1
|
Danek V, Tureckova J, Huebner K, Erlenbach-Wuensch K, Baranova P, Dobes J, Balounova J, Simova M, Novosadova V, Madureira Trufen CE, Prochazkova M, Talacko P, Harant K, Barinka C, Beck IM, Schneider-Stock R, Sedlacek R, Prochazka J. CUL4A exhibits tumor-suppressing role via regulation of HUWE1-mediated SMAD3 intracellular shuttling. Cancer Lett 2025; 621:217663. [PMID: 40120800 DOI: 10.1016/j.canlet.2025.217663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/06/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Changes in cellular physiology and proteomic homeostasis accompanied the initiation and progression of colorectal cancer. Thus, ubiquitination represents a central regulatory mechanism in proteome dynamics. However, the complexity of the ubiquitinating network involved in carcinogenesis remains unclear. This study revealed the tumor-suppressive role of the ubiquitin ligase Cullin4A (CUL4A) in the intestine. We showed that simultaneous loss of CUL4A and hyperactivation of the Wnt pathway promotes tumor development in the distal colon. This tumor development is caused by an accumulation of the inactive SMAD3, a TGF-β pathway mediator. Depletion of CUL4A resulted in stabilization of HUWE1, which attenuated SMAD3 function. We showed a correlation between the intracellular localization of CUL4A and colorectal cancer progression, where nuclear CUL4A localization correlates with advanced colorectal cancer progression. In summary, we identified CUL4A as an important regulator of SMAD3 signal transduction competence in a HUWE1-dependent manner and demonstrated a critical role for the crosstalk between ubiquitination and the Wnt/TGF-β signaling pathways in gastrointestinal homeostasis.
Collapse
Affiliation(s)
- Veronika Danek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Jolana Tureckova
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Kerstin Huebner
- Experimental Tumorpathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, 91054, Erlangen, Germany
| | | | - Petra Baranova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, 252 50, Czech Republic
| | - Jan Dobes
- Department of Cell Biology, Faculty of Science, Charles University, 128 00, Prague, Czech Republic
| | - Jana Balounova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Michaela Simova
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Vendula Novosadova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Carlos Eduardo Madureira Trufen
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Michaela Prochazkova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Pavel Talacko
- BIOCEV Proteomics Core Facility, Faculty of Science, Charles University, Vestec, 252 50, Czech Republic
| | - Karel Harant
- BIOCEV Proteomics Core Facility, Faculty of Science, Charles University, Vestec, 252 50, Czech Republic
| | - Cyril Barinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, 252 50, Czech Republic
| | - Inken M Beck
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic; Animal Research Centre, Ulm University, Ulm, Germany
| | - Regine Schneider-Stock
- Experimental Tumorpathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, 91054, Erlangen, Germany; Institute of Pathology, FAU Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Radislav Sedlacek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic; Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Jan Prochazka
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic; Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic.
| |
Collapse
|
2
|
Cervera-Juanes RP, Zimmerman KD, Wilhelm LJ, Lowe CC, Gonzales SW, Carlson T, Hitzemann R, Ferguson BM, Grant KA. Pre-existing DNA methylation signatures in the prefrontal cortex of alcohol-naïve nonhuman primates define neural vulnerability for future risky ethanol consumption. Neurobiol Dis 2025; 209:106886. [PMID: 40139280 PMCID: PMC12044430 DOI: 10.1016/j.nbd.2025.106886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/13/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Alcohol use disorder (AUD) is a highly prevalent, complex, multifactorial and heterogeneous disorder, with 11 % and 30 % of adults meeting criteria for past-year and lifetime AUD, respectively. Identification of the molecular mechanisms underlying risk for AUD would facilitate effective deployment of personalized interventions. Studies using rhesus monkeys and rats, have demonstrated that individuals with low cognitive flexibility and a predisposition towards habitual behaviors show an increased risk for future heavy drinking. Further, low cognitive flexibility is associated with reduced dorsolateral prefrontal cortex (dlPFC) function in rhesus monkeys. To explore the underlying unique molecular signatures that increase risk for chronic heavy drinking, a genome-wide DNA methylation (DNAm) analysis of the alcohol-naïve dlPFC-A46 biopsy prior to chronic alcohol self-administration was conducted. The DNAm profile provides a molecular snapshot of the alcohol-naïve dlPFC, with mapped genes and associated signaling pathways that vary across individuals. The analysis identified 1,463 differentially methylated regions (DMRs) related to unique genes that were strongly associated with average ethanol intake consumed over 6 months of voluntary self-administration. These findings translate behavioral phenotypes into neural markers of risk for AUD, and hold promise for parallel discoveries in risk for other disorders involving impaired cognitive flexibility. SIGNIFICANCE: Alcohol use disorder (AUD) is a highly prevalent and heterogeneous disorder. Prevention strategies to accurately identify individuals with a high risk for AUD, would help reduce the prevalence, and severity of AUD. Our novel epigenomic analysis of the alcohol-naïve nonhuman primate cortex provides a molecular snapshot of the vulnerable brain, pointing to circuitry and molecular mechanisms associated with cortical development, synaptic functions, glutamatergic signaling and coordinated signaling pathways. With a complex disorder like AUD, having the ability to identify the molecular mechanisms underlying AUD risk is critical for better development of personalized effective treatments.
Collapse
Affiliation(s)
- Rita P Cervera-Juanes
- Department of Translational Neuroscience, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, United States of America; Center for Precision Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, United States of America.
| | - Kip D Zimmerman
- Center for Precision Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, United States of America; Department of Internal Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157, United States of America
| | - Larry J Wilhelm
- Department of Translational Neuroscience, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, United States of America
| | - Clara Christine Lowe
- Department of Translational Neuroscience, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, United States of America
| | - Steven W Gonzales
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States of America
| | - Tim Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States of America
| | - Robert Hitzemann
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States of America; Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, United States of America
| | - Betsy M Ferguson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States of America; Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States of America
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States of America; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States of America; Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, United States of America
| |
Collapse
|
3
|
Surakhy M, Matheson J, Barnes DJ, Carter EJ, Hughes J, Bühnemann C, Sanegre S, Morreau H, Metz P, Imianowski CJ, Hassan AB. Smad4 and TGFβ1 dependent gene expression signatures in conditional intestinal adenoma, organoids and colorectal cancer. Sci Rep 2025; 15:16330. [PMID: 40348815 PMCID: PMC12065906 DOI: 10.1038/s41598-025-00908-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 05/02/2025] [Indexed: 05/14/2025] Open
Abstract
TGF-β ligands suppress growth yet can paradoxically and potently promote cancer invasion and metastasis depending on downstream pathway mutational context, such as loss of Mothers against decapentaplegic homolog 4 (Smad4). Here, we characterised phenotypes and associated gene expression signatures in conditional murine intestinal adenoma with and without Smad4. Conditional Lgr5-CreERT2 activation in Apcfl/flSmad4fl/fl mice resulted in homozygote floxed alleles (ApcΔ/ΔSmad4Δ/Δ) and adenoma formation. The adenoma phenotype was discordant, with reduced small intestinal adenoma burden yet development of large non-metastatic caecal adenoma with nuclear localisation of phospho-Smad2/3. Derived ApcΔ/ΔSmad4Δ/Δ adenoma organoids resisted TGF-β1 dose dependent growth arrest and cell death (IC50 534 pM) compared to ApcΔ/ΔSmad4+/+ (IC50 24 pM). TGF-β1 (390 pM) altered adenoma bulk mRNA expression most significantly for Id1low and Spp1high in ApcΔ/ΔSmad4Δ/Δ. Single cell RNAseq of caecal adenoma identified expansion of Lgr5low, Pak3high and Id1low progenitor populations in ApcΔ/ΔSmad4Δ/Δ. Of the 76 Smad4 and TGF-β1 dependent genes identified in Apcfl/flSmad4fl/fl adenoma organoids, only 7 human equivalent genes were differentially expressed in SMAD4 mutated colorectal cancer (TCGA cohorts), including ID1low. SMAD4low, ID1low SPP1high and PAK3high all correlated with poorer survival. Murine adenoma identified Smad4 dependent gene expression signatures that require further evaluation as functional biomarker classifiers of SMAD4 mutated cancer subtypes.
Collapse
Affiliation(s)
- Mirvat Surakhy
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Julia Matheson
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - David J Barnes
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Emma J Carter
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Jennifer Hughes
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Claudia Bühnemann
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Sabina Sanegre
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Paul Metz
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Charlotte J Imianowski
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Andrew Bassim Hassan
- Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| |
Collapse
|
4
|
Osburn SC, Smith ME, Wahl D, LaRocca TJ. Novel effects of reverse transcriptase inhibitor supplementation in skeletal muscle of old mice. Physiol Genomics 2025; 57:308-320. [PMID: 40062980 DOI: 10.1152/physiolgenomics.00115.2024] [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: 08/05/2024] [Revised: 10/08/2024] [Accepted: 02/15/2025] [Indexed: 04/01/2025] Open
Abstract
Aging is the primary risk factor for the development of many chronic diseases, including dementias, cardiovascular disease, and diabetes. There is significant interest in identifying novel "geroprotective" agents, including by repurposing existing drugs, but such treatments may affect organ systems differently. One current example is the nucleoside reverse transcriptase inhibitor 3TC, which has been increasingly studied as a potential gerotherapeutic. Recent data suggest that 3TC may reduce inflammation and improve cognitive function in older mice; however, the effects of 3TC on other tissues in aged animals are less well characterized. Here, we use transcriptomics (RNA-seq) and targeted metabolomics to investigate the influence of 3TC supplementation on skeletal muscle in older mice. We show that 3TC 1) does not overtly affect muscle mass or functional/health markers, 2) largely reverses age-related changes in gene expression and metabolite signatures, and 3) is potentially beneficial for mitochondrial function in old animals via increases in antioxidant enzymes and decreases in mitochondrial reactive oxygen species. Collectively, our results suggest that, in addition to its protective effects in other tissues, 3TC supplementation does not have adverse effects in aged muscle and may even protect muscle/mitochondrial health in this context.NEW & NOTEWORTHY Recent studies suggest that the nucleoside reverse transcriptase inhibitor 3TC may improve brain health and cognitive function in old mice, but its effects on other aging tissues have not been comprehensively studied. This is the first study to use a multiomics approach to investigate the effects of 3TC treatment on skeletal muscle of old mice. The results suggest that 3TC reverses age-related transcriptomic and metabolite signatures and is potentially beneficial for mitochondrial function in aged muscle.
Collapse
Affiliation(s)
- Shelby C Osburn
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| | - Meghan E Smith
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| | - Devin Wahl
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| | - Thomas J LaRocca
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| |
Collapse
|
5
|
McNamara HM, Guyer AM, Jia BZ, Parot VJ, Dobbs CD, Schier AF, Cohen AE, Lord ND. Optogenetic control of Nodal signaling patterns. Development 2025; 152:dev204506. [PMID: 40145591 PMCID: PMC12070070 DOI: 10.1242/dev.204506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/20/2025] [Indexed: 03/28/2025]
Abstract
A crucial step in early embryogenesis is the establishment of spatial patterns of signaling activity. Tools to perturb morphogen signals with high resolution in space and time can help reveal how embryonic cells decode these signals to make appropriate fate decisions. Here, we present new optogenetic reagents and an experimental pipeline for creating designer Nodal signaling patterns in live zebrafish embryos. Nodal receptors were fused to the light-sensitive heterodimerizing pair Cry2/CIB1N, and the type II receptor was sequestered to the cytosol. The improved optoNodal2 reagents eliminate dark activity and improve response kinetics, without sacrificing dynamic range. We adapted an ultra-widefield microscopy platform for parallel light patterning in up to 36 embryos, and demonstrated precise spatial control over Nodal signaling activity and downstream gene expression. Patterned Nodal activation drove precisely controlled internalization of endodermal precursors. Furthermore, we used patterned illumination to generate synthetic signaling patterns in Nodal signaling mutants, rescuing several characteristic developmental defects. This study establishes an experimental toolkit for systematic exploration of Nodal signaling patterns in live embryos.
Collapse
Affiliation(s)
| | - Alison M. Guyer
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Bill Z. Jia
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Vicente J. Parot
- Institute for Biological and Medical Engineering, Pontificia Universidad Católica de Chile, Santiago 7820244, Chile
| | - Caleb D. Dobbs
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Adam E. Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Nathan D. Lord
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
6
|
Fei X, Song C, Cui J, Li Y, Lei X, Tang H. The role of deubiquitinases in cardiovascular diseases: mechanisms and therapeutic implications. Front Cardiovasc Med 2025; 12:1582049. [PMID: 40376148 PMCID: PMC12078317 DOI: 10.3389/fcvm.2025.1582049] [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: 02/23/2025] [Accepted: 04/22/2025] [Indexed: 05/18/2025] Open
Abstract
Cardiovascular diseases (CVDs) have become the leading cause of death globally, surpassing infectious diseases and other chronic illnesses. The incidence and mortality rates of CVDs are rising worldwide, posing a key challenge in public health. The ubiquitination system is a vast and complex. It is an important post-translational modification that plays a crucial role in various cellular processes. Deubiquitination is catalyzed by deubiquitinases (DUBs), which remove ubiquitin (Ub) from ubiquitinated proteins, thereby reversing the ubiquitination process. DUBs play an important role in many biological processes, such as DNA repair, cell metabolism, differentiation, epigenetic regulation, and protein stability control. They also participate in the regulation of many signaling pathways associated with the development and progression of CVDs. In this review, we primarily focus on the role of DUBs in various key pathological mechanisms of atherosclerosis (AS), such as foam cell formation, vascular remodeling (VR), endothelial-to-mesenchymal transition (End-MT), and clonal hematopoiesis (CH). In the heart, we summarize the involvement of DUBs in diseases and pathological processes, including heart failure (HF), myocardial infarction (MI), myocardial hypertrophy (MH) and ischemia/reperfusion (I/R) injury. Additionally, we also explore the diabetic cardiomyopathy (DCM) and the use of doxorubicin-induced cardiotoxicity in clinical settings. A comprehensive understanding of deubiquitination may provide new insights for the treatment and drug design of CVDs.
