101
|
Du Y, Fan X, Song C, Chang W, Xiong J, Deng L, Ji WK. Sec23IP recruits VPS13B/COH1 to ER exit site-Golgi interface for tubular ERGIC formation. J Cell Biol 2024; 223:e202402083. [PMID: 39352497 PMCID: PMC11457499 DOI: 10.1083/jcb.202402083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/25/2024] [Accepted: 08/07/2024] [Indexed: 10/09/2024] Open
Abstract
VPS13B/COH1 is the only known causative factor for Cohen syndrome, an early-onset autosomal recessive developmental disorder with intellectual inability, developmental delay, joint hypermobility, myopia, and facial dysmorphism as common features, but the molecular basis of VPS13B/COH1 in pathogenesis remains largely unclear. Here, we identify Sec23 interacting protein (Sec23IP) at the ER exit site (ERES) as a VPS13B adaptor that recruits VPS13B to ERES-Golgi interfaces. VPS13B interacts directly with Sec23IP via the VPS13 adaptor binding domain (VAB), and the interaction promotes the association between ERES and the Golgi. Disease-associated missense mutations of VPS13B-VAB impair the interaction with Sec23IP. Knockout of VPS13B or Sec23IP blocks the formation of tubular ERGIC, an unconventional cargo carrier that expedites ER-to-Golgi transport. In addition, depletion of VPS13B or Sec23IP delays ER export of procollagen, suggesting a link between procollagen secretion and joint laxity in patients with Cohen disease. Together, our study reveals a crucial role of VPS13B-Sec23IP interaction at the ERES-Golgi interface in the pathogenesis of Cohen syndrome.
Collapse
Affiliation(s)
- Yuanjiao Du
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyu Fan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | | | | | - Juan Xiong
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Deng
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Wei-Ke Ji
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, China
- Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
102
|
Silverman JB, Krystofiak EE, Caplan LR, Lau KS, Tyska MJ. Organization of a cytoskeletal superstructure in the apical domain of intestinal tuft cells. J Cell Biol 2024; 223:e202404070. [PMID: 39352498 PMCID: PMC11457492 DOI: 10.1083/jcb.202404070] [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: 04/13/2024] [Revised: 08/26/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024] Open
Abstract
Tuft cells are a rare epithelial cell type that play important roles in sensing and responding to luminal antigens. A defining morphological feature of this lineage is the actin-rich apical "tuft," which contains large fingerlike protrusions. However, details of the cytoskeletal ultrastructure underpinning the tuft, the molecules involved in building this structure, or how it supports tuft cell biology remain unclear. In the context of the small intestine, we found that tuft cell protrusions are supported by long-core bundles that consist of F-actin crosslinked in a parallel and polarized configuration; they also contain a tuft cell-specific complement of actin-binding proteins that exhibit regionalized localization along the bundle axis. Remarkably, in the sub-apical cytoplasm, the array of core actin bundles interdigitates and co-aligns with a highly ordered network of microtubules. The resulting cytoskeletal superstructure is well positioned to support subcellular transport and, in turn, the dynamic sensing functions of the tuft cell that are critical for intestinal homeostasis.
Collapse
Affiliation(s)
- Jennifer B. Silverman
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Evan E. Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Cell Imaging Shared Resource, Vanderbilt University, Nashville, TN, USA
| | - Leah R. Caplan
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ken S. Lau
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
103
|
Rötte M, Höhne MY, Klug D, Ramlow K, Zedler C, Lehne F, Schneider M, Bischoff MC, Bogdan S. CYRI controls epidermal wound closure and cohesion of invasive border cell cluster in Drosophila. J Cell Biol 2024; 223:e202310153. [PMID: 39453414 PMCID: PMC11519390 DOI: 10.1083/jcb.202310153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 07/19/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024] Open
Abstract
Cell motility is crucial for many biological processes including morphogenesis, wound healing, and cancer invasion. The WAVE regulatory complex (WRC) is a central Arp2/3 regulator driving cell motility downstream of activation by Rac GTPase. CYFIP-related Rac1 interactor (CYRI) proteins are thought to compete with WRC for interaction with Rac1 in a feedback loop regulating lamellipodia dynamics. However, the physiological role of CYRI proteins in vivo in healthy tissues is unclear. Here, we used Drosophila as a model system to study CYRI function at the cellular and organismal levels. We found that CYRI is not only a potent WRC regulator in single macrophages that controls lamellipodial spreading but also identified CYRI as a molecular brake on the Rac-WRC-Arp2/3 pathway to slow down epidermal wound healing. In addition, we found that CYRI limits invasive border cell migration by controlling cluster cohesion and migration. Thus, our data highlight CYRI as an important regulator of cellular and epithelial tissue dynamics conserved across species.
Collapse
Affiliation(s)
- Marvin Rötte
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Mila Y. Höhne
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Dennis Klug
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Kirsten Ramlow
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Caroline Zedler
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Franziska Lehne
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Meike Schneider
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Maik C. Bischoff
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Sven Bogdan
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
104
|
Barefield DY. Myosin-binding protein-H: Not just filler. J Gen Physiol 2024; 156:e202413622. [PMID: 39485243 PMCID: PMC11533577 DOI: 10.1085/jgp.202413622] [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] [Indexed: 11/03/2024] Open
Abstract
Decades of research into striated muscle have provided a robust understanding of the structure and function of the sarcomere and its protein constituents. However, a handful of sarcomere proteins remain that have had little to no functional characterization. These are typically proteins that are highly muscle-type specific or are products of alternative start sites or alternative splicing.
Collapse
Affiliation(s)
- David Y. Barefield
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
105
|
Matakatsu H, Fehon RG. Dachsous and Fat coordinately repress the Dachs-Dlish-Approximated complex to control growth. J Cell Biol 2024; 223:e202406119. [PMID: 39373700 PMCID: PMC11461286 DOI: 10.1083/jcb.202406119] [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: 06/19/2024] [Revised: 08/28/2024] [Accepted: 09/11/2024] [Indexed: 10/08/2024] Open
Abstract
Two protocadherins, Dachsous and Fat, regulate organ growth in Drosophila via the Hippo pathway. Dachsous and Fat bind heterotypically to regulate the abundance and subcellular localization of a "core complex" consisting of Dachs, Dlish, and Approximated. This complex localizes to the junctional cortex where it represses Warts. Dachsous is believed to promote growth by recruiting and stabilizing this complex, while Fat represses growth by promoting its degradation. Here, we examine the functional relationships between the intracellular domains of Dachsous and Fat and the core complex. While Dachsous promotes the accumulation of core complex proteins in puncta, it is not required for their assembly. Indeed, the core complex accumulates maximally in the absence of both Dachsous and Fat. Furthermore, Dachsous represses growth in the absence of Fat by removing the core complex from the junctional cortex. Fat similarly recruits core complex components but promotes their degradation. Our findings reveal that Dachsous and Fat coordinately constrain tissue growth by repressing the core complex.
Collapse
Affiliation(s)
- Hitoshi Matakatsu
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Richard G. Fehon
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
106
|
Sho T, Li Y, Jiao H, Yu L. Migratory autolysosome disposal mitigates lysosome damage. J Cell Biol 2024; 223:e202403195. [PMID: 39347717 PMCID: PMC11457477 DOI: 10.1083/jcb.202403195] [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: 03/30/2024] [Revised: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 10/01/2024] Open
Abstract
Lysosomes, essential for intracellular degradation and recycling, employ damage-control strategies such as lysophagy and membrane repair mechanisms to maintain functionality and cellular homeostasis. Our study unveils migratory autolysosome disposal (MAD), a response to lysosomal damage where cells expel LAMP1-LC3 positive structures via autolysosome exocytosis, requiring autophagy machinery, SNARE proteins, and cell migration. This mechanism, crucial for mitigating lysosomal damage, underscores the role of cell migration in lysosome damage control and facilitates the release of small extracellular vesicles, highlighting the intricate relationship between cell migration, organelle quality control, and extracellular vesicle release.
Collapse
Affiliation(s)
- Takami Sho
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Li
- Cryo-EM Facility of Tsinghua University, Branch of National Protein Science Facility, Tsinghua University, Beijing, China
| | - Haifeng Jiao
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Li Yu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
107
|
Mead AF, Wood NB, Nelson SR, Palmer BM, Yang L, Previs SB, Ploysangngam A, Kennedy GG, McAdow JF, Tremble SM, Zimmermann MA, Cipolla MJ, Ebert AM, Johnson AN, Gurnett CA, Previs MJ, Warshaw DM. Functional role of myosin-binding protein H in thick filaments of developing vertebrate fast-twitch skeletal muscle. J Gen Physiol 2024; 156:e202413604. [PMID: 39373654 PMCID: PMC11461142 DOI: 10.1085/jgp.202413604] [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: 05/14/2024] [Revised: 08/01/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024] Open
Abstract
Myosin-binding protein H (MyBP-H) is a component of the vertebrate skeletal muscle sarcomere with sequence and domain homology to myosin-binding protein C (MyBP-C). Whereas skeletal muscle isoforms of MyBP-C (fMyBP-C, sMyBP-C) modulate muscle contractility via interactions with actin thin filaments and myosin motors within the muscle sarcomere "C-zone," MyBP-H has no known function. This is in part due to MyBP-H having limited expression in adult fast-twitch muscle and no known involvement in muscle disease. Quantitative proteomics reported here reveal that MyBP-H is highly expressed in prenatal rat fast-twitch muscles and larval zebrafish, suggesting a conserved role in muscle development and prompting studies to define its function. We take advantage of the genetic control of the zebrafish model and a combination of structural, functional, and biophysical techniques to interrogate the role of MyBP-H. Transgenic, FLAG-tagged MyBP-H or fMyBP-C both localize to the C-zones in larval myofibers, whereas genetic depletion of endogenous MyBP-H or fMyBP-C leads to increased accumulation of the other, suggesting competition for C-zone binding sites. Does MyBP-H modulate contractility in the C-zone? Globular domains critical to MyBP-C's modulatory functions are absent from MyBP-H, suggesting that MyBP-H may be functionally silent. However, our results suggest an active role. In vitro motility experiments indicate MyBP-H shares MyBP-C's capacity as a molecular "brake." These results provide new insights and raise questions about the role of the C-zone during muscle development.
