Pub Date : 2024-09-16DOI: 10.1016/j.ydbio.2024.09.005
Rohit Kumar, S. Srikrishna
Cachexia and systemic organ wasting are metabolic syndrome often associated with cancer. However, the exact mechanism of cancer associated cachexia like syndrome still remain elusive. In this study, we utilized a scribble (scrib) knockdown induced hindgut tumor to investigate the role of JNK kinase in cachexia like syndrome. Scrib, a cell polarity regulator, also acts as a tumor suppressor gene. Its loss and mis-localization are reported in various type of malignant cancer-like breast, colon and prostate cancer. The scrib knockdown flies exhibited male lethality, reduced life span, systemic organ wasting and increased pJNK level in hindgut of female flies. Interestingly, knocking down of human JNK Kinase analogue, hep, in scrib knockdown background in hindgut leads to restoration of loss of scrib mediated lethality and systemic organ wasting. Our data showed that scrib loss in hindgut is capable of inducing cancer associated cachexia like syndrome. Here, we firstly report that blocking the JNK signaling pathway effectively rescued the cancer cachexia induced by scrib knockdown, along with its associated gut barrier disruption. These findings have significantly advanced our understanding of cancer cachexia and have potential implications for the development of therapeutic strategies. However, more research is needed to fully understand the complex mechanisms underlying this condition.
{"title":"JNK Kinase regulates cachexia like syndrome in scribble knockdown tumor model of Drosophila melanogaster","authors":"Rohit Kumar, S. Srikrishna","doi":"10.1016/j.ydbio.2024.09.005","DOIUrl":"10.1016/j.ydbio.2024.09.005","url":null,"abstract":"<div><p>Cachexia and systemic organ wasting are metabolic syndrome often associated with cancer. However, the exact mechanism of cancer associated cachexia like syndrome still remain elusive. In this study, we utilized a <em>scribble</em> (<em>scrib</em>) knockdown induced hindgut tumor to investigate the role of JNK kinase in cachexia like syndrome. <em>Scrib</em>, a cell polarity regulator, also acts as a tumor suppressor gene. Its loss and mis-localization are reported in various type of malignant cancer-like breast, colon and prostate cancer. The <em>scrib</em> knockdown flies exhibited male lethality, reduced life span, systemic organ wasting and increased pJNK level in hindgut of female flies. Interestingly, knocking down of human JNK Kinase analogue, <em>hep</em>, in <em>scrib</em> knockdown background in hindgut leads to restoration of loss of <em>scrib</em> mediated lethality and systemic organ wasting. Our data showed that <em>scrib</em> loss in hindgut is capable of inducing cancer associated cachexia like syndrome. Here, we firstly report that blocking the JNK signaling pathway effectively rescued the cancer cachexia induced by <em>scrib</em> knockdown, along with its associated gut barrier disruption. These findings have significantly advanced our understanding of cancer cachexia and have potential implications for the development of therapeutic strategies. However, more research is needed to fully understand the complex mechanisms underlying this condition.</p></div>","PeriodicalId":11070,"journal":{"name":"Developmental biology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142242396","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-14DOI: 10.1016/S0012-1606(24)00226-4
{"title":"Outside Back Cover - Graphical abstract TOC/TOC in double column/Cover image legend if applicable, Bar code, Abstracting and Indexing information","authors":"","doi":"10.1016/S0012-1606(24)00226-4","DOIUrl":"10.1016/S0012-1606(24)00226-4","url":null,"abstract":"","PeriodicalId":11070,"journal":{"name":"Developmental biology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142229803","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-14DOI: 10.1016/j.ydbio.2024.09.003
Seo-Hee Cho , Ji Hyang Kim , Seonhee Kim
Combined removal of Crb1 and Crb2 from the developing optic vesicle evokes cellular and laminar disorganization by disrupting the apical cell-cell adhesion in developing retinal epithelium. As a result, at postnatal stages, affected mouse retinas show temporarily thickened, coarsely laminated retinas in addition to functional deficits, including a severely abnormal electroretinogram and decreased visual acuity. These features are reminiscent of Leber congenital amaurosis 8, which is caused in humans by subsets of Crb1 mutations. However, the cellular basis of the abnormalities in retinal progenitor cells (RPCs) that lead to retinal disorganization is largely unknown. In this study, we analyze specific features of RPCs in mutant retinas, including maintenance of the progenitor pool, cell cycle progression, cell cycle phase-dependent nuclear positioning, cell survival, and generation of mature retinal cell types. We find crucial defects in the mutant RPCs. Upon removal of CRB1 and CRB2, apical