Collapse
Affiliation(s)
- Xiangyu Fei
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Chao Song
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Jian Cui
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Yuqing Li
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Xiaoyong Lei
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Huifang Tang
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| |
Collapse
|
7
|
Haantjes RR, Strik J, de Visser J, Postma M, van Amerongen R, van Boxtel AL. Towards an integrated view and understanding of embryonic signalling during murine gastrulation. Cells Dev 2025:204028. [PMID: 40316255 DOI: 10.1016/j.cdev.2025.204028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
At the onset of mammalian gastrulation, secreted signalling molecules belonging to the Bmp, Wnt, Nodal and Fgf signalling pathways induce and pattern the primitive streak, marking the start for the cellular rearrangements that generate the body plan. Our current understanding of how signalling specifies and organises the germ layers in three dimensions, was mainly derived from genetic experimentation using mouse embryos performed over many decades. However, the exact spatiotemporal sequence of events is still poorly understood, both because of a lack of tractable models that allow for real time visualisation of signalling and differentiation and because of the molecular and cellular complexity of these early developmental events. In recent years, a new wave of in vitro embryo models has begun to shed light on the dynamics of signalling during primitive streak formation. Here we discuss the similarities and differences between a widely adopted mouse embryo model, termed gastruloids, and real embryos from a signalling perspective. We focus on the gene regulatory networks that underlie signalling pathway interactions and outline some of the challenges ahead. Finally, we provide a perspective on how embryo models may be used to advance our understanding of signalling dynamics through computational modelling.
Collapse
Affiliation(s)
- Rhanna R Haantjes
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| | - Jeske Strik
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands; Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, 6525GA Nijmegen, the Netherlands.
| | - Joëlle de Visser
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| | - Marten Postma
- Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| | - Renée van Amerongen
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| | - Antonius L van Boxtel
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Mallo M. The Link Between TGF-β Signaling and Chromatin Dynamics Takes an Unexpected Path. Bioessays 2025:e70013. [PMID: 40296304 DOI: 10.1002/bies.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
The TGF-β/BMP signaling superfamily plays critical roles in regulating a wide range of physiological and pathological processes. A recent study on Tgfbr1 function during mouse embryonic development found that, in the absence of this gene, the primordium of the external genitalia changes its fate, forming an additional set of hindlimbs and resulting in the so-called "six-legged" mice. Analysis of these embryos demonstrated that Tgfbr1 induces global remodeling of chromatin accessibility, establishing distinct regulatory elements that other morphogenetic factors use to drive either hindlimb or genital development. While global changes in chromatin structure have been previously associated with TGF-β signaling, the alterations seen in six-legged embryos differ significantly from previously described mechanisms. In this review, I discuss these distinctions and underscore the importance of studying biological processes within their natural tissue context, where essential tissue interactions are preserved.
Collapse
Affiliation(s)
- Moises Mallo
- Gulbenkian Institute for Molecular Medicine, Lisboa, Portugal
| |
Collapse
|
9
|
Caraballo LAS, Filho LASF, Sena LS, Biagiotti D, de Moura JDS, de Sousa Júnior A, Rocha AO, de Sousa FCB, da Silva Santos NP, Sarmento JLR. Genome-wide association study applied to prolificacy in Santa Inês sheep. Trop Anim Health Prod 2025; 57:169. [PMID: 40214842 DOI: 10.1007/s11250-025-04424-5] [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: 10/29/2024] [Accepted: 04/04/2025] [Indexed: 05/01/2025]
Abstract
This study aimed to identify genomic regions associated with prolificacy in Santa Inês sheep raised in tropical conditions. The prolificacy of the dam was defined as single (only one lamb born per ewe per lambing) or multiple (more than one animal born per ewe per lambing). After quality control of phenotypic data, 1584 lambing records of 715 females occurred between the years 2000 and 2018 were used. The animals were genotyped with the OvineSNP50 BeadChip panel (Illumina Inc.). After quality control of genomic data, information of 46,714 SNPs and 388 samples and females was used for the subsequent analyses. The single-step GWAS (ssGWAS) methodology was used to estimate the effects of genetic markers and their association with the prolificacy. A total of 21 windows of 10 adjacent SNPs that explained at least 0.5% of the additive genetic variance for prolificacy were identified. In such regions, genes associated with different reproductive functions in the female were found: CACNA1E, NTRK1, PLCH1, SMAD3, CENPF, TOPBP1, IL33, DRD2, MID1, HCCS, and ARHGAP6. Some candidate regions related to prolificacy harbor genes that were not previously described and genes without known functions. These results can help to identify genes associated with prolificacy and could be used in genomic reproductive studies on prolificacy, as well as in the selection of the most prolific ewes in the population.
Collapse
Affiliation(s)
- Luis Andrés Salazar Caraballo
- Agrarian Sciences Center (CCA), Federal University of Piauí (UFPI), Campus Universitário Ministro Petrônio Portella, Teresina, PI, Brazil
| | | | - Luciano Silva Sena
- Phd in Animal Science, Graduate Program in Animal Science, Agrarian Sciences Center (CCA), UFPI, Campus Universitário Ministro Petrônio Portella, Teresina, Brazil
| | - Daniel Biagiotti
- Technical College of Teresina, UFPI, Campus Ministro Petrônio Portella, Teresina, PI, Brazil
| | - José Dos Santos de Moura
- Federal Institute of Education, Science and Technology of Piauí, IFPI, José de Freitas, PI, Brazil
| | - Antônio de Sousa Júnior
- Technical College of Teresina, UFPI, Campus Ministro Petrônio Portella, Teresina, PI, Brazil
| | | | | | | | | |
Collapse
|
10
|
Ji Z, Siu WYS, Dueñas ME, Müller L, Trost M, Carvalho P. Suppression of TGF-β/SMAD signaling by an inner nuclear membrane phosphatase complex. Nat Commun 2025; 16:3474. [PMID: 40216785 PMCID: PMC11992160 DOI: 10.1038/s41467-025-58681-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: 08/08/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Cytokines of the TGF-β superfamily control essential cell fate decisions via receptor regulated SMAD (R-SMAD) transcription factors. Ligand-induced R-SMAD phosphorylation in the cytosol triggers their activation and nuclear accumulation. We determine how R-SMADs are inactivated by dephosphorylation in the cell nucleus to counteract signaling by TGF-β superfamily ligands. We show that R-SMAD dephosphorylation is mediated by an inner nuclear membrane associated complex containing the scaffold protein MAN1 and the CTDNEP1-NEP1R1 phosphatase. Structural prediction, domain mapping and mutagenesis reveals that MAN1 binds independently to the CTDNEP1-NEP1R1 phosphatase and R-SMADs to promote their inactivation by dephosphorylation. Disruption of this complex causes nuclear accumulation of R-SMADs and aberrant signaling, even in the absence of TGF-β ligands. These findings establish CTDNEP1-NEP1R1 as the R-SMAD phosphatase, reveal the mechanistic basis for TGF-β signaling inactivation and highlight how this process is disrupted by disease-associated MAN1 mutations.
Collapse
Affiliation(s)
- Zhe Ji
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | | - Maria Emilia Dueñas
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- The Kids Research Institute Australia, Perth Children's Hospital, Nedlands, Australia
| | - Leonie Müller
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Matthias Trost
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Pedro Carvalho
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
11
|
Yang W, Cheng R, Qin M, Pan X, Tan Y, Feng K, Zhang J, Huang J. Tricellulin facilitates colorectal cancer metastasis through activation of the TGFβ/SMAD2/3 signalling pathway. Front Oncol 2025; 15:1562976. [PMID: 40291908 PMCID: PMC12021625 DOI: 10.3389/fonc.2025.1562976] [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: 01/18/2025] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Background Tricellulin belongs to the TAMP family of proteins and is primarily localized at the tricellular tight junctions. While its role in the progression of cancer has been reported, its importance in the progression of colorectal cancer (CRC) remains unclear. Objective This study aimed to determine the function and mechanism of tricellulin in CRC progression. Methods The proteins expression in cells and/or tissues was determined by Western blot, immunohistochemistry staining, and/or RT-qPCR analyses. The biological functions of tricellulin were investigated through in vitro assays (CCK-8, Transwell migration, and colony formation assays) and in vivo xenograft models. Tricellulin was significantly upregulated in CRC tissues compared to adjacent normal tissues. The expression of tricellulin was correlated with poor prognosis in patients with CRC. Results In vitro assays showed that tricellulin enhanced CRC cell proliferation, migration, and invasion. Mechanistically, tricellulin activated the TGFb1/SMAD2/3 pathway, while TGFb1 reciprocally controlled the expression of tricellulin. Also, tricellulin promotes CRC cell migration/invasion through EMT. In vivo models confirmed that the overexpression of tricellulin facilitated tumor growth and activated the TGFb1/ SMAD2/3 pathway in CRC. Conclusion Our findings demonstrate thatTricellulin promotes the metastasis of colorectal cancer by activating the TGF-β/SMAD2/3 signaling pathway, and TGF-β1 can reciprocally regulate the expression of tricellulin.We have revealed a novel mechanism by which tricellulin forms a positive feedback loop to promote the growth and metastasis of CRC. This mechanism provides novel insights into CRC progression and suggests potential therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jinxiu Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiean Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
12
|
Sadri S, Aghajani A, Soleimani H, Ghorbani Kalkhajeh S, Nazari H, Brouki Milan P, Peyravian N, Pezeshkian Z, Malekzadeh Kebria M, Shirazi F, Shams E, Naderi Noukabadi F, Nazemalhosseini-Mojarad E, Salehi Z. Exploring the Role of the TGF-β Signaling Pathway in Colorectal Precancerous Polyps Biochemical Genetics. Biochem Genet 2025; 63:1116-1148. [PMID: 39636332 DOI: 10.1007/s10528-024-10988-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: 09/01/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Colorectal cancer (CRC) is an important public health issue and is the third most common cancer, accounting for approximately 10% of all cancer cases worldwide. CRC results from the accumulation of multiple genetic and epigenetic alterations in the normal epithelial cells of the colon and rectum, leading to the development of colorectal polyps and invasive carcinomas. The transforming growth factor-beta (TGF-β) pathway is regulated in many diseases, such as cancer. This factor can show tumor suppressant function in the early stages in healthy and cancer cells. It can be regulated and affected by different factors, including noncoding RNAs, which are the remarkable regulators for this pathway. The most prominent functions of this factor are cell cycle arrest and apoptosis in cancer cells. However, activating at the final stages of the cell cycle can cause tumor metastasis. Thus, the dual function of TGF-β and the pleiotropic nature of this signaling make it a crucial challenge for cancer treatment. Accurately studying the TGF-β signaling pathway is critical to determine its role. One of the roles of TGF-β signaling is its significant effect on colorectal polyp malignancy and cancer. In this article, we review the published scientific papers regarding the TGF-β signaling pathway, its related genes, and their contribution to precancerous conditions and colorectal cancer progression. The complex interaction of the TGF-β signaling pathway with noncoding RNAs, such as lncRNA TUG1 and miR-21, significantly influences colorectal polyp and cancer progression. Identifying dysregulated TGF-β-related noncoding RNAs offers promising therapeutic avenues for colorectal cancer. Comprehending TGF-β's connection to other molecular mechanisms is crucial for advancing effective therapeutic strategies.
Collapse
Affiliation(s)
- Shadi Sadri
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 19835-178, Iran
| | - Ali Aghajani
- School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hiva Soleimani
- Department of General Biology, Faculty of Fundamental Science, Islamic Azad University of Shahr-E Qods, Tehran, 37515-374, Iran
| | - Sourena Ghorbani Kalkhajeh
- Department of Radiologic Technology, School of Allied Medical Sciences, Ahvaz Jundi-Shapour University of Medical Sciences, Ahvaz, Iran
| | - Haniyeh Nazari
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, 19395-1495, Iran
| | - Peiman Brouki Milan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Noshad Peyravian
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Pezeshkian
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Maziar Malekzadeh Kebria
- Cancer Research Center, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Shirazi
- Division of Genetics, Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, 817467344, Iran
| | - Elahe Shams
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 19835-178, Iran
| | - Fatemeh Naderi Noukabadi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 19835-178, Iran
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 19835-178, Iran.
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.
| | - Zahra Salehi
- Department of Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, 14114, Iran.
| |
Collapse
|
13
|
Japa O, Phuangsri C, Klinbumrung K, Prakhammin K. Receptor-regulated smads (R-Smads) in the liver fluke Fasciola gigantica: Characterization, comparative sequence analysis, and life stage-specific expression. Acta Trop 2025; 264:107603. [PMID: 40157585 DOI: 10.1016/j.actatropica.2025.107603] [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: 02/11/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
The transforming growth factor-beta (TGF-β) signaling pathway is a key conserved developmental pathway, demonstrating remarkable conservation in its components, structures, and functions across diverse animal species. This study presents a comprehensive characterization and comparative analysis of receptor-regulated Smads (R-Smads) in Fasciola gigantica, a major parasite affecting cattle, buffalo, and ruminants in tropical regions. Four R-Smads were identified: FgSmad1/5, FgSmad2, FgSmad3, and FgSmad8. Full-length cDNAs for these R-Smads, isolated using the rapid amplification of cDNA ends (RACE) approach, were 3348, 3010, 5172, and 1991 bp in length, encoding proteins of 464, 506, 678, and 404 amino acids, respectively. FgSmad1/5 and FgSmad8 were classified as bone morphogenetic protein (BMP)-specific R-Smads (BR-Smads) due to their conserved signature motifs and similarity to those found in parasitic platyhelminths and mammalian hosts. Conversely, FgSmad2 and FgSmad3 were identified as TGF-β/activin-specific R-Smads (AR-Smads), displaying a distinctive pattern of conserved motifs commonly observed among trematode parasites. Notably, FgSmad2 exhibited an atypical feature for R-Smads, as it lacked the MH1 domain. The expressions of F. gigantica R-Smad genes were observed across all developmental stages, with the highest levels of most R-Smads occurring in the unembryonated egg stage (primarily BR-Smads) and the metacercaria stage (predominantly AR-Smads), underscoring a significant shift in TGF-β and BMP signaling dynamics during development. These findings greatly advance our understanding of R-Smad proteins within the TGF-β signaling pathway of F. gigantica and provide a solid foundation for future research into their roles and regulatory mechanisms.
