51
|
Matsuoka R, Kitajima K, Nii T, Zou Z, Tanaka K, Joo K, Ohkawa Y, Ohga S, Meno C. Hyperglycaemia induces diet-dependent defects of the left-right axis by lowering intracellular pH. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167550. [PMID: 39442590 DOI: 10.1016/j.bbadis.2024.167550] [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: 08/06/2024] [Revised: 10/02/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Pregestational diabetes is a risk factor for congenital anomalies, including heterotaxy syndrome, a rare birth defect characterized by the abnormal arrangement of organs relative to the left-right (L-R) body axis. To provide insight into the underlying mechanism by which diabetes induces heterotaxy, we here analyzed the L-R axis of mouse embryos of diabetic dams. Various Pitx2 expression patterns indicative of disruption of L-R axis formation were apparent in such embryos. Expression of Nodal at the node, which triggers a Nodal-Pitx2 expression cascade in lateral plate mesoderm, showed marked regression associated with L-R axis malformation. This regression was similar to that apparent in Wnt3a-/- embryos, and canonical Wnt signalling was indeed found to be downregulated in embryos of diabetic dams. RNA sequencing revealed dysregulation of glycolysis in embryos of diabetic dams, and high glucose lowered intracellular pH in the primitive streak, leading to the suppression of Wnt signalling and the regression of Nodal expression. Of note, maternal vitamin A intake increased the incidence of L-R axis defects in embryos of diabetic dams, with dysregulation of retinoic acid metabolism being apparent in these embryos and in Wnt3a-/- embryos. Our results shed light on the mechanisms underlying embryopathies associated with maternal diabetes and suggest the importance of diet for prevention of heterotaxy.
Collapse
Affiliation(s)
- Ryohei Matsuoka
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Keiko Kitajima
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takenobu Nii
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Zhaonan Zou
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kaori Tanaka
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Kunihiko Joo
- Department of Cardiovascular Surgery, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Chikara Meno
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
52
|
Hong Y, Lin Q, Zhang Y, Liu J, Zheng Z. Research Progress of Ribosomal Proteins in Reproductive Development. Int J Mol Sci 2024; 25:13151. [PMID: 39684863 DOI: 10.3390/ijms252313151] [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: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Ribosomal proteins constitute the principal components of ribosomes, and their functions span a wide spectrum. Recent investigations have unveiled their involvement in oocyte and embryo development, playing a pivotal role in reproductive development. Numerous pieces of evidence indicate that ribosomal proteins participate in the regulation of various cellular activities, including nucleolar stress, oxidative stress, cell proliferation and autophagy. Despite these findings, the precise mechanisms through which ribosomal proteins influence reproductive development via these cellular activities remain elusive. Therefore, elucidating the mechanisms of action is essential for a comprehensive understanding of the role and function of ribosomal proteins in reproductive development. This paper systematically reviews the progress in research on nucleolar stress, oxidative stress, cell proliferation and autophagy concerning ribosomal proteins during reproductive development. Furthermore, we explore the potential of ribosomal proteins as diagnostic markers for various diseases. Additionally, we propose the development of drugs and therapies targeting ribosomal proteins, underscoring the potential for novel medical interventions in the context of reproductive health.
Collapse
Affiliation(s)
- Yuqi Hong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Qisheng Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yuan Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jilong Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhanhong Zheng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
53
|
Wu L, Wang J, Chai L, Chen J, Jin X. Roles of deubiquitinases in urologic cancers (Review). Oncol Lett 2024; 28:609. [PMID: 39525605 PMCID: PMC11544529 DOI: 10.3892/ol.2024.14743] [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: 07/23/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Human health is endangered by the occurrence and progression of urological cancers, including renal cell carcinoma, prostate cancer and bladder cancer, which are usually associated with the activation of oncogenic factors and inhibition of cancer suppressors. The primary mechanism for protein breakdown in cells is the ubiquitin-proteasome system, whilst deubiquitinases contribute to the reversal of this process. However, both are important for protein homeostasis. Deubiquitination may also be involved in the control of the cell cycle, proliferation and apoptosis, and dysregulated deubiquitination is associated with the malignant transformation, invasion and metastasis of urologic malignancies. Therefore, a comprehensive summary of the mechanisms underlying deubiquitination in urological cancers may provide novel strategies and insights for diagnosis and treatment. The present review aimed to methodically clarify the role of deubiquitinating enzymes in urinary system cancers as well as their prospective application prospects for clinical treatment.
Collapse
Affiliation(s)
- Liangpei Wu
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jiahui Wang
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Lin Chai
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jun Chen
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
54
|
Zhang K, Zhang Y, Qin J, Zhu H, Liu N, Sun D, Yin Y, Mao S, Zhu W, Huang Z, Liu J. Early concentrate starter introduction induces rumen epithelial parakeratosis by blocking keratinocyte differentiation with excessive ruminal butyrate accumulation. J Adv Res 2024; 66:71-86. [PMID: 38128723 PMCID: PMC11674766 DOI: 10.1016/j.jare.2023.12.016] [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: 08/04/2023] [Revised: 11/27/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023] Open
Abstract
INTRODUCTION Rumen epithelial parakeratosis, a common disease in ruminants caused by abnormalities in the ruminal stratified squamous epithelial keratinization process, negatively impacts ruminant health and performance. However, we still lack a comprehensive perception of the underlying mechanisms and the predisposing factors for this disorder. OBJECTIVES Here, we investigated rumen epithelial cell heterogeneity, differentiation trajectories, and cornification to clarify the rumen epithelial keratinization process and discern the key ruminal metabolites contributing to rumen epithelial parakeratosis. METHODS Twenty-four 14-day-old lambs were divided into three groups, including only milk feeding, milk plus alfalfa hay feeding, and milk plus corn-soybean concentrate starter feeding. At 42 days of age, the lambs were slaughtered, and rumen tissues were collected for single-cell RNA-sequencing (scRNA-seq), immunofluorescence, and quantitative real-time PCR (qRT-PCR) analyses. Ruminal fluid samples were collected for metabolomic analyses. Rumen epithelial organoid was used to verify the key ruminal metabolites contributing to parakeratosis. RESULTS As expected, we observed that concentrate starter introduction resulted in rumen epithelial parakeratosis. Moreover, scRNA-seq analysis revealed a developmental impediment in the transition from differentiated keratinocytes to terminally differentiated keratinocytes (TDK) in lambs with concentrate starter introduction. Immunofluorescence and qRT-PCR analyses further verified the location and expression of marker genes of TDK. Metabolomic analysis showed a robust positive correlation between ruminal butyrate levels and rumen epithelial keratinization. More importantly, we successfully established a rumen organoid model capable of facilitating the study of the keratinization process in the rumen epithelia and further confirmed that high dose butyrate indeed contributed to rumen epithelial parakeratosis. CONCLUSION Collectively, concentrate starter introduction induces ruminal epithelial parakeratosis by blocking keratinocyte differentiation with excessive ruminal butyrate accumulation in a neonatal lamb model. These findings enhance our understanding of rumen epithelial keratinization and provide valuable insights for addressing rumen epithelial parakeratosis using early nutritional intervention strategies.
Collapse
Affiliation(s)
- Kai Zhang
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yali Zhang
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jing Qin
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Haining Zhu
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ning Liu
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Daming Sun
- Laboratory of Metabolism and Drug Target Discovery, State Key Laboratory of Natural Medicines, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yuyang Yin
- Huzhou Academy of Agricultural Sciences, Huzhou 313000, China
| | - Shengyong Mao
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weiyun Zhu
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zan Huang
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Junhua Liu
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
55
|
Yang S, Sun M, Chen L, Zhang H, Sun L, Liu E, Tian X, Hou X, Lin Y, Lu M. WNT inhibitory factor 1 (WIF1) is a novel fusion partner of RUNX family transcription factor 1 (RUNX1) in acute myeloid leukemia with t(12;21)(q14;q22). J Hematop 2024; 17:245-249. [PMID: 39066949 DOI: 10.1007/s12308-024-00597-4] [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: 11/14/2023] [Accepted: 07/14/2024] [Indexed: 07/30/2024] Open
Abstract
As a member of the core transcription factor family, RUNX1 plays an important role in stem cell differentiation. RUNX1 rearrangements are common in myeloid and lymphoid tumors [1]. (Blood 129(15):2070-2082, 2017). One of the most commonly detected abnormalities in acute myeloid leukemia (AML) is the translocation t(8;21)(q22;q22) (Blood Adv 4(1):229-238, 2020), resulting in a RUNX1::RUNX1T1 fusion. Occasionally, RUNX1 is translocated with other genes. This article describes an AML patient with a specific chromosomal translocation involving the RUNX1 gene and the identification of the RUNX1::WIF1 fusion. Chromosomal abnormalities were detected through karyotype analysis, break gene involved was identified via fluorescence in situ hybridization (FISH), and the novel fusion was identified through transcriptome sequencing and subsequently confirmed through reverse transcription-polymerase chain reaction (RT-PCR) and Sanger sequencing. A 79-year-old female patient diagnosed with AML was found to have a t(12;21)(q14;q12) translocation. FISH analysis provided evidence of RUNX1 gene rearrangement. Additionally, transcriptomic sequencing revealed a novel fusion known as RUNX1::WIF1, which consists of RUNX1 exon 2 and WIF1 exon 3. The novel fusion was further confirmed through RT-PCR and Sanger sequencing. We identified WIF1 as a novel fusion partner of RUNX1 in AML. Additionally, this is the first report of a RUNX1 fusion gene with the break point in intron 2, resulting in an out-of-frame fusion. Further research is needed to investigate the impact of this novel fusion on the establishment and progression of the disease.
Collapse
Affiliation(s)
- Shaobin Yang
- Sino-US Diagnostics Lab, Tianjin Enterprise Key Laboratory of Al-Aided Hematopathology Diagnosis, Tianjin, China
| | - Ming Sun
- Department of Hematology, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Long Chen
- Sino-US Diagnostics Lab, Tianjin Enterprise Key Laboratory of Al-Aided Hematopathology Diagnosis, Tianjin, China
| | - Hong Zhang
- Sino-US Diagnostics Lab, Tianjin Enterprise Key Laboratory of Al-Aided Hematopathology Diagnosis, Tianjin, China
| | - Lidan Sun
- Sino-US Diagnostics Lab, Tianjin Enterprise Key Laboratory of Al-Aided Hematopathology Diagnosis, Tianjin, China
| | - Enbin Liu
- Sino-US Diagnostics Lab, Tianjin Enterprise Key Laboratory of Al-Aided Hematopathology Diagnosis, Tianjin, China
| | - Xin Tian
- Sino-US Diagnostics Lab, Tianjin Enterprise Key Laboratory of Al-Aided Hematopathology Diagnosis, Tianjin, China
| | - Xiaoju Hou
- Sino-US Diagnostics Lab, Tianjin Enterprise Key Laboratory of Al-Aided Hematopathology Diagnosis, Tianjin, China
| | - Yani Lin
- Sino-US Diagnostics Lab, Tianjin Enterprise Key Laboratory of Al-Aided Hematopathology Diagnosis, Tianjin, China
| | - Mize Lu
- Department of Hematology, Affiliated Wuxi People's Hospital, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, China.
| |
Collapse
|
56
|
Nadhan R, Isidoro C, Song YS, Dhanasekaran DN. LncRNAs and the cancer epigenome: Mechanisms and therapeutic potential. Cancer Lett 2024; 605:217297. [PMID: 39424260 DOI: 10.1016/j.canlet.2024.217297] [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: 08/23/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as critical regulators of epigenome, modulating gene expression through DNA methylation, histone modification, and/or chromosome remodeling. Dysregulated lncRNAs act as oncogenes or tumor suppressors, driving tumor progression by shaping the cancer epigenome. By interacting with the writers, readers, and erasers of the epigenetic script, lncRNAs induce epigenetic modifications that bring about changes in cancer cell proliferation, apoptosis, epithelial-mesenchymal transition, migration, invasion, metastasis, cancer stemness and chemoresistance. This review analyzes and discusses the multifaceted role of lncRNAs in cancer pathobiology, from cancer genesis and progression through metastasis and therapy resistance. It also explores the therapeutic potential of targeting lncRNAs through innovative diagnostic, prognostic, and therapeutic strategies. Understanding the dynamic interplay between lncRNAs and epigenome is crucial for developing personalized therapeutic strategies, offering new avenues for precision cancer medicine.
Collapse
Affiliation(s)
- Revathy Nadhan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Ciro Isidoro
- Laboratory of Molecular Pathology and NanoBioImaging, Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Cancer Research Institute, College of Medicine, Seoul National University, Seoul, 151-921, South Korea.
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
57
|
Carroll SH, Schafer S, Kawasaki K, Tsimbal C, Jule AM, Hallett SA, Li E, Liao EC. Genetic requirement of dact1/2 to regulate noncanonical Wnt signaling and calpain 8 during embryonic convergent extension and craniofacial morphogenesis. eLife 2024; 13:RP91648. [PMID: 39570288 PMCID: PMC11581427 DOI: 10.7554/elife.91648] [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] [Indexed: 11/22/2024] Open
Abstract
Wnt signaling plays crucial roles in embryonic patterning including the regulation of convergent extension (CE) during gastrulation, the establishment of the dorsal axis, and later, craniofacial morphogenesis. Further, Wnt signaling is a crucial regulator of craniofacial morphogenesis. The adapter proteins Dact1 and Dact2 modulate the Wnt signaling pathway through binding to Disheveled. However, the distinct relative functions of Dact1 and Dact2 during embryogenesis remain unclear. We found that dact1 and dact2 genes have dynamic spatiotemporal expression domains that are reciprocal to one another suggesting distinct functions during zebrafish embryogenesis. Both dact1 and dact2 contribute to axis extension, with compound mutants exhibiting a similar CE defect and craniofacial phenotype to the wnt11f2 mutant. Utilizing single-cell RNAseq and an established noncanonical Wnt pathway mutant with a shortened axis (gpc4), we identified dact1/2-specific roles during early development. Comparative whole transcriptome analysis between wildtype and gpc4 and wildtype and dact1/2 compound mutants revealed a novel role for dact1/2 in regulating the mRNA expression of the classical calpain capn8. Overexpression of capn8 phenocopies dact1/2 craniofacial dysmorphology. These results identify a previously unappreciated role of capn8 and calcium-dependent proteolysis during embryogenesis. Taken together, our findings highlight the distinct and overlapping roles of dact1 and dact2 in embryonic craniofacial development, providing new insights into the multifaceted regulation of Wnt signaling.