Collapse
Affiliation(s)
- Andrew F. Mead
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Neil B. Wood
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Shane R. Nelson
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Bradley M. Palmer
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Lin Yang
- National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY, USA
| | - Samantha Beck Previs
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Angela Ploysangngam
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Guy G. Kennedy
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Jennifer F. McAdow
- Department of Neurlogical Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Sarah M. Tremble
- Department of Electrical and Biomedical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT, USA
| | - Marcus A. Zimmermann
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Marilyn J. Cipolla
- Department of Electrical and Biomedical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT, USA
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Alicia M. Ebert
- Department of Biology, College of Arts and Sciences, University of Vermont, Burlington, VT, USA
| | - Aaron N. Johnson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Christina A. Gurnett
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Michael J. Previs
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - David M. Warshaw
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| |
Collapse
|
108
|
de Leeuw NF, Budhathoki R, Russell LJ, Loerke D, Blankenship JT. Nuclei as mechanical bumpers during epithelial remodeling. J Cell Biol 2024; 223:e202405078. [PMID: 39325019 PMCID: PMC11450824 DOI: 10.1083/jcb.202405078] [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: 05/13/2024] [Revised: 08/21/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024] Open
Abstract
The morphogenesis of developing tissues relies on extensive cellular rearrangements in shape, position, and identity. A key process in reshaping tissues is cell intercalation-driven elongation, where epithelial cells align and intercalate along a common axis. Typically, analyses focus on how peripheral cortical forces influence cell shape changes. Less attention is given to how inhomogeneities in internal structures, particularly the nucleus, impact cell shaping. Here, we examine how pulsed contractile and extension dynamics interact with the nucleus in elongating Drosophila embryos. Our data show that tightly packed nuclei in apical layers hinder tissue remodeling/oscillatory behaviors. We identify two mechanisms for resolving internuclear tensions: nuclear deformation and dispersion. Embryos with non-deformable nuclei use nuclear dispersion to maintain near-normal extensile rates, while those with non-dispersible nuclei due to microtubule inhibition exhibit disruptions in contractile behaviors. Disrupting both mechanisms leads to severe tissue extension defects and cell extrusion. These findings highlight the critical role of nuclear shape and positioning in topological remodeling of epithelia.
Collapse
Affiliation(s)
- Noah F. de Leeuw
- Department of Physics and Astronomy, University of Denver, Denver, CO, USA
| | - Rashmi Budhathoki
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Liam J. Russell
- Department of Physics and Astronomy, University of Denver, Denver, CO, USA
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Dinah Loerke
- Department of Physics and Astronomy, University of Denver, Denver, CO, USA
| | | |
Collapse
|
109
|
Smith M, Gay L, Babst M. ER-plasma membrane contact sites deliver ER lipids and proteins for rapid cell surface expansion. J Cell Biol 2024; 223:e202308137. [PMID: 39302311 DOI: 10.1083/jcb.202308137] [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: 08/28/2023] [Revised: 07/16/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
As a consequence of hypoosmotic shock, yeast cells swell rapidly and increase the surface area by ∼20% in 20 s. Approximately, 35% of this surface increase is mediated by the ER-plasma membrane contact sites, specifically the tricalbins, which are required for the delivery of both lipids and the GPI-anchored protein Crh2 from the cortical ER to the plasma membrane. Therefore, we propose a new function for the tricalbins: mediating the fusion of the ER to the plasma membrane at contact sites. This proposed fusion is triggered by calcium influx via the stretch-gated channel Cch1 and is supported by the anoctamin Ist2.
Collapse
Affiliation(s)
- Madison Smith
- Henry Eyring Center for Cell and Genome Science, University of Utah , Salt Lake City, UT, USA
| | - Lincoln Gay
- Henry Eyring Center for Cell and Genome Science, University of Utah , Salt Lake City, UT, USA
| | - Markus Babst
- Henry Eyring Center for Cell and Genome Science, University of Utah , Salt Lake City, UT, USA
| |
Collapse
|
110
|
Dias Araújo AR, Bello AA, Bigay J, Franckhauser C, Gautier R, Cazareth J, Kovács D, Brau F, Fuggetta N, Čopič A, Antonny B. Surface tension-driven sorting of human perilipins on lipid droplets. J Cell Biol 2024; 223:e202403064. [PMID: 39297796 PMCID: PMC11413419 DOI: 10.1083/jcb.202403064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024] Open
Abstract
Perilipins (PLINs), the most abundant proteins on lipid droplets (LDs), display similar domain organization including amphipathic helices (AH). However, the five human PLINs bind different LDs, suggesting different modes of interaction. We established a minimal system whereby artificial LDs covered with defined polar lipids were transiently deformed to promote surface tension. Binding of purified PLIN3 and PLIN4 AH was strongly facilitated by tension but was poorly sensitive to phospholipid composition and to the presence of diacylglycerol. Accordingly, LD coverage by PLIN3 increased as phospholipid coverage decreased. In contrast, PLIN1 bound readily to LDs fully covered by phospholipids; PLIN2 showed an intermediate behavior between PLIN1 and PLIN3. In human adipocytes, PLIN3/4 were found in a soluble pool and relocated to LDs upon stimulation of fast triglyceride synthesis, whereas PLIN1 and PLIN2 localized to pre-existing LDs, consistent with the large difference in LD avidity observed in vitro. We conclude that the PLIN repertoire is adapted to handling LDs with different surface properties.
Collapse
Affiliation(s)
- Ana Rita Dias Araújo
- Université Côte d’Azur, CNRS and Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Sophia Antipolis, France
| | - Abdoul Akim Bello
- Université Côte d’Azur, CNRS and Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Sophia Antipolis, France
| | - Joëlle Bigay
- Université Côte d’Azur, CNRS and Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Sophia Antipolis, France
| | - Céline Franckhauser
- Centre de Recherche en Biologie Cellulaire de Montpellier-CRBM, Université de Montpellier, CNRS, UMR 5237, Montpellier, France
| | - Romain Gautier
- Université Côte d’Azur, CNRS and Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Sophia Antipolis, France
| | - Julie Cazareth
- Université Côte d’Azur, CNRS and Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Sophia Antipolis, France
| | - Dávid Kovács
- Université Côte d’Azur, CNRS and Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Sophia Antipolis, France
| | - Frédéric Brau
- Université Côte d’Azur, CNRS and Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Sophia Antipolis, France
| | - Nicolas Fuggetta
- Centre de Recherche en Biologie Cellulaire de Montpellier-CRBM, Université de Montpellier, CNRS, UMR 5237, Montpellier, France
| | - Alenka Čopič
- Centre de Recherche en Biologie Cellulaire de Montpellier-CRBM, Université de Montpellier, CNRS, UMR 5237, Montpellier, France
| | - Bruno Antonny
- Université Côte d’Azur, CNRS and Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Sophia Antipolis, France
| |
Collapse
|
111
|
Mercado-Perez A, Hernandez JP, Fedyshyn Y, Treichel AJ, Joshi V, Kossick K, Betageri KR, Farrugia G, Druliner B, Beyder A. Piezo2 interacts with E-cadherin in specialized gastrointestinal epithelial mechanoreceptors. J Gen Physiol 2024; 156:e202213324. [PMID: 39495178 PMCID: PMC11536063 DOI: 10.1085/jgp.202213324] [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: 12/22/2022] [Revised: 01/20/2024] [Accepted: 09/19/2024] [Indexed: 11/05/2024] Open
Abstract
Piezo2 is a mechanically gated ion channel most commonly expressed by specialized mechanoreceptors, such as the enteroendocrine cells (EECs) of the gastrointestinal epithelium. A subpopulation of EECs expresses Piezo2 and functionally resembles the skin's touch sensors, called Merkel cells. Low-magnitude mechanical stimuli delivered to the mucosal layer are primarily sensed by mechanosensitive EECs in a process we term "gut touch." Piezo2 transduces cellular forces into ionic currents, a process that is sensitive to bilayer tension and cytoskeletal depolymerization. E-cadherin is a widely expressed protein that mediates cell-cell adhesion in epithelia and interacts with scaffold proteins that anchor it to actin fibers. E-cadherin was shown to interact with Piezo2 in immortalized cell models. We hypothesized that the Piezo2-E-cadherin interaction is important for EEC mechanosensitivity. To test this, we used super-resolution imaging, co-immunoprecipitation, and functional assays in primary tissues from mice and gut organoids. In tissue EECs and intestinal organoids, we observed multiple Piezo2 cellular pools, including one that overlaps with actin and E-cadherin at the cells' lateral walls. Further, E-cadherin co-immunoprecipitated with Piezo2 in the primary colonic epithelium. We found that E-cadherin knockdown decreases mechanosensitive calcium responses in mechanically stimulated primary EECs. In all, our results demonstrate that Piezo2 localizes to the lateral wall of EECs, where it physically interacts with E-cadherin and actin. They suggest that the Piezo2-E-cadherin-actin interaction is important for mechanosensitivity in the gut epithelium and possibly in tissues where E-cadherin and Piezo2 are co-expressed in epithelial mechanoreceptors, such as skin, lung, and bladder.
Collapse
Affiliation(s)
- Arnaldo Mercado-Perez
- Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, MN, USA
- Medical Scientist Training Program (MSTP), Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Jeric P. Hernandez
- Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yaroslav Fedyshyn
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | | | - Vikram Joshi
- Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kimberlee Kossick
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kalpana R. Betageri
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Gianrico Farrugia
- Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Brooke Druliner
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Arthur Beyder
- Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
112
|
Song Y, Lan Y, Li K, Qiao D, Cao Y, Xu H. Regulation of a novel DsGATA1 from Dunaliella salina on the synthesis of carotenoids under red light. Appl Microbiol Biotechnol 2024; 108:82. [PMID: 38189955 DOI: 10.1007/s00253-023-12894-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/31/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024]
Abstract
Dunaliella salina is a high-quality industrial effector for carotenoid production. The mechanism by which red light regulates carotenoid synthesis is still unclear. In this study, a transcription factor of DsGATA1 with a distinct structure was discovered in D. salina. The recognition motif of DsGATA1 was comparable to that of plant and fungal GATA, despite its evolutionary proximity to animal-derived GATA. The expression of DsGATA1 in D. salina was still noticeably decreased when exposed to red light. Analysis of physiological and biochemical transcriptomic data from overexpressed, interfering, and wild-type strains of DsGATA1 revealed that DsGATA1 acts as a global regulator of D. salina carotenoid synthesis. The upregulated genes in the CBP pathway by DsGATA1 were involved in its regulation of the synthesis of carotenoids. DsGATA1 also enhanced carotenoid accumulation under red light by affecting N metabolism. DsGATA1 was found to directly bind to the promoter of nitrate reductase to activate its expression, promoting D. salina nitrate uptake and accelerating biomass accumulation. DsGATA1 affected the expression of the genes encoding GOGAT, GDH, and ammonia transporter proteins. Moreover, our study revealed that the regulation of N metabolism by DsGATA1 led to the production of NO molecules that inhibited carotenoid synthesis. However, DsGATA1 significantly enhanced carotenoid synthesis by NO scavenger removal of NO. The D. salina carotenoid accumulation under red light was elevated by 46% in the presence of overexpression of DsGATA1 and NO scavenger. Nevertheless, our results indicated that DsGATA1 could be an important target for engineering carotenoid production. KEY POINTS: • DsGATA1 with a distinct structure and recognition motif was found in D. salina • DsGATA1 enhanced carotenoid production and biomass in D. salina under red light • DsGATA1 is involved in the regulation of N metabolism and carotenoid synthesis.