Collapse
Affiliation(s)
- Ornampai Japa
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao, Thailand; Scientific Instrument and Product Standard Quality Inspection Center, University of Phayao, Phayao, Thailand.
| | - Chorpaka Phuangsri
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Khuruwan Klinbumrung
- Scientific Instrument and Product Standard Quality Inspection Center, University of Phayao, Phayao, Thailand
| | - Khanuengnij Prakhammin
- Department of Applied Statistics, Rajamangala University of Technology Isan, Khon Kaen Campus, Khon Kaen, Thailand
| |
Collapse
|
14
|
Chia ZJ, Kumarapperuma H, Zhang R, Little PJ, Kamato D. Smad transcription factors as mediators of 7 transmembrane G protein-coupled receptor signalling. Acta Pharmacol Sin 2025; 46:795-804. [PMID: 39506064 PMCID: PMC11950520 DOI: 10.1038/s41401-024-01413-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024]
Abstract
The Smad transcription factors are well known for their role at the core of transforming growth factor-β (TGF-β) signalling. However, recent evidence shows that the Smad transcription factors play a vital role downstream of other classes of receptors including G protein-coupled receptors (GPCR). The versatility of Smad transcription factors originated from the two regions that can be differently activated by the TGF-β receptor superfamily or through the recruitment of intracellular kinases stimulated by other receptors classes such as GPCRs. The classic GPCR signalling cascade is further expanded to conditional adoption of the Smad transcription factor under the stimulation of Akt, demonstrating the unique involvement of the Smad transcription factor in GPCR signalling pathways in disease environments. In this review, we provide a summary of the signalling pathways of the Smad transcription factors as an important downstream mediator of GPCRs, presenting exciting opportunities for discovering new therapeutic targets for diseases.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Hirushi Kumarapperuma
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Ruizhi Zhang
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Environment and Science, Griffith Sciences, Griffith University, Nathan, QLD, Australia
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia.
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
- School of Environment and Science, Griffith Sciences, Griffith University, Nathan, QLD, Australia.
| |
Collapse
|
15
|
Pfeiffer F, Boshans LL, Jukkola P, Beebe M, Akinlaja Y, Aksoy EB, Sun Y, Baez CBR, Hedrich UBS, Arbelo U, Nishiyama A. Oligodendrocyte precursor cells establish regulatory contacts with the soma of active neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.646001. [PMID: 40236059 PMCID: PMC11996299 DOI: 10.1101/2025.03.28.646001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Oligodendrocyte precursor cells (OPCs) can generate myelinating oligodendrocytes life-long, a process that is dependent on neuronal activity. However, it is possible that OPCs have additional functions, influencing neuronal functions directly. We have used a mouse genetic model of juvenile seizures and chemical induction of neuronal activity to examine the morphological and molecular changes in OPCs around activated principal neurons. We found an increase in process extension of OPCs specifically toward the soma of activated neurons. Moreover, we found that the close proximity of OPC processes around neurons expressing the immediate early gene c-Fos decreased the calcium transients in these neurons, indicating a regulative function of OPCs. Analyses of transcriptome and chromatin accessibility revealed significant changes in genes involved in transforming growth factor beta (TGFβ) signaling. Extracellular matrix genes, particularly those encoding type VI collagen, an established binding partner for the OPC surface protein NG2, was increased around active neurons. Our findings indicate that OPCs are an integral part of the neural network and may help to decrease the activity of neurons that have previously been over-excited, in order to protect these neurons.
Collapse
|
16
|
Ward T, Morton SU, Venturini G, Tai W, Jang MY, Gorham J, Delaughter D, Wasson LK, Khazal Z, Homsy J, Gelb BD, Chung WK, Bruneau BG, Brueckner M, Tristani-Firouzi M, DePalma SR, Seidman C, Seidman JG. Modeling SMAD2 Mutations in Induced Pluripotent Stem Cells Provides Insights Into Cardiovascular Disease Pathogenesis. J Am Heart Assoc 2025; 14:e036860. [PMID: 40028843 DOI: 10.1161/jaha.124.036860] [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: 11/05/2024] [Accepted: 01/17/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND SMAD2 is a coregulator that binds a variety of transcription factors in human development. Heterozygous SMAD2 loss-of-function and missense variants are identified in patients with congenital heart disease (CHD) or arterial aneurysms. Mechanisms that cause distinct cardiovascular phenotypes remain unknown. We aimed to define transcriptional and epigenetic effects of SMAD2 variants and their role in CHD. We also assessed the function of SMAD2 missense variants of uncertain significance. METHODS AND RESULTS Rare SMAD2 variants (minor allele frequency ≤10-5) were identified in exome sequencing of 11 336 participants with CHD. We constructed isogenic induced pluripotent stem cells with heterozygous or homozygous loss-of-function and missense SMAD2 variants identified in CHD probands. Wild-type and mutant induced pluripotent stem cells were analyzed using bulk RNA sequencing, chromatin accessibility (Assay for Transposase-Accessible Chromatin With Sequencing), and integrated with published SMAD2/3 chromatin immunoprecipitation data. Cardiomyocyte differentiation and contractility were evaluated. Thirty participants with CHD had heterozygous loss-of-function or missense SMAD2 variants. SMAD2 haploinsufficiency altered chromatin accessibility at promoters and dysregulated expression of 385 SMAD regulated genes, including 10 CHD-associated genes. Motifs enriched in differential Assay for Transposase-Accessible Chromatin peaks predicted that SMAD2 haploinsufficiency disrupts interactions with transcription factors NANOG (homeobox protein NANOG), ETS, TEAD3/4 (transcriptional enhanced associate domain 3/4), CREB1 (cAMP response element binding protein 1), and AP1 (activator protein 1). Compared with SMAD2-haploinsufficient cells, induced pluripotent stem cells with R114C or W274C variants exhibited distinct and shared chromatin accessibility and transcription factor binding changes. CONCLUSIONS SMAD2 haploinsufficiency disrupts transcription factor binding and chromatin interactions critical for cardiovascular development. Differences between the molecular consequences of loss-of-function and missense variants likely contribute to phenotypic heterogeneity. These findings indicate opportunities for molecular analyses to improve reclassification of SMAD2 variants of uncertain clinical significance.
Collapse
Affiliation(s)
- Tarsha Ward
- Department of Genetics Harvard Medical School Boston MA USA
| | - Sarah U Morton
- Department of Genetics Harvard Medical School Boston MA USA
- Division of Newborn Medicine Boston Children's Hospital Boston MA USA
| | | | - Warren Tai
- Department of Genetics Harvard Medical School Boston MA USA
| | - Min Young Jang
- Department of Genetics Harvard Medical School Boston MA USA
| | - Joshua Gorham
- Department of Genetics Harvard Medical School Boston MA USA
| | - Dan Delaughter
- Department of Genetics Harvard Medical School Boston MA USA
| | | | - Zahra Khazal
- Department of Genetics Harvard Medical School Boston MA USA
| | - Jason Homsy
- Department of Genetics Harvard Medical School Boston MA USA
- Cardurion Pharmaceuticals, Inc. Burlington MA USA
| | - Bruce D Gelb
- Mindich Child Health and Development Institute and the Department of Pediatrics and Genetics and Genomic Sciences Icahn School of Medicine at Mount Sinai New York NY USA
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital Harvard Medical School Boston MA USA
| | - Benoit G Bruneau
- Gladstone Institutes San Francisco CA USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone San Francisco CA USA
- Department of Pediatrics, Cardiovascular Research Institute, Institute for Human Genetics, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research University of California San Francisco CA USA
| | - Martina Brueckner
- Department of Genetics and Pediatrics Yale University School of Medicine New Haven CT USA
| | - Martin Tristani-Firouzi
- Division of Pediatric Cardiology University of Utah and School of Medicine Salt Lake City UT USA
| | | | - Christine Seidman
- Department of Genetics Harvard Medical School Boston MA USA
- Department of Medicine Brigham and Women's Hospital Boston MA USA
- Howard Hughes Medical Institute Harvard Medical School Boston MA USA
| | - J G Seidman
- Department of Genetics Harvard Medical School Boston MA USA
| |
Collapse
|
17
|
Gallardo FS, Cruz-Soca M, Bock-Pereda A, Faundez-Contreras J, Gutiérrez-Rojas C, Gandin A, Torresan V, Casar JC, Ravasio A, Brandan E. Role of TGF-β/SMAD/YAP/TAZ signaling in skeletal muscle fibrosis. Am J Physiol Cell Physiol 2025; 328:C1015-C1028. [PMID: 39925133 DOI: 10.1152/ajpcell.00541.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/22/2024] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Skeletal muscle fibrosis is strongly associated with the differentiation of its resident multipotent fibro/adipogenic progenitors (FAPs) toward the myofibroblast phenotype. Although transforming growth factor type β (TGF-β) signaling is well-known for driving FAPs differentiation and fibrosis, due to its pleiotropic functions its complete inhibition is not suitable for treating fibrotic disorders such as muscular dystrophies. Here, we describe that TGF-β operates through the mechanosensitive transcriptional regulators Yes-associated protein (YAP)/ transcriptional coactivator with PDZ-binding motif (TAZ) to determine the myofibroblast fate of FAPs and skeletal muscle fibrosis. Spatial transcriptomics analyses of dystrophic and acute injured muscles showed that areas with active fibrosis and TGF-β signaling displayed high YAP/TAZ activity. Using a TGF-β-driven fibrotic mouse model, we found that activation of YAP/TAZ in activated FAPs is associated with the fibrotic process. Mechanistically, primary culture of FAPs reveals the remarkable ability of TGF-β1 to activate YAP/TAZ through its canonical SMAD3 pathway. Moreover, inhibition of YAP/TAZ, either by disrupting its activity (with Verteporfin) or cellular mechanotransduction (with the Rho inhibitor C3 or soft matrices), decreased TGF-β1-dependent FAPs differentiation into myofibroblasts. In vivo, administration of Verteporfin in mice limits the deposition of collagen and fibronectin, and the activation of FAPs during the development of fibrosis. Overall, our work provides robust evidence for considering YAP/TAZ as a potential target in muscular fibroproliferative disorders.NEW & NOTEWORTHY The understanding of the nuclear factors governing the differentiation of muscular fibro/adipogenic progenitors (FAPs) into myofibroblasts is in its infancy. Here, we comprehensively elucidate the status, regulation, and role of the mechanotransducers Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) in the muscular fibrotic process. Our findings reveal that inhibiting cellular mechanotransduction limits FAP differentiation and the extent of muscular fibrosis exerted by transforming growth factor type β (TGF-β). This research shed new lights on the molecular mechanisms dictating the cell fate of FAPs and the muscular fibrosis.
Collapse
Affiliation(s)
- Felipe S Gallardo
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Meilyn Cruz-Soca
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Alexia Bock-Pereda
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Jennifer Faundez-Contreras
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
- Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Cristian Gutiérrez-Rojas
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
- Escuela de Kinesiología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alessandro Gandin
- Department of Industrial Engineering, University of Padova and INSTM, Padova, Italy
| | - Veronica Torresan
- Department of Industrial Engineering, University of Padova and INSTM, Padova, Italy
| | - Juan Carlos Casar
- Department of Neurology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrea Ravasio
- Institute for Biological and Medical Engineering, School of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrique Brandan
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
- Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
18
|
Pan M, Zhang Y, Wright WC, Liu X, Passaia B, Currier D, Low J, Chapple RH, Steele JA, Connelly JP, Ju B, Plyler E, Lu M, Loughran AJ, Yang L, Abraham BJ, Pruett-Miller SM, Freeman B, Campbell GE, Dyer MA, Chen T, Stewart E, Koo S, Sheppard H, Easton J, Geeleher P. Bone morphogenetic protein (BMP) signaling determines neuroblastoma cell fate and sensitivity to retinoic acid. Nat Commun 2025; 16:2036. [PMID: 40021625 PMCID: PMC11871043 DOI: 10.1038/s41467-025-57185-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025] Open
Abstract
Retinoic acid (RA) is a standard-of-care neuroblastoma drug thought to be effective by inducing differentiation. Curiously, RA has little effect on primary human tumors during upfront treatment but can eliminate neuroblastoma cells from the bone marrow during post-chemo maintenance therapy-a discrepancy that has never been explained. To investigate this, we treat a large cohort of neuroblastoma cell lines with RA and observe that the most RA-sensitive cells predominantly undergo apoptosis or senescence, rather than differentiation. We conduct genome-wide CRISPR knockout screens under RA treatment, which identify bone morphogenic protein (BMP) signaling as controlling the apoptosis/senescence vs differentiation cell fate decision and determining RA's overall potency. We then discover that BMP signaling activity is markedly higher in neuroblastoma patient samples at bone marrow metastatic sites, providing a plausible explanation for RA's ability to clear neuroblastoma cells specifically from the bone marrow, by seemingly mimicking interactions between BMP and RA during normal development.
Collapse
Affiliation(s)
- Min Pan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Yinwen Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - William C Wright
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xueying Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Barbara Passaia
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Duane Currier
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jonathan Low
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Richard H Chapple
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jacob A Steele
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jon P Connelly
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Bensheng Ju
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Emily Plyler
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Meifen Lu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Allister J Loughran
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Brian J Abraham
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Burgess Freeman
- Preclinical Pharmacokinetic Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - George E Campbell
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Elizabeth Stewart
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Selene Koo
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Heather Sheppard
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Paul Geeleher
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
19
|
Wu B, Neupane J, Zhou Y, Zhang J, Chen Y, Surani MA, Zhang Y, Bao S, Li X. Stem cell-based embryo models: a tool to study early human development. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2741-1. [PMID: 39969747 DOI: 10.1007/s11427-024-2741-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/23/2024] [Indexed: 02/20/2025]
Abstract
How a mammalian fertilized egg acquires totipotency and develops into a full-term offspring is a fundamental scientific question. Human embryonic development is difficult to study due to limited resources, technical challenges and ethics. Moreover, the precise regulatory mechanism underlying early human embryonic development remains unknown. In recent years, the emergence of stem cell-based embryo models (SCBEM) provides the opportunity to reconstitute pre- to post-implantation development in vitro. These models to some extent mimic the embryo morphologically and transcriptionally, and thus may be used to study key events in mammalian pre- and post-implantation development. Many groups have successfully generated SCBEM of the mouse and human. Here, we provide a comparative review of the mouse and human SCBEM, discuss the capability of these models to mimic natural embryos and give a perspective on their potential future applications.
