1
|
Tiberio F, Polito L, Salvati M, Di Pietro L, Massimi L, Parolini O, Tamburrini G, Lattanzi W. Current Understanding of Crouzon Syndrome Pathophysiology and New Therapeutic Approaches. J Craniofac Surg 2025:00001665-990000000-02627. [PMID: 40227035 DOI: 10.1097/scs.0000000000011376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 04/15/2025] Open
Abstract
Crouzon syndrome (CS) is a rare genetic disorder characterized by the premature fusion of cranial sutures, leading to craniofacial abnormalities and potential neurological complications. CS is caused primarily by gain-of-function mutations in the FGFR2 gene and, less commonly, by mutations in the FGFR3 gene (specifically associated with CS with acanthosis nigricans). Managing CS requires a multidisciplinary approach, combining early and later surgical interventions to prevent intracranial hypertension and correct craniofacial deformities, along with ongoing care to address associated complications. Recent advancements in CS classification on the basis of cranial suture involvement have refined phenotype-genotype correlations, improving personalized therapeutic strategies. This review aims to provide a comprehensive and updated overview of CS, including detailed insights into molecular genetics and biological mechanisms underlying its pathophysiology, and a depiction of the clinical features, diagnosis, and surgical aspects of CS. In addition, we delve into innovative theranostic views, where molecular genetic testing allows the design of personalized noninvasive therapeutic approaches based on innovative biotechnologies, including RNA-interference molecules, pharmacological modulation of FGFR signaling pathways, and recombinant proteins. These advancements underscore the importance of integrating molecular studies into diagnostic and therapeutic protocols to increase the precision and effectiveness of nonsurgical treatments for CS.
Collapse
Affiliation(s)
- Federica Tiberio
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Polito
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Martina Salvati
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorena Di Pietro
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Massimi
- Unità Operativa Complessa di Neurochirurgia Infantile, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ornella Parolini
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gianpiero Tamburrini
- Unità Operativa Complessa di Neurochirurgia Infantile, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Wanda Lattanzi
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
2
|
Meyfarth SRS, Baratto-Filho F, Locks MEN, Proff P, Zandoná GO, Fernandes TDO, de França PHC, Kirschneck C, Antunes LS, Küchler EC. Fibroblast growth factor receptor 2 (FGFR2) genetic polymorphisms contribute to fused roots in human molars. PLoS One 2025; 20:e0316904. [PMID: 40208883 PMCID: PMC11984737 DOI: 10.1371/journal.pone.0316904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 12/18/2024] [Indexed: 04/12/2025] Open
Abstract
Fibroblast growth factors (FGFRs) signaling are required for human tooth development. Its dysregulation affects tooth formation and patients with FGFR2 mutations often present dental anomalies in the spectrum of the syndrome. This study aimed to investigate whether genetic polymorphisms in FGFR2 are associated with molar fused roots. The null hypothesis is that genetic variations in FGFR2 are not associated with isolated cases (non-syndromic) of molars fused roots. Panoramic radiographs of non-syndromic patients were used to assess the occurrence of fused roots in molars. Genomic DNA analysis was performed to investigate polymorphisms within the candidate gene. The association between fused roots and genetic polymorphisms was analyzed using allelic and genotypic distributions, and haplotype frequencies. Odds ratio and 95% confidence interval were calculated to assess the chance of presenting fused roots. The significance level was set at p < 0.05 for all the analysis. A total of 170 patients were included. Statistically significant differences in genotype distribution were observed in rs10736303 and rs2162540. Individuals carrying at least one G allele of rs10736303 had an increased chance to present fused roots. A total of 154 haplotype combinations demonstrated statistically significant associations. The polymorphisms rs10736303 and rs2162540 in FGFR2 were associated with fused roots in human molars.
Collapse
Affiliation(s)
| | - Flares Baratto-Filho
- Department of Dentistry, University of Joinville Region (Univille), Joinville, South Carolina, Brazil
- School of Dentistry, Tuiuti University from Paraná, Curitiba, PR, Brazil
| | - Maria Eduarda Nunis Locks
- Department of Dentistry, University of Joinville Region (Univille), Joinville, South Carolina, Brazil
| | - Peter Proff
- Department of Orthodontics, University of Regensburg, Regensburg, Germany
| | | | - Thaís de Oliveira Fernandes
- Postgraduation Program, Fluminense Federal University, Nova Friburgo Health Institute, Nova Friburgo, RJ, Brazil
| | | | | | | | | |
Collapse
|
3
|
Grochot R, Joshi K, Cammarota A, Woodford R, Sathanantham G, Williams A, Arkenau T, Subbiah V, Swanton C, Fontana E. Safety and Activity of Fibroblast Growth Factor Receptor Inhibitors in Advanced Malignancies: A Pooled Analysis of Early-Phase Clinical Trials. JCO Precis Oncol 2025; 9:e2400896. [PMID: 40239140 DOI: 10.1200/po-24-00896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/28/2025] [Accepted: 02/19/2025] [Indexed: 04/18/2025] Open
Abstract
PURPOSE Aberrant signaling through the fibroblast growth factor receptor (FGFR) due to activating somatic alterations has been associated with multiple malignancies. FGFR inhibitors (FGFRi) with distinct profiles recently entered standard of care. This work summarizes the experience of a dedicated clinical trial unit with FGFRi developed in the last decade within the context of clinical trials. METHODS Demographic and clinical data were collected for patients enrolled in FGFR-targeting phase I to II trials conducted at Sarah Cannon Research Institute, United Kingdom between January 2012 and August 2023. RESULTS Fifty-four patients across seven trials were identified: 50% male; median age 55 years. An FGFR alteration was present in 81% of cases; rearrangements, amplifications, and mutations were present in 59%, 43%, and 9.1% of the cases, respectively, with coexisting alterations in 27%. The most frequent primary tumors were cholangiocarcinomas (31%), urothelial (15%), and colorectal (15%); 85% of the patients were FGFRi-naïve. The most common adverse events (AEs) were hyperphosphatemia (42%), dry mouth (35%), fatigue (24%), mucositis (24%), nail changes (22%), and palmar-plantar erythrodysesthesia (20%), with significant differences between pan-FGFRi and FGFR-2i. The rate of G3 AEs was 22%; no G4-5 events were observed. The median time on treatment was 3.5 months (0.2-72.8). Higher disease control rate was observed in the presence of any FGFR alteration, compared with all-comers (odds ratio [OR], 7; P = .0226). The objective response rate was 38%, 25%, and 25% in patients with gene rearrangements, amplification, and mutations, respectively. The median duration of response was 2.3 months (1.6-7.7). After a median follow-up time of 20 months (95% CI, 12.9 to 71.8), median progression-free survival (mPFS) was 3.2 months (95% CI, 1.9 to 4.6) and median overall survival was 13 months (95% CI, 6.4 to 19.6). PFS was significantly different by response, FGFR status, and tumor type. Patients who experienced a G2-3 AE were more likely to achieve a response (OR, 5.24; P = .0256). CONCLUSION FGFRi are effective treatment strategies for patients with advanced solid tumors harboring FGFR alterations, with manageable toxicities in most patients.
Collapse
Affiliation(s)
- Rafael Grochot
- Sarah Cannon Research Institute (SCRI), London, United Kingdom
| | - Kroopa Joshi
- Sarah Cannon Research Institute (SCRI), London, United Kingdom
| | - Antonella Cammarota
- Sarah Cannon Research Institute (SCRI), London, United Kingdom
- Humanitas University, Milan, Italy
| | - Rachel Woodford
- Sarah Cannon Research Institute (SCRI), London, United Kingdom
| | | | - Anja Williams
- Sarah Cannon Research Institute (SCRI), London, United Kingdom
| | | | | | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Elisa Fontana
- Sarah Cannon Research Institute (SCRI), London, United Kingdom
| |
Collapse
|
4
|
Chen P, Li B, Lu Z, Xu Q, Zheng H, Jiang S, Jiang L, Zheng X. PCBP2 as an intrinsic agi ng factor regulates the senescence of hBMSCs through the ROS-FGF2 signaling axis. eLife 2025; 13:RP92419. [PMID: 40053388 PMCID: PMC11888601 DOI: 10.7554/elife.92419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
Background It has been reported that loss of PCBP2 led to increased reactive oxygen species (ROS) production and accelerated cell aging. Knockdown of PCBP2 in HCT116 cells leads to significant downregulation of fibroblast growth factor 2 (FGF2). Here, we tried to elucidate the intrinsic factors and potential mechanisms of bone marrow mesenchymal stromal cells (BMSCs) aging from the interactions among PCBP2, ROS, and FGF2. Methods Unlabeled quantitative proteomics were performed to show differentially expressed proteins in the replicative senescent human bone marrow mesenchymal stromal cells (RS-hBMSCs). ROS and FGF2 were detected in the loss-and-gain cell function experiments of PCBP2. The functional recovery experiments were performed to verify whether PCBP2 regulates cell function through ROS/FGF2-dependent ways. Results PCBP2 expression was significantly lower in P10-hBMSCs. Knocking down the expression of PCBP2 inhibited the proliferation while accentuated the apoptosis and cell arrest of RS-hBMSCs. PCBP2 silence could increase the production of ROS. On the contrary, overexpression of PCBP2 increased the viability of both P3-hBMSCs and P10-hBMSCs significantly. Meanwhile, overexpression of PCBP2 led to significantly reduced expression of FGF2. Overexpression of FGF2 significantly offset the effect of PCBP2 overexpression in P10-hBMSCs, leading to decreased cell proliferation, increased apoptosis, and reduced G0/G1 phase ratio of the cells. Conclusions This study initially elucidates that PCBP2 as an intrinsic aging factor regulates the replicative senescence of hBMSCs through the ROS-FGF2 signaling axis. Funding This study was supported by the National Natural Science Foundation of China (82172474).
Collapse
Affiliation(s)
- Pengbo Chen
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Bo Li
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Zeyu Lu
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Qingyin Xu
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Huoliang Zheng
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Shengdan Jiang
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Leisheng Jiang
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Xinfeng Zheng
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| |
Collapse
|
5
|
Sisakht MM, Gholizadeh F, Shahravi Z, Doust-Vaghe YK, Nilforoushzadeh MA, Amirkhani MA. Sodium Alginate/Poly (Acrylicacid) Hydrogel Composite, Potential Carrier for Fibroblast Growth Factor1 (FGF1) Delivery. Chem Biodivers 2025; 22:e202401738. [PMID: 39340197 DOI: 10.1002/cbdv.202401738] [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: 07/17/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 09/30/2024]
Abstract
Fibroblast growth factor1 is a powerful signaling molecule that plays a critical role in injury repair of diverse tissue by stimulating cell growth and angiogenesis. FGF1 has significant role in the cell fate and regulating inflammation with short half-life and poor in vivo stability. The encapsulation of the growth factor in the hydrogel led to peptide protect from the degradation and/or immune recognition and enable controlled drug delivery over a longer period of time. The aim of this study is to develop and evaluate a hydrogel carrier with adjustable release rate while maintaining bioactivity of FGF1. Here we describe an optimal ratio of sodium alginate and polyacrylic acid without additional cross linker containing optimum amount of FGF1 with the potential of sustained release to be used as a therapeutic agent. The carrier was characterized by FTIR, contact angle and swelling ratio. The activity of FGF1 after release from the hydrogel was confirmed by ELISA and Western blot. Further assessment of genes related to inflammation were evaluated by RTPCR. This hydrogel is able to deliver growth factors by restricting the essential proteins within the matrix to prevent rapid proteolysis and explosive release and is therefore widely applicable.
Collapse
Affiliation(s)
- Mahsa Mollapour Sisakht
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Stem Cell and Regenerative Medicine Innovation Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Gholizadeh
- Stem Cell and Regenerative Medicine Innovation Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Shahravi
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Kiani Doust-Vaghe
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Nilforoushzadeh
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Skin Repair Research Center, Sarvsan-e Pars Health Development company, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
6
|
Lee J, Park JC, Kim H, Bae HS, Lee DS. Nuclear factor I-C regulates intramembranous bone formation via control of FGF signalling. Heliyon 2025; 11:e41789. [PMID: 39882457 PMCID: PMC11774937 DOI: 10.1016/j.heliyon.2025.e41789] [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: 10/18/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Our previous studies indicate that NFI-C is essential for tooth root development and endochondral ossification. However, its exact role in calvarial intramembranous bone formation remains unclear. In this study, we demonstrate that the disruption of the Nfic gene leads to defects in intramembranous bone formation, characterized by decreased osteogenic proliferative activity and reduced osteoblast differentiation during postnatal osteogenesis. Additionally, Nfic-deficient mice exhibited incomplete suture closure, although Nfic disruption did not affect prenatal calvarial bone development. We found that the expression levels of Fgfr1 and Fgfr2 were reduced in the primary calvarial mesenchymal cells of Nfic-deficient mice. In contrast, NFI-C overexpression in human bone marrow stromal cells (hBMSCs) significantly increased the expression of these factors. Furthermore, NFI-C regulates FGFR1 expression by directly binding to its promoter. These results indicate that NFI-C is crucial in regulating calvarial bone formation and suture closure by controlling Fgfr1 expression and cellular proliferation.