Collapse
Affiliation(s)
- Yao Song
- Microbiology and Metabolic Engineering Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China
| | - Yanhong Lan
- Microbiology and Metabolic Engineering Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China
| | - Ke Li
- Microbiology and Metabolic Engineering Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China
| | - Dairong Qiao
- Microbiology and Metabolic Engineering Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China
| | - Yi Cao
- Microbiology and Metabolic Engineering Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China
| | - Hui Xu
- Microbiology and Metabolic Engineering Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China.
| |
Collapse
|
113
|
Wang C, Chen Z, Copenhaver GP, Wang Y. Heterochromatin in plant meiosis. Nucleus 2024; 15:2328719. [PMID: 38488152 PMCID: PMC10950279 DOI: 10.1080/19491034.2024.2328719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
Heterochromatin is an organizational property of eukaryotic chromosomes, characterized by extensive DNA and histone modifications, that is associated with the silencing of transposable elements and repetitive sequences. Maintaining heterochromatin is crucial for ensuring genomic integrity and stability during the cell cycle. During meiosis, heterochromatin is important for homologous chromosome synapsis, recombination, and segregation, but our understanding of meiotic heterochromatin formation and condensation is limited. In this review, we focus on the dynamics and features of heterochromatin and how it condenses during meiosis in plants. We also discuss how meiotic heterochromatin influences the interaction and recombination of homologous chromosomes during prophase I.
Collapse
Affiliation(s)
- Cong Wang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China
| | - Zhiyu Chen
- State Key Laboratory of Genetic Engineering, Institute of Plant Biology, School of Life Sciences, Fudan University, Shanghai, China
| | - Gregory P. Copenhaver
- Department of Biology and the Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Yingxiang Wang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China
- State Key Laboratory of Genetic Engineering, Institute of Plant Biology, School of Life Sciences, Fudan University, Shanghai, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
114
|
Fernandez MK, Sinha M, Zidan M, Renz M. Nuclear actin filaments - a historical perspective. Nucleus 2024; 15:2320656. [PMID: 38384139 PMCID: PMC10885181 DOI: 10.1080/19491034.2024.2320656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/14/2024] [Indexed: 02/23/2024] Open
Abstract
The view on nuclear filaments formed by non-skeletal β-actin has significantly changed over the decades. Initially, filamentous actin was observed in amphibian oocyte nuclei and only under specific cell stress conditions in mammalian cell nuclei. Improved labeling and imaging technologies have permitted insights into a transient but microscopically apparent filament network that is relevant for chromatin organization, biomechanics of the mammalian cell nucleus, gene expression, and DNA damage repair. Here, we will provide a historical perspective on the developing insight into nuclear actin filaments.
Collapse
Affiliation(s)
| | - Molika Sinha
- Gynecologic Oncology Division, School of Medicine Stanford University, Palo Alto, CA, USA
| | - Mia Zidan
- Gynecologic Oncology Division, School of Medicine Stanford University, Palo Alto, CA, USA
| | - Malte Renz
- Gynecologic Oncology Division, School of Medicine Stanford University, Palo Alto, CA, USA
| |
Collapse
|
115
|
Mizoguchi T. In vivo dynamics of hard tissue-forming cell origins: Insights from Cre/loxP-based cell lineage tracing studies. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:109-119. [PMID: 38406212 PMCID: PMC10885318 DOI: 10.1016/j.jdsr.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
Bone tissue provides structural support for our bodies, with the inner bone marrow (BM) acting as a hematopoietic organ. Within the BM tissue, two types of stem cells play crucial roles: mesenchymal stem cells (MSCs) (or skeletal stem cells) and hematopoietic stem cells (HSCs). These stem cells are intricately connected, where BM-MSCs give rise to bone-forming osteoblasts and serve as essential components in the BM microenvironment for sustaining HSCs. Despite the mid-20th century proposal of BM-MSCs, their in vivo identification remained elusive owing to a lack of tools for analyzing stemness, specifically self-renewal and multipotency. To address this challenge, Cre/loxP-based cell lineage tracing analyses are being employed. This technology facilitated the in vivo labeling of specific cells, enabling the tracking of their lineage, determining their stemness, and providing a deeper understanding of the in vivo dynamics governing stem cell populations responsible for maintaining hard tissues. This review delves into cell lineage tracing studies conducted using commonly employed genetically modified mice expressing Cre under the influence of LepR, Gli1, and Axin2 genes. These studies focus on research fields spanning long bones and oral/maxillofacial hard tissues, offering insights into the in vivo dynamics of stem cell populations crucial for hard tissue homeostasis.
Collapse
|
116
|
Yang Y, Chen Q, Fan S, Lu Y, Huang Q, Liu X, Peng X. Glutamine sustains energy metabolism and alleviates liver injury in burn sepsis by promoting the assembly of mitochondrial HSP60-HSP10 complex via SIRT4 dependent protein deacetylation. Redox Rep 2024; 29:2312320. [PMID: 38329114 PMCID: PMC10854458 DOI: 10.1080/13510002.2024.2312320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Burns and burn sepsis, characterized by persistent and profound hypercatabolism, cause energy metabolism dysfunction that worsens organ injury and systemic disorders. Glutamine (Gln) is a key nutrient that remarkably replenishes energy metabolism in burn and sepsis patients, but its exact roles beyond substrate supply is unclear. In this study, we demonstrated that Gln alleviated liver injury by sustaining energy supply and restoring redox balance. Meanwhile, Gln also rescued the dysfunctional mitochondrial electron transport chain (ETC) complexes, improved ATP production, reduced oxidative stress, and protected hepatocytes from burn sepsis injury. Mechanistically, we revealed that Gln could activate SIRT4 by upregulating its protein synthesis and increasing the level of Nicotinamide adenine dinucleotide (NAD+), a co-enzyme that sustains the activity of SIRT4. This, in turn, reduced the acetylation of shock protein (HSP) 60 to facilitate the assembly of the HSP60-HSP10 complex, which maintains the activity of ETC complex II and III and thus sustain ATP generation and reduce reactive oxygen species release. Overall, our study uncovers a previously unknown pharmacological mechanism involving the regulation of HSP60-HSP10 assembly by which Gln recovers mitochondrial complex activity, sustains cellular energy metabolism and exerts a hepato-protective role in burn sepsis.
Collapse
Affiliation(s)
- Yongjun Yang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People’s Republic of China
| | - Qian Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People’s Republic of China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People’s Republic of China
| | - Yongling Lu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People’s Republic of China
| | - Qianyin Huang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People’s Republic of China
| | - Xin Liu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People’s Republic of China
| | - Xi Peng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People’s Republic of China
- State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), ChongqingPeople’s Republic of China
| |
Collapse
|
117
|
Lv L, Yang C, Zhang X, Chen T, Luo M, Yu G, Chen Q. Autophagy-related protein PlATG2 regulates the vegetative growth, sporangial cleavage, autophagosome formation, and pathogenicity of peronophythora litchii. Virulence 2024; 15:2322183. [PMID: 38438325 PMCID: PMC10913709 DOI: 10.1080/21505594.2024.2322183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/18/2024] [Indexed: 03/06/2024] Open
Abstract
Autophagy is an intracellular degradation process that is important for the development and pathogenicity of phytopathogenic fungi and for the defence response of plants. However, the molecular mechanisms underlying autophagy in the pathogenicity of the plant pathogenic oomycete Peronophythora litchii, the causal agent of litchi downy blight, have not been well characterized. In this study, the autophagy-related protein ATG2 homolog, PlATG2, was identified and characterized using a CRISPR/Cas9-mediated gene replacement strategy in P. litchii. A monodansylcadaverine (MDC) staining assay indicated that deletion of PlATG2 abolished autophagosome formation. Infection assays demonstrated that ΔPlatg2 mutants showed significantly impaired pathogenicity in litchi leaves and fruits. Further studies have revealed that PlATG2 participates in radial growth and asexual/sexual development of P. litchii. Moreover, zoospore release and cytoplasmic cleavage of sporangia were considerably lower in the ΔPlatg2 mutants than in the wild-type strain by FM4-64 staining. Taken together, our results revealed that PlATG2 plays a pivotal role in vegetative growth, sporangia and oospore production, zoospore release, sporangial cleavage, and plant infection of P. litchii. This study advances our understanding of the pathogenicity mechanisms of the phytopathogenic oomycete P. litchii and is conducive to the development of effective control strategies.
Collapse
Affiliation(s)
- Lin Lv
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Chengdong Yang
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Xue Zhang
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Taixu Chen
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Manfei Luo
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Ge Yu
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Qinghe Chen
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| |
Collapse
|
118
|
Hu XM, Zheng S, Zhang Q, Wan X, Li J, Mao R, Yang R, Xiong K. PANoptosis signaling enables broad immune response in psoriasis: From pathogenesis to new therapeutic strategies. Comput Struct Biotechnol J 2024; 23:64-76. [PMID: 38125299 PMCID: PMC10730955 DOI: 10.1016/j.csbj.2023.11.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Background Accumulating evidence suggests that regulated cell death, such as pyroptosis, apoptosis, and necroptosis, is deeply involved in the pathogenesis of psoriasis. As a newly recognized form of systematic cell death, PANoptosis is involved in a variety of inflammatory disorders through amplifying inflammatory and immune cascades, but its role in psoriasis remains elusive. Objectives To reveal the role of PANoptosis in psoriasis for a potential therapeutic strategy. Methods Multitranscriptomic analysis and experimental validation were used to identify PANoptosis signaling in psoriasis. RNA-seq and scRNA-seq analyses were performed to establish a PANoptosis-mediated immune response in psoriasis, which revealed hub genes through WGCNA and predicted disulfiram as a potential drug. The effect and mechanism of disulfiram were verified in imiquimod (IMQ)-induced psoriasis. Results Here, we found a highlighted PANoptosis signature in psoriasis patients through multitranscriptomic analysis and experimental validation. Based on this, two distinct PANoptosis patterns (non/high) were identified, which were the options for clinical classification. The high-PANoptosis-related group had a higher response rate to immune cell infiltration (such as M1 macrophages and keratinocytes). Subsequently, WGCNA showed the hub genes (e.g., S100A12, CYCS, NOD2, STAT1, HSPA4, AIM2, MAPK7), which were significantly associated with clinical phenotype, PANoptosis signature, and identified immune response in psoriasis. Finally, we explored disulfiram (DSF) as a candidate drug for psoriasis through network pharmacology, which ameliorated IMQ-mediated psoriatic symptoms through antipyroptosis-mediated inflammation and enhanced apoptotic progression. By analyzing the specific ligand-receptor interaction pairs within and between cell lineages, we speculated that DSF might exert its effects by targeting keratinocytes directly or targeting M1 macrophages to downregulate the proliferation of keratinocytes. Conclusions PANoptosis with its mediated immune cell infiltration provides a roadmap for research on the pathogenesis and therapeutic strategies of psoriasis.