Collapse
Affiliation(s)
- Baojiang Wu
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Jitesh Neupane
- The Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Yang Zhou
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Jingcheng Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Yanglin Chen
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - M Azim Surani
- The Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Yong Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China.
| | - Siqin Bao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
| | - Xihe Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animals, Hohhot, 011517, China.
| |
Collapse
|
20
|
Zhao N, Nie X, Yan Y, Liu Z, Chen X, Shu P, Zhong J. α-arbutin prevents UVA-induced skin photodamage via alleviating DNA damage and collagen degradation in NIH-3T3 cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 263:113100. [PMID: 39787978 DOI: 10.1016/j.jphotobiol.2025.113100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/20/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Ultraviolet radiation (UV) causes certain side effects to the skin, and their accumulation to a certain extent can lead to accelerated aging of the skin. Recent studies suggest that α-arbutin may be useful in various disorders such as hyperpigmentation disorders, wound healing, and antioxidant activity. However, the role of α-arbutin in skin photodamage is unclear. In this study, under UVA-induced photodamage conditions, α-arbutin treated mouse skin fibroblasts (NIH-3T3) can repair DNA damage and resist apoptosis by reducing the production of reactive oxygen species (ROS) and increasing the phosphorylation of glycogen synthase kinase 3 beta (GSK3β) to orchestra AKT/GSK3β pathway. Meanwhile, α-arbutin can also regulate collagen metabolism and facilitate the replenishment of collagen by targeting the phosphorylation of SMAD3 to mediate the TGFβ/SMAD pathway in NIH-3T3. In conclusion, we found that α-arbutin can mitigate the detrimental effects of skin photodamage induced by UVA irradiation, and provides a theoretical basis for the use of α-arbutin in the treatment of skin photodamage.
Collapse
Affiliation(s)
- Nan Zhao
- HBN Research Institute and Biological Laboratory, Shenzhen Hujia Technology Co., Ltd., 518000 Shenzhen, Guangdong, PR China
| | - Xin Nie
- HBN Research Institute and Biological Laboratory, Shenzhen Hujia Technology Co., Ltd., 518000 Shenzhen, Guangdong, PR China
| | - Yizhen Yan
- HBN Research Institute and Biological Laboratory, Shenzhen Hujia Technology Co., Ltd., 518000 Shenzhen, Guangdong, PR China
| | - Zhao Liu
- HBN Research Institute and Biological Laboratory, Shenzhen Hujia Technology Co., Ltd., 518000 Shenzhen, Guangdong, PR China
| | - Xueqing Chen
- HBN Research Institute and Biological Laboratory, Shenzhen Hujia Technology Co., Ltd., 518000 Shenzhen, Guangdong, PR China
| | - Peng Shu
- HBN Research Institute and Biological Laboratory, Shenzhen Hujia Technology Co., Ltd., 518000 Shenzhen, Guangdong, PR China.
| | - Jiangming Zhong
- HBN Research Institute and Biological Laboratory, Shenzhen Hujia Technology Co., Ltd., 518000 Shenzhen, Guangdong, PR China.
| |
Collapse
|
21
|
Lei J, Chen J, Yu W, Wu Q, Jing S, Tang Y, Lin L, Hu M. Portrait of WWP1: the current state in human cancer. Front Cell Dev Biol 2025; 12:1516613. [PMID: 39949609 PMCID: PMC11821962 DOI: 10.3389/fcell.2024.1516613] [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: 10/24/2024] [Accepted: 12/31/2024] [Indexed: 02/16/2025] Open
Abstract
WWP1, a member of the C2-WW-HECT E3 ligase family, is an E3 ubiquitin-protein ligase containing WW domains. This enzyme plays a critical role in regulating diverse cellular processes. Its expression is modulated by various factors and non-coding RNAs, resulting in ubiquitination that affects substrate protein degradation. WWP1 demonstrates a dual function, acting predominantly as an oncogene in tumors but occasionally as a tumor suppressor. This review summarizes WWP1's biological roles, therapeutic potential in oncology, upstream regulatory factors, and downstream substrates. It aims to promote research on WWP1's antitumor effects, improve understanding of its role in tumorigenesis, and support the development of targeted therapies.
Collapse
Affiliation(s)
- Jiaming Lei
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Jun Chen
- The Central Hospital of Ezhou, Affiliated Hospital of Hubei University of Science and Technology, Ezhou, Hubei, China
| | - Wenwen Yu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Qing Wu
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shuang Jing
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yuanguang Tang
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Li Lin
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Meichun Hu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| |
Collapse
|
22
|
Dong DL, Jin GZ. Targeting Chondrocyte Hypertrophy as Strategies for the Treatment of Osteoarthritis. Bioengineering (Basel) 2025; 12:77. [PMID: 39851351 PMCID: PMC11760869 DOI: 10.3390/bioengineering12010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Osteoarthritis (OA) is a common joint disease characterized by pain and functional impairment, which severely impacts the quality of life of middle-aged and elderly individuals. During normal bone development, chondrocyte hypertrophy is a natural physiological process. However, in the progression of OA, chondrocyte hypertrophy becomes one of its key pathological features. Although there is no definitive evidence to date confirming that chondrocyte hypertrophy is the direct cause of OA, substantial experimental data indicate that it plays an important role in the disease's pathogenesis. In this review, we first explore the mechanisms underlying chondrocyte hypertrophy in OA and offer new insights. We then propose strategies for inhibiting chondrocyte hypertrophy from the perspectives of targeting signaling pathways and tissue engineering, ultimately envisioning the future prospects of OA treatment.
Collapse
Affiliation(s)
- Da-Long Dong
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea;
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea;
| |
Collapse
|
23
|
Cao D, Grünig E, Sirenko Y, Radchenko G, Gall H, Ahmed A, Theiß S, Lankeit M, Meder B, Laugsch M, Eichstaedt CA. SMAD5 as a novel gene for familial pulmonary arterial hypertension. Clin Sci (Lond) 2025; 139:15-27. [PMID: 39631055 DOI: 10.1042/cs20241340] [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: 07/10/2024] [Revised: 10/24/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024]
Abstract
Genetic diagnostic testing of 325 pulmonary arterial hypertension (PAH) patients using a PAH specific gene panel including 18 known PAH genes revealed mutations in 23%. Further PAH candidate genes were sequenced in the remaining patients exposing two SMAD5 variants, which were clinically and functionally characterized. We first recorded familial cosegregation and clinical parameters. Functional tests were performed following transient over-expression of the two SMAD5 variants in pulmonary artery smooth muscle cells (PASMCs). Expression of these variants was confirmed by quantitative PCR, Sanger sequencing, and Western blotting. Cell viability was evaluated using cell counting kit 8, cell proliferation by bromodeoxyuridine (BrdU), and apoptosis by annexin V assay. Both SMAD5 missense variants were absent in healthy controls and predicted to be pathogenic. The variant c.1175T>C p.(Leu392Pro) was identified in a heritable PAH patient and her healthy son. The mother had died of suspected PAH at age 42. The expression of this variant in PASMCs led to significantly higher cell viability due to higher proliferation in comparison with SMAD5 wild-type cells. The second variant c.277T>A p.(Trp93Arg) was identified in a patient with congenital heart disease associated PAH with a surgically repaired ventricular septal defect. Its expression led to significantly lower cell viability due to increased apoptosis in comparison with wild-type SMAD5 cells. Taking into account familial aggregation, clinical findings, and functional evidence, both variants could be classified as likely pathogenic. This is the first description of SMAD5 as a potential novel PAH gene for genetic diagnostic testing.
Collapse
Affiliation(s)
- Ding Cao
- Center for Pulmonary Hypertension, Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Ekkehard Grünig
- Center for Pulmonary Hypertension, Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Yuriy Sirenko
- Pulmonary Hypertension Center, SI NSC named after M.D. Starzhesko of NAMS of Ukraine, Kyiv, Ukraine
| | - Ganna Radchenko
- Pulmonary Hypertension Center, SI NSC named after M.D. Starzhesko of NAMS of Ukraine, Kyiv, Ukraine
| | - Henning Gall
- Department of Pneumology, Medical and Policlinic II, University Hospital of Giessen and Marburg, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
| | - Ayat Ahmed
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Susanne Theiß
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Mareike Lankeit
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Benjamin Meder
- Department of Internal Medicine III, Precision Digital Health, University of Heidelberg and Informatics for Life and German Center for Cardiovascular Research (DZHK), Heidelberg, Germany
| | - Magdalena Laugsch
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Christina A Eichstaedt
- Center for Pulmonary Hypertension, Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
24
|
Qian C, Dong G, Yang C, Zheng W, Zhong C, Shen Q, Lu Y, Zhao Y. Broadening horizons: molecular mechanisms and disease implications of endothelial-to-mesenchymal transition. Cell Commun Signal 2025; 23:16. [PMID: 39789529 PMCID: PMC11720945 DOI: 10.1186/s12964-025-02028-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is defined as an important process of cellular differentiation by which endothelial cells (ECs) are prone to lose their characteristics and transform into mesenchymal cells. During EndMT, reduced expression of endothelial adhesion molecules disrupts intercellular adhesion, triggering cytoskeletal reorganization and mesenchymal transition. Numerous studies have proved that EndMT is a multifaceted biological event driven primarily by cytokines such as TGF-β, TNF-α, and IL-1β, alongside signaling pathways like WNT, Smad, MEK-ERK, and Notch. Nevertheless, the exact roles of EndMT in complicated diseases have not been comprehensively reviewed. In this review, we summarize the predominant molecular regulatory mechanisms and signaling pathways that contribute to the development of EndMT, as well as highlight the contributions of a series of imperative non-coding RNAs in curbing the initiation of EndMT. Furthermore, we discuss the significant impact of EndMT on worsening vasculature-related diseases, including cancer, cardiovascular diseases, atherosclerosis, pulmonary vascular diseases, diabetes-associated fibrotic conditions, and cerebral cavernous malformation, providing the implications that targeting EndMT holds promise as a therapeutic strategy to mitigate disease progression.
Collapse
Affiliation(s)
- Cheng Qian
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guanglu Dong
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chunmei Yang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weiwei Zheng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chongjin Zhong
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiuhong Shen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yang Zhao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
25
|
Lee CM, Lee HY, Jarrell ZR, Smith MR, Jones DP, Go YM. Mechanistic role for mTORC1 signaling in profibrotic toxicity of low-dose cadmium. Toxicol Appl Pharmacol 2025; 494:117159. [PMID: 39557346 DOI: 10.1016/j.taap.2024.117159] [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: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Cadmium (Cd) is a toxic environmental metal that occurs naturally in food and drinking water. Cd is of increasing concern to human health due to its association with age-related diseases and long biological half-life. Previous studies show that low-dose Cd exposure via drinking water induces mechanistic target of rapamycin complex 1 (mTORC1) signaling in mice; however, the role of mTORC1 pathway in Cd-induced pro-fibrotic responses has not been established. In the present study, we used human lung fibroblasts to examine whether inhibiting the mTORC1 pathway prevents lung fibrosis signaling induced by low-dose Cd exposure. Results show that rapamycin, a pharmacological inhibitor of mTORC1, inhibited Cd-dependent phosphorylation of ribosomal protein S6, a downstream marker of mTORC1 activation. Rapamycin also decreased Cd-dependent increases in pro-fibrotic markers, α-smooth muscle actin, collagen 1α1 and fibronectin. Cd activated mitochondrial spare respiratory capacity in association with increased cell proliferation. Rapamycin decreased these responses, showing that mTORC1 signaling supports mitochondrial energy supply for cell proliferation, an important step in fibroblast trans-differentiation into myofibroblasts. Collectively, these results establish a key mechanistic role for mTORC1 activation in environmental Cd-dependent lung fibrosis.
Collapse
Affiliation(s)
- Choon-Myung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, United States of America
| | - Ho Young Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, United States of America
| | - Zachery R Jarrell
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, United States of America
| | - M Ryan Smith
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, United States of America; VA Healthcare System of Atlanta, Decatur, GA 30033, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, United States of America.
| | - Young-Mi Go
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, United States of America.
| |
Collapse
|
26
|
Geng Y, Bao C, Chen Y, Yan Z, Miao F, Wang T, Li Y, Li L, Sun W, Xu Y. NLRP3 deficiency improves bone healing of tooth extraction sockets through SMAD2/3-RUNX2-mediated osteoblast differentiation. Stem Cells 2024; 42:1085-1099. [PMID: 39404121 DOI: 10.1093/stmcls/sxae064] [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: 05/31/2024] [Accepted: 08/26/2024] [Indexed: 12/12/2024]
Abstract
Impaired bone healing following tooth extraction poses a significant challenge for implantation. As a crucial component of the natural immune system, the NLRP3 inflammasome is one of the most extensively studied pattern-recognition receptors, and is involved in multiple diseases. Yet, the role of NLRP3 in bone healing remains to be clarified. Here, to investigate the effect of NLRP3 on bone healing, we established a maxillary first molar extraction model in wild-type and NLRP3KO mice using minimally invasive techniques. We observed that NLRP3 was activated during the bone repair phase, and its depletion enhanced socket bone formation and osteoblast differentiation. Moreover, NLRP3 inflammasome activation was found to inhibit osteogenic differentiation in alveolar bone-derived mesenchymal stem cells (aBMSCs), an effect mitigated by NLRP3 deficiency. Mechanistically, we established that the SMAD2/3-RUNX2 signaling pathway is a downstream target of NLRP3 inflammasome activation, and SMAD2/3 knockdown partially reversed the significant decrease in expression of RUNX2, OSX, and ALP induced by NLRP3. Thus, our findings demonstrate that NLRP3 negatively modulates alveolar socket bone healing and contributes to the understanding of the NLRP3-induced signaling pathways involved in osteogenesis regulation.
Collapse
Affiliation(s)
- Ying Geng
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 212000, People's Republic of China
| | - Chen Bao
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
| | - Yue Chen
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
| | - Ziwei Yan
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
| | - Fen Miao
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
- Department of Stomatology, Children's Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
| | - Ting Wang
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
| | - Yingyi Li
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
| | - Lu Li
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 212000, People's Republic of China
| | - Wen Sun
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 212000, People's Republic of China
| | - Yan Xu
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 212000, People's Republic of China
| |
Collapse
|
27
|
Wu D, Sun X, Li X, Zuo Z, Yan D, Yin W. RRM2 Regulates Hepatocellular Carcinoma Progression Through Activation of TGF-β/Smad Signaling and Hepatitis B Virus Transcription. Genes (Basel) 2024; 15:1575. [PMID: 39766842 PMCID: PMC11675542 DOI: 10.3390/genes15121575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a type of malignant tumor with high morbidity and mortality. Untimely treatment and high recurrence are currently the major challenges for HCC. The identification of potential targets of HCC progression is crucial for the development of new therapeutic strategies. METHODS Bioinformatics analyses have been employed to discover genes that are differentially expressed in clinical cases of HCC. A variety of pharmacological methods, such as MTT, colony formation, EdU, Western blotting, Q-PCR, wound healing, Transwell, cytoskeleton F-actin filaments, immunohistochemistry (IHC), hematoxylin-eosin (HE) staining, and dual-luciferase reporter assay analyses, were utilized to study the pharmacological effects and potential mechanisms of ribonucleotide reductase regulatory subunit M2 (RRM2) in HCC. RESULTS RRM2 expression is significantly elevated in HCC, which is well correlated with poor clinical outcomes. Both in vitro and in vivo experiments demonstrated that RRM2 promoted HCC cell growth and metastasis. Mechanistically, RRM2 modulates the EMT phenotype of HCC, and further studies have shown that RRM2 facilitates the activation of the TGF-β/Smad signaling pathway. SB431542, an inhibitor of TGF-β signaling, significantly inhibited RRM2-induced cell migration. Furthermore, RRM2 expression was correlated with diminished survival in HBV-associated HCC patients. RRM2 knockdown decreased the levels of HBV RNA, pgRNA, cccDNA, and HBV DNA in HepG2.2.15 cells exhibiting sustained HBV infection, while RRM2 knockdown inhibited the activity of the HBV Cp, Xp, and SpI promoters. CONCLUSION RRM2 is involved in the progression of HCC by activating the TGF-β/Smad signaling pathway. RRM2 increases HBV transcription in HBV-expressing HCC cells. Targeting RRM2 may be of potential value in the treatment of HCC.