Collapse
Affiliation(s)
- Jieun Lee
- Department of Oral Hygiene, Namseoul University, Cheonan, Republic of Korea
| | - Joo-Cheol Park
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
- Regenerative Dental Medicine R and D Center, Hysensbio Co., Ltd., Seoul, Republic of Korea
| | - Heung‐Joong Kim
- Department of Anatomy and Orofacial Development School of Dentistry Chosun University, Dong-gu, Gwangju, Republic of Korea
| | - Hyun Sook Bae
- Department of Oral Hygiene, Namseoul University, Cheonan, Republic of Korea
| | - Dong-Seol Lee
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
- Regenerative Dental Medicine R and D Center, Hysensbio Co., Ltd., Seoul, Republic of Korea
| |
Collapse
|
7
|
Morice A, de La Seiglière A, Kany A, Khonsari RH, Bensidhoum M, Puig-Lombardi ME, Legeai Mallet L. FGFR antagonists restore defective mandibular bone repair in a mouse model of osteochondrodysplasia. Bone Res 2025; 13:12. [PMID: 39837840 PMCID: PMC11751307 DOI: 10.1038/s41413-024-00385-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 01/30/2025] Open
Abstract
Gain-of-function mutations in fibroblast growth factor receptor (FGFR) genes lead to chondrodysplasia and craniosynostoses. FGFR signaling has a key role in the formation and repair of the craniofacial skeleton. Here, we analyzed the impact of Fgfr2- and Fgfr3-activating mutations on mandibular bone formation and endochondral bone repair after non-stabilized mandibular fractures in mouse models of Crouzon syndrome (Crz) and hypochondroplasia (Hch). Bone mineralization of the calluses was abnormally high in Crz mice and abnormally low in Hch mice. The latter model presented pseudarthrosis and impaired chondrocyte differentiation. Spatial transcriptomic analyses of the Hch callus revealed abnormally low expression of Col11, Col1a, Dmp1 genes in mature chondrocytes. We found that the expression of genes involved in autophagy and apoptosis (Smad1, Comp, Birc2) was significantly perturbed and that the Dusp3, Dusp9, and Socs3 genes controlling the mitogen-activated protein kinase pathway were overexpressed. Lastly, we found that treatment with a tyrosine kinase inhibitor (BGJ398, infigratinib) or a C-type natriuretic peptide (BMN111, vosoritide) fully rescued the defective endochondral bone repair observed in Hch mice. Taken as a whole, our findings show that FGFR3 is a critical orchestrator of bone repair and provide a rationale for the development of potential treatments for patients with FGFR3-osteochondrodysplasia.
Collapse
Affiliation(s)
- Anne Morice
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Paris, France
| | - Amélie de La Seiglière
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Paris, France
| | - Alexia Kany
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Paris, France
| | - Roman H Khonsari
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Paris, France
| | | | - Maria-Emilia Puig-Lombardi
- Bioinformatics Core Platform, Imagine Institute, INSERM UMR1163 and Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR3633, Université Paris Cité, Paris, France
| | - Laurence Legeai Mallet
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Paris, France.
| |
Collapse
|
8
|
Sánchez-Serna G, Badia-Ramentol J, Bujosa P, Ferrández-Roldán A, Torres-Águila NP, Fabregà-Torrus M, Wibisana JN, Mansfield MJ, Plessy C, Luscombe NM, Albalat R, Cañestro C. Less, but More: New Insights From Appendicularians on Chordate Fgf Evolution and the Divergence of Tunicate Lifestyles. Mol Biol Evol 2025; 42:msae260. [PMID: 39686543 PMCID: PMC11733497 DOI: 10.1093/molbev/msae260] [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: 09/02/2024] [Revised: 11/17/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The impact of gene loss on the diversification of taxa and the emergence of evolutionary innovations remains poorly understood. Here, our investigation on the evolution of the Fibroblast Growth Factors (FGFs) in appendicularian tunicates as a case study reveals a scenario of "less, but more" characterized by massive losses of all Fgf gene subfamilies, except for the Fgf9/16/20 and Fgf11/12/13/14, which in turn underwent two bursts of duplications. Through phylogenetic analysis, synteny conservation, and gene and protein structure, we reconstruct the history of appendicularian Fgf genes, highlighting their paracrine and intracellular functions. An exhaustive analysis of developmental Fgf expression in Oikopleura dioica allows us to identify four associated evolutionary patterns characterizing the "less, but more" conceptual framework: conservation of ancestral functions; function shuffling between paralogs linked to gene losses; innovation of new functions after the duplication bursts; and function extinctions linked to gene losses. Our findings allow us to formulate novel hypotheses about the impact of Fgf losses and duplications on the transition from an ancestral ascidian-like biphasic lifestyle to the fully free-living appendicularians. These hypotheses include massive co-options of Fgfs for the development of the oikoblast and the tail fin; recruitment of Fgf11/12/13/14s into the evolution of a new mouth, and their role modulating neuronal excitability; the evolutionary innovation of an anterior tail FGF signaling source upon the loss of retinoic acid signaling; and the potential link between the loss of Fgf7/10/22 and Fgf8/17/18 and the loss of drastic metamorphosis and tail absorption in appendicularians, in contrast to ascidians.
Collapse
Affiliation(s)
- Gaspar Sánchez-Serna
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona 08028, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona (UB), Barcelona, Spain
| | - Jordi Badia-Ramentol
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona 08028, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona (UB), Barcelona, Spain
| | - Paula Bujosa
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona 08028, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona (UB), Barcelona, Spain
| | - Alfonso Ferrández-Roldán
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona 08028, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona (UB), Barcelona, Spain
| | - Nuria P Torres-Águila
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona 08028, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona (UB), Barcelona, Spain
| | - Marc Fabregà-Torrus
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona 08028, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona (UB), Barcelona, Spain
| | - Johannes N Wibisana
- Genomics and Regulatory Systems Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Michael J Mansfield
- Genomics and Regulatory Systems Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Charles Plessy
- Genomics and Regulatory Systems Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Nicholas M Luscombe
- Genomics and Regulatory Systems Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Ricard Albalat
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona 08028, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona (UB), Barcelona, Spain
| | - Cristian Cañestro
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona 08028, Spain
- Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona (UB), Barcelona, Spain
| |
Collapse
|
9
|
Lee ZM, Goh BH, Khaw KY. Aloe vera and the Proliferative Phase of Cutaneous Wound Healing: Status Quo Report on Active Principles, Mechanisms, and Applications. PLANTA MEDICA 2025; 91:4-18. [PMID: 39566518 DOI: 10.1055/a-2446-2146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Aloe vera is commonly used as traditional medicine for cutaneous wound healing. Nonetheless, the wound healing mechanisms of Aloe vera remain unclear. This review aims to provide insight into the molecular mechanisms of Aloe vera in promoting cutaneous wound healing, with particular emphasis on the mechanisms that stimulate cell proliferation and migration. Aloe vera has been shown to upregulate growth factors such as keratinocyte growth factor-1 (KGF-1), transforming growth factor-β (TGF-β), cyclin D1, insulin-like growth factor 1 (IGF-1), vascular endothelial growth factor (VEGF), basic fibroblastic growth factor (bFGF), and microfibril-associated glycoprotein 4 (MFAP4), as well as collagen, fibrillin, elastin, α-smooth muscle actin (α-SMA), integrins, and platelet endothelial cell adhesion molecule 1 (PECAM-1, also known as CD31), while downregulating the expression of matrix metalloproteinases (MMPs). In addition, Aloe vera was also found to upregulate PI3K/Akt and MAPK pathways, as well as the TGF-β signalling pathway via Smad proteins. Furthermore, molecular docking studies revealed that certain chemical constituents of Aloe vera bind to some of the forementioned growth factors or signalling molecules. With regards to current applications, although human clinical trials have reported positive results from using Aloe vera in healing open wounds and burns and alleviating some inflammatory skin diseases, the current commercial uses of Aloe vera remain largely focused on cosmetic products. Thus, greater advances are required to promote the use of Aloe vera products in clinical settings.
Collapse
Affiliation(s)
- Zhuang Min Lee
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Subang Jaya, Malaysia
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Subang Jaya, Malaysia
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
| | - Kooi Yeong Khaw
- Biopharmaceutical Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| |
Collapse
|
10
|
Han S, Chen Q, Zhu Q, Han W. Circulating inflammatory cytokines and the risk of cerebral small vessel disease: a bidirectional Mendelian randomization analysis. J Stroke Cerebrovasc Dis 2025; 34:108163. [PMID: 39637729 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108163] [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: 06/15/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND A correlation between inflammation and cerebral small vessel disease (CSVD) has been hypothesized by earlier observational research, while this correlation has not been well established. Considering the significant clinical value of this causality determination, Mendelian randomization (MR) was implemented to investigate the causality between inflammatory cytokines and CSVD radiological lesions. METHODS Using the publicly available Genome-Wide Association Study (GWAS) datasets, a bidirectional two-sample MR analysis was employed to infer causality between 91 inflammatory cytokines and CSVD phenotypes [white matter hyperintensity (WHM), fractional anisotropy (FA), mean diffusivity (MD), cerebral microbleeds (CMBs), and lacunar stroke]. A set of methods was used for sensitivity analysis, including Cochran's Q test, MR-Egger intercept method, and MR pleiotropy residual sum and outlier (MR-PRESSO) global test. Furthermore, the strength of causality was assessed using the Bonferroni correction. RESULTS Our research discovered a mutually predictive bidirectional link between CSVD phenotypes and inflammatory cytokines. Following the application of the Bonferroni correction, fibroblast growth factor 21 (FGF-21) was significantly inversely correlated with an increased risk of CMBs (OR = 0.579, 95 % CI = 0.425-0.789, P = 0.00055). Using sensitivity analysis, heterogeneity, and horizontal pleiotropy were not detected. CONCLUSION In this investigation, we established the causality between CSVD and inflammatory cytokines, with FGF-21 in particular significantly reducing the risk of CMBs. With further validation, these findings may provide new targets for the prevention, detection, and intervention of CSVD.
Collapse
Affiliation(s)
- Shasha Han
- Department of Neurology, Jiaozhou Central Hosptital of Qingdao, Qingdao 266300, Shandong, China.
| | - Qiong Chen
- Department of Neurology, Jiaozhou Central Hosptital of Qingdao, Qingdao 266300, Shandong, China.
| | - Qiang Zhu
- Department of Emergency, Jiaozhou Central Hosptital of Qingdao, Qingdao 266300, Shandong, China.
| | - Wenxiu Han
- Translational Pharmaceutical Laboratory, Jining NO.1 People's Hospital, Shandong First Medical University, Jining 272000, Shandong, China; Institute of Translational Pharmacy, Jining Medical Research Academy, Jining 272000, Shandong, China.
| |
Collapse
|
11
|
Rodriguez Torres CS, Wicker NB, Puccini de Castro V, Stefinko M, Bennett DC, Bernhardt B, Garcia Montes de Oca M, Jallow S, Flitcroft K, Palalay JJS, Payán Parra OA, Stern YE, Koelle MR, Voisine C, Woods IG, Lo TW, Stern MJ, de la Cova CC. The Caenorhabditis elegans protein SOC-3 permits an alternative mode of signal transduction by the EGL-15 FGF receptor. Dev Biol 2024; 516:183-195. [PMID: 39173814 PMCID: PMC11488645 DOI: 10.1016/j.ydbio.2024.08.014] [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: 05/14/2024] [Revised: 08/02/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Fibroblast Growth Factors and their receptors (FGFRs) comprise a cell signaling module that can stimulate signaling by Ras and the kinases Raf, MEK, and ERK to regulate animal development and homeostatic functions. In Caenorhabditis elegans, the sole FGFR ortholog EGL-15 acts with the GRB2 ortholog SEM-5 to promote chemoattraction and migration by the sex myoblasts (SMs) and fluid homeostasis by the hypodermis (Hyp7). Cell-specific differences in EGL-15 signaling were suggested by the phenotypes caused by egl-15(n1457), an allele that removes a region of its C-terminal domain (CTD) known to bind SEM-5. To determine how mutations altered EGL-15 activity in the SMs and Hyp7, we used the kinase reporter ERK-KTR to measure activation of the ERK ortholog MPK-1. Consequences of egl-15(n1457) were cell-specific, resulting in loss of MPK-1 activity in the SMs and elevated activity in Hyp7. Previous studies of Hyp7 showed that loss of the CLR-1 phosphatase causes a fluid homeostasis defect termed "Clear" that is suppressed by reduction of EGL-15 signaling, a phenotype termed "Suppressor of Clear" (Soc). To identify mechanisms that permit EGL-15 signaling in Hyp7, we conducted a genetic screen for Soc mutants in the clr-1; egl-15(n1457) genotype. We report the identification of SOC-3, a protein with putative SEM-5-binding motifs and PH and PTB domains similar to DOK and IRS proteins. In combination with the egl-15(n1457) mutation, loss of either soc-3, the GAB1 ortholog soc-1, or the SHP2 ortholog ptp-2, reduced MPK-1 activation. We generated alleles of soc-3 to test the requirement for the SEM-5-binding motifs, finding that residue Tyr356 is required for function. We propose that EGL-15-mediated SM chemoattraction relies solely on the direct interaction between SEM-5 and the EGL-15 CTD. In Hyp7, EGL-15 signaling uses two mechanisms: the direct SEM-5 binding mechanism; and an alternative, CTD-independent mechanism involving SOC-3, SOC-1, and PTP-2. This work demonstrates that FGF signaling uses distinct, tissue-specific mechanisms in development, and identifies SOC-3 as a potential adaptor that facilitates Ras pathway activation by FGFR.
