1
|
Duan Z, Cao H, Xu M, Huang W, Peng Y, Shen Z, Hu S, Han Y. Chemogenetic ablation and regeneration of arterial valve in zebrafish. Biochem Biophys Res Commun 2025; 762:151786. [PMID: 40209504 DOI: 10.1016/j.bbrc.2025.151786] [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: 03/28/2025] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Aortic valve diseases are prevalent and severe cardiovascular conditions with limited treatment options beyond surgical intervention. The ability to regenerate aortic valves would revolutionize the management of these diseases. Utilizing the zebrafish model, which possesses remarkable regenerative capacities, we developed a chemogenetic arterial valve ablation model using a zebrafish-codon optimized nitroreductase. We found that arterial valve ablation led to blood regurgitation and impaired cardiac function, which are commonly associated with aortic valve diseases. Following ablation, zebrafish arterial valve could fully regenerate and restore valvular and cardiac function. Moreover, suppression of blood flow significantly impedes valve regeneration, indicating the importance of hemodynamic forces in this process. Our research has successfully established a robust aortic valve injury model to study the cellular and molecular mechanisms underlying its regeneration process which will facilitate the development of innovative therapeutic strategies tailored for aortic valve diseases.
Collapse
Affiliation(s)
- Zongyi Duan
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Hao Cao
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Mengting Xu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Wenping Huang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Yuanhui Peng
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Zhenya Shen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Shijun Hu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Yanchao Han
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
2
|
Kelly RG. On the cusps of the second heart field: insights from zebrafish into arterial valve origins and disease. Cardiovasc Res 2025; 121:6-7. [PMID: 39569430 DOI: 10.1093/cvr/cvae249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 11/22/2024] Open
Affiliation(s)
- Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| |
Collapse
|
3
|
Derrick CJ, Eley L, Alqahtani A, Henderson DJ, Chaudhry B. Zebrafish arterial valve development occurs through direct differentiation of second heart field progenitors. Cardiovasc Res 2025; 121:157-173. [PMID: 39460530 PMCID: PMC11998914 DOI: 10.1093/cvr/cvae230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/03/2024] [Accepted: 09/15/2024] [Indexed: 10/28/2024] Open
Abstract
AIMS Bicuspid aortic valve (BAV) is the most common congenital heart defect, affecting at least 2% of the population. The embryonic origins of BAV remain poorly understood, with few assays for validating patient variants, limiting the identification of causative genes for BAV. In both human and mouse, the left and right leaflets of the arterial valves arise from the outflow tract cushions, with interstitial cells originating from neural crest cells and the overlying endocardium through endothelial-to-mesenchymal transition (EndoMT). In contrast, an EndoMT-independent mechanism of direct differentiation of cardiac progenitors from the second heart field (SHF) is responsible for the formation of the anterior and posterior leaflets. Defects in either of these developmental mechanisms can result in BAV. Although zebrafish have been suggested as a model for human variant testing, their naturally bicuspid arterial valve has not been considered suitable for understanding human arterial valve development. Here, we have set out to investigate to what extent the processes involved in arterial valve development are conserved in zebrafish and, ultimately, whether functional testing of BAV variants could be carried out. METHODS AND RESULTS Using a combination of live imaging, immunohistochemistry, and Cre-mediated lineage tracing, we show that the zebrafish arterial valve primordia develop directly from SHF progenitors with no contribution from EndoMT or neural crest, in keeping with the human and mouse anterior and posterior leaflets. Moreover, once formed, these primordia share common subsequent developmental events with all three aortic valve leaflets. CONCLUSION Our work highlights a conserved ancestral mechanism of arterial valve leaflet formation from the SHF and identifies that development of the arterial valve is distinct from that of the atrioventricular valve in zebrafish. Crucially, this confirms the utility of zebrafish for understanding the development of specific BAV subtypes and arterial valve dysplasia, offering potential for high-throughput variant testing.
Collapse
Affiliation(s)
- Christopher J Derrick
- International Centre for Life, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Lorraine Eley
- International Centre for Life, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Ahlam Alqahtani
- International Centre for Life, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Deborah J Henderson
- International Centre for Life, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Bill Chaudhry
- International Centre for Life, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| |
Collapse
|
4
|
Chalitsios CV, Pagkalidou E, Papagiannopoulos CK, Markozannes G, Bouras E, Watts EL, Richmond RC, Tsilidis KK. The role of sleep traits in prostate, endometrial, and epithelial ovarian cancers: An observational and Mendelian randomisation study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.10.25325598. [PMID: 40297455 PMCID: PMC12036389 DOI: 10.1101/2025.04.10.25325598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background Sleep traits may influence cancer risk; however, their associations with prostate (PCa), endometrial (ECa), and epithelial ovarian (EOCa) cancer remain unclear. Methods We conducted an observational analysis using the UK Biobank cohort and a two-sample Mendelian randomisation (MR) analysis to investigate the association of six sleep traits-duration, chronotype, insomnia, daytime napping, daytime sleepiness, and snoring-with PCa, ECa, and EOCa risk. Cox proportional hazards models were used for the observational analysis, while the inverse variance-weighted (IVW) method was applied in MR, with multiple sensitivity analyses. A Bonferroni correction accounted for multiple testing. Results Among 8,608 PCa, 1,079 ECa, and 680 EOCa incident diagnoses (median follow-up: 6.9 years), snoring was associated with reduced EOCa risk (HR=0.78, 95%CI: 0.62-0.98), while daytime sleepiness was associated with increased EOCa risk (HR=1.23, 95%CI: 1.03-1.47). However, these associations were not confirmed in MR. MR suggested higher odds of PCa (OR IVW =1.05, 95%CI: 1.01-1.11) and aggressive PCa (OR IVW =1.10, 95%CI: 1.02-1.19) for evening compared to morning chronotype. None of the findings survived multiple testing correction. Conclusion Sleep traits were not associated with PCa, ECa, or EOCa risk, but evening chronotype may increase PCa risk. Further research is needed to verify this association and investigate potential underlying mechanisms. Impact The proposed results have potential utility in reproductive cancer prevention. What is already known on this topic Sleep traits have been implicated in cancer risk, but their associations with prostate, endometrial, and epithelial ovarian cancer remain unclear. What this study adds This study found suggestive evidence that an evening chronotype may be associated with an increased risk of overall and aggressive prostate cancer. How this study might affect research practice or policy Further research is needed to confirm the potential association between chronotype and prostate cancer risk, which could inform personalised cancer prevention strategies.
Collapse
|
5
|
Eggan P, Gordon SE, Zagotta WN. Domain Coupling in Allosteric Regulation of SthK Measured Using Time-Resolved Transition Metal Ion FRET. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646362. [PMID: 40236086 PMCID: PMC11996379 DOI: 10.1101/2025.03.31.646362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cyclic nucleotide-binding domain (CNBD) ion channels are vital for cellular signaling and excitability, with activation regulated by cyclic adenosine- or guanosine-monophosphate (cAMP, cGMP) binding. However, the allosteric mechanisms underlying this activation, particularly the energetics that describe conformational changes within individual domains and between domains, remain unclear. The prokaryotic CNBD channel SthK has been a useful model for better understanding these allosteric mechanisms. Here, we applied time-resolved transition metal ion Förster resonance energy transfer (tmFRET) to investigate the conformational dynamics and energetics in the CNBD of SthK in both a soluble C-terminal fragment of the protein, SthK Cterm , and in the full-length channel, SthK Full . We incorporated the noncanonical amino acid Acd as a FRET donor and a metal bound to a chelator conjugated to a cysteine as an acceptor. We used time correlated single photon counting (TCSPC) to measure time-resolved FRET and fit the TCSPC data to obtain donor-acceptor distance distributions in the absence and presence of cAMP. The distance distributions allowed us to quantify the energetics of coupling between the C-terminal domains and the transmembrane domains by comparing the donor-acceptor distance distributions for SthK Cterm and SthK Full . Our data indicate that the presence of the SthK transmembrane domains makes the activating conformational change in the CNBD more favorable. These findings highlight the power of time-resolved tmFRET to uncover the structural and energetic landscapes of allosteric proteins and of the ligand-mediated mechanism in CNBD channels specifically.
Collapse
|
6
|
Li S, Park J, Phan TM, Egelman EH, Bird JE, Shin JB. Tonotopic Specialization of MYO7A Isoforms in Auditory Hair Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646665. [PMID: 40236041 PMCID: PMC11996455 DOI: 10.1101/2025.04.01.646665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
1. Mutations in Myo7a cause Usher syndrome type 1B and non-syndromic deafness, but the precise function of MYO7A in sensory hair cells remains unclear. We identify and characterize a novel isoform, MYO7A-N, expressed in auditory hair cells alongside the canonical MYO7A-C. Isoform-specific knock-in mice reveal that inner hair cells primarily express MYO7A-C, while outer hair cells express both isoforms in opposing tonotopic gradients. Both localize to the upper tip-link insertion site, consistent with a role in the tip link for mechanotransduction. Loss of MYO7A-N leads to outer hair cell degeneration and progressive hearing loss. Cryo-EM structures reveal isoform-specific differences at actomyosin interfaces, correlating with distinct ATPase activities. These findings reveal an unexpected layer of molecular diversity within the mechanotransduction machinery. We propose that MYO7A isoform specialization enables fine-tuning of tip-link tension, thus hearing sensitivity, and contributes to the frequency-resolving power of the cochlea.
Collapse
|
7
|
Himmelberg MM, Kwak Y, Carrasco M, Winawer J. Preferred spatial frequency covaries with cortical magnification in human primary visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644195. [PMID: 40166269 PMCID: PMC11957105 DOI: 10.1101/2025.03.19.644195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Primary visual cortex (V1) has played a key role in understanding the organization of cerebral cortex. Both structural and functional properties vary sharply throughout the human V1 map. Despite large variation, underlying constancies computed from the covariation pattern of V1 properties have been proposed. Such constancies would imply that V1 is composed of multiple identical units whose visual properties differ only due to differences in their inputs. To test this, we used fMRI to investigate how V1 cortical magnification and preferred spatial frequency covary across eccentricity and polar angle, and across individual observers (n=40). The two properties correlated across individuals, such that those with higher overall cortical magnification (i.e., larger V1 maps) had higher preferred spatial frequency (integrated across the map). Although correlated, the two properties were not proportional, and hence their ratio (mm of cortex per stimulus cycle) was not constant. Cortical magnification and preferred spatial frequency were strongly correlated across eccentricity and across polar angle, however their relation differed between these dimensions: they were proportional across eccentricity but not polar angle. The constant ratio of cortical magnification to preferred spatial frequency across eccentricity suggests a shared underlying cause of variation in the two properties, e.g., the gradient of retinal ganglion cell density across eccentricity. In contrast, the deviation from proportionality around polar angle implies that cortical variation differs from that in retina along this dimension. Thus, a constancy hypothesis is supported for one of the two spatial dimensions of V1, highlighting the importance of examining the full 2D-map, in multiple individuals, to understand how V1 is organized.
Collapse
Affiliation(s)
- Marc M. Himmelberg
- Department of Psychology, New York University, New York, NY, USA, 10003
- Center for Neural Science, New York University, New York, NY, USA, 10003
| | - Yuna Kwak
- Department of Psychology, New York University, New York, NY, USA, 10003
| | - Marisa Carrasco
- Department of Psychology, New York University, New York, NY, USA, 10003
- Center for Neural Science, New York University, New York, NY, USA, 10003
| | - Jonathan Winawer
- Department of Psychology, New York University, New York, NY, USA, 10003
- Center for Neural Science, New York University, New York, NY, USA, 10003
| |
Collapse
|
8
|
Xie W, Wardle SG, Langbein J, Fruchet O, Baumhauer M, Phan A, Tong AP, Japee S, Inati SK, Baker CI, Zaghloul KA. The role of the parahippocampal cortex in memory consolidation for scenes. Learn Mem 2025; 32:a054053. [PMID: 40316419 PMCID: PMC12052091 DOI: 10.1101/lm.054053.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 03/06/2025] [Indexed: 05/04/2025]
Abstract
Classic models propose that forming lasting visual memories involves coordinated interactions between visually selective neocortical structures and the hippocampus during memory consolidation. However, the precise role of visually selective neocortical structures in memory consolidation remains elusive, given their potential contributions spanning from initial perceptual encoding to subsequent memory reactivation. We capitalized on a unique opportunity, involving direct recording from the posterior parahippocampus and its subsequent resection in a neurological patient, to investigate the impact of scene-selective neocortical lesions on visual memory consolidation. First, with intracranial EEG, we confirmed the functional relevance of the patient's resected tissues in representing a specific visual category, in this case, scene images. Subsequently, we identified disruption of memory for scenes relative to faces and objects during the participant's postoperative visit. This finding prompted a comprehensive analysis of visual memory across different visual categories in this participant, as well as an examination of similar functions in other neurological patients with intact parahippocampi and a cohort of online participants. Through these within- and between-participant comparisons, we identified greater time-dependent reduction in visual memory for scene images following the resection of the posterior parahippocampus. Importantly, these changes in memory retention could not be attributed to a general reduction in initial memory encoding following neocortical lesions. Our findings, therefore, suggest that reactivating scene-selective neocortical areas is essential for converting transient visual perceptual experiences into lasting long-term scene memories.
