1
|
Bhardwaj K, Jha A, Roy A, Kumar H. The crucial role of VPS35 and SHH in Parkinson's disease: Understanding the mechanisms behind the neurodegenerative disorder. Brain Res 2024; 1845:149204. [PMID: 39197569 DOI: 10.1016/j.brainres.2024.149204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/10/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
Parkinson's disease (PD) is indeed a complex neurodegenerative disorder recognized by the progressive depletion of dopaminergic neurons in the brain, particularly in the substantia nigra region, leading to motor impairments and other symptoms. But at the molecular level, the study about PD still lacks. As the number of cases worldwide continues to increase, it is critical to focus on the cellular and molecular mechanisms of the disease's presentation and neurodegeneration to develop novel therapeutic approaches. At the molecular level, the complexity is more due to the involvement of vacuolar protein sorting 35 (VPS35) and sonic hedgehog (SHH) signaling in PD (directly or indirectly), leading to one of the most prominent hallmarks of the disease, which is an accumulation of α-synuclein. This elevated pathogenesis may result from impaired autophagy due to mutation in the case of VPS35 and impairment in SHH signaling at the molecular level. The traditional understanding of PD is marked by the disruption of dopaminergic neurons and dopaminergic signaling, which exacerbates symptoms of motor function deficits. However, the changes at the molecular level that are being disregarded also impact the overall health of the dopaminergic system. Gaining insight into these two unique mechanisms is essential to determine whether they give neuroprotection or have no effect on the health of neurons. Hence, here we tried to simplify the understanding of the role of VPS35 and SHH signaling to comprehend it in one direction.
Collapse
Affiliation(s)
- Kritika Bhardwaj
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Akanksha Jha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
2
|
Reding K, Chung M, Heath A, Hotopp JD, Pick L. Same rule, different genes: Blimp1 is a pair-rule gene in the milkweed bug Oncopeltus fasciatus. SCIENCE ADVANCES 2024; 10:eadq9045. [PMID: 39546609 PMCID: PMC11566998 DOI: 10.1126/sciadv.adq9045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024]
Abstract
Morphological features of organismal body plans are often highly conserved within large taxa. For example, segmentation is a shared and defining feature of all insects. Screens in Drosophila identified genes responsible for the development of body segments, including the "pair-rule" genes (PRGs), which subdivide embryos into double-segment units in a previously unexpected pre-patterning step. Here we show that the milkweed bug Oncopeltus fasciatus also uses a pair rule for embryo subdivision but Oncopeltus employs different genes for this process. We identified the gene Blimp1 as an Oncopeltus PRG based on its expression pattern, tested its function with RNA interference and CRISPR-Cas9, and generated the first PR mutant in this species. Although it does not have PR function in Drosophila, like Drosophila PRGs, Blimp1 encodes a transcription factor required for embryonic viability. Thus, pair-rule subdivision of the insect body plan is more highly conserved than the factors mediating this process, suggesting a developmental constraint on this pre-patterning step.
Collapse
Affiliation(s)
- Katie Reding
- Department of Entomology, University of Maryland, 4291 Fieldhouse Dr., College Park, MD 20742, USA
| | - Matthew Chung
- Microbiology and Immunology and Institute for Genome Sciences, University of Maryland School of Medicine, 670 West Baltimore St., Baltimore, MD 21201, USA
| | - Abigail Heath
- Department of Entomology, University of Maryland, 4291 Fieldhouse Dr., College Park, MD 20742, USA
| | - Julie Dunning Hotopp
- Microbiology and Immunology and Institute for Genome Sciences, University of Maryland School of Medicine, 670 West Baltimore St., Baltimore, MD 21201, USA
| | - Leslie Pick
- Department of Entomology, University of Maryland, 4291 Fieldhouse Dr., College Park, MD 20742, USA
| |
Collapse
|
3
|
Peifer M. 40 years since the Heidelberg genetic screen that revolutionized developmental and cell biology. Development 2024; 151:dev204433. [PMID: 39431300 DOI: 10.1242/dev.204433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Our current understanding of the molecular basis of embryonic development and the shared machinery underlying this remarkable process has its roots in three papers published 40 years ago, which summarize the results of the Nobel Prize-winning 'Heidelberg screen'. The genesis of these experiments that empowered us and the stories behind the experiments are worth revisiting.
Collapse
Affiliation(s)
- Mark Peifer
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
4
|
Arrías PN, Osmanli Z, Peralta E, Chinestrad PM, Monzon AM, Tosatto SCE. Diversity and structural-functional insights of alpha-solenoid proteins. Protein Sci 2024; 33:e5189. [PMID: 39465903 PMCID: PMC11514114 DOI: 10.1002/pro.5189] [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: 05/06/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/29/2024]
Abstract
Alpha-solenoids are a significant and diverse subset of structured tandem repeat proteins (STRPs) that are important in various domains of life. This review examines their structural and functional diversity and highlights their role in critical cellular processes such as signaling, apoptosis, and transcriptional regulation. Alpha-solenoids can be classified into three geometric folds: low curvature, high curvature, and corkscrew, as well as eight subfolds: ankyrin repeats; Huntingtin, elongation factor 3, protein phosphatase 2A, and target of rapamycin; armadillo repeats; tetratricopeptide repeats; pentatricopeptide repeats; Pumilio repeats; transcription activator-like; and Sel-1 and Sel-1-like repeats. These subfolds represent distinct protein families with unique structural properties and functions, highlighting the versatility of alpha-solenoids. The review also discusses their association with disease, highlighting their potential as therapeutic targets and their role in protein design. Advances in state-of-the-art structure prediction methods provide new opportunities and challenges in the functional characterization and classification of this kind of fold, emphasizing the need for continued development of methods for their identification and proper data curation and deposition in the main databases.
Collapse
Affiliation(s)
- Paula Nazarena Arrías
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- Department of Protein ScienceKTH Royal Institute of TechnologyStockholmSweden
| | - Zarifa Osmanli
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Estefanía Peralta
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias ExactasUniversidad Nacional de La PlataLa PlataBuenos AiresArgentina
| | | | | | - Silvio C. E. Tosatto
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- Institute of Biomembranes, Bioenergetics and Molecular BiotechnologiesNational Research Council (CNR‐IBIOM)BariItaly
| |
Collapse
|
5
|
Pallavi S, Jain S, Mohanty SK, Andrabi SW, Rajender S. Retinoic Acid Regulates Spermiogenesis Via Hoxb1 and Shh Signaling in Testicular Germ Cells. Reprod Sci 2024; 31:3400-3412. [PMID: 39080234 DOI: 10.1007/s43032-024-01648-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: 04/05/2024] [Accepted: 07/08/2024] [Indexed: 11/02/2024]
Abstract
Retinoic acid (RA) regulates all four major events in spermatogenesis; spermatogonial differentiation, meiotic entry, spermiogenesis, and spermiation. For the pre-meiotic phase, Sertoli cells are the source of RA and for the post-meiotic phase, pachytene spermatocytes are the source of RA. While the entire spermatogenic process is regulated by RA, how each of these phases is regulated by RA remains completely unknown. Homeobox B1 (Hoxb1) has two retinoic acid response elements (RARE) upstream and downstream of the gene. In this study, we investigated if RA facilitates spermatogenesis by its action on Hoxb1. The expressions of the Hoxb1 and Sonic hedgehog (Shh) genes were analyzed in the post-natal mouse testes and the testicular localizations of Hoxb1, Shh and Gli1 were analyzed by immunohistochemistry in the adult rat testis. To delineate the signaling mechanisms, Hoxb1 expression was altered in vitro and in vivo using retinoic acid and miR-361-3p. Finally, the levels of miR-361-3p and HOXB1 were analyzed in infertile human sperm samples. Hoxb1 and Shh gene expressions were found to be low in the testis of post-natal Swiss mice of 7, 14, 28, 35, and 60 days, after which the expressions of both spiked. Immunohistochemistry in the adult mouse testis showed the expressions of Hoxb1, Shh, and Gli1 in the elongating spermatids. Exposure of GC2 cells to RA and in vivo IP RA injection upregulated Hoxb1 and Shh signaling in the testis with increased expressions of Shh, Gli1, and Hdac1. Retinoic acid administration in Swiss mice compromised sperm production and reduced epididymal sperm count. The analysis of infertile human semen samples revealed an increased level of HOXB1 and a decreased level of miR-361-3p as compared to fertile controls. We conclude that retinoic acid regulates late stage of spermatogenesis (spermiogenesis) by affecting Hoxb1 and Shh signaling.
Collapse
Affiliation(s)
- Saini Pallavi
- Division of Endocrinology, Central Drug Research Institute, Uttar Pradesh, Lucknow, India
- Academy of Scientific and Innovative Research, Uttar Pradesh, Ghaziabad, India
| | - Simran Jain
- Division of Endocrinology, Central Drug Research Institute, Uttar Pradesh, Lucknow, India
| | - Sujit Kumar Mohanty
- Division of Endocrinology, Central Drug Research Institute, Uttar Pradesh, Lucknow, India
| | | | - Singh Rajender
- Division of Endocrinology, Central Drug Research Institute, Uttar Pradesh, Lucknow, India.
- Academy of Scientific and Innovative Research, Uttar Pradesh, Ghaziabad, India.
| |
Collapse
|
6
|
Ali M, Younas L, Liu J, He H, Zhang X, Zhou Q. Development and evolution of Drosophila chromatin landscape in a 3D genome context. Nat Commun 2024; 15:9452. [PMID: 39487148 PMCID: PMC11530545 DOI: 10.1038/s41467-024-53892-0] [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: 02/27/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024] Open
Abstract
Little is known about how the epigenomic states change during development and evolution in a 3D genome context. Here we use Drosophila pseudoobscura with complex turnover of sex chromosomes as a model to address this, by collecting massive epigenomic and Hi-C data from five developmental stages and three adult tissues. We reveal that over 60% of the genes and transposable elements (TE) exhibit at least one developmental transition of chromatin state. Transitions on specific but not housekeeping enhancers are associated with specific chromatin loops and topologically associated domain borders (TABs). While evolutionarily young TEs are generally silenced, old TEs more often have been domesticated as interacting TABs or specific enhancers. But on the recently evolved X chromosome, young TEs are instead often active and recruited as TABs, due to acquisition of dosage compensation. Overall we characterize how Drosophila epigenomic landscapes change during development and in response to chromosome evolution, and highlight the important roles of TEs in genome organization and regulation.