Collapse
Affiliation(s)
| | - Nicole B Wicker
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | | | - Mariya Stefinko
- Department of Biology, Northeastern Illinois University, Chicago, IL, 60625, USA
| | | | | | | | - Sainabou Jallow
- Department of Biology, Ithaca College, Ithaca, NY, 14850, USA
| | - Katelyn Flitcroft
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | | | - Omar A Payán Parra
- Department of Biology, Northeastern Illinois University, Chicago, IL, 60625, USA
| | - Yaakov E Stern
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | | | - Cindy Voisine
- Department of Biology, Northeastern Illinois University, Chicago, IL, 60625, USA
| | - Ian G Woods
- Department of Biology, Ithaca College, Ithaca, NY, 14850, USA
| | - Te-Wen Lo
- Department of Biology, Ithaca College, Ithaca, NY, 14850, USA
| | - Michael J Stern
- Department of Biology, Northeastern Illinois University, Chicago, IL, 60625, USA
| | - Claire C de la Cova
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA.
| |
Collapse
|
12
|
Siripoon T, O'Donnell C, Jin Z, Mahipal A. Fibroblast growth factor therapies in biliary tract cancers: current and future state. Expert Opin Investig Drugs 2024; 33:1245-1255. [PMID: 39629832 DOI: 10.1080/13543784.2024.2430201] [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: 10/02/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024]
Abstract
INTRODUCTION Cholangiocarcinoma is the rare and aggressive tumor with poor prognosis and limited therapeutic options. Recently, there have been promising developments in molecular targeted therapies for patients following the progression of first-line chemotherapy and immunotherapy combinations. Dysregulation of fibroblast Growth Factor Receptor (FGFR) signaling is significantly associated with tumorigenesis of intrahepatic cholangiocarcinoma and has been identified as a targetable alteration. This was possible through the discovery of crucial insights into the biochemical mechanisms and pathophysiology of the FGFR pathway. AREAS COVERED This review summarizes the current state of FGFR targeted therapies, mechanisms of resistance, and future directions for FGFR-targeted therapies in patients with cholangiocarcinoma. EXPERT OPINION Currently, pemigatinib and futibatinib are FDA approved FGFR-targeted therapies that have demonstrated remarkable responses. However, there is still a significant proportion of patients whose disease remains intrinsically resistant to treatment and most patients eventually develop secondary resistance after an initial response. Additionally, unique side effects of FGFR inhibitors may limit their efficacy in clinical practice and can have detrimental effects on quality of life. Several novel FGFR inhibitors are currently being investigated to overcome resistance mechanisms and reduce toxicities.
Collapse
Affiliation(s)
| | | | - Zhaohui Jin
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Amit Mahipal
- Department of Oncology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
13
|
Rzhanova LA, Alpeeva EV, Aleksandrova MA. Using Small Molecules to Reprogram RPE Cells in Regenerative Medicine for Degenerative Eye Disease. Cells 2024; 13:1931. [PMID: 39682681 PMCID: PMC11640686 DOI: 10.3390/cells13231931] [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: 09/26/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
The main purpose of regenerative medicine for degenerative eye diseases is to create cells to replace lost or damaged ones. Due to their anatomical, genetic, and epigenetic features, characteristics of origin, evolutionary inheritance, capacity for dedifferentiation, proliferation, and plasticity, mammalian and human RPE cells are of great interest as endogenous sources of new photoreceptors and other neurons for the degrading retina. Promising methods for the reprogramming of RPE cells into retinal cells include genetic methods and chemical methods under the influence of certain low-molecular-weight compounds, so-called small molecules. Depending on the goal, which can be the preservation or the replacement of lost RPE cells and cellular structures, various small molecules are used to influence certain biological processes at different levels of cellular regulation. This review discusses the potential of the chemical reprogramming of RPE cells in comparison with other somatic cells and induced pluripotent stem cells (iPSCs) into neural cells of the brain and retina. Possible mechanisms of the chemically induced reprogramming of somatic cells under the influence of small molecules are explored and compared. This review also considers other possibilities in using them in the treatment of retinal degenerative diseases based on the protection, preservation, and support of survived RPE and retinal cells.
Collapse
Affiliation(s)
- Lyubov A. Rzhanova
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia;
| | - Elena V. Alpeeva
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia;
| | | |
Collapse
|
14
|
Zhang S, Wang H, Meng Y, Li M, Li Y, Ye X, Duan S, Xiao S, Lu H, Zhong K. Ethyl butyrate inhibits caudal fin regeneration in adult zebrafish by disrupting extracellular matrix remodeling. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 276:107111. [PMID: 39366190 DOI: 10.1016/j.aquatox.2024.107111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/14/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
Wound healing and tissue regeneration are influenced by a variety of factors. Adverse lifestyle habits, such as excessive alcohol consumption, delay wound healing and increase the risk of secondary infections. Ethyl butyrate is a common food additive widely used to enhance the aroma of alcoholic beverages. This additive is generally considered harmless to human health in both industrial and domestic settings. However, the ecotoxicity and its effects on wound healing have not been elucidated. In this study, we used zebrafish as the experimental animal, and the caudal fins were amputated to explore the effects of ethyl butyrate on wound healing and tissue regeneration. The effect of ethyl butyrate on blastema and bone regeneration and its impact on the transcriptional levels of regeneration-related genes and inflammation-related genes were evaluated. RNA-seq was conducted to determine the differentially expressed genes (DEGs) between the treatment and the control groups. KEGG and GO analysis was conducted to explore the functions of DEGs. Significantly enriched GO terms and KEGG pathways were identified to explore the molecular mechanism underlying the inhibition of zebrafish caudal fin regeneration by ethyl butyrate. The results demonstrated that ethyl butyrate significantly inhibited the regeneration of zebrafish caudal fins, including blastema and bone regeneration. Ethyl butyrate exposure significantly downregulated the expression of genes associated with bone and blastema regeneration and inflammation response. KEGG and GO functional analyses revealed that the DEGs were associated with significant enrichment of extracellular matrix-receptor interactions. Ethyl butyrate treatment downregulated the expression of most extracellular matrix-related genes. These findings indicate that ethyl butyrate potentially modulates pathways associated with the structure, adhesion, modification, and degradation of the extracellular matrix, thereby disrupting extracellular matrix remodeling, inhibiting wound inflammation, impairing blastema and bone regeneration and ultimately hindering caudal fin regeneration. In summary, the findings demonstrate that ethyl butyrate disrupts extracellular matrix remodeling and inhibits the regeneration of zebrafish caudal fins. These results provide valuable insights into the rational use of ethyl butyrate and further investigation of wound healing mechanisms.
Collapse
Affiliation(s)
- Sijie Zhang
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Shiyuan South Rd, Ganzhou, Jiangxi 341000, China
| | - Hao Wang
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Shiyuan South Rd, Ganzhou, Jiangxi 341000, China
| | - Yunlong Meng
- School of Medicine, Tongji University, Shanghai 200000, China
| | - Mijia Li
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Shiyuan South Rd, Ganzhou, Jiangxi 341000, China
| | - Yang Li
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Shiyuan South Rd, Ganzhou, Jiangxi 341000, China
| | - Xinhao Ye
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Shiyuan South Rd, Ganzhou, Jiangxi 341000, China
| | - Shiyi Duan
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Shiyuan South Rd, Ganzhou, Jiangxi 341000, China
| | - Shimei Xiao
- National Center of Quality Testing and Inspection for Tungsten and Rare Earth Products, Ganzhou 341000, China; Jiangxi Institute of Tungsten and Rare Earth, Ganzhou 341000, China
| | - Huiqiang Lu
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Shiyuan South Rd, Ganzhou, Jiangxi 341000, China
| | - Keyuan Zhong
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Shiyuan South Rd, Ganzhou, Jiangxi 341000, China.
| |
Collapse
|
15
|
Khetchoumian K, Sochodolsky K, Lafont C, Gouhier A, Bemmo A, Kherdjemil Y, Kmita M, Le Tissier P, Mollard P, Christian H, Drouin J. Paracrine FGF1 signaling directs pituitary architecture and size. Proc Natl Acad Sci U S A 2024; 121:e2410269121. [PMID: 39320918 PMCID: PMC11459159 DOI: 10.1073/pnas.2410269121] [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: 05/22/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Organ architecture is established during development through intricate cell-cell communication mechanisms, yet the specific signals mediating these communications often remain elusive. Here, we used the anterior pituitary gland that harbors different interdigitated hormone-secreting homotypic cell networks to dissect cell-cell communication mechanisms operating during late development. We show that blocking differentiation of corticotrope cells leads to pituitary hypoplasia with a major effect on somatotrope cells that directly contact corticotropes. Gene knockout of the corticotrope-restricted transcription factor Tpit results in fewer somatotropes, with less secretory granules and a loss of cell polarity, resulting in systemic growth retardation. Single-cell transcriptomic analyses identified FGF1 as a corticotrope-specific Tpit dosage-dependent target gene responsible for these phenotypes. Consistently, genetic ablation of FGF1 in mice phenocopies pituitary hypoplasia and growth impairment observed in Tpit-deficient mice. These findings reveal FGF1 produced by the corticotrope cell network as an essential paracrine signaling molecule participating in pituitary architecture and size.
Collapse
Affiliation(s)
- Konstantin Khetchoumian
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Kevin Sochodolsky
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Chrystel Lafont
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
- BioCampus Montpellier, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
| | - Arthur Gouhier
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Amandine Bemmo
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Yacine Kherdjemil
- Disease Modeling and Genome Editing platform, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Marie Kmita
- Laboratoire de recherche en génétique et développement, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Paul Le Tissier
- Centre for Integrative Physiology, University of Edinburgh, EdinburghEH8 9XD, United Kingdom
| | - Patrice Mollard
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
- BioCampus Montpellier, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
| | - Helen Christian
- Department of Physiology, Anatomy and Genetics, Oxford University, OxfordOX1 3QX, United Kingdom
| | - Jacques Drouin
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| |
Collapse
|
16
|
Zhu X, Wang L, Wang K, Yao Y, Zhou F. Erdafitinib promotes ferroptosis in human uveal melanoma by inducing ferritinophagy and lysosome biogenesis via modulating the FGFR1/mTORC1/TFEB signaling axis. Free Radic Biol Med 2024; 222:552-568. [PMID: 38971541 DOI: 10.1016/j.freeradbiomed.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Uveal melanoma (UM) is a rare yet lethal primary intraocular malignancy affecting adults. Analysis of data from The Cancer Genome Atlas (TCGA) database revealed that FGFR1 expression was increased in UM tumor tissues and was linked to aggressive behavior and a poor prognosis. This study assessed the anti-tumor effects of Erdafitinib, a selective pan-FGFR inhibitor, in both in vitro and in vivo UM models. Erdafitinib exhibited a robust anti-cancer activity in UM through inducing ferroptosis in the FGFR1-dependent manner. Transcriptomic data revealed that Erdafitinib mediated its anti-cancer effects via modulating the ferritinophagy/lysosome biogenesis. Subsequent research revealed that Erdafitinib exerted its effects by reducing the expression of FGFR1 and inhibiting the activity of mTORC1 in UM cells. Concurrently, it enhanced the dephosphorylation, nuclear translocation, and transcriptional activity of TFEB. The aggregation of TFEB in nucleus triggered FTH1-dependent ferritinophagy, leading to lysosomal activation and iron overload. Conversely, the overexpression of FGFR1 served to mitigate the effects of Erdafitinib on ferritinophagy, lysosome biogenesis, and the activation of the mTORC1/TFEB signaling pathway. In vivo experiments have convincingly shown that Erdafitinib markedly curtails tumor growth in an UM xenograft mouse model, an effect that is closely correlated with a decrease in FGFR1 expression levels. The present study is the first to demonstrate that Erdafitinib powerfully induces ferroptosis in UM by orchestrating the ferritinophagy and lysosome biogenesis via modulating the FGFR1/mTORC1/TFEB signaling. Consequently, Erdafitinib emerges as a strong candidate for clinical trial investigation, and FGFR1 emerges as a novel and promising therapeutic target in the treatment of UM.