structures of the RPCs, determined by markers of cilia and centrosomes, are basally shifted. In addition, the positioning of the somata of the M-phase cells, normally localized at the apical surface of the retinal epithelium, is basally shifted in a nearly randomized pattern along the apico-basal axis. Consequently, we propose that positioning of RPCs is desynchronized from cell cycle phase and largely randomized during embryonic development at E17.5. Because the resultant postmitotic cells inevitably lose positional information, the outer and inner nuclear layers (ONL and INL) fail to form from ONBL during neonatal development and retinal cells become mixed locally and globally. Additional results of the lost tissue polarity in Crb1/Crb2 dKO retinas include atypical formation of heterotopic cell patches containing photoreceptor cells in the ganglion cell layer and acellular patches filled with neural processes. Collectively, these changes lead to a mouse model of LCA8-like pathology. LCA8-like pathology differs substantially from the well-characterized, broad range of degeneration phenotypes that arise during the differentiation of photoreceptor and Muller glial cells in retinitis pigmentosa 12, a closely related disease caused by mutated human Crb1.
Importantly, the present results suggest that Crb1/Crb2 serve indispensable functions in maintaining cell-cycle phase-dependent positioning of RPCs along the apico-basal axis, regulating cell cycle progression, and maintaining structural laminar integrity without significantly affecting the size of the RPC pools, generation of the subsets of the retinal cell types, or the distribution of cell cycle phases during RPC division. Taken together, these findings provide the crucial cellular basis of the thickening and severely disorganized lamination that are the unique features of the retinal abnormalities in LCA8 patients.
{"title":"Perturbed cell cycle phase-dependent positioning and nuclear migration of retinal progenitors along the apico-basal axis underlie global retinal disorganization in the LCA8-like mouse model","authors":"Seo-Hee Cho , Ji Hyang Kim , Seonhee Kim","doi":"10.1016/j.ydbio.2024.09.003","DOIUrl":"10.1016/j.ydbio.2024.09.003","url":null,"abstract":"<div><div>Combined removal of Crb1 and Crb2 from the developing optic vesicle evokes cellular and laminar disorganization by disrupting the apical cell-cell adhesion in developing retinal epithelium. As a result, at postnatal stages, affected mouse retinas show temporarily thickened, coarsely laminated retinas in addition to functional deficits, including a severely abnormal electroretinogram and decreased visual acuity. These features are reminiscent of Leber congenital amaurosis 8, which is caused in humans by subsets of Crb1 mutations. However, the cellular basis of the abnormalities in retinal progenitor cells (RPCs) that lead to retinal disorganization is largely unknown. In this study, we analyze specific features of RPCs in mutant retinas, including maintenance of the progenitor pool, cell cycle progression, cell cycle phase-dependent nuclear positioning, cell survival, and generation of mature retinal cell types. We find crucial defects in the mutant RPCs. Upon removal of CRB1 and CRB2, apical structures of the RPCs, determined by markers of cilia and centrosomes, are basally shifted. In addition, the positioning of the somata of the M-phase cells, normally localized at the apical surface of the retinal epithelium, is basally shifted in a nearly randomized pattern along the apico-basal axis. Consequently, we propose that positioning of RPCs is desynchronized from cell cycle phase and largely randomized during embryonic development at E17.5. Because the resultant postmitotic cells inevitably lose positional information, the outer and inner nuclear layers (ONL and INL) fail to form from ONBL during neonatal development and retinal cells become mixed locally and globally. Additional results of the lost tissue polarity in Crb1/Crb2 dKO retinas include atypical formation of heterotopic cell patches containing photoreceptor cells in the ganglion cell layer and acellular patches filled with neural processes. Collectively, these changes lead to a mouse model of LCA8-like pathology. LCA8-like pathology differs substantially from the well-characterized, broad range of degeneration phenotypes that arise during the differentiation of photoreceptor and Muller glial cells in retinitis pigmentosa 12, a closely related disease caused by mutated human Crb1.</div><div>Importantly, the present results suggest that Crb1/Crb2 serve indispensable functions in maintaining cell-cycle phase-dependent positioning of RPCs along the apico-basal axis, regulating cell cycle progression, and maintaining structural laminar integrity without significantly affecting the size of the RPC pools, generation of the subsets of the retinal cell types, or the distribution of cell cycle phases during RPC division. Taken together, these findings provide the crucial cellular basis of the thickening and severely disorganized lamination that are the unique features of the retinal abnormalities in LCA8 patients.</div></div>","PeriodicalId":11070,"journal":{"name":"Developmental biology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142281783","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-13DOI: 10.1016/j.ydbio.2024.09.004