Collapse
Affiliation(s)
- Xi-min Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Shengyuan Zheng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Xinxing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rui Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510000, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
119
|
Kim AH, Lee HM, Kim HS, Jeong SW, Jun JK, Jang J. CRISPR/Cas9-mediated knock-in of a fluorescent reporter into the target locus of interest in human pluripotent stem cells. MethodsX 2024; 13:102807. [PMID: 39036607 PMCID: PMC11259911 DOI: 10.1016/j.mex.2024.102807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/14/2024] [Indexed: 07/23/2024] Open
Abstract
The method presented herein is associated with the Lab Resource article titled "Generation of αMHC-EGFP knock-in in human pluripotent stem cell line, SNUe003-A-3, using CRISPR/CAS9-based gene targeting" [1]. The cardiac muscle-specific protein, α-myosin heavy chain (αMHC), is encoded by the human MYH6 gene, which is expressed in both the atria and ventricles during embryonic development and is predominantly expressed in the atria after birth [2]. Herein, the methods used to achieve CRISPR/SpCas9-mediated introduction of an EGFP reporter into αMHC, the target locus in human pluripotent stem cells (hPSCs) for cardiac lineage tracing and clinical cell sorting are described. The CRISPR-Cas9 system enables efficient replacement of the stop codon in the last exon of αMHC with a 2A non-joining peptide (T2A)-EGFP cassette. First, hPSCs are transfected with the donor construct and Cas9/sgRNA plasmids via electroporation and selected with neomycin for approximately 3 weeks. Thereafter, the established cell line exhibits typical characteristics of human embryonic stem cells (hESCs). When these cells differentiate into cardiomyocytes, the expression of EGFP is confirmed using confocal microscopy, flow cytometry analysis, and immunostaining.•The line enables monitoring of cell maturation events during human cardiac development.•The line is a valuable platform for cardiotoxicity tests and drug screening.•This method has already been employed in two original studies, as previously reported for reporter cell line generation using CRISPR/Cas9.
Collapse
Affiliation(s)
- A-Hyeon Kim
- Department of Biomedical Sciences, College of Medical Convergence, Catholic Kwandong University, Gangneung-si, Gangwon-do, Republic of Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Dept. of Obstetrics & Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ha Myoung Lee
- Department of Biomedical Sciences, College of Medical Convergence, Catholic Kwandong University, Gangneung-si, Gangwon-do, Republic of Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Dept. of Obstetrics & Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hong-Sik Kim
- Department of Biomedical Sciences, College of Medical Convergence, Catholic Kwandong University, Gangneung-si, Gangwon-do, Republic of Korea
| | - Seong Woo Jeong
- Department of Biomedical Sciences, College of Medical Convergence, Catholic Kwandong University, Gangneung-si, Gangwon-do, Republic of Korea
| | - Jong Kwan Jun
- Institute of Reproductive Medicine and Population, Medical Research Center, Dept. of Obstetrics & Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jiho Jang
- Department of Biomedical Sciences, College of Medical Convergence, Catholic Kwandong University, Gangneung-si, Gangwon-do, Republic of Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Dept. of Obstetrics & Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| |
Collapse
|
120
|
Li Y, Yang X, Shi C, Wang L, Wang Y, Zhang W, Wang P, Zhang H, Yang X, Wen P. Insights into the microscopic heterogeneity of whey proteins between yak colostrum and mature milk based on 4D lable-free quantitative phosphoproteomics. Food Chem 2024; 460:140679. [PMID: 39106750 DOI: 10.1016/j.foodchem.2024.140679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/09/2024]
Abstract
This study aimed to reveal the change patterns of the phosphorylation modification status of yak whey phosphoproteins during lactation and their physiological effects. Herein, we comprehensively characterized whey phosphoproteome in yak colostrum and mature milk using an ultra-high throughput phosphoproteomics approach incorporating trapped ion mobility technology. A total of 344 phosphorylation sites from 206 phosphoproteins were identified, with individual site modification predominating. Notably, 117 significantly different phosphorylation sites were distributed on 89 whey phosphoproteins. Gene ontology analysis indicated that these significantly different whey phosphoproteins (SDWPPs) were mainly annotated to carbohydrate metabolic process, membrane, extracellular region and calcium ion binding. Metabolic pathway enrichment analysis demonstrated that SDWPPs were critically involved in protein processing in endoplasmic reticulum, NOD-like receptor signaling pathway and N-glycan biosynthesis. Our results elucidate the phosphorylation profiles of yak whey phosphoproteins at different lactations and their adaptive regulatory role in meeting the nutritional requirements of yak calves during development.
Collapse
Affiliation(s)
- Yiheng Li
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Xue Yang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Chengrui Shi
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Longlin Wang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Ying Wang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Weibing Zhang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Pengjie Wang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Hao Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Xiaoli Yang
- Gansu Institute of Business and Technology, Lanzhou 730010, China.
| | - Pengcheng Wen
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China.
| |
Collapse
|
121
|
Nakajima D, Takahashi N, Inoue T, Nomura SIM, Matsubayashi HT. A unified purification method for actin-binding proteins using a TEV-cleavable His-Strep-tag. MethodsX 2024; 13:102884. [PMID: 39224451 PMCID: PMC11367271 DOI: 10.1016/j.mex.2024.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The actin cytoskeleton governs the dynamic functions of cells, ranging from motility to phagocytosis and cell division. To elucidate the molecular mechanism, in vitro reconstructions of the actin cytoskeleton and its force generation process have played essential roles, highlighting the importance of efficient purification methods for actin-binding proteins. In this study, we introduce a unified purification method for actin-binding proteins, including capping protein (CP), cofilin, ADF, profilin, fascin, and VASP, key regulators in force generation of the actin cytoskeleton. Exploiting a His-Strep-tag combined with a TEV protease cleavage site, we purified these diverse actin-binding proteins through a simple two-column purification process: initial purification through a Strep-Tactin column and subsequent tag removal through the reverse purification by a Ni-NTA column. Biochemical and microscopic assays validated the functionality of the purified proteins, demonstrating the versatility of the approach. Our methods not only delineate critical steps for the efficient preparation of actin-binding proteins but also hold the potential to advance investigations of mutants, isoforms, various source species, and engineered proteins involved in actin cytoskeletal dynamics.•Unified purification method for various actin-binding proteins.•His-Strep-tag and TEV protease cleavage for efficient purification.•Functional validation through biochemical and microscopic assays.
Collapse
Affiliation(s)
- Daichi Nakajima
- Molecular Robotics Laboratory, Department of Robotics, Graduate School of Engineering, Tohoku University, Aoba 6-6-01 Aramaki Aoba-ku, Mechanical Eng. Research Bldg. 2 (A 03), Sendai, Miyagi, 980-8579, Japan
| | - Nozomi Takahashi
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aoba 6-3 Aramaki Aoba-ku, Research Bldg. (G 06), Sendai, Miyagi, 980-8579, Japan
| | - Takanari Inoue
- Department of Cell Biology, Johns Hopkins University School of Medicine, 855 N. Wolfe St. 476 Rangos Building, Baltimore, MD, 21205, USA
| | - Shin-ichiro M. Nomura
- Molecular Robotics Laboratory, Department of Robotics, Graduate School of Engineering, Tohoku University, Aoba 6-6-01 Aramaki Aoba-ku, Mechanical Eng. Research Bldg. 2 (A 03), Sendai, Miyagi, 980-8579, Japan
| | - Hideaki T. Matsubayashi
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aoba 6-3 Aramaki Aoba-ku, Research Bldg. (G 06), Sendai, Miyagi, 980-8579, Japan
| |
Collapse
|
122
|
VijayKumar S, Borja M, Neff N, Royer LA, Lange M. Maximizing single cell dissociation protocol for individual zebrafish embryo. MethodsX 2024; 13:102958. [PMID: 39329153 PMCID: PMC11426156 DOI: 10.1016/j.mex.2024.102958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Single-cell sequencing has revolutionized our understanding of cellular heterogeneity and cell state, enabling investigations across diverse fields such as developmental biology, immunology, and cancer biology. However, obtaining a high-quality single-cell suspension is still challenging, particularly when starting with limited materials like Zebrafish embryos, a powerful animal model for studying developmental processes and human diseases. Here, we present an optimized protocol for isolating single cells from individual zebrafish embryos, offering a valuable resource for researchers interested in working with limited starting material. The protocol facilitates unique investigations utilizing individual embryos, such as inter-individual genetic differences and embryo-specific lineage tracing analysis. Using a refined single-cell isolation protocol alongside zebrafish as a model organism, researchers can access a resource for exploring the emergence of all types and states of cells, advancing our understanding of cellular processes and disease mechanisms.
Collapse
Affiliation(s)
| | | | - Norma Neff
- Chan Zuckerberg Biohub, San Francisco, USA
| | | | | |
Collapse
|
123
|
Yao M, Zhang L, Teng X, Lei Y, Xing X, Ren T, Pan Y, Zhang L, Li Z, Lin J, Zheng Y, Xing L, Zhou J, Wu C. Transcriptomic profiling of Dip2a in the neural differentiation of mouse embryonic stem cells. Comput Struct Biotechnol J 2024; 23:700-710. [PMID: 38292475 PMCID: PMC10825174 DOI: 10.1016/j.csbj.2023.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction The disconnected-interacting protein 2 homolog A (DIP2A), a member of disconnected-interacting 2 protein family, has been shown to be involved in human nervous system-related mental illness. This protein is highly expressed in the nervous system of mouse. Mutation of mouse DIP2A causes defects in spine morphology and synaptic transmission, autism-like behaviors, and defective social novelty [5], [27], indicating that DIP2A is critical to the maintenance of neural development. However, the role of DIP2A in neural differentiation has yet to be investigated. Objective To determine the role of DIP2A in neural differentiation, a neural differentiation model was established using mouse embryonic stem cells (mESCs) and studied by using gene-knockout technology and RNA-sequencing-based transcriptome analysis. Results We found that DIP2A is not required for mESCs pluripotency maintenance, but loss of DIP2A causes the neural differentiation abnormalities in both N2B27 and KSR medium. Functional knockout of Dip2a gene also decreased proliferation of mESCs by perturbation of the cell cycle and profoundly inhibited the expression of a large number of neural development-associated genes which mainly enriched in spinal cord development and postsynapse assembly. Conclusions The results of this report demonstrate that DIP2A plays an essential role in regulating differentiation of mESCs towards the neural fate.