Collapse
Affiliation(s)
- Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Ling Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China.
| | - Ying Yao
- Department of Pharmacy, Wuxi Maternity and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214002, Jiangsu Province, China.
| | - Fanfan Zhou
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
17
|
Manole CG, Voiculescu VM, Soare C, Ceafalan LC, Gherghiceanu M, Hinescu ME. Skin Telocytes Could Fundament the Cellular Mechanisms of Wound Healing in Platelet-Rich Plasma Administration. Cells 2024; 13:1321. [PMID: 39195210 PMCID: PMC11353115 DOI: 10.3390/cells13161321] [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: 06/12/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
For more than 40 years, autologous platelet concentrates have been used in clinical medicine. Since the first formula used, namely platelet-rich plasma (PRP), other platelet concentrates have been experimented with, including platelet-rich fibrin and concentrated growth factor. Platelet concentrates have three standard characteristics: they act as scaffolds, they serve as a source of growth factors and cytokines, and they contain live cells. PRP has become extensively used in regenerative medicine for the successful treatment of a variety of clinical (non-)dermatological conditions like alopecies, acne scars, skin burns, skin ulcers, muscle, cartilage, and bone repair, and as an adjuvant in post-surgery wound healing, with obvious benefits in terms of functionality and aesthetic recovery of affected tissues/organs. These indications were well documented, and a large amount of evidence has already been published supporting the efficacy of this method. The primordial principle behind minimally invasive PRP treatments is the usage of the patient's own platelets. The benefits of the autologous transplantation of thrombocytes are significant, representing a fast and economic method that requires only basic equipment and training, and it is biocompatible, thus being a low risk for the patient (infection and immunological reactions can be virtually disregarded). Usually, the structural benefits of applying PRP are attributed to fibroblasts only, as they are considered the most numerous cell population within the interstitium. However, this apparent simplistic explanation is still eluding those different types of interstitial cells (distinct from fibroblasts) that are residing within stromal tissue, e.g., telocytes (TCs). Moreover, dermal TCs have an already documented potential in angiogenesis (extra-cutaneous, but also within skin), and their implication in skin recovery in a few dermatological conditions was attested and described ultrastructurally and immunophenotypically. Interestingly, PRP biochemically consists of a series of growth factors, cytokines, and other molecules, to which TCs have also proven to have a positive expression. Thus, it is attractive to hypothesize and to document any tissular collaboration between cutaneous administered PRP and local dermal TCs in skin recovery/repair/regeneration. Therefore, TCs could be perceived as the missing link necessary to provide a solid explanation of the good results achieved by administering PRP in skin-repairing processes.
Collapse
Affiliation(s)
- Catalin G. Manole
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Ultrastructural Pathology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Vlad M. Voiculescu
- Department of Oncological Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Cristina Soare
- Department of Oncological Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Laura Cristina Ceafalan
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Cell Biology, Neurosciences and Experimental Myology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihaela Gherghiceanu
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Ultrastructural Pathology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihail E. Hinescu
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
18
|
Dermitzakis I, Chatzi D, Kyriakoudi SA, Evangelidis N, Vakirlis E, Meditskou S, Theotokis P, Manthou ME. Skin Development and Disease: A Molecular Perspective. Curr Issues Mol Biol 2024; 46:8239-8267. [PMID: 39194704 DOI: 10.3390/cimb46080487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/27/2024] [Accepted: 07/28/2024] [Indexed: 08/29/2024] Open
Abstract
Skin, the largest organ in the human body, is a crucial protective barrier that plays essential roles in thermoregulation, sensation, and immune defence. This complex organ undergoes intricate processes of development. Skin development initiates during the embryonic stage, orchestrated by molecular cues that control epidermal specification, commitment, stratification, terminal differentiation, and appendage growth. Key signalling pathways are integral in coordinating the development of the epidermis, hair follicles, and sweat glands. The complex interplay among these pathways is vital for the appropriate formation and functionality of the skin. Disruptions in multiple molecular pathways can give rise to a spectrum of skin diseases, from congenital skin disorders to cancers. By delving into the molecular mechanisms implicated in developmental processes, as well as in the pathogenesis of diseases, this narrative review aims to present a comprehensive understanding of these aspects. Such knowledge paves the way for developing innovative targeted therapies and personalised treatment approaches for various skin conditions.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Despoina Chatzi
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Stella Aikaterini Kyriakoudi
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Nikolaos Evangelidis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Efstratios Vakirlis
- First Department of Dermatology and Venereology, School of Medicine, Aristotle University of Thessaloniki, 54643 Thessaloniki, Greece
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
19
|
Mustafa M, Abbas K, Alam M, Habib S, Zulfareen, Hasan GM, Islam S, Shamsi A, Hassan I. Investigating underlying molecular mechanisms, signaling pathways, emerging therapeutic approaches in pancreatic cancer. Front Oncol 2024; 14:1427802. [PMID: 39087024 PMCID: PMC11288929 DOI: 10.3389/fonc.2024.1427802] [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: 05/04/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Pancreatic adenocarcinoma, a clinically challenging malignancy constitutes a significant contributor to cancer-related mortality, characterized by an inherently poor prognosis. This review aims to provide a comprehensive understanding of pancreatic adenocarcinoma by examining its multifaceted etiologies, including genetic mutations and environmental factors. The review explains the complex molecular mechanisms underlying its pathogenesis and summarizes current therapeutic strategies, including surgery, chemotherapy, and emerging modalities such as immunotherapy. Critical molecular pathways driving pancreatic cancer development, including KRAS, Notch, and Hedgehog, are discussed. Current therapeutic strategies, including surgery, chemotherapy, and radiation, are discussed, with an emphasis on their limitations, particularly in terms of postoperative relapse. Promising research areas, including liquid biopsies, personalized medicine, and gene editing, are explored, demonstrating the significant potential for enhancing diagnosis and treatment. While immunotherapy presents promising prospects, it faces challenges related to immune evasion mechanisms. Emerging research directions, encompassing liquid biopsies, personalized medicine, CRISPR/Cas9 genome editing, and computational intelligence applications, hold promise for refining diagnostic approaches and therapeutic interventions. By integrating insights from genetic, molecular, and clinical research, innovative strategies that improve patient outcomes can be developed. Ongoing research in these emerging fields holds significant promise for advancing the diagnosis and treatment of this formidable malignancy.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Zulfareen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sidra Islam
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
20
|
Li HZ, Zhang JL, Yuan DL, Xie WQ, Ladel CH, Mobasheri A, Li YS. Role of signaling pathways in age-related orthopedic diseases: focus on the fibroblast growth factor family. Mil Med Res 2024; 11:40. [PMID: 38902808 PMCID: PMC11191355 DOI: 10.1186/s40779-024-00544-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Fibroblast growth factor (FGF) signaling encompasses a multitude of functions, including regulation of cell proliferation, differentiation, morphogenesis, and patterning. FGFs and their receptors (FGFR) are crucial for adult tissue repair processes. Aberrant FGF signal transduction is associated with various pathological conditions such as cartilage damage, bone loss, muscle reduction, and other core pathological changes observed in orthopedic degenerative diseases like osteoarthritis (OA), intervertebral disc degeneration (IVDD), osteoporosis (OP), and sarcopenia. In OA and IVDD pathologies specifically, FGF1, FGF2, FGF8, FGF9, FGF18, FGF21, and FGF23 regulate the synthesis, catabolism, and ossification of cartilage tissue. Additionally, the dysregulation of FGFR expression (FGFR1 and FGFR3) promotes the pathological process of cartilage degradation. In OP and sarcopenia, endocrine-derived FGFs (FGF19, FGF21, and FGF23) modulate bone mineral synthesis and decomposition as well as muscle tissues. FGF2 and other FGFs also exert regulatory roles. A growing body of research has focused on understanding the implications of FGF signaling in orthopedic degeneration. Moreover, an increasing number of potential targets within the FGF signaling have been identified, such as FGF9, FGF18, and FGF23. However, it should be noted that most of these discoveries are still in the experimental stage, and further studies are needed before clinical application can be considered. Presently, this review aims to document the association between the FGF signaling pathway and the development and progression of orthopedic diseases. Besides, current therapeutic strategies targeting the FGF signaling pathway to prevent and treat orthopedic degeneration will be evaluated.
Collapse
Affiliation(s)
- Heng-Zhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing-Lve Zhang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Dong-Liang Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Wen-Qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | | | - Ali Mobasheri
- Faculty of Medicine, Research Unit of Health Sciences and Technology, University of Oulu, 90014, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406, Vilnius, Lithuania.
- Department of Rheumatology and Clinical Immunology, Universitair Medisch Centrum Utrecht, Utrecht, 3508, GA, the Netherlands.
- Department of Joint Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000, Liège, Belgium.
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
21
|
Shen L, Li Y, Zhao H. Fibroblast growth factor signaling in macrophage polarization: impact on health and diseases. Front Immunol 2024; 15:1390453. [PMID: 38962005 PMCID: PMC11219802 DOI: 10.3389/fimmu.2024.1390453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/29/2024] [Indexed: 07/05/2024] Open
Abstract
Fibroblast growth factors (FGFs) are a versatile family of peptide growth factors that are involved in various biological functions, including cell growth and differentiation, embryonic development, angiogenesis, and metabolism. Abnormal FGF/FGF receptor (FGFR) signaling has been implicated in the pathogenesis of multiple diseases such as cancer, metabolic diseases, and inflammatory diseases. It is worth noting that macrophage polarization, which involves distinct functional phenotypes, plays a crucial role in tissue repair, homeostasis maintenance, and immune responses. Recent evidence suggests that FGF/FGFR signaling closely participates in the polarization of macrophages, indicating that they could be potential targets for therapeutic manipulation of diseases associated with dysfunctional macrophages. In this article, we provide an overview of the structure, function, and downstream regulatory pathways of FGFs, as well as crosstalk between FGF signaling and macrophage polarization. Additionally, we summarize the potential application of harnessing FGF signaling to modulate macrophage polarization.
Collapse
Affiliation(s)
- Luyao Shen
- The Second Affiliated Hospital & Yuying Children’s Hospital/The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yongsheng Li
- The Second Affiliated Hospital & Yuying Children’s Hospital/The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Huakan Zhao
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
22
|
Lana JF, Navani A, Jeyaraman M, Santos N, Pires L, Santos GS, Rodrigues IJ, Santos D, Mosaner T, Azzini G, da Fonseca LF, de Macedo AP, Huber SC, de Moraes Ferreira Jorge D, Purita J. Sacral Bioneuromodulation: The Role of Bone Marrow Aspirate in Spinal Cord Injuries. Bioengineering (Basel) 2024; 11:461. [PMID: 38790327 PMCID: PMC11118755 DOI: 10.3390/bioengineering11050461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Spinal cord injury (SCI) represents a severe trauma to the nervous system, leading to significant neurological damage, chronic inflammation, and persistent neuropathic pain. Current treatments, including pharmacotherapy, immobilization, physical therapy, and surgical interventions, often fall short in fully addressing the underlying pathophysiology and resultant disabilities. Emerging research in the field of regenerative medicine has introduced innovative approaches such as autologous orthobiologic therapies, with bone marrow aspirate (BMA) being particularly notable for its regenerative and anti-inflammatory properties. This review focuses on the potential of BMA to modulate inflammatory pathways, enhance tissue regeneration, and restore neurological function disrupted by SCI. We hypothesize that BMA's bioactive components may stimulate reparative processes at the cellular level, particularly when applied at strategic sites like the sacral hiatus to influence lumbar centers and higher neurological structures. By exploring the mechanisms through which BMA influences spinal repair, this review aims to establish a foundation for its application in clinical settings, potentially offering a transformative approach to SCI management that extends beyond symptomatic relief to promoting functional recovery.
Collapse
Affiliation(s)
- José Fábio Lana
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Clinical Research, Anna Vitória Lana Institute (IAVL), Indaiatuba 13334-170, SP, Brazil
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
| | - Annu Navani
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
- Comprehensive Spine & Sports Center, Campbell, CA 95008, USA
| | - Madhan Jeyaraman
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Department of Orthopaedics, ACS Medical College and Hospital, Chennai 600077, Tamil Nadu, India
| | - Napoliane Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Luyddy Pires
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Gabriel Silva Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Izair Jefthé Rodrigues
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Douglas Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Tomas Mosaner
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Gabriel Azzini
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Lucas Furtado da Fonseca
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Medical School, Federal University of São Paulo (UNIFESP), São Paulo 04024-002, SP, Brazil
| | - Alex Pontes de Macedo
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Stephany Cares Huber
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Daniel de Moraes Ferreira Jorge
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Joseph Purita
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
| |
Collapse
|
23
|
Juma SN, Liao J, Huang Y, Vlashi R, Wang Q, Wu B, Wang D, Wu M, Chen G. Osteoarthritis versus psoriasis arthritis: Physiopathology, cellular signaling, and therapeutic strategies. Genes Dis 2024; 11:100986. [PMID: 38292181 PMCID: PMC10825447 DOI: 10.1016/j.gendis.2023.04.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/15/2023] [Indexed: 02/01/2024] Open
Abstract
Osteoarthritis and psoriasis arthritis are two degenerative forms of arthritis that share similar yet also different manifestations at the histological, cellular, and clinical levels. Rheumatologists have marked them as two entirely distinct arthropathies. Given recent discoveries in disease initiation and progression, potential mechanisms, cellular signaling pathways, and ongoing clinical therapeutics, there are now more opportunities for discovering osteoarthritis drugs. This review summarized the osteoarthritis and psoriasis arthritis signaling pathways, crosstalk between BMP, WNT, TGF-β, VEGF, TLR, and FGF signaling pathways, biomarkers, and anatomical pathologies. Through bench research, we demonstrated that regenerative medicine is a promising alternative for treating osteoarthritis by highlighting significant scientific discoveries on entheses, multiple signaling blockers, and novel molecules such as immunoglobulin new antigen receptors targeted for potential drug evaluation. Furthermore, we offered valuable therapeutic approaches with a multidisciplinary strategy to treat patients with osteoarthritis or psoriasis arthritis in the coming future in the clinic.