Collapse
Affiliation(s)
- Weizhen Xie
- Department of Psychology, University of Maryland, College Park, Maryland 20742, USA
- Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA
- Department of Neurosurgery, University of Maryland, Baltimore, Maryland 21201, USA
| | - Susan G Wardle
- Laboratory of Brain and Cognition, NIMH, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jenna Langbein
- Department of Neurosurgery, University of Maryland, Baltimore, Maryland 21201, USA
| | - Oceane Fruchet
- Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Molly Baumhauer
- Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Audrey Phan
- Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ai Phuong Tong
- Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Shruti Japee
- Laboratory of Brain and Cognition, NIMH, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Sara K Inati
- Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Chris I Baker
- Laboratory of Brain and Cognition, NIMH, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kareem A Zaghloul
- Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
9
|
Jain N, Ogbonna EC, Maliga Z, Jacobson C, Zhang L, Shih A, Rosenberg J, Kalam H, Gagné A, Solomon IH, Santagata S, Sorger PK, Aldridge BB, Martinot AJ. Multiomic analysis identifies suppressive myeloid cell populations in human TB granulomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642376. [PMID: 40161687 PMCID: PMC11952478 DOI: 10.1101/2025.03.10.642376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Tuberculosis (TB) remains a major global health challenge, particularly in the context of multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb). Host-directed therapies (HDTs) have been proposed as adjunctive therapy to enhance immune control of infection. Recently, one such HDT, pharmacologic modulation of myeloid-derived suppressor cells (MDSCs), has been proposed to treat MDR-TB. While MDSCs have been well characterized in cancer, their role in TB pathogenesis remains unclear. To investigate whether MDSCs or other myeloid suppressor populations contribute to TB granuloma microenvironments (GME), we performed spatial transcriptional profiling and single-cell immunophenotyping on eighty-four granulomas in lung specimens from three individuals with active disease. Granulomas were histologically classified based on H&E staining, and transcriptional signatures were compared across regions of interest (ROIs) at different states of granuloma maturation. Our analysis revealed that immune suppression within granuloma was not primarily driven by classical MDSCs but rather by multiple myeloid cell subsets, including dendritic cells expressing indoleamine 2,3 dioxygenase-1 expressing (IDO1+ DCs). IDO1+ DCs were the most frequently observed suppressive myeloid cells, particularly in cellular regions, and their spatial proximity to activated T cells suggested localized immunosuppression. Importantly, granulomas at different stages contained distinct proportions of suppressor myeloid cells, with necrotic and cellular regions showing different myeloid phenotypes that may influence granuloma progression. Gene set enrichment analysis (GSEA) further indicated that elevated IDO1 expression was associated with a complex immune response that balanced suppressive signaling, immune activation, and cellular metabolism. These findings suggest that classical MDSCs, as defined in tumor microenvironments, likely play a minor role in TB, whereas IDO1+ DCs may be key regulators of immune suppression in granulomas influencing local Mtb control in infected lung. A deeper understanding of the role of IDO1+ suppressive myeloid cells in TB granulomas is essential to assessing their potential as therapeutic targets in TB treatment.
Collapse
|
10
|
Lynch KM, Custer RM, Sepehrband F, Toga AW, Choupan J. Perivascular and parenchymal fluid characteristics are related to age and cognitive performance across the lifespan. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.14.616343. [PMID: 40060450 PMCID: PMC11888170 DOI: 10.1101/2024.10.14.616343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Perivascular spaces (PVS) play a critical role in fluid transfer and waste clearance in the brain, but few studies have explored how alterations to perivascular fluid flow may impact brain maturation and behavior across the lifespan. This study aims to characterize age-related alterations to perivascular and parenchymal fluid flow characteristics across the lifespan in typically developing children (8-21 years) and aging adults (35-90 years) and assess their contribution to cognition. In this study, we employ multi-compartment diffusion models, neurite orientation dispersion and density imaging (NODDI) and tissue tensor imaging (TTI), to quantify free water diffusion characteristics within automatically defined perivascular spaces, the surrounding parenchyma, and at variable distances from the PVS. Our findings show free water diffusion characteristics within the PVS and surrounding parenchyma are associated with age in both developing children and in aging adults. Additionally, age was associated with accelerated change in free water diffusion measures with distance from the PVS. There was no direct effect of free water diffusion measures on cognitive scores across subjects; however, a more complex relationship emerged such that age modified the relationship between free water diffusion measures and cognition. Together, these findings provide evidence of age-associated alterations to fluid flow dynamics and cognition that may be related to the waste clearance system.
Collapse
Affiliation(s)
- Kirsten M Lynch
- Laboratory of Neuro Imaging (LONI), USC Mark and Mary Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Los Angeles, CA, 90033
| | - Rachel M Custer
- Laboratory of Neuro Imaging (LONI), USC Mark and Mary Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Los Angeles, CA, 90033
| | - Farshid Sepehrband
- Laboratory of Neuro Imaging (LONI), USC Mark and Mary Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Los Angeles, CA, 90033
- NeuroScope Inc., New York, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging (LONI), USC Mark and Mary Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Los Angeles, CA, 90033
| | - Jeiran Choupan
- Laboratory of Neuro Imaging (LONI), USC Mark and Mary Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Los Angeles, CA, 90033
- NeuroScope Inc., New York, USA
| |
Collapse
|
11
|
Hogrebe NJ, Schmidt MD, Augsornworawat P, Gale SE, Shunkarova M, Millman JR. Depolymerizing F-actin accelerates the exit from pluripotency to enhance stem cell-derived islet differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.21.618465. [PMID: 39484596 PMCID: PMC11526947 DOI: 10.1101/2024.10.21.618465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
In this study, we demonstrate that cytoskeletal state at the onset of directed differentiation is critical for the specification of human pluripotent stem cells (hPSCs) to all three germ layers. In particular, a polymerized actin cytoskeleton facilitates directed ectoderm differentiation, while depolymerizing F-actin promotes mesendoderm lineages. Applying this concept to a stem cell-derived islet (SC-islet) differentiation protocol, we show that depolymerizing F-actin with latrunculin A (latA) during the first 24 hours of definitive endoderm formation facilitates rapid exit from pluripotency and alters Activin/Nodal, BMP, JNK-JUN, and WNT pathway signaling dynamics. These signaling changes influence downstream patterning of the gut tube, leading to improved pancreatic progenitor identity and decreased expression of markers associated with other endodermal lineages. Continued differentiation generates islets containing a higher percentage of β cells that exhibit improved maturation, insulin secretion, and ability to reverse hyperglycemia. Furthermore, this latA treatment reduces enterochromaffin cells in the final cell population and corrects differentiations from hPSC lines that otherwise fail to consistently produce pancreatic islets, highlighting the importance of cytoskeletal signaling at the onset of directed differentiation.
Collapse
Affiliation(s)
- Nathaniel J. Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Mason D. Schmidt
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Punn Augsornworawat
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sarah E. Gale
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Mira Shunkarova
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Jeffrey R. Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, USA
| |
Collapse
|
12
|
Crawford KJ, Humphrey KS, Cortes E, Wang J, Feigin ME, Witkiewicz AK, Knudsen ES, Abel EV. Targeting FGFR4 Abrogates HNF1A-driven Metastasis in Pancreatic Ductal Adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636643. [PMID: 39974881 PMCID: PMC11839031 DOI: 10.1101/2025.02.06.636643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Purpose We previously identified an oncogenic role for the transcription factor HNF1A in pancreatic ductal adenocarcinoma (PDAC). However, the role of HNF1A in the metastatic progression of PDAC remains unknown and targeting modalities for HNF1A -dependent phenotypes have yet to be identified. Experimental Design Transwell chambers were used to assess the effects of HNF1A and FGFR4 modulation on the migration and invasion of ATCC and patient-derived PDAC cells in vitro . An intrasplenic injection xenograft model was used to evaluate the impact of HNF1A knockdown and overexpression on metastatic tumor burden. Single-cell RNA sequencing, tissue microarray (TMA) data, and UMAP spatial profiling were used to identify FGFR4 as an HNF1A target gene upregulated in metastatic cells. RNAi and two FGFR4 inhibiting modalities (H3B-6527 and U3- 1784) were utilized to demonstrate the efficacy of FGFR4 inhibiting agents at reducing HNF1A- driven metastasis. Results Knockdown of HNF1A significantly decreases and HNF1A overexpression significantly increases PDAC cell migration and invasion. In vivo studies show that HNF1A knockdown significantly abrogates metastasis, while overexpression significantly promotes metastasis. Single-cell RNAseq shows that FGFR4 is upregulated in metastatic PDAC cells and staining for HNF1A and FGFR4 in a PDAC TMA reveals significant correlation between HNF1A and FGFR4 in PDAC patients. Further, knockdown and inhibition of FGFR4 significantly decreases HNF1A- mediated cell migration and invasion, and blocks HNF1A-driven metastasis in vivo . Conclusions These findings demonstrate that HNF1A drives PDAC metastasis via upregulation of FGFR4, and FGFR4 inhibition is a potential mechanism to target metastasis in PDAC patients. Translational Relevance Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, made even more devastating when metastases overwhelm major organs. The vast majority of PDAC patients either present with metastases or will relapse with recurrent metastatic PDAC after primary tumor resection. Unfortunately, toxic and largely ineffective chemotherapies are currently the only approved treatment options for these patients and therefore there exists a critical and unmet clinical need for targeted therapies against pro-metastatic pathways in PDAC. In the current study, we identify HNF1A as an oncogenic transcription factor that drives metastasis in PDAC, and it does so through upregulation of the receptor tyrosine kinase FGFR4. Importantly, FGFR4 is a targetable vulnerability and treatment with an FGFR4 blocking antibody reduces HNF1A-driven metastasis. These findings suggest that FGFR4 inhibitors could be an efficacious treatment for PDAC patients for the prevention or delay of metastatic tumor development.
Collapse
|
13
|
Burger WAC, Mobbs JI, Rana B, Wang J, Joshi K, Gentry PR, Yeasmin M, Venugopal H, Bender AM, Lindsley CW, Miao Y, Christopoulos A, Valant C, Thal DM. Cryo-EM reveals a new allosteric binding site at the M 5 mAChR. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636602. [PMID: 39975287 PMCID: PMC11838558 DOI: 10.1101/2025.02.05.636602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The M5 muscarinic acetylcholine receptor (M5 mAChR) represents a promising therapeutic target for neurological disorders. However, the high conservation of its orthosteric binding site has posed significant challenges for drug development. While selective positive allosteric modulators (PAMs) offer a potential solution, a structural understanding of the M5 mAChR and its allosteric binding sites has remained limited. Here, we present a 2.8 Å cryo-electron microscopy structure of the M5 mAChR complexed with heterotrimeric Gq protein and the agonist iperoxo, completing the active-state structural characterization of the mAChR family. To identify the binding site of M5-selective PAMs, we implemented an integrated approach combining mutagenesis, pharmacological assays, structural biology, and molecular dynamics simulations. Our mutagenesis studies revealed that selective M5 PAMs bind outside previously characterized M5 mAChR allosteric sites. Subsequently, we obtained a 2.1 Å structure of M5 mAChR co-bound with acetylcholine and the selective PAM VU6007678, revealing a novel allosteric pocket at the extrahelical interface between transmembrane domains 3 and 4 that was confirmed through mutagenesis and simulations. These findings demonstrate the diverse mechanisms of allosteric regulation in mAChRs and highlight the value of integrating pharmacological and structural approaches to identify novel allosteric binding sites.