Collapse
Affiliation(s)
- Shannon H Carroll
- Center for Craniofacial Innovation, Children’s Hospital of Philadelphia Research, Institute, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Shriners Hospital for ChildrenTampaUnited States
| | - Sogand Schafer
- Center for Craniofacial Innovation, Children’s Hospital of Philadelphia Research, Institute, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Kenta Kawasaki
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Shriners Hospital for ChildrenTampaUnited States
| | - Casey Tsimbal
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Shriners Hospital for ChildrenTampaUnited States
| | - Amelie M Jule
- Department of Biostatistics, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - Shawn A Hallett
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Shriners Hospital for ChildrenTampaUnited States
| | - Edward Li
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Eric C Liao
- Center for Craniofacial Innovation, Children’s Hospital of Philadelphia Research, Institute, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Shriners Hospital for ChildrenTampaUnited States
| |
Collapse
|
58
|
Adler M, Medzhitov R. Recurrent hyper-motif circuits in developmental programs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624466. [PMID: 39605580 PMCID: PMC11601646 DOI: 10.1101/2024.11.20.624466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
During embryogenesis, homogenous groups of cells self-organize into stereotypic spatial and temporal patterns that make up tissues and organs. These emergent patterns are controlled by transcription factors and secreted signals that regulate cellular fate and behaviors through intracellular regulatory circuits and cell-cell communication circuits. However, the principles of these circuits and how their properties are combined to provide the spatio-temporal properties of tissues remain unclear. Here we develop a framework to explore building-block circuits of developmental programs. We use single-cell gene expression data across developmental stages of the human intestine to infer the key intra- and inter-cellular circuits that control developmental programs. We study how these circuits are joined into higher-level hyper-motif circuits and explore their emergent dynamical properties. This framework uncovers design principles of developmental programs and reveals the rules that allow cells to develop robust and diverse patterns.
Collapse
Affiliation(s)
- Miri Adler
- Department of Genetics, Silberman Institute of Life Science, Edmond J. Safra Campus, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
- Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
59
|
Wu Y, Wang Y, Dong Y, Sun LV, Zheng Y. Lactate promotes H3K18 lactylation in human neuroectoderm differentiation. Cell Mol Life Sci 2024; 81:459. [PMID: 39562370 DOI: 10.1007/s00018-024-05510-x] [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: 06/21/2024] [Revised: 10/01/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
In mammals, early embryonic gastrulation process is high energy demanding. Previous studies showed that, unlike endoderm and mesoderm cells, neuroectoderm differentiated from human embryonic stem cells relied on aerobic glycolysis as the major energy metabolic process, which generates lactate as the final product. Here we explored the function of intracellular lactate during neuroectoderm differentiation. Our results revealed that the intracellular lactate level was elevated in neuroectoderm and exogenous lactate could further promote hESCs differentiation towards neuroectoderm. Changing intracellular lactate levels by sodium lactate or LDHA inhibitors had no obvious effect on BMP or WNT/β-catenin signaling during neuroectoderm differentiation. Notably, histone lactylation, especially H3K18 lactylation was significant upregulated during this process. We further performed CUT&Tag experiments and the results showed that H3K18la is highly enriched at gene promoter regions. By analyzing data from CUT&Tag and RNA-seq experiments, we further identified that four genes, including PAX6, were transcriptionally upregulated by lactate during neuroectoderm differentiation. A H3K18la modification site at PAX6 promoter was verified and exogenous lactate could also rescue the level of PAX6 after shPAX6 inhibition.
Collapse
Affiliation(s)
- Yu Wu
- Obstetrics and Gynecology Hospital, Institute of Developmental Biology & Molecular Medicine, Department of Cellular & Developmental Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Yumeng Wang
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuhao Dong
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 21 Lower Kent Ridge Rd, Singapore, 119077, Singapore
| | - Ling V Sun
- Obstetrics and Gynecology Hospital, Institute of Developmental Biology & Molecular Medicine, Department of Cellular & Developmental Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Yufang Zheng
- Obstetrics and Gynecology Hospital, Institute of Developmental Biology & Molecular Medicine, Department of Cellular & Developmental Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
60
|
Parodi J, Mira RG, Fuenzalida M, Cerpa W, Serrano FG, Tapia-Rojas C, Martinez-Torres A, Inestrosa NC. Wnt-5a Signaling Mediates Metaplasticity at Hippocampal CA3-CA1 Synapses in Mice. Cell Mol Neurobiol 2024; 44:76. [PMID: 39535658 PMCID: PMC11561030 DOI: 10.1007/s10571-024-01512-2] [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: 08/22/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Wnt signaling plays a role in synaptic plasticity, but the specific cellular events and molecular components involved in Wnt signaling-mediated synaptic plasticity are not well defined. Here, we report a change in the threshold required to induce synaptic plasticity that facilitates the induction of long-term potentiation (LTP) and inhibits the induction of long-term depression (LTD) during brief exposure to the noncanonical ligand Wnt-5a. Both effects are related to the metaplastic switch of hippocampal CA3-CA1 synaptic transmission, a complex mechanism underlying the regulation of the threshold required to induce synaptic plasticity and of synaptic efficacy. We observed an early increase in the amplitude of field excitatory postsynaptic potentials (fEPSPs) that persisted over time, including after washout. The first phase involves an increase in the fEPSP amplitude that is required to trigger a spontaneous second phase that depends on Jun N-terminal kinase (JNK) and N-methyl D-aspartate receptor (NMDAR) activity. These changes are prevented by treatment with secreted frizzled-related protein 2 (sFRP-2), an endogenous antagonist of Wnt ligands. Here, we demonstrate the contribution of Wnt-5a signaling to a process associated with metaplasticity at CA3-CA1 synapses that favors LTP over LTD.
Collapse
Affiliation(s)
- Jorge Parodi
- Departamento de Análisis de Datos, Facultad de Ciencias Sociales, Universidad Autónoma de Chile, Temuco, Chile
| | - Rodrigo G Mira
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Millenium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Waldo Cerpa
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe G Serrano
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cheril Tapia-Rojas
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Ataulfo Martinez-Torres
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Nibaldo C Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile.
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
61
|
Schaefer T, Mittal N, Wang H, Ataman M, Candido S, Lötscher J, Velychko S, Tintignac L, Bock T, Börsch A, Baßler J, Rao TN, Zmajkovic J, Roffeis S, Löliger J, Jacob F, Dumlin A, Schürch C, Schmidt A, Skoda RC, Wymann MP, Hess C, Schöler HR, Zaehres H, Hurt E, Zavolan M, Lengerke C. Nuclear and cytosolic fractions of SOX2 synergize as transcriptional and translational co-regulators of cell fate. Cell Rep 2024; 43:114807. [PMID: 39368083 DOI: 10.1016/j.celrep.2024.114807] [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: 07/14/2023] [Revised: 01/28/2024] [Accepted: 09/13/2024] [Indexed: 10/07/2024] Open
Abstract
Stemness and pluripotency are mediated by transcriptional master regulators that promote self-renewal and repress cell differentiation, among which is the high-mobility group (HMG) box transcription factor SOX2. Dysregulated SOX2 expression, by contrast, leads to transcriptional aberrations relevant to oncogenic transformation, cancer progression, metastasis, therapy resistance, and relapse. Here, we report a post-transcriptional mechanism by which the cytosolic pool of SOX2 contributes to these events in an unsuspected manner. Specifically, a low-complexity region within SOX2's C-terminal segment connects to the ribosome to modulate the expression of cognate downstream factors. Independent of nuclear structures or DNA, this C-terminal functionality alone changes metabolic properties and induces non-adhesive growth when expressed in the cytosol of SOX2 knockout cells. We thus propose a revised model of SOX2 action where nuclear and cytosolic fractions cooperate to impose cell fate decisions via both transcriptional and translational mechanisms.
Collapse
Affiliation(s)
- Thorsten Schaefer
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland.
| | | | - Hui Wang
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Meric Ataman
- Biozentrum, University of Basel, Basel, Switzerland
| | - Silvia Candido
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Jonas Lötscher
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Sergiy Velychko
- Max Planck Institute for Molecular Biomedicine, Münster, Germany; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Lionel Tintignac
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Thomas Bock
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Anastasiya Börsch
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Jochen Baßler
- Biochemistry Center Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Tata Nageswara Rao
- Medical Research Center, Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland; Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Jakub Zmajkovic
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Sarah Roffeis
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Jordan Löliger
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Francis Jacob
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Alain Dumlin
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Christoph Schürch
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Radek C Skoda
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Matthias P Wymann
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Christoph Hess
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; CITIID, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Hans R Schöler
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Holm Zaehres
- Max Planck Institute for Molecular Biomedicine, Münster, Germany; Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Ed Hurt
- Biochemistry Center Heidelberg, Heidelberg University, Heidelberg, Germany
| | | | - Claudia Lengerke
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
62
|
Hasan S, Mahmud Z, Hossain M, Islam S. Harnessing the role of aberrant cell signaling pathways in glioblastoma multiforme: a prospect towards the targeted therapy. Mol Biol Rep 2024; 51:1069. [PMID: 39424705 DOI: 10.1007/s11033-024-09996-3] [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: 06/19/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
Glioblastoma Multiforme (GBM), designated as grade IV by the World Health Organization, is the most aggressive and challenging brain tumor within the central nervous system. Around 80% of GBM patients have a poor prognosis, with a median survival of 12-15 months. Approximately 90% of GBM cases originate from normal glial cells via oncogenic processes, while the remainder arise from low-grade tumors. GBM is notorious for its heterogeneity, high recurrence rates, invasiveness, and aggressive behavior. Its malignancy is driven by increased invasive migration, proliferation, angiogenesis, and reduced apoptosis. Throughout various stages of central nervous system (CNS) development, pivotal signaling pathways, including Wnt/β-catenin, Sonic hedgehog signaling (Shh), PI3K/AKT/mTOR, Ras/Raf/MAPK/ERK, STAT3, NF-КB, TGF-β, and Notch signaling, orchestrate the growth, proliferation, differentiation, and migration of neural progenitor cells in the brain. Numerous upstream and downstream regulators within these signaling pathways have been identified as significant contributors to the development of human malignancies. Disruptions or aberrant activations in these pathways are linked to gliomagenesis, enhancing the invasiveness, progression, and aggressiveness of GBM, along with epithelial to mesenchymal transition (EMT) and the presence of glioma stem cells (GSCs). Traditional GBM treatment involves surgery, radiotherapy, and chemotherapy with Temozolomide (TMZ). However, most patients experience tumor recurrence, leading to low survival rates. This review provides an overview of the major cell signaling pathways involved in gliomagenesis. Furthermore, we explore the signaling pathways leading to therapy resistance and target key molecules within these signaling pathways, paving the way for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Subbrina Hasan
- Laboratory of Neuroscience and Neurogenetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Mahmud Hossain
- Laboratory of Neuroscience and Neurogenetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Sohidul Islam
- Department of Biochemistry & Microbiology, North South University, Dhaka, 1229, Bangladesh
| |
Collapse
|
63
|
Luo D, Zheng J, Lv S, Sheng R, Chen M, He X, Zhang X. Wnt specifically induces FZD5/8 endocytosis and degradation and the involvement of RSPO-ZNRF3/RNF43 and DVL. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619000. [PMID: 39463927 PMCID: PMC11507892 DOI: 10.1101/2024.10.18.619000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Frizzled (FZD) proteins are the principal receptors of the Wnt signaling pathway. However, whether Wnt ligands induce FZD endocytosis and degradation remains elusive. The transmembrane E3 ubiquitin ligases ZNRF3 and RNF43 promote the endocytosis and degradation of FZD receptors to inhibit Wnt signaling, and their function is antagonized by R-spondin (RSPO) proteins. However, the dependency of RSPO-ZNRF3/RNF43-mediated FZD endocytosis and degradation on Wnt stimulation, as well as the specificity of this degradation for different FZD, remains unclear. Here, we demonstrated that Wnt specifically induces FZD5/8 endocytosis and degradation in a ZNRF3/RNF43-dependent manner. ZNRF3/RNF43 selectively targets FZD5/8 for degradation upon Wnt stimulation. RSPO1 enhances Wnt signaling by specifically stabilizing FZD5/8. Wnt promotes the interaction between FZD5 and RNF43. We further demonstrated that DVL proteins promote ligand-independent endocytosis of FZD but are dispensable for Wnt-induced FZD5/8 endocytosis and degradation. Our results reveal a novel negative regulatory mechanism of Wnt signaling at the receptor level and illuminate the mechanism by which RSPO-ZNRF3/RNF43 regulates Wnt signaling, which may provide new insights into regenerative medicine and cancer therapy.