Collapse
Affiliation(s)
- Salma Nassor Juma
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Junguang Liao
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Yuping Huang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Qingwan Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Bocong Wu
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Dan Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Mengrui Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| |
Collapse
|
24
|
Gurugubelli KR, Ballambattu VB. Perspectives on folate with special reference to epigenetics and neural tube defects. Reprod Toxicol 2024; 125:108576. [PMID: 38479591 DOI: 10.1016/j.reprotox.2024.108576] [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: 12/27/2023] [Revised: 03/07/2024] [Accepted: 03/10/2024] [Indexed: 03/22/2024]
Abstract
Folate is a micronutrient essential for DNA synthesis, cell division, fetal growth and development. Folate deficiency leads to genomic instability. Inadequate intake of folate during conception may lead to neural tube defects (NTDs) in the offspring. Folate influences the DNA methylation, histone methylation and homocysteine mediated gene methylation. DNA methylation influences the expression of microRNAs (miRNAs). Folate deficiency may be associated with miRNAs misregulation leading to NTDs. Mitochondrial epigenetics and folate metabolism has proved to be involved in embryogenesis and neural tube development. Folate related genetic variants also cause the occurrence of NTDs. Unmetabolized excessive folate may affect health adversely. Hence estimation of folate levels in the blood plays an important role in high-risk cases.
Collapse
Affiliation(s)
- Krishna Rao Gurugubelli
- Department of Biochemistry, Andhra Medical College (AMC), Visakhapatnam, Andhra Pradesh, India
| | - Vishnu Bhat Ballambattu
- Aarupadai Veedu Medical College & Hospital (AVMC & H), Vinayaka Mission's Research Foundation (DU), Kirumambakkam, Puducherry, India.
| |
Collapse
|
25
|
Kespohl B, Hegele AL, Düsterhöft S, Bakker H, Buettner FFR, Hartig R, Lokau J, Garbers C. Molecular characterization of the craniosynostosis-associated interleukin-11 receptor variants p.T306_S308dup and p.E364_V368del. FEBS J 2024; 291:1667-1683. [PMID: 37994264 DOI: 10.1111/febs.17015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/02/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
Interleukin-11 (IL-11) is a member of the IL-6 family of cytokines and is an important factor for bone homeostasis. IL-11 binds to and signals via the membrane-bound IL-11 receptor (IL-11R, classic signaling) or soluble forms of the IL-11R (sIL-11R, trans-signaling). Mutations in the IL11RA gene, which encodes the IL-11R, are associated with craniosynostosis, a human condition in which one or several of the sutures close prematurely, resulting in malformation of the skull. The biological mechanisms of how mutations within the IL-11R are linked to craniosynostosis are mostly unexplored. In this study, we analyze two variants of the IL-11R described in craniosynostosis patients: p.T306_S308dup, which results in a duplication of three amino-acid residues within the membrane-proximal fibronectin type III domain, and p.E364_V368del, which results in a deletion of five amino-acid residues in the so-called stalk region adjacent to the plasma membrane. The stalk region connects the three extracellular domains to the transmembrane and intracellular region of the IL-11R and contains cleavage sites for different proteases that generate sIL-11R variants. Using a combination of bioinformatics and different biochemical, molecular, and cell biology methods, we show that the IL-11R-T306_S308dup variant does not mature correctly, is intracellularly retained, and does not reach the cell surface. In contrast, the IL-11R-E364_V368del variant is fully biologically active and processed normally by proteases, thus allowing classic and trans-signaling of IL-11. Our results provide evidence that mutations within the IL11RA gene may not be causative for craniosynostosis and suggest that other regulatory mechanism(s) are involved but remain to be identified.
Collapse
Affiliation(s)
- Birte Kespohl
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Germany
| | - Anna-Lena Hegele
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Germany
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, RWTH Aachen University, Germany
| | - Hans Bakker
- Institute of Clinical Biochemistry, Hannover Medical School, Germany
| | - Falk F R Buettner
- Institute of Clinical Biochemistry, Hannover Medical School, Germany
| | - Roland Hartig
- Institute for Molecular and Clinical Immunology and Service Unit Multiparametric Bioimaging and Cytometry, Medical Faculty, University of Magdeburg, Germany
| | - Juliane Lokau
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Germany
- Institute of Clinical Biochemistry, Hannover Medical School, Germany
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, Germany
| |
Collapse
|
26
|
Shan KS, Dalal S, Thaw Dar NN, McLish O, Salzberg M, Pico BA. Molecular Targeting of the Fibroblast Growth Factor Receptor Pathway across Various Cancers. Int J Mol Sci 2024; 25:849. [PMID: 38255923 PMCID: PMC10815772 DOI: 10.3390/ijms25020849] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/19/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are a family of receptor tyrosine kinases that are involved in the regulation of cell proliferation, survival, and development. FGFR alterations including amplifications, fusions, rearrangements, and mutations can result in the downstream activation of tyrosine kinases, leading to tumor development. Targeting these FGFR alterations has shown to be effective in treating cholangiocarcinoma, urothelial carcinoma, and myeloid/lymphoid neoplasms, and there are currently four FGFR inhibitors approved by the Food and Drug Administration (FDA). There have been developments in multiple agents targeting the FGFR pathway, including selective FGFR inhibitors, ligand traps, monoclonal antibodies, and antibody-drug conjugates. However, most of these agents have variable and low responses, with some intolerable toxicities and acquired resistances. This review will summarize previous clinical experiences and current developments in agents targeting the FGFR pathway, and will also discuss future directions for FGFR-targeting agents.
Collapse
Affiliation(s)
- Khine S. Shan
- Memorial Health Care, Division of Hematology and Oncology, Pembroke Pines, FL 33028, USA; (S.D.); (N.N.T.D.); (O.M.); (M.S.)
| | | | | | | | | | | |
Collapse
|
27
|
Cooper EJ, Scholpp S. Transport and gradient formation of Wnt and Fgf in the early zebrafish gastrula. Curr Top Dev Biol 2023; 157:125-153. [PMID: 38556457 DOI: 10.1016/bs.ctdb.2023.12.003] [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] [Indexed: 04/02/2024]
Abstract
Within embryonic development, the occurrence of gastrulation is critical in the formation of multiple germ layers with many differentiative abilities. These cells are instructed through exposure to signalling molecules called morphogens. The secretion of morphogens from a source tissue creates a concentration gradient that allows distinct pattern formation in the receiving tissue. This review focuses on the morphogens Wnt and Fgf in zebrafish development. Wnt has been shown to have critical roles throughout gastrulation, including in anteroposterior patterning and neural posterisation. Fgf is also a vital signal, contributing to involution and mesodermal specification. Both morphogens have also been found to work in finely balanced synergy for processes such as neural induction. Thus, the signalling range of Wnts and Fgfs must be strictly controlled to target the correct target cells. Fgf and Wnts signal to local cells as well as to cells in the distance in a highly regulated way, requiring specific dissemination mechanisms that allow efficient and precise signalling over short and long distances. Multiple transportation mechanisms have been discovered to aid in producing a stable morphogen gradient, including short-range diffusion, filopodia-like extensions called cytonemes and extracellular vesicles, mainly exosomes. These mechanisms are specific to the morphogen that they transport and the intended signalling range. This review article discusses how spreading mechanisms in these two morphogenetic systems differ and the consequences on paracrine signalling, hence tissue patterning.
Collapse
Affiliation(s)
- Emma J Cooper
- Living Systems Institute, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Steffen Scholpp
- Living Systems Institute, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom.
| |
Collapse
|
28
|
Elhai M, Micheroli R, Houtman M, Mirrahimi M, Moser L, Pauli C, Bürki K, Laimbacher A, Kania G, Klein K, Schätzle P, Frank Bertoncelj M, Edalat SG, Keusch L, Khmelevskaya A, Toitou M, Geiss C, Rauer T, Sakkou M, Kollias G, Armaka M, Distler O, Ospelt C. The long non-coding RNA HOTAIR contributes to joint-specific gene expression in rheumatoid arthritis. Nat Commun 2023; 14:8172. [PMID: 38071204 PMCID: PMC10710443 DOI: 10.1038/s41467-023-44053-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Although patients with rheumatoid arthritis (RA) typically exhibit symmetrical joint involvement, some patients develop alternative disease patterns in response to treatment, suggesting that different molecular mechanism may underlie disease progression depending on joint location. Here, we identify joint-specific changes in RA synovium and synovial fibroblasts (SF) between knee and hand joints. We show that the long non-coding RNA HOTAIR, which is only expressed in knee SF, regulates more than 50% of this site-specific gene expression in SF. HOTAIR is downregulated after stimulation with pro-inflammatory cytokines and is expressed at lower levels in knee samples from patients with RA, compared with osteoarthritis. Knockdown of HOTAIR in knee SF increases PI-Akt signalling and IL-6 production, but reduces Wnt signalling. Silencing HOTAIR inhibits the migratory function of SF, decreases SF-mediated osteoclastogenesis, and increases the recruitment of B cells by SF. We propose that HOTAIR is an important epigenetic factor in joint-specific gene expression in RA.
Collapse
Affiliation(s)
- Muriel Elhai
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Raphael Micheroli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Miranda Houtman
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Masoumeh Mirrahimi
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Larissa Moser
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Chantal Pauli
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Kristina Bürki
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Andrea Laimbacher
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Gabriela Kania
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Kerstin Klein
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Mojca Frank Bertoncelj
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Sam G Edalat
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Leandra Keusch
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Alexandra Khmelevskaya
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Melpomeni Toitou
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Celina Geiss
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas Rauer
- Department of Trauma Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Maria Sakkou
- Institute for Bioinnovation, Biomedical Sciences Research Center (BSRC) 'Alexander Fleming', Vari, Greece
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - George Kollias
- Institute for Bioinnovation, Biomedical Sciences Research Center (BSRC) 'Alexander Fleming', Vari, Greece
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Marietta Armaka
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Caroline Ospelt
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
29
|
Harish RK, Gupta M, Zöller D, Hartmann H, Gheisari A, Machate A, Hans S, Brand M. Real-time monitoring of an endogenous Fgf8a gradient attests to its role as a morphogen during zebrafish gastrulation. Development 2023; 150:dev201559. [PMID: 37665167 PMCID: PMC10565248 DOI: 10.1242/dev.201559] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Morphogen gradients impart positional information to cells in a homogenous tissue field. Fgf8a, a highly conserved growth factor, has been proposed to act as a morphogen during zebrafish gastrulation. However, technical limitations have so far prevented direct visualization of the endogenous Fgf8a gradient and confirmation of its morphogenic activity. Here, we monitor Fgf8a propagation in the developing neural plate using a CRISPR/Cas9-mediated EGFP knock-in at the endogenous fgf8a locus. By combining sensitive imaging with single-molecule fluorescence correlation spectroscopy, we demonstrate that Fgf8a, which is produced at the embryonic margin, propagates by diffusion through the extracellular space and forms a graded distribution towards the animal pole. Overlaying the Fgf8a gradient curve with expression profiles of its downstream targets determines the precise input-output relationship of Fgf8a-mediated patterning. Manipulation of the extracellular Fgf8a levels alters the signaling outcome, thus establishing Fgf8a as a bona fide morphogen during zebrafish gastrulation. Furthermore, by hindering Fgf8a diffusion, we demonstrate that extracellular diffusion of the protein from the source is crucial for it to achieve its morphogenic potential.
Collapse
Affiliation(s)
- Rohit Krishnan Harish
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Mansi Gupta
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Daniela Zöller
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Hella Hartmann
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- CMCB Technology Platform, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Ali Gheisari
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- CMCB Technology Platform, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Anja Machate
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Stefan Hans
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Michael Brand
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| |
Collapse
|
30
|
Li D, Liu L, He X, Wang N, Sun R, Li X, Yu T, Chu XM. Roles of long non-coding RNAs in angiogenesis-related diseases: Focusing on non-neoplastic aspects. Life Sci 2023; 330:122006. [PMID: 37544376 DOI: 10.1016/j.lfs.2023.122006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/28/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Angiogenesis is a key process in organ and tissue morphogenesis, as well as growth during human development, and is coordinated by pro- and anti-angiogenic factors. When this balance is affected, the related physiological and pathological changes lead to disease. Long non-coding RNAs (lncRNAs) are an important class of non-coding RNAs that do not encode proteins, but play a dynamic role in regulating gene expression. LncRNAs have been reported to be extensively involved in angiogenesis, particularly tumor angiogenesis. The non-tumor aspects have received relatively little attention and summary, but there is a broad space for research and exploration on lncRNA-targeted angiogenesis in this area. In this review, we focus on lncRNAs in angiogenesis-related diseases other than tumors, such as atherosclerosis, myocardial infarction, stroke, diabetic complications, hypertension, osteoporosis, dermatosis, as well as, endocrine, neurological, and other systemic disorders. Moreover, multiple cell types have been implicated in lncRNA-targeted angiogenesis, but only endothelial cells have attracted widespread attention. Thus, we explore the roles of other cells. Finally, we summarize the potential research directions in the area of lncRNAs and angiogenesis that can be undertaken by combining cutting-edge technology and interdisciplinary research, which will provide new insights into the involvement of lncRNAs in angiogenesis-related diseases.