Collapse
Affiliation(s)
- Mujahid Ali
- Center for Reproductive Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria
- Institute of Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Lubna Younas
- Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria
| | - Jing Liu
- Center for Evolutionary & Organismal Biology & Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Huangyi He
- MOE Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xinpei Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Qi Zhou
- Center for Reproductive Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria.
- Center for Evolutionary & Organismal Biology & Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- MOE Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China.
| |
Collapse
|
7
|
Yan RE, Greenfield JP. Challenges and Outlooks in Precision Medicine: Expectations Versus Reality. World Neurosurg 2024; 190:573-581. [PMID: 39425299 DOI: 10.1016/j.wneu.2024.06.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 10/21/2024]
Abstract
Recent developments in technology have led to rapid advances in precision medicine, especially due to the rise of next-generation sequencing and molecular profiling. These technological advances have led to rapid advances in research, including increased tumor subtype resolution, new therapeutic agents, and mechanistic insights. Certain therapies have even been approved for molecular biomarkers across histopathological diagnoses; however, translation of research findings to the clinic still faces a number of challenges. In this review, the authors discuss several key challenges to the clinical integration of precision medicine, including the blood-brain barrier, both a lack and excess of molecular targets, and tumor heterogeneity/escape from therapy. They also highlight a few key efforts to address these challenges, including new frontiers in drug delivery, a rapidly expanding treatment repertoire, and improvements in active response monitoring. With continued improvements and developments, the authors anticipate that precision medicine will increasingly become the gold standard for clinical care.
Collapse
Affiliation(s)
- Rachel E Yan
- Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Jeffrey P Greenfield
- Department of Neurological Surgery, NewYork-Presbyterian Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
8
|
Schnider ST, Vigano MA, Affolter M, Aguilar G. Functionalized Protein Binders in Developmental Biology. Annu Rev Cell Dev Biol 2024; 40:119-142. [PMID: 39038471 DOI: 10.1146/annurev-cellbio-112122-025214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Developmental biology has greatly profited from genetic and reverse genetic approaches to indirectly studying protein function. More recently, nanobodies and other protein binders derived from different synthetic scaffolds have been used to directly dissect protein function. Protein binders have been fused to functional domains, such as to lead to protein degradation, relocalization, visualization, or posttranslational modification of the target protein upon binding. The use of such functionalized protein binders has allowed the study of the proteome during development in an unprecedented manner. In the coming years, the advent of the computational design of protein binders, together with further advances in scaffold engineering and synthetic biology, will fuel the development of novel protein binder-based technologies. Studying the proteome with increased precision will contribute to a better understanding of the immense molecular complexities hidden in each step along the way to generate form and function during development.
Collapse
Affiliation(s)
| | | | | | - Gustavo Aguilar
- Current affiliation: Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
- Biozentrum, Universität Basel, Basel, Switzerland;
| |
Collapse
|
9
|
Gallo E, De Renzis S, Sharpe J, Mayor R, Hartmann J. Versatile system cores as a conceptual basis for generality in cell and developmental biology. Cell Syst 2024; 15:790-807. [PMID: 39236709 DOI: 10.1016/j.cels.2024.08.001] [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: 08/16/2023] [Revised: 05/26/2024] [Accepted: 08/07/2024] [Indexed: 09/07/2024]
Abstract
The discovery of general principles underlying the complexity and diversity of cellular and developmental systems is a central and long-standing aim of biology. While new technologies collect data at an ever-accelerating rate, there is growing concern that conceptual progress is not keeping pace. We contend that this is due to a paucity of conceptual frameworks that support meaningful generalizations. This led us to develop the core and periphery (C&P) hypothesis, which posits that many biological systems can be decomposed into a highly versatile core with a large behavioral repertoire and a specific periphery that configures said core to perform one particular function. Versatile cores tend to be widely reused across biology, which confers generality to theories describing them. Here, we introduce this concept and describe examples at multiple scales, including Turing patterning, actomyosin dynamics, multi-cellular morphogenesis, and vertebrate gastrulation. We also sketch its evolutionary basis and discuss key implications and open questions. We propose that the C&P hypothesis could unlock new avenues of conceptual progress in mesoscale biology.
Collapse
Affiliation(s)
- Elisa Gallo
- Institute of Molecular Life Sciences, University of Zurich (UZH), 8057 Zurich, Switzerland
| | - Stefano De Renzis
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - James Sharpe
- EMBL Barcelona, European Molecular Biology Laboratory (EMBL), 08003 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Jonas Hartmann
- Institute of Molecular Life Sciences, University of Zurich (UZH), 8057 Zurich, Switzerland; Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany; EMBL Barcelona, European Molecular Biology Laboratory (EMBL), 08003 Barcelona, Spain; Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK; Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL 60208, USA; Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
10
|
Falconi J, Strobel K, Djiane A, Lassus P. [Drosophila as a model to study cancer biology]. Bull Cancer 2024; 111:880-892. [PMID: 38960821 DOI: 10.1016/j.bulcan.2024.05.005] [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: 01/30/2024] [Revised: 04/18/2024] [Accepted: 05/03/2024] [Indexed: 07/05/2024]
Abstract
The rising global incidence of cancer makes it the second leading cause of death worldwide. Over the past decades, significant progress has been made in both basic knowledge and the discovery of new therapeutic approaches. However, the complexity of mechanisms related to tumor development requires the use of sophisticated and adapted research tools. Among these, the fruitfly Drosophila melanogaster represents a powerful genetic model with numerous practical and conceptual advantages. Indeed, the conservation of genes implicated in cancer between this insect and mammals places Drosophila as a crucial genetic tool for understanding the fundamental mechanisms governing tumorigenesis and identifying new therapeutic targets. This review aims to describe this original model and demonstrate its relevance for studying cancer biology.
Collapse
Affiliation(s)
- Jennifer Falconi
- IRCM, Inserm, ICM, université de Montpellier, Montpellier, France
| | - Katrin Strobel
- IRCM, Inserm, ICM, université de Montpellier, Montpellier, France
| | - Alexandre Djiane
- IRCM, Inserm, ICM, université de Montpellier, Montpellier, France
| | - Patrice Lassus
- IRCM, Inserm U1194, ICM, CNRS, université de Montpellier, 208, rue des Apothicaires, 34298 Montpellier cedex, France.
| |
Collapse
|
11
|
Sánchez Arteaga J. "The Logic of Monsters:" Pere Alberch and the Evolutionary Significance of Experimental Teratology. JOURNAL OF THE HISTORY OF BIOLOGY 2024; 57:379-401. [PMID: 39212877 PMCID: PMC11438615 DOI: 10.1007/s10739-024-09783-7] [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] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
This paper offers an historical introduction to Pere Alberch's evolutionary thought and his contributions to Evo-Devo, based on his unique approach to experimental teratology. We will take as our point of reference the teratogenic experiments developed by Alberch and Emily A. Gale during the 1980s, aimed at producing monstrous variants of frogs and salamanders. We will analyze his interpretation of the results of these experiments within the framework of the emergence of evolutionary developmental biology (or "Evo-Devo"). The aim is understand how Alberch interpreted teratological anomalies as highly revealing objects of study for understanding the development of organic form, not only in an ontogenetic sense-throughout embryonic development-but also phylogenetically-throughout the evolution of species. Alberch's interpretation of monsters reflects the influence of a long tradition of non-Darwinian evolutionary thought, which began in the nineteenth century and was continued in the twentieth century by people such as Richard Goldschmidt, Conrad H. Waddington, and Stephen Jay Gould. They all proposed various non-gradualist models of evolution, in which embryonic development played a central role. Following this tradition, Alberch argued that, in order to attain a correct understanding of the role of embryological development in evolution, it was necessary to renounce the gradualist paradigm associated with the Darwinian interpretation of evolution, which understood nature as a continuum. According to Alberch, the study of monstrous abnormalities was of great value in understanding how certain epigenetic restrictions in development could give rise to discontinuities and directionality in morphological transformations throughout evolution.
Collapse
Affiliation(s)
- Juanma Sánchez Arteaga
- Institute of History (IH), Spanish National Research Council (CSIC), Madrid, Spain.
- Centro de Ciencias Humanas y Sociales - Consejo Superior de Investigaciones Científicas (CCHS/CSIC), Despacho 2F2, Calle de Albasanz, 26, San Blas-Canillejas, 28037, Madrid, Spain.
| |
Collapse
|
12
|
Jiang L, Yang S, Deng L, Luo J, Zhang X, Chen S, Dong Z. ARL13B promotes cell cycle through the sonic hedgehog signaling pathway to alleviate nerve damage during cerebral ischemia/reperfusion in rats. Biochem Pharmacol 2024; 227:116446. [PMID: 39038552 DOI: 10.1016/j.bcp.2024.116446] [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: 03/12/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Cerebral ischemia/reperfusion (CIRI) is a leading cause of death worldwide. A small GTPase known as ADP-ribosylation factor-like protein 13B (ARL13B) is essential in several illnesses. The role of ARL13B in CIRI remains unknown, though. A middle cerebral artery occlusion/reperfusion (MCAO/R) in rats as well as an oxygen-glucose deprivation/reoxygenation (OGD/R) models in PC12 cells were constructed. The neuroprotective effects of ARL13B against MCAO/R were evaluated using neurological scores, TTC staining, rotarod testing, H&E staining, and Nissl staining. To detect the expression of proteins associated with the SHH pathway and apoptosis, western blotting and immunofluorescence were employed. Apoptosis was detected using TUNEL assays and flow cytometry. There was increased expression of ARL13B in cerebral ischemia/reperfusion models. However, ARL13B knockdown aggravated CIRI nerve injury by inhibiting the sonic hedgehog (SHH) pathway. In addition, the use of SHH pathway agonist (SAG) can increased ARL13B expression, reverse the effects of ARL13B knockdown exacerbating CIRI nerve injury. ARL13B alleviated cerebral infarction and pathological injury and played a protective role against MCAO/R. Furthermore, ARL13B significantly increased the expression of SHH pathway-related proteins and the anti-apoptotic protein BCL-2, while decreased the expression of pro-apoptotic protein BAX, thus reducing apoptosis. The results from the OGD/R model in PC12 cells were consistent with those obtained in vivo. Surprisingly, we demonstrated that ARL13B regulates the cell cycle to protect against CIRI nerve injury. Our findings indicate that ARL13B protects against CIRI by reducing apoptosis through SHH-dependent pathway activation, and suggest that ARL13B plays a crucial role in CIRI pathogenesis.
Collapse
Affiliation(s)
- Lu Jiang
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Shaonan Yang
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Ling Deng
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Jingjing Luo
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Xiaoling Zhang
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Sha Chen
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Zhi Dong
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China.
| |
Collapse
|
13
|
Li XC, Srinivasan V, Laiker I, Misunou N, Frankel N, Pallares LF, Crocker J. TF-High-Evolutionary: In Vivo Mutagenesis of Gene Regulatory Networks for the Study of the Genetics and Evolution of the Drosophila Regulatory Genome. Mol Biol Evol 2024; 41:msae167. [PMID: 39117360 PMCID: PMC11342961 DOI: 10.1093/molbev/msae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 08/10/2024] Open
Abstract
Understanding the evolutionary potential of mutations in gene regulatory networks is essential to furthering the study of evolution and development. However, in multicellular systems, genetic manipulation of regulatory networks in a targeted and high-throughput way remains challenging. In this study, we designed TF-High-Evolutionary (HighEvo), a transcription factor (TF) fused with a base editor (activation-induced deaminase), to continuously induce germline mutations at TF-binding sites across regulatory networks in Drosophila. Populations of flies expressing TF-HighEvo in their germlines accumulated mutations at rates an order of magnitude higher than natural populations. Importantly, these mutations accumulated around the targeted TF-binding sites across the genome, leading to distinct morphological phenotypes consistent with the developmental roles of the tagged TFs. As such, this TF-HighEvo method allows the interrogation of the mutational space of gene regulatory networks at scale and can serve as a powerful reagent for experimental evolution and genetic screens focused on the regulatory genome.
Collapse
Affiliation(s)
- Xueying C Li
- European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Ian Laiker
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Universidad de Buenos Aires (UBA), Buenos Aires 1428, Argentina
| | | | - Nicolás Frankel
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Universidad de Buenos Aires (UBA), Buenos Aires 1428, Argentina
| | - Luisa F Pallares
- Friedrich Miescher Laboratory, Max Planck Society, Tübingen, Germany
| | - Justin Crocker
- European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
14
|
Shi DL. Canonical and Non-Canonical Wnt Signaling Generates Molecular and Cellular Asymmetries to Establish Embryonic Axes. J Dev Biol 2024; 12:20. [PMID: 39189260 PMCID: PMC11348223 DOI: 10.3390/jdb12030020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
The formation of embryonic axes is a critical step during animal development, which contributes to establishing the basic body plan in each particular organism. Wnt signaling pathways play pivotal roles in this fundamental process. Canonical Wnt signaling that is dependent on β-catenin regulates the patterning of dorsoventral, anteroposterior, and left-right axes. Non-canonical Wnt signaling that is independent of β-catenin modulates cytoskeletal organization to coordinate cell polarity changes and asymmetric cell movements. It is now well documented that components of these Wnt pathways biochemically and functionally interact to mediate cell-cell communications and instruct cellular polarization in breaking the embryonic symmetry. The dysfunction of Wnt signaling disrupts embryonic axis specification and proper tissue morphogenesis, and mutations of Wnt pathway genes are associated with birth defects in humans. This review discusses the regulatory roles of Wnt pathway components in embryonic axis formation by focusing on vertebrate models. It highlights current progress in decoding conserved mechanisms underlying the establishment of asymmetry along the three primary body axes. By providing an in-depth analysis of canonical and non-canonical pathways in regulating cell fates and cellular behaviors, this work offers insights into the intricate processes that contribute to setting up the basic body plan in vertebrate embryos.
Collapse
Affiliation(s)
- De-Li Shi
- Department of Medical Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China;
- Laboratory of Developmental Biology, Centre National de la Recherche Scientifique (CNRS), UMR7622, Institut de Biologie Paris-Seine (IBPS), Sorbonne University, 75005 Paris, France
| |
Collapse
|
15
|
Guo Z, Wang S, Wang Y, Wang Z, Ou G. A machine learning enhanced EMS mutagenesis probability map for efficient identification of causal mutations in Caenorhabditis elegans. PLoS Genet 2024; 20:e1011377. [PMID: 39186782 PMCID: PMC11379379 DOI: 10.1371/journal.pgen.1011377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 09/06/2024] [Accepted: 07/27/2024] [Indexed: 08/28/2024] Open
Abstract
Chemical mutagenesis-driven forward genetic screens are pivotal in unveiling gene functions, yet identifying causal mutations behind phenotypes remains laborious, hindering their high-throughput application. Here, we reveal a non-uniform mutation rate caused by Ethyl Methane Sulfonate (EMS) mutagenesis in the C. elegans genome, indicating that mutation frequency is influenced by proximate sequence context and chromatin status. Leveraging these factors, we developed a machine learning enhanced pipeline to create a comprehensive EMS mutagenesis probability map for the C. elegans genome. This map operates on the principle that causative mutations are enriched in genetic screens targeting specific phenotypes among random mutations. Applying this map to Whole Genome Sequencing (WGS) data of genetic suppressors that rescue a C. elegans ciliary kinesin mutant, we successfully pinpointed causal mutations without generating recombinant inbred lines. This method can be adapted in other species, offering a scalable approach for identifying causal genes and revitalizing the effectiveness of forward genetic screens.
Collapse
Affiliation(s)
- Zhengyang Guo
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Shimin Wang
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Yang Wang
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Zi Wang
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Guangshuo Ou
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| |
Collapse
|
16
|
Deichmann U. Contrasting philosophical and scientific views in the long history of studying the generation of form in development. Biosystems 2024; 242:105260. [PMID: 38925338 DOI: 10.1016/j.biosystems.2024.105260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/23/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
Focusing on the opposing ways of thinking of philosophers and scientists to explain the generation of form in biological development, I show that today's controversies over explanations of early development bear fundamental similarities to the dichotomy of preformation theory versus epigenesis in Greek antiquity. They are related to the acceptance or rejection of the idea of a physical form of what today would be called information for the generating of the embryo as a necessary pre-requisite for specific development and heredity. As a recent example, I scrutinize the dichotomy of genomic causality versus self-organization in 20th and 21st century theories of the generation of form. On the one hand, the generation of patterns and form, as well as the constant outcome in development, are proposed to be causally related to something that is "preformed" in the germ cells, the nucleus of germ cells, or the genome. On the other hand, it is proposed that there is no pre-existing form or information, and development is seen as a process where genuinely new characters emerge from formless matter, either by immaterial "forces of life," or by physical-chemical processes of self-organization. I also argue that these different ways of thinking and the research practices associated with them are not equivalent, and maintain that it is impossible to explain the generation of form and constant outcome of development without the assumption of the transmission of pre-existing information in the form of DNA sequences in the genome. Only in this framework of "preformed" information can "epigenesis" in the form of physical and chemical processes of self-organization play an important role.
Collapse
Affiliation(s)
- Ute Deichmann
- Jacques Loeb Centre for the History and Philosophy of Science, Ben-Gurion University of the Negev, Beer Sheva, 8410500, Israel.
| |
Collapse
|
17
|
Tümen D, Heumann P, Huber J, Hahn N, Macek C, Ernst M, Kandulski A, Kunst C, Gülow K. Unraveling Cancer's Wnt Signaling: Dynamic Control through Protein Kinase Regulation. Cancers (Basel) 2024; 16:2686. [PMID: 39123414 PMCID: PMC11312265 DOI: 10.3390/cancers16152686] [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: 07/08/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Since the initial identification of oncogenic Wnt in mice and Drosophila, the Wnt signaling pathway has been subjected to thorough and extensive investigation. Persistent activation of Wnt signaling exerts diverse cancer characteristics, encompassing tumor initiation, tumor growth, cell senescence, cell death, differentiation, and metastasis. Here we review the principal signaling mechanisms and the regulatory influence of pathway-intrinsic and extrinsic kinases on cancer progression. Additionally, we underscore the divergences and intricate interplays of the canonical and non-canonical Wnt signaling pathways and their critical influence in cancer pathophysiology, exhibiting both growth-promoting and growth-suppressing roles across diverse cancer types.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Karsten Gülow
- Department of Internal Medicine I Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (D.T.); (N.H.)
| |
Collapse
|
18
|
David OG, Arce AV, Costa-da-Silva AL, Bellantuono AJ, DeGennaro M. Fertility decline in Aedes aegypti (Diptera: Culicidae) mosquitoes is associated with reduced maternal transcript deposition and does not depend on female age. JOURNAL OF MEDICAL ENTOMOLOGY 2024; 61:1064-1070. [PMID: 38757780 PMCID: PMC11239790 DOI: 10.1093/jme/tjae058] [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: 02/07/2024] [Revised: 04/02/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024]
Abstract
Female mosquitoes undergo multiple rounds of reproduction known as gonotrophic cycles (GC). A gonotrophic cycle spans the period from blood meal intake to egg laying. Nutrients from vertebrate host blood are necessary for completing egg development. During oogenesis, a female prepackages mRNA into her oocytes, and these maternal transcripts drive the first 2 h of embryonic development prior to zygotic genome activation. In this study, we profiled transcriptional changes in 1-2 h of Aedes aegypti (Diptera: Culicidae) embryos across 2 GC. We found that homeotic genes which are regulators of embryogenesis are downregulated in embryos from the second gonotrophic cycle. Interestingly, embryos produced by Ae. aegypti females progressively reduced their ability to hatch as the number of GC increased. We show that this fertility decline is due to increased reproductive output and not the mosquitoes' age. Moreover, we found a similar decline in fertility and fecundity across 3 GC in Aedes albopictus. Our results are useful for predicting mosquito population dynamics to inform vector control efforts.
