elastin Homoeostasis as Well as Fibroblast Mobility. J INVEST SURG 2020; 35:203-211. [PMID: 33131351 DOI: 10.1080/08941939.2020.1811812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AIM This basic research aimed to detect the inner-correlation of EGF containing fibulin extracellular matrix protein 1 (EFEMP1), TIMP metallopeptidase inhibitor 3 (TIMP3), matrix metalloprotease 9 (MMP9), elastin (ELN) in direct inguinal hernia (IH), and their effect on fibroblasts motility. METHODS Transversalis fascia samples from 20 direct IH patients and 20 varicocele (served as controls) patients were collected for detecting EFEMP1, TIMP3, MMP9 and ELN expressions by immunohistochemistry assay. Fibroblasts L929 cells were transfected with EFEMP1 overexpression plasmid or knock-down plasmid to investigate the influence of EFEMP1 dysregulation on L929 cell migration, invasion, TIMP3, MMP9 and ELN expressions. Additionally, rescue experiments were performed by adding TIMP3 knockdown plasmid to the EFEMP1-overexpressed L929 cells. RESULTS Transversalis fascia EFEMP1, TIMP3 and ELN expressions were decreased, but MMP9 expression was increased in IH patients compared with controls. In IH patients, EFEMP1 was not correlated with TIMP3, but positively correlated with ELN and negatively correlated with MMP9; TIMP3 negatively correlated with MMP9, but positively correlated with ELN. Overexpression of EFEMP1 did not affect TIMP3 expression but increased ELN expression and decreased MMP9 expression in L929 cells. In addition, EFEMP1 suppressed L929 cell migration and invasion. The following rescue experiments indicated that silencing TIMP3 attenuated the effect of EFEMP1 overexpression on MMP9 and ELN expressions as well as the effect of EFEMP1 overexpression on cell migration and invasion in L929 cells. CONCLUSIONS EFEMP1 is downregulated in direct IH, and it regulates ELN homoeostasis as well as fibroblast mobility via interacting with TIMP3.
Collapse
Affiliation(s)
- Xiaohui Peng
- Department of General Surgery, Xiamen Fifth Hospital, Xiamen, China
| | - Zhongwu Guo
- Department of General Surgery, Daqing Oil Field General Hospital, Daqing, Heilongjiang, China
| | - Yinlong Zhang
- Central Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Baichen Sun
- Department of Pathology, Xiamen Fifth Hospital, Xiamen, Fujian, China
| | - Qi Zhang
- Department of General Surgery, Daqing Oil Field General Hospital, Daqing, Heilongjiang, China
| |
Collapse
|
47
|
Noda K, Kitagawa K, Miki T, Horiguchi M, Akama TO, Taniguchi T, Taniguchi H, Takahashi K, Ogra Y, Mecham RP, Terajima M, Yamauchi M, Nakamura T. A matricellular protein fibulin-4 is essential for the activation of lysyl oxidase. SCIENCE ADVANCES 2020; 6:6/48/eabc1404. [PMID: 33239290 PMCID: PMC7688322 DOI: 10.1126/sciadv.abc1404] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/09/2020] [Indexed: 06/11/2023]
Abstract
Fibulin-4 is a matricellular protein required for extracellular matrix (ECM) assembly. Mice deficient in fibulin-4 (Fbln4-/- ) have disrupted collagen and elastin fibers and die shortly after birth from aortic and diaphragmatic rupture. The function of fibulin-4 in ECM assembly, however, remains elusive. Here, we show that fibulin-4 is required for the activity of lysyl oxidase (LOX), a copper-containing enzyme that catalyzes the covalent cross-linking of elastin and collagen. LOX produced by Fbln4-/- cells had lower activity than LOX produced by wild-type cells due to the absence of lysine tyrosyl quinone (LTQ), a unique cofactor required for LOX activity. Our studies showed that fibulin-4 is required for copper ion transfer from the copper transporter ATP7A to LOX in the trans-Golgi network (TGN), which is a necessary step for LTQ formation. These results uncover a pivotal role for fibulin-4 in the activation of LOX and, hence, in ECM assembly.
Collapse
Affiliation(s)
- Kazuo Noda
- Department of Pharmacology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Kaori Kitagawa
- Department of Pharmacology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Takao Miki
- Department of Pharmacology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Masahito Horiguchi
- Department of Emergency Medicine, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto 605-0981, Japan
| | - Tomoya O Akama
- Department of Pharmacology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Takako Taniguchi
- Division of Disease Proteomics, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
| | - Hisaaki Taniguchi
- Division of Disease Proteomics, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
| | - Kazuaki Takahashi
- Laboratory of Toxicology and Environmental Health, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Yasumitsu Ogra
- Laboratory of Toxicology and Environmental Health, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Masahiko Terajima
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Mitsuo Yamauchi
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Tomoyuki Nakamura
- Department of Pharmacology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan.