Collapse
Affiliation(s)
- Dong Luo
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China; The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- These authors contributed equally to this work
| | - Jing Zheng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China; The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- These authors contributed equally to this work
| | - Shuning Lv
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China; The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ren Sheng
- College of Life and Health Science, Northeastern University, Shenyang, China
| | - Maorong Chen
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Xi He
- The F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Xinjun Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China; The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
64
|
Chang X, Li W, Matsui S, Huynh C, Cederquist GY, Studer L, Iwafuchi M, Shillington A, Chronis C, Tchieu J. ZMYND11 Functions in Bimodal Regulation of Latent Genes and Brain-like Splicing to Safeguard Corticogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618524. [PMID: 39464123 PMCID: PMC11507784 DOI: 10.1101/2024.10.15.618524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Despite the litany of pathogenic variants linked to neurodevelopmental disorders (NDD) including autism (ASD) and intellectual disability 1,2 , our understanding of the underlying mechanisms caused by risk genes remain unclear. Here, we leveraged a human pluripotent stem cell model to uncover the neurodevelopmental consequences of mutations in ZMYND11 , a newly implicated risk gene 3,4 . ZMYND11, known for its tumor suppressor function, encodes a histone-reader that recognizes sites of transcriptional elongation and acts as a co-repressor 5,6 . Our findings reveal that ZMYND11-deficient cortical neural stem cells showed upregulation of latent developmental pathways, impairing progenitor and neuron production. In addition to its role on histones, ZMYND11 controls a brain-specific isoform switch involving the splicing regulator RBFOX2. Extending our findings to other chromatin-related ASD risk factors revealed similar developmental pathway activation and splicing dysregulation, partially rescuable through ZMYND11's regulatory functions.
Collapse
|
65
|
Schulte G. International Union of Basic and Clinical Pharmacology CXV: The Class F of G Protein-Coupled Receptors. Pharmacol Rev 2024; 76:1009-1037. [PMID: 38955509 DOI: 10.1124/pharmrev.124.001062] [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: 01/03/2024] [Revised: 04/10/2024] [Accepted: 05/17/2024] [Indexed: 07/04/2024] Open
Abstract
The class F of G protein-coupled receptors (GPCRs) consists of 10 Frizzleds (FZD1-10) and Smoothened (SMO). FZDs bind and are activated by secreted lipoglycoproteins of the Wingless/Int-1 (WNT) family, and SMO is indirectly activated by the Hedgehog (Hh) family of morphogens acting on the transmembrane protein Patched. The advance of our understanding of FZDs and SMO as dynamic transmembrane receptors and molecular machines, which emerged during the past 14 years since the first-class F GPCR IUPHAR nomenclature report, justifies an update. This article focuses on the advances in molecular pharmacology and structural biology providing new mechanistic insight into ligand recognition, receptor activation mechanisms, signal initiation, and signal specification. Furthermore, class F GPCRs continue to develop as drug targets, and novel technologies and tools such as genetically encoded biosensors and CRISP/Cas9 edited cell systems have contributed to refined functional analysis of these receptors. Also, advances in crystal structure analysis and cryogenic electron microscopy contribute to the rapid development of our knowledge about structure-function relationships, providing a great starting point for drug development. Despite the progress, questions and challenges remain to fully understand the complexity of the WNT/FZD and Hh/SMO signaling systems. SIGNIFICANCE STATEMENT: The recent years of research have brought about substantial functional and structural insight into mechanisms of activation of Frizzleds and Smoothened. While the advance furthers our mechanistic understanding of ligand recognition, receptor activation, signal specification, and initiation, broader opportunities emerge that allow targeting class F GPCRs for therapy and regenerative medicine employing both biologics and small molecule compounds.
Collapse
Affiliation(s)
- Gunnar Schulte
- Karolinska Institutet, Department of Physiology & Pharmacology, Receptor Biology & Signaling, Biomedicum, Stockholm, Sweden
| |
Collapse
|
66
|
Takahashi M, Isagawa T, Sato T, Takeda N, Kawakami K. Lineage tracing using Wnt2b-2A-CreERT2 knock-in mice reveals the contributions of Wnt2b-expressing cells to novel subpopulations of mesothelial/epicardial cell lineages during mouse development. Genes Cells 2024; 29:854-875. [PMID: 39109760 DOI: 10.1111/gtc.13147] [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: 06/08/2023] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 10/04/2024]
Abstract
Mesothelial and epicardial cells give rise to various types of mesenchymal cells via epithelial (mesothelial)-to-mesenchymal transition during development. However, the genes controlling the differentiation and diversification of mesothelial/epicardial cells remain unclear. Here, we examined Wnt2b expression in the embryonic mesothelium and epicardium and performed lineage tracing of Wnt2b-expressing cells by using novel Wnt2b-2A-CreERT2 knock-in and LacZ-reporter mice. Wnt2b was expressed in mesothelial cells covering visceral organs, but the expression was restricted in their subpopulations. Wnt2b-expressing cells labeled at embryonic day (E) 10.5 were distributed to the mesothelium and mesenchyme in the lungs, abdominal wall, stomach, and spleen in Wnt2b2A-CreERT2/+;R26RLacZ/+ mice at E13.0. Wnt2b was initially expressed in the proepicardial organ (PEO) at E9.5 and then in the epicardium after E10.0. Wnt2b-expressing PEO cells labeled at E9.5 differentiated into a small fraction of cardiac fibroblasts and preferentially localized at the left side of the postnatal heart. LacZ+ epicardium-derived cells labeled at E10.5 differentiated into a small fraction of fibroblasts and smooth muscle cells in the postnatal heart. Taken together, our results reveal novel subpopulations of PEO and mesothelial/epicardial cells that are distinguishable by Wnt2b expression and elucidate the unique contribution of Wnt2b-expressing PEO and epicardial cells to the postnatal heart.
Collapse
Affiliation(s)
- Masanori Takahashi
- Department of Anatomy, Division of Bioimaging and Neuro-cell Science, Jichi Medical University, Shimotsuke, Japan
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Takayuki Isagawa
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
- Data Science Center, Jichi Medical University, Shimotsuke, Japan
| | - Tatsuyuki Sato
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Norihiko Takeda
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | | |
Collapse
|
67
|
Wang R, Wu NS, Wang L, Zhang ZZ, Wang CF, Wang Y, Liang Y, Zhang Y, Qi XW. A pan-cancer analysis of Wnt family member 7B in human cancers. CANCER INNOVATION 2024; 3:e139. [PMID: 39257440 PMCID: PMC11386237 DOI: 10.1002/cai2.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/27/2024] [Accepted: 04/29/2024] [Indexed: 09/12/2024]
Abstract
Background Previous studies have highlighted the crucial role of Wnt7B in the development of various cancers, including breast, pancreatic, and gastric cancers. However, research into the involvement of Wnt7B is often confined to specific tumor types, with a noticeable lack of comprehensive studies spanning multiple cancer forms. The potential of Wnt7B as a diagnostic or prognostic cancer biomarker has not been fully explored. Methods In this study, we combined bioinformatics and immunohistochemistry analyses to examine the expression patterns and functions of Wnt7B in cancerous and adjacent noncancerous tissues across a range of tumors. Results Our data indicate that Wnt7B may serve as a novel prognostic biomarker and therapeutic target in certain cancers. Conclusion We found significant upregulation of Wnt7B expression levels in the majority of cancer cases examined. Furthermore, Wnt7B can influence cancer prognosis by modulating the tumor microenvironment, immune cell infiltration, and tumor stemness, among other factors. Additionally, we examined the associations between anticancer drug sensitivity and Wnt7B expression, which could aid in the development of more precise clinical therapies.
Collapse
Affiliation(s)
- Rui Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital Army Medical University Chongqing China
| | - Ni-Sha Wu
- Department of Breast and Thyroid Surgery, Southwest Hospital Army Medical University Chongqing China
| | - Li Wang
- Department of Infection China Academy of Chinese Medical Sciences, Guang'anmen Hospital Beijing China
| | - Zhi-Zhao Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital Army Medical University Chongqing China
| | - Cheng-Fang Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital Army Medical University Chongqing China
| | - Yan Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital Army Medical University Chongqing China
| | - Yan Liang
- Department of Breast and Thyroid Surgery, Southwest Hospital Army Medical University Chongqing China
| | - Yi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital Army Medical University Chongqing China
| | - Xiao-Wei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital Army Medical University Chongqing China
| |
Collapse
|
68
|
Mehner LM, Munoz-Sagredo L, Sonnentag SJ, Treffert SM, Orian-Rousseau V. Targeting CD44 and other pleiotropic co-receptors as a means for broad inhibition of tumor growth and metastasis. Clin Exp Metastasis 2024; 41:599-611. [PMID: 38761292 PMCID: PMC11499327 DOI: 10.1007/s10585-024-10292-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 05/02/2024] [Indexed: 05/20/2024]
Abstract
Although progress has been made in the treatment of cancer, particularly for the four major types of cancers affecting the lungs, colon, breast and prostate, resistance to cancer treatment often emerges upon inhibition of major signaling pathways, which leads to the activation of additional pathways as a last-resort survival mechanism by the cancer cells. This signaling plasticity provides cancer cells with a level of operational freedom, reducing treatment efficacy. Plasticity is a characteristic of cancer cells that are not only able to switch signaling pathways but also from one cellular state (differentiated cells to stem cells or vice versa) to another. It seems implausible that the inhibition of one or a few signaling pathways of heterogeneous and plastic tumors can sustain a durable effect. We propose that inhibiting molecules with pleiotropic functions such as cell surface co-receptors can be a key to preventing therapy escape instead of targeting bona fide receptors. Therefore, we ask the question whether co-receptors often considered as "accessory molecules" are an overlooked key to control cancer cell behavior.
Collapse
Affiliation(s)
- Lisa-Marie Mehner
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Leonel Munoz-Sagredo
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
- School of Medicine, Universidad de Valparaiso, Valparaiso, Chile
| | - Steffen Joachim Sonnentag
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Sven Máté Treffert
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Véronique Orian-Rousseau
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany.
| |
Collapse
|
69
|
Ajabnoor R. Different Shades of Desmoid-Type Fibromatosis (DTF): Detection of Noval Mutations in the Clinicopathologic Analysis of 32 Cases. Diagnostics (Basel) 2024; 14:2161. [PMID: 39410565 PMCID: PMC11476057 DOI: 10.3390/diagnostics14192161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/30/2024] [Accepted: 09/13/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Desmoid-type fibromatosis (DTF) is a locally aggressive myofibroblastic/fibroblastic neoplasm with a high risk of local recurrence. It has a variety of histologic features that might confuse diagnosis, especially when detected during core needle biopsy. The Wnt/β-catenin pathway is strongly linked to the pathogenesis of DT fibromatosis. METHOD This study examined 33 desmoid-type fibromatoses (DTFs) from 32 patients, analyzing its clinical characteristics, histologic patterns, occurrence rates, relationship with clinical outcomes, immunohistochemical and molecular findings. RESULTS The DTFs exhibit a range of 1 to 7 histologic patterns per tumor, including conventional, hypercellular, myxoid, hyalinized/hypocellular, staghorn/hemangiopericytomatous blood vessels pattern, nodular fasciitis-like, and keloid-like morphology. No substantial association was found between the existence of different histologic patterns and the clinical outcome. All thirty-three (100%) samples of DTF had a variable percentage of cells that were nuclear positive for β-catenin. An NGS analysis detected novel non-CTNNB1 mutations in two DTFs, including BCL10, MPL, and RBM10 gene mutations. CONCLUSIONS This study reveals a diverse morphology of DTFs that could result in misdiagnosis. Therefore, surgical pathologists must comprehend this thoroughly. Also, the importance of the newly identified non-CTNNB1 gene mutations is still unclear. More research and analyses are needed to completely grasp the clinical implications of these mutations.
Collapse
Affiliation(s)
- Rana Ajabnoor
- Department of Pathology, Faculty of Medicine, King Abdulaziz University and King Abdulaziz University Hospital, Jeddah 22252, Saudi Arabia
| |
Collapse
|
70
|
Grätz L, Voss JH, Schulte G. Class-Wide Analysis of Frizzled-Dishevelled Interactions Using BRET Biosensors Reveals Functional Differences among Receptor Paralogs. ACS Sens 2024; 9:4626-4636. [PMID: 39213612 PMCID: PMC11443525 DOI: 10.1021/acssensors.4c00806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Wingless/Int-1 (WNT) signaling is mediated by WNT binding to 10 Frizzleds (FZD1-10), which propagate the signal inside the cell by interacting with different transducers, most prominently the phosphoprotein Dishevelled (DVL). Despite recent progress, questions about WNT/FZD selectivity and paralog-dependent differences in the FZD/DVL interaction remain unanswered. Here, we present a class-wide analysis of the FZD/DVL interaction using the DEP domain of DVL as a proxy in bioluminescence resonance energy transfer (BRET) techniques. Most FZDs engage in a constitutive high-affinity interaction with DEP. Stimulation of unimolecular FZD/DEP BRET sensors with different ligands revealed that most paralogs are dynamic in the FZD/DEP interface, showing distinct profiles in terms of ligand selectivity and signal kinetics. This study underlines mechanistic differences in terms of how allosteric communication between FZDs and their main signal transducer DVL occurs. Moreover, the unimolecular sensors represent the first receptor-focused biosensors to surpass the requirements for high-throughput screening, facilitating FZD-targeted drug discovery.
Collapse
Affiliation(s)
- Lukas Grätz
- Department of Physiology & Pharmacology, Section of Receptor Biology & Signaling, Biomedicum, Karolinska Institutet, S-17165 Stockholm, Sweden
| | - Jan H Voss
- Department of Physiology & Pharmacology, Section of Receptor Biology & Signaling, Biomedicum, Karolinska Institutet, S-17165 Stockholm, Sweden
| | - Gunnar Schulte
- Department of Physiology & Pharmacology, Section of Receptor Biology & Signaling, Biomedicum, Karolinska Institutet, S-17165 Stockholm, Sweden
| |
Collapse
|
71
|
Wu Y, Lyu Z, Hu F, Yang L, Yang K, Chen M, Wang Y. A chondroitin sulphate hydrogel with sustained release of SDF-1α for extensive cartilage defect repair through induction of cell homing and promotion of chondrogenesis. J Mater Chem B 2024; 12:8672-8687. [PMID: 39115288 DOI: 10.1039/d4tb00624k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Articular cartilage damage represents a prevalent clinical disease in orthopedics, with its regeneration and repair constituting a central focus in ongoing research endeavors. While hydrogel technology has achieved notable progress in the field of cartilage regeneration, addressing the repair of larger cartilage defects remains a significant and formidable challenge. In pursuit of achieving the repair of extensive cartilage defects, this study designed a polydopamine-modified chondroitin sulfate hydrogel loaded with SDF-1α (P-SCMA). This hydrogel, capable of directly providing glycosaminoglycans (GAGs), served as a platform for carrying growth factors and attracting mesenchymal stem cells for the in situ reconstruction of extensive cartilage defects. The results indicate that the P-SCMA hydrogel is capable of not only directly providing GAGs but also sustainably releasing SDF-1α. In the early stages, it promotes cell adhesion and proliferation and induces cell homing, while in the later stages, it further induces chondrogenesis by inhibiting the Wnt/β-catenin pathway. This bioactive hydrogel, which possesses the functions of providing GAGs, promoting cell proliferation, inducing cell homing and chondrogenesis, is capable of promoting cartilage repair in multiple ways, providing new perspectives for the repair of extensive cartilage defects.