Collapse
Affiliation(s)
- Mingze Yao
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Lei Zhang
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
- Center of Reproductive Medicine, Children's Hospital of Shanxi and Women Health Center of Shanxi, Taiyuan 030006, China
| | - Xiaojuan Teng
- Dermatology Hospital, Southern Medical University, Guangzhou 510000, China
| | - Yu Lei
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Xiaoyu Xing
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Tinglin Ren
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Yuanqing Pan
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Liwen Zhang
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Zhengfeng Li
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510000, China
| | - Jingxia Lin
- Dermatology Hospital, Southern Medical University, Guangzhou 510000, China
| | - Yaowu Zheng
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Li Xing
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Jiajian Zhou
- Dermatology Hospital, Southern Medical University, Guangzhou 510000, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
124
|
Kong L, Yang J, Yang H, Xu B, Yang T, Liu W. Research advances on CaMKs-mediated neurodevelopmental injury. Arch Toxicol 2024; 98:3933-3947. [PMID: 39292234 DOI: 10.1007/s00204-024-03865-5] [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: 07/13/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Calcium/calmodulin-dependent protein kinases (CaMKs) are important proteins in the calcium signaling cascade response pathway, which can broadly regulate biological functions in vivo. Multifunctional CaMKs play key roles in neural development, including neuronal circuit building, synaptic plasticity establishment, and neurotrophic factor secretion. Currently, four familial proteins, calcium/calmodulin-dependent protein kinase I (CaMKI), calcium/calmodulin-dependent protein kinase II (CaMKII), eukaryotic elongation factor 2 kinase (eEF2K, popularly known as CaMKIII) and calcium/calmodulin-dependent protein kinase IV (CaMKIV), are thought to have been the most extensively studied during neurodevelopment. Although their spatial structures are extremely similar, as well as the initial starting point of activation, both require the activation of calcium and calmodulin (CaM) complexes to be involved in the process, and the phosphorylation sites and modes of each member are different. Furthermore, due to the high structural similarity of CaMKs, their members may play synergistic roles in the regulation of neural development, but different CaMKs also have their own means of regulating neural development. In this review, we first describe the visualized protein structural forms of CaMKI, CaMKII, eEF2K and CaMKIV, and then describe the functions of each kinase in neurodevelopment. After that, we focus on four main mechanisms of neurodevelopmental damage caused by CaMKs: CaMKI/ERK/CREB pathway inhibition leading to dendritic spine structural damage; Ca2+/CaM/CaMKII through induction of mitochondrial kinetic disorders leading to neurodevelopmental damage; CaMKIII/eEF2 hyperphosphorylation affects the establishment of synaptic plasticity; and CaMKIV/JNK/NF-κB through induction of an inflammatory response leading to neurodevelopmental damage. In conclusion, we briefly discuss the pathophysiological significance of aberrant CaMK family expression in neurodevelopmental disorders, as well as the protective effects of conventional CaMKII and CaMKIII antagonists against neurodevelopmental injury.
Collapse
Affiliation(s)
- Lingxu Kong
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Jing Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Huajie Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Bin Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Tianyao Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China.
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China.
| | - Wei Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China.
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
125
|
Ferraguto C, Piquemal-Lagoueillat M, Lemaire V, Moreau MM, Trazzi S, Uguagliati B, Ciani E, Bertrand SS, Louette E, Bontempi B, Pietropaolo S. Therapeutic efficacy of the BKCa channel opener chlorzoxazone in a mouse model of Fragile X syndrome. Neuropsychopharmacology 2024; 49:2032-2041. [PMID: 39223257 PMCID: PMC11480417 DOI: 10.1038/s41386-024-01956-6] [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: 03/14/2024] [Revised: 06/30/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Fragile X syndrome (FXS) is an X-linked neurodevelopmental disorder characterized by several behavioral abnormalities, including hyperactivity, anxiety, sensory hyper-responsiveness, and autistic-like symptoms such as social deficits. Despite considerable efforts, effective pharmacological treatments are still lacking, prompting the need for exploring the therapeutic value of existing drugs beyond their original approved use. One such repurposed drug is chlorzoxazone which is classified as a large-conductance calcium-dependent potassium (BKCa) channel opener. Reduced BKCa channel functionality has been reported in FXS patients, suggesting that molecules activating these channels could serve as promising treatments for this syndrome. Here, we sought to characterize the therapeutic potential of chlorzoxazone using the Fmr1-KO mouse model of FXS which recapitulates the main phenotypes of FXS, including BKCa channel alterations. Chlorzoxazone, administered either acutely or chronically, rescued hyperactivity and acoustic hyper-responsiveness as well as impaired social interactions exhibited by Fmr1-KO mice. Chlorzoxazone was more efficacious in alleviating these phenotypes than gaboxadol and metformin, two repurposed treatments for FXS that do not target BKCa channels. Systemic administration of chlorzoxazone modulated the neuronal activity-dependent gene c-fos in selected brain areas of Fmr1-KO mice, corrected aberrant hippocampal dendritic spines, and was able to rescue impaired BKCa currents recorded from hippocampal and cortical neurons of these mutants. Collectively, these findings provide further preclinical support for BKCa channels as a valuable therapeutic target for treating FXS and encourage the repurposing of chlorzoxazone for clinical applications in FXS and other related neurodevelopmental diseases.
Collapse
Affiliation(s)
| | | | - Valerie Lemaire
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France
| | - Maïté M Moreau
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Stefania Trazzi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Elisabetta Ciani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | | | | - Bruno Bontempi
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France
| | | |
Collapse
|
126
|
Wang L, Dai Z, Bi J, Chen Y, Wang Z, Sun Z, Ji Z, Wang H, Zhang Y, Wang L, Mao J, Yang J. Polydopamine-functionalized calcium-deficient hydroxyapatite 3D-printed scaffold with sustained doxorubicin release for synergistic chemo-photothermal therapy of osteosarcoma and accelerated bone regeneration. Mater Today Bio 2024; 29:101253. [PMID: 39399244 PMCID: PMC11470592 DOI: 10.1016/j.mtbio.2024.101253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/25/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
Interior bone-tissue regeneration and rapid tumor recurrence post-resection are critical challenges in osteosarcoma and other bone cancers. Conventional bone tissue engineering scaffolds lack inhibitory effects on bone tumor recurrence. Herein, multifunctional scaffolds (named DOX/PDA@CDHA) were designed through the spontaneous polymerization of Dopamine (PDA) on the surface of Calcium Deficient Hydroxyapatite (CDHA) scaffolds, followed by in situ loading of the chemotherapeutic drug Doxorubicin (DOX). The PDA coating endowed the scaffolds with significant photothermal properties, while the gradual release of DOX provided an effective chemotherapeutic effect. The on-demand release of DOX at tumor sites, triggered by dual stimulation (near-infrared (NIR) light and the acidic pH typical of tumor microenvironments), specifically targets cancer cells, thereby mitigating systemic side effects. These unique characteristics facilitated effective osteosarcoma eradication both in vitro and in vivo. Moreover, the scaffold's composition, which mimics the mineral phase of natural bone and is enhanced by PDA's biocompatibility, promotes critical osteogenic and angiogenic processes. This facilitates not only tumor eradication but also the regeneration of healthy bone tissue. Collectively, this study presents a potent candidate for the regeneration of bone defects induced by osteosarcoma.
Collapse
Affiliation(s)
- Lu Wang
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials (Ministry of Education), Shandong University, Jinan, 250061, PR China
- Schools of Materials Science and Engineering, Shandong University, Jinan, 250061, PR China
| | - Zihan Dai
- Department of Orthopedics, Qilu Hospital of Shandong University, #107 Wenhuaxi Road, Jinan, 250061, PR China
- Cheeloo College of Medicine, Shandong University, Jinan, 250061, PR China
| | - Jianqiang Bi
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials (Ministry of Education), Shandong University, Jinan, 250061, PR China
- Schools of Materials Science and Engineering, Shandong University, Jinan, 250061, PR China
| | - Yunzhen Chen
- Department of Orthopedics, Qilu Hospital of Shandong University, #107 Wenhuaxi Road, Jinan, 250061, PR China
| | - Ziyu Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, 100191, PR China
| | - Zhenqian Sun
- Department of Orthopedics, Qilu Hospital of Shandong University, #107 Wenhuaxi Road, Jinan, 250061, PR China
- Cheeloo College of Medicine, Shandong University, Jinan, 250061, PR China
| | - Zhongjie Ji
- Department of Orthopedics, Qilu Hospital of Shandong University, #107 Wenhuaxi Road, Jinan, 250061, PR China
- Cheeloo College of Medicine, Shandong University, Jinan, 250061, PR China
| | - Hongliang Wang
- Department of Orthopedics, Qilu Hospital of Shandong University, #107 Wenhuaxi Road, Jinan, 250061, PR China
- Cheeloo College of Medicine, Shandong University, Jinan, 250061, PR China
| | - Yan Zhang
- Advanced Medical Research Institute/Translational Medicine Core Facility of Advanced Medical Research Institute, Shandong University, PR China
| | - Limei Wang
- Advanced Medical Research Institute/Translational Medicine Core Facility of Advanced Medical Research Institute, Shandong University, PR China
| | - Junjie Mao
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials (Ministry of Education), Shandong University, Jinan, 250061, PR China
- Schools of Materials Science and Engineering, Shandong University, Jinan, 250061, PR China
| | - Junxing Yang
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials (Ministry of Education), Shandong University, Jinan, 250061, PR China
- Schools of Materials Science and Engineering, Shandong University, Jinan, 250061, PR China
| |
Collapse
|
127
|
Chauhan P, Bhardwaj N, Rajaura S, Chandra H, Singh A, Babu R, Gupta NJ. Benzo (A) pyrene exposure alters alveolar epithelial and macrophage cells diversity and induces antioxidant responses in lungs. Toxicol Rep 2024; 13:101777. [PMID: 39506978 PMCID: PMC11539143 DOI: 10.1016/j.toxrep.2024.101777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
This study was designed to investigate the toxic effects of benzo (a) pyrene (BaP) in the lungs. Mice were repeatedly treated orally with BaP (50 mg/kg body weight, twice a week for four weeks) to induce a tumour. After 4 months of BaP administration, tumours were visible beneath the skin. The histopathological section of the lungs shows congestion of pulmonary blood vessels, alveolar hyperplasia, and concurrent epithelial hyperplasia with infiltrates of inflammatory cells also seen. Thereafter, a single-cell suspension of lung tissues was stained with fluorescently conjugated antibodies for the demarcation of alveolar epithelial (anti-mouse CD74 and podoplanin) and macrophage (F4/80 and CD11b) cells and measured by flow cytometry. The expression of antioxidant genes was assessed by qRT-PCR. The number of alveolar epithelial cells 1 (AEC1) increased, but the number of alveolar epithelial cells 2 (AEC2) and transitional alveolar epithelial cells (TAEC) was significantly decreased in tumour-bearing mice. The proportion of CD11b+ alveolar macrophages (AM) and interstitial macrophages (IM) was increased, but the proportion of F4/80+ AM cells was reduced. The BaP administration significantly increased the ROS production in alveolar cells. The relative expression levels of antioxidant genes (SOD1, catalase, GPX1, and HIF-1α) were increased, but NRF2 expression was decreased in BaP-treated alveolar cells. The expression of anti-inflammatory (NF-κB) was also significantly increased. In conclusion, BaP exposure induced an inflammatory response, altered alveolar epithelial cell and macrophage diversity, and increased antioxidant responses in the lungs.