| |
Collapse
|
48
|
Fibulin-3 knockout mice demonstrate corneal dysfunction but maintain normal retinal integrity. J Mol Med (Berl) 2020; 98:1639-1656. [PMID: 32964303 DOI: 10.1007/s00109-020-01974-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/16/2020] [Accepted: 09/01/2020] [Indexed: 12/17/2022]
Abstract
Fibulin-3 (F3) is an extracellular matrix glycoprotein found in basement membranes across the body. An autosomal dominant R345W mutation in F3 causes a macular dystrophy resembling dry age-related macular degeneration (AMD), whereas genetic removal of wild-type (WT) F3 protects mice from sub-retinal pigment epithelium (RPE) deposit formation. These observations suggest that F3 is a protein which can regulate pathogenic sub-RPE deposit formation in the eye. Yet the precise role of WT F3 within the eye is still largely unknown. We found that F3 is expressed throughout the mouse eye (cornea, trabecular meshwork (TM) ring, neural retina, RPE/choroid, and optic nerve). We next performed a thorough structural and functional characterization of each of these tissues in WT and homozygous (F3-/-) knockout mice. The corneal stroma in F3-/- mice progressively thins beginning at 2 months, and the development of corneal opacity and vascularization starts at 9 months, which worsens with age. However, in all other tissues (TM, neural retina, RPE, and optic nerve), gross structural anatomy and functionality were similar across WT and F3-/- mice when evaluated using SD-OCT, histological analyses, electron microscopy, scotopic electroretinogram, optokinetic response, and axonal anterograde transport. The lack of noticeable retinal abnormalities in F3-/- mice was confirmed in a human patient with biallelic loss-of-function mutations in F3. These data suggest that (i) F3 is important for maintaining the structural integrity of the cornea, (ii) absence of F3 does not affect the structure or function of any other ocular tissue in which it is expressed, and (iii) targeted silencing of F3 in the retina and/or RPE will likely be well-tolerated, serving as a safe therapeutic strategy for reducing sub-RPE deposit formation in disease. KEY MESSAGES: • Fibulins are expressed throughout the body at varying levels. • Fibulin-3 has a tissue-specific pattern of expression within the eye. • Lack of fibulin-3 leads to structural deformities in the cornea. • The retina and RPE remain structurally and functionally healthy in the absence of fibulin-3 in both mice and humans.
Collapse
|
49
|
Livingstone I, Uversky VN, Furniss D, Wiberg A. The Pathophysiological Significance of Fibulin-3. Biomolecules 2020; 10:E1294. [PMID: 32911658 PMCID: PMC7563619 DOI: 10.3390/biom10091294] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023] Open
Abstract
Fibulin-3 (also known as EGF-containing fibulin extracellular matrix protein 1 (EFEMP1)) is a secreted extracellular matrix glycoprotein, encoded by the EFEMP1 gene that belongs to the eight-membered fibulin protein family. It has emerged as a functionally unique member of this family, with a diverse array of pathophysiological associations predominantly centered on its role as a modulator of extracellular matrix (ECM) biology. Fibulin-3 is widely expressed in the human body, especially in elastic-fibre-rich tissues and ocular structures, and interacts with enzymatic ECM regulators, including tissue inhibitor of metalloproteinase-3 (TIMP-3). A point mutation in EFEMP1 causes an inherited early-onset form of macular degeneration called Malattia Leventinese/Doyne honeycomb retinal dystrophy (ML/DHRD). EFEMP1 genetic variants have also been associated in genome-wide association studies with numerous complex inherited phenotypes, both physiological (namely, developmental anthropometric traits) and pathological (many of which involve abnormalities of connective tissue function). Furthermore, EFEMP1 expression changes are implicated in the progression of numerous types of cancer, an area in which fibulin-3 has putative significance as a therapeutic target. Here we discuss the potential mechanistic roles of fibulin-3 in these pathologies and highlight how it may contribute to the development, structural integrity, and emergent functionality of the ECM and connective tissues across a range of anatomical locations. Its myriad of aetiological roles positions fibulin-3 as a molecule of interest across numerous research fields and may inform our future understanding and therapeutic approach to many human diseases in clinical settings.
Collapse
Affiliation(s)
- Imogen Livingstone
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford OX3 7LD, UK; (I.L.); (D.F.)
| | - Vladimir N. Uversky
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino 142290, Moscow Region, Russia;
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Dominic Furniss
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford OX3 7LD, UK; (I.L.); (D.F.)
- Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Akira Wiberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford OX3 7LD, UK; (I.L.); (D.F.)
- Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
50
|
Johnson AA, Shokhirev MN, Wyss-Coray T, Lehallier B. Systematic review and analysis of human proteomics aging studies unveils a novel proteomic aging clock and identifies key processes that change with age. Ageing Res Rev 2020; 60:101070. [PMID: 32311500 DOI: 10.1016/j.arr.2020.101070] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
The development of clinical interventions that significantly improve human healthspan requires robust markers of biological age as well as thoughtful therapeutic targets. To promote these goals, we performed a systematic review and analysis of human aging and proteomics studies. The systematic review includes 36 different proteomics analyses, each of which identified proteins that significantly changed with age. We discovered 1,128 proteins that had been reported by at least two or more analyses and 32 proteins that had been reported by five or more analyses. Each of these 32 proteins has known connections relevant to aging and age-related disease. GDF15, for example, extends both lifespan and healthspan when overexpressed in mice and is additionally required for the anti-diabetic drug metformin to exert beneficial effects on body weight and energy balance. Bioinformatic enrichment analyses of our 1,128 commonly identified proteins heavily implicated processes relevant to inflammation, the extracellular matrix, and gene regulation. We additionally propose a novel proteomic aging clock comprised of proteins that were reported to change with age in plasma in three or more different studies. Using a large patient cohort comprised of 3,301 subjects (aged 18-76 years), we demonstrate that this clock is able to accurately predict human age.
Collapse
|