Collapse
Affiliation(s)
- Yuezhou Wu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Zhuocheng Lyu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Fei Hu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Linjun Yang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Ke Yang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Mo Chen
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - You Wang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| |
Collapse
|
72
|
Boonsawat P, Asadollahi R, Niedrist D, Steindl K, Begemann A, Joset P, Bhoj EJ, Li D, Zackai E, Vetro A, Barba C, Guerrini R, Whalen S, Keren B, Khan A, Jing D, Palomares Bralo M, Rikeros Orozco E, Hao Q, Schlott Kristiansen B, Zheng B, Donnelly D, Clowes V, Zweier M, Papik M, Siegel G, Sabatino V, Mocera M, Horn AHC, Sticht H, Rauch A. Deleterious ZNRF3 germline variants cause neurodevelopmental disorders with mirror brain phenotypes via domain-specific effects on Wnt/β-catenin signaling. Am J Hum Genet 2024; 111:1994-2011. [PMID: 39168120 PMCID: PMC11393693 DOI: 10.1016/j.ajhg.2024.07.016] [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: 04/29/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Zinc and RING finger 3 (ZNRF3) is a negative-feedback regulator of Wnt/β-catenin signaling, which plays an important role in human brain development. Although somatically frequently mutated in cancer, germline variants in ZNRF3 have not been established as causative for neurodevelopmental disorders (NDDs). We identified 12 individuals with ZNRF3 variants and various phenotypes via GeneMatcher/Decipher and evaluated genotype-phenotype correlation. We performed structural modeling and representative deleterious and control variants were assessed using in vitro transcriptional reporter assays with and without Wnt-ligand Wnt3a and/or Wnt-potentiator R-spondin (RSPO). Eight individuals harbored de novo missense variants and presented with NDD. We found missense variants associated with macrocephalic NDD to cluster in the RING ligase domain. Structural modeling predicted disruption of the ubiquitin ligase function likely compromising Wnt receptor turnover. Accordingly, the functional assays showed enhanced Wnt/β-catenin signaling for these variants in a dominant negative manner. Contrarily, an individual with microcephalic NDD harbored a missense variant in the RSPO-binding domain predicted to disrupt binding affinity to RSPO and showed attenuated Wnt/β-catenin signaling in the same assays. Additionally, four individuals harbored de novo truncating or de novo or inherited large in-frame deletion variants with non-NDD phenotypes, including heart, adrenal, or nephrotic problems. In contrast to NDD-associated missense variants, the effects on Wnt/β-catenin signaling were comparable between the truncating variant and the empty vector and between benign variants and the wild type. In summary, we provide evidence for mirror brain size phenotypes caused by distinct pathomechanisms in Wnt/β-catenin signaling through protein domain-specific deleterious ZNRF3 germline missense variants.
Collapse
Affiliation(s)
| | - Reza Asadollahi
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Faculty of Engineering and Science, University of Greenwich London, Medway Campus, Chatham Maritime ME4 4TB, UK
| | - Dunja Niedrist
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Katharina Steindl
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Anaïs Begemann
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Pascal Joset
- Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Elizabeth J Bhoj
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dong Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elaine Zackai
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Annalisa Vetro
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Carmen Barba
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy; University of Florence, Florence, Italy
| | - Renzo Guerrini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Sandra Whalen
- Unité Fonctionnelle de Génétique Odellin, Hôpital Armand Trousseau, Paris, France
| | - Boris Keren
- Département de Génétique, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Amjad Khan
- Faculty of Science, Department of Biological Science (Zoology), University of Lakki Marwat, Khyber Pakhtunkhwa 28420, Pakistan
| | - Duan Jing
- Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - María Palomares Bralo
- Instituto de Genética Médica y Molecular (INGEMM), Unidad de Trastornos Del Neurodesarrollo, Hospital Universitario La Paz, Madrid, Spain
| | - Emi Rikeros Orozco
- Instituto de Genética Médica y Molecular (INGEMM), Unidad de Trastornos Del Neurodesarrollo, Hospital Universitario La Paz, Madrid, Spain
| | - Qin Hao
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | | | - Bixia Zheng
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Deirdre Donnelly
- Northern Ireland Regional Genetics Centre, Belfast Health & Social Care Trust, Belfast, Northern Ireland
| | - Virginia Clowes
- Thames Regional Genetics Service, North West University Healthcare NHS Trust, London, UK
| | - Markus Zweier
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Michael Papik
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Gabriele Siegel
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Valeria Sabatino
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Martina Mocera
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Anselm H C Horn
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Heinrich Sticht
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Pediatric University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
73
|
Shen L, Han F, Pan L, Du L, Sun P, Zhang K, Wu X, Pang K, Zhu J. Construction of tissue engineered cornea with skin-derived corneal endothelial-like cell and mechanism research for the cell differentiation. Front Med (Lausanne) 2024; 11:1448248. [PMID: 39286645 PMCID: PMC11402686 DOI: 10.3389/fmed.2024.1448248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Corneal endothelial transplantation accounts for most of corneal transplantation for treating corneal diseases, however severe shortage of corneal donors is the biggest obstacle. In our previous study, we differentiated human skin-derived precursors (SKPs) into corneal endothelial cell (CEC)-like cells with a co-culture system. In this study, we aimed to investigate cell differentiation molecular mechanism and evaluate the function of CEC-like cells by developing tissue-engineered corneas in order to improve cell production efficiency and provide basic research for clinical transformation. Methods We performed transcriptome sequencing of SKPs and CEC-like cells. Further, we focused on the possible enriching pathways, including PI3K/Akt, MAPK/Erk, WNT/β-catenin, and important transcription factors Pitx2 and Foxc1. The PI3K and β-catenin inhibitors were also added to the culture system to observe the differentiation alteration. We developed a graft for a tissue-engineered cornea (TEC) using CEC-like cells and acellular porcine cornea matrix scaffold. The tissue-engineered corneas were transplanted into rabbits via penetrating keratoplasty. Results The PI3K/Akt, MAPK/Erk, and WNT/β-catenin pathways play important roles during the differentiation of SKPs into CEC-like cells. Crosstalk existed between the PI3K/Akt and MAPK/Erk pathways. The PI3K/Akt and WNT/β-catenin pathways were connected. Pitx2 and Foxc1 were subject to temporal and spatial controls of the WNT/β-catenin pathway. The inhibition of the PI3K/Akt and WNT/β-catenin pathways both prevented cell differentiation. CEC-like cells grew well on the acellular porcine cornea matrix scaffold, and the tissue-engineered corneal graft performed well after transplantation into rabbits. Conclusion We provide experimental basis for CEC-like cell industrial production and drive the cells to be clinically applied in cellular replacement therapy or alternative graft substitution for treating corneal diseases in the future.
Collapse
Affiliation(s)
- Lin Shen
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Fang Han
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Lijie Pan
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Liqun Du
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Peng Sun
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Kai Zhang
- Department of Ophthalmology, Shandong Second Provincial General Hospital, Jinan, China
| | - Xinyi Wu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Kunpeng Pang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Zhu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
74
|
Sanchez-Ruiz JA, Treviño-Alvarez AM, Zambrano-Lucio M, Lozano Díaz ST, Wang N, Biernacka JM, Tye SJ, Cuellar-Barboza AB. The Wnt signaling pathway in major depressive disorder: A systematic review of human studies. Psychiatry Res 2024; 339:115983. [PMID: 38870775 DOI: 10.1016/j.psychres.2024.115983] [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: 03/01/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024]
Abstract
Despite uncertainty about the specific molecular mechanisms driving major depressive disorder (MDD), the Wnt signaling pathway stands out as a potentially influential factor in the pathogenesis of MDD. Known for its role in intercellular communication, cell proliferation, and fate, Wnt signaling has been implicated in diverse biological phenomena associated with MDD, spanning neurodevelopmental to neurodegenerative processes. In this systematic review, we summarize the functional differences in protein and gene expression of the Wnt signaling pathway, and targeted genetic association studies, to provide an integrated synthesis of available human data examining Wnt signaling in MDD. Thirty-three studies evaluating protein expression (n = 15), gene expression (n = 9), or genetic associations (n = 9) were included. Only fifteen demonstrated a consistently low overall risk of bias in selection, comparability, and exposure. We found conflicting observations of limited and distinct Wnt signaling components across diverse tissue sources. These data do not demonstrate involvement of Wnt signaling dysregulation in MDD. Given the well-established role of Wnt signaling in antidepressant response, we propose that a more targeted and functional assessment of Wnt signaling is needed to understand its role in depression pathophysiology. Future studies should include more components, assess multiple tissues concurrently, and follow a standardized approach.
Collapse
Affiliation(s)
- Jorge A Sanchez-Ruiz
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | | | | | - Sofía T Lozano Díaz
- Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Ning Wang
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Joanna M Biernacka
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Susannah J Tye
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia; Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA; Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA
| | - Alfredo B Cuellar-Barboza
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry, Universidad Autónoma de Nuevo León, Monterrey, Mexico.
| |
Collapse
|
75
|
Sun H, Gao Y, Ma X, Deng Y, Bi L, Li L. Mechanism and application of feedback loops formed by mechanotransduction and histone modifications. Genes Dis 2024; 11:101061. [PMID: 39071110 PMCID: PMC11282412 DOI: 10.1016/j.gendis.2023.06.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 03/24/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2024] Open
Abstract
Mechanical stimulation is the key physical factor in cell environment. Mechanotransduction acts as a fundamental regulator of cell behavior, regulating cell proliferation, differentiation, apoptosis, and exhibiting specific signature alterations during the pathological process. As research continues, the role of epigenetic science in mechanotransduction is attracting attention. However, the molecular mechanism of the synergistic effect between mechanotransduction and epigenetics in physiological and pathological processes has not been clarified. We focus on how histone modifications, as important components of epigenetics, are coordinated with multiple signaling pathways to control cell fate and disease progression. Specifically, we propose that histone modifications can form regulatory feedback loops with signaling pathways, that is, histone modifications can not only serve as downstream regulators of signaling pathways for target gene transcription but also provide feedback to regulate signaling pathways. Mechanotransduction and epigenetic changes could be potential markers and therapeutic targets in clinical practice.
Collapse
Affiliation(s)
- Han Sun
- Department of Hematology and Oncology, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Yafang Gao
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Xinyu Ma
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Yizhou Deng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Lintao Bi
- Department of Hematology and Oncology, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
76
|
Xu L, Shi Y, Huang J, Feng L, Wang Y, Sik AG, Chen X, Liu K, Wang R, Jin M. Developmental toxicity assay of xanthatin in zebrafish embryos. Comp Biochem Physiol C Toxicol Pharmacol 2024; 283:109957. [PMID: 38857669 DOI: 10.1016/j.cbpc.2024.109957] [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: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
Xanthatin (XAN), a xanthanolide sesquiterpene lactone, isolated from Chinese herb, Xanthium strumarium L, has various pharmacological activities, such as antitumor activity and anti-inflammatory. However, little is known about its potential toxicity and the mechanism. Here, zebrafish model was used to study the developmental toxicity in vivo. Our results indicated that xanthatin increased the mortality and led to the morphological abnormalities including pericardial edema, yolk sac edema, curved body shape and hatching delay. Furthermore, xanthatin damaged the normal structure and/or function of heart, liver, immune and nervous system. ROS elevation and much more apoptosis cells were observed after xanthatin exposure. Gene expression results showed that oxidative stress-related genes nrf2 was inhibited, while oxidative stress-related genes (keap1 and nqo1) and apoptotic genes (caspase3, caspase9 and p53) were increased after xanthatin exposure. Mitophagy related genes pink1 and parkin, and wnt pathway (β-catenin, wnt8a and wnt11) were significantly increased after xanthatin exposure. Taken together, our finding indicated that xanthatin induced developmental toxicity, and the ROS elevation, apoptosis activation, dysregulation of mitophagy and wnt pathways were involved in the toxicity caused by xanthatin.