Collapse
Affiliation(s)
- Dandan Wu
- State Key Lab of Pharmaceutical Biotechnology (SKLPB), College of Life Sciences in Nanjing University (Xianlin Campus), Nanjing University, Nanjing 210046, China; (D.W.); (X.S.); (X.L.)
| | - Xinning Sun
- State Key Lab of Pharmaceutical Biotechnology (SKLPB), College of Life Sciences in Nanjing University (Xianlin Campus), Nanjing University, Nanjing 210046, China; (D.W.); (X.S.); (X.L.)
| | - Xin Li
- State Key Lab of Pharmaceutical Biotechnology (SKLPB), College of Life Sciences in Nanjing University (Xianlin Campus), Nanjing University, Nanjing 210046, China; (D.W.); (X.S.); (X.L.)
| | - Zongchao Zuo
- The First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China;
| | - Dong Yan
- Department of Cardiology, Affiliated Hospital of Nanjing University of TCM, Nanjing 210023, China;
| | - Wu Yin
- State Key Lab of Pharmaceutical Biotechnology (SKLPB), College of Life Sciences in Nanjing University (Xianlin Campus), Nanjing University, Nanjing 210046, China; (D.W.); (X.S.); (X.L.)
| |
Collapse
|
28
|
Zhou L, Wang N, Feng W, Liu X, Wu Q, Chen J, Jiao X, Ning X, Qi Z, Xu Z, Jiang X, Zhao Q. Soluble TGF-β decoy receptor TGFBR3 exacerbates Alzheimer's disease pathology by modifying microglial function. Glia 2024; 72:2201-2216. [PMID: 39137117 DOI: 10.1002/glia.24606] [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: 01/09/2024] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024]
Abstract
Alzheimer's disease (AD) is a major cause of progressive dementia characterized by memory loss and progressive neurocognitive dysfunction. However, the molecular mechanisms are not fully understood. To elucidate the molecular mechanism contributing to AD, an integrated analytical workflow was deployed to identify pivotal regulatory target within the RNA-sequencing (RNA-seq) data of the temporal cortex from AD patients. Soluble transforming growth factor beta receptor 3 (sTGFBR3) was identified as a critical target in AD, which was abnormally elevated in AD patients and AD mouse models. We then demonstrated that sTGFBR3 deficiency restored spatial learning and memory deficits in amyloid precursor protein (APP)/PS1 and streptozotocin (STZ)-induced neuronal impairment mice after its expression was disrupted by a lentiviral (LV) vector expressing shRNA. Mechanistically, sTGFBR3 deficiency augments TGF-β signaling and suppressing the NF-κB pathway, thereby reduced the number of disease-associated microglia (DAMs), inhibited proinflammatory activity and increased the phagocytic activity of DAMs. Moreover, sTGFBR3 deficiency significantly mitigated acute neuroinflammation provoked by lipopolysaccharide (LPS) and alleviated neuronal dysfunction induced by STZ. Collectively, these results position sTGFBR3 as a promising candidate for therapeutic intervention in AD.
Collapse
Affiliation(s)
- Lijun Zhou
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Nan Wang
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Wenzheng Feng
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Xin Liu
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Qiong Wu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Jiangxia Chen
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Xinming Jiao
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Xinyue Ning
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Zhentong Qi
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Zihua Xu
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Xiaowen Jiang
- College of Traditional Chinese Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Qingchun Zhao
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| |
Collapse
|
29
|
El Hamamy A, Iqbal Z, Le NM, Ranjan A, Zhang Y, Lin HW, Tan C, Sumani D, Patrizz A, McCullough LD, Li J. Targeted TGF-βR2 Silencing in the Retrotrapezoid Nucleus Mitigates Respiratory Dysfunction and Cognitive Decline in a Mouse Model of Cerebral Amyloid Angiopathy with and without Stroke. Transl Stroke Res 2024:10.1007/s12975-024-01306-0. [PMID: 39543011 DOI: 10.1007/s12975-024-01306-0] [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: 05/17/2024] [Revised: 10/07/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by the deposition of amyloid-beta peptides within cerebral blood vessels, leading to neurovascular complications. Ischemic strokes result from acute disruptions in cerebral blood flow, triggering metabolic disturbances and neurodegeneration. Both conditions often co-occur and are associated with respiratory dysfunctions. The retrotrapezoid nucleus (RTN), which is crucial for CO2 sensing and breathing regulation in the brainstem, may play a key role in breathing disorders seen in these conditions. This study aims to investigate the role of Transforming Growth Factor Beta (TGF-β) signaling in the RTN on respiratory and cognitive functions in CAA, both with and without concurrent ischemic stroke. Adult male Tg-SwDI (CAA model) mice and C57BL/6 wild-type controls underwent stereotaxic injections of lentivirus targeting TGF-βR2 in the RTN. Stroke was induced by middle cerebral artery occlusion using a monofilament. Respiratory functions were assessed using whole-body plethysmography, while cognitive functions were evaluated through the Barnes Maze and Novel Object Recognition Test (NORT). Immunohistochemical analysis was conducted to measure TGF-βR2 and GFAP expressions in the RTN. CAA mice exhibited significant respiratory dysfunctions, including reduced respiratory rates and increased apnea frequency, as well as impaired cognitive performance. TGF-βR2 silencing in the RTN improved respiratory functions and cognitive outcomes in CAA mice. In CAA mice with concurrent stroke, TGF-βR2 silencing similarly enhanced respiratory and cognitive functions. Immunohistochemistry confirmed reduced TGF-βR2 and GFAP expressions in the RTN following silencing. Our findings demonstrate that increased TGF-β signaling and gliosis in the RTN contribute to respiratory and cognitive dysfunctions in CAA and CAA with stroke. Targeting TGF-βR2 signaling in the RTN offers a promising therapeutic strategy to mitigate these impairments. This study is the first to report a causal link between brainstem gliosis and both respiratory and cognitive dysfunctions in CAA and stroke models.
Collapse
Affiliation(s)
- Ahmad El Hamamy
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zahid Iqbal
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ngoc Mai Le
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Arya Ranjan
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - YuXing Zhang
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hung Wen Lin
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chunfeng Tan
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Destiny Sumani
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anthony Patrizz
- Department of Neurological Surgery and Brain Repair, University of South Florida at Tampa, Tampa, FL, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jun Li
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
30
|
Papismadov N, Levi N, Roitman L, Agrawal A, Ovadya Y, Cherqui U, Yosef R, Akiva H, Gal H, Krizhanovsky V. p21 regulates expression of ECM components and promotes pulmonary fibrosis via CDK4 and Rb. EMBO J 2024; 43:5360-5380. [PMID: 39349844 PMCID: PMC11574164 DOI: 10.1038/s44318-024-00246-7] [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: 05/21/2024] [Revised: 08/05/2024] [Accepted: 09/05/2024] [Indexed: 11/20/2024] Open
Abstract
Fibrosis and accumulation of senescent cells are common tissue changes associated with aging. Here, we show that the CDK inhibitor p21 (CDKN1A), known to regulate the cell cycle and the viability of senescent cells, also controls the expression of extracellular matrix (ECM) components in senescent and proliferating cells of the fibrotic lung, in a manner dependent on CDK4 and Rb phosphorylation. p21 knockout protects mice from the induction of lung fibrosis. Moreover, inducible p21 silencing during fibrosis development alleviates disease pathology, decreasing the inflammatory response and ECM accumulation in the lung, and reducing the amount of senescent cells. Furthermore, p21 silencing limits fibrosis progression even when introduced during disease development. These findings show that one common mechanism regulates both cell cycle progression and expression of ECM components, and suggest that targeting p21 might be a new approach for treating age-related fibrotic pathologies.
Collapse
Affiliation(s)
- Nurit Papismadov
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Naama Levi
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Lior Roitman
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Amit Agrawal
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Yossi Ovadya
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Ulysse Cherqui
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Reut Yosef
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Hagay Akiva
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Hilah Gal
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Valery Krizhanovsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel.
| |
Collapse
|
31
|
Baek JS, Lee JH, Kim JH, Cho SS, Kim YS, Yang JH, Shin EJ, Kang HG, Kim SJ, Ahn SG, Park EY, Baek DJ, Yim SK, Kang KW, Ki SH, Kim KM. An inducible sphingosine kinase 1 in hepatic stellate cells potentiates liver fibrosis. Biochem Pharmacol 2024; 229:116520. [PMID: 39236934 DOI: 10.1016/j.bcp.2024.116520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
Hepatic stellate cells (HSCs) play a role in hepatic fibrosis and sphingosine kinase (SphK) is involved in biological processes. As studies on the regulatory mechanisms and functions of SphK in HSCs during liver fibrosis are currently limited, this study aimed to elucidate the regulatory mechanism and connected pathways of SphK upon HSC activation. The expression of SphK1 was higher in HSCs than in hepatocytes, and upregulated in activated primary HSCs. SphK1 was also increased in liver homogenates of carbon tetrachloride-treated or bile duct ligated mice and in transforming growth factor-β (TGF-β)-treated LX-2 cells. TGF-β-mediated SphK1 induction was due to Smad3 signaling in LX-2 cells. SphK1 modulation altered the expression of liver fibrogenesis-related genes. This SphK1-mediated profibrogenic effect was dependent on SphK1/sphingosine-1-phosphate/sphingosine-1-phosphate receptor signaling through ERK. Epigallocatechin gallate blocked TGF-β-induced SphK1 expression and hepatic fibrogenesis by attenuating Smad and MAPK activation. SphK1 induced by TGF-β facilitates HSC activation and liver fibrogenesis, which is reversed by epigallocatechin gallate. Accordingly, SphK1 and related signal transduction may be utilized to treat liver fibrosis.
Collapse
Affiliation(s)
- Jin Sol Baek
- MRC-OSTRC, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju 61452, South Korea
| | - Ji Hyun Lee
- MRC-OSTRC, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju 61452, South Korea
| | - Ji Hye Kim
- MRC-OSTRC, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju 61452, South Korea
| | - Sam Seok Cho
- MRC-OSTRC, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju 61452, South Korea; Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju 61452, Republic of Korea; Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju 61452, Republic of Korea
| | - Yun Seok Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Pharmacy, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do 58245, Republic of Korea
| | - Eun Jin Shin
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju 61452, Republic of Korea; Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea; Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju 61452, Republic of Korea
| | - Hyeon-Gu Kang
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju 61452, Republic of Korea; Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea
| | - Seok-Jun Kim
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju 61452, Republic of Korea; Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea; Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju 61452, Republic of Korea
| | - Sang-Gun Ahn
- Department of Pathology, School of Dentistry, Chosun University, Gwangju 61452, Republic of Korea
| | - Eun Young Park
- College of Pharmacy, Mokpo National University, Muan-gun, Jeollanam-do 58554, Republic of Korea
| | - Dong Jae Baek
- College of Pharmacy, Mokpo National University, Muan-gun, Jeollanam-do 58554, Republic of Korea
| | - Sung-Kun Yim
- Marine Biotechnology Research Center, Jeonnam Bioindustry Foundation, 21-7, Nonggongdanji 4Gil, Wando-eup, Wando-gun, Jeollanam-do 59108, Republic of Korea
| | - Keon Wook Kang
- Department of Pharmacy, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sung Hwan Ki
- MRC-OSTRC, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju 61452, South Korea
| | - Kyu Min Kim
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju 61452, Republic of Korea; Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea; Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju 61452, Republic of Korea.
| |
Collapse
|
32
|
Lee JH, Sánchez-Rivera FJ, He L, Basnet H, Chen FX, Spina E, Li L, Torner C, Chan JE, Yarlagadda DVK, Park JS, Sussman C, Rudin CM, Lowe SW, Tammela T, Macias MJ, Koche RP, Massagué J. TGF-β and RAS jointly unmask primed enhancers to drive metastasis. Cell 2024; 187:6182-6199.e29. [PMID: 39243762 PMCID: PMC12035776 DOI: 10.1016/j.cell.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/08/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024]
Abstract
Epithelial-to-mesenchymal transitions (EMTs) and extracellular matrix (ECM) remodeling are distinct yet important processes during carcinoma invasion and metastasis. Transforming growth factor β (TGF-β) and RAS, signaling through SMAD and RAS-responsive element-binding protein 1 (RREB1), jointly trigger expression of EMT and fibrogenic factors as two discrete arms of a common transcriptional response in carcinoma cells. Here, we demonstrate that both arms come together to form a program for lung adenocarcinoma metastasis and identify chromatin determinants tying the expression of the constituent genes to TGF-β and RAS inputs. RREB1 localizes to H4K16acK20ac marks in histone H2A.Z-loaded nucleosomes at enhancers in the fibrogenic genes interleukin-11 (IL11), platelet-derived growth factor-B (PDGFB), and hyaluronan synthase 2 (HAS2), as well as the EMT transcription factor SNAI1, priming these enhancers for activation by a SMAD4-INO80 nucleosome remodeling complex in response to TGF-β. These regulatory properties segregate the fibrogenic EMT program from RAS-independent TGF-β gene responses and illuminate the operation and vulnerabilities of a bifunctional program that promotes metastatic outgrowth.