Collapse
Affiliation(s)
- Wessel A. C. Burger
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- These authors contributed equally: Wessel A. C. Burger, Jesse I Mobbs, Bhavika Rana, Jinan Wang
| | - Jesse I. Mobbs
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- These authors contributed equally: Wessel A. C. Burger, Jesse I Mobbs, Bhavika Rana, Jinan Wang
| | - Bhavika Rana
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- These authors contributed equally: Wessel A. C. Burger, Jesse I Mobbs, Bhavika Rana, Jinan Wang
| | - Jinan Wang
- Computational Medicine Program and Department of Pharmacology, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina 27599, United States
- These authors contributed equally: Wessel A. C. Burger, Jesse I Mobbs, Bhavika Rana, Jinan Wang
| | - Keya Joshi
- Computational Medicine Program and Department of Pharmacology, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Patrick R. Gentry
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Mahmuda Yeasmin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Hariprasad Venugopal
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, Victoria, Australia
| | - Aaron M. Bender
- Department of Pharmacology, Warren Center for Neuroscience Drug Discovery and Department of Chemistry, Vanderbilt University, Nashville, United States
| | - Craig W. Lindsley
- Department of Pharmacology, Warren Center for Neuroscience Drug Discovery and Department of Chemistry, Vanderbilt University, Nashville, United States
| | - Yinglong Miao
- Computational Medicine Program and Department of Pharmacology, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Australia
| | - Celine Valant
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - David M. Thal
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Sutton PJ, Mosqueda N, Brownlee CW. Palmitoylated Importin α Regulates Mitotic Spindle Orientation Through Interaction with NuMA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.25.620315. [PMID: 39484393 PMCID: PMC11527331 DOI: 10.1101/2024.10.25.620315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Regulation of cell division orientation is a fundamental process critical to differentiation and tissue homeostasis. Microtubules emanating from the mitotic spindle pole bind a conserved complex of proteins at the cell cortex which orients the spindle and ultimately the cell division plane. Control of spindle orientation is of particular importance in developing tissues, such as the developing brain. Misorientation of the mitotic spindle and thus subsequent division plane misalignment can contribute to improper segregation of cell fate determinants in developing neuroblasts, leading to a rare neurological disorder known as microcephaly. We demonstrate that the nuclear transport protein importin α, when palmitoylated, plays a critical role in mitotic spindle orientation through localizing factors, such as NuMA, to the cell cortex. We also observe craniofacial developmental defects in Xenopus laevis when importin α palmitoylation is abrogated, including smaller head and brains, a hallmark of spindle misorientation and microcephaly. These findings characterize not only a role for importin α in spindle orientation, but also a broader role for importin α palmitoylation which has significance for many cellular processes.
Collapse
|
15
|
van der Molen T, Spaeth A, Chini M, Hernandez S, Kaurala GA, Schweiger HE, Duncan C, McKenna S, Geng J, Lim M, Bartram J, Dendukuri A, Zhang Z, Gonzalez-Ferrer J, Bhaskaran-Nair K, Blauvelt LJ, Harder CR, Petzold LR, Alam El Din DM, Laird J, Schenke M, Smirnova L, Colquitt BM, Mostajo-Radji MA, Hansma PK, Teodorescu M, Hierlemann A, Hengen KB, Hanganu-Opatz IL, Kosik KS, Sharf T. Protosequences in brain organoids model intrinsic brain states Authors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.29.573646. [PMID: 38234832 PMCID: PMC10793448 DOI: 10.1101/2023.12.29.573646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Neuronal firing sequences are thought to be the basic building blocks of neural coding and information broadcasting within the brain. However, when sequences emerge during neurodevelopment remains unknown. We demonstrate that structured firing sequences are present in spontaneous activity of human and murine brain organoids and ex vivo neonatal brain slices from the murine somatosensory cortex. We observed a balance between temporally rigid and flexible firing patterns that are emergent phenomena in human and murine brain organoids and early postnatal murine somatosensory cortex, but not in primary dissociated cortical cultures. Our findings suggest that temporal sequences do not arise in an experience-dependent manner, but are rather constrained by an innate preconfigured architecture established during neurogenesis. These findings highlight the potential for brain organoids to further explore how exogenous inputs can be used to refine neuronal circuits and enable new studies into the genetic mechanisms that govern assembly of functional circuitry during early human brain development.
Collapse
Affiliation(s)
- Tjitse van der Molen
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Alex Spaeth
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Mattia Chini
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sebastian Hernandez
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Gregory A. Kaurala
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Hunter E. Schweiger
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Cole Duncan
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sawyer McKenna
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jinghui Geng
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Max Lim
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Julian Bartram
- Department of Biosystems Science and Engineering, ETH Zürich, Klingelbergstrasse 48, 4056 Basel, Switzerland
| | - Aditya Dendukuri
- Department of Computer Science, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Zongren Zhang
- Department of Physics, University of California Santa Barbara, Santa Barbara, CA 93106
| | - Jesus Gonzalez-Ferrer
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Kiran Bhaskaran-Nair
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Lon J. Blauvelt
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
| | - Cole R.K. Harder
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Linda R. Petzold
- Department of Computer Science, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Dowlette-Mary Alam El Din
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jason Laird
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Maren Schenke
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bradley M. Colquitt
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
- Institute for the Biology of Stem Cells, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | | | - Paul K. Hansma
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Physics, University of California Santa Barbara, Santa Barbara, CA 93106
| | - Mircea Teodorescu
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Andreas Hierlemann
- Department of Biosystems Science and Engineering, ETH Zürich, Klingelbergstrasse 48, 4056 Basel, Switzerland
| | - Keith B. Hengen
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Ileana L. Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Kenneth S. Kosik
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Tal Sharf
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Institute for the Biology of Stem Cells, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
16
|
Bahuguna J, Verstynen T, Rubin JE. How cortico-basal ganglia-thalamic subnetworks can shift decision policies to maximize reward rate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.21.595174. [PMID: 38826315 PMCID: PMC11142098 DOI: 10.1101/2024.05.21.595174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
All mammals exhibit flexible decision policies that depend, at least in part, on the cortico-basal ganglia-thalamic (CBGT) pathways. Yet understanding how the complex connectivity, dynamics, and plasticity of CBGT circuits translate into experience-dependent shifts of decision policies represents a longstanding challenge in neuroscience. Here we present the results of a computational approach to address this problem. Specifically, we simulated decisions driven by CBGT circuits under baseline, unrewarded conditions using a spiking neural network, and fit an evidence accumulation model to the resulting behavior. Using canonical correlation analysis, we then replicated the identification of three control ensembles (responsiveness, pliancy and choice) within CBGT circuits, with each of these subnetworks mapping to a specific configuration of the evidence accumulation process. We subsequently simulated learning in a simple two-choice task with one optimal (i.e., rewarded) target and found that feedback-driven dopaminergic plasticity on cortico-striatal synapses effectively manages the speed-accuracy tradeoff so as to increase reward rate over time. The learning-related changes in the decision policy can be decomposed in terms of the contributions of each control ensemble, whose influence is driven by sequential reward prediction errors on individual trials. Our results provide a clear and simple mechanism for how dopaminergic plasticity shifts subnetworks within CBGT circuits so as to maximize reward rate by strategically modulating how evidence is used to drive decisions.
Collapse
Affiliation(s)
- Jyotika Bahuguna
- Department of Psychology & Neuroscience Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Timothy Verstynen
- Department of Psychology & Neuroscience Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- Center for the Neural Basis of Cognition, Pittsburgh, Pennsylvania, United States of America
| | - Jonathan E Rubin
- Center for the Neural Basis of Cognition, Pittsburgh, Pennsylvania, United States of America
- Department of Mathematics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
17
|
Dutcher HA, Gasch AP. Investigating the role of RNA-binding protein Ssd1 in aneuploidy tolerance through network analysis. RNA (NEW YORK, N.Y.) 2024; 31:100-112. [PMID: 39471998 DOI: 10.1261/rna.080199.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/15/2024] [Indexed: 11/06/2024]
Abstract
RNA-binding proteins (RBPs) play critical cellular roles by mediating various stages of RNA life cycles. Ssd1, an RBP with pleiotropic effects, has been implicated in aneuploidy tolerance in Saccharomyces cerevisiae but its mechanistic role remains unclear. Here, we used a network-based approach to inform on Ssd1's role in aneuploidy tolerance, by identifying and experimentally perturbing a network of RBPs that share mRNA targets with Ssd1. We identified RBPs whose bound mRNA targets significantly overlap with Ssd1 targets. For 14 identified RBPs, we then used a genetic approach to generate all combinations of genotypes for euploid and aneuploid yeast with an extra copy of chromosome XII, with and without SSD1 and/or the RBP of interest. Deletion of 10 RBPs either exacerbated or alleviated the sensitivity of wild-type and/or ssd1Δ cells to chromosome XII duplication, in several cases indicating genetic interactions with SSD1 in the context of aneuploidy. We integrated these findings with results from a global overexpression screen that identified genes whose duplication complements ssd1Δ aneuploid sensitivity. The resulting network points to a subgroup of proteins with shared roles in translational repression and P-body formation, implicating these functions in aneuploidy tolerance. Our results reveal a role for new RBPs in aneuploidy tolerance and support a model in which Ssd1 mitigates translation-related stresses in aneuploid cells.
Collapse
Affiliation(s)
- H Auguste Dutcher
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Audrey P Gasch
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
18
|
Liu N, Kawamura R, Qiang W, Balboula A, Marko JF, Qiao H. Isolation and manipulation of meiotic spindles from mouse oocytes reveals migration regulated by pulling force during asymmetric division. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627260. [PMID: 39677774 PMCID: PMC11643109 DOI: 10.1101/2024.12.06.627260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Spindles are essential for accurate chromosome segregation in all eukaryotic cells. This study presents a novel approach for isolating fresh mammalian spindles from mouse oocytes, establishing it as a valuable in vitro model system for a wide range of possible studies. Our method enables the investigation of the physical properties and migration force of meiotic spindles in oocytes. We found that the spindle length decreases upon isolation from the oocyte. Combining this observation with direct measurements of spindle mechanics, we examined the forces governing spindle migration during oocyte asymmetric division. Our findings suggest that the spindle migration is regulated by a pulling force and a net tensile force of approximately 680 pN is applied to the spindle in vivo during the migration process. This method, unveiling insights into spindle dynamics, holds promise as a robust model for future investigations into spindle formation and chromosome separation. We also found that the same approach could not isolate spindles from somatic cells, indicative of mammalian oocytes having a unique spindle organization amenable to isolation.
Collapse
Affiliation(s)
- Ning Liu
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Ryo Kawamura
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Wenan Qiang
- Center for Developmental Therapeutics, Northwestern University, Evanston, IL, USA
| | - Ahmed Balboula
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - John F Marko
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
- Department of Physics and Astronomy, Northwestern University, Evanston, IL, USA
| | - Huanyu Qiao
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
19
|
Cornell CE, Chorlay A, Krishnamurthy D, Martin NR, Baldauf L, Fletcher DA. Target cell tension regulates macrophage trogocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626490. [PMID: 39677802 PMCID: PMC11642796 DOI: 10.1101/2024.12.02.626490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Macrophages are known to engulf small membrane fragments, or trogocytose, target cells and pathogens, rather than fully phagocytose them. However, little is known about what causes macrophages to choose trogocytosis versus phagocytosis. Here, we report that cortical tension of target cells is a key regulator of macrophage trogocytosis. At low tension, macrophages will preferentially trogocytose antibody-opsonized cells, while at high tension they tend towards phagocytosis. Using model vesicles, we demonstrate that macrophages will rapidly switch from trogocytosis to phagocytosis when membrane tension is increased. Stiffening the cortex of target cells also biases macrophages to phagocytose them, a trend that can be countered by increasing antibody surface density and is captured in a mechanical model of trogocytosis. This work suggests that a distinct molecular pathway for trogocytosis is not required to explain differences in trogocytosis among target cell types and points to a mechanism for target cells to modulate trogocytosis.
Collapse
|
20
|
Kraus F, Ross B, Herrmann B, Obleser J. Neurophysiology of Effortful Listening: Decoupling Motivational Modulation from Task Demands. J Neurosci 2024; 44:e0589242024. [PMID: 39261007 PMCID: PMC11529814 DOI: 10.1523/jneurosci.0589-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/13/2024] Open
Abstract
In demanding listening situations, a listener's motivational state may affect their cognitive investment. Here, we aim to delineate how domain-specific sensory processing, domain-general neural alpha power, and pupil size as a proxy for cognitive investment encode influences of motivational state under demanding listening. Participants (male and female) performed an auditory gap-detection task while the pupil size and the magnetoencephalogram were simultaneously recorded. Task demand and a listener's motivational state were orthogonally manipulated through changes in gap duration and monetary-reward prospect, respectively. Whereas task difficulty impaired performance, reward prospect enhanced it. The pupil size reliably indicated the modulatory impact of an individual's motivational state. At the neural level, the motivational state did not affect auditory sensory processing directly but impacted attentional postprocessing of an auditory event as reflected in the late evoked-response field and alpha-power change. Both pregap pupil dilation and higher parietal alpha power predicted better performance at the single-trial level. The current data support a framework wherein the motivational state acts as an attentional top-down neural means of postprocessing the auditory input in challenging listening situations.
Collapse
Affiliation(s)
- Frauke Kraus
- Department of Psychology, University of Lübeck, 23562 Lübeck, Germany
- Center of Brain, Behavior, and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Bernhard Ross
- Rotman Research Institute, Baycrest Academy for Research and Education, Toronto, Ontario M6A 2E1, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Björn Herrmann
- Rotman Research Institute, Baycrest Academy for Research and Education, Toronto, Ontario M6A 2E1, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Jonas Obleser
- Department of Psychology, University of Lübeck, 23562 Lübeck, Germany
- Center of Brain, Behavior, and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
21
|
Nasim S, Abujamra BA, Chaparro D, Nogueira PDS, Riva A, Hutcheson JD, Kos L. Multiple cell types including melanocytes contribute to elastogenesis in the developing murine aortic valve. Sci Rep 2024; 14:25481. [PMID: 39461968 PMCID: PMC11513076 DOI: 10.1038/s41598-024-73673-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
Elastic fibers are crucial for aortic valve (AoV) function and are generated and maintained by valvular interstitial cells (VICs). VICs exhibit diverse phenotypes, yet the specific subpopulation responsible for producing and regulating elastic fibers remains unclear. This gap in knowledge is significant, given that elastin (Eln) abnormalities lead to congenital AoV defects and initiate AoV diseases. This study characterizes the timing of Eln expression in murine AoV, revealing it peaks during late embryogenesis and early postnatal stages, decreasing in adulthood. Spatial transcriptomics and RT-qPCR indicate that Eln expression correlates with genes associated to elastogenesis, including Acta2, a smooth muscle cell marker. While Eln expression is not exclusive to a single VIC subpopulation, RNAscope and immunofluorescence demonstrate a population of Eln-expressing VICs that co-express alpha smooth muscle actin and melanocytic markers. As previously reported in adult mice, we show a relationship between AoV pigment and elastic fiber patterning during early postnatal stages and further show that melanocytes may play a critical role in elastogenesis. In summary, Eln is expressed in the AoV during early postnatal stages by cells co-expressing markers of various types, highlighting the complexity of VICs phenotypes and their role in elastic fiber regulation.
Collapse
Affiliation(s)
- Sana Nasim
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA
| | - Beatriz Abdo Abujamra
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | - Daniel Chaparro
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA
| | - Perony Da Silva Nogueira
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | | | - Joshua D Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA.
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA.
| | - Lidia Kos
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA.
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
22
|
Qiu W, Dincer AB, Janizek JD, Celik S, Pittet M, Naxerova K, Lee SI. A deep profile of gene expression across 18 human cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.17.585426. [PMID: 38559197 PMCID: PMC10980029 DOI: 10.1101/2024.03.17.585426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Clinically and biologically valuable information may reside untapped in large cancer gene expression data sets. Deep unsupervised learning has the potential to extract this information with unprecedented efficacy but has thus far been hampered by a lack of biological interpretability and robustness. Here, we present DeepProfile, a comprehensive framework that addresses current challenges in applying unsupervised deep learning to gene expression profiles. We use DeepProfile to learn low-dimensional latent spaces for 18 human cancers from 50,211 transcriptomes. DeepProfile outperforms existing dimensionality reduction methods with respect to biological interpretability. Using DeepProfile interpretability methods, we show that genes that are universally important in defining the latent spaces across all cancer types control immune cell activation, while cancer type-specific genes and pathways define molecular disease subtypes. By linking DeepProfile latent variables to secondary tumor characteristics, we discover that tumor mutation burden is closely associated with the expression of cell cycle-related genes. DNA mismatch repair and MHC class II antigen presentation pathway expression, on the other hand, are consistently associated with patient survival. We validate these results through Kaplan-Meier analyses and nominate tumor-associated macrophages as an important source of survival-correlated MHC class II transcripts. Our results illustrate the power of unsupervised deep learning for discovery of cancer biology from existing gene expression data.
Collapse
Affiliation(s)
- Wei Qiu
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA
| | - Ayse B. Dincer
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA
| | - Joseph D. Janizek
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA
- Medical Scientist Training Program, University of Washington, Seattle, WA
| | | | - Mikael Pittet
- Department of Pathology and Immunology, University of Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Switzerland
| | - Kamila Naxerova
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Su-In Lee
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA
| |
Collapse
|
23
|
Lynch KM, Bodison SC, Cabeen RP, Toga AW, Voelker CC. The spatial organization of ascending auditory pathway microstructural maturation from infancy through adolescence using a novel fiber tracking approach. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.597798. [PMID: 38915661 PMCID: PMC11195149 DOI: 10.1101/2024.06.10.597798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Auditory perception is established through experience-dependent stimuli exposure during sensitive developmental periods; however, little is known regarding the structural development of the central auditory pathway in humans. The present study characterized the regional developmental trajectories of the ascending auditory pathway from the brainstem to the auditory cortex from infancy through adolescence using a novel diffusion MRI-based tractography approach and along-tract analyses. We used diffusion tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI) to quantify the magnitude and timing of auditory pathway microstructural maturation. We found spatially varying patterns of white matter maturation along the length of the tract, with inferior brainstem regions developing earlier than thalamocortical projections and left hemisphere tracts developing earlier than the right. These results help to characterize the processes that give rise to functional auditory processing and may provide a baseline for detecting abnormal development.
Collapse
Affiliation(s)
- Kirsten M. Lynch
- Laboratory of Neuro Imaging (LONI), USC Mark and Mary Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Stefanie C. Bodison
- Department of Occupational Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
| | - Ryan P. Cabeen
- Laboratory of Neuro Imaging (LONI), USC Mark and Mary Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging (LONI), USC Mark and Mary Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Los Angeles, CA, USA
| | | |
Collapse
|
24
|
Nascimento F, Özyurt MG, Halablab K, Bhumbra GS, Caron G, Bączyk M, Zytnicki D, Manuel M, Roselli F, Brownstone R, Beato M. Spinal microcircuits go through multiphasic homeostatic compensations in a mouse model of motoneuron degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588918. [PMID: 38645210 PMCID: PMC11030447 DOI: 10.1101/2024.04.10.588918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
In many neurological conditions, early-stage neural circuit adaption can preserve relatively normal behaviour. In some diseases, spinal motoneurons progressively degenerate yet movement is initially preserved. We therefore investigated whether these neurons and associated microcircuits adapt in a mouse model of progressive motoneuron degeneration. Using a combination of in vitro and in vivo electrophysiology and super-resolution microscopy, we found that, early in the disease, neurotransmission in a key pre-motor circuit, the recurrent inhibition mediated by Renshaw cells, is reduced by half due to impaired quantal size associated with decreased glycine receptor density. This impairment is specific, and not a widespread feature of spinal inhibitory circuits. Furthermore, it recovers at later stages of disease. Additionally, an increased probability of release from proprioceptive afferents leads to increased monosynaptic excitation of motoneurons. We reveal that in motoneuron degenerative conditions, spinal microcircuits undergo specific multiphasic homeostatic compensations that may contribute to preservation of force output.
Collapse
Affiliation(s)
- Filipe Nascimento
- Department of Neuroscience Physiology and Pharmacology (NPP), Gower Street, University College London, WC1E 6BT, UK
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - M. Görkem Özyurt
- Department of Neuroscience Physiology and Pharmacology (NPP), Gower Street, University College London, WC1E 6BT, UK
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Kareen Halablab
- Department of Neurology, Ulm University, Ulm, Germany
- German Centre for Neurodegenerative Diseases-Ulm (DZNE-Ulm), Ulm, Germany
| | - Gardave Singh Bhumbra
- Department of Neuroscience Physiology and Pharmacology (NPP), Gower Street, University College London, WC1E 6BT, UK
| | - Guillaume Caron
- Saints-Pères Paris Institute for the Neurosciences (SPPIN), Université Paris Cité, Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Marcin Bączyk
- Department of Neurobiology, Poznań University of Physical Education, Poznań, Poland
| | - Daniel Zytnicki
- Saints-Pères Paris Institute for the Neurosciences (SPPIN), Université Paris Cité, Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Marin Manuel
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, USA
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany
- German Centre for Neurodegenerative Diseases-Ulm (DZNE-Ulm), Ulm, Germany
| | - Rob Brownstone
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Marco Beato
- Department of Neuroscience Physiology and Pharmacology (NPP), Gower Street, University College London, WC1E 6BT, UK
| |
Collapse
|
25
|
Eggan P, Gordon SE, Zagotta WN. Ligand-Coupled Conformational Changes in a Cyclic Nucleotide-Gated Ion Channel Revealed by Time-Resolved Transition Metal Ion FRET. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.591185. [PMID: 39411160 PMCID: PMC11475872 DOI: 10.1101/2024.04.25.591185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Cyclic nucleotide-binding domain (CNBD) ion channels play crucial roles in cellular-signaling and excitability and are regulated by the direct binding of cyclic adenosine- or guanosine-monophosphate (cAMP, cGMP). However, the precise allosteric mechanism governing channel activation upon ligand binding, particularly the energetic changes within domains, remains poorly understood. The prokaryotic CNBD channel SthK offers a valuable model for investigating this allosteric mechanism. In this study, we investigated the conformational dynamics and energetics of the SthK C-terminal region using a combination of steady-state and time-resolved transition metal ion Förster resonance energy transfer (tmFRET) experiments. We engineered donor-acceptor pairs at specific sites within a SthK C-terminal fragment by incorporating a fluorescent noncanonical amino acid donor and metal ion acceptors. Measuring tmFRET with fluorescence lifetimes, we determined intramolecular distance distributions in the absence and presence of cAMP or cGMP. The probability distributions between conformational states without and with ligand were used to calculate the changes in free energy ( ΔG ) and differences in free energy change ( ΔΔG ) in the context of a simple four-state model. Our findings reveal that cAMP binding produces large structural changes, with a very favorable ΔΔG . In contrast to cAMP, cGMP behaved as a partial agonist and only weakly promoted the active state. Furthermore, we assessed the impact of protein oligomerization and ionic strength on the structure and energetics of the conformational states. This study demonstrates the effectiveness of time-resolved tmFRET in determining the conformational states and the ligand-dependent energetics of the SthK C-terminal region.
Collapse
Affiliation(s)
- Pierce Eggan
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Sharona E. Gordon
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - William N. Zagotta
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| |
Collapse
|
26
|
Anderson RH, Kerwin J, Lamers WH, Hikspoors JPJM, Mohun TJ, Chaudhry B, Lisgo S, Henderson DJ. Cardiac development demystified by use of the HDBR atlas. J Anat 2024; 245:517-534. [PMID: 38783643 PMCID: PMC11424819 DOI: 10.1111/joa.14066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Much has been learned over the last half century regarding the molecular and genetic changes that take place during cardiac development. As yet, however, these advances have not been translated into knowledge regarding the marked changes that take place in the anatomical arrangements of the different cardiac components. As such, therefore, many aspects of cardiac development are still described on the basis of speculation rather than evidence. In this review, we show how controversial aspects of development can readily be arbitrated by the interested spectator by taking advantage of the material now gathered together in the Human Developmental Biology Resource; HDBR. We use the material to demonstrate the changes taking place during the formation of the ventricular loop, the expansion of the atrioventricular canal, the incorporation of the systemic venous sinus, the formation of the pulmonary vein, the process of atrial septation, the remodelling of the pharyngeal arches, the major changes occurring during formation of the outflow tract, the closure of the embryonic interventricular communication, and the formation of the ventricular walls. We suggest that access to the resource makes it possible for the interested observer to arbitrate, for themselves, the ongoing controversies that continue to plague the understanding of cardiac development.