Collapse
Affiliation(s)
- Liyan Xu
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Yuxin Shi
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Jing Huang
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Lixin Feng
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Yuxin Wang
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Attila Gabor Sik
- Institute of Physiology, Medical School, University of Pecs, Pecs H-7624, Hungary; Szentagothai Research Centre, University of Pecs, Pecs H-7624, Hungary
| | - Xiqiang Chen
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China.
| | - Meng Jin
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China.
| |
Collapse
|
77
|
Hadjiargyrou M, Kotsiopriftis M, Lauzier D, Hamdy RC, Kloen P. Activation of Wnt signaling in human fracture callus and nonunion tissues. Bone Rep 2024; 22:101780. [PMID: 39005846 PMCID: PMC11245924 DOI: 10.1016/j.bonr.2024.101780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
The Wnt signaling pathway is a key molecular process during fracture repair. Although much of what we now know about the role of this pathway in bone is derived from in vitro and animal studies, the same cannot be said about humans. As such, we hypothesized that Wnt signaling will also be a key process in humans during physiological fracture healing as well as in the development of a nonunion (hypertrophic and oligotrophic). We further hypothesized that the expression of Wnt-signaling pathway genes/proteins would exhibit a differential expression pattern between physiological fracture callus and the pathological nonunion tissues. We tested these two hypotheses by examining the mRNA levels of key Wnt-signaling related genes: ligands (WNT4, WNT10a), receptors (FZD4, LRP5, LRP6), inhibitors (DKK1, SOST) and modulators (CTNNB1 and PORCN). RNA sequencing from calluses as well as from the two nonunion tissue types, revealed that all of these genes were expressed at about the same level in these three tissue types. Further, spatial expression experiments identified the cells responsible of producing these proteins. Robust expression was detected in osteoblasts for the majority of these genes except SOST which displayed low expression, but in contrast, was mostly detected in osteocytes. Many of these genes were also expressed by callus chondrocytes as well. Taken together, these results confirm that Wnt signaling is indeed active during both human physiological fracture healing as well as in pathological nonunions.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Maria Kotsiopriftis
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, Montreal, QC H4A 0A9, Canada
| | - Dominique Lauzier
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, Montreal, QC H4A 0A9, Canada
| | - Reggie C Hamdy
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, Montreal, QC H4A 0A9, Canada
| | - Peter Kloen
- Department of Orthopedic Surgery and Sports Medicine, Amsterdam UMC, location Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, (Tissue Function and Regeneration), Amsterdam, the Netherlands
| |
Collapse
|
78
|
Borges KS, Little DW, Magalhães TDA, Ribeiro C, Dumontet T, Lapensee C, Basham KJ, Seth A, Azova S, Guagliardo NA, Barrett PQ, Berber M, O'Connell AE, Turcu AF, Lerario AM, Mohan DR, Rainey W, Carlone DL, Hirschhorn JN, Salic A, Breault DT, Hammer GD. Non-canonical Wnt signaling triggered by WNT2B drives adrenal aldosterone production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609423. [PMID: 39229119 PMCID: PMC11370552 DOI: 10.1101/2024.08.23.609423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The steroid hormone aldosterone, produced by the zona glomerulosa (zG) of the adrenal gland, is a master regulator of plasma electrolytes and blood pressure. While aldosterone control by the renin-angiotensin system is well understood, other key regulatory factors have remained elusive. Here, we replicated a prior association between a non-coding variant in WNT2B and an increased risk of primary aldosteronism, a prevalent and debilitating disease caused by excessive aldosterone production. We further show that in both mice and humans, WNT2B is expressed in the mesenchymal capsule surrounding the adrenal cortex, in close proximity to the zG. Global loss of Wnt2b in the mouse results in a dysmorphic and hypocellular zG, with impaired aldosterone production. Similarly, humans harboring WNT2B loss-of-function mutations develop a novel form of Familial Hyperreninemic Hypoaldosteronism, designated here as Type 4. Additionally, we demonstrate that WNT2B signals by activating the non-canonical Wnt/planar cell polarity pathway. Our findings identify WNT2B as a key regulator of zG function and aldosterone production with important clinical implications.
Collapse
Affiliation(s)
- Kleiton S Borges
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Donald W Little
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Claudio Ribeiro
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Typhanie Dumontet
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chris Lapensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Aishwarya Seth
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
| | - Svetlana Azova
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Nick A Guagliardo
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0735, USA
| | - Paula Q Barrett
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0735, USA
| | - Mesut Berber
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Amy E O'Connell
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Adina F Turcu
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Antonio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dipika R Mohan
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - William Rainey
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Joel N Hirschhorn
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
79
|
Grass T, Dokuzluoglu Z, Buchner F, Rosignol I, Thomas J, Caldarelli A, Dalinskaya A, Becker J, Rost F, Marass M, Wirth B, Beyer M, Bonaguro L, Rodriguez-Muela N. Isogenic patient-derived organoids reveal early neurodevelopmental defects in spinal muscular atrophy initiation. Cell Rep Med 2024; 5:101659. [PMID: 39067446 PMCID: PMC11384962 DOI: 10.1016/j.xcrm.2024.101659] [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: 07/25/2023] [Revised: 02/26/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024]
Abstract
Whether neurodevelopmental defects underlie postnatal neuronal death in neurodegeneration is an intriguing hypothesis only recently explored. Here, we focus on spinal muscular atrophy (SMA), a neuromuscular disorder caused by reduced survival of motor neuron (SMN) protein levels leading to spinal motor neuron (MN) loss and muscle wasting. Using the first isogenic patient-derived induced pluripotent stem cell (iPSC) model and a spinal cord organoid (SCO) system, we show that SMA SCOs exhibit abnormal morphological development, reduced expression of early neural progenitor markers, and accelerated expression of MN progenitor and MN markers. Longitudinal single-cell RNA sequencing reveals marked defects in neural stem cell specification and fewer MNs, favoring mesodermal progenitors and muscle cells, a bias also seen in early SMA mouse embryos. Surprisingly, SMN2-to-SMN1 conversion does not fully reverse these developmental abnormalities. These suggest that early neurodevelopmental defects may underlie later MN degeneration, indicating that postnatal SMN-increasing interventions might not completely amend SMA pathology in all patients.
Collapse
Affiliation(s)
- Tobias Grass
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany.
| | - Zeynep Dokuzluoglu
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Felix Buchner
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Ines Rosignol
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany; Technische Universität Dresden (TUD), Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Joshua Thomas
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Antonio Caldarelli
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Anna Dalinskaya
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Jutta Becker
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany
| | - Fabian Rost
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering, TUD, Dresden, Germany
| | - Michele Marass
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Center for Rare Diseases, University Hospital of Cologne, Cologne, Germany
| | - Marc Beyer
- Systems Medicine, DZNE, Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE & University of Bonn and West German Genome Center, Bonn, Germany; Immunogenomics & Neurodegeneration, DZNE, Bonn, Germany
| | - Lorenzo Bonaguro
- Systems Medicine, DZNE, Bonn, Germany; Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Natalia Rodriguez-Muela
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany; Technische Universität Dresden (TUD), Center for Regenerative Therapies Dresden, Dresden, Germany; Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
80
|
D’Antonio DL, Fantini F, Moscatello C, Ferrone A, Scaringi S, Valanzano R, Ficari F, Efthymakis K, Neri M, Aceto GM, Curia MC. The Interplay among Wnt/β-catenin Family Members in Colorectal Adenomas and Surrounding Tissues. Biomedicines 2024; 12:1730. [PMID: 39200196 PMCID: PMC11352173 DOI: 10.3390/biomedicines12081730] [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: 06/23/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 09/02/2024] Open
Abstract
BACKGROUND The colorectal adenoma undergoes neoplastic progression via the normal epithelium-adenoma-adenocarcinoma sequence as reported in the Vogelgram. The hazard of developing a tumor is deeply associated with the number and size of adenomas and their subtype. Adenomatous polyps are histologically categorized as follows: approximately 80-90% are tubular, 5-15% are villous, and 5-10% are tubular/villous. Given the higher risk of a malignant transformation observed in tubular/villous adenomas, patients diagnosed with adenomatous polyposis are at an improved risk of developing CRC. The Wnt/β-catenin pathway plays a key role in the onset of colorectal adenoma; in particular, intestinal cells first acquire loss-of-function mutations in the APC gene that induce the formation of adenomas. METHODS Wnt/β-catenin pathway APC, Wnt3a, Wnt5a, LEF1, and BCL9 genes and protein expression analyses were conducted by qRT-PCR and western blot in 68 colonic samples (polyps and adjacent mucosa) from 41 patients, of which 17 were affected by FAP. Ten normal colonic mucosal samples were collected from 10 healthy donors. RESULTS In this study, both the APC gene and protein were less expressed in the colon tumor compared to the adjacent colonic mucosa. Conversely, the activated β-catenin was more expressed in polyps than in the adjacent mucosa. All results confirmed the literature data on carcinomas. A statistically significant correlation between Wnt3a and BCL9 both in polyps and in the adjacent mucosa underlines that the canonical Wnt pathway is activated in early colon carcinogenesis and that the adjacent mucosa is already altered. CONCLUSION This is the first study analyzing the difference in expression of the Wnt/β-catenin pathway in human colorectal adenomas. Understanding the progression from adenomas to colorectal carcinomas is essential for the development of new therapeutic strategies and improving clinical outcomes with the use of APC and β-catenin as biomarkers.
Collapse
Affiliation(s)
- Domenica Lucia D’Antonio
- Department of Medical, Oral and Biotechnological Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.L.D.); (F.F.); (C.M.); (A.F.); (G.M.A.)
- Villa Serena Foundation for Research, Via Leonardo Petruzzi 42, 65013 Città Sant’Angelo, Italy
| | - Fabiana Fantini
- Department of Medical, Oral and Biotechnological Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.L.D.); (F.F.); (C.M.); (A.F.); (G.M.A.)
| | - Carmelo Moscatello
- Department of Medical, Oral and Biotechnological Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.L.D.); (F.F.); (C.M.); (A.F.); (G.M.A.)
| | - Alessio Ferrone
- Department of Medical, Oral and Biotechnological Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.L.D.); (F.F.); (C.M.); (A.F.); (G.M.A.)
| | - Stefano Scaringi
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134 Firenze, Italy; (S.S.); (R.V.); (F.F.)
| | - Rosa Valanzano
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134 Firenze, Italy; (S.S.); (R.V.); (F.F.)
| | - Ferdinando Ficari
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134 Firenze, Italy; (S.S.); (R.V.); (F.F.)
| | - Konstantinos Efthymakis
- Department of Medicine and Aging Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (K.E.); (M.N.)
| | - Matteo Neri
- Department of Medicine and Aging Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (K.E.); (M.N.)
| | - Gitana Maria Aceto
- Department of Medical, Oral and Biotechnological Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.L.D.); (F.F.); (C.M.); (A.F.); (G.M.A.)
| | - Maria Cristina Curia
- Department of Medical, Oral and Biotechnological Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.L.D.); (F.F.); (C.M.); (A.F.); (G.M.A.)
| |
Collapse
|
81
|
Shi DL. Canonical and Non-Canonical Wnt Signaling Generates Molecular and Cellular Asymmetries to Establish Embryonic Axes. J Dev Biol 2024; 12:20. [PMID: 39189260 PMCID: PMC11348223 DOI: 10.3390/jdb12030020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
The formation of embryonic axes is a critical step during animal development, which contributes to establishing the basic body plan in each particular organism. Wnt signaling pathways play pivotal roles in this fundamental process. Canonical Wnt signaling that is dependent on β-catenin regulates the patterning of dorsoventral, anteroposterior, and left-right axes. Non-canonical Wnt signaling that is independent of β-catenin modulates cytoskeletal organization to coordinate cell polarity changes and asymmetric cell movements. It is now well documented that components of these Wnt pathways biochemically and functionally interact to mediate cell-cell communications and instruct cellular polarization in breaking the embryonic symmetry. The dysfunction of Wnt signaling disrupts embryonic axis specification and proper tissue morphogenesis, and mutations of Wnt pathway genes are associated with birth defects in humans. This review discusses the regulatory roles of Wnt pathway components in embryonic axis formation by focusing on vertebrate models. It highlights current progress in decoding conserved mechanisms underlying the establishment of asymmetry along the three primary body axes. By providing an in-depth analysis of canonical and non-canonical pathways in regulating cell fates and cellular behaviors, this work offers insights into the intricate processes that contribute to setting up the basic body plan in vertebrate embryos.
Collapse
Affiliation(s)
- De-Li Shi
- Department of Medical Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China;
- Laboratory of Developmental Biology, Centre National de la Recherche Scientifique (CNRS), UMR7622, Institut de Biologie Paris-Seine (IBPS), Sorbonne University, 75005 Paris, France
| |
Collapse
|
82
|
Wu Z, Zang Y, Li C, He Z, Liu J, Du Z, Ma X, Jing L, Duan H, Feng J, Yan X. CD146, a therapeutic target involved in cell plasticity. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1563-1578. [PMID: 38613742 DOI: 10.1007/s11427-023-2521-x] [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: 07/31/2023] [Accepted: 12/28/2023] [Indexed: 04/15/2024]
Abstract
Since its identification as a marker for advanced melanoma in the 1980s, CD146 has been found to have multiple functions in both physiological and pathological processes, including embryonic development, tissue repair and regeneration, tumor progression, fibrosis disease, and inflammations. Subsequent research has revealed that CD146 is involved in various signaling pathways as a receptor or co-receptor in these processes. This correlation between CD146 and multiple diseases has sparked interest in its potential applications in diagnosis, prognosis, and targeted therapy. To better comprehend the versatile roles of CD146, we have summarized its research history and synthesized findings from numerous reports, proposing that cell plasticity serves as the underlying mechanism through which CD146 contributes to development, regeneration, and various diseases. Targeting CD146 would consequently halt cell state shifting during the onset and progression of these related diseases. Therefore, the development of therapy targeting CD146 holds significant practical value.
Collapse
Affiliation(s)
- Zhenzhen Wu
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuzhe Zang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chuyi Li
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhiheng He
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jingyu Liu
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhaoqi Du
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinran Ma
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lin Jing
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hongxia Duan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China.
| | - Jing Feng
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China.