Armin P. Moczek
Understanding the origins of novel complex traits, the evolutionary transitions they enabled, and how those shaped the subsequent course of evolution, are all foundational objectives of evolutionary biology. Yet how developmental systems may transform to yield the first eye, limb, or placenta remains poorly understood. Seminal work by Courtney Clark-Hachtel, David Linz, and Yoshinori Tomoyasu published in the Proceedings of the National Academy of Sciences in 2013 used the origins of insect wings - one of the most impactful innovations of animal life on Earth - to provide both a case study and a new way of thinking of how novel complex traits may come into being. This paradigm-setting study not only transformed the way we view insect wings, their origins, and their affinities to other morphological structures; even more importantly, it created entryways to envision innovation as emerging gradually, not somehow divorced from ancestral homology, but through it via the differential modification, fusion, and elaboration of ancestral component parts. In a conceptual universe of descent with modification, where everything new must ultimately emerge from the old, this work thereby established a powerful bridge connecting ancestral homology and novelty through a gradual process of innovation, sparking much creative and groundbreaking work to follow since its publication just a little over a decade ago.
{"title":"Taking flight!","authors":"Armin P. Moczek","doi":"10.1016/j.ydbio.2024.09.004","DOIUrl":"10.1016/j.ydbio.2024.09.004","url":null,"abstract":"<div><p>Understanding the origins of novel complex traits, the evolutionary transitions they enabled, and how those shaped the subsequent course of evolution, are all foundational objectives of evolutionary biology. Yet how developmental systems may transform to yield the first eye, limb, or placenta remains poorly understood. Seminal work by Courtney Clark-Hachtel, David Linz, and Yoshinori Tomoyasu published in the <em>Proceedings of the National Academy of Sciences</em> in 2013 used the origins of insect wings - one of the most impactful innovations of animal life on Earth - to provide both a case study and a new way of thinking of how novel complex traits may come into being. This paradigm-setting study not only transformed the way we view insect wings, their origins, and their affinities to other morphological structures; even more importantly, it created entryways to envision innovation as emerging gradually, not somehow divorced from ancestral homology, but <em>through it</em> via the differential modification, fusion, and elaboration of ancestral component parts. In a conceptual universe of <em>descent with modification</em>, where everything new must ultimately emerge from the old, this work thereby established a powerful bridge connecting ancestral homology and novelty through a gradual process of innovation, sparking much creative and groundbreaking work to follow since its publication just a little over a decade ago.</p></div>","PeriodicalId":11070,"journal":{"name":"Developmental biology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142242395","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-06DOI: 10.1016/j.ydbio.2024.09.002
Camilo V. Echeverria Jr. , Tess A. Leathers , Crystal D. Rogers
The choice of fixation method significantly impacts tissue morphology and visualization of gene expression and proteins after in situ hybridization chain reaction (HCR) or immunohistochemistry (IHC), respectively. In this study, we compared the effects of paraformaldehyde (PFA) and trichloroacetic acid (TCA) fixation techniques prior to HCR and IHC on chicken embryos. Our findings underscore the importance of optimizing fixation methods for accurate visualization and subsequent interpretation of HCR and IHC results, with implications for probe and antibody validation and tissue-specific protein localization studies. We found that TCA fixation resulted in larger and more circular nuclei and neural tubes compared to PFA fixation. Additionally, TCA fixation altered the subcellular fluorescence signal intensity of various proteins, including transcription factors, cytoskeletal proteins, and cadherins. Notably, TCA fixation revealed protein signals in tissues that may be inaccessible with PFA fixation. In contrast, TCA fixation proved ineffective for mRNA visualization. These results highlight the need for optimization of fixation protocols depending on the target and model system, emphasizing the importance of methodological considerations in biological analyses.