Collapse
Affiliation(s)
- Olayinka G David
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA
| | - Andrea V Arce
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA
| | - Andre Luis Costa-da-Silva
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA
| | - Anthony J Bellantuono
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA
| | - Matthew DeGennaro
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
19
|
Liberali P, Schier AF. The evolution of developmental biology through conceptual and technological revolutions. Cell 2024; 187:3461-3495. [PMID: 38906136 DOI: 10.1016/j.cell.2024.05.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Developmental biology-the study of the processes by which cells, tissues, and organisms develop and change over time-has entered a new golden age. After the molecular genetics revolution in the 80s and 90s and the diversification of the field in the early 21st century, we have entered a phase when powerful technologies provide new approaches and open unexplored avenues. Progress in the field has been accelerated by advances in genomics, imaging, engineering, and computational biology and by emerging model systems ranging from tardigrades to organoids. We summarize how revolutionary technologies have led to remarkable progress in understanding animal development. We describe how classic questions in gene regulation, pattern formation, morphogenesis, organogenesis, and stem cell biology are being revisited. We discuss the connections of development with evolution, self-organization, metabolism, time, and ecology. We speculate how developmental biology might evolve in an era of synthetic biology, artificial intelligence, and human engineering.
Collapse
Affiliation(s)
- Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; University of Basel, Basel, Switzerland.
| | | |
Collapse
|
20
|
Hu L, Chen W, Qian A, Li YP. Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and disease. Bone Res 2024; 12:39. [PMID: 38987555 PMCID: PMC11237130 DOI: 10.1038/s41413-024-00342-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/27/2024] [Accepted: 05/12/2024] [Indexed: 07/12/2024] Open
Abstract
Wnts are secreted, lipid-modified proteins that bind to different receptors on the cell surface to activate canonical or non-canonical Wnt signaling pathways, which control various biological processes throughout embryonic development and adult life. Aberrant Wnt signaling pathway underlies a wide range of human disease pathogeneses. In this review, we provide an update of Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and diseases. The Wnt proteins, receptors, activators, inhibitors, and the crosstalk of Wnt signaling pathways with other signaling pathways are summarized and discussed. We mainly review Wnt signaling functions in bone formation, homeostasis, and related diseases, and summarize mouse models carrying genetic modifications of Wnt signaling components. Moreover, the therapeutic strategies for treating bone diseases by targeting Wnt signaling, including the extracellular molecules, cytosol components, and nuclear components of Wnt signaling are reviewed. In summary, this paper reviews our current understanding of the mechanisms by which Wnt signaling regulates bone formation, homeostasis, and the efforts targeting Wnt signaling for treating bone diseases. Finally, the paper evaluates the important questions in Wnt signaling to be further explored based on the progress of new biological analytical technologies.
Collapse
Affiliation(s)
- Lifang Hu
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Airong Qian
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
21
|
Sigafoos AN, Tolosa EJ, Carr RM, Fernandez-Barrena MG, Almada LL, Pease DR, Hogenson TL, Raja Arul GL, Mousavi F, Sen S, Vera RE, Marks DL, Flores LF, LaRue-Nolan KC, Wu C, Bamlet WR, Vrabel AM, Sicotte H, Schenk EL, Smyrk TC, Zhang L, Rabe KG, Oberg AL, Zaphiropoulos PG, Chevet E, Graham RP, Hagen CE, di Magliano MP, Elsawa SF, Pin CL, Mao J, McWilliams RR, Fernandez-Zapico ME. KRAS Promotes GLI2-Dependent Transcription during Pancreatic Carcinogenesis. CANCER RESEARCH COMMUNICATIONS 2024; 4:1677-1689. [PMID: 38896052 PMCID: PMC11232480 DOI: 10.1158/2767-9764.crc-23-0464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/19/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
Aberrant activation of GLI transcription factors has been implicated in the pathogenesis of different tumor types including pancreatic ductal adenocarcinoma. However, the mechanistic link with established drivers of this disease remains in part elusive. In this study, using a new genetically engineered mouse model overexpressing constitutively active mouse form of GLI2 and a combination of genome-wide assays, we provide evidence of a novel mechanism underlying the interplay between KRAS, a major driver of pancreatic ductal adenocarcinoma development, and GLI2 to control oncogenic gene expression. These mice, also expressing KrasG12D, show significantly reduced median survival rate and accelerated tumorigenesis compared with the KrasG12D only expressing mice. Analysis of the mechanism using RNA sequencing demonstrate higher levels of GLI2 targets, particularly tumor growth-promoting genes, including Ccnd1, N-Myc, and Bcl2, in KrasG12D mutant cells. Furthermore, chromatin immunoprecipitation sequencing studies showed that in these cells KrasG12D increases the levels of trimethylation of lysine 4 of the histone 3 (H3K4me3) at the promoter of GLI2 targets without affecting significantly the levels of other major active chromatin marks. Importantly, Gli2 knockdown reduces H3K4me3 enrichment and gene expression induced by mutant Kras. In summary, we demonstrate that Gli2 plays a significant role in pancreatic carcinogenesis by acting as a downstream effector of KrasG12D to control gene expression.
Collapse
Affiliation(s)
- Ashley N. Sigafoos
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Ezequiel J. Tolosa
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Ryan M. Carr
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Maite G. Fernandez-Barrena
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Luciana L. Almada
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - David R. Pease
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Tara L. Hogenson
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Glancis L. Raja Arul
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Fatemeh Mousavi
- Department of Physiology and Pharmacology, University of Western Ontario, London, Canada.
- Department of Oncology, University of Western Ontario, London, Canada.
| | - Sandhya Sen
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Renzo E. Vera
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - David L. Marks
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Luis F. Flores
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Kayla C. LaRue-Nolan
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Chen Wu
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - William R. Bamlet
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota.
| | - Anne M. Vrabel
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Hugues Sicotte
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota.
| | - Erin L. Schenk
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Thomas C. Smyrk
- Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota.
| | - Lizhi Zhang
- Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota.
| | - Kari G. Rabe
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota.
| | - Ann L. Oberg
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota.
| | | | - Eric Chevet
- Université de Rennes, CEDEX, Rennes, France.
| | | | | | - Marina P. di Magliano
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan.
| | - Sherine F. Elsawa
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire.
| | - Christopher L. Pin
- Department of Physiology and Pharmacology, University of Western Ontario, London, Canada.
- Department of Oncology, University of Western Ontario, London, Canada.
| | - Junhao Mao
- University of Massachusetts Medical School, Worcester, Massachusetts.
| | | | - Martin E. Fernandez-Zapico
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
22
|
Pinot M, Le Borgne R. Spatio-Temporal Regulation of Notch Activation in Asymmetrically Dividing Sensory Organ Precursor Cells in Drosophila melanogaster Epithelium. Cells 2024; 13:1133. [PMID: 38994985 PMCID: PMC11240559 DOI: 10.3390/cells13131133] [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: 05/31/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
The Notch communication pathway, discovered in Drosophila over 100 years ago, regulates a wide range of intra-lineage decisions in metazoans. The division of the Drosophila mechanosensory organ precursor is the archetype of asymmetric cell division in which differential Notch activation takes place at cytokinesis. Here, we review the molecular mechanisms by which epithelial cell polarity, cell cycle and intracellular trafficking participate in controlling the directionality, subcellular localization and temporality of mechanosensitive Notch receptor activation in cytokinesis.
Collapse
Affiliation(s)
| | - Roland Le Borgne
- Univ Rennes, Centre National de la Recherche Scientifique UMR 6290, IGDR (Institut de Génétique et Développement de Rennes), F-35000 Rennes, France
| |
Collapse
|
23
|
Kaufholz F, Ulrich J, Hakeemi MS, Bucher G. Temporal control of RNAi reveals both robust and labile feedback loops in the segmentation clock of the red flour beetle. Proc Natl Acad Sci U S A 2024; 121:e2318229121. [PMID: 38865277 PMCID: PMC11194489 DOI: 10.1073/pnas.2318229121] [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: 10/19/2023] [Accepted: 05/03/2024] [Indexed: 06/14/2024] Open
Abstract
Animals from all major clades have evolved a segmented trunk, reflected in the human spine or the insect segments. These units emerge during embryogenesis from a posterior segment addition zone (SAZ), where repetitive gene activity is regulated by a mechanism described by the clock and wavefront/speed gradient model. In the red flour beetle Tribolium castaneum, RNA interference (RNAi) has been used to continuously knock down the function of primary pair-rule genes (pPRGs), caudal or Wnt pathway components, which has led to the complete breakdown of segmentation. However, it has remained untested, if this breakdown was reversible by bringing the missing gene function back to the system. To fill this gap, we established a transgenic system in T. castaneum, which allows blocking an ongoing RNAi effect with temporal control by expressing a viral inhibitor of RNAi via heat shock. We show that the T. castaneum segmentation machinery was able to reestablish after RNAi targeting the pPRGs Tc-eve, Tc-odd, and Tc-runt was blocked. However, we observed no rescue after blocking RNAi targeting Wnt pathway components. We conclude that the insect segmentation system contains both robust feedback loops that can reestablish and labile feedback loops that break down irreversibly. This combination may reconcile conflicting needs of the system: Labile systems controlling initiation and maintenance of the SAZ ensure that only one SAZ is formed. Robust feedback loops confer developmental robustness toward external disturbances.