- Joint Laboratory of Nanozymes in Zhengzhou University, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
83
|
Guo S, Tong Y, Li T, Yang K, Gao W, Peng F, Zou X. Endoplasmic Reticulum Stress-Mediated Cell Death in Renal Fibrosis. Biomolecules 2024; 14:919. [PMID: 39199307 PMCID: PMC11352060 DOI: 10.3390/biom14080919] [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: 05/17/2024] [Revised: 07/04/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
The endoplasmic reticulum (ER) is indispensable for maintaining normal life activities. Dysregulation of the ER function results in the accumulation of harmful proteins and lipids and the disruption of intracellular signaling pathways, leading to cellular dysfunction and eventual death. Protein misfolding within the ER disrupts its delicate balance, resulting in the accumulation of misfolded or unfolded proteins, a condition known as endoplasmic reticulum stress (ERS). Renal fibrosis, characterized by the aberrant proliferation of fibrotic tissue in the renal interstitium, stands as a grave consequence of numerous kidney disorders, precipitating a gradual decline in renal function. Renal fibrosis is a serious complication of many kidney conditions and is characterized by the overgrowth of fibrotic tissue in the glomerular and tubular interstitium, leading to the progressive failure of renal function. Studies have shown that, during the onset and progression of kidney disease, ERS causes various problems in the kidneys, a process that can lead to kidney fibrosis. This article elucidates the underlying intracellular signaling pathways modulated by ERS, delineating its role in triggering diverse forms of cell death. Additionally, it comprehensively explores a spectrum of potential pharmacological agents and molecular interventions aimed at mitigating ERS, thereby charting novel research avenues and therapeutic advancements in the management of renal fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiangyu Zou
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China; (S.G.); (Y.T.); (T.L.); (K.Y.); (W.G.); (F.P.)
| |
Collapse
|
84
|
Li W, Wang H, Zhao J, Xia J, Sun X. scHyper: reconstructing cell-cell communication through hypergraph neural networks. Brief Bioinform 2024; 25:bbae436. [PMID: 39276328 PMCID: PMC11401449 DOI: 10.1093/bib/bbae436] [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: 05/26/2024] [Revised: 07/14/2024] [Accepted: 08/07/2024] [Indexed: 09/16/2024] Open
Abstract
Cell-cell communications is crucial for the regulation of cellular life and the establishment of cellular relationships. Most approaches of inferring intercellular communications from single-cell RNA sequencing (scRNA-seq) data lack a comprehensive global network view of multilayered communications. In this context, we propose scHyper, a new method that can infer intercellular communications from a global network perspective and identify the potential impact of all cells, ligand, and receptor expression on the communication score. scHyper designed a new way to represent tripartite relationships, by extracting a heterogeneous hypergraph that includes the source (ligand expression), the target (receptor expression), and the relevant ligand-receptor (L-R) pairs. scHyper is based on hypergraph representation learning, which measures the degree of match between the intrinsic attributes (static embeddings) of nodes and their observed behaviors (dynamic embeddings) in the context (hyperedges), quantifies the probability of forming hyperedges, and thus reconstructs the cell-cell communication score. Additionally, to effectively mine the key mechanisms of signal transmission, we collect a rich dataset of multisubunit complex L-R pairs and propose a nonparametric test to determine significant intercellular communications. Comparing with other tools indicates that scHyper exhibits superior performance and functionality. Experimental results on the human tumor microenvironment and immune cells demonstrate that scHyper offers reliable and unique capabilities for analyzing intercellular communication networks. Therefore, we introduced an effective strategy that can build high-order interaction patterns, surpassing the limitations of most methods that can only handle low-order interactions, thus more accurately interpreting the complexity of intercellular communications.
Collapse
Affiliation(s)
- Wenying Li
- School of Mathematics and System Science, Xinjiang University, No. 777 Huarui Street, Shuimogou District, Urumqi, Xinjiang 830017, China
| | - Haiyun Wang
- School of Mathematics and System Science, Xinjiang University, No. 777 Huarui Street, Shuimogou District, Urumqi, Xinjiang 830017, China
| | - Jianping Zhao
- School of Mathematics and System Science, Xinjiang University, No. 777 Huarui Street, Shuimogou District, Urumqi, Xinjiang 830017, China
| | - Junfeng Xia
- School of Mathematics and System Science, Xinjiang University, No. 777 Huarui Street, Shuimogou District, Urumqi, Xinjiang 830017, China
- Institute of Physical Science and Information Technology, Anhui University, No. 111 Jiulong Road, Shushan District, Hefei, Anhui 230601, China
| | - Xiaoqiang Sun
- School of Mathematics, Sun Yat-sen University, No. 135 Xingang Xi Road, Haizhu District, Guangzhou, Guangdong 510275, China
| |
Collapse
|
85
|
Gerdol M, Greco S, Marino R, Locascio A, Plateroti M, Sirakov M. Conserved Signaling Pathways in the Ciona robusta Gut. Int J Mol Sci 2024; 25:7846. [PMID: 39063090 PMCID: PMC11277035 DOI: 10.3390/ijms25147846] [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: 06/03/2024] [Revised: 07/04/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
The urochordate Ciona robusta exhibits numerous functional and morphogenetic traits that are shared with vertebrate models. While prior investigations have identified several analogies between the gastrointestinal tract (i.e., gut) of Ciona and mice, the molecular mechanisms responsible for these similarities remain poorly understood. This study seeks to address this knowledge gap by investigating the transcriptional landscape of the adult stage gut. Through comparative genomics analyses, we identified several evolutionarily conserved components of signaling pathways of pivotal importance for gut development (such as WNT, Notch, and TGFβ-BMP) and further evaluated their expression in three distinct sections of the gastrointestinal tract by RNA-seq. Despite the presence of lineage-specific gene gains, losses, and often unclear orthology relationships, the investigated pathways were characterized by well-conserved molecular machinery, with most components being expressed at significant levels throughout the entire intestinal tract of C. robusta. We also showed significant differences in the transcriptional landscape of the stomach and intestinal tract, which were much less pronounced between the proximal and distal portions of the intestine. This study confirms that C. robusta is a reliable model system for comparative studies, supporting the use of ascidians as a model to study gut physiology.
Collapse
Affiliation(s)
- Marco Gerdol
- Department of Life Sciences, Università degli Studi di Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy; (M.G.); (S.G.)
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy; (R.M.); (A.L.)
| | - Samuele Greco
- Department of Life Sciences, Università degli Studi di Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy; (M.G.); (S.G.)
| | - Rita Marino
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy; (R.M.); (A.L.)
| | - Annamaria Locascio
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy; (R.M.); (A.L.)
| | - Michelina Plateroti
- Institute of Genetics and Molecular and Cellular Biology, CNRS UMR7104–INSERM U1258–Université de Strasbourg, 1 Rue Laurent Fries, 67404 Illkirch, France
| | - Maria Sirakov
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy; (R.M.); (A.L.)
| |
Collapse
|
86
|
Hu L, Chen W, Qian A, Li YP. Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and disease. Bone Res 2024; 12:39. [PMID: 38987555 PMCID: PMC11237130 DOI: 10.1038/s41413-024-00342-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/27/2024] [Accepted: 05/12/2024] [Indexed: 07/12/2024] Open
Abstract
Wnts are secreted, lipid-modified proteins that bind to different receptors on the cell surface to activate canonical or non-canonical Wnt signaling pathways, which control various biological processes throughout embryonic development and adult life. Aberrant Wnt signaling pathway underlies a wide range of human disease pathogeneses. In this review, we provide an update of Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and diseases. The Wnt proteins, receptors, activators, inhibitors, and the crosstalk of Wnt signaling pathways with other signaling pathways are summarized and discussed. We mainly review Wnt signaling functions in bone formation, homeostasis, and related diseases, and summarize mouse models carrying genetic modifications of Wnt signaling components. Moreover, the therapeutic strategies for treating bone diseases by targeting Wnt signaling, including the extracellular molecules, cytosol components, and nuclear components of Wnt signaling are reviewed. In summary, this paper reviews our current understanding of the mechanisms by which Wnt signaling regulates bone formation, homeostasis, and the efforts targeting Wnt signaling for treating bone diseases. Finally, the paper evaluates the important questions in Wnt signaling to be further explored based on the progress of new biological analytical technologies.
Collapse
Affiliation(s)
- Lifang Hu
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Airong Qian
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
87
|
Wang Q, Lu W, Lu L, Wu R, Wu D. miR-575/RIPK4 axis modulates cell cycle progression and proliferation by inactivating the Wnt/β-catenin signaling pathway through inhibiting RUNX1 in colon cancer. Mol Cell Biochem 2024; 479:1747-1766. [PMID: 38480605 DOI: 10.1007/s11010-024-04938-w] [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] [Accepted: 01/10/2024] [Indexed: 07/18/2024]
Abstract
Receptor interacting protein serine/threonine kinase 4 (RIPK4) is widely involved in human cancer development. Nevertheless, its role in colon cancer (COAD) has not been elucidated till now. Our research aimed at exploring the function and underlying molecular mechanism of RIPK4 in COAD progression. Through bioinformatic analyses and RT-qPCR, RIPK4 was discovered to be increased in COAD cells and tissues, and its high level predicted poor prognosis. Loss-of-function assays revealed that RIPK4 silencing suppressed COAD cell growth, induced cell cycle arrest, and enhanced cell apoptosis. In vivo experiments also proved that tumor growth was inhibited by silencing of RIPK4. Luciferase reporter assay validated that RIPK4 was targeted and negatively regulated by miR-575. Western blotting demonstrated that Wnt3a, phosphorylated (p)-GSK-3β, and cytoplasmic and nuclear β-catenin protein levels, β-catenin nuclear translocation, and Cyclin D1, CDK4, Cyclin E, and c-Myc protein levels were reduced by RIPK4 knockdown, which however was reversed by treatment with LiCl, the Wnt/β-catenin pathway activator. LiCl also offset the influence of RIPK4 knockdown on COAD cell growth, cell cycle process, and apoptosis. Finally, RIPK4 downregulation reduced RUNX1 level, which was upregulated in COAD and its high level predicted poor prognosis. RIPK4 is positively associated with RUNX1 in COAD. Overexpressing RUNX1 antagonized the suppression of RIPK4 knockdown on RUNX1, Wnt3a, p-GSK-3β, cytoplasmic β-catenin, nuclear β-catenin, Cyclin D1, CDK4, Cyclin E, and c-Myc levels. Collectively, miR-575/RIPK4 axis repressed COAD progression via inactivating the Wnt/β-catenin pathway through downregulating RUNX1.
Collapse
Affiliation(s)
- Qun Wang
- Department of Hepatopancreatobiliary Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 16 Zhuodaoquan South Road, Hongshan District, Wuhan, 430079, China.
- Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, 430079, China.
- Colorectal Cancer Clinical Research Center of Hubei Province, Wuhan, 430079, China.
| | - Weijun Lu
- Department of Hepatopancreatobiliary Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 16 Zhuodaoquan South Road, Hongshan District, Wuhan, 430079, China
- Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, 430079, China
| | - Li Lu
- Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, 430079, China
- Department of Gastrointestinal Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Ruopu Wu
- Tianjin Medical University, Tianjin, 300070, China
| | - Dongde Wu
- Department of Hepatopancreatobiliary Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 16 Zhuodaoquan South Road, Hongshan District, Wuhan, 430079, China.
| |
Collapse
|
88
|
Ding Y, Chen ZQ, Pan WF, Chen HJ, Wu M, Lyu YQ, Xie H, Huang YC, Chen ZZ, Chen F. The association and underlying mechanism of the digit ratio (2D:4D) in hypospadias. Asian J Androl 2024; 26:356-365. [PMID: 38563741 PMCID: PMC11280205 DOI: 10.4103/aja202377] [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: 07/29/2023] [Accepted: 01/14/2024] [Indexed: 04/04/2024] Open
Abstract
The second-to-fourth digit (2D:4D) ratio is thought to be associated with prenatal androgen exposure. However, the relationship between the 2D:4D ratio and hypospadias is poorly understood, and its molecular mechanism is not clear. In this study, by analyzing the hand digit length of 142 boys with hypospadias (23 distal, 68 middle, and 51 proximal) and 196 controls enrolled in Shanghai Children's Hospital (Shanghai, China) from December 2020 to December 2021, we found that the 2D:4D ratio was significantly increased in boys with hypospadias ( P < 0.001) and it was positively correlated with the severity of the hypospadias. This was further verified by the comparison of control mice and prenatal low testosterone mice model obtained by knocking out the risk gene (dynein axonemal heavy chain 8 [ DNAH8 ]) associated with hypospadias. Furthermore, the discrepancy was mainly caused by a shift in 4D. Proteomic characterization of a mouse model validated that low testosterone levels during pregnancy can impair the growth and development of 4D. Comprehensive mechanistic explorations revealed that during the androgen-sensitive window, the downregulation of the androgen receptor (AR) caused by low testosterone levels, as well as the suppressed expression of chondrocyte proliferation-related genes such as Wnt family member 5a ( Wnt5a ), Wnt5b , Smad family member 2 ( Smad2 ), and Smad3 ; mitochondrial function-related genes in cartilage such as AMP-activated protein kinase ( AMPK ) and nuclear respiratory factor 1 ( Nrf-1 ); and vascular development-related genes such as myosin light chain ( MLC ), notch receptor 3 ( Notch3 ), and sphingosine kinase 1 ( Sphk1 ), are responsible for the limitation of 4D growth, which results in a higher 2D:4D ratio in boys with hypospadias via decreased endochondral ossification. This study indicates that the ratio of 2D:4D is a risk marker of hypospadias and provides a potential molecular mechanism.
Collapse
Affiliation(s)
- Yu Ding
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Zu-Quan Chen
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Wen-Feng Pan
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Hao-Jie Chen
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Min Wu
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Yi-Qing Lyu
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Hua Xie
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Yi-Chen Huang
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Zhong-Zhong Chen
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Urogenital Development Research Center, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Fang Chen
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Clinical Research Center for Hypospadias, Pediatric College, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China
| |
Collapse
|
89
|
de Nys R, Gardner A, van Eyk C, Mincheva-Tasheva S, Thomas P, Bhattacharjee R, Jolly L, Martinez-Garay I, Fox IWJ, Kamath KS, Kumar R, Gecz J. Proteomic analysis of the developing mammalian brain links PCDH19 to the Wnt/β-catenin signalling pathway. Mol Psychiatry 2024; 29:2199-2210. [PMID: 38454084 PMCID: PMC11408250 DOI: 10.1038/s41380-024-02482-z] [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: 07/06/2023] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 03/09/2024]
Abstract
Clustering Epilepsy (CE) is a neurological disorder caused by pathogenic variants of the Protocadherin 19 (PCDH19) gene. PCDH19 encodes a protein involved in cell adhesion and Estrogen Receptor α mediated-gene regulation. To gain further insights into the molecular role of PCDH19 in the brain, we investigated the PCDH19 interactome in the developing mouse hippocampus and cortex. Combined with a meta-analysis of all reported PCDH19 interacting proteins, our results show that PCDH19 interacts with proteins involved in actin, microtubule, and gene regulation. We report CAPZA1, αN-catenin and, importantly, β-catenin as novel PCDH19 interacting proteins. Furthermore, we show that PCDH19 is a regulator of β-catenin transcriptional activity, and that this pathway is disrupted in CE individuals. Overall, our results support the involvement of PCDH19 in the cytoskeletal network and point to signalling pathways where PCDH19 plays critical roles.