Collapse
Affiliation(s)
- Pooja Chauhan
- Department of Zoology and Environmental Science, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Nitin Bhardwaj
- Department of Zoology and Environmental Science, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Sumit Rajaura
- Department of Zoology and Environmental Science, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Harish Chandra
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Ashutosh Singh
- Department of Biochemistry, Lucknow University, Lucknow, Uttar Pradesh, India
| | - Ram Babu
- Department of Botany, Kirori Mal College, New Delhi, India
| | - Neelu Jain Gupta
- Department of Zoology, CCS University Campus Meerut, Uttar Pradesh, India
| |
Collapse
|
128
|
Keeley O, Coyne AN. Nuclear and degradative functions of the ESCRT-III pathway: implications for neurodegenerative disease. Nucleus 2024; 15:2349085. [PMID: 38700207 PMCID: PMC11073439 DOI: 10.1080/19491034.2024.2349085] [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: 02/11/2024] [Accepted: 04/24/2024] [Indexed: 05/05/2024] Open
Abstract
The ESCRT machinery plays a pivotal role in membrane-remodeling events across multiple cellular processes including nuclear envelope repair and reformation, nuclear pore complex surveillance, endolysosomal trafficking, and neuronal pruning. Alterations in ESCRT-III functionality have been associated with neurodegenerative diseases including Frontotemporal Dementia (FTD), Amyotrophic Lateral Sclerosis (ALS), and Alzheimer's Disease (AD). In addition, mutations in specific ESCRT-III proteins have been identified in FTD/ALS. Thus, understanding how disruptions in the fundamental functions of this pathway and its individual protein components in the human central nervous system (CNS) may offer valuable insights into mechanisms underlying neurodegenerative disease pathogenesis and identification of potential therapeutic targets. In this review, we discuss ESCRT components, dynamics, and functions, with a focus on the ESCRT-III pathway. In addition, we explore the implications of altered ESCRT-III function for neurodegeneration with a primary emphasis on nuclear surveillance and endolysosomal trafficking within the CNS.
Collapse
Affiliation(s)
- Olivia Keeley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alyssa N. Coyne
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
129
|
Ireland J, Kilian KA. The importance of matrix in cardiomyogenesis: Defined substrates for maturation and chamber specificity. Matrix Biol Plus 2024; 24:100160. [PMID: 39291079 PMCID: PMC11403269 DOI: 10.1016/j.mbplus.2024.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Human embryonic stem cell-derived cardiomyocytes (hESC-CM) are a promising source of cardiac cells for disease modelling and regenerative medicine. However, current protocols invariably lead to mixed population of cardiac cell types and often generate cells that resemble embryonic phenotypes. Here we developed a combinatorial approach to assess the importance of extracellular matrix proteins (ECMP) in directing the differentiation of cardiomyocytes from human embryonic stem cells (hESC). We did this by focusing on combinations of ECMP commonly found in the developing heart with a broad goal of identifying combinations that promote maturation and influence chamber specific differentiation. We formulated 63 unique ECMP combinations fabricated from collagen 1, collagen 3, collagen 4, fibronectin, laminin, and vitronectin, presented alone and in combinations, leading to the identification of specific ECMP combinations that promote hESC proliferation, pluripotency, and germ layer specification. When hESC were subjected to a differentiation protocol on the ECMP combinations, it revealed precise protein combinations that enhance differentiation as determined by the expression of cardiac progenitor markers kinase insert domain receptor (KDR) and mesoderm posterior transcription factor 1 (MESP1). High expression of cardiac troponin (cTnT) and the relative expression of myosin light chain isoforms (MLC2a and MLC2v) led to the identification of three surfaces that promote a mature cardiomyocyte phenotype. Action potential morphology was used to assess chamber specificity, which led to the identification of matrices that promote chamber-specific cardiomyocytes. This study provides a matrix-based approach to improve control over cardiomyocyte phenotypes during differentiation, with the scope for translation to cardiac laboratory models and for the generation of functional chamber specific cardiomyocytes for regenerative therapies.
Collapse
Affiliation(s)
- Jake Ireland
- School of Chemistry, UNSW Sydney, Sydney, New South Wales, Australia
| | - Kristopher A Kilian
- School of Chemistry, UNSW Sydney, Sydney, New South Wales, Australia
- School of Materials Science and Engineering, UNSW Sydney, Sydney, New South Wales, Australia
- Australian Centre for NanoMedicine, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
130
|
Jalal MS, Duttaroy A. Maternal Spargel/dPGC-1 is critical for embryonic development and influences chorion gene amplification via Cyclin E activity. Dev Biol 2024; 516:158-166. [PMID: 39173813 DOI: 10.1016/j.ydbio.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
The function of spargel/dPGC-1 in Drosophila oogenesis has been unequivocally established. Here, we sought to assess whether Spargel protein or RNA is essential for developmentally competent eggs. The trans-heterozygotic combination of two spargel mutant alleles allowed us to decrease Spargel expression to very low levels. Using this model, we now demonstrated the requirement for Spargel in eggshell patterning and embryonic development, which led us to establish that spargel is a maternal effect gene. Further examination of Spargel's potential mechanism of action in eggshell biogenesis revealed that low levels of Spargel in the adult ovary cause diminished Cyclin E activity, resulting in reduced chorion gene amplification levels, leading to eggshell biogenesis defects. Thus, another novel role for spargel/dPGC-1 is exposed whereby, through Cyclin E activity, this conserved transcriptional coactivator regulates the chorion gene amplification process.
Collapse
|
131
|
Yue T, Zhang W, Pei H, Danzeng D, He J, Yang J, Luo Y, Zhang Z, Xiong S, Yang X, Ji Q, Yang Z, Hou J. Monascus pigment-protected bone marrow-derived stem cells for heart failure treatment. Bioact Mater 2024; 42:270-283. [PMID: 39285916 PMCID: PMC11403898 DOI: 10.1016/j.bioactmat.2024.08.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have demonstrated significant therapeutic potential in heart failure (HF) treatment. However, their clinical application is impeded by low retention rate and low cellular activity of MSCs caused by high inflammatory and reactive oxygen species (ROS) microenvironment. In this study, monascus pigment (MP) nanoparticle (PPM) was proposed for improving adverse microenvironment and assisting in transplantation of bone marrow-derived MSCs (BMSCs). Meanwhile, in order to load PPM and reduce the mechanical damage of BMSCs, injectable hydrogels based on Schiff base cross-linking were prepared. The PPM displays ROS-scavenging and macrophage phenotype-regulating capabilities, significantly enhancing BMSCs survival and activity in HF microenvironment. This hydrogel demonstrates superior biocompatibility, injectability, and tissue adhesion. With the synergistic effects of injectable, adhesive hydrogel and the microenvironment-modulating properties of MP, cardiac function was effectively improved in the pericardial sac of rats. Our results offer insights into advancing BMSCs-based HF therapies and their clinical applications.
Collapse
Affiliation(s)
- Tian Yue
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Wentai Zhang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, Guangdong, 523000, China
| | - Haifeng Pei
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, China
| | - Dunzhu Danzeng
- School of Medicine, Tibet University, Lhasa, Tibet, 850000, China
| | - Jian He
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Jiali Yang
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Yong Luo
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Zhen Zhang
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Shiqiang Xiong
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| | - Xiangbo Yang
- Ya'an Xunkang Pharmaceutical Co., LTD, Ya'an, Sichuan, 625015, China
| | - Qisen Ji
- Ya'an Xunkang Pharmaceutical Co., LTD, Ya'an, Sichuan, 625015, China
| | - Zhilu Yang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, Guangdong, 523000, China
| | - Jun Hou
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu Institute of Cardiovascular Disease, Chengdu, Sichuan, 610031, China
| |
Collapse
|
132
|
King MR, Ruff KM, Pappu RV. Emergent microenvironments of nucleoli. Nucleus 2024; 15:2319957. [PMID: 38443761 PMCID: PMC10936679 DOI: 10.1080/19491034.2024.2319957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/13/2024] [Indexed: 03/07/2024] Open
Abstract
In higher eukaryotes, the nucleolus harbors at least three sub-phases that facilitate multiple functionalities including ribosome biogenesis. The three prominent coexisting sub-phases are the fibrillar center (FC), the dense fibrillar component (DFC), and the granular component (GC). Here, we review recent efforts in profiling sub-phase compositions that shed light on the types of physicochemical properties that emerge from compositional biases and territorial organization of specific types of macromolecules. We highlight roles played by molecular grammars which refers to protein sequence features including the substrate binding domains, the sequence features of intrinsically disordered regions, and the multivalence of these distinct types of domains / regions. We introduce the concept of a barcode of emergent physicochemical properties of nucleoli. Although our knowledge of the full barcode remains incomplete, we hope that the concept prompts investigations into undiscovered emergent properties and engenders an appreciation for how and why unique microenvironments control biochemical reactions.
Collapse
Affiliation(s)
- Matthew R. King
- Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, Campus, MO, USA
| | - Kiersten M. Ruff
- Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, Campus, MO, USA
| | - Rohit V. Pappu
- Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, Campus, MO, USA
| |
Collapse
|
133
|
Ohno M, Higuchi Y, Yamai K, Fuchigami S, Sasaki T, Oda Y, Hayashi I. Structural analysis of microtubule binding by minus-end targeting protein Spiral2. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119858. [PMID: 39370045 DOI: 10.1016/j.bbamcr.2024.119858] [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: 02/26/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
Microtubules (MTs) are dynamic cytoskeletal polymers that play a critical role in determining cell polarity and shape. In plant cells, acentrosomal MTs are localized on the cell surface and are referred to as cortical MTs. Cortical MTs nucleate in the cell cortex and detach from nucleation sites. The released MT filaments perform treadmilling, with the plus-ends of MTs polymerizing and the minus-ends depolymerizing. Minus-end targeting proteins, -TIPs, include Spiral2, which regulates the minus-end dynamics of acentrosomal MTs. Spiral2 accumulates autonomously at MT minus-ends and inhibits filament shrinkage, but the mechanism by which Spiral2 specifically recognizes minus-ends of MTs remains unknown. Here we describe the crystal structure of Spiral2's N-terminal MT-binding domain. The structural properties of this domain resemble those of the HEAT repeat structure of the tumor overexpressed gene (TOG) domain, but the number of HEAT repeats is different and the conformation is highly arched. Gel filtration and co-sedimentation analyses demonstrate that the domain binds preferentially to MT filaments rather than the tubulin dimer, and that the tubulin-binding mode of Spiral2 via the basic surface is similar to that of the TOG domain. We constructed an in silico model of the Spiral2-tubulin complex to identify residues that potentially recognize tubulin. Mutational analysis revealed that the key residues inferred in the model are involved in microtubule recognition, and provide insight into the mechanism by which end-targeting proteins stabilize MT ends.