Collapse
Affiliation(s)
- Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266100, People's Republic of China
| | - Lili Liu
- School of Basic Medicine, Qingdao University, No. 308 Ningxia Road, Qingdao, People's Republic of China
| | - Xiangqin He
- Department of Echocardiography, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Ni Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266100, People's Republic of China
| | - Ruicong Sun
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266100, People's Republic of China
| | - Xiaolu Li
- Department of Echocardiography, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Tao Yu
- Institute for Translational Medicine, Qingdao University, No. 38 Dengzhou Road, 266021, People's Republic of China.
| | - Xian-Ming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266100, People's Republic of China; Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, No. 5 Zhiquan Road, Qingdao 266000, People's Republic of China.
| |
Collapse
|
31
|
Gong X, Han Z, Fan H, Wu Y, He Y, Fu Y, Zhu T, Li H. The interplay of inflammation and remodeling in the pathogenesis of chronic rhinosinusitis: current understanding and future directions. Front Immunol 2023; 14:1238673. [PMID: 37771597 PMCID: PMC10523020 DOI: 10.3389/fimmu.2023.1238673] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Chronic rhinosinusitis (CRS), a common clinical condition characterized by persistent mucosal inflammation and tissue remodeling, has a complex pathogenesis that is intricately linked to innate and adaptive immunity. A number of studies have demonstrated that a variety of immune cells and cytokines that play a vital role in mediating inflammation in CRS are also involved in remodeling of the nasal mucosa and the cells as well as different cytokines involved in remodeling in CRS are also able to exert some influence on inflammation, even though the exact relationship between inflammation and remodeling in CRS has not yet been fully elucidated. In this review, the potential role of immune cells and cytokines in regulating inflammation and remodeling of CRS mucosa has been described, starting with the immune cells and cytokines that act together in inflammation and remodeling. The goal is to aid researchers in understanding intimate connection between inflammation and remodeling of CRS and to offer novel ideas for future research.
Collapse
Affiliation(s)
- Xinru Gong
- Health and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhoutong Han
- Health and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Hongli Fan
- Health and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yuqi Wu
- Health and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yuanqiong He
- Health and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yijie Fu
- School of Preclinical Medicine, Chengdu University, Chengdu, China
| | - Tianmin Zhu
- Health and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Hui Li
- School of Preclinical Medicine, Chengdu University, Chengdu, China
| |
Collapse
|
32
|
Yoshida YG, Yan S, Xu H, Yang J. Novel Metal Nanomaterials to Promote Angiogenesis in Tissue Regeneration. ENGINEERED REGENERATION 2023; 4:265-276. [PMID: 37234753 PMCID: PMC10207714 DOI: 10.1016/j.engreg.2023.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
Angiogenesis-the formation of new blood vessels from existing blood vessels-has drawn significant attention in medical research. New techniques have been developed to control proangiogenic factors to obtain desired effects. Two important research areas are 1) understanding cellular mechanisms and signaling pathways involved in angiogenesis and 2) discovering new biomaterials and nanomaterials with proangiogenic effects. This paper reviews recent developments in controlling angiogenesis in the context of regenerative medicine and wound healing. We focus on novel proangiogenic materials that will advance the field of regenerative medicine. Specifically, we mainly focus on metal nanomaterials. We also discuss novel technologies developed to carry these proangiogenic inorganic molecules efficiently to target sites. We offer a comprehensive overview by combining existing knowledge regarding metal nanomaterials with novel developments that are still being refined to identify new nanomaterials.
Collapse
Affiliation(s)
- Yuki G. Yoshida
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Su Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Hui Xu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jian Yang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
33
|
Ahmad I, Lokau J, Kespohl B, Malik NA, Baig SM, Hartig R, Behme D, Schwab R, Altmüller J, Jameel M, Mucha S, Thiele H, Tariq M, Nürnberg P, Erdmann J, Garbers C. The interleukin-11 receptor variant p.W307R results in craniosynostosis in humans. Sci Rep 2023; 13:13479. [PMID: 37596289 PMCID: PMC10439179 DOI: 10.1038/s41598-023-39466-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 07/26/2023] [Indexed: 08/20/2023] Open
Abstract
Craniosynostosis is characterized by the premature fusion and ossification of one or more of the sutures of the calvaria, often resulting in abnormal features of the face and the skull. In cases in which growth of the brain supersedes available space within the skull, developmental delay or cognitive impairment can occur. A complex interplay of different cell types and multiple signaling pathways are required for correct craniofacial development. In this study, we report on two siblings with craniosynostosis and a homozygous missense pathogenic variant within the IL11RA gene (c.919 T > C; p.W307R). The patients present with craniosynostosis, exophthalmos, delayed tooth eruption, mild platybasia, and a basilar invagination. The p.W307R variant is located within the arginine-tryptophan-zipper within the D3 domain of the IL-11R, a structural element known to be important for the stability of the cytokine receptor. Expression of IL-11R-W307R in cells shows impaired maturation of the IL-11R, no transport to the cell surface and intracellular retention. Accordingly, cells stably expressing IL-11R-W307R do not respond when stimulated with IL-11, arguing for a loss-of-function mutation. In summary, the IL-11R-W307R variant, reported here for the first time to our knowledge, is most likely the causative variant underlying craniosynostosis in these patients.
Collapse
Affiliation(s)
- Ilyas Ahmad
- Institute for Cardiogenetics, and University Heart Center, University of Lübeck, Building 67, BMF, Ratzeburger Allee 160, 23562, Lübeck, Germany.
- DZHK (German Research Center for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, 23562, Lübeck, Germany.
- Cologne Center for Genomics (CCG) and Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany.
| | - Juliane Lokau
- Department of Pathology, Medical Faculty, Otto-Von-Guericke-University, 39120, Magdeburg, Germany
| | - Birte Kespohl
- Department of Pathology, Medical Faculty, Otto-Von-Guericke-University, 39120, Magdeburg, Germany
| | - Naveed Altaf Malik
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, 38000, Pakistan
| | - Shahid Mahmood Baig
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, 38000, Pakistan
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, 74800, Pakistan
| | - Roland Hartig
- Institute for Molecular and Clinical Immunology and Service Unit Multiparametric Bioimaging and Cytometry, Medical Faculty, Otto-Von-Guericke-University, 39120, Magdeburg, Germany
| | - Daniel Behme
- University Clinic for Neuroradiology, Medical Faculty, Otto-Von-Guericke-University, 39120, Magdeburg, Germany
| | - Roland Schwab
- University Clinic for Neuroradiology, Medical Faculty, Otto-Von-Guericke-University, 39120, Magdeburg, Germany
| | - Janine Altmüller
- Cologne Center for Genomics (CCG) and Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
- Core Facility Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Muhammad Jameel
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, 38000, Pakistan
- Centre for Regenerative Medicine and Stem Cell Research, Aga Khan University, Karachi, 74800, Pakistan
| | - Sören Mucha
- Institute for Cardiogenetics, and University Heart Center, University of Lübeck, Building 67, BMF, Ratzeburger Allee 160, 23562, Lübeck, Germany
- DZHK (German Research Center for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, 23562, Lübeck, Germany
- Institute of Epidemiology, Kiel University, 24105, Kiel, Germany
| | - Holger Thiele
- Cologne Center for Genomics (CCG) and Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Muhammad Tariq
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, 38000, Pakistan
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG) and Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, and University Heart Center, University of Lübeck, Building 67, BMF, Ratzeburger Allee 160, 23562, Lübeck, Germany
- DZHK (German Research Center for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, 23562, Lübeck, Germany
| | - Christoph Garbers
- Department of Pathology, Medical Faculty, Otto-Von-Guericke-University, 39120, Magdeburg, Germany.
- Health Campus Immunology, Infectiology and Inflammation (GC:I3), Otto-Von-Guericke-University, 39120, Magdeburg, Germany.
- Center for Health and Medical Prevention (ChaMP), Otto-Von-Guericke-University, 39120, Magdeburg, Germany.
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
34
|
Wang X, Wang T, Liang H, Wang L, Akhtar F, Shi X, Ren W, Huang B, Kou X, Chen Y, Zhan Y, Wang C. A novel SNP in NKX1-2 gene is associated with carcass traits in Dezhou donkey. BMC Genom Data 2023; 24:41. [PMID: 37550632 PMCID: PMC10408065 DOI: 10.1186/s12863-023-01145-2] [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: 03/15/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND At present, donkey meat in the market shows an imbalance between supply and demand, and there is an urgent need to cultivate a meat-type Dezhou donkey breed. On the one hand, it can improve the imbalance in the market, and on the other hand, it can promote the rapid development of the donkey industry. This study aimed to reveal significant genetic variation in the NK1 homeobox 2 gene (NKX1-2) of Dezhou donkeys and investigate the association between genotype and body size in Dezhou donkeys. RESULTS In this study, a SNP (g.54704925 A > G) was identified at the exon4 by high-depth resequencing of the Dezhou donkey NKX1-2 gene. The AA genotype is the dominant genotype. The g.54704925 A > G site was significantly associated with body length, thoracic girth, and hide weight (P < 0.05), while it was highly significantly associated with body height and carcass weight (P < 0.01) in Dezhou donkeys. CONCLUSION Overall, the results of this study showed that the NKX1-2 gene could be a candidate gene for breeding meat-type Dezhou donkeys, and the g.54704925 A > G locus could be used as a marker locus for selection and breeding.
Collapse
Affiliation(s)
- Xinrui Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Tianqi Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Huili Liang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Liyuan Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Faheem Akhtar
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Xiaoyuan Shi
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Wei Ren
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Bingjian Huang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Xiyan Kou
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Yinghui Chen
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Yandong Zhan
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Changfa Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy and Agricultural Engineering, Liaocheng University, Liaocheng, 252059, China.
| |
Collapse
|
35
|
Silva-García CG. Devo-Aging: Intersections Between Development and Aging. GeroScience 2023; 45:2145-2159. [PMID: 37160658 PMCID: PMC10651630 DOI: 10.1007/s11357-023-00809-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/25/2023] [Indexed: 05/11/2023] Open
Abstract
There are two fundamental questions in developmental biology. How does a single fertilized cell give rise to a whole body? and how does this body later produce progeny? Synchronization of these embryonic and postembryonic developments ensures continuity of life from one generation to the next. An enormous amount of work has been done to unravel the molecular mechanisms behind these processes, but more recently, modern developmental biology has been expanded to study development in wider contexts, including regeneration, environment, disease, and even aging. However, we have just started to understand how the mechanisms that govern development also regulate aging. This review discusses examples of signaling pathways involved in development to elucidate how their regulation influences healthspan and lifespan. Therefore, a better knowledge of developmental signaling pathways stresses the possibility of using them as innovative biomarkers and targets for aging and age-related diseases.
Collapse
Affiliation(s)
- Carlos Giovanni Silva-García
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA.
- Center on the Biology of Aging, Brown University, Providence, RI, USA.
| |
Collapse
|
36
|
Lana JF, Purita J, Everts PA, De Mendonça Neto PAT, de Moraes Ferreira Jorge D, Mosaner T, Huber SC, Azzini GOM, da Fonseca LF, Jeyaraman M, Dallo I, Santos GS. Platelet-Rich Plasma Power-Mix Gel (ppm)-An Orthobiologic Optimization Protocol Rich in Growth Factors and Fibrin. Gels 2023; 9:553. [PMID: 37504432 PMCID: PMC10379106 DOI: 10.3390/gels9070553] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023] Open
Abstract
Platelet- and fibrin-rich orthobiologic products, such as autologous platelet concentrates, have been extensively studied and appreciated for their beneficial effects on multiple conditions. Platelet-rich plasma (PRP) and its derivatives, including platelet-rich fibrin (PRF), have demonstrated encouraging outcomes in clinical and laboratory settings, particularly in the treatment of musculoskeletal disorders such as osteoarthritis (OA). Although PRP and PRF have distinct characteristics, they share similar properties. The relative abundance of platelets, peripheral blood cells, and molecular components in these orthobiologic products stimulates numerous biological pathways. These include inflammatory modulation, augmented neovascularization, and the delivery of pro-anabolic stimuli that regulate cell recruitment, proliferation, and differentiation. Furthermore, the fibrinolytic system, which is sometimes overlooked, plays a crucial role in musculoskeletal regenerative medicine by regulating proteolytic activity and promoting the recruitment of inflammatory cells and mesenchymal stem cells (MSCs) in areas of tissue regeneration, such as bone, cartilage, and muscle. PRP acts as a potent signaling agent; however, it diffuses easily, while the fibrin from PRF offers a durable scaffolding effect that promotes cell activity. The combination of fibrin with hyaluronic acid (HA), another well-studied orthobiologic product, has been shown to improve its scaffolding properties, leading to more robust fibrin polymerization. This supports cell survival, attachment, migration, and proliferation. Therefore, the administration of the "power mix" containing HA and autologous PRP + PRF may prove to be a safe and cost-effective approach in regenerative medicine.
Collapse
Affiliation(s)
- José Fábio Lana
- OrthoRegen Group, Max-Planck University, Indaiatuba 13343-060, Brazil
| | | | | | | | | | - Tomas Mosaner
- Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil
| | - Stephany Cares Huber
- Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil
| | | | | | - Madhan Jeyaraman
- Department of Orthopaedics, Faculty of Medicine, Sri Lalithambigai Medical College and Hospital, Tamil Nadu 600095, India
| | - Ignacio Dallo
- SportMe Medical Center, Department of Orthopaedic Surgery and Sports Medicine, Unit of Biological Therapies and MSK Interventionism, 41013 Seville, Spain
| | - Gabriel Silva Santos
- Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil
| |
Collapse
|
37
|
Derham JM, Kalsotra A. The discovery, function, and regulation of epithelial splicing regulatory proteins (ESRP) 1 and 2. Biochem Soc Trans 2023; 51:1097-1109. [PMID: 37314029 PMCID: PMC11298080 DOI: 10.1042/bst20221124] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023]
Abstract
Alternative splicing is a broad and evolutionarily conserved mechanism to diversify gene expression and functionality. The process relies on RNA binding proteins (RBPs) to recognize and bind target sequences in pre-mRNAs, which allows for the inclusion or skipping of various alternative exons. One recently discovered family of RBPs is the epithelial splicing regulatory proteins (ESRP) 1 and 2. Here, we discuss the structure and physiological function of the ESRPs in a variety of contexts. We emphasize the current understanding of their splicing activities, using the classic example of fibroblast growth factor receptor 2 mutually exclusive splicing. We also describe the mechanistic roles of ESRPs in coordinating the splicing and functional output of key signaling pathways that support the maintenance of, or shift between, epithelial and mesenchymal cell states. In particular, we highlight their functions in the development of mammalian limbs, the inner ear, and craniofacial structure while discussing the genetic and biochemical evidence that showcases their conserved roles in tissue regeneration, disease, and cancer pathogenesis.
Collapse
Affiliation(s)
- Jessica M. Derham
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Auinash Kalsotra
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Cancer Center @ Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute of Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
38
|
Myo AC, Kobayashi Y, Niki Y, Kamimoto H, Moriyama K. Exosome-mediated small interfering RNA delivery inhibits aberrant osteoblast differentiation in Apert syndrome model mice. Arch Oral Biol 2023; 153:105753. [PMID: 37348363 DOI: 10.1016/j.archoralbio.2023.105753] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/06/2023] [Accepted: 06/17/2023] [Indexed: 06/24/2023]
Abstract
OBJECTIVE Apert syndrome, an autosomal dominant congenital disorder characterized by craniosynostosis, is caused by a missense mutation (S252W or P253R) in fibroblast growth factor receptor 2 (FGFR2). Exosomes are naturally occurring carriers that deliver nucleic acids, including small interfering RNA (siRNA), to induce gene silencing. This study aimed to develop siRNA-loaded exosomes (Ex-siRNAFgfr2S252W) to silence the Fgfr2S252W gain-of-function mutation, thereby inhibiting the increased osteoblastic differentiation caused by the constitutive activation of FGFR2 signaling in calvarial osteoblastic cells isolated from Apert syndrome model mice. DESIGN Primary calvarial osteoblast-like cells were isolated from the embryonic calvarial sutures of the Apert syndrome model (Fgfr2S252W/+) and littermate wild-type mice (Ap-Ob and Wt-Ob, respectively). Exosomes were extracted from the serum of wild-type mice, validated using biomarkers, and used to encapsulate siRNAs. After exosome-mediated siRNA transfection, cells were analyzed under a fluorescence microscope to validate the delivery of Ex-siRNAFgfr2S252W, followed by western blot and real-time reverse transcription polymerase chain reaction analyses. RESULTS After 24 h of Ex-siRNAFgfr2S252W delivery in both Ap-Ob and Wt-Ob, siRNA-loaded exosome delivery was validated. Moreover, p44/42 mitogen-activated protein kinase (MAPK) phosphorylation, runt-related transcription factor 2 (Runx2), and collagen type 1 alpha 1 (Col1a1) mRNA expression, and alkaline phosphatase (ALP) activity were significantly increased in Ap-Ob. The levels of phospho-p44/42 protein, Runx2, Col1a1, and ALP were significantly decreased after Ex-siRNAFgfr2S252W transfection but did not affect Wt-Ob. CONCLUSIONS These results indicate that exosome-mediated delivery of siRNA targeting Fgfr2S252W is a potential non-invasive treatment for aberrant FGF/FGFR signaling.
Collapse
Affiliation(s)
- Aye Chan Myo
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45, Bunkyo-ku, Tokyo 113-8549, Japan
| | - Yukiho Kobayashi
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45, Bunkyo-ku, Tokyo 113-8549, Japan.
| | - Yuki Niki
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45, Bunkyo-ku, Tokyo 113-8549, Japan
| | - Hiroyuki Kamimoto
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45, Bunkyo-ku, Tokyo 113-8549, Japan
| | - Keiji Moriyama
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45, Bunkyo-ku, Tokyo 113-8549, Japan
| |
Collapse
|
39
|
Zhao X, Erhardt S, Sung K, Wang J. FGF signaling in cranial suture development and related diseases. Front Cell Dev Biol 2023; 11:1112890. [PMID: 37325554 PMCID: PMC10267317 DOI: 10.3389/fcell.2023.1112890] [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: 11/30/2022] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Suture mesenchymal stem cells (SMSCs) are a heterogeneous stem cell population with the ability to self-renew and differentiate into multiple cell lineages. The cranial suture provides a niche for SMSCs to maintain suture patency, allowing for cranial bone repair and regeneration. In addition, the cranial suture functions as an intramembranous bone growth site during craniofacial bone development. Defects in suture development have been implicated in various congenital diseases, such as sutural agenesis and craniosynostosis. However, it remains largely unknown how intricate signaling pathways orchestrate suture and SMSC function in craniofacial bone development, homeostasis, repair and diseases. Studies in patients with syndromic craniosynostosis identified fibroblast growth factor (FGF) signaling as an important signaling pathway that regulates cranial vault development. A series of in vitro and in vivo studies have since revealed the critical roles of FGF signaling in SMSCs, cranial suture and cranial skeleton development, and the pathogenesis of related diseases. Here, we summarize the characteristics of cranial sutures and SMSCs, and the important functions of the FGF signaling pathway in SMSC and cranial suture development as well as diseases caused by suture dysfunction. We also discuss emerging current and future studies of signaling regulation in SMSCs.
Collapse
Affiliation(s)
- Xiaolei Zhao
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- MD Anderson Cancer Center and UT Health Graduate School of Biomedical Sciences, The University of Texas, Houston, TX, United States
| | - Kihan Sung
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- MD Anderson Cancer Center and UT Health Graduate School of Biomedical Sciences, The University of Texas, Houston, TX, United States
| |
Collapse
|
40
|
Goutam RS, Kumar V, Lee U, Kim J. Exploring the Structural and Functional Diversity among FGF Signals: A Comparative Study of Human, Mouse, and Xenopus FGF Ligands in Embryonic Development and Cancer Pathogenesis. Int J Mol Sci 2023; 24:ijms24087556. [PMID: 37108717 PMCID: PMC10146080 DOI: 10.3390/ijms24087556] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Fibroblast growth factors (FGFs) encode a large family of growth factor proteins that activate several intracellular signaling pathways to control diverse physiological functions. The human genome encodes 22 FGFs that share a high sequence and structural homology with those of other vertebrates. FGFs orchestrate diverse biological functions by regulating cellular differentiation, proliferation, and migration. Dysregulated FGF signaling may contribute to several pathological conditions, including cancer. Notably, FGFs exhibit wide functional diversity among different vertebrates spatiotemporally. A comparative study of FGF receptor ligands and their diverse roles in vertebrates ranging from embryonic development to pathological conditions may expand our understanding of FGF. Moreover, targeting diverse FGF signals requires knowledge regarding their structural and functional heterogeneity among vertebrates. This study summarizes the current understanding of human FGF signals and correlates them with those in mouse and Xenopus models, thereby facilitating the identification of therapeutic targets for various human disorders.
Collapse
Affiliation(s)
- Ravi Shankar Goutam
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- iPS Bio, Inc., 3F, 16 Daewangpangyo-ro 712 Beon-gil, Bundang-gu, Seongnam-si 13522, Republic of Korea
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
41
|
Ghaly M, Proulx J, Borgmann K, Park IW. Novel role of HIV-1 Nef in regulating the ubiquitination of cellular proteins. Front Cell Infect Microbiol 2023; 13:1106591. [PMID: 36968110 PMCID: PMC10031067 DOI: 10.3389/fcimb.2023.1106591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/16/2023] [Indexed: 03/10/2023] Open
Abstract
Our recent data established that HIV-1 Nef is pivotal in determining the fate of cellular proteins by modulating ubiquitination. However, it is unknown which proteins are ubiquitinated in the presence of Nef, a question critical for understanding the proliferation/restriction strategies of HIV-1 in infected cells. To identify cellular proteins ubiquitinated by Nef, we conducted a proteomic analysis of cellular proteins in the presence and absence of Nef. Proteomic analysis in HEK293T cells indicated that 93 proteins were upregulated and 232 were downregulated in their ubiquitination status by Nef. Computational analysis classified these proteins based on molecular function, biological process, subcellular localization, and biological pathway. Of those proteins, we found a majority of molecular functions to be involved in binding and catalytic activity. With respect to biological processes, a significant portion of the proteins identified were related to cellular and metabolic processes. Subcellular localization analysis showed the bulk of proteins to be localized to the cytosol and cytosolic compartments, which is consistent with the known function and location of Nef during HIV-1 infection. As for biological pathways, the wide range of affected proteins was denoted by the multiple modes to fulfill function, as distinguished from a strictly singular means, which was not detected. Among these ubiquitinated proteins, six were found to directly interact with Nef, wherein two were upregulated and four downregulated. We also identified 14 proteins involved in protein stability through directly participating in the Ubiquitin Proteasome System (UPS)-mediated proteasomal degradation pathway. Of those proteins, we found six upregulated and eight downregulated. Taken together, these analyses indicate that HIV-1 Nef is integral to regulating the stability of various cellular proteins via modulating ubiquitination. The molecular mechanisms directing Nef-triggered regulation of cellular protein ubiquitination are currently under investigation.
Collapse
|
42
|
Numakawa T, Kajihara R. Neurotrophins and Other Growth Factors in the Pathogenesis of Alzheimer’s Disease. Life (Basel) 2023; 13:life13030647. [PMID: 36983803 PMCID: PMC10051261 DOI: 10.3390/life13030647] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/12/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
The involvement of the changed expression/function of neurotrophic factors in the pathogenesis of neurodegenerative diseases, including Alzheimer’s disease (AD), has been suggested. AD is one of the age-related dementias, and is characterized by cognitive impairment with decreased memory function. Developing evidence demonstrates that decreased cell survival, synaptic dysfunction, and reduced neurogenesis are involved in the pathogenesis of AD. On the other hand, it is well known that neurotrophic factors, especially brain-derived neurotrophic factor (BDNF) and its high-affinity receptor TrkB, have multiple roles in the central nervous system (CNS), including neuronal maintenance, synaptic plasticity, and neurogenesis, which are closely linked to learning and memory function. Thus, many investigations regarding therapeutic approaches to AD, and/or the screening of novel drug candidates for its treatment, focus on upregulation of the BDNF/TrkB system. Furthermore, current studies also demonstrate that GDNF, IGF1, and bFGF, which play roles in neuroprotection, are associated with AD. In this review, we introduce data demonstrating close relationships between the pathogenesis of AD, neurotrophic factors, and drug candidates, including natural compounds that upregulate the BDNF-mediated neurotrophic system.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Correspondence:
| | - Ryutaro Kajihara
- Department of Biomedical Laboratory Sciences, Faculty of Life Science, Kumamoto University, Kumamoto 862-0976, Japan
| |
Collapse
|
43
|
Alam MK, Alfawzan AA, Abutayyem H, Kanwal B, Alswairki HJ, Verma S, Ganji KK, Munisekhar MS, Siddiqui AA, Fahim A. Craniofacial characteristics in Crouzon's syndrome: A systematic review and meta-analysis. Sci Prog 2023; 106:368504231156297. [PMID: 36803068 PMCID: PMC10358517 DOI: 10.1177/00368504231156297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The purpose of this meta-analysis was to strengthen the credibility of primary research results by combining open-source scientific material, namely a comparison of craniofacial features (Cfc) between Crouzon's syndrome (CS) patients and non-CS populations. All articles published up to October 7, 2021, were included in the search of PubMed, Google Scholar, Scopus, Medline, and Web of Science. The PRISMA guidelines were followed to conduct this study. PECO framework was applied in the following ways: Those who have CS are denoted by the letter P, those who have been diagnosed with CS via clinical or genetic means by the letter E, those who do not have CS by the letter C, and those who have a Cfc of CS by the letter O. Independent reviewers collected the data and ranked the publications based on their adherence to the Newcastle-Ottawa Quality Assessment Scale. A total of six case-control studies were reviewed for this meta-analysis. Due to the large variation in cephalometric measures, only those published in at least two previous studies were included. This analysis found that CS patients had a smaller skull and mandible volumes than those without CS.in terms of SNA° (MD = -2.33, p = <0.001, I2 = 83.6%) and ANB°(MD = -1.89, p = <0.005, I2 = 93.1%)), as well as ANS (MD = -1.87, p = 0.001, I2 = 96.5%)) and SN/PP (MD = -1.99, p = 0.036, I2 = 77.3%)). In comparison to the general population, people with CS tend to have shorter and flatter cranial bases, smaller orbital volumes, and cleft palates. They differ from the general population in having a shorter skull base and more V-shaped maxillary arches.