{"title":"Comparative analysis of fixation techniques for signal detection in avian embryos","authors":"Camilo V. Echeverria Jr. , Tess A. Leathers , Crystal D. Rogers","doi":"10.1016/j.ydbio.2024.09.002","DOIUrl":"10.1016/j.ydbio.2024.09.002","url":null,"abstract":"<div><p>The choice of fixation method significantly impacts tissue morphology and visualization of gene expression and proteins after <em>in situ</em> hybridization chain reaction (HCR) or immunohistochemistry (IHC), respectively. In this study, we compared the effects of paraformaldehyde (PFA) and trichloroacetic acid (TCA) fixation techniques prior to HCR and IHC on chicken embryos. Our findings underscore the importance of optimizing fixation methods for accurate visualization and subsequent interpretation of HCR and IHC results, with implications for probe and antibody validation and tissue-specific protein localization studies. We found that TCA fixation resulted in larger and more circular nuclei and neural tubes compared to PFA fixation. Additionally, TCA fixation altered the subcellular fluorescence signal intensity of various proteins, including transcription factors, cytoskeletal proteins, and cadherins. Notably, TCA fixation revealed protein signals in tissues that may be inaccessible with PFA fixation. In contrast, TCA fixation proved ineffective for mRNA visualization. These results highlight the need for optimization of fixation protocols depending on the target and model system, emphasizing the importance of methodological considerations in biological analyses.</p></div>","PeriodicalId":11070,"journal":{"name":"Developmental biology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0012160624002276/pdfft?md5=a5f642eeed16694517cf22ae7be25c9c&pid=1-s2.0-S0012160624002276-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153396","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-05DOI: 10.1016/j.ydbio.2024.09.001
B. Camia , M. Longo , A. Bergonzi , I. Dezza , M. Biggiogera , C.A. Redi , A. Casasco , M. Monti
Clathrin is one of the leading players in the endocytic process during oocyte maturation. Immunofluorescence and transmission electron analysis on fully-grown germinal vesicle (GV) mouse oocytes shows Clathrin localization on the cortical region with three peculiar patterns: complete, incomplete, and half-moon. The first configuration is characterized by Clathrin lattices along the cortex; the second is represented by Clathrin lattices interrupted by invaginations forming coated vesicles as an indication of active endocytosis. The half-moon profile, the less frequent but the most interesting one, refers to Clathrin lattices distributed to one-half of the cell.
The in vivo analysis of organelles' positioning and cytoplasmic rearrangements, performed to understand the possible relation between endocytosis and oocyte maturation, suggests that the half-moon pattern indicates those fully-grown oocytes that may have likely undergone Germinal Vesicle Breakdown, MI, and MII. Our results show that, before oocytes undergo maturation, Clathrin localizes on the side of the cell, opposite to future spindle migration, thus marking spindle orientation in mouse oocytes.