Collapse
Affiliation(s)
| | - Janet Kerwin
- Human Developmental Biology Resource, Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Wouter H. Lamers
- Department of Anatomy and EmbryologyMaastricht UniversityMaastrichtThe Netherlands
| | | | | | - Bill Chaudhry
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Steven Lisgo
- Human Developmental Biology Resource, Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Deborah J. Henderson
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
- Human Developmental Biology Resource, Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
27
|
Feng H, Yang S, Zhang L, Zhu J, Li J, Yang Z. A new Prdm1-Cre line is suitable for studying the second heart field development. Dev Biol 2024; 514:78-86. [PMID: 38880275 DOI: 10.1016/j.ydbio.2024.06.007] [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: 01/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
The second heart field (SHF) plays a pivotal role in heart development, particularly in outflow tract (OFT) morphogenesis and septation, as well as in the expansion of the right ventricle (RV). Two mouse Cre lines, the Mef2c-AHF-Cre (Mef2c-Cre) and Isl1-Cre, have been widely used to study the SHF development. However, Cre activity is triggered not only in the SHF but also in the RV in the Mef2c-Cre mice, and in the Isl1-Cre mice, Cre activation is not SHF-specific. Therefore, a more suitable SHF-Cre line is desirable for better understanding SHF development. Here, we generated and characterized the Prdm1-Cre knock-in mice. In comparison with Mef2c-Cre mice, the Cre activity is similar in the pharyngeal and splanchnic mesoderm, and in the OFT of the Prdm1-Cre mice. Nonetheless, it was noticed that Cre expression is largely reduced in the RV of Prdm1-Cre mice compared to the Mef2c-Cre mice. Furthermore, we deleted Hand2, Nkx2-5, Pdk1 and Tbx20 using both Mef2c-Cre and Prdm1-Cre mice to study OFT morphogenesis and septation, making a comparison between these two Cre lines. New insights were obtained in understanding SHF development including differentiation into cardiomyocytes in the OFT using Prdm1-Cre mice. In conclusion, we found that Prdm1-Cre mouse line is a more appropriate tool to monitor SHF development, while the Mef2c-Cre mice are excellent in studying the role and function of the SHF in OFT morphogenesis and septation.
Collapse
Affiliation(s)
- Haiyue Feng
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Suming Yang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lijun Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Jingai Zhu
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
28
|
Whedon SD, Lee K, Wang ZA, Zahn E, Lu C, Yapa-Abeywardana M, Fairall L, Nam E, Dubois-Coyne S, Ioannes PD, Sheng X, Andrei A, Lundberg E, Jiang J, Armache KJ, Zhao Y, Schwabe JWR, Wu M, Garcia BA, Cole PA. A circular engineered sortase for interrogating histone H3 in chromatin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612318. [PMID: 39372790 PMCID: PMC11451751 DOI: 10.1101/2024.09.10.612318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Reversible modification of the histone H3 N-terminal tail is critical in regulating chromatin structure, gene expression, and cell states, while its dysregulation contributes to disease pathogenesis. Understanding the crosstalk between H3 tail modifications in nucleosomes constitutes a central challenge in epigenetics. Here we describe an engineered sortase transpeptidase, cW11, that displays highly favorable properties for introducing scarless H3 tails onto nucleosomes. This approach significantly accelerates the production of both symmetrically and asymmetrically modified nucleosomes. We demonstrate the utility of asymmetrically modified nucleosomes produced in this way in dissecting the impact of multiple modifications on eraser enzyme processing and molecular recognition by a reader protein. Moreover, we show that cW11 sortase is very effective at cutting and tagging histone H3 tails from endogenous histones, facilitating multiplex "cut-and-paste" middle down proteomics with tandem mass tags. This cut-and- paste proteomics approach permits the quantitative analysis of histone H3 modification crosstalk after treatment with different histone deacetylase inhibitors. We propose that these chemoenzymatic tail isolation and modification strategies made possible with cW11 sortase will broadly power epigenetics discovery and therapeutic development.
Collapse
Affiliation(s)
- Samuel D Whedon
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Kwangwoon Lee
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Zhipeng A Wang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Emily Zahn
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Congcong Lu
- Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Maheeshi Yapa-Abeywardana
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Louise Fairall
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Eunju Nam
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sarah Dubois-Coyne
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Pablo De Ioannes
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, United States
| | - Xinlei Sheng
- The Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, United States
| | - Adelina Andrei
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Emily Lundberg
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jennifer Jiang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Karim-Jean Armache
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, United States
| | - Yingming Zhao
- The Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, United States
| | - John W R Schwabe
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Mingxuan Wu
- Department of Chemistry, School of Science, Westlake University, Hangzhou 310030, China
| | - Benjamin A Garcia
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Philip A Cole
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
29
|
Stebbins K, Somaiya RD, Sabbagh U, Khaksar P, Liang Y, Su J, Fox MA. Retinal Input Is Required for the Maintenance of Neuronal Laminae in the Ventrolateral Geniculate Nucleus. eNeuro 2024; 11:ENEURO.0022-24.2024. [PMID: 39160068 PMCID: PMC11373735 DOI: 10.1523/eneuro.0022-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/24/2024] [Accepted: 07/02/2024] [Indexed: 08/21/2024] Open
Abstract
Retinal ganglion cell (RGC) axons provide direct input into several brain regions, including the dorsal lateral geniculate nucleus (dLGN), which is important for image-forming vision, and the ventrolateral geniculate nucleus (vLGN), which is associated with nonimage-forming vision. Through both activity- and morphogen-dependent mechanisms, retinal inputs play important roles in the development of dLGN, including the refinement of retinal projections, morphological development of thalamocortical relay cells (TRCs), timing of corticogeniculate innervation, and recruitment and distribution of inhibitory interneurons. In contrast, little is known about the role of retinal inputs in the development of vLGN. Grossly, vLGN is divided into two domains, the retinorecipient external vLGN (vLGNe) and nonretinorecipient internal vLGN (vLGNi). Studies previously found that vLGNe consists of transcriptionally distinct GABAergic subtypes distributed into at least four adjacent laminae. At present, it remains unclear whether retinal inputs influence the development of these cell-type-specific neuronal laminae in vLGNe. Here, we elucidated the developmental timeline for these laminae in the mouse vLGNe, and results indicate that these laminae are specified at or before birth. We observed that mutant mice without retinal inputs have a normal laminar distribution of GABAergic cells at birth; however, after the first week of postnatal development, these mutants exhibited a dramatic disruption in the laminar organization of inhibitory neurons and clear boundaries between vLGNe and vLGNi. Overall, our results show that while the formation of cell-type-specific layers in mouse vLGNe does not depend on RGC inputs, retinal signals are critical for their maintenance.
Collapse
Affiliation(s)
- Katelyn Stebbins
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia 24061
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia 24016
| | - Rachana Deven Somaiya
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia 24061
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Ubadah Sabbagh
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia 24061
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02138
| | - Parsa Khaksar
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia 24016
| | - Yanping Liang
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
| | - Jianmin Su
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia 24061
| | - Michael A Fox
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, Virginia 24061
- Department of Biology, College of Natural Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
30
|
LeBlang CJ, Pazyra-Murphy MF, Silagi ES, Dasgupta S, Tsolias M, Miller T, Petrova V, Zhen S, Jovanovic V, Castellano D, Gerrish K, Ormanoglu P, Tristan C, Singeç I, Woolf CJ, Tasdemir-Yilmaz O, Segal RA. Satellite glial contact enhances differentiation and maturation of human iPSC-derived sensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604966. [PMID: 39211268 PMCID: PMC11361066 DOI: 10.1101/2024.07.24.604966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sensory neurons generated from induced pluripotent stem cells (iSNs) are used to model human peripheral neuropathies, however current differentiation protocols produce sensory neurons with an embryonic phenotype. Peripheral glial cells contact sensory neurons early in development and contribute to formation of the canonical pseudounipolar morphology, but these signals are not encompassed in current iSN differentiation protocols. Here, we show that terminal differentiation of iSNs in co-culture with rodent Dorsal Root Ganglion satellite glia (rSG) advances their differentiation and maturation. Co-cultured iSNs develop a pseudounipolar morphology through contact with rSGs. This transition depends on semaphorin-plexin guidance cues and on glial gap junction signaling. In addition to morphological changes, iSNs terminally differentiated in co-culture exhibit enhanced spontaneous action potential firing, more mature gene expression, and increased susceptibility to paclitaxel induced axonal degeneration. Thus, iSNs differentiated in coculture with rSGs provide a better model for investigating human peripheral neuropathies.
Collapse
|
31
|
McLeod MJ, Barwell SAE, Holyoak T, Thorne RE. A structural perspective on the temperature-dependent activity of enzymes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609221. [PMID: 39229032 PMCID: PMC11370597 DOI: 10.1101/2024.08.23.609221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Enzymes are biomolecular catalysts whose activity varies with temperature. Unlike for small-molecule catalysts, the structural ensembles of enzymes can vary substantially with temperature, and it is in general unclear how this modulates the temperature dependence of activity. Here multi-temperature X-ray crystallography was used to record structural changes from -20°C to 40°C for a mesophilic enzyme in complex with inhibitors mimicking substrate-, intermediate-, and product-bound states, representative of major complexes underlying the kinetic constantk c a t . Both inhibitors, substrates and catalytically relevant loop motifs increasingly populate catalytically competent conformations as temperature increases. These changes occur even in temperature ranges where kinetic measurements show roughly linear Arrhenius/Eyring behavior where parameters characterizing the system are assumed to be temperature independent. Simple analysis shows that linear Arrhenius/Eyring behavior can still be observed when the underlying activation energy / enthalpy values vary with temperature, e.g., due to structural changes, and that the underlying thermodynamic parameters can be far from values derived from Arrhenius/Eyring model fits. Our results indicate a critical role for temperature-dependent atomic-resolution structural data in interpreting temperature-dependent kinetic data from enzymatic systems.
Collapse
Affiliation(s)
| | | | - Todd Holyoak
- University of Waterloo, Waterloo Ontario, Canada. Department of Biology
| | | |
Collapse
|
32
|
Rubio LS, Mohajan S, Gross DS. Heat Shock Factor 1 forms nuclear condensates and restructures the yeast genome before activating target genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.28.560064. [PMID: 37808805 PMCID: PMC10557744 DOI: 10.1101/2023.09.28.560064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
In insects and mammals, 3D genome topology has been linked to transcriptional states yet whether this link holds for other eukaryotes is unclear. Using both ligation proximity and fluorescence microscopy assays, we show that in Saccharomyces cerevisiae, Heat Shock Response (HSR) genes dispersed across multiple chromosomes and under the control of Heat Shock Factor (Hsf1) rapidly reposition in cells exposed to acute ethanol stress and engage in concerted, Hsf1-dependent intergenic interactions. Accompanying 3D genome reconfiguration is equally rapid formation of Hsf1-containing condensates. However, in contrast to the transience of Hsf1-driven intergenic interactions that peak within 10-20 min and dissipate within 1 h in the presence of 8.5% (v/v) ethanol, transcriptional condensates are stably maintained for hours. Moreover, under the same conditions, Pol II occupancy of HSR genes, chromatin remodeling, and RNA expression are detectable only later in the response and peak much later (>1 h). This contrasts with the coordinate response of HSR genes to thermal stress (39°C) where Pol II occupancy, transcription, histone eviction, intergenic interactions, and formation of Hsf1 condensates are all rapid yet transient (peak within 2.5-10 min and dissipate within 1 h). Therefore, Hsf1 forms condensates, restructures the genome and transcriptionally activates HSR genes in response to both forms of proteotoxic stress but does so with strikingly different kinetics. In cells subjected to ethanol stress, Hsf1 forms condensates and repositions target genes before transcriptionally activating them.
Collapse
Affiliation(s)
- Linda S. Rubio
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130
| | - Suman Mohajan
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130
| | - David S. Gross
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130
| |
Collapse
|
33
|
Ye Z, Wessel R, Franken TP. Brain-like border ownership signals support prediction of natural videos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.11.607040. [PMID: 39185218 PMCID: PMC11343161 DOI: 10.1101/2024.08.11.607040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
To make sense of visual scenes, the brain must segment foreground from background. This is thought to be facilitated by neurons in the primate visual system that encode border ownership (BOS), i.e. whether a local border is part of an object on one or the other side of the border. It is unclear how these signals emerge in neural networks without a teaching signal of what is foreground and background. In this study, we investigated whether BOS signals exist in PredNet, a self-supervised artificial neural network trained to predict the next image frame of natural video sequences. We found that a significant number of units in PredNet are selective for BOS. Moreover these units share several other properties with the BOS neurons in the brain, including robustness to scene variations that constitute common object transformations in natural videos, and hysteresis of BOS signals. Finally, we performed ablation experiments and found that BOS units contribute more to prediction than non-BOS units for videos with moving objects. Our findings indicate that BOS units are especially useful to predict future input in natural videos, even when networks are not required to segment foreground from background. This suggests that BOS neurons in the brain might be the result of evolutionary or developmental pressure to predict future input in natural, complex dynamic visual environments.
Collapse
Affiliation(s)
- Zeyuan Ye
- Department of Physics, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ralf Wessel
- Department of Physics, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Tom P. Franken
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
34
|
Warder BN, Nelson KA, Sui J, Anllo L, DiNardo S. An actomyosin network organizes niche morphology and responds to feedback from recruited stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.08.556877. [PMID: 38746236 PMCID: PMC11092431 DOI: 10.1101/2023.09.08.556877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Stem cells often rely on signals from a niche, which in many tissues adopts a precise morphology. What remains elusive is how niches are formed, and how morphology impacts function. To address this, we leverage the Drosophila gonadal niche, which affords genetic tractability and live-imaging. We have previously shown mechanisms dictating niche cell migration to their appropriate position within the gonad, and the resultant consequences on niche function. Here, we show that once positioned, niche cells robustly polarize filamentous actin (F-actin) and Non-muscle Myosin II (MyoII) towards neighboring germ cells. Actomyosin tension along the niche periphery generates a highly reproducible smoothened contour. Without contractility, niches are misshapen and exhibit defects in their ability to regulate germline stem cell behavior. We additionally show that germ cells aid in polarizing MyoII within niche cells, and that extrinsic input is required for niche morphogenesis and function. Our work reveals a feedback mechanism where stem cells shape the niche that guides their behavior.
Collapse
Affiliation(s)
- Bailey N. Warder
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kara A. Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justin Sui
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
35
|
Piñeiro-Sabarís R, MacGrogan D, de la Pompa JL. Intricate MIB1-NOTCH-GATA6 Interactions in Cardiac Valvular and Septal Development. J Cardiovasc Dev Dis 2024; 11:223. [PMID: 39057643 PMCID: PMC11277162 DOI: 10.3390/jcdd11070223] [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: 06/05/2024] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Genome-wide association studies and experimental mouse models implicate the MIB1 and GATA6 genes in congenital heart disease (CHD). Their close physical proximity and conserved synteny suggest that these two genes might be involved in analogous cardiac developmental processes. Heterozygous Gata6 loss-of-function mutations alone or humanized Mib1 mutations in a NOTCH1-sensitized genetic background cause bicuspid aortic valve (BAV) and a membranous ventricular septal defect (VSD), consistent with MIB1 and NOTCH1 functioning in the same pathway. To determine if MIB1-NOTCH and GATA6 interact in valvular and septal development, we generated compound heterozygote mice carrying different Mib1 missense (Mib1K735R and Mib1V943F) or nonsense (Mib1R530X) mutations with the Gata6STOP/+ heterozygous null mutation. Combining Mib1R530X/+ or Mib1K735R/+ with Gata6STOP/+ does not affect Gata6STOP/+ single mutant phenotypes. In contrast, combining Mib1V943F/+ with Gata6STOP/+ decreases the incidence of BAV and VSD by 50%, suggesting a suppressive effect of Mib1V943F/+ on Gata6STOP/+. Transcriptomic and functional analyses revealed that while the EMT pathway term is depleted in the Gata6STOP/+ mutant, introducing the Mib1V943F variant robustly enriches this term, consistent with the Mib1V943F/+ phenotypic suppression of Gata6STOP/+. Interestingly, combined Notch1 and Gata6 insufficiency led to a nearly fully penetrant VSD but did not affect the BAV phenotype, underscoring the complex functional relationship between MIB1, NOTCH, and GATA6 in valvular and septal development.
Collapse
Affiliation(s)
- Rebeca Piñeiro-Sabarís
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain;
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Donal MacGrogan
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain;
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain;
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| |
Collapse
|
36
|
Grosely R, Alvarado C, Ivanov IP, Nicholson OB, Puglisi JD, Dever TE, Lapointe CP. eIF1 and eIF5 dynamically control translation start site fidelity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602410. [PMID: 39026837 PMCID: PMC11257575 DOI: 10.1101/2024.07.10.602410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Translation initiation defines the identity of a synthesized protein through selection of a translation start site on a messenger RNA. This process is essential to well-controlled protein synthesis, modulated by stress responses, and dysregulated in many human diseases. The eukaryotic initiation factors eIF1 and eIF5 interact with the initiator methionyl-tRNAi Met on the 40S ribosomal subunit to coordinate start site selection. Here, using single-molecule analysis of in vitro reconstituted human initiation combined with translation assays in cells, we examine eIF1 and eIF5 function. During translation initiation on a panel of RNAs, we monitored both proteins directly and in real time using single-molecule fluorescence. As expected, eIF1 loaded onto mRNAs as a component of the 43S initiation complex. Rapid (~ 2 s) eIF1 departure required a translation start site and was delayed by alternative start sites and a longer 5' untranslated region (5'UTR). After its initial departure, eIF1 rapidly and transiently sampled initiation complexes, with more prolonged sampling events on alternative start sites. By contrast, eIF5 only transiently bound initiation complexes late in initiation immediately prior to association of eIF5B, which allowed joining of the 60S ribosomal subunit. eIF5 association required the presence of a translation start site and was inhibited and destabilized by alternative start sites. Using both knockdown and overexpression experiments in human cells, we validated that eIF1 and eIF5 have opposing roles during initiation. Collectively, our findings demonstrate how multiple eIF1 and eIF5 binding events control start-site selection fidelity throughout initiation, which is tuned in response to changes in the levels of both proteins.
Collapse
Affiliation(s)
- Rosslyn Grosely
- Dept. of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Carlos Alvarado
- Dept. of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ivaylo P. Ivanov
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | | | - Joseph D. Puglisi
- Dept. of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas E. Dever
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
37
|
Akirtava C, May G, McManus CJ. Deciphering the cis-regulatory landscape of natural yeast Transcript Leaders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.03.601937. [PMID: 39005336 PMCID: PMC11245039 DOI: 10.1101/2024.07.03.601937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Protein synthesis is a vital process that is highly regulated at the initiation step of translation. Eukaryotic 5' transcript leaders (TLs) contain a variety of cis-regulatory features that influence translation and mRNA stability. However, the relative influences of these features in natural TLs are poorly characterized. To address this, we used massively parallel reporter assays (MPRAs) to quantify RNA levels, ribosome loading, and protein levels from 11,027 natural yeast TLs in vivo and systematically compared the relative impacts of their sequence features on gene expression. We found that yeast TLs influence gene expression over two orders of magnitude. While a leaky scanning model using Kozak contexts and uAUGs explained half of the variance in expression across transcript leaders, the addition of other features explained ~70% of gene expression variation. Our analyses detected key cis-acting sequence features, quantified their effects in vivo, and compared their roles to motifs reported from an in vitro study of ribosome recruitment. In addition, our work quantitated the effects of alternative transcription start site usage on gene expression in yeast. Thus, our study provides new quantitative insights into the roles of TL cis-acting sequences in regulating gene expression.
Collapse
Affiliation(s)
- Christina Akirtava
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- RNA Bioscience Initiative, University of Colorado - Anshutz, Aurora, CO, 80045, USA
| | - Gemma May
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - C Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| |
Collapse
|
38
|
Lobel JH, Ingolia NT. Precise measurement of molecular phenotypes with barcode-based CRISPRi systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600132. [PMID: 38948701 PMCID: PMC11213135 DOI: 10.1101/2024.06.21.600132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Genome-wide CRISPR-Cas9 screens have untangled regulatory networks and revealed the genetic underpinnings of diverse biological processes. Their success relies on experimental designs that interrogate specific molecular phenotypes and distinguish key regulators from background effects. Here, we realize these goals with a generalizable platform for CRISPR interference with barcoded expression reporter sequencing (CiBER-seq) that dramatically improves the sensitivity and scope of genome-wide screens. We systematically address technical factors that distort phenotypic measurements by normalizing expression reporters against closely-matched control promoters, integrated together into the genome at single copy. To test our ability to capture post-transcriptional and post-translational regulation through sequencing, we screened for genes that affected nonsense-mediated mRNA decay and Doa10-mediated cytosolic protein decay. Our optimized CiBER-seq screens accurately capture the known components of well-studied RNA and protein quality control pathways with minimal background. These results demonstrate the precision and versatility of CiBER-seq for dissecting the genetic networks controlling cellular behaviors.
Collapse
Affiliation(s)
- Joseph H. Lobel
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Nicholas T. Ingolia
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Lead contact
| |
Collapse
|
39
|
Packer J, Gubieda AG, Brooks A, Deutz LN, Squires I, Ellison S, Schneider C, Naganathan SR, Wollman AJ, Dickinson DJ, Rodriguez J. Atypical Protein Kinase C Promotes its own Asymmetric Localisation by Phosphorylating Cdc42 in the C. elegans zygote. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.27.563985. [PMID: 38009101 PMCID: PMC10675845 DOI: 10.1101/2023.10.27.563985] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
Atypical protein kinase C (aPKC) is a major regulator of cell polarity. Acting in conjunction with Par6, Par3 and the small GTPase Cdc42, aPKC becomes asymmetrically localised and drives the polarisation of cells. aPKC activity is crucial for its own asymmetric localisation, suggesting a hitherto unknown feedback mechanism contributing to polarisation. Here we show in the C. elegans zygote that the feedback relies on aPKC phosphorylation of Cdc42 at serine 71. The turnover of CDC-42 phosphorylation ensures optimal aPKC asymmetry and activity throughout polarisation by tuning Par6/aPKC association with Par3 and Cdc42. Moreover, turnover of Cdc42 phosphorylation regulates actomyosin cortex dynamics that are known to drive aPKC asymmetry. Given the widespread role of aPKC and Cdc42 in cell polarity, this form of self-regulation of aPKC may be vital for the robust control of polarisation in many cell types.
Collapse
Affiliation(s)
- John Packer
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- These authors contributed equally
| | - Alicia G. Gubieda
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- These authors contributed equally
| | - Aaron Brooks
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- These authors contributed equally
| | - Lars N. Deutz
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
- These authors contributed equally
| | - Iolo Squires
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- These authors contributed equally
| | | | | | - Sundar Ram Naganathan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Adam J.M. Wollman
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Daniel J. Dickinson
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Josana Rodriguez
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Lead contact
| |
Collapse
|
40
|
Prescott NA, Mansisidor A, Bram Y, Biaco T, Rendleman J, Faulkner SC, Lemmon AA, Lim C, Hamard PJ, Koche RP, Risca VI, Schwartz RE, David Y. A nucleosome switch primes Hepatitis B Virus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.03.531011. [PMID: 38915612 PMCID: PMC11195122 DOI: 10.1101/2023.03.03.531011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Chronic hepatitis B virus (HBV) infection is an incurable global health threat responsible for causing liver disease and hepatocellular carcinoma. During the genesis of infection, HBV establishes an independent minichromosome consisting of the viral covalently closed circular DNA (cccDNA) genome and host histones. The viral X gene must be expressed immediately upon infection to induce degradation of the host silencing factor, Smc5/6. However, the relationship between cccDNA chromatinization and X gene transcription remains poorly understood. Establishing a reconstituted viral minichromosome platform, we found that nucleosome occupancy in cccDNA drives X transcription. We corroborated these findings in cells and further showed that the chromatin destabilizing molecule CBL137 inhibits X transcription and HBV infection in hepatocytes. Our results shed light on a long-standing paradox and represent a potential new therapeutic avenue for the treatment of chronic HBV infection.
Collapse
Affiliation(s)
- Nicholas A. Prescott
- Tri-Institutional PhD Program in Chemical Biology; New York, NY 10065, USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Andrés Mansisidor
- Laboratory of Genome Architecture and Dynamics, The Rockefeller University; New York, NY 10065, USA
- These authors contributed equally
| | - Yaron Bram
- Division of Gastroenterology & Hepatology, Department of Medicine, Weill Cornell Medicine; New York, NY 10065, USA
- These authors contributed equally
| | - Tracy Biaco
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
- Department of Pharmacology, Weill Cornell Medicine; New York, NY 10065, USA
- These authors contributed equally
| | - Justin Rendleman
- Laboratory of Genome Architecture and Dynamics, The Rockefeller University; New York, NY 10065, USA
| | - Sarah C. Faulkner
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Abigail A. Lemmon
- Tri-Institutional PhD Program in Chemical Biology; New York, NY 10065, USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Christine Lim
- Division of Gastroenterology & Hepatology, Department of Medicine, Weill Cornell Medicine; New York, NY 10065, USA
| | - Pierre-Jacques Hamard
- Epigenetics Research Innovation Lab, Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Richard P. Koche
- Epigenetics Research Innovation Lab, Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Viviana I. Risca
- Laboratory of Genome Architecture and Dynamics, The Rockefeller University; New York, NY 10065, USA
| | - Robert E. Schwartz
- Division of Gastroenterology & Hepatology, Department of Medicine, Weill Cornell Medicine; New York, NY 10065, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine; New York, NY 10065, USA
| | - Yael David
- Tri-Institutional PhD Program in Chemical Biology; New York, NY 10065, USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
- Department of Pharmacology, Weill Cornell Medicine; New York, NY 10065, USA
- Lead Contact
| |
Collapse
|
41
|
Schuck RJ, Ward AE, Sahoo AR, Rybak JA, Pyron RJ, Trybala TN, Simmons TB, Baccile JA, Sgouralis I, Buck M, Lamichhane R, Barrera FN. Cholesterol inhibits assembly and activation of the EphA2 receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598255. [PMID: 38915729 PMCID: PMC11195142 DOI: 10.1101/2024.06.10.598255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The receptor tyrosine kinase EphA2 drives cancer malignancy by facilitating metastasis. EphA2 can be found in different self-assembly states: as a monomer, dimer, and oligomer. However, our understanding remains limited regarding which EphA2 state is responsible for driving pro-metastatic signaling. To address this limitation, we have developed SiMPull-POP, a single-molecule method for accurate quantification of membrane protein self-assembly. Our experiments revealed that a reduction of plasma membrane cholesterol strongly promoted EphA2 self-assembly. Indeed, low cholesterol caused a similar effect to the EphA2 ligand ephrinA1-Fc. These results indicate that cholesterol inhibits EphA2 assembly. Phosphorylation studies in different cell lines revealed that low cholesterol increased phospho-serine levels, the signature of oncogenic signaling. Investigation of the mechanism that cholesterol uses to inhibit the assembly and activity of EphA2 indicate an in-trans effect, where EphA2 is phosphorylated by protein kinase A downstream of beta-adrenergic receptor activity, which cholesterol also inhibits. Our study not only provides new mechanistic insights on EphA2 oncogenic function, but also suggests that cholesterol acts as a molecular safeguard mechanism that prevents uncontrolled self-assembly and activation of EphA2.
Collapse
Affiliation(s)
- Ryan J Schuck
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, USA
| | - Alyssa E Ward
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, USA
| | - Amita R Sahoo
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, USA
| | - Jennifer A Rybak
- Genome Science and Technology, University of Tennessee, Knoxville, USA
| | - Robert J Pyron
- Genome Science and Technology, University of Tennessee, Knoxville, USA
| | - Thomas N Trybala
- Department of Chemistry, University of Tennessee, Knoxville, USA
| | - Timothy B Simmons
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, USA
| | - Joshua A Baccile
- Department of Chemistry, University of Tennessee, Knoxville, USA
| | | | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, USA
| | - Rajan Lamichhane
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, USA
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, USA
| |
Collapse
|
42
|
Dintzner E, Bandekar SJ, Leon K, Cechova K, Vafabakhsh R, Araç D. The far extracellular CUB domain of the adhesion GPCR ADGRG6/GPR126 is a key regulator of receptor signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.16.580607. [PMID: 38766069 PMCID: PMC11100614 DOI: 10.1101/2024.02.16.580607] [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
Adhesion G Protein-coupled receptors (aGPCRs) transduce extracellular adhesion signals into cytoplasmic signaling pathways. ADGRG6/GPR126 is an aGPCR critical for axon myelination, heart development and ear development; and is associated with developmental diseases and cancers. ADGRG6 has a large, alternatively-spliced, five-domain extracellular region (ECR) that samples different conformations and regulates receptor signaling. However, the molecular details of how the ECR regulates signaling are unclear. Herein, we studied the conformational dynamics of the conserved CUB domain which is located at the distal N-terminus of the ECR and is deleted in an alternatively-spliced isoform ( Δ CUB). We showed that the Δ CUB isoform has decreased signaling. Molecular dynamics simulations suggest that the CUB domain is involved in interdomain contacts to maintain a compact ECR conformation. A cancer-associated CUB domain mutant, C94Y, drastically perturbs the ECR conformation and results in elevated signaling, whereas another CUB mutant, Y96A, located near a conserved Ca 2+ -binding site, decreases signaling. Our results suggest an ECR-mediated mechanism for ADGRG6 regulation in which the CUB domain instructs conformational changes within the ECR to regulate receptor signaling.
Collapse
|
43
|
Dutcher HA, Hose J, Howe H, Rojas J, Gasch AP. The response to single-gene duplication implicates translation as a key vulnerability in aneuploid yeast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589582. [PMID: 38659764 PMCID: PMC11042342 DOI: 10.1101/2024.04.15.589582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Aneuploidy produces myriad consequences in health and disease, yet models of the deleterious effects of chromosome amplification are still widely debated. To distinguish the molecular determinants of aneuploidy stress, we measured the effects of duplicating individual genes in cells with varying chromosome duplications, in wild-type cells and cells sensitized to aneuploidy by deletion of RNA-binding protein Ssd1. We identified gene duplications that are nearly neutral in wild-type euploid cells but significantly deleterious in euploids lacking SSD1 or SSD1+ aneuploid cells with different chromosome duplications. Several of the most deleterious genes are linked to translation; in contrast, duplication of other translational regulators, including eI5Fa Hyp2, benefit ssd1Δ aneuploids over controls. Using modeling of aneuploid growth defects, we propose that the deleterious effects of aneuploidy emerge from an interaction between the cumulative burden of many amplified genes on a chromosome and a subset of duplicated genes that become toxic in that context. Our results suggest that the mechanism behind their toxicity is linked to a key vulnerability in translation in aneuploid cells. These findings provide a perspective on the dual impact of individual genes and overall genomic burden, offering new avenues for understanding aneuploidy and its cellular consequences.
Collapse
|
44
|
Birch S, McGee L, Provencher C, DeMio C, Plachetzki D. Phototactic preference and its genetic basis in the planulae of the colonial Hydrozoan Hydractinia symbiolongicarpus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.585045. [PMID: 38617216 PMCID: PMC11014542 DOI: 10.1101/2024.03.28.585045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Background Marine organisms with sessile adults commonly possess motile larval stages that make settlement decisions based on integrating environmental sensory cues. Phototaxis, the movement toward or away from light, is a common behavioral characteristic of aquatic and marine metazoan larvae, and of algae, protists, and fungi. In cnidarians, behavioral genomic investigations of motile planulae larvae have been conducted in anthozoans (corals and sea anemones) and scyphozoans (true jellyfish), but such studies are presently lacking in hydrozoans. Here, we examined the behavioral genomics of phototaxis in planulae of the hydrozoan Hydractinia symbiolongicarpus. Results A behavioral phototaxis study of day 3 planulae indicated preferential phototaxis to green (523 nm) and blue (470 nm) wavelengths of light, but not red (625 nm) wavelengths. A developmental transcriptome study where planula larvae were collected from four developmental time points for RNA-seq revealed that many genes critical to the physiology and development of ciliary photosensory systems are dynamically expressed in planula development and correspond to the expression of phototactic behavior. Microscopical investigations using immunohistochemistry and in situ hybridization demonstrated that several transcripts with predicted function in photoreceptors, including cnidops class opsin, CNG ion channel, and CRX-like transcription factor, localize to ciliated bipolar sensory neurons of the aboral sensory neural plexus, which is associated with the direction of phototaxis and the site of settlement. Conclusions The phototactic preference displayed by planulae is consistent with the shallow sandy marine habitats they experience in nature. Our genomic investigations add further evidence of similarities between cnidops-mediated photoreceptors of hydrozoans and other cnidarians and ciliary photoreceptors as found in the eyes of humans and other bilaterians, suggesting aspects of their shared evolutionary history.
Collapse
Affiliation(s)
- Sydney Birch
- Department of Molecular, Cellular, and Biomedical Sciences; University of New Hampshire; Durham, NH, 03824; USA
- Department of Biological Sciences; University of North Carolina Charlotte; Charlotte, NC, 28223; USA
| | - Lindy McGee
- Department of Molecular, Cellular, and Biomedical Sciences; University of New Hampshire; Durham, NH, 03824; USA
| | - Curtis Provencher
- Department of Molecular, Cellular, and Biomedical Sciences; University of New Hampshire; Durham, NH, 03824; USA
| | - Christine DeMio
- Department of Molecular, Cellular, and Biomedical Sciences; University of New Hampshire; Durham, NH, 03824; USA
| | - David Plachetzki
- Department of Molecular, Cellular, and Biomedical Sciences; University of New Hampshire; Durham, NH, 03824; USA
| |
Collapse
|
45
|
Anderson RH, Lamers WH, Hikspoors JPJM, Mohun TJ, Bamforth SD, Chaudhry B, Eley L, Kerwin J, Crosier M, Henderson DJ. Development of the arterial roots and ventricular outflow tracts. J Anat 2024; 244:497-513. [PMID: 37957890 PMCID: PMC10862166 DOI: 10.1111/joa.13973] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/05/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
The separation of the outflow tract of the developing heart into the systemic and pulmonary arterial channels remains controversial and poorly understood. The definitive outflow tracts have three components. The developing outflow tract, in contrast, has usually been described in two parts. When the tract has exclusively myocardial walls, such bipartite description is justified, with an obvious dogleg bend separating proximal and distal components. With the addition of non-myocardial walls distally, it becomes possible to recognise three parts. The middle part, which initially still has myocardial walls, contains within its lumen a pair of intercalated valvar swellings. The swellings interdigitate with the distal ends of major outflow cushions, formed by the remodelling of cardiac jelly, to form the primordiums of the arterial roots. The proximal parts of the major cushions, occupying the proximal part of the outflow tract, which also has myocardial walls, themselves fuse and muscularise. The myocardial shelf thus formed remodels to become the free-standing subpulmonary infundibulum. Details of all these processes are currently lacking. In this account, we describe the anatomical changes seen during the overall remodelling. Our interpretations are based on the interrogation of serially sectioned histological and high-resolution episcopic microscopy datasets prepared from developing human and mouse embryos, with some of the datasets processed and reconstructed to reveal the specific nature of the tissues contributing to the separation of the outflow channels. Our findings confirm that the tripartite postnatal arrangement can be correlated with the changes occurring during development.
Collapse
Affiliation(s)
| | - Wouter H. Lamers
- Department of Anatomy & EmbryologyMaastricht UniversityMaastrichtThe Netherlands
| | | | | | | | - Bill Chaudhry
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Lorraine Eley
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Janet Kerwin
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Moira Crosier
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | | |
Collapse
|
46
|
Jones DL, Morley MP, Li X, Ying Y, Cardenas-Diaz FL, Li S, Zhou S, Schaefer SE, Chembazhi UV, Nottingham A, Lin S, Cantu E, Diamond JM, Basil MC, Vaughan AE, Morrisey EE. An injury-induced tissue niche shaped by mesenchymal plasticity coordinates the regenerative and disease response in the lung. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582147. [PMID: 38529490 PMCID: PMC10962740 DOI: 10.1101/2024.02.26.582147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Severe lung injury causes basal stem cells to migrate and outcompete alveolar stem cells resulting in dysplastic repair and a loss of gas exchange function. This "stem cell collision" is part of a multistep process that is now revealed to generate an injury-induced tissue niche (iTCH) containing Keratin 5+ epithelial cells and plastic Pdgfra+ mesenchymal cells. Temporal and spatial single cell analysis reveals that iTCHs are governed by mesenchymal proliferation and Notch signaling, which suppresses Wnt and Fgf signaling in iTCHs. Conversely, loss of Notch in iTCHs rewires alveolar signaling patterns to promote euplastic regeneration and gas exchange. The signaling patterns of iTCHs can differentially phenotype fibrotic from degenerative human lung diseases, through apposing flows of FGF and WNT signaling. These data reveal the emergence of an injury and disease associated iTCH in the lung and the ability of using iTCH specific signaling patterns to discriminate human lung disease phenotypes.
Collapse
Affiliation(s)
- Dakota L. Jones
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P. Morley
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Xinyuan Li
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yun Ying
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fabian L. Cardenas-Diaz
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shanru Li
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Su Zhou
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah E. Schaefer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ullas V. Chembazhi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ana Nottingham
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan Lin
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Cantu
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua M. Diamond
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria C. Basil
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew E. Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward E. Morrisey
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
47
|
Lin Y, Yang Q, Lin X, Liu X, Qian Y, Xu D, Cao N, Han X, Zhu Y, Hu W, He X, Yu Z, Kong X, Zhu L, Zhong Z, Liu K, Zhou B, Wang Y, Peng J, Zhu W, Wang J. Extracellular Matrix Disorganization Caused by ADAMTS16 Deficiency Leads to Bicuspid Aortic Valve With Raphe Formation. Circulation 2024; 149:605-626. [PMID: 38018454 DOI: 10.1161/circulationaha.123.065458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 11/03/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND A better understanding of the molecular mechanism of aortic valve development and bicuspid aortic valve (BAV) formation would significantly improve and optimize the therapeutic strategy for BAV treatment. Over the past decade, the genes involved in aortic valve development and BAV formation have been increasingly recognized. On the other hand, ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) gene family members have been reported to be able to modulate cardiovascular development and diseases. The present study aimed to further investigate the roles of ADAMTS family members in aortic valve development and BAV formation. METHODS Morpholino-based ADAMTS family gene-targeted screening for zebrafish heart outflow tract phenotypes combined with DNA sequencing in a 304 cohort BAV patient registry study was initially carried out to identify potentially related genes. Both ADAMTS gene-specific fluorescence in situ hybridization assay and genetic tracing experiments were performed to evaluate the expression pattern in the aortic valve. Accordingly, related genetic mouse models (both knockout and knockin) were generated using the CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9) method to further study the roles of ADAMTS family genes. The lineage-tracing technique was used again to evaluate how the cellular activity of specific progenitor cells was regulated by ADAMTS genes. Bulk RNA sequencing was used to investigate the signaling pathways involved. Inducible pluripotent stem cells derived from both BAV patients and genetic mouse tissue were used to study the molecular mechanism of ADAMTS. Immunohistochemistry was performed to examine the phenotype of cardiac valve anomalies, especially in the extracellular matrix components. RESULTS ADAMTS genes targeting and phenotype screening in zebrafish and targeted DNA sequencing on a cohort of patients with BAV identified ADAMTS16 (a disintegrin and metalloproteinase with thrombospondin motifs 16) as a BAV-causing gene and found the ADAMTS16 p. H357Q variant in an inherited BAV family. Both in situ hybridization and genetic tracing studies described a unique spatiotemporal pattern of ADAMTS16 expression during aortic valve development. Adamts16+/- and Adamts16+/H355Q mouse models both exhibited a right coronary cusp-noncoronary cusp fusion-type BAV phenotype, with progressive aortic valve thickening associated with raphe formation (fusion of the commissure). Further, ADAMTS16 deficiency in Tie2 lineage cells recapitulated the BAV phenotype. This was confirmed in lineage-tracing mouse models in which Adamts16 deficiency affected endothelial and second heart field cells, not the neural crest cells. Accordingly, the changes were mainly detected in the noncoronary and right coronary leaflets. Bulk RNA sequencing using inducible pluripotent stem cells-derived endothelial cells and genetic mouse embryonic heart tissue unveiled enhanced FAK (focal adhesion kinase) signaling, which was accompanied by elevated fibronectin levels. Both in vitro inducible pluripotent stem cells-derived endothelial cells culture and ex vivo embryonic outflow tract explant studies validated the altered FAK signaling. CONCLUSIONS Our present study identified a novel BAV-causing ADAMTS16 p. H357Q variant. ADAMTS16 deficiency led to BAV formation.
Collapse
Affiliation(s)
- Ying Lin
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Qifan Yang
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xiaoping Lin
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xianbao Liu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Yi Qian
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Dilin Xu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Naifang Cao
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (X.H.)
| | - Yanqing Zhu
- Ministry of Education Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network (Y.Z., K.L., J.P.), Hangzhou, China
| | - Wangxing Hu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xiaopeng He
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Zhengyang Yu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xiangmin Kong
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Lianlian Zhu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Zhiwei Zhong
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Kai Liu
- Ministry of Education Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network (Y.Z., K.L., J.P.), Hangzhou, China
| | - Bin Zhou
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences (B.Z.)
| | - Yidong Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University Health Science Center, China (Y.W.)
| | - Jinrong Peng
- Ministry of Education Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network (Y.Z., K.L., J.P.), Hangzhou, China
| | - Wei Zhu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Jian'an Wang
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- Research Center for Life Science and Human Health, Binjiang Institute (J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| |
Collapse
|
48
|
Alahmari AA, Chaubey AH, Jonnakuti VS, Tisdale AA, Schwarz CD, Cornwell AC, Maraszek KE, Paterson EJ, Kim M, Venkat S, Gomez EC, Wang J, Gurova KV, Yalamanchili HK, Feigin ME. CPSF3 inhibition blocks pancreatic cancer cell proliferation through disruption of core histone mRNA processing. RNA (NEW YORK, N.Y.) 2024; 30:281-297. [PMID: 38191171 PMCID: PMC10870380 DOI: 10.1261/rna.079931.123] [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: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 01/10/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with limited effective treatment options, potentiating the importance of uncovering novel drug targets. Here, we target cleavage and polyadenylation specificity factor 3 (CPSF3), the 3' endonuclease that catalyzes mRNA cleavage during polyadenylation and histone mRNA processing. We find that CPSF3 is highly expressed in PDAC and is associated with poor prognosis. CPSF3 knockdown blocks PDAC cell proliferation and colony formation in vitro and tumor growth in vivo. Chemical inhibition of CPSF3 by the small molecule JTE-607 also attenuates PDAC cell proliferation and colony formation, while it has no effect on cell proliferation of nontransformed immortalized control pancreatic cells. Mechanistically, JTE-607 induces transcriptional readthrough in replication-dependent histones, reduces core histone expression, destabilizes chromatin structure, and arrests cells in the S-phase of the cell cycle. Therefore, CPSF3 represents a potential therapeutic target for the treatment of PDAC.
Collapse
Affiliation(s)
- Abdulrahman A Alahmari
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Aditi H Chaubey
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Venkata S Jonnakuti
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
- Program in Quantitative and Computational Biology, Baylor College of Medicine, Houston, Texas 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Arwen A Tisdale
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Carla D Schwarz
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Abigail C Cornwell
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Kathryn E Maraszek
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Emily J Paterson
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Minsuh Kim
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Swati Venkat
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Katerina V Gurova
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| | - Hari Krishna Yalamanchili
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030, USA
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Michael E Feigin
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, USA
| |
Collapse
|
49
|
Park JH, Hothi P, Lopez Garcia de Lomana A, Pan M, Calder R, Turkarslan S, Wu WJ, Lee H, Patel AP, Cobbs C, Huang S, Baliga NS. Gene regulatory network topology governs resistance and treatment escape in glioma stem-like cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578510. [PMID: 38370784 PMCID: PMC10871280 DOI: 10.1101/2024.02.02.578510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Poor prognosis and drug resistance in glioblastoma (GBM) can result from cellular heterogeneity and treatment-induced shifts in phenotypic states of tumor cells, including dedifferentiation into glioma stem-like cells (GSCs). This rare tumorigenic cell subpopulation resists temozolomide, undergoes proneural-to-mesenchymal transition (PMT) to evade therapy, and drives recurrence. Through inference of transcriptional regulatory networks (TRNs) of patient-derived GSCs (PD-GSCs) at single-cell resolution, we demonstrate how the topology of transcription factor interaction networks drives distinct trajectories of cell state transitions in PD-GSCs resistant or susceptible to cytotoxic drug treatment. By experimentally testing predictions based on TRN simulations, we show that drug treatment drives surviving PD-GSCs along a trajectory of intermediate states, exposing vulnerability to potentiated killing by siRNA or a second drug targeting treatment-induced transcriptional programs governing non-genetic cell plasticity. Our findings demonstrate an approach to uncover TRN topology and use it to rationally predict combinatorial treatments that disrupts acquired resistance in GBM.
Collapse
Affiliation(s)
| | - Parvinder Hothi
- Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA
| | | | - Min Pan
- Institute for Systems Biology, Seattle, WA
| | | | | | - Wei-Ju Wu
- Institute for Systems Biology, Seattle, WA
| | - Hwahyung Lee
- Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA
| | - Anoop P Patel
- Department of Neurosurgery, Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC
- Center for Advanced Genomic Technologies, Duke University, Durham, NC
| | - Charles Cobbs
- Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA
| | - Sui Huang
- Institute for Systems Biology, Seattle, WA
| | - Nitin S Baliga
- Institute for Systems Biology, Seattle, WA
- Departments of Microbiology, Biology, and Molecular Engineering Sciences, University of Washington, Seattle, WA
| |
Collapse
|
50
|
Kratz JC, Banerjee S. Gene expression tradeoffs determine bacterial survival and adaptation to antibiotic stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.20.576495. [PMID: 38328084 PMCID: PMC10849509 DOI: 10.1101/2024.01.20.576495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
To optimize their fitness, cells face the crucial task of efficiently responding to various stresses. This necessitates striking a balance between conserving resources for survival and allocating resources for growth and division. The fundamental principles governing these tradeoffs is an outstanding challenge in the physics of living systems. In this study, we introduce a coarse-grained theoretical framework for bacterial physiology that establishes a connection between the physiological state of cells and their survival outcomes in dynamic environments, particularly in the context of antibiotic exposure. Predicting bacterial survival responses to varying antibiotic doses proves challenging due to the profound influence of the physiological state on critical parameters, such as the Minimum Inhibitory Concentration (MIC) and killing rates, even within an isogenic cell population. Our proposed theoretical model bridges the gap by linking extracellular antibiotic concentration and nutrient quality to intracellular damage accumulation and gene expression. This framework allows us to predict and explain the control of cellular growth rate, death rate, MIC and survival fraction in a wide range of time-varying environments. Surprisingly, our model reveals that cell death is rarely due to antibiotic levels being above the maximum physiological limit, but instead survival is limited by the inability to alter gene expression sufficiently quickly to transition to a less susceptible physiological state. Moreover, bacteria tend to overexpress stress response genes at the expense of reduced growth, conferring greater protection against further antibiotic exposure. This strategy is in contrast to those employed in different nutrient environments, in which bacteria allocate resources to maximize growth rate. This highlights an important tradeoff between the cellular capacity for growth and the ability to survive antibiotic exposure.
Collapse
Affiliation(s)
- Josiah C. Kratz
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Shiladitya Banerjee
- Department of Physics, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|