Collapse
Affiliation(s)
- Mohammad Khursheed Alam
- Orthodontics, Preventive Dentistry Department, College of Dentistry, Jouf University, Saudi Arabia
| | - Ahmed Ali Alfawzan
- Department of Preventive Dentistry, College of Dentistry in Ar Rass, Qassim University, Ar Rass, Saudi Arabia
| | - Huda Abutayyem
- Department of Clinical Sciences, Center of Medical and Bio-Allied Health Sciences Research, College of Dentistry, Ajman University, Ajman, United Arab Emirates
| | | | | | - Swati Verma
- Division of Orthodontics and Dentofacial Deformities, Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi, India
| | - Kiran Kumar Ganji
- Preventive Dentistry Department, College of Dentistry, Jouf University, Saudi Arabia
| | | | - Ammar A Siddiqui
- Preventive Dentistry Department, College of Dentistry, University of Ha'il, Saudi Arabia
| | - Ayesha Fahim
- College of Dentistry, University of Lahore, Lahore, Pakistan
| |
Collapse
|
44
|
Colin-Pierre C, El Baraka O, Danoux L, Bardey V, André V, Ramont L, Brézillon S. Regulation of stem cell fate by HSPGs: implication in hair follicle cycling. NPJ Regen Med 2022; 7:77. [PMID: 36577752 PMCID: PMC9797564 DOI: 10.1038/s41536-022-00267-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 11/30/2022] [Indexed: 12/29/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are part of proteoglycan family. They are composed of heparan sulfate (HS)-type glycosaminoglycan (GAG) chains covalently linked to a core protein. By interacting with growth factors and/or receptors, they regulate numerous pathways including Wnt, hedgehog (Hh), bone morphogenic protein (BMP) and fibroblast growth factor (FGF) pathways. They act as inhibitor or activator of these pathways to modulate embryonic and adult stem cell fate during organ morphogenesis, regeneration and homeostasis. This review summarizes the knowledge on HSPG structure and classification and explores several signaling pathways regulated by HSPGs in stem cell fate. A specific focus on hair follicle stem cell fate and the possibility to target HSPGs in order to tackle hair loss are discussed in more dermatological and cosmeceutical perspectives.
Collapse
Affiliation(s)
- Charlie Colin-Pierre
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.
- BASF Beauty Care Solutions France SAS, Pulnoy, France.
| | | | - Louis Danoux
- BASF Beauty Care Solutions France SAS, Pulnoy, France
| | | | - Valérie André
- BASF Beauty Care Solutions France SAS, Pulnoy, France
| | - Laurent Ramont
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
- CHU de Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
| |
Collapse
|
45
|
Beird HC, Bielack SS, Flanagan AM, Gill J, Heymann D, Janeway KA, Livingston JA, Roberts RD, Strauss SJ, Gorlick R. Osteosarcoma. Nat Rev Dis Primers 2022; 8:77. [PMID: 36481668 DOI: 10.1038/s41572-022-00409-y] [Citation(s) in RCA: 219] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2022] [Indexed: 12/13/2022]
Abstract
Osteosarcoma is the most common primary malignant tumour of the bone. Osteosarcoma incidence is bimodal, peaking at 18 and 60 years of age, and is slightly more common in males. The key pathophysiological mechanism involves several possible genetic drivers of disease linked to bone formation, causing malignant progression and metastasis. While there have been significant improvements in the outcome of patients with localized disease, with event-free survival outcomes exceeding 60%, in patients with metastatic disease, event-free survival outcomes remain poor at less than 30%. The suspicion of osteosarcoma based on radiographs still requires pathological evaluation of a bone biopsy specimen for definitive diagnosis and CT imaging of the chest should be performed to identify lung nodules. So far, population-based screening and surveillance strategies have not been implemented due to the rarity of osteosarcoma and the lack of reliable markers. Current screening focuses only on groups at high risk such as patients with genetic cancer predisposition syndromes. Management of osteosarcoma requires a multidisciplinary team of paediatric and medical oncologists, orthopaedic and general surgeons, pathologists, radiologists and specialist nurses. Survivors of osteosarcoma require specialized medical follow-up, as curative treatment consisting of chemotherapy and surgery has long-term adverse effects, which also affect the quality of life of patients. The development of osteosarcoma model systems and related research as well as the evaluation of new treatment approaches are ongoing to improve disease outcomes, especially for patients with metastases.
Collapse
Affiliation(s)
- Hannah C Beird
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stefan S Bielack
- Pediatric Oncology, Hematology, Immunology, Klinikum Stuttgart - Olgahospital, Stuttgart Cancer Center, Stuttgart, Germany
| | - Adrienne M Flanagan
- Research Department of Pathology, Cancer Institute, University College London, London, UK
| | - Jonathan Gill
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dominique Heymann
- Nantes Université, CNRS, UMR6286, US2B, Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Katherine A Janeway
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - J Andrew Livingston
- Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ryan D Roberts
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sandra J Strauss
- University College London Hospitals NHS Foundation Trust, University College London, London, UK
| | - Richard Gorlick
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA. .,Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
46
|
FGFR Inhibitors in Cholangiocarcinoma-A Novel Yet Primary Approach: Where Do We Stand Now and Where to Head Next in Targeting This Axis? Cells 2022; 11:cells11233929. [PMID: 36497187 PMCID: PMC9737583 DOI: 10.3390/cells11233929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/20/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinomas (CCAs) are rare but aggressive tumours with poor diagnosis and limited treatment options. Molecular targeted therapies became a promising proposal for patients after progression under first-line chemical treatment. In light of an escalating prevalence of CCA, it is crucial to fully comprehend its pathophysiology, aetiology, and possible targets in therapy. Such knowledge would play a pivotal role in searching for new therapeutic approaches concerning diseases' symptoms and their underlying causes. Growing evidence showed that fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) pathway dysregulation is involved in a variety of processes during embryonic development and homeostasis as well as tumorigenesis. CCA is known for its close correlation with the FGF/FGFR pathway and targeting this axis has been proposed in treatment guidelines. Bearing in mind the significance of molecular targeted therapies in different neoplasms, it seems most reasonable to move towards intensive research and testing on these in the case of CCA. However, there is still a need for more data covering this topic. Although positive results of many pre-clinical and clinical studies are discussed in this review, many difficulties lie ahead. Furthermore, this review presents up-to-date literature regarding the outcomes of the latest clinical data and discussion over future directions of FGFR-directed therapies in patients with CCA.
Collapse
|
47
|
Fibroblast growth factor 5 overexpression ameliorated lipopolysaccharide-induced apoptosis of hepatocytes through regulation of the phosphoinositide-3-kinase/protein kinase B pathway. Chin Med J (Engl) 2022; 135:2859-2868. [PMID: 36728504 PMCID: PMC9943982 DOI: 10.1097/cm9.0000000000002540] [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: 08/19/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Sepsis is a systemic inflammatory syndrome induced by several infectious agents. Multiple organs are affected by sepsis, including the liver, which plays an important role in metabolism and immune homeostasis. Fibroblast growth factors (FGFs) participate in several biological processes, although the role of FGF5 in sepsis is unclear. METHODS In this study, lipopolysaccharide (LPS) was administrated to mice to establish a sepsis-induced liver injury. A similar in vitro study was conducted using L-02 hepatocytes. Western blot and immunohistochemistry staining were performed to evaluate the FGF5 expression level in liver tissues and cells. Inflammatory cell infiltrations, cleaved-caspase-3 expressions, reactive oxygen species and levels of inflammatory cytokines were detected by immunofluorescence, dihydroethidium staining, and reverse transcription quantitative polymerase chain reaction analysis, respectively. Flow cytometry was used to detect the apoptosis level of cells. In addition, ribonucleic acid (RNA)-sequencing was applied to explore the possible mechanism by which FGF5 exerted effects. RESULTS LPS administration caused FGF5 down-regulation in the mouse liver as well as in L-02 hepatocytes. Additionally, with FGF5 overexpression, liver injury and the level of hepatocyte apoptosis were ameliorated. Further, RNA sequencing performed in hepatocytes revealed the phosphoinositide-3-kinase/protein kinase B (PI3K/AKT) pathway as a possible pathway regulated by FGF5 . This was supported using an inhibitor of the PI3K/AKT pathway, which abrogated the protective effect of FGF5 in LPS-induced hepatocyte injury. CONCLUSION The anti-apoptotic effect of FGF5 on hepatocytes suffering from LPS has been demonstrated and was dependent on the activation of the PI3K/AKT signaling pathway.
Collapse
|
48
|
Bölük A, Yavuz M, Demircan T. Axolotl: A resourceful vertebrate model for regeneration and beyond. Dev Dyn 2022; 251:1914-1933. [PMID: 35906989 DOI: 10.1002/dvdy.520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/04/2022] [Accepted: 07/21/2022] [Indexed: 01/30/2023] Open
Abstract
The regenerative capacity varies significantly among the animal kingdom. Successful regeneration program in some animals results in the functional restoration of tissues and lost structures. Among the highly regenerative animals, axolotl provides multiple experimental advantages with its many extraordinary characteristics. It has been positioned as a regeneration model organism due to its exceptional renewal capacity, including the internal organs, central nervous system, and appendages, in a scar-free manner. In addition to this unique regeneration ability, the observed low cancer incidence, its resistance to carcinogens, and the reversing effect of its cell extract on neoplasms strongly suggest its usability in cancer research. Axolotl's longevity and efficient utilization of several anti-aging mechanisms underline its potential to be employed in aging studies.
Collapse
Affiliation(s)
- Aydın Bölük
- School of Medicine, Muğla Sıtkı Koçman University, Muğla, Turkey
| | - Mervenur Yavuz
- Institute of Health Sciences, Muğla Sıtkı Koçman University, Muğla, Turkey
| | - Turan Demircan
- Department of Medical Biology, School of Medicine, Muğla Sıtkı Koçman University, Muğla, Turkey
| |
Collapse
|
49
|
Abu-Elmagd M, Assidi M, Alrefaei AF, Rebai A. Editorial: Advances in genomic and genetic tools, and their applications for understanding embryonic development and human diseases. Front Cell Dev Biol 2022; 10:1016400. [PMID: 36478744 PMCID: PMC9720382 DOI: 10.3389/fcell.2022.1016400] [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: 08/10/2022] [Accepted: 10/04/2022] [Indexed: 10/10/2023] Open
Abstract
Significant advances have been recently made in the development of the genetic and genomic platforms. This has greatly contributed to a better understanding of gene expression and regulation machinery. Consequently, this led to considerable progress in unraveling evidence of the genotype-phenotype correlation between normal/abnormal embryonic development and human disease complexity. For example, advanced genomic tools such as next-generation sequencing, and microarray-based CGH have substantially helped in the identification of gene and copy number variants associated with diseases as well as in the discovery of causal gene mutations. In addition, bioinformatic analysis tools of genome annotation and comparison have greatly aided in data analysis for the interpretation of the genetic variants at the individual level. This has unlocked potential possibilities for real advances toward new therapies in personalized medicine for the targeted treatment of human diseases. However, each of these genomic and bioinformatics tools has its limitations and hence further efforts are required to implement novel approaches to overcome these limitations. It could be possible that the use of more than one platform for genotype-phenotype deep analysis is an effective approach to disentangling the cause and treatment of the disease complexities. Our research topic aimed at deciphering these complexities by shedding some light on the recent applications of the basic and advanced genetic/genomic and bioinformatics approaches. These include studying gene-gene, protein-protein, and gene-environment interactions. We, in addition, aimed at a better understanding of the link between normal/abnormal embryonic development and the cause of human disease induction.
Collapse
Affiliation(s)
- Muhammad Abu-Elmagd
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mourad Assidi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulmajeed F. Alrefaei
- Department of Biology, Jamoum University College, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Ahmed Rebai
- Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
50
|
Lee DG, Kim YK, Baek KH. The bHLH Transcription Factors in Neural Development and Therapeutic Applications for Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms232213936. [PMID: 36430421 PMCID: PMC9696289 DOI: 10.3390/ijms232213936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
The development of functional neural circuits in the central nervous system (CNS) requires the production of sufficient numbers of various types of neurons and glial cells, such as astrocytes and oligodendrocytes, at the appropriate periods and regions. Hence, severe neuronal loss of the circuits can cause neurodegenerative diseases such as Huntington's disease (HD), Parkinson's disease (PD), Alzheimer's disease (AD), and Amyotrophic Lateral Sclerosis (ALS). Treatment of such neurodegenerative diseases caused by neuronal loss includes some strategies of cell therapy employing stem cells (such as neural progenitor cells (NPCs)) and gene therapy through cell fate conversion. In this report, we review how bHLH acts as a regulator in neuronal differentiation, reprogramming, and cell fate determination. Moreover, several different researchers are conducting studies to determine the importance of bHLH factors to direct neuronal and glial cell fate specification and differentiation. Therefore, we also investigated the limitations and future directions of conversion or transdifferentiation using bHLH factors.
Collapse
Affiliation(s)
- Dong Gi Lee
- Joint Section of Science in Environmental Technology, Food Technology, and Molecular Biotechnology, Ghent University, Incheon 21569, Korea
| | - Young-Kwang Kim
- Department of Biomedical Science, CHA Stem Cell Institute, CHA University, Seongnam 13488, Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA Stem Cell Institute, CHA University, Seongnam 13488, Korea
- Correspondence: ; Tel.: +82-31-881-7134
| |
Collapse
|