Collapse
Affiliation(s)
- Rebekah de Nys
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Alison Gardner
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Clare van Eyk
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Stefka Mincheva-Tasheva
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Paul Thomas
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Rudrarup Bhattacharjee
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Lachlan Jolly
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Isabel Martinez-Garay
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Ian W J Fox
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, Wales, United Kingdom
| | | | - Raman Kumar
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Jozef Gecz
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
- School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
90
|
Chi J, Bi W, Lou K, Ma J, Wu J, Cui Y. Research advances in Peyronie's disease: a comprehensive review on genomics, pathways, phenotypic manifestation, and therapeutic targets. Sex Med Rev 2024; 12:477-490. [PMID: 38456235 DOI: 10.1093/sxmrev/qeae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/02/2024] [Accepted: 01/26/2024] [Indexed: 03/09/2024]
Abstract
INTRODUCTION Penile induration disease, commonly known as Peyronie's disease (PD), is a connective tissue disorder that affects the penis, leading to the development of fibrous plaques, penile curvature, and erectile dysfunction. PD is a common male reproductive system disease with a complex etiology involving multiple genes, signaling pathways, and different phenotypes. OBJECTIVES The etiology and pathogenesis of PD remain poorly understood, hindering the development of effective treatment strategies. By understanding the underlying mechanisms of PD, we can pave the way for targeted therapies and improved patient outcomes. METHODS We reviewed the epidemiology and pathophysiology of PD. We performed database searches on Google Scholar, PubMed, Medline, and Web of Science from inception to September 2023. The literature reviewed included priapism guidelines, review articles, current trial studies, and various literature related to PD. RESULTS This article provides a comprehensive overview of the current research progress on the disease, focusing on its genetic factors, signaling pathways, cellular mechanisms, phenotypic manifestations, and therapeutic targets. It can help identify individuals at higher risk, aid in early detection and intervention, and provide insights into fibrosis and tissue remodeling. It can also reveal potential therapeutic targets, guide accurate diagnoses and treatment strategies, and address the impact of the disease on patients' quality of life. CONCLUSION By integrating insights from genomics, molecular pathways, clinical phenotypes, and therapeutic potentials, our research aims to achieve a deeper and more comprehensive understanding of PD, propelling the field toward innovative strategies that enhance the lives of those affected by PD. The complex manifestations and pathogenesis of PD necessitate the use of multiple treatment methods for personalized care.
Collapse
Affiliation(s)
- Junpeng Chi
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Wenhua Bi
- Department of Urology, Weifang Hospital of Traditional Chinese Medicine, Weifang, 265400, China
| | - Keyuan Lou
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Jian Ma
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Jitao Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Yuanshan Cui
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| |
Collapse
|
91
|
Kschonsak YT, Gao X, Miller SE, Hwang S, Marei H, Wu P, Li Y, Ruiz K, Dorighi K, Holokai L, Perampalam P, Tsai WTK, Kee YS, Agard NJ, Harris SF, Hannoush RN, de Sousa E Melo F. Potent and selective binders of the E3 ubiquitin ligase ZNRF3 stimulate Wnt signaling and intestinal organoid growth. Cell Chem Biol 2024; 31:1176-1187.e10. [PMID: 38056465 DOI: 10.1016/j.chembiol.2023.11.006] [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: 02/13/2023] [Revised: 08/21/2023] [Accepted: 11/10/2023] [Indexed: 12/08/2023]
Abstract
Selective and precise activation of signaling transduction cascades is key for cellular reprogramming and tissue regeneration. However, the development of small- or large-molecule agonists for many signaling pathways has remained elusive and is rate limiting to realize the full clinical potential of regenerative medicine. Focusing on the Wnt pathway, here we describe a series of disulfide-constrained peptides (DCPs) that promote Wnt signaling activity by modulating the cell surface levels of ZNRF3, an E3 ubiquitin ligase that controls the abundance of the Wnt receptor complex FZD/LRP at the plasma membrane. Mechanistically, monomeric DCPs induce ZNRF3 ubiquitination, leading to its cell surface clearance, ultimately resulting in FZD stabilization. Furthermore, we engineered multimeric DCPs that induce expansive growth of human intestinal organoids, revealing a dependence between valency and ZNRF3 clearance. Our work highlights a strategy for the development of potent, biologically active Wnt signaling pathway agonists via targeting of ZNRF3.
Collapse
Affiliation(s)
- Yvonne T Kschonsak
- Department of Discovery Oncology, Genentech Inc, South San Francisco, CA 94080, USA.
| | - Xinxin Gao
- Department of Early Discovery Biochemistry and Peptide Therapeutics, Genentech Inc, South San Francisco, CA 94080, USA.
| | - Stephen E Miller
- Department of Early Discovery Biochemistry and Peptide Therapeutics, Genentech Inc, South San Francisco, CA 94080, USA
| | - Sunhee Hwang
- Department of Early Discovery Biochemistry and Peptide Therapeutics, Genentech Inc, South San Francisco, CA 94080, USA
| | - Hadir Marei
- Department of Discovery Oncology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Ping Wu
- Department of Structural Biology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Yanjie Li
- Department of Early Discovery Biochemistry and Peptide Therapeutics, Genentech Inc, South San Francisco, CA 94080, USA
| | - Karen Ruiz
- Department of Discovery Oncology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Kristel Dorighi
- Department of Molecular Biology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Loryn Holokai
- Department of Biomarker Discovery, Genentech Inc, South San Francisco, CA 94080, USA
| | - Pirunthan Perampalam
- ProCogia Inc. under contract to Hoffmann-La Roche Limited, Toronto, Ontario M5J2P1, Canada
| | - Wen-Ting K Tsai
- Department of Antibody Engineering, Genentech Inc, South San Francisco, CA 94080, USA
| | - Yee-Seir Kee
- Department of Antibody Engineering, Genentech Inc, South San Francisco, CA 94080, USA
| | - Nicholas J Agard
- Department of Antibody Engineering, Genentech Inc, South San Francisco, CA 94080, USA
| | - Seth F Harris
- Department of Structural Biology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Rami N Hannoush
- Department of Early Discovery Biochemistry and Peptide Therapeutics, Genentech Inc, South San Francisco, CA 94080, USA.
| | | |
Collapse
|
92
|
Liu ZH, Xia Y, Ai S, Wang HL. Health risks of Bisphenol-A exposure: From Wnt signaling perspective. ENVIRONMENTAL RESEARCH 2024; 251:118752. [PMID: 38513750 DOI: 10.1016/j.envres.2024.118752] [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: 11/28/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
Human beings are routinely exposed to chronic and low dose of Bisphenols (BPs) due to their widely pervasiveness in the environment. BPs hold similar chemical structures to 17β-estradiol (E2) and thyroid hormone, thus posing threats to human health by rendering the endocrine system dysfunctional. Among BPs, Bisphenol-A (BPA) is the best-known and extensively studied endocrine disrupting compound (EDC). BPA possesses multisystem toxicity, including reproductive toxicity, neurotoxicity, hepatoxicity and nephrotoxicity. Particularly, the central nervous system (CNS), especially the developing one, is vulnerable to BPA exposure. This review describes our current knowledge of BPA toxicity and the related molecular mechanisms, with an emphasis on the role of Wnt signaling in the related processes. We also discuss the role of oxidative stress, endocrine signaling and epigenetics in the regulation of Wnt signaling by BPA exposure. In summary, dysfunction of Wnt signaling plays a key role in BPA toxicity and thus can be a potential target to alleviate EDCs induced damage to organisms.
Collapse
Affiliation(s)
- Zhi-Hua Liu
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
| | - Yanzhou Xia
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
| | - Shu Ai
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
| | - Hui-Li Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China.
| |
Collapse
|
93
|
Sampathkumar P, Jung H, Chen H, Zhang Z, Suen N, Yang Y, Huang Z, Lopez T, Benisch R, Lee SJ, Ye J, Yeh WC, Li Y. Targeted protein degradation systems to enhance Wnt signaling. eLife 2024; 13:RP93908. [PMID: 38847394 PMCID: PMC11161174 DOI: 10.7554/elife.93908] [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] [Indexed: 06/09/2024] Open
Abstract
Molecules that facilitate targeted protein degradation (TPD) offer great promise as novel therapeutics. The human hepatic lectin asialoglycoprotein receptor (ASGR) is selectively expressed on hepatocytes. We have previously engineered an anti-ASGR1 antibody-mutant RSPO2 (RSPO2RA) fusion protein (called SWEETS) to drive tissue-specific degradation of ZNRF3/RNF43 E3 ubiquitin ligases, which achieved hepatocyte-specific enhanced Wnt signaling, proliferation, and restored liver function in mouse models, and an antibody-RSPO2RA fusion molecule is currently in human clinical trials. In the current study, we identified two new ASGR1- and ASGR1/2-specific antibodies, 8M24 and 8G8. High-resolution crystal structures of ASGR1:8M24 and ASGR2:8G8 complexes revealed that these antibodies bind to distinct epitopes on opposing sides of ASGR, away from the substrate-binding site. Both antibodies enhanced Wnt activity when assembled as SWEETS molecules with RSPO2RA through specific effects sequestering E3 ligases. In addition, 8M24-RSPO2RA and 8G8-RSPO2RA efficiently downregulate ASGR1 through TPD mechanisms. These results demonstrate the possibility of combining different therapeutic effects and degradation mechanisms in a single molecule.
Collapse
Affiliation(s)
| | | | - Hui Chen
- Surrozen, IncSouth San FranciscoUnited States
| | | | | | - Yiran Yang
- Surrozen, IncSouth San FranciscoUnited States
| | - Zhong Huang
- Surrozen, IncSouth San FranciscoUnited States
| | - Tom Lopez
- Surrozen, IncSouth San FranciscoUnited States
| | | | | | - Jay Ye
- Surrozen, IncSouth San FranciscoUnited States
| | | | - Yang Li
- Surrozen, IncSouth San FranciscoUnited States
| |
Collapse
|
94
|
Du F, Li J, Zhang S, Zeng X, Nie J, Li Z. Oxidative stress in hair follicle development and hair growth: Signalling pathways, intervening mechanisms and potential of natural antioxidants. J Cell Mol Med 2024; 28:e18486. [PMID: 38923380 PMCID: PMC11196958 DOI: 10.1111/jcmm.18486] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/02/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Hair follicle development and hair growth are regulated by multiple factors and multiple signalling pathways. The hair follicle, as an important skin appendage, is the basis for hair growth, and it has the functions of safeguarding the body, perceiving the environment and regulating body temperature. Hair growth undergoes a regular hair cycle, including anagen, catagen and telogen. A small amount of physiological shedding of hair occurs under normal conditions, always in a dynamic equilibrium. Hair loss occurs when the skin or hair follicles are stimulated by oxidative stress, inflammation or hormonal disorders that disrupt the homeostasis of the hair follicles. Numerous researches have indicated that oxidative stress is an important factor causing hair loss. Here, we summarize the signalling pathways and intervention mechanisms by which oxidative stress affects hair follicle development and hair growth, discuss existing treatments for hair loss via the antioxidant pathway and provide our own insights. In addition, we collate antioxidant natural products promoting hair growth in recent years and discuss the limitations and perspectives of current hair loss prevention and treatment.
Collapse
Affiliation(s)
- Fanpan Du
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Basic Pharmacology of Guizhou ProvinceZunyi Medical UniversityZunyiChina
- Department of Pharmacology, School of PharmacyZunyi Medical UniversityZunyiChina
| | - Jingjie Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Basic Pharmacology of Guizhou ProvinceZunyi Medical UniversityZunyiChina
- Department of Pharmacology, School of PharmacyZunyi Medical UniversityZunyiChina
| | - Shiqian Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Basic Pharmacology of Guizhou ProvinceZunyi Medical UniversityZunyiChina
- Department of Pharmacology, School of PharmacyZunyi Medical UniversityZunyiChina
| | - Xuemei Zeng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Basic Pharmacology of Guizhou ProvinceZunyi Medical UniversityZunyiChina
- Department of Pharmacology, School of PharmacyZunyi Medical UniversityZunyiChina
| | - Jing Nie
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Basic Pharmacology of Guizhou ProvinceZunyi Medical UniversityZunyiChina
- Department of Pharmacology, School of PharmacyZunyi Medical UniversityZunyiChina
| | - Zheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Basic Pharmacology of Guizhou ProvinceZunyi Medical UniversityZunyiChina
- Department of Pharmacology, School of PharmacyZunyi Medical UniversityZunyiChina
| |
Collapse
|
95
|
Wrynn T, Min S, Horeth E, Osinski J, Sinha S, Romano RA. ΔNp63 regulates Sfrp1 expression to direct salivary gland branching morphogenesis. PLoS One 2024; 19:e0301082. [PMID: 38722977 PMCID: PMC11081224 DOI: 10.1371/journal.pone.0301082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/08/2024] [Indexed: 05/13/2024] Open
Abstract
Branching morphogenesis is a complex process shared by many organs including the lungs, kidney, prostate, as well as several exocrine organs including the salivary, mammary and lacrimal glands. This critical developmental program ensures the expansion of an organ's surface area thereby maximizing processes of cellular secretion or absorption. It is guided by reciprocal signaling from the epithelial and mesenchymal cells. While signaling pathways driving salivary gland branching morphogenesis have been relatively well-studied, our understanding of the underlying transcriptional regulatory mechanisms directing this program, is limited. Here, we performed in vivo and ex vivo studies of the embryonic mouse submandibular gland to determine the function of the transcription factor ΔNp63, in directing branching morphogenesis. Our studies show that loss of ΔNp63 results in alterations in the differentiation program of the ductal cells which is accompanied by a dramatic reduction in branching morphogenesis that is mediated by dysregulation of WNT signaling. We show that ΔNp63 modulates WNT signaling to promote branching morphogenesis by directly regulating Sfrp1 expression. Collectively, our findings have revealed a novel role for ΔNp63 in the regulation of this critical process and offers a better understanding of the transcriptional networks involved in branching morphogenesis.