Collapse
Affiliation(s)
- Jun Ho Lee
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Francisco J Sánchez-Rivera
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Lan He
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Harihar Basnet
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Fei Xavier Chen
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elena Spina
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Liangji Li
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carles Torner
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Jason E Chan
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dig Vijay Kumar Yarlagadda
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Tri-Institutional Graduate Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jin Suk Park
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carleigh Sussman
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tuomas Tammela
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maria J Macias
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona 08028, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
33
|
Mogollón García HD, de Andrade Ferrazza R, Ochoa JC, de Athayde FF, Vidigal PMP, Wiltbank M, Kastelic JP, Sartori R, Ferreira JCP. Landscape transcriptomic analysis of bovine follicular cells during key phases of ovarian follicular development. Biol Res 2024; 57:76. [PMID: 39468655 PMCID: PMC11514973 DOI: 10.1186/s40659-024-00558-2] [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: 05/24/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND There are many gaps in our understanding of the mechanisms involved in ovarian follicular development in cattle, particularly regarding follicular deviation, acquisition of ovulatory capacity, and preovulatory changes. Molecular evaluations of ovarian follicular cells during follicular development in cattle, especially serial transcriptomic analyses across key growth phases, have not been reported. This study aims to address this gap by analyzing gene expression using RNA-seq in granulosa and antral cells recovered from ovarian follicular fluid during critical phases of ovarian follicular development in Holstein cows. RESULTS Integrated analysis of gene ontology (GO), gene set enrichment (GSEA), protein-protein interaction (PPI), and gene topology identified that differentially expressed genes (DEGs) in the largest ovarian follicles at deviation (Dev) were primarily involved in FSH-negative feedback, steroidogenesis, cell proliferation, apoptosis, and the prevention of early follicle rupture. In contrast, DEGs in the second largest follicles (DevF2) were mainly related to loss of cell viability, apoptosis, and immune cell invasion. In the dominant (PostDev) and preovulatory (PreOv) follicles, DEGs were associated with vascular changes and inflammatory responses. CONCLUSIONS The transcriptome of ovarian follicular fluid cells had a predominance of granulosa cells in the dominant follicle at deviation, with upregulation of genes involved in cell viability, steroidogenesis, and apoptosis prevention, whereas in the non-selected follicle there was upregulation of cell death-related transcripts. Immune cell transcripts increased significantly after deviation, particularly in preovulatory follicles, indicating strong intrafollicular chemotactic activity. We inferred that immune cell invasion occurred despite an intact basal lamina, contributing to follicular maturation.
Collapse
Affiliation(s)
- Henry David Mogollón García
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil
- Department of Genetic, Evolution, Microbiology and Immunology. Biology Institute, Campinas State University, Campinas, São Paulo, Brazil
- Computational Systems Biology Laboratory (CSBL), Institut Pasteur, University of São Paulo (USP), São Paulo, Brazil
| | | | - Julian Camilo Ochoa
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil
| | - Flávia Florencio de Athayde
- Department of Animal Production and Health, School of Veterinary Medicine, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | | | - Milo Wiltbank
- Department of Animal & Dairy Sciences, University of Wisconsin-Madison, Madison, USA
| | | | - Roberto Sartori
- Department of Animal Science, Luiz de Queiroz College of Agriculture (ESALQ), University of São Paulo, Piracicaba, São Paulo, Brazil
| | - João Carlos Pinheiro Ferreira
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil.
| |
Collapse
|
34
|
Miyoshi Y, Lucena-Cacace A, Tian Y, Matsumura Y, Tani K, Nishikawa M, Narita M, Kimura T, Ono K, Yoshida Y. SMAD3 mediates the specification of human induced pluripotent stem cell-derived epicardium into progenitors for the cardiac pericyte lineage. Stem Cell Reports 2024; 19:1399-1416. [PMID: 39332407 PMCID: PMC11561457 DOI: 10.1016/j.stemcr.2024.08.008] [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: 03/26/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/29/2024] Open
Abstract
Understanding the molecular mechanisms of epicardial epithelial-to-mesenchymal transition (EMT), particularly in directing cell fate toward epicardial derivatives, is crucial for regenerative medicine using human induced pluripotent stem cell (iPSC)-derived epicardium. Although transforming growth factor β (TGF-β) plays a pivotal role in epicardial biology, orchestrating EMT during embryonic development via downstream signaling through SMAD proteins, the function of SMAD proteins in the epicardium in maintaining vascular homeostasis or mediating the differentiation of various epicardial-derived cells (EPDCs) is not yet well understood. Our study reveals that TGF-β-independent SMAD3 expression autonomously predicts epicardial cell specification and lineage maintenance, acting as a key mediator in promoting the angiogenic-oriented specification of the epicardium into cardiac pericyte progenitors. This finding uncovers a novel role for SMAD3 in the human epicardium, particularly in generating cardiac pericyte progenitors that enhance cardiac microvasculature angiogenesis. This insight opens new avenues for leveraging epicardial biology in developing more effective cardiac regeneration strategies.
Collapse
Affiliation(s)
- Yutaro Miyoshi
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Antonio Lucena-Cacace
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan.
| | - Yu Tian
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Yasuko Matsumura
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Kanae Tani
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Misato Nishikawa
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Megumi Narita
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yoshinori Yoshida
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
35
|
Zhang J, Jiang Y, Zhang Z, Li S, Fan H, Gu J, Mao R, Xu X. Repulsive guidance molecules b (RGMb): molecular mechanism, function and role in diseases. Expert Rev Mol Med 2024; 26:e24. [PMID: 39375839 PMCID: PMC11488336 DOI: 10.1017/erm.2024.24] [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: 08/26/2023] [Revised: 12/23/2023] [Accepted: 06/11/2024] [Indexed: 10/09/2024]
Abstract
Repulsive guidance molecule b (RGMb), a glycosylphosphatidylinositol-anchored member of the RGM family, is initially identified as a co-receptor of bone morphogenetic protein (BMP) in the nervous system. The expression of RGMb is transcriptionally regulated by dorsal root ganglion 11 (DRG11), which is a transcription factor expressed in embryonic DRG and dorsal horn neurons and plays an important role in the development of sensory circuits. RGMb is involved in important physiological processes such as embryonic development, immune response, intercellular adhesion and tumorigenesis. Furthermore, RGMb is mainly involved in the regulation of RGMb-neogenin-Rho and BMP signalling pathways. The recent discovery of programmed death-ligand 2 (PD-L2)-RGMb binding reveals that the cell signalling network and functional regulation centred on RGMb are extremely complex. The latest report suggests that down-regulation of the PD-L2-RGMb pathway in the gut microbiota promotes an anti-tumour immune response, which defines a potentially effective immune strategy. However, the biological function of RGMb in a variety of human diseases has not been fully determined, and will remain an active research field. This article reviews the properties and functions of RGMb, focusing on its role under various physiological and pathological conditions.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Yijing Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Zijian Zhang
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Shilin Li
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Haowen Fan
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jinhua Gu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Xiaohong Xu
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong University, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
36
|
Luo X, Ni X, Zhi J, Jiang X, Bai R. Small molecule agents against alopecia: Potential targets and related pathways. Eur J Med Chem 2024; 276:116666. [PMID: 39002436 DOI: 10.1016/j.ejmech.2024.116666] [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: 05/14/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Alopecia has emerged as a global concern, extending beyond the middle-aged and elderly population and increasingly affecting younger individuals. Despite its growing prevalence, the treatment options and effective drugs for alopecia remain limited due to the incomplete understanding of its underlying mechanisms. Therefore, it is urgent to explore the pathogenesis of alopecia and discover novel and safer therapeutic agents. This review provided an overview of the prevailing clinical disorders of alopecia, and the key pathways and targets involved in hair growth process. Additionally, it discusses FDA-approved drugs and clinical candidates for the treatment of alopecia, and explores small molecule compounds with anti-alopecia potential in the drug discovery phase. These endeavors are expected to provide researchers with valuable scientific insights and practical information for anti-alopecia drug discovery.
Collapse
Affiliation(s)
- Xinyu Luo
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Xinhua Ni
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Jia Zhi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Xiaoying Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China.
| |
Collapse
|
37
|
Geng W, An J, Dong K, Zhang H, Zhang X, Liu Y, Xu R, Liu Y, Huang X, Song H, Yan W, Sun A, He F, Wang J, Gao H, Tian C. ZNF8 Orchestrates with Smad3 to Promote Lung Metastasis by Recruiting SMYD3 in Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404904. [PMID: 39225541 PMCID: PMC11515916 DOI: 10.1002/advs.202404904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/03/2024] [Indexed: 09/04/2024]
Abstract
Most deaths in breast cancer patients are attributed to metastasis, and lung metastasis is associated with a particularly poor prognosis; therefore it is imperative to identify potential target for intervention. The transforming growth factor-β (TGF-β) pathway plays a vital role in breast cancer metastasis, in which Smad3 is the key mediator and performs specific functions by binding with different cofactors. However, Smad3 cofactors involved in lung metastasis have not yet been identified. This study first establishes the interactome of Smad3 in breast cancer cells and identifies ZNF8 as a novel Smad3 cofactor. Furthermore, the results reveal that ZNF8 is closely associated with breast cancer lung metastasis prognosis, and specifically facilitates TGF-β pathway-mediated breast cancer lung metastasis by participating in multiple processes. Mechanistically, ZNF8 binds with Smad3 to enhance the H3K4me3 modification and promote the expression of lung metastasis signature genes by recruiting SMYD3. SMYD3 inhibition by BCI121 effectively prevents ZNF8-mediated lung metastasis. Overall, the study identifies a novel cofactor of TGF-β/Smad3 that promotes lung metastasis in breast cancer and introduces potential therapeutic strategies for the early management of breast cancer lung metastasis.
Collapse
Affiliation(s)
- Wenwen Geng
- Department of Breast SurgeryQilu Hospital (Qingdao)Cheeloo College of MedicineShandong UniversityQingdaoShandong266000China
- Laboratory of OncologyQilu Hospital (Qingdao)Cheeloo College of MedicineShandong UniversityQingdaoShandong266000China
| | - Junhua An
- Department of Breast SurgeryQilu Hospital (Qingdao)Cheeloo College of MedicineShandong UniversityQingdaoShandong266000China
- Laboratory of OncologyQilu Hospital (Qingdao)Cheeloo College of MedicineShandong UniversityQingdaoShandong266000China
| | - Ke Dong
- Department of Breast SurgeryQilu Hospital (Qingdao)Cheeloo College of MedicineShandong UniversityQingdaoShandong266000China
- Laboratory of OncologyQilu Hospital (Qingdao)Cheeloo College of MedicineShandong UniversityQingdaoShandong266000China
| | - Hailu Zhang
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
- College of Life SciencesHebei UniversityBaodingHebei071002China
| | - Xiuyuan Zhang
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Yuchen Liu
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Rong Xu
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Yifan Liu
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Xiaofen Huang
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
- College of Life SciencesHebei UniversityBaodingHebei071002China
| | - Haiyun Song
- Department of PathologyQilu Hospital (Qingdao)Cheeloo College of MedicineShandong UniversityQingdaoShandong266000China
| | - Wei Yan
- The First Medical Center of Chinese PLA General HospitalBeijing100036China
| | - Aihua Sun
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
- College of Life SciencesHebei UniversityBaodingHebei071002China
- Research Unit of Proteomics Dirven Cancer Precision MedicineChinese Academy of Medical SciencesBeijing102206China
| | - Fuchu He
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
- Research Unit of Proteomics Dirven Cancer Precision MedicineChinese Academy of Medical SciencesBeijing102206China
| | - Jian Wang
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
- College of Life SciencesHebei UniversityBaodingHebei071002China
| | - Haidong Gao
- Department of Breast SurgeryQilu Hospital (Qingdao)Cheeloo College of MedicineShandong UniversityQingdaoShandong266000China
- Laboratory of OncologyQilu Hospital (Qingdao)Cheeloo College of MedicineShandong UniversityQingdaoShandong266000China
| | - Chunyan Tian
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
- College of Life SciencesHebei UniversityBaodingHebei071002China
- Research Unit of Proteomics Dirven Cancer Precision MedicineChinese Academy of Medical SciencesBeijing102206China
| |
Collapse
|
38
|
Chen S, Hayoun-Neeman D, Nagar M, Pinyan S, Hadad L, Yaacobov L, Alon L, Shachar LE, Swissa T, Kryukov O, Gershoni-Yahalom O, Rosental B, Cohen S, Lichtenstein RG. Terminal α1,2-fucosylation of glycosphingolipids by FUT1 is a key regulator in early cell-fate decisions. EMBO Rep 2024; 25:4433-4464. [PMID: 39256596 PMCID: PMC11467398 DOI: 10.1038/s44319-024-00243-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024] Open
Abstract
The embryonic cell surface is rich in glycosphingolipids (GSLs), which change during differentiation. The reasons for GSL subgroup variation during early embryogenesis remain elusive. By combining genomic approaches, flow cytometry, confocal imaging, and transcriptomic data analysis, we discovered that α1,2-fucosylated GSLs control the differentiation of human pluripotent cells (hPCs) into germ layer tissues. Overexpression of α1,2-fucosylated GSLs disrupts hPC differentiation into mesodermal lineage and reduces differentiation into cardiomyocytes. Conversely, reducing α1,2-fucosylated groups promotes hPC differentiation and mesoderm commitment in response to external signals. We find that bone morphogenetic protein 4 (BMP4), a mesodermal gene inducer, suppresses α1,2-fucosylated GSL expression. Overexpression of α1,2-fucosylated GSLs impairs SMAD activation despite BMP4 presence, suggesting α-fucosyl end groups as BMP pathway regulators. Additionally, the absence of α1,2-fucosylated GSLs in early/late mesoderm and primitive streak stages in mouse embryos aligns with the hPC results. Thus, α1,2-fucosylated GSLs may regulate early cell-fate decisions and embryo development by modulating cell signaling.
Collapse
Affiliation(s)
- Saray Chen
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Dana Hayoun-Neeman
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Michal Nagar
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Sapir Pinyan
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Limor Hadad
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Liat Yaacobov
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Lilach Alon
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Liraz Efrat Shachar
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Tair Swissa
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Olga Kryukov
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Orly Gershoni-Yahalom
- Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Benyamin Rosental
- Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Smadar Cohen
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- The Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Rachel G Lichtenstein
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel.
- Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel.
| |
Collapse
|
39
|
Skubic C, Trček H, Nassib P, Kreft T, Walakira A, Pohar K, Petek S, Režen T, Ihan A, Rozman D. Knockouts of CYP51A1, DHCR24, or SC5D from cholesterol synthesis reveal pathways modulated by sterol intermediates. iScience 2024; 27:110651. [PMID: 39262789 PMCID: PMC11387598 DOI: 10.1016/j.isci.2024.110651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/20/2024] [Accepted: 07/31/2024] [Indexed: 09/13/2024] Open
Abstract
Sterols from cholesterol synthesis are crucial for cholesterol production, but also have individual roles difficult to assess in vivo due to essentiality of cholesterol. We developed HepG2 cell models with knockouts (KOs) for three enzymes of cholesterol synthesis, each accumulating specific sterols. Surprisingly, KOs of CYP51, DHCR24, and SC5D shared only 9% of differentially expressed genes. The most striking was the phenotype of CYP51 KO with highly elevated lanosterol and 24,25-dihydrolanosterol, significant increase in G2+M phase and enhanced cancer and cell cycle pathways. Comparisons with mouse liver Cyp51 KO data suggest 24,25-dihydrolanosterol activates similar cell proliferation pathways, possibly via elevated LEF1 and WNT/NFKB signaling. In contrast, SC5D and DHCR24 KO cells with elevated lathosterol or desmosterol proliferated slowly, with downregulated E2F, mitosis, and enriched HNF1A. These findings demonstrate that increase of lanosterol and 24,25-dihydrolanosterol, but not other sterols, promotes cell proliferation in hepatocytes.
Collapse
Affiliation(s)
- Cene Skubic
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Hana Trček
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Petra Nassib
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Tinkara Kreft
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Andrew Walakira
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Katka Pohar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Sara Petek
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Tadeja Režen
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Alojz Ihan
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Damjana Rozman
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| |
Collapse
|
40
|
Bustamante A, Baritaki S, Zaravinos A, Bonavida B. Relationship of Signaling Pathways between RKIP Expression and the Inhibition of EMT-Inducing Transcription Factors SNAIL1/2, TWIST1/2 and ZEB1/2. Cancers (Basel) 2024; 16:3180. [PMID: 39335152 PMCID: PMC11430682 DOI: 10.3390/cancers16183180] [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: 07/25/2024] [Revised: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Untreated primary carcinomas often lead to progression, invasion and metastasis, a process that involves the epithelial-to-mesenchymal transition (EMT). Several transcription factors (TFs) mediate the development of EMT, including SNAIL1/SNAIL2, TWIST1/TWIST2 and ZEB1/ZEB2, which are overexpressed in various carcinomas along with the under expression of the metastasis suppressor Raf Kinase Inhibitor Protein (RKIP). Overexpression of RKIP inhibits EMT and the above associated TFs. We, therefore, hypothesized that there are inhibitory cross-talk signaling pathways between RKIP and these TFs. Accordingly, we analyzed the various properties and biomarkers associated with the epithelial and mesenchymal tissues and the various molecular signaling pathways that trigger the EMT phenotype such as the TGF-β, the RTK and the Wnt pathways. We also presented the various functions and the transcriptional, post-transcriptional and epigenetic regulations for the expression of each of the EMT TFs. Likewise, we describe the transcriptional, post-transcriptional and epigenetic regulations of RKIP expression. Various signaling pathways mediated by RKIP, including the Raf/MEK/ERK pathway, inhibit the TFs associated with EMT and the stabilization of epithelial E-Cadherin expression. The inverse relationship between RKIP and the TF expressions and the cross-talks were further analyzed by bioinformatic analysis. High mRNA levels of RKIP correlated negatively with those of SNAIL1, SNAIL2, TWIST1, TWIST2, ZEB1, and ZEB2 in several but not all carcinomas. However, in these carcinomas, high levels of RKIP were associated with good prognosis, whereas high levels of the above transcription factors were associated with poor prognosis. Based on the inverse relationship between RKIP and EMT TFs, it is postulated that the expression level of RKIP in various carcinomas is clinically relevant as both a prognostic and diagnostic biomarker. In addition, targeting RKIP induction by agonists, gene therapy and immunotherapy will result not only in the inhibition of EMT and metastases in carcinomas, but also in the inhibition of tumor growth and reversal of resistance to various therapeutic strategies. However, such targeting strategies must be better investigated as a result of tumor heterogeneities and inherent resistance and should be better adapted as personalized medicine.
Collapse
Affiliation(s)
- Andrew Bustamante
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Stavroula Baritaki
- Laboratory of Experimental Oncology, Division of Surgery, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Apostolos Zaravinos
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus
| | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
41
|
Shaikh R, Larson NJ, Kam J, Hanjaya-Putra D, Zartman J, Umulis DM, Li L, Reeves GT. Optimal performance objectives in the highly conserved bone morphogenetic protein signaling pathway. NPJ Syst Biol Appl 2024; 10:103. [PMID: 39277657 PMCID: PMC11401948 DOI: 10.1038/s41540-024-00430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/21/2024] [Indexed: 09/17/2024] Open
Abstract
Throughout development, complex networks of cell signaling pathways drive cellular decision-making across different tissues and contexts. The transforming growth factor β (TGF-β) pathways, including the BMP/Smad pathway, play crucial roles in determining cellular responses. However, as the Smad pathway is used reiteratively throughout the life cycle of all animals, its systems-level behavior varies from one context to another, despite the pathway connectivity remaining nearly constant. For instance, some cellular systems require a rapid response, while others require high noise filtering. In this paper, we examine how the BMP-Smad pathway balances trade-offs among three such systems-level behaviors, or "Performance Objectives (POs)": response speed, noise amplification, and the sensitivity of pathway output to receptor input. Using a Smad pathway model fit to human cell data, we show that varying non-conserved parameters (NCPs) such as protein concentrations, the Smad pathway can be tuned to emphasize any of the three POs and that the concentration of nuclear phosphatase has the greatest effect on tuning the POs. However, due to competition among the POs, the pathway cannot simultaneously optimize all three, but at best must balance trade-offs among the POs. We applied the multi-objective optimization concept of the Pareto Front, a widely used concept in economics to identify optimal trade-offs among various requirements. We show that the BMP pathway efficiently balances competing POs across species and is largely Pareto optimal. Our findings reveal that varying the concentration of NCPs allows the Smad signaling pathway to generate a diverse range of POs. This insight identifies how signaling pathways can be optimally tuned for each context.
Collapse
Affiliation(s)
- Razeen Shaikh
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, TX, USA
| | - Nissa J Larson
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Jayden Kam
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, TX, USA
| | - Donny Hanjaya-Putra
- Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA
| | - Jeremiah Zartman
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA
- Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - David M Umulis
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| | - Linlin Li
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| | - Gregory T Reeves
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, TX, USA.
- Faculty of Genetics and Genomics, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
42
|
Wood KA, Tong RS, Motta M, Cordeddu V, Scimone ER, Bush SJ, Maxwell DW, Giannoulatou E, Caputo V, Traversa A, Mancini C, Ferrero GB, Benedicenti F, Grammatico P, Melis D, Steindl K, Brunetti-Pierri N, Trevisson E, Wilkie AO, Lin AE, Cormier-Daire V, Twigg SR, Tartaglia M, Goriely A. SMAD4 mutations causing Myhre syndrome are under positive selection in the male germline. Am J Hum Genet 2024; 111:1953-1969. [PMID: 39116879 PMCID: PMC11444041 DOI: 10.1016/j.ajhg.2024.07.006] [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: 06/07/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
While it is widely thought that de novo mutations (DNMs) occur randomly, we previously showed that some DNMs are enriched because they are positively selected in the testes of aging men. These "selfish" mutations cause disorders with a shared presentation of features, including exclusive paternal origin, significant increase of the father's age, and high apparent germline mutation rate. To date, all known selfish mutations cluster within the components of the RTK-RAS-MAPK signaling pathway, a critical modulator of testicular homeostasis. Here, we demonstrate the selfish nature of the SMAD4 DNMs causing Myhre syndrome (MYHRS). By analyzing 16 informative trios, we show that MYHRS-causing DNMs originated on the paternally derived allele in all cases. We document a statistically significant epidemiological paternal age effect of 6.3 years excess for fathers of MYHRS probands. We developed an ultra-sensitive assay to quantify spontaneous MYHRS-causing SMAD4 variants in sperm and show that pathogenic variants at codon 500 are found at elevated level in sperm of most men and exhibit a strong positive correlation with donor's age, indicative of a high apparent germline mutation rate. Finally, we performed in vitro assays to validate the peculiar functional behavior of the clonally selected DNMs and explored the basis of the pathophysiology of the different SMAD4 sperm-enriched variants. Taken together, these data provide compelling evidence that SMAD4, a gene operating outside the canonical RAS-MAPK signaling pathway, is associated with selfish spermatogonial selection and raises the possibility that other genes/pathways are under positive selection in the aging human testis.
Collapse
Affiliation(s)
- Katherine A Wood
- MRC Weatherall Institute of Molecular Medicine, Oxford OX39DS, UK; Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX39DS, UK; NIHR Oxford Biomedical Research Centre, Oxford OX39DU, UK
| | - R Spencer Tong
- MRC Weatherall Institute of Molecular Medicine, Oxford OX39DS, UK; Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX39DS, UK; NIHR Oxford Biomedical Research Centre, Oxford OX39DU, UK
| | - Marialetizia Motta
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Viviana Cordeddu
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Eleanor R Scimone
- Medical Genetics, Mass General Brigham, Harvard Medical School, Harvard University, Boston, MA 02114, USA
| | - Stephen J Bush
- MRC Weatherall Institute of Molecular Medicine, Oxford OX39DS, UK; Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX39DS, UK; NIHR Oxford Biomedical Research Centre, Oxford OX39DU, UK
| | - Dale W Maxwell
- MRC Weatherall Institute of Molecular Medicine, Oxford OX39DS, UK; Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX39DS, UK; NIHR Oxford Biomedical Research Centre, Oxford OX39DU, UK
| | - Eleni Giannoulatou
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, NSW 2010, Australia; School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Viviana Caputo
- Department of Experimental Medicine, Sapienza University, 00161 Rome, Italy
| | - Alice Traversa
- Department of Experimental Medicine, Sapienza University, 00161 Rome, Italy
| | - Cecilia Mancini
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Giovanni B Ferrero
- Department of Clinical and Biological Science, University of Torino, 10126 Turin, Italy
| | | | - Paola Grammatico
- Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, 00152 Rome, Italy
| | - Daniela Melis
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Salerno, Italy
| | - Katharina Steindl
- Institute of Medical Genetics, University of Zurich, 8952 Schlieren-Zurich, Switzerland
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, 80131 Naples, Italy; Telethon Institute of Genetics and Medicine, Pozzuoli, Italy; Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, University of Naples Federico II, Naples, Italy
| | - Eva Trevisson
- Department of Women's and Children's Health, University of Padova, 35128 Padua, Italy
| | - Andrew Om Wilkie
- MRC Weatherall Institute of Molecular Medicine, Oxford OX39DS, UK; Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX39DS, UK; NIHR Oxford Biomedical Research Centre, Oxford OX39DU, UK
| | - Angela E Lin
- Medical Genetics, Mass General Brigham, Harvard Medical School, Harvard University, Boston, MA 02114, USA
| | - Valerie Cormier-Daire
- Université Paris Cité, Service de Médecine Génomique des Maladies Rares, INSERM UMR 1163, Institut Imagine, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Stephen Rf Twigg
- MRC Weatherall Institute of Molecular Medicine, Oxford OX39DS, UK; Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX39DS, UK; NIHR Oxford Biomedical Research Centre, Oxford OX39DU, UK
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy.
| | - Anne Goriely
- MRC Weatherall Institute of Molecular Medicine, Oxford OX39DS, UK; Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX39DS, UK; NIHR Oxford Biomedical Research Centre, Oxford OX39DU, UK.
| |
Collapse
|
43
|
Johansen AM, Forsythe SD, McGrath CT, Barker G, Jimenez H, Paluri RK, Pasche BC. TGFβ in Pancreas and Colorectal Cancer: Opportunities to Overcome Therapeutic Resistance. Clin Cancer Res 2024; 30:3676-3687. [PMID: 38916900 PMCID: PMC11371528 DOI: 10.1158/1078-0432.ccr-24-0468] [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: 02/09/2024] [Revised: 04/16/2024] [Accepted: 05/30/2024] [Indexed: 06/26/2024]
Abstract
TGFβ is a pleiotropic signaling pathway that plays a pivotal role in regulating a multitude of cellular functions. TGFβ has a dual role in cell regulation where it induces growth inhibition and cell death; however, it can switch to a growth-promoting state under cancerous conditions. TGFβ is upregulated in colorectal cancer and pancreatic cancer, altering the tumor microenvironment and immune system and promoting a mesenchymal state. The upregulation of TGFβ in certain cancers leads to resistance to immunotherapy, and attempts to inhibit TGFβ expression have led to reduced therapeutic resistance when combined with chemotherapy and immunotherapy. Here, we review the current TGFβ inhibitor drugs in clinical trials for pancreatic and colorectal cancer, with the goal of uncovering advances in improving clinical efficacy for TGFβ combinational treatments in patients. Furthermore, we discuss the relevance of alterations in TGFβ signaling and germline variants in the context of personalizing treatment for patients who show lack of response to current therapeutics.
Collapse
Affiliation(s)
- Allan M. Johansen
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157-1082, USA
| | - Steven D. Forsythe
- Neuroendocrine Therapy Section, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Callum T. McGrath
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157-1082, USA
| | - Grayson Barker
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157-1082, USA
| | - Hugo Jimenez
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan 48201, USA
| | - Ravi K. Paluri
- Section of Hematology/Oncology, Wake Forest School of Medicine, Winston-Salem, NC, 27157-1082. USA
| | - Boris C. Pasche
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan 48201, USA
| |
Collapse
|
44
|
Poyrazlı F, Okuyan D, Köçkar F, Türkoğlu SA. Hypoxic Regulation of the KLK4 Gene in two Different Prostate Cancer Cells Treated with TGF- β. Cell Biochem Biophys 2024; 82:2797-2812. [PMID: 39026058 DOI: 10.1007/s12013-024-01396-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2024] [Indexed: 07/20/2024]
Abstract
The human kallikrein-related peptidase (KLK) family which consists of 15 members is associated with prostate cancer and other cancers. It has been reported that overexpression of KLK4 in prostate cancer correlates with bone metastasis or advanced stage. Hypoxia occurs in the early stages of prostate cancer due to the accumulation of acidic metabolites or reactive oxygen species (ROS). In our study, KLK4 gene expression in hypoxic conditions in PC-3 and LNCaP cells which are treated with TGF-β was evaluated with mRNA, protein, and promoter activity levels. A chemical hypoxia model was created and confirmed at mRNA and protein level. No statistically significant cytotoxic effect of CoCl2 and TGF-β was observed in PC-3 and LNCaP cells with the MTT test. Four different truncated KLK4 gene promoter constructs were cloned in pmetLuc expression vector and basal activities of all promoter fragments were analyzed. The activities of P1 (-447/ + 657), P2 (-103/ + 657), and P3 (-267/ + 657) promoter fragments increased in hypoxic conditions except P4 (+555/ + 657), which does not contain the SMAD and HRE region. KLK4 mRNA levels in both PC-3 and LNCaP cells increased in the hypoxia and hypoxia/TGF groups compared to the non-treated groups. The stimulating effect of TGF-β is correlated with the increase in SMAD2/3 mRNA levels. KLK4 expression is up-regulated by TGF-β, especially under hypoxic conditions, and its interaction with the SMAD pathway is determined with different inhibitor experiments. HIF-1α and SMAD transcription factors bind to the KLK4 promoter showing the direct interaction of HIF-1α (-80/-52) and SMAD (+163/+194) regions with EMSA.