Collapse
Affiliation(s)
- Felix Kaufholz
- Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences, Göttingen37077, Germany
- Department of Evolutionary Developmental Genetics, University of Göttingen, Johann-Friedrich-Blumenbach Institute, Göttingen Center for Molecular Biosciences, Göttingen37077, Germany
| | - Julia Ulrich
- Department of Evolutionary Developmental Genetics, University of Göttingen, Johann-Friedrich-Blumenbach Institute, Göttingen Center for Molecular Biosciences, Göttingen37077, Germany
| | - Muhammad Salim Hakeemi
- Department of Evolutionary Developmental Genetics, University of Göttingen, Johann-Friedrich-Blumenbach Institute, Göttingen Center for Molecular Biosciences, Göttingen37077, Germany
| | - Gregor Bucher
- Department of Evolutionary Developmental Genetics, University of Göttingen, Johann-Friedrich-Blumenbach Institute, Göttingen Center for Molecular Biosciences, Göttingen37077, Germany
| |
Collapse
|
24
|
Baheux Blin M, Loreau V, Schnorrer F, Mangeol P. PatternJ: an ImageJ toolset for the automated and quantitative analysis of regular spatial patterns found in sarcomeres, axons, somites, and more. Biol Open 2024; 13:bio060548. [PMID: 38887972 PMCID: PMC11212633 DOI: 10.1242/bio.060548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
Regular spatial patterns are ubiquitous forms of organization in nature. In animals, regular patterns can be found from the cellular scale to the tissue scale, and from early stages of development to adulthood. To understand the formation of these patterns, how they assemble and mature, and how they are affected by perturbations, a precise quantitative description of the patterns is essential. However, accessible tools that offer in-depth analysis without the need for computational skills are lacking for biologists. Here, we present PatternJ, a novel toolset to analyze regular one-dimensional patterns precisely and automatically. This toolset, to be used with the popular imaging processing program ImageJ/Fiji, facilitates the extraction of key geometric features within and between pattern repeats in static images and time-lapse series. We validate PatternJ with simulated data and test it on images of sarcomeres from insect muscles and contracting cardiomyocytes, actin rings in neurons, and somites from zebrafish embryos obtained using confocal fluorescence microscopy, STORM, electron microscopy, and brightfield imaging. We show that the toolset delivers subpixel feature extraction reliably even with images of low signal-to-noise ratio. PatternJ's straightforward use and functionalities make it valuable for various scientific fields requiring quantitative one-dimensional pattern analysis, including the sarcomere biology of muscles or the patterning of mammalian axons, speeding up discoveries with the bonus of high reproducibility.
Collapse
Affiliation(s)
- Mélina Baheux Blin
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM-UMR7288, Marseille 13009, France
| | - Vincent Loreau
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM-UMR7288, Marseille 13009, France
| | - Frank Schnorrer
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM-UMR7288, Marseille 13009, France
| | - Pierre Mangeol
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM-UMR7288, Marseille 13009, France
| |
Collapse
|
25
|
Han R, Yang J, Zhu Y, Gan R. Wnt signaling in gastric cancer: current progress and future prospects. Front Oncol 2024; 14:1410513. [PMID: 38952556 PMCID: PMC11216096 DOI: 10.3389/fonc.2024.1410513] [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: 04/01/2024] [Accepted: 05/13/2024] [Indexed: 07/03/2024] Open
Abstract
Levels of the Wnt pathway components are abnormally altered in gastric cancer cells, leading to malignant cell proliferation, invasion and metastasis, poor prognosis and chemoresistance. Therefore, it is important to understand the mechanism of Wnt signaling pathway in gastric cancer. We systematically reviewed the molecular mechanisms of the Wnt pathway in gastric cancer development; and summarize the progression and the challenges of research on molecular agents of the Wnt pathway.
Collapse
Affiliation(s)
- Ruyue Han
- Cancer Research Institute, Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jing Yang
- Department of Gastroenterology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yingying Zhu
- Cancer Research Institute, Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Runliang Gan
- Cancer Research Institute, Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
26
|
Doe CQ, Thor S. 40 years of homeodomain transcription factors in the Drosophila nervous system. Development 2024; 151:dev202910. [PMID: 38819456 PMCID: PMC11190446 DOI: 10.1242/dev.202910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Drosophila nervous system development progresses through a series of well-characterized steps in which homeodomain transcription factors (HDTFs) play key roles during most, if not all, phases. Strikingly, although some HDTFs have only one role, many others are involved in multiple steps of the developmental process. Most Drosophila HDTFs engaged in nervous system development are conserved in vertebrates and often play similar roles during vertebrate development. In this Spotlight, we focus on the role of HDTFs during embryogenesis, where they were first characterized.
Collapse
Affiliation(s)
- Chris Q. Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | - Stefan Thor
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
27
|
Holstein TW. The significance of Ethel Browne's research on Hydra for the organizer concept. Cells Dev 2024; 178:203907. [PMID: 38417631 DOI: 10.1016/j.cdev.2024.203907] [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: 01/25/2024] [Revised: 02/17/2024] [Accepted: 02/17/2024] [Indexed: 03/01/2024]
Abstract
This article focuses on the roots of the organizer concept, which was developed by Hans Spemann during his studies of early embryonic development in amphibians. The fundamental properties of this axis-inducing signaling center have been elucidated through pioneering molecular research by Eddy De Robertis' laboratory and other researchers. Evolutionary comparisons have disclosed the presence of this signaling center, involving the interaction of Wnt and TGF-beta signaling pathways, existed not only in vertebrates but also in basal Metazoa such as Cnidaria. - Notably, even prior to the groundbreaking experiments conducted by Hilde Mangold and Hans Spemann, Ethel Browne conducted similar transplantation experiments on Hydra polyps. They were performed under the guidance of Thomas H Morgan and in the laboratory of Edmund B Wilson. Howard Lenhoff was the first to draw connections between Ethel Browne's transplantation experiments and those of Spemann and Mangold, igniting a vivid debate on the precedence of the organizer concept and its recognition in Nobel Prize considerations. This review critically compares the experiments conducted by Spemann and Mangold with those preceding their seminal work, concluding that the organizer concept clearly builds upon earlier research aimed at understanding developmental gradients, such as in the simple model Hydra. However, these approaches were not pursued further by Morgan, who shifted his focus towards unraveling the genetic control of development in flies, an approach that ultimately revealed the molecular identity of the Spemann organizer in vertebrates.
Collapse
Affiliation(s)
- Thomas W Holstein
- Heidelberg University, Centre for Organismal Studies (COS), Molecular Evolution and Genomics, Im Neuenheimer Feld 230, D-69120 Heidelberg, Germany.
| |
Collapse
|
28
|
Yang Y, Li S, Luo L. Responses of organ precursors to correct and incorrect inductive signals. Trends Cell Biol 2024; 34:484-495. [PMID: 37739814 DOI: 10.1016/j.tcb.2023.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/14/2023] [Accepted: 08/31/2023] [Indexed: 09/24/2023]
Abstract
During embryonic development, the inductive molecules produced by local origins normally arrive at their target tissues in a nondirectional, diffusion manner. The target organ precursor cells must correctly interpret these inductive signals to ensure proper specification/differentiation, which is dependent on two prerequisites: (i) obtaining cell-intrinsic competence; and (ii) receiving correct inductive signals while resisting incorrect ones. Gain of intrinsic competence could avoid a large number of misinductions because the incompetent cells are nonresponsive to inductive signals. However, in cases of different precursor cells with similar competence and located in close proximity, resistance to incorrect inductive signals is essential for accurate determination of cell fate. Here we outline the mechanisms of how organ precursors respond to correct and incorrect inductive signals.
Collapse
Affiliation(s)
- Yun Yang
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing, China
| | - Shuang Li
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing, China
| | - Lingfei Luo
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing, China; School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
29
|
Li Y, Jiang Z, Xu Y, Yan J, Wu Q, Huang S, Wang L, Xie Y, Wu X, Wang Y, Li Y, Fan X, Li F, Yuan W. Pygo-F773W Mutation Reveals Novel Functions beyond Wnt Signaling in Drosophila. Int J Mol Sci 2024; 25:5998. [PMID: 38892188 PMCID: PMC11172468 DOI: 10.3390/ijms25115998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Pygopus (Pygo) has been identified as a specific nuclear co-activator of the canonical Wingless (Wg)/Wnt signaling pathway in Drosophila melanogaster. Pygo proteins consist of two conserved domains: an N-terminal homologous domain (NHD) and a C-terminal plant homologous domain (PHD). The PHD's ability to bind to di- and trimethylated lysine 4 of histone H3 (H3K4me2/3) appears to be independent of Wnt signaling. There is ongoing debate regarding the significance of Pygo's histone-binding capacity. Drosophila Pygo orthologs have a tryptophan (W) > phenylalanine (F) substitution in their histone pocket-divider compared to vertebrates, leading to reduced histone affinity. In this research, we utilized CRISPR/Cas9 technology to introduce the Pygo-F773W point mutation in Drosophila, successfully establishing a viable homozygous Pygo mutant line for the first time. Adult mutant flies displayed noticeable abnormalities in reproduction, locomotion, heart function, and lifespan. RNA-seq and cluster analysis indicated that the mutation primarily affected pathways related to immunity, metabolism, and posttranslational modification in adult flies rather than the Wnt signaling pathway. Additionally, a reduction in H3K9 acetylation levels during the embryonic stage was observed in the mutant strains. These findings support the notion that Pygo plays a wider role in chromatin remodeling, with its involvement in Wnt signaling representing only a specific aspect of its chromatin-related functions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Fang Li
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (Y.L.); (Z.J.); (X.F.)
| | - Wuzhou Yuan
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (Y.L.); (Z.J.); (X.F.)
| |
Collapse
|
30
|
Li XC, Gandara L, Ekelöf M, Richter K, Alexandrov T, Crocker J. Rapid response of fly populations to gene dosage across development and generations. Nat Commun 2024; 15:4551. [PMID: 38811562 PMCID: PMC11137061 DOI: 10.1038/s41467-024-48960-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
Although the effects of genetic and environmental perturbations on multicellular organisms are rarely restricted to single phenotypic layers, our current understanding of how developmental programs react to these challenges remains limited. Here, we have examined the phenotypic consequences of disturbing the bicoid regulatory network in early Drosophila embryos. We generated flies with two extra copies of bicoid, which causes a posterior shift of the network's regulatory outputs and a decrease in fitness. We subjected these flies to EMS mutagenesis, followed by experimental evolution. After only 8-15 generations, experimental populations have normalized patterns of gene expression and increased survival. Using a phenomics approach, we find that populations were normalized through rapid increases in embryo size driven by maternal changes in metabolism and ovariole development. We extend our results to additional populations of flies, demonstrating predictability. Together, our results necessitate a broader view of regulatory network evolution at the systems level.