{"title":"The localization and function of the moonlighting protein Clathrin during oocyte maturation","authors":"B. Camia , M. Longo , A. Bergonzi , I. Dezza , M. Biggiogera , C.A. Redi , A. Casasco , M. Monti","doi":"10.1016/j.ydbio.2024.09.001","DOIUrl":"10.1016/j.ydbio.2024.09.001","url":null,"abstract":"<div><p>Clathrin is one of the leading players in the endocytic process during oocyte maturation. Immunofluorescence and transmission electron analysis on fully-grown germinal vesicle (GV) mouse oocytes shows Clathrin localization on the cortical region with three peculiar patterns: complete, incomplete, and half-moon. The first configuration is characterized by Clathrin lattices along the cortex; the second is represented by Clathrin lattices interrupted by invaginations forming coated vesicles as an indication of active endocytosis. The half-moon profile, the less frequent but the most interesting one, refers to Clathrin lattices distributed to one-half of the cell.</p><p>The <em>in vivo</em> analysis of organelles' positioning and cytoplasmic rearrangements, performed to understand the possible relation between endocytosis and oocyte maturation, suggests that the half-moon pattern indicates those fully-grown oocytes that may have likely undergone Germinal Vesicle Breakdown, MI, and MII. Our results show that, before oocytes undergo maturation, Clathrin localizes on the side of the cell, opposite to future spindle migration, thus marking spindle orientation in mouse oocytes.</p></div>","PeriodicalId":11070,"journal":{"name":"Developmental biology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145372","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-28DOI: 10.1016/j.ydbio.2024.08.006
Matthew T. Biegler , Kirubel Belay , Wei Wang , Christina Szialta , Paul Collier , Ji-Dung Luo , Bettina Haase , Gregory L. Gedman , Asha V. Sidhu , Elijah Harter , Carlos Rivera-López , Kwame Amoako-Boadu , Olivier Fedrigo , Hagen U. Tilgner , Thomas Carroll , Erich D. Jarvis , Anna L. Keyte
The diversity of germ cell developmental strategies has been well documented across many vertebrate clades. However, much of our understanding of avian primordial germ cell (PGC) specification and differentiation has derived from only one species, the chicken (Gallus gallus). Of the three major classes of birds, chickens belong to Galloanserae, representing less than 4% of species, while nearly 95% of extant bird species belong to Neoaves. This represents a significant gap in our knowledge of germ cell development across avian species, hampering efforts to adapt genome editing and reproductive technologies developed in chicken to other birds. We therefore applied single-cell RNA sequencing to investigate inter-species differences in germ cell development between chicken and zebra finch (Taeniopygia castanotis), a Neoaves songbird species and a common model of vocal learning. Analysis of early embryonic male and female gonads revealed the presence of two distinct early germ cell types in zebra finch and only one in chicken. Both germ cell types expressed zebra finch Germline Restricted Chromosome (GRC) genes, present only in songbirds among birds. One of the zebra finch germ cell types expressed the canonical PGC markers, as did chicken, but with expression differences in several signaling pathways and biological processes. The second zebra finch germ cell cluster was marked by proliferation and fate determination markers, indicating beginning of differentiation. Notably, these two zebra finch germ cell populations were present in both male and female zebra finch gonads as early as HH25. Using additional chicken developmental stages, similar germ cell heterogeneity was identified in the more developed gonads of females, but not males. Overall, our study demonstrates a substantial heterochrony in zebra finch germ cell development compared to chicken, indicating a richer diversity of avian germ cell developmental strategies than previously known.
{"title":"Pronounced early differentiation underlies zebra finch gonadal germ cell development","authors":"Matthew T. Biegler , Kirubel Belay , Wei Wang , Christina Szialta , Paul Collier , Ji-Dung Luo , Bettina Haase , Gregory L. Gedman , Asha V. Sidhu , Elijah Harter , Carlos Rivera-López , Kwame Amoako-Boadu , Olivier Fedrigo , Hagen U. Tilgner , Thomas Carroll , Erich D. Jarvis , Anna L. Keyte","doi":"10.1016/j.ydbio.2024.08.006","DOIUrl":"10.1016/j.ydbio.2024.08.006","url":null,"abstract":"<div><div>The diversity of germ cell developmental strategies has been well documented across many vertebrate clades. However, much of our understanding of avian primordial germ cell (PGC) specification and differentiation