Collapse
Affiliation(s)
- Fatma Poyrazlı
- University of Balikesir, Faculty of Science and Literature, Department of Molecular Biology and Genetics, Balikesir, Turkey
| | - Derya Okuyan
- University of Bandırma, Susurluk Vocational Training Schools, Laboratory and Veterinary Health Program, Balikesir, Turkey
| | - Feray Köçkar
- University of Balikesir, Faculty of Science and Literature, Department of Molecular Biology and Genetics, Balikesir, Turkey
| | - Sümeyye Aydoğan Türkoğlu
- University of Balikesir, Faculty of Science and Literature, Department of Molecular Biology and Genetics, Balikesir, Turkey.
| |
Collapse
|
45
|
Islam MA, Sultana OF, Bandari M, Kshirsagar S, Manna PR, Reddy PH. MicroRNA-455-3P as a peripheral biomarker and therapeutic target for mild cognitive impairment and Alzheimer's disease. Ageing Res Rev 2024; 100:102459. [PMID: 39153602 PMCID: PMC11383742 DOI: 10.1016/j.arr.2024.102459] [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: 06/29/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
MicroRNAs are small non-coding RNAs evolutionary conserved molecules. They regulate cellular processes, including RNA silencing, post-translational gene expression and neurodegeneration. MicroRNAs are involved with human diseases such as cancer, Alzheimer's disease (AD) and others. Interestingly, cerebrospinal fluids (CSF) and the blood of AD patients have altered expressions of many RNAs, which may serve as potential peripheral biomarkers. The intensive investigation from our lab revealed that microRNA-455-3 P (miR-455-3p) is a strong candidate as a potential biomarker and therapeutic target for AD. Several genes implicated in the pathogenesis of AD are directly targeted by miR-455-3p. Several years of our lab research revealed that miR-455-3p regulates important physiological processes associated with AD, such as the processing of the amyloid precursor protein (APP), TGF-β signaling, the regulation of oxidative stress, mitochondrial biogenesis, and synaptic damages. The expression of miR-455-3p in mild cognitive impaired subjects and AD patients pointed out its involvement in AD progression. Recently, our lab generated both transgenic and knockout mice for miR-455-3p. Interestingly miR-455-3p transgenic mice showed superior cognitive learning, improved memory and extended lifespan compared to age matched wild-type mice, whereas miR-455-3-p knockout mice showed cognitive decline and reduced lifespan. Information derived from mouse models further demonstrated the advantageous impact of miR-455-3p on dendritic growth, synaptogenesis, and mitochondrial biogenesis in preventing the onset and progression of AD. The identification of miR-455-3p as a biomarker was suggested by its presence in postmortem AD brains, B-lymphocytes, and fibroblasts. Our hypothesis that miR-455-3p could be a peripheral biomarker and therapeutic target for AD.
Collapse
Affiliation(s)
- Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Madhuri Bandari
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Pulak R Manna
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA 5. Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
46
|
Zelisko N, Lesyk R, Stoika R. Structure, unique biological properties, and mechanisms of action of transforming growth factor β. Bioorg Chem 2024; 150:107611. [PMID: 38964148 DOI: 10.1016/j.bioorg.2024.107611] [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: 01/21/2024] [Revised: 06/07/2024] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
Transforming growth factor β (TGF-β) is a ubiquitous molecule that is extremely conserved structurally and plays a systemic role in human organism. TGF-β is a homodimeric molecule consisting of two subunits joined through a disulphide bond. In mammals, three genes code for TGF-β1, TGF-β2, and TGF-β3 isoforms of this cytokine with a dominating expression of TGF-β1. Virtually, all normal cells contain TGF-β and its specific receptors. Considering the exceptional role of fine balance played by the TGF-β in anumber of physiological and pathological processes in human body, this cytokine may be proposed for use in medicine as an immunosuppressant in transplantology, wound healing and bone repair. TGFb itself is an important target in oncology. Strategies for blocking members of TGF-β signaling pathway as therapeutic targets have been considered. In this review, signalling mechanisms of TGF-β1 action are addressed, and their role in physiology and pathology with main focus on carcinogenesis are described.
Collapse
Affiliation(s)
- Nataliya Zelisko
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine.
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine
| |
Collapse
|
47
|
Dave JM, Chakraborty R, Agyemang A, Ntokou A, Saito J, Ballabh P, Martin KA, Greif DM. Loss of TGFβ-Mediated Repression of Angiopoietin-2 in Pericytes Underlies Germinal Matrix Hemorrhage Pathogenesis. Stroke 2024; 55:2340-2352. [PMID: 39129597 PMCID: PMC11347087 DOI: 10.1161/strokeaha.123.045248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND TGF (transforming growth factor)-β pathway is central to blood-brain barrier development as it regulates cross talk between pericytes and endothelial cells. Murine embryos lacking TGFβ receptor Alk5 (activin receptor-like kinase 5) in brain pericytes (mutants) display endothelial cell hyperproliferation, abnormal vessel morphology, and gross germinal matrix hemorrhage-intraventricular hemorrhage (GMH-IVH), leading to perinatal lethality. Mechanisms underlying how ALK5 signaling in pericytes noncell autonomously regulates endothelial cell behavior remain elusive. METHODS Transcriptomic analysis of human brain pericytes with ALK5 silencing identified differential gene expression. Brain vascular cells isolated from mutant embryonic mice with GMH-IVH and preterm human IVH brain samples were utilized for target validation. Finally, pharmacological and genetic inhibition was used to study the therapeutic effects on GMH-IVH pathology. RESULTS Herein, we establish that the TGFβ/ALK5 pathway robustly represses ANGPT2 (angiopoietin-2) in pericytes via epigenetic remodeling. TGFβ-driven SMAD (suppressor of mothers against decapentaplegic) 3/4 associates with TGIF1 (TGFβ-induced factor homeobox 1) and HDAC (histone deacetylase) 5 to form a corepressor complex at the Angpt2 promoter, resulting in promoter deacetylation and gene repression. Moreover, murine and human germinal matrix vessels display increased ANGPT2 expression during GMH-IVH. Isolation of vascular cells from murine germinal matrix identifies pericytes as a cellular source of excessive ANGPT2. In addition, mutant endothelial cells exhibit higher phosphorylated TIE2 (tyrosine protein kinase receptor). Pharmacological or genetic inhibition of ANGPT2 in mutants improves germinal matrix vessel morphology and attenuates GMH pathogenesis. Importantly, genetic ablation of Angpt2 in mutant pericytes prevents perinatal lethality, prolonging survival. CONCLUSIONS This study demonstrates that TGFβ-mediated ANGPT2 repression in pericytes is critical for maintaining blood-brain barrier integrity and identifies pericyte-derived ANGPT2 as an important pathological target for GMH-IVH.
Collapse
Affiliation(s)
- Jui M. Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Raja Chakraborty
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
| | - Alex Agyemang
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aglaia Ntokou
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Junichi Saito
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Praveen Ballabh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kathleen A. Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
48
|
Brewer A, Zhao JF, Fasimoye R, Shpiro N, Macartney TJ, Wood NT, Wightman M, Alessi DR, Sapkota GP. Targeted dephosphorylation of SMAD3 as an approach to impede TGF-β signaling. iScience 2024; 27:110423. [PMID: 39104417 PMCID: PMC11298613 DOI: 10.1016/j.isci.2024.110423] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/09/2024] [Accepted: 06/27/2024] [Indexed: 08/07/2024] Open
Abstract
TGF-β (transforming growth factor-β) signaling is involved in a myriad of cellular processes and its dysregulation has been implicated in many human diseases, including fibrosis and cancer. TGF-β transcriptional responses are controlled by tail phosphorylation of transcription factors SMAD2 and SMAD3 (mothers against decapentaplegic homolog 2/3). Therefore, targeted dephosphorylation of phospho-SMAD3 could provide an innovative mechanism to block some TGF-β-induced transcriptional responses, such as the transcription of SERPINE-1, which encodes plasminogen activator inhibitor 1 (PAI-1). Here, by developing and employing a bifunctional molecule, BDPIC (bromoTAG-dTAG proximity-inducing chimera), we redirected multiple phosphatases, tagged with bromoTAG, to dephosphorylate phospho-SMAD3, tagged with dTAG. Using CRISPR-Cas9 technology, we generated homozygous double knock-in A549 bromoTAG/bromoTAG PPM1H/ dTAG/dTAG SMAD3 cells, in which the BDPIC-induced proximity between bromoTAG-PPM1H and dTAG-SMAD3 led to a robust dephosphorylation of dTAG-SMAD3 and a significant decrease in SERPINE-1 transcription. Our work demonstrates targeted dephosphorylation of phospho-proteins as an exciting modality for rewiring cell signaling.
Collapse
Affiliation(s)
- Abigail Brewer
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Jin-Feng Zhao
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Rotimi Fasimoye
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Natalia Shpiro
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Thomas J. Macartney
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Nicola T. Wood
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Melanie Wightman
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Dario R. Alessi
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Gopal P. Sapkota
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
49
|
Itoh Y, Miyake K, Koinuma D, Omata C, Saitoh M, Miyazawa K. Analysis of the DNA-binding properties of TGF-β-activated Smad complexes unveils a possible molecular basis for cellular context-dependent signaling. FASEB J 2024; 38:e23877. [PMID: 39114961 DOI: 10.1096/fj.202400978r] [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: 04/30/2024] [Revised: 07/10/2024] [Accepted: 07/30/2024] [Indexed: 09/18/2024]
Abstract
Transforming growth factor-β (TGF-β) is a pleiotropic cytokine that modulates a wide variety of cellular responses by regulating target gene expression. It principally transmits signals via receptor-activated transcription factors Smad2 and Smad3, which form trimeric complexes with Smad4 upon activation and regulate gene expression by binding to genomic DNA. Here, we examined the mechanisms by which TGF-β regulates the transcription of target genes in a cell context-dependent manner by screening a double-stranded DNA oligonucleotide library for DNA sequences bound to endogenous activated Smad complexes. Screening was performed by cyclic amplification of selected targets (CASTing) using an anti-Smad2/3 antibody and nuclear extracts isolated from three cell lines (A549, HepG2, and HaCaT) stimulated with TGF-β. The preference of the activated Smad complexes for conventional Smad-binding motifs such as Smad-binding element (SBE) and CAGA motifs was different in HepG2 than in the other two cell lines, which may indicate the distinct composition of the activated Smad complexes. Several transcription factor-binding motifs other than SBE or CAGA, including the Fos/Jun-binding motifs, were detected in the enriched sequences. Reporter assays using sequences containing these transcription factor-binding motifs together with Smad-binding motifs indicated that some of the motifs may be involved in cell type-dependent transcriptional activation by TGF-β. The results suggest that the CASTing method is useful for elucidating the molecular basis of context-dependent Smad signaling.
Collapse
Affiliation(s)
- Yuka Itoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kunio Miyake
- Department of Epidemiology and Environmental Medicine, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Daizo Koinuma
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Chiho Omata
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Masao Saitoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- Center for Medical Education and Sciences, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Keiji Miyazawa
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
50
|
Przybyszewski O, Mik M, Nowicki M, Kusiński M, Mikołajczyk-Solińska M, Śliwińska A. Using microRNAs Networks to Understand Pancreatic Cancer-A Literature Review. Biomedicines 2024; 12:1713. [PMID: 39200178 PMCID: PMC11351910 DOI: 10.3390/biomedicines12081713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic cancer is a severe disease, challenging to diagnose and treat, and thereby characterized by a poor prognosis and a high mortality rate. Pancreatic ductal adenocarcinoma (PDAC) represents approximately 90% of pancreatic cancer cases, while other cases include neuroendocrine carcinoma. Despite the growing knowledge of the pathophysiology of this cancer, the mortality rate caused by it has not been effectively reduced. Recently, microRNAs have aroused great interest among scientists and clinicians, as they are negative regulators of gene expression, which participate in many processes, including those related to the development of pancreatic cancer. The aim of this review is to show how microRNAs (miRNAs) affect key signaling pathways and related cellular processes in pancreatic cancer development, progression, diagnosis and treatment. We included the results of in vitro studies, animal model of pancreatic cancer and those performed on blood, saliva and tumor tissue isolated from patients suffering from PDAC. Our investigation identified numerous dysregulated miRNAs involved in KRAS, JAK/STAT, PI3/AKT, Wnt/β-catenin and TGF-β signaling pathways participating in cell cycle control, proliferation, differentiation, apoptosis and metastasis. Moreover, some miRNAs (miRNA-23a, miRNA-24, miRNA-29c, miRNA-216a) seem to be engaged in a crosstalk between signaling pathways. Evidence concerning the utility of microRNAs in the diagnosis and therapy of this cancer is poor. Therefore, despite growing knowledge of the involvement of miRNAs in several processes associated with pancreatic cancer, we are beginning to recognize and understand their role and usefulness in clinical practice.
Collapse
Affiliation(s)
- Oskar Przybyszewski
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska St., 92-213 Lodz, Poland
| | - Michał Mik
- Department of General and Colorectal Surgery, Medical University of Lodz, 113 Stefana Żeromskiego St., 90-549 Lodz, Poland; (M.M.); (M.N.)
| | - Michał Nowicki
- Department of General and Colorectal Surgery, Medical University of Lodz, 113 Stefana Żeromskiego St., 90-549 Lodz, Poland; (M.M.); (M.N.)
| | - Michał Kusiński
- Department of Endocrinological, General and Oncological Surgery, Medical University of Lodz, 62 Pabianicka St., 93-513 Lodz, Poland;
| | - Melania Mikołajczyk-Solińska
- Department of Internal Medicine, Diabetology and Clinical Pharmacology, Medical University of Lodz, 251 Pomorska St., 92-213 Lodz, Poland;
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska St., 92-213 Lodz, Poland
| |
Collapse
|