Collapse
Affiliation(s)
- Xueying C Li
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- College of Life Sciences, Beijing Normal University, Beijing, China.
| | - Lautaro Gandara
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Måns Ekelöf
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Kerstin Richter
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Theodore Alexandrov
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Molecular Medicine Partnership Unit between EMBL and Heidelberg University, Heidelberg, Germany
- BioInnovation Institute, Copenhagen, Denmark
| | - Justin Crocker
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| |
Collapse
|
31
|
Guo J, Yan W, Duan H, Wang D, Zhou Y, Feng D, Zheng Y, Zhou S, Liu G, Qin X. Therapeutic Effects of Natural Products on Liver Cancer and Their Potential Mechanisms. Nutrients 2024; 16:1642. [PMID: 38892575 PMCID: PMC11174683 DOI: 10.3390/nu16111642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Liver cancer ranks third globally among causes of cancer-related deaths, posing a significant public health challenge. However, current treatments are inadequate, prompting a growing demand for novel, safe, and effective therapies. Natural products (NPs) have emerged as promising candidates in drug development due to their diverse biological activities, low toxicity, and minimal side effects. This paper begins by reviewing existing treatment methods and drugs for liver cancer. It then summarizes the therapeutic effects of NPs sourced from various origins on liver cancer. Finally, we analyze the potential mechanisms of NPs in treating liver cancer, including inhibition of angiogenesis, migration, and invasion; regulation of the cell cycle; induction of apoptosis, autophagy, pyroptosis, and ferroptosis; influence on tumor metabolism; immune regulation; regulation of intestinal function; and regulation of key signaling pathways. This systematic review aims to provide a comprehensive overview of NPs research in liver cancer treatment, offering a foundation for further development and application in pharmaceuticals and functional foods.
Collapse
Affiliation(s)
- Jinhong Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Wenjie Yan
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Hao Duan
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Diandian Wang
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Yaxi Zhou
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Duo Feng
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing 100081, China;
| | - Yue Zheng
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China;
| | - Shiqi Zhou
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Gaigai Liu
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Xia Qin
- Graduate Department, Beijing Union University, Beijing 100101, China
| |
Collapse
|
32
|
Zhu W, Knoll P, Steinbock O. Exploring the Synthesis of Self-Organization and Active Motion. J Phys Chem Lett 2024; 15:5476-5487. [PMID: 38748082 DOI: 10.1021/acs.jpclett.4c01031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Proteins, genetic material, and membranes are fundamental to all known organisms, yet these components alone do not constitute life. Life emerges from the dynamic processes of self-organization, assembly, and active motion, suggesting the existence of similar artificial systems. Against this backdrop, our Perspective explores a variety of chemical phenomena illustrating how nonequilibrium self-organization and micromotors contribute to life-like behavior and functionalities. After explaining key terms, we discuss specific examples including enzymatic motion, diffusiophoretic and bubble-driven self-propulsion, pattern-forming reaction-diffusion systems, self-assembling inorganic aggregates, and hierarchically emergent phenomena. We also provide a roadmap for combining self-organization and active motion and discuss possible outcomes through biological analogs. We suggest that this research direction, deeply rooted in physical chemistry, offers opportunities for further development with broad impacts on related sciences and technologies.
Collapse
Affiliation(s)
- Wen Zhu
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306-4390, United States
| | - Pamela Knoll
- UK Centre for Astrobiology, School of Physics and Astronomy, Institute for Condensed Matter and Complex Systems, University of Edinburgh, Edinburgh EH9 3FD, U.K
| | - Oliver Steinbock
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306-4390, United States
| |
Collapse
|
33
|
Dong J, Scott TG, Mukherjee R, Guertin MJ. ZNF143 binds DNA and stimulates transcripstion initiation to activate and repress direct target genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.594008. [PMID: 38798607 PMCID: PMC11118474 DOI: 10.1101/2024.05.13.594008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Transcription factors bind to sequence motifs and act as activators or repressors. Transcription factors interface with a constellation of accessory cofactors to regulate distinct mechanistic steps to regulate transcription. We rapidly degraded the essential and ubiquitously expressed transcription factor ZNF143 to determine its function in the transcription cycle. ZNF143 facilitates RNA Polymerase initiation and activates gene expression. ZNF143 binds the promoter of nearly all its activated target genes. ZNF143 also binds near the site of genic transcription initiation to directly repress a subset of genes. Although ZNF143 stimulates initiation at ZNF143-repressed genes (i.e. those that increase expression upon ZNF143 depletion), the molecular context of binding leads to cis repression. ZNF143 competes with other more efficient activators for promoter access, physically occludes transcription initiation sites and promoter-proximal sequence elements, and acts as a molecular roadblock to RNA Polymerases during early elongation. The term context specific is often invoked to describe transcription factors that have both activation and repression functions. We define the context and molecular mechanisms of ZNF143-mediated cis activation and repression.
Collapse
Affiliation(s)
- Jinhong Dong
- Center for Cell Analysis and Modeling, University of Connecticut, Farmington, Connecticut, United States of America
| | - Thomas G Scott
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Rudradeep Mukherjee
- Center for Cell Analysis and Modeling, University of Connecticut, Farmington, Connecticut, United States of America
| | - Michael J Guertin
- Center for Cell Analysis and Modeling, University of Connecticut, Farmington, Connecticut, United States of America
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, Connecticut, United States of America
| |
Collapse
|
34
|
VanDerMolen KR, Newman MA, Breen PC, Huff LA, Dowen RH. Non-cell-autonomous regulation of mTORC2 by Hedgehog signaling maintains lipid homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592795. [PMID: 38766075 PMCID: PMC11100691 DOI: 10.1101/2024.05.06.592795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Organisms must appropriately allocate energetic resources between essential cellular processes to maintain homeostasis and in turn, maximize fitness. The nutritional and homeostatic regulators of energy homeostasis have been studied in detail; however, how developmental signals might impinge on these pathways to govern cellular metabolism is poorly understood. Here, we identify a non-canonical role for Hedgehog (Hh), a classic regulator of development, in maintaining intestinal lipid homeostasis in C. elegans . We find that expression of two Hh ligands, GRD-3 and GRD-4, is controlled by the LIN-29/EGR transcription factor in the hypodermis, where the Hh secretion factor CHE-14/Dispatched also facilitates non-cell autonomous Hh signaling. We demonstrate, using C. elegans and mouse hepatocytes, that Hh metabolic regulation does not occur through the canonical Hh transcription factor, TRA-1/GLI, but rather through non-canonical signaling that engages mTOR Complex 2 (mTORC2) in the intestine. Hh mutants display impaired lipid homeostasis, including reduced lipoprotein synthesis and fat accumulation, decreased growth, and upregulation of autophagy factors, mimicking loss of mTORC2. Additionally, we found that Hh inhibits p38 MAPK signaling in parallel to mTORC2 activation and that both pathways act together to modulate of lipid homeostasis. Our findings show a non-canonical role for Hedgehog signaling in lipid metabolism via regulation of core homeostatic pathways and reveal a new mechanism by which developmental timing events govern metabolic decisions.
Collapse
|
35
|
Gerges A, Canning U. Neuroblastoma and its Target Therapies: A Medicinal Chemistry Review. ChemMedChem 2024; 19:e202300535. [PMID: 38340043 DOI: 10.1002/cmdc.202300535] [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: 10/06/2023] [Revised: 01/22/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Neuroblastoma (NB) is a childhood malignant tumour belonging to a group of embryonic tumours originating from progenitor cells of the sympathoadrenal lineage. The heterogeneity of NB is reflected in the survival rates of those with low and intermediate risk diseases who have survival rates ranging from 85 to 90 %. However, for those identified with high-risk Stage 4 NB, the treatment options are much more limited. For this group, current treatment consists of immunotherapy (monoclonal antibodies) in combination with anti-cancer drugs and has a 40 to 50 % survival rate. The purpose of this review is to summarise NB research from a medicinal chemistry perspective and to highlight advances in targeted drug therapy in the field. The review examines the medicinal chemistry of a number of drugs tested in research, some of which are currently under clinical trial. It concludes by proposing that future medicinal chemistry research into NB should consider other possible target therapies and adopt a multi-target drug approach rather than a one-drug-one-target approach for improved efficacy and less drug-drug interaction for the treatment of NB Stage 4 (NBS4) patients.
Collapse
Affiliation(s)
- A Gerges
- Bioscience Department, London Metropolitan University, 166-220 Holloway Road, London, N7 8DB, England, United Kingdom
| | - U Canning
- Bioscience Department, London Metropolitan University, 166-220 Holloway Road, London, N7 8DB, England, United Kingdom
| |
Collapse
|
36
|
Tsukiyama T. New insights in ubiquitin-dependent Wnt receptor regulation in tumorigenesis. In Vitro Cell Dev Biol Anim 2024; 60:449-465. [PMID: 38383910 PMCID: PMC11126518 DOI: 10.1007/s11626-024-00855-w] [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: 10/24/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024]
Abstract
Wnt signaling plays a crucial role in embryonic development and homeostasis maintenance. Delicate and sensitive fine-tuning of Wnt signaling based on the proper timings and positions is required to balance cell proliferation and differentiation and maintain individual health. Therefore, homeostasis is broken by tissue hypoplasia or tumor formation once Wnt signal dysregulation disturbs the balance of cell proliferation. The well-known regulatory mechanism of Wnt signaling is the molecular reaction associated with the cytoplasmic accumulation of effector β-catenin. In addition to β-catenin, most Wnt effector proteins are also regulated by ubiquitin-dependent modification, both qualitatively and quantitatively. This review will explain the regulation of the whole Wnt signal in four regulatory phases, as well as the different ubiquitin ligases and the function of deubiquitinating enzymes in each phase. Along with the recent results, the mechanism by which RNF43 negatively regulates the surface expression of Wnt receptors, which has recently been well understood, will be detailed. Many RNF43 mutations have been identified in pancreatic and gastrointestinal cancers and examined for their functional alteration in Wnt signaling. Several mutations facilitate or activate the Wnt signal, reversing the RNF43 tumor suppressor function into an oncogene. RNF43 may simultaneously play different roles in classical multistep tumorigenesis, as both wild-type and mutant RNF43 suppress the p53 pathway. We hope that the knowledge obtained from further research in RNF43 will be applied to cancer treatment in the future despite the fully unclear function of RNF43.
Collapse
Affiliation(s)
- Tadasuke Tsukiyama
- Department of Biochemistry, Graduate School of Medicine, Hokkaido University, 15NW7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan.
| |
Collapse
|
37
|
Ambrogi M, Vezina CM. Roles of airway and intestinal epithelia in responding to pathogens and maintaining tissue homeostasis. Front Cell Infect Microbiol 2024; 14:1346087. [PMID: 38736751 PMCID: PMC11082347 DOI: 10.3389/fcimb.2024.1346087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/10/2024] [Indexed: 05/14/2024] Open
Abstract
Epithelial cells form a resilient barrier and orchestrate defensive and reparative mechanisms to maintain tissue stability. This review focuses on gut and airway epithelia, which are positioned where the body interfaces with the outside world. We review the many signaling pathways and mechanisms by which epithelial cells at the interface respond to invading pathogens to mount an innate immune response and initiate adaptive immunity and communicate with other cells, including resident microbiota, to heal damaged tissue and maintain homeostasis. We compare and contrast how airway and gut epithelial cells detect pathogens, release antimicrobial effectors, collaborate with macrophages, Tregs and epithelial stem cells to mount an immune response and orchestrate tissue repair. We also describe advanced research models for studying epithelial communication and behaviors during inflammation, tissue injury and disease.
Collapse
Affiliation(s)
| | - Chad M. Vezina
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
38
|
Zandawala M, Gera J. Leptin- and cytokine-like unpaired signaling in Drosophila. Mol Cell Endocrinol 2024; 584:112165. [PMID: 38266772 DOI: 10.1016/j.mce.2024.112165] [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: 11/15/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
Animals have evolved a multitude of signaling pathways that enable them to orchestrate diverse physiological processes to tightly regulate systemic homeostasis. This signaling is mediated by various families of peptide hormones and cytokines that are conserved across the animal kingdom. In this review, we primarily focus on the unpaired (Upd) family of proteins in Drosophila which are evolutionarily related to mammalian leptin and the cytokine interleukin 6. We summarize expression patterns of Upd in Drosophila and discuss the parallels in structure, signaling pathway, and functions between Upd and their mammalian counterparts. In particular, we focus on the roles of Upd in governing metabolic homeostasis, growth and development, and immune responses. We aim to stimulate future studies on leptin-like signaling in other phyla which can help bridge the evolutionary gap between insect Upd and vertebrate leptin and cytokines like interleukin 6.
Collapse
Affiliation(s)
- Meet Zandawala
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany; Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, 89557, USA.
| | - Jayati Gera
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| |
Collapse
|
39
|
Kong L, Jin X. Dysregulation of deubiquitination in breast cancer. Gene 2024; 902:148175. [PMID: 38242375 DOI: 10.1016/j.gene.2024.148175] [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: 10/25/2023] [Revised: 12/04/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
Breast cancer (BC) is a highly frequent malignant tumor that poses a serious threat to women's health and has different molecular subtypes, histological subtypes, and biological features, which act by activating oncogenic factors and suppressing cancer inhibitors. The ubiquitin-proteasome system (UPS) is the main process contributing to protein degradation, and deubiquitinases (DUBs) are reverse enzymes that counteract this process. There is growing evidence that dysregulation of DUBs is involved in the occurrence of BC. Herein, we review recent research findings in BC-associated DUBs, describe their nature, classification, and functions, and discuss the potential mechanisms of DUB-related dysregulation in BC. Furthermore, we present the successful treatment of malignant cancer with DUB inhibitors, as well as analyzing the status of targeting aberrant DUBs in BC.
Collapse
Affiliation(s)
- Lili Kong
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo 315211, Zhejiang, China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
40
|
Seli DA, Prendergast A, Ergun Y, Tyagi A, Taylor HS. High NaCl Concentrations in Water Are Associated with Developmental Abnormalities and Altered Gene Expression in Zebrafish. Int J Mol Sci 2024; 25:4104. [PMID: 38612913 PMCID: PMC11012806 DOI: 10.3390/ijms25074104] [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: 01/07/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Salt is frequently introduced in ecosystems, where it acts as a pollutant. This study examined how changes in salinity affect the survival and development of zebrafish from the two-cell to the blastocyst stage and from the blastocyst to the larval stage. Control zebrafish embryos were cultured in E3 medium containing 5 mM Sodium Chloride (NaCl), 0.17 mM Potassium Chloride (KCL), 0.33 mM Calcium Chloride (CaCl2), and 0.33 mM Magnesium Sulfade (MgSO4). Experiments were conducted using increasing concentrations of each individual salt at 5×, 10×, 50×, and 100× the concentration found in E3 medium. KCL, CaCl2, and MgSO4 did not result in lethal abnormalities and did not affect early embryo growth at any of the concentrations tested. Concentrations of 50× and 100× NaCl caused embryonic death in both stages of development. Concentrations of 5× and 10× NaCl resulted in uninflated swim bladders in 12% and 65% of larvae, compared to 4.2% of controls, and caused 1654 and 2628 genes to be differentially expressed in blastocysts, respectively. The ATM signaling pathway was affected, and the Sonic Hedgehog pathway genes Shh and Ptc1 implicated in swim bladder development were downregulated. Our findings suggest that increased NaCl concentrations may alter gene expression and cause developmental abnormalities in animals found in affected ecosystems.
Collapse
Affiliation(s)
- Denis A. Seli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06520, USA
| | - Andrew Prendergast
- Yale Zebrafish Phenotyping Core, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yagmur Ergun
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06520, USA
- IVIRMA Global Research Alliance, IVIRMA New Jersey, Marlton, NJ 08053, USA
| | - Antariksh Tyagi
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Hugh S. Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
41
|
Hu K, Li W, Ma S, Fang D, Xu J. The identification and classification of candidate genes during the zygotic genome activation in the mammals. ZYGOTE 2024; 32:119-129. [PMID: 38248909 DOI: 10.1017/s0967199423000631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Zygotic genome activation (ZGA) is a critical event in early embryonic development, and thousands of genes are involved in this delicate and sophisticated biological process. To date, however, only a handful of these genes have revealed their core functions in this special process, and therefore the roles of other genes still remain unclear. In the present study, we used previously published transcriptome profiling to identify potential key genes (candidate genes) in minor ZGA and major ZGA in both human and mouse specimens, and further identified the conserved genes across species. Our results showed that 887 and 760 genes, respectively, were thought to be specific to human and mouse in major ZGA, and the other 135 genes were considered to be orthologous genes. Moreover, the conserved genes were most enriched in rRNA processing in the nucleus and cytosol, ribonucleoprotein complex biogenesis, ribonucleoprotein complex assembly and ribosome large subunit biogenesis. The findings of this first comprehensive identification and characterization of candidate genes in minor and major ZGA provide relevant insights for future studies on ZGA.
Collapse
Affiliation(s)
- Kaiyue Hu
- Luoyang maternal and Child Health Hospital, 206, Tongqu Road, Luoyang, Henan, 47100China
| | - Wenbo Li
- The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, Henan, 450052China
| | - Shuxia Ma
- Luoyang maternal and Child Health Hospital, 206, Tongqu Road, Luoyang, Henan, 47100China
| | - Dong Fang
- Luoyang maternal and Child Health Hospital, 206, Tongqu Road, Luoyang, Henan, 47100China
| | - Jiawei Xu
- The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, Henan, 450052China
| |
Collapse
|
42
|
Singh R, Ray A. Therapeutic potential of hedgehog signaling in advanced cancer types. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 386:49-80. [PMID: 38782501 DOI: 10.1016/bs.ircmb.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
In this chapter, we have made an attempt to elucidate the relevance of hedgehog signaling pathway in tumorigenesis. Here, we have described different types of hedgehog signaling (canonical and non-canonical) with emphasis on the different mechanisms (mutation-driven, autocrine, paracrine and reverse paracrine) it adopts during tumorigenesis. We have discussed the role of hedgehog signaling in regulating cell proliferation, invasion and epithelial-to-mesenchymal transition in both local and advanced cancer types, as reported in different studies based on preclinical and clinical models. We have specifically addressed the role of hedgehog signaling in aggressive neuroendocrine tumors as well. We have also elaborated on the studies showing therapeutic relevance of the inhibitors of hedgehog signaling in cancer. Evidence of the crosstalk of hedgehog signaling components with other signaling pathways and treatment resistance due to tumor heterogeneity have also been briefly discussed. Together, we have tried to put forward a compilation of the studies on therapeutic potential of hedgehog signaling in various cancers, specifically aggressive tumor types with a perspective into what is lacking and demands further investigation.
Collapse
Affiliation(s)
- Richa Singh
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States.
| | - Anindita Ray
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| |
Collapse
|
43
|
Andreas E, Cummins B, Gedeon T. Quantifying robustness of the gap gene network. J Theor Biol 2024; 580:111720. [PMID: 38211890 DOI: 10.1016/j.jtbi.2024.111720] [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: 06/02/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/13/2024]
Abstract
Early development of Drosophila melanogaster (fruit fly) facilitated by the gap gene network has been shown to be incredibly robust, and the same patterns emerge even when the process is seriously disrupted. We investigate this robustness using a previously developed computational framework called DSGRN (Dynamic Signatures Generated by Regulatory Networks). Our mathematical innovations include the conceptual extension of this established modeling technique to enable modeling of spatially monotone environmental effects, as well as the development of a collection of graph theoretic robustness scores for network models. This allows us to rank order the robustness of network models of cellular systems where each cell contains the same genetic network topology but operates under a parameter regime that changes continuously from cell to cell. We demonstrate the power of this method by comparing the robustness of two previously introduced network models of gap gene expression along the anterior-posterior axis of the fruit fly embryo, both to each other and to a random sample of networks with same number of nodes and edges. We observe that there is a substantial difference in robustness scores between the two models. Our biological insight is that random network topologies are in general capable of reproducing complex patterns of expression, but that using measures of robustness to rank order networks permits a large reduction in hypothesis space for highly conserved systems such as developmental networks.