has derived from only one species, the chicken (<em>Gallus gallus</em>). Of the three major classes of birds, chickens belong to Galloanserae, representing less than 4% of species, while nearly 95% of extant bird species belong to Neoaves. This represents a significant gap in our knowledge of germ cell development across avian species, hampering efforts to adapt genome editing and reproductive technologies developed in chicken to other birds. We therefore applied single-cell RNA sequencing to investigate inter-species differences in germ cell development between chicken and zebra finch (<em>Taeniopygia castanotis</em>), a Neoaves songbird species and a common model of vocal learning. Analysis of early embryonic male and female gonads revealed the presence of two distinct early germ cell types in zebra finch and only one in chicken. Both germ cell types expressed zebra finch Germline Restricted Chromosome (GRC) genes, present only in songbirds among birds. One of the zebra finch germ cell types expressed the canonical PGC markers, as did chicken, but with expression differences in several signaling pathways and biological processes. The second zebra finch germ cell cluster was marked by proliferation and fate determination markers, indicating beginning of differentiation. Notably, these two zebra finch germ cell populations were present in both male and female zebra finch gonads as early as HH25. Using additional chicken developmental stages, similar germ cell heterogeneity was identified in the more developed gonads of females, but not males. Overall, our study demonstrates a substantial heterochrony in zebra finch germ cell development compared to chicken, indicating a richer diversity of avian germ cell developmental strategies than previously known.</div></div>","PeriodicalId":11070,"journal":{"name":"Developmental biology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142105157","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-23DOI: 10.1016/j.ydbio.2024.08.012
David Lagman , Anthony Leon , Nadia Cieminska , Wei Deng , Marios Chatzigeorgiou , Simon Henriet , Daniel Chourrout
Larvacean tunicates feature a spectacular innovation not seen in other animals - the trunk oikoplastic epithelium (OE). This epithelium produces a house, a large and complex extracellular structure used for filtering and concentrating food particles. Previously we identified several homeobox transcription factor genes expressed during early OE patterning. Among these are two Pax3/7 copies that we named pax37A and pax37B. The vertebrate homologs, PAX3 and PAX7 are involved in developmental processes related to neural crest and muscles. In the ascidian tunicate Ciona intestinalis, Pax3/7 plays a role in the development of cells deriving from the neural plate border, including trunk epidermal sensory neurons and tail nerve cord neurons, as well as in the neural tube closure. Here we have investigated the roles of Oikopleura dioica pax37A and pax37B in the development of the OE, by using CRISPR-Cas9 mutant lines and analyzing scRNA-seq data from wild-type animals. We found that pax37B but not pax37A is essential for the differentiation of cell fields that produce the food concentrating filter of the house: the anterior Fol, giant Fol and Nasse cells. Trajectory analysis supported a neuroepithelial-like or a preplacodal ectoderm transcriptional signature in these cells. We propose that the highly specialized secretory epithelial cells of the Fol region either maintained or evolved neuroepithelial features. This is supported by a fragmented gene regulatory network involved in their development that also operates in ascidian epidermal neurons.
{"title":"Pax3/7 gene function in Oikopleura dioica supports a neuroepithelial-like origin for its house-making Fol territory","authors":"David Lagman , Anthony Leon , Nadia Cieminska , Wei Deng , Marios Chatzigeorgiou , Simon Henriet , Daniel Chourrout","doi":"10.1016/j.ydbio.2024.08.012","DOIUrl":"10.1016/j.ydbio.2024.08.012","url":null,"abstract":"<div><p>Larvacean tunicates feature a spectacular innovation not seen in other animals - the trunk oikoplastic epithelium (OE). This epithelium produces a house, a large and complex extracellular structure used for filtering and concentrating food particles. Previously we identified several homeobox transcription factor genes expressed during early OE patterning. Among these are two <em>Pax3/7</em> copies that we named <em>pax37A</em> and <em>pax37B</em>. The vertebrate homologs, <em>PAX3</em> and <em>PAX7</em> are involved in developmental processes related to neural crest and muscles. In the ascidian tunicate <em>Ciona intestinalis</em>, <em>Pax3/7</em> plays a role in