Collapse
Affiliation(s)
- Theresa Wrynn
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Sangwon Min
- Department of Stem Cell and Regenerative Biology, Faculty of Arts and Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Erich Horeth
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Jason Osinski
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Satrajit Sinha
- Department of Biochemistry, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Rose-Anne Romano
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
96
|
Dutt P, Haider N, Mouaaz S, Podmore L, Stambolic V. β-catenin turnover is regulated by Nek10-mediated tyrosine phosphorylation in A549 lung adenocarcinoma cells. Proc Natl Acad Sci U S A 2024; 121:e2300606121. [PMID: 38683979 PMCID: PMC11087748 DOI: 10.1073/pnas.2300606121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/08/2024] [Indexed: 05/02/2024] Open
Abstract
β-catenin has influential roles affecting embryonic development, tissue homeostasis, and human diseases including cancer. Cellular β-catenin levels are exquisitely controlled by a variety of regulatory mechanisms. In the course of exploring the functions of the Nek10 tyrosine kinase, we observed that deletion of Nek10 in lung adenocarcinoma cells resulted in dramatic stabilization of β-catenin, suggestive of a Nek10 role in the control of β-catenin turnover. Nek10-deficient cells exhibited diminished ability to form tumorspheres in suspension, grow in soft agar, and colonize mouse lung tissue following tail vein injection. Mechanistically, Nek10 associates with the Axin complex, responsible for β-catenin degradation, where it phosphorylates β-catenin at Tyr30, located within the regulatory region governing β-catenin turnover. In the absence of Nek10 phosphorylation, GSK3-mediated phosphorylation of β-catenin, a prerequisite for its turnover, is impaired. This represents a divergent function within the Nek family, whose other members are serine-threonine kinases involved in different elements of the centrosomal cycle, primary cilia function, and DNA damage responses.
Collapse
Affiliation(s)
- Previn Dutt
- Princess Margaret Cancer Centre, University Health Network, Princess Margaret Cancer Research Tower, Toronto, ONM5G 1L7, Canada
| | - Nasir Haider
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Research Tower, Toronto, ONM5G 1L7, Canada
| | - Samar Mouaaz
- Princess Margaret Cancer Centre, University Health Network, Princess Margaret Cancer Research Tower, Toronto, ONM5G 1L7, Canada
| | - Lauren Podmore
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Research Tower, Toronto, ONM5G 1L7, Canada
| | - Vuk Stambolic
- Princess Margaret Cancer Centre, University Health Network, Princess Margaret Cancer Research Tower, Toronto, ONM5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Research Tower, Toronto, ONM5G 1L7, Canada
| |
Collapse
|
97
|
Loukas AT, Papadourakis M, Panagiotopoulos V, Zarmpala A, Chontzopoulou E, Christodoulou S, Katsila T, Zoumpoulakis P, Matsoukas MT. Natural Compounds for Bone Remodeling: A Computational and Experimental Approach Targeting Bone Metabolism-Related Proteins. Int J Mol Sci 2024; 25:5047. [PMID: 38732267 PMCID: PMC11084538 DOI: 10.3390/ijms25095047] [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: 03/15/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Osteoporosis, characterized by reduced bone density and increased fracture risk, affects over 200 million people worldwide, predominantly older adults and postmenopausal women. The disruption of the balance between bone-forming osteoblasts and bone-resorbing osteoclasts underlies osteoporosis pathophysiology. Standard treatment includes lifestyle modifications, calcium and vitamin D supplementation and specific drugs that either inhibit osteoclasts or stimulate osteoblasts. However, these treatments have limitations, including side effects and compliance issues. Natural products have emerged as potential osteoporosis therapeutics, but their mechanisms of action remain poorly understood. In this study, we investigate the efficacy of natural compounds in modulating molecular targets relevant to osteoporosis, focusing on the Mitogen-Activated Protein Kinase (MAPK) pathway and the gut microbiome's influence on bone homeostasis. Using an in silico and in vitro methodology, we have identified quercetin as a promising candidate in modulating MAPK activity, offering a potential therapeutic perspective for osteoporosis treatment.
Collapse
Affiliation(s)
- Alexandros-Timotheos Loukas
- Department of Food Science and Technology, University of West Attica, Ag. Spyridonos, 12243 Egaleo, Greece; (A.-T.L.); (P.Z.)
- Cloudpharm Private Company, Kifissias Avenue 44, 15125 Marousi, Greece; (V.P.); (A.Z.); (E.C.); (S.C.)
| | - Michail Papadourakis
- Cloudpharm Private Company, Kifissias Avenue 44, 15125 Marousi, Greece; (V.P.); (A.Z.); (E.C.); (S.C.)
| | - Vasilis Panagiotopoulos
- Cloudpharm Private Company, Kifissias Avenue 44, 15125 Marousi, Greece; (V.P.); (A.Z.); (E.C.); (S.C.)
- Department of Biomedical Engineering, University of West Attica, Ag. Spyridonos, 12243 Egaleo, Greece
| | - Apostolia Zarmpala
- Cloudpharm Private Company, Kifissias Avenue 44, 15125 Marousi, Greece; (V.P.); (A.Z.); (E.C.); (S.C.)
| | - Eleni Chontzopoulou
- Cloudpharm Private Company, Kifissias Avenue 44, 15125 Marousi, Greece; (V.P.); (A.Z.); (E.C.); (S.C.)
| | - Stephanos Christodoulou
- Cloudpharm Private Company, Kifissias Avenue 44, 15125 Marousi, Greece; (V.P.); (A.Z.); (E.C.); (S.C.)
| | - Theodora Katsila
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece;
| | - Panagiotis Zoumpoulakis
- Department of Food Science and Technology, University of West Attica, Ag. Spyridonos, 12243 Egaleo, Greece; (A.-T.L.); (P.Z.)
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece;
| | - Minos-Timotheos Matsoukas
- Cloudpharm Private Company, Kifissias Avenue 44, 15125 Marousi, Greece; (V.P.); (A.Z.); (E.C.); (S.C.)
- Department of Biomedical Engineering, University of West Attica, Ag. Spyridonos, 12243 Egaleo, Greece
| |
Collapse
|
98
|
Powell GT, Faro A, Zhao Y, Stickney H, Novellasdemunt L, Henriques P, Gestri G, White ER, Ren J, Lu W, Young RM, Hawkins TA, Cavodeassi F, Schwarz Q, Dreosti E, Raible DW, Li VSW, Wright GJ, Jones EY, Wilson SW. Cachd1 interacts with Wnt receptors and regulates neuronal asymmetry in the zebrafish brain. Science 2024; 384:573-579. [PMID: 38696577 PMCID: PMC7615972 DOI: 10.1126/science.ade6970] [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: 09/02/2022] [Accepted: 03/27/2024] [Indexed: 05/04/2024]
Abstract
Neurons on the left and right sides of the nervous system often show asymmetric properties, but how such differences arise is poorly understood. Genetic screening in zebrafish revealed that loss of function of the transmembrane protein Cachd1 resulted in right-sided habenula neurons adopting left-sided identity. Cachd1 is expressed in neuronal progenitors, functions downstream of asymmetric environmental signals, and influences timing of the normally asymmetric patterns of neurogenesis. Biochemical and structural analyses demonstrated that Cachd1 can bind simultaneously to Lrp6 and Frizzled family Wnt co-receptors. Consistent with this, lrp6 mutant zebrafish lose asymmetry in the habenulae, and epistasis experiments support a role for Cachd1 in modulating Wnt pathway activity in the brain. These studies identify Cachd1 as a conserved Wnt receptor-interacting protein that regulates lateralized neuronal identity in the zebrafish brain.
Collapse
Affiliation(s)
- Gareth T. Powell
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Wellcome Trust Sanger Institute; Cambridge CB10 1SA, UK
| | - Ana Faro
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Yuguang Zhao
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Heather Stickney
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Departments of Otolaryngology-HNS and Biological Structure, University of Washington; Seattle, WA 98195-7420, USA
- Ambry Genetics; Aliso Viejo, CA 92656, USA
| | - Laura Novellasdemunt
- The Francis Crick Institute; London, NW1 1AT, UK
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology; 08028, Barcelona, Spain
| | - Pedro Henriques
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Gaia Gestri
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | | | - Jingshan Ren
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Weixian Lu
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Rodrigo M. Young
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- Institute of Ophthalmology, University College London; London, EC1V 9EL, UK
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor; Camino La Piramide 5750, 8580745, Santiago, Chile
| | - Thomas A. Hawkins
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - Florencia Cavodeassi
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
- St. George’s, University of London; London, SW17 0RE, UK
| | - Quenten Schwarz
- Institute of Ophthalmology, University College London; London, EC1V 9EL, UK
| | - Elena Dreosti
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| | - David W. Raible
- Departments of Otolaryngology-HNS and Biological Structure, University of Washington; Seattle, WA 98195-7420, USA
| | | | - Gavin J. Wright
- Wellcome Trust Sanger Institute; Cambridge CB10 1SA, UK
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York; York, YO10 5DD, UK
| | - E. Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Stephen W. Wilson
- Cell and Developmental Biology, University College London; London, WC1E 6BT, UK
| |
Collapse
|
99
|
Gong H, Zhu C, Han D, Liu S. Secreted Glycoproteins That Regulate Synaptic Function: the Dispatchers in the Central Nervous System. Mol Neurobiol 2024; 61:2719-2727. [PMID: 37924485 DOI: 10.1007/s12035-023-03731-y] [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: 02/07/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023]
Abstract
Glycoproteins are proteins that contain oligosaccharide chains. As widely distributed functional proteins in the body, glycoproteins are essential for cellular development, cellular function maintenance, and intercellular communication. Glycoproteins not only play a role in the cell and the membrane, but they are also secreted in the intercell. These secreted glycoproteins are critical to the central nervous system for neurodevelopment and synaptic transmission. More specifically, secreted glycoproteins play indispensable roles in neurite growth mediation, axon guiding, synaptogenesis, neuronal differentiation, the release of synaptic vesicles, subunit composition of neurotransmitter receptors, and neurotransmitter receptor trafficking among other things. Abnormal expressions of secreted glycoproteins in the central nervous system are associated with abnormal neuron development, impaired synaptic organization/transmission, and neuropsychiatric disorders. This article reviews the secreted glycoproteins that regulate neuronal development and synaptic function in the central nervous system, and the molecular mechanism of these regulations, providing reference for research about synaptic function regulation and related central nervous system diseases.
Collapse
Affiliation(s)
- Haiying Gong
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Conglei Zhu
- Department of Pharmacy, Fuyang People's Hospital, Fuyang, Anhui, China
| | - Di Han
- Department of Respiratory and Critical Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Sen Liu
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
100
|
Fomo KN, Perumal N, Manicam C, Pfeiffer N, Grus FH. Neuroretinal Cell Culture Model as a Tool for the Development of New Therapeutic Approaches for Oxidative Stress-Induced Ocular Diseases, with a Focus on Glaucoma. Cells 2024; 13:775. [PMID: 38727311 PMCID: PMC11083839 DOI: 10.3390/cells13090775] [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: 03/04/2024] [Revised: 04/15/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Glaucoma is a heterogeneous group of optic neuropathies characterized by a progressive degeneration of the retinal ganglion cells (RGCs), leading to irreversible vision loss. Nowadays, the traditional therapeutic approach to glaucoma consists of lowering the intraocular pressure (IOP), which does not address the neurodegenerative features of the disease. Besides animal models of glaucoma, there is a considerable need for in vitro experimental models to propose new therapeutic strategies for this ocular disease. In this study, we elucidated the pathological mechanisms leading to neuroretinal R28 cell death after exposure to glutamate and hydrogen peroxide (H2O2) in order to develop new therapeutic approaches for oxidative stress-induced retinal diseases, including glaucoma. We were able to show that glutamate and H2O2 can induce a decrease in R28 cell viability in a concentration-dependent manner. A cell viability of about 42% was found after exposure to 3 mM of glutamate and about 56% after exposure to 100 µM of H2O2 (n = 4). Label-free quantitative mass spectrometry analysis revealed differential alterations of 193 and 311 proteins in R28 cells exposed to 3 mM of glutamate and 100 µM of H2O2, respectively (FDR < 1%; p < 0.05). Bioinformatics analysis indicated that the protein changes were associated with the dysregulation of signaling pathways, which was similar to those observed in glaucoma. Thus, the proteomic alteration induced by glutamate was associated with the inhibition of the PI3K/AKT signaling pathway. On the other hand, H2O2-induced toxicity in R28 cells was linked to the activation of apoptosis signaling and the inhibition of the mTOR and ERK/MAPK signaling pathways. Furthermore, the data show a similarity in the inhibition of the EIF2 and AMPK signaling pathways and the activation of the sumoylation and WNT/β-catenin signaling pathways in both groups. Our findings suggest that the exposure of R28 cells to glutamate and H2O2 could induce glaucoma-like neurodegenerative features and potentially provide a suitable tool for the development of new therapeutic strategies for retinal diseases.
Collapse
Affiliation(s)
| | | | | | | | - Franz H. Grus
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (K.N.F.); (N.P.); (C.M.); (N.P.)
| |
Collapse
|