Collapse
Affiliation(s)
- Elizabeth Andreas
- Department of Mathematical Sciences, Montana State University, Bozeman, 59718, MT, USA.
| | - Breschine Cummins
- Department of Mathematical Sciences, Montana State University, Bozeman, 59718, MT, USA
| | - Tomáš Gedeon
- Department of Mathematical Sciences, Montana State University, Bozeman, 59718, MT, USA
| |
Collapse
|
44
|
Chi C, Roland TJ, Song K. Differentiation of Pluripotent Stem Cells for Disease Modeling: Learning from Heart Development. Pharmaceuticals (Basel) 2024; 17:337. [PMID: 38543122 PMCID: PMC10975450 DOI: 10.3390/ph17030337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 04/01/2024] Open
Abstract
Heart disease is a pressing public health problem and the leading cause of death worldwide. The heart is the first organ to gain function during embryogenesis in mammals. Heart development involves cell determination, expansion, migration, and crosstalk, which are orchestrated by numerous signaling pathways, such as the Wnt, TGF-β, IGF, and Retinoic acid signaling pathways. Human-induced pluripotent stem cell-based platforms are emerging as promising approaches for modeling heart disease in vitro. Understanding the signaling pathways that are essential for cardiac development has shed light on the molecular mechanisms of congenital heart defects and postnatal heart diseases, significantly advancing stem cell-based platforms to model heart diseases. This review summarizes signaling pathways that are crucial for heart development and discusses how these findings improve the strategies for modeling human heart disease in vitro.
Collapse
Affiliation(s)
- Congwu Chi
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Truman J. Roland
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Kunhua Song
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| |
Collapse
|
45
|
Harland RM, Wallingford JB. "And it was the worst possible result, because it actually worked:" An interview with Richard Harland. Cells Dev 2024:203910. [PMID: 38452855 DOI: 10.1016/j.cdev.2024.203910] [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: 01/10/2024] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
One hundred years ago, Hilde Mangold and Hans Spemann published their seminal paper on what came to be known as The Organizer, but seven decades would pass before the molecular basis of this remarkable phenomenon was revealed. Richard Harland and his laboratory played a key role in that discovery, and in this interview he discusses not just the science and the people but also other important factors like mental health and luck.
Collapse
Affiliation(s)
- Richard M Harland
- Dept. of Molecular and Cell Biology, University of California at Berkeley, United States of America
| | - John B Wallingford
- Dept. of Molecular Biosciences, University of Texas at Austin, United States of America.
| |
Collapse
|
46
|
Li Q, Li W, Hu K, Wang Y, Li Y, Xu J. A de novo variant in RERE causes autistic behavior by disrupting related genes and signaling pathway. Clin Genet 2024; 105:273-282. [PMID: 38018232 DOI: 10.1111/cge.14461] [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: 09/07/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023]
Abstract
Autism spectrum disorder (ASD) is a highly variable neurodevelopmental disorder that typically manifests childhood, characterized by a triad of symptoms: impaired social interaction, communication difficulties, and restricted interests with repetitive behaviors. De novo variants in related genes can cause ASD. We present the case of a 6-year-old Chinese boy with autistic behavior, including language communication impairments, intellectual disabilities, stunted development, and irritability in social interactions. Using Sanger sequencing, we confirmed a pathogenic in the RERE gene (NM_012102.4) (c.3732delC, p.Tyr1245Thrfs*12; EX21; Het). Subsequently, we generated an RERE point mutation cell line (ReMut) using CRISPR/Cas9 Targeted Genome Editing. Immunofluorescence was conducted to determine the location of the mutant RERE. RNA-sequencing and mass spectrometry analyses were performed to elucidate the ASD-related genes and signaling pathways disrupted by this variant in RERE. We identified 3790 differentially expressed genes and 684 differentially expressed proteins. The SHH signaling pathway was found to be downregulated, and the Hippo pathway was upregulated in ReMut. Genes implicated in autism, such as CNTNAP2, STX1A, FARP2, and GPC1, were significantly downregulated. Simultaneously, we noted alterations in HDAC1 and HDAC2, which are members of the WHHERE complex, suggesting their role in the pathogenesis of this patient. In conclusion, we report a de novo variant in RERE associated with autistic behavior. The finding that ASD is associated with RERE variants underscore the role of genetic factors in ASD and provides insights regarding the mechanisms underlying RERE variants in disease onset.
Collapse
Affiliation(s)
- Qian Li
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
- NHC Key Laboratory of Birth Defects Prevention, Zhengzhou, Henan, China
- Jining No. 1 People's Hospital, Jining, Shandong, China
| | - Wenbo Li
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
- NHC Key Laboratory of Birth Defects Prevention, Zhengzhou, Henan, China
| | - Kaiyue Hu
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
- NHC Key Laboratory of Birth Defects Prevention, Zhengzhou, Henan, China
| | - Yaqian Wang
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
- NHC Key Laboratory of Birth Defects Prevention, Zhengzhou, Henan, China
| | - Yang Li
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
- NHC Key Laboratory of Birth Defects Prevention, Zhengzhou, Henan, China
| | - Jiawei Xu
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
- NHC Key Laboratory of Birth Defects Prevention, Zhengzhou, Henan, China
| |
Collapse
|
47
|
Lee C, Maier W, Jiang YY, Nakano K, Lechtreck KF, Gaertig J. Global and local functions of the Fused kinase ortholog CdaH in intracellular patterning in Tetrahymena. J Cell Sci 2024; 137:jcs261256. [PMID: 37667859 PMCID: PMC10565251 DOI: 10.1242/jcs.261256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023] Open
Abstract
Ciliates assemble numerous microtubular structures into complex cortical patterns. During ciliate division, the pattern is duplicated by intracellular segmentation that produces a tandem of daughter cells. In Tetrahymena thermophila, the induction and positioning of the division boundary involves two mutually antagonistic factors: posterior CdaA (cyclin E) and anterior CdaI (Hippo kinase). Here, we characterized the related cdaH-1 allele, which confers a pleiotropic patterning phenotype including an absence of the division boundary and an anterior-posterior mispositioning of the new oral apparatus. CdaH is a Fused or Stk36 kinase ortholog that localizes to multiple sites that correlate with the effects of its loss, including the division boundary and the new oral apparatus. CdaH acts downstream of CdaA to induce the division boundary and drives asymmetric cytokinesis at the tip of the posterior daughter. CdaH both maintains the anterior-posterior position of the new oral apparatus and interacts with CdaI to pattern ciliary rows within the oral apparatus. Thus, CdaH acts at multiple scales, from induction and positioning of structures on the cell-wide polarity axis to local organelle-level patterning.
Collapse
Affiliation(s)
- Chinkyu Lee
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Wolfgang Maier
- Bioinformatics, University of Freiburg, 79110 Freiburg, Germany
| | - Yu-Yang Jiang
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Kentaro Nakano
- Degree Programs in Biology, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Karl F Lechtreck
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Jacek Gaertig
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
48
|
Tan QH, Otgonbaatar A, Kaur P, Ga AF, Harmston NP, Tolwinski NS. The Wnt Co-Receptor PTK7/Otk and Its Homolog Otk-2 in Neurogenesis and Patterning. Cells 2024; 13:365. [PMID: 38474329 PMCID: PMC10930971 DOI: 10.3390/cells13050365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Wnt signaling is a highly conserved metazoan pathway that plays a crucial role in cell fate determination and morphogenesis during development. Wnt ligands can induce disparate cellular responses. The exact mechanism behind these different outcomes is not fully understood but may be due to interactions with different receptors on the cell membrane. PTK7/Otk is a transmembrane receptor that is implicated in various developmental and physiological processes including cell polarity, cell migration, and invasion. Here, we examine two roles of Otk-1 and Otk-2 in patterning and neurogenesis. We find that Otk-1 is a positive regulator of signaling and Otk-2 functions as its inhibitor. We propose that PTK7/Otk functions in signaling, cell migration, and polarity contributing to the diversity of cellular responses seen in Wnt-mediated processes.
Collapse
Affiliation(s)
- Qian Hui Tan
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
| | - Agimaa Otgonbaatar
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
| | - Prameet Kaur
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
| | - Angelica Faye Ga
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
| | - Nathan P. Harmston
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
- Molecular Biosciences Division, Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Nicholas S. Tolwinski
- Division of Science, Yale-NUS College, Singapore 138527, Singapore (A.O.); (P.K.); (A.F.G.); (N.P.H.)
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
49
|
Thiery JP, Sheng G, Shu X, Runyan R. How studies in developmental epithelial-mesenchymal transition and mesenchymal-epithelial transition inspired new research paradigms in biomedicine. Development 2024; 151:dev200128. [PMID: 38300897 DOI: 10.1242/dev.200128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Epithelial-mesenchymal transition (EMT) and its reverse mechanism, mesenchymal-epithelial transition (MET), are evolutionarily conserved mechanisms initially identified in studies of early metazoan development. EMT may even have been established in choanoflagellates, the closest unicellular relative of Metazoa. These crucial morphological transitions operate during body plan formation and subsequently in organogenesis. These findings have prompted an increasing number of investigators in biomedicine to assess the importance of such mechanisms that drive epithelial cell plasticity in multiple diseases associated with congenital disabilities and fibrosis, and, most importantly, in the progression of carcinoma. EMT and MET also play crucial roles in regenerative medicine, notably by contributing epigenetic changes in somatic cells to initiate reprogramming into stem cells and their subsequent differentiation into distinct lineages.
Collapse
Affiliation(s)
| | - Guojun Sheng
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Xiaodong Shu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Raymond Runyan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
50
|
Li X, Levine M. What are tethering elements? Curr Opin Genet Dev 2024; 84:102151. [PMID: 38237456 DOI: 10.1016/j.gde.2023.102151] [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: 10/05/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 02/12/2024]
Abstract
High-resolution Micro-C maps identified a specialized class of regulatory DNAs termed 'tethering elements' (TEs) in Drosophila. These 300-500-bp elements facilitate specific long-range genomic associations or loops. The POZ-containing transcription factor GAF (GAGA-associated factor) contributes to loop formation. Tether-tether interactions accelerate Hox gene activation by distal enhancers, and coordinate transcription of duplicated genes (paralogs) through promoter-promoter associations. Some TEs engage in ultra-long-range enhancer-promoter and promoter-promoter interactions (meta-loops) in the Drosophila brain. We discuss the basis for tether-tether specificity and speculate on the occurrence of similar elements in vertebrate genomes.
Collapse
Affiliation(s)
- Xiao Li
- Lewis-Sigler Institute, Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA. https://twitter.com/@XiaoLi5525
| | - Michael Levine
- Lewis-Sigler Institute, Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA.
| |
Collapse
|