the development of cells deriving from the neural plate border, including trunk epidermal sensory neurons and tail nerve cord neurons, as well as in the neural tube closure. Here we have investigated the roles of <em>Oikopleura dioica pax37A</em> and <em>pax37B</em> in the development of the OE, by using CRISPR-Cas9 mutant lines and analyzing scRNA-seq data from wild-type animals. We found that <em>pax37B</em> but not <em>pax37A</em> is essential for the differentiation of cell fields that produce the food concentrating filter of the house: the anterior Fol, giant Fol and Nasse cells. Trajectory analysis supported a neuroepithelial-like or a preplacodal ectoderm transcriptional signature in these cells. We propose that the highly specialized secretory epithelial cells of the Fol region either maintained or evolved neuroepithelial features. This is supported by a fragmented gene regulatory network involved in their development that also operates in ascidian epidermal neurons.</p></div>","PeriodicalId":11070,"journal":{"name":"Developmental biology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-08-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0012160624002173/pdfft?md5=64a013109c29b07527c91d9aace535e5&pid=1-s2.0-S0012160624002173-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142055207","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-22DOI: 10.1016/j.ydbio.2024.08.007
Salvador Rojas , Paul G. Barghouth , Peter Karabinis , Néstor J. Oviedo
The precise regulation of transcription is required for embryonic development, adult tissue turnover, and regeneration. Epigenetic modifications play a crucial role in orchestrating and regulating the transcription of genes. These modifications are important in the transition of pluripotent stem cells and their progeny. Methylation, a key epigenetic modification, influences gene expression through changes in DNA methylation. Work in different organisms has shown that the DNA methyltransferase-1-associated protein (DMAP1) may associate with other molecules to repress transcription through DNA methylation. Thus, DMAP1 is a versatile protein implicated in a myriad of events, including pluripotency maintenance, DNA damage repair, and tumor suppression. While DMAP1 has been extensively studied in vitro, its complex regulation in the context of the adult organism remains unclear. To gain insights into the possible roles of DMAP1 at the organismal level, we used planarian flatworms that possess remarkable regenerative capabilities driven by pluripotent stem cells called neoblast. Our findings demonstrate the evolutionary conservation of DMAP1 in the planarian Schmidtea mediterranea. Functional disruption of DMAP1 through RNA interference revealed its critical role in tissue maintenance, neoblast differentiation, and regeneration in S. mediterranea. Moreover, our analysis unveiled a novel function for DMAP1 in regulating cell death in response to DNA damage and influencing the expression of axial polarity markers. Our findings provide a simplified paradigm for studying DMAP1's function in adult tissues.
胚胎发育、成体组织更替和再生都需要对转录进行精确调控。表观遗传修饰在协调和调节基因转录方面发挥着至关重要的作用。这些修饰在多能干细胞及其后代的转变过程中非常重要。甲基化是一种关键的表观遗传修饰,通过 DNA 甲基化的变化影响基因表达。不同生物体的研究表明,DNA甲基转移酶-1相关蛋白(DMAP1)可与其他分子结合,通过DNA甲基化抑制转录。因此,DMAP1 是一种多用途蛋白质,与包括多能性维持、DNA 损伤修复和肿瘤抑制在内的无数事件有关。虽然 DMAP1 已在体外得到广泛研究,但其在成体中的复杂调控仍不清楚。为了深入了解DMAP1在生物体水平上可能发挥的作用,我们利用了扁平足类动物,这种动物在多能干细胞(称为neoblast)的驱动下具有非凡的再生能力。我们的研究结果表明,DMAP1在扁形动物Schmidtea mediterranea中具有进化保守性。通过RNA干扰对DMAP1进行功能性破坏,发现了它在S. mediterranea的组织维持、新母细胞分化和再生中的关键作用。此外,我们的分析还揭示了 DMAP1 在 DNA 损伤时调控细胞死亡以及影响轴向极性标记表达的新功能。我们的发现为研究DMAP1在成体组织中的功能提供了一个简化的范例。
{"title":"The DNA methyltransferase DMAP1 is required for tissue maintenance and planarian regeneration","authors":"Salvador Rojas , Paul G. Barghouth , Peter Karabinis , Néstor J. Oviedo","doi":"10.1016/j.ydbio.2024.08.007","DOIUrl":"10.1016/j.ydbio.2024.08.007","url":null,"abstract":"<div><p>The precise regulation of transcription is required for embryonic development, adult tissue turnover, and regeneration. Epigenetic modifications play a crucial role in orchestrating and regulating the transcription of genes. These modifications are important in the transition of pluripotent stem cells and their progeny. Methylation, a key epigenetic modification, influences gene expression through changes in DNA methylation. Work in different organisms has shown that the DNA methyltransferase-1-associated protein (DMAP1) may associate with other molecules to repress transcription through DNA methylation. Thus, DMAP1 is a versatile protein implicated in a myriad of events, including pluripotency maintenance, DNA damage repair, and tumor suppression. While DMAP1 has been extensively studied <em>in vitro</em>, its complex regulation in the context of the adult organism remains unclear. To gain insights into the possible roles of DMAP1 at the organismal level, we used planarian flatworms that possess remarkable regenerative capabilities driven by pluripotent stem cells called neoblast. Our findings demonstrate the evolutionary conservation of DMAP1 in the planarian <em>Schmidtea mediterranea</em>. Functional disruption of DMAP1 through RNA interference revealed its critical role in tissue maintenance, neoblast differentiation, and regeneration in <em>S. mediterranea</em>. Moreover, our analysis unveiled a novel function for DMAP1 in regulating cell death in response to DNA damage and influencing the expression of axial polarity markers. Our findings provide a simplified paradigm for studying DMAP1's function in adult tissues.</p></div>","PeriodicalId":11070,"journal":{"name":"Developmental biology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142046451","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-21DOI: 10.1016/j.ydbio.2024.08.008
Corine M. van der Weele, Katrina C. Hospes, Katherine E. Rowe, William R. Jeffery
The teleost Astyanax mexicanus consists of surface dwelling (surface fish) and cave dwelling (cavefish) forms. Cavefish have evolved in subterranean habitats characterized by reduced oxygen levels (hypoxia) and exhibit a subset of phenotypic traits controlled by increased Sonic hedgehog (Shh) signaling along the embryonic midline. The enhancement of primitive hematopoietic domains, which are formed bilaterally in the anterior and posterior lateral plate mesoderm, are responsible for the development of more larval erythrocytes in cavefish relative to surface fish. In this study, we determine the role of hypoxia and Shh signaling in the development and evolution of primitive hematopoiesis in cavefish. We show that hypoxia treatment during embryogenesis increases primitive hematopoiesis and erythrocyte development in surface fish. We also demonstrate that upregulation of Shh midline signaling by the Smoothened agonist SAG increases primitive hematopoiesis and erythrocyte development in surface fish, whereas Shh downregulation via treatment with the Smoothened inhibitor cyclopamine decreases these traits in cavefish. Together these results suggest that hematopoietic enhancement is regulated by hypoxia and Shh signaling. Lastly, we demonstrate that hypoxia enhances expression of Shh signaling along the midline of surface fish embryos. We conclude that hypoxia-mediated Shh plasticity may be a driving force for the adaptive evolution of primitive hematopoiesis and erythrocyte development in cavefish.
{"title":"Hypoxia-sonic hedgehog axis as a driver of primitive hematopoiesis development and evolution in cavefish","authors":"Corine M. van der Weele, Katrina C. Hospes, Katherine E. Rowe, William R. Jeffery","doi":"10.1016/j.ydbio.2024.08.008","DOIUrl":"10.1016/j.ydbio.2024.08.008","url":null,"abstract":"<div><p>The teleost <em>Astyanax mexicanus</em> consists of surface dwelling (surface fish) and cave dwelling (cavefish) forms. Cavefish have evolved in subterranean habitats characterized by reduced oxygen levels (hypoxia) and exhibit a subset of phenotypic traits controlled by increased Sonic hedgehog (Shh) signaling along the embryonic midline. The enhancement of primitive hematopoietic domains, which are formed bilaterally in the anterior and posterior lateral plate mesoderm, are responsible for the development of more larval erythrocytes in cavefish relative to surface fish. In this study, we determine the role of hypoxia and Shh signaling in the development and evolution of primitive hematopoiesis in cavefish. We show that hypoxia treatment during embryogenesis increases primitive hematopoiesis and erythrocyte development in surface fish. We also demonstrate that upregulation of Shh midline signaling by the Smoothened agonist SAG increases primitive hematopoiesis and erythrocyte development in surface fish, whereas Shh downregulation via treatment with the Smoothened inhibitor cyclopamine decreases these traits in cavefish. Together these results suggest that hematopoietic enhancement is regulated by hypoxia and Shh signaling. Lastly, we demonstrate that hypoxia enhances expression of Shh signaling along the midline of surface fish embryos. We conclude that hypoxia-mediated Shh plasticity may be a driving force for the adaptive evolution of primitive hematopoiesis and erythrocyte development in cavefish.</p></div>","PeriodicalId":11070,"journal":{"name":"Developmental biology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142021438","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}