Collapse
Affiliation(s)
- Marina Ohno
- Department of Medical Life Science, Yokohama City University, 1-7-29 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Yuuki Higuchi
- Department of Medical Life Science, Yokohama City University, 1-7-29 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Kazune Yamai
- Department of Medical Life Science, Yokohama City University, 1-7-29 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Sotaro Fuchigami
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga, Shizuoka, Japan
| | - Takema Sasaki
- Department of Biological Science, Graduate School of Science, Nagoya University, Furo-cho, Chikusa, Nagoya, Aichi, Japan
| | - Yoshihisa Oda
- Department of Biological Science, Graduate School of Science, Nagoya University, Furo-cho, Chikusa, Nagoya, Aichi, Japan
| | - Ikuko Hayashi
- Department of Medical Life Science, Yokohama City University, 1-7-29 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.
| |
Collapse
|
134
|
Li T, Zhou X, Wang Y, Liu X, Fan Y, Li R, Zhang H, Xu Y. AtCIPK20 regulates microtubule stability to mediate stomatal closure under drought stress in Arabidopsis. PLANT, CELL & ENVIRONMENT 2024; 47:5297-5314. [PMID: 39189953 DOI: 10.1111/pce.15112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024]
Abstract
Drought stress is a common abiotic challenge that profoundly impacts plant growth and development. As sessile organisms, plants rely on various physiological and morphological adaptations to cope with drought conditions. The CIPK (calcineurin B-like protein-interacting protein kinase) family proteins play a pivotal role in mediating plant responses to abiotic stress through modulation of cellular membrane events via the CBL-CIPK complex. However, reports documenting the CIPKs' regulation of non-membrane events are scant. In this study, we discovered a novel subcellular localisation pattern of the AtCIPK20 protein of Arabidopsis, specifically to cortical microtubules (cMT), which is distinct from previously reported localisation patterns of plant CIPKs. AtCIPK20 regulates ABA-induced loss of cMT organisation in guard cells, thereby facilitating stomatal closure, mitigating leaf water loss, and protecting plants from drought stress in Arabidopsis. The C-terminal regulatory domain of AtCIPK20 governs its cMT targeting, whereas the interaction of AtCIPK20 with its CBL partners disrupts this localisation. Notably, the cMT targeting characteristic of AtCIPK20 is not exclusive, as several other CIPK members in Arabidopsis, maize, and rice exhibit similar localisation patterns. These findings broaden our current understanding of the role of plant CIPK members in abiotic stress resistance and suggest that future exploration of CIPK molecular functions should adopt a more comprehensive perspective.
Collapse
Affiliation(s)
- Tao Li
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Xuna Zhou
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Yixiao Wang
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Xueqin Liu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Yudong Fan
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Ruiqi Li
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Huiyong Zhang
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Yufang Xu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
135
|
Fujiwara H. Dynamic duo: Cell-extracellular matrix interactions in hair follicle development and regeneration. Dev Biol 2024; 516:20-34. [PMID: 39059679 DOI: 10.1016/j.ydbio.2024.07.012] [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: 01/06/2024] [Revised: 06/20/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Ectodermal organs, such as hair follicles, originate from simple epithelial and mesenchymal sheets through a complex developmental process driven by interactions between these cell types. This process involves dermal condensation, placode formation, bud morphogenesis, and organogenesis, and all of these processes require intricate interactions among various tissues. Recent research has emphasized the crucial role of reciprocal and dynamic interactions between cells and the extracellular matrix (ECM), referred to as the "dynamic duo", in the development of ectodermal organs. These interactions provide spatially and temporally changing biophysical and biochemical cues within tissues. Using the hair follicle as an example, this review highlights two types of cell-ECM adhesion units-focal adhesion-type and hemidesmosome-type adhesion units-that facilitate communication between epithelial and mesenchymal cells. This review further explores how these adhesion units, along with other cell-ECM interactions, evolve during hair follicle development and regeneration, underscoring their importance in guiding both developmental and regenerative processes.
Collapse
|
136
|
Van Wauwe J, Mahy A, Craps S, Ekhteraei-Tousi S, Vrancaert P, Kemps H, Dheedene W, Doñate Puertas R, Trenson S, Roderick HL, Beerens M, Luttun A. PRDM16 determines specification of ventricular cardiomyocytes by suppressing alternative cell fates. Life Sci Alliance 2024; 7:e202402719. [PMID: 39304345 PMCID: PMC11415600 DOI: 10.26508/lsa.202402719] [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: 03/14/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
PRDM16 is a transcription factor with histone methyltransferase activity expressed at the earliest stages of cardiac development. Pathogenic mutations in humans lead to cardiomyopathy, conduction abnormalities, and heart failure. PRDM16 is specifically expressed in ventricular but not atrial cardiomyocytes, and its expression declines postnatally. Because in other tissues PRDM16 is best known for its role in binary cell fate decisions, we hypothesized a similar decision-making function in cardiomyocytes. Here, we demonstrated that cardiomyocyte-specific deletion of Prdm16 during cardiac development results in contractile dysfunction and abnormal electrophysiology of the postnatal heart, resulting in premature death. By combined RNA+ATAC single-cell sequencing, we found that PRDM16 favors ventricular working cardiomyocyte identity, by opposing the activity of master regulators of ventricular conduction and atrial fate. Myocardial loss of PRDM16 during development resulted in hyperplasia of the (distal) ventricular conduction system. Hence, PRDM16 plays an indispensable role during cardiac development by driving ventricular working cardiomyocyte identity.
Collapse
Affiliation(s)
- Jore Van Wauwe
- https://ror.org/05f950310 Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Alexia Mahy
- https://ror.org/05f950310 Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Sander Craps
- https://ror.org/05f950310 Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Samaneh Ekhteraei-Tousi
- https://ror.org/05f950310 Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Pieter Vrancaert
- https://ror.org/05f950310 Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Hannelore Kemps
- https://ror.org/05f950310 Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Wouter Dheedene
- https://ror.org/05f950310 Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Rosa Doñate Puertas
- https://ror.org/05f950310 Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Sander Trenson
- https://ror.org/05f950310 Cardiology Lab, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - H Llewelyn Roderick
- https://ror.org/05f950310 Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Manu Beerens
- Institute for Clinical Chemistry and Laboratory Medicine, Medizinische Klinik und Poliklinik Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Aernout Luttun
- https://ror.org/05f950310 Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
137
|
Hadjikypri X, Theofanous C, Christodoulidi A, Georgiades P. New findings on the orientation of the mouse anterior-posterior (A-P) axis before and during the initiation of gastrulation using a more refined embryo staging. Biochem Biophys Rep 2024; 40:101817. [PMID: 39286290 PMCID: PMC11402440 DOI: 10.1016/j.bbrep.2024.101817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/11/2024] [Accepted: 08/25/2024] [Indexed: 09/19/2024] Open
Abstract
A clinically significant event of early mammalian embryogenesis is the generation and early development of the anterior-posterior (A-P) axis, the imaginary line along which the structures from head to tail will form. This axis not only appears before gastrulation but is also oriented in a specific way in relation to the long and short diameters of the bilaterally symmetric epiblast. In mice, the most widely used mammalian in vivo model of early embryogenesis, the A-P axis is normally aligned with the long epiblast diameter by the early streak (ES) stage, a time during early gastrulation around embryonic day 6.5 (E6.5). Incorrect orientation of the A-P axis by the ES stage, that is, being aligned with the short epiblast diameter, leads to failure in completing gastrulation and results in embryo death soon after. Knowing the orientation of this axis from when it forms before gastrulation (around E5.5) until just before the ES stage is crucial for: (a) understanding the ill-defined factors involved in its formation and early development since they must be spatially related to it, and (b) providing explanations for the underlying mechanism when it is incorrectly orientated. However, the orientation of the A-P axis in pre-ES embryos of the E5.5-E6.5 period remains unclear. Specifically, although it is thought that this axis initially aligns with the short epiblast diameter and subsequently changes its orientation to become aligned with the long diameter by an unidentified pre-gastrulation stage before the ES stage, this proposition remains unresolved. This is largely due to the lack of clearly defined morphological criteria for staging certain periods of pre-ES mouse embryos (especially when the A-P axis initiates and when gastrulation begins prior to the ES stage), which are a prerequisite for identifying A-P axis orientation at specific pre-ES stages. Furthermore, although the orientation of an extraembryonic trophoblast asymmetry, specifically the tilt of the ectoplacental cone (EPC), coincides with that of the A-P axis by the ES stage, it is unknown whether such an association also exists at pre-gastrulation stages during A-P axis formation. Knowing this would exclude or implicate this trophoblast asymmetry as an upstream factor in orientating the A-P axis when it forms. To address these issues, we established a more refined embryo staging for the E5.5-E6.5 period using a novel combination of live morphological criteria and used it to examine the orientation of the A-P axis and that of the EPC tilt at specific stages. First, contrary to current thinking, we show that when the A-P axis first appears at our newly described anterior visceral endoderm-1 (AVE-1) and AVE-2 stages, it aligns with the long epiblast diameter in all embryos. This orientation is maintained in most embryos at all subsequent pre-gastrulation stages, specifically at our AVE-3 and pre-streak stages (the remaining embryos of these stages had this axis aligned with the short epiblast diameter). Second, we identified for the first time the pre-ES stage when gastrulation initiates, which we named the nascent streak (NS) stage, and further subdivided it into NS-1 and NS-2. At variance with current belief, we provide evidence that the earliest stage just before the ES stage when all embryos align their A-P axis with the long epiblast diameter is not a pre-gastrulation stage, but the NS-2 stage (at NS-1, most but not all embryos had this A-P axis orientation). Third, we implicate the EPC tilt as a possible extraembryonic factor in promoting correct A-P axis orientation, as this tilt exists before the AVE-1 stage and its orientation coincided with that of the A-P axis in all embryos at AVE-1, AVE-2 and ES stages and almost all embryos at AVE-3, pre-streak and NS stages. Overall, our work: (a) identified the previously unresolved orientation of the mouse A-P axis within the epiblast before the ES stage during the E5.5-E6.5 period; (b) provides an alternative explanation for when this axis is incorrectly oriented by the ES stage, namely, its defective alignment with the short epiblast diameter by this stage could be due to its failure to align with the long epiblast diameter from the time of its formation; and (c) implicates the pre-existing orientation of the EPC tilt as a possible factor in orientating the newly formed A-P axis.
Collapse
Affiliation(s)
- Xenia Hadjikypri
- Department of Biological Sciences, University of Cyprus, University Campus, P.O. Box 20537, 1678, Nicosia, Cyprus
| | - Christina Theofanous
- Department of Biological Sciences, University of Cyprus, University Campus, P.O. Box 20537, 1678, Nicosia, Cyprus
| | - Antonia Christodoulidi
- Department of Biological Sciences, University of Cyprus, University Campus, P.O. Box 20537, 1678, Nicosia, Cyprus
| | - Pantelis Georgiades
- Department of Biological Sciences, University of Cyprus, University Campus, P.O. Box 20537, 1678, Nicosia, Cyprus
| |
Collapse
|
138
|
Furze A, Waldron A, Yajima M. Visualizing metabolic regulation using metabolic biosensors during sea urchin embryogenesis. Dev Biol 2024; 516:122-129. [PMID: 39117030 PMCID: PMC11402557 DOI: 10.1016/j.ydbio.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024]
Abstract
Growing evidence suggests that metabolic regulation directly influences cellular function and development and thus may be more dynamic than previously expected. In vivo and in real-time analysis of metabolite activities during development is crucial to test this idea directly. In this study, we employ two metabolic biosensors to track the dynamics of pyruvate and oxidative phosphorylation (Oxphos) during the early embryogenesis of the sea urchin. A pyruvate sensor, PyronicSF, shows the signal enrichment on the mitotic apparatus, which is consistent with the localization patterns of the corresponding enzyme, pyruvate kinase (PKM). The addition of pyruvate increases the PyronicSF signal, while PKM knockdown decreases its signal, responding to the pyruvate level in the cell. Similarly, a ratio-metric sensor, Grx-roGFP, that reads the redox potential of the cell responds to DTT and H2O2, the known reducer and inducer of Oxphos. These observations suggest that these metabolic biosensors faithfully reflect the metabolic status in the cell during embryogenesis. The time-lapse imaging of these biosensors suggests that pyruvate and Oxphos levels change both spatially and temporarily during embryonic development. Pyruvate level is increased first in micromeres compared to other blastomeres at the 16-cell stage and remains high in ectoderm while decreasing in endomesoderm during gastrulation. In contrast, the Oxphos signal first decreases in micromeres at the 16-cell stage, while it increases in the endomesoderm during gastrulation, showing the opposite trend of the pyruvate signal. These results suggest that metabolic regulation is indeed both temporally and spatially dynamic during embryogenesis, and these biosensors are a valuable tool to monitor metabolic activities in real-time in developing embryos.
Collapse
Affiliation(s)
- Aidan Furze
- Department of Molecular Biology Cell Biology Biochemistry, Brown University, 185 Meeting Street, BOX-GL277, Providence, RI, 02912, USA
| | - Ashley Waldron
- Department of Molecular Biology Cell Biology Biochemistry, Brown University, 185 Meeting Street, BOX-GL277, Providence, RI, 02912, USA
| | - Mamiko Yajima
- Department of Molecular Biology Cell Biology Biochemistry, Brown University, 185 Meeting Street, BOX-GL277, Providence, RI, 02912, USA.
| |
Collapse
|
139
|
Wang H, Li X, You X, Zhao G. Harnessing the power of artificial intelligence for human living organoid research. Bioact Mater 2024; 42:140-164. [PMID: 39280585 PMCID: PMC11402070 DOI: 10.1016/j.bioactmat.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/21/2024] [Accepted: 08/26/2024] [Indexed: 09/18/2024] Open
Abstract
As a powerful paradigm, artificial intelligence (AI) is rapidly impacting every aspect of our day-to-day life and scientific research through interdisciplinary transformations. Living human organoids (LOs) have a great potential for in vitro reshaping many aspects of in vivo true human organs, including organ development, disease occurrence, and drug responses. To date, AI has driven the revolutionary advances of human organoids in life science, precision medicine and pharmaceutical science in an unprecedented way. Herein, we provide a forward-looking review, the frontiers of LOs, covering the engineered construction strategies and multidisciplinary technologies for developing LOs, highlighting the cutting-edge achievements and the prospective applications of AI in LOs, particularly in biological study, disease occurrence, disease diagnosis and prediction and drug screening in preclinical assay. Moreover, we shed light on the new research trends harnessing the power of AI for LO research in the context of multidisciplinary technologies. The aim of this paper is to motivate researchers to explore organ function throughout the human life cycle, narrow the gap between in vitro microphysiological models and the real human body, accurately predict human-related responses to external stimuli (cues and drugs), accelerate the preclinical-to-clinical transformation, and ultimately enhance the health and well-being of patients.
Collapse
Affiliation(s)
- Hui Wang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, PR China
| | - Xiangyang Li
- Henan Engineering Research Center of Food Microbiology, College of food and bioengineering, Henan University of Science and Technology, Luoyang, 471023, PR China
- Haihe Laboratory of Synthetic Biology, Tianjin, 300308, PR China
| | - Xiaoyan You
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, PR China
- Henan Engineering Research Center of Food Microbiology, College of food and bioengineering, Henan University of Science and Technology, Luoyang, 471023, PR China
| | - Guoping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, PR China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, PR China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, PR China
| |
Collapse
|
140
|
Huang N, Chan BP. A 3D micro-printed single cell micro-niche with asymmetric niche signals - An in vitro model for asymmetric cell division study. Biomaterials 2024; 311:122684. [PMID: 38971120 DOI: 10.1016/j.biomaterials.2024.122684] [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: 03/16/2024] [Revised: 05/31/2024] [Accepted: 06/23/2024] [Indexed: 07/08/2024]
Abstract
Intricate microenvironment signals orchestrate to affect cell behavior and fate during tissue morphogenesis. However, the underlying mechanisms on how specific local niche signals influence cell behavior and fate are not fully understood, owing to the lack of in vitro platform able to precisely, quantitatively, spatially, and independently manipulate individual niche signals. Here, microarrays of protein-based 3D single cell micro-niche (3D-SCμN), with precisely engineered biophysical and biochemical niche signals, are micro-printed by a multiphoton microfabrication and micropatterning technology. Mouse embryonic stem cell (mESC) is used as the model cell to study how local niche signals affect stem cell behavior and fate. By precisely engineering the internal microstructures of the 3D SCμNs, we demonstrate that the cell division direction can be controlled by the biophysical niche signals, in a cell shape-independent manner. After confining the cell division direction to a dominating axis, single mESCs are exposed to asymmetric biochemical niche signals, specifically, cell-cell adhesion molecule on one side and extracellular matrix on the other side. We demonstrate that, symmetry-breaking (asymmetric) niche signals successfully trigger cell polarity formation and bias the orientation of asymmetric cell division, the mitosis process resulting in two daughter cells with differential fates, in mESCs.
Collapse
Affiliation(s)
- Nan Huang
- Tissue Engineering Laboratory, Biomedical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China; Tissue Engineering Laboratory, School of Biomedical Sciences, Institute of Tissue Engineering and Regenerative Medicine, And Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong Special Administrative Region of China
| | - Barbara Pui Chan
- Tissue Engineering Laboratory, Biomedical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China; Tissue Engineering Laboratory, School of Biomedical Sciences, Institute of Tissue Engineering and Regenerative Medicine, And Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong Special Administrative Region of China.
| |
Collapse
|
141
|
Abedimanesh S, Safaralizadeh R, Jahanafrooz Z, Najafi S, Amini M, Nazarloo SS, Bahojb Mahdavi SZ, Baradaran B, Jebelli A, Mokhtarzadeh AA. Interaction of noncoding RNAs with hippo signaling pathway in cancer cells and cancer stem cells. Noncoding RNA Res 2024; 9:1292-1307. [PMID: 39045083 PMCID: PMC11263728 DOI: 10.1016/j.ncrna.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 07/25/2024] Open
Abstract
The Hippo signaling pathway has a regulatory function in the organogenesis process and cellular homeostasis, switching the cascade reactions of crucial kinases acts to turn off/on the Hippo pathway, altering the downstream gene expression and thereby regulating proliferation, apoptosis, or stemness. Disruption of this pathway can lead to the occurrence of various disorders and different types of cancer. Recent findings highlight the importance of ncRNAs, such as microRNA, circular RNA, and lncRNAs, in modulating the Hippo pathway. Defects in ncRNAs can disrupt Hippo pathway balance, increasing tumor cells, tumorigenesis, and chemotherapeutic resistance. This review summarizes ncRNAs' inhibitory or stimulatory role in - Hippo pathway regulation in cancer and stem cells. Identifying the relation between ncRNAs and the components of this pathway could pave the way for developing new biomarkers in the treatment and diagnosis of cancers.
Collapse
Affiliation(s)
- Saba Abedimanesh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Zohreh Jahanafrooz
- Department of Biology, Faculty of Sciences, University of Maragheh, Maragheh, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Soltani Nazarloo
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asiyeh Jebelli
- Department of Biological Sciences, Faculty of Basic Sciences, Higher Education Institute of Rab-Rashid, Tabriz, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
142
|
Eldeeb D, Ikeda Y, Hojo H, Ohba S. Unraveling the hidden complexity: Exploring dental tissues through single-cell transcriptional profiling. Regen Ther 2024; 27:218-229. [PMID: 38596822 PMCID: PMC11002530 DOI: 10.1016/j.reth.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/11/2024] Open
Abstract
Understanding the composition and function of cells constituting tissues and organs is vital for unraveling biological processes. Single-cell analysis has allowed us to move beyond traditional methods of categorizing cell types. This innovative technology allows the transcriptional and epigenetic profiling of numerous individual cells, leading to significant insights into the development, homeostasis, and pathology of various organs and tissues in both animal models and human samples. In this review, we delve into the outcomes of major investigations using single-cell transcriptomics to decipher the cellular composition of mammalian teeth and periodontal tissues. The recent single-cell transcriptome-based studies have traced in detail the dental epithelium-ameloblast lineage and dental mesenchyme lineages in the mouse incisors and the tooth germ of both mice and humans; unraveled the microenvironment, the identity of niche cells, and cellular intricacies in the dental pulp; shed light on the molecular mechanisms orchestrating root formation; and characterized cellular dynamics of the periodontal ligament. Additionally, cellular components in dental pulps were compared between healthy and carious teeth at a single-cell level. Each section of this review contributes to a comprehensive understanding of tooth biology, offering valuable insights into developmental processes, niche cell identification, and the molecular secrets of the dental environment.
Collapse
Affiliation(s)
- Dahlia Eldeeb
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Japan
- Department of Physiology, Division of Biomedical Sciences, Nihon University School of Medicine, Japan
- Department of Oral Biology, Faculty of Dentistry, Cairo University, Egypt
| | - Yuki Ikeda
- Department of Tissue and Developmental Biology, Graduate School of Dentistry, Osaka University, Japan
| | - Hironori Hojo
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Japan
| | - Shinsuke Ohba
- Department of Tissue and Developmental Biology, Graduate School of Dentistry, Osaka University, Japan
| |
Collapse
|
143
|
Rodriguez Torres CS, Wicker NB, Puccini de Castro V, Stefinko M, Bennett DC, Bernhardt B, Garcia Montes de Oca M, Jallow S, Flitcroft K, Palalay JJS, Payán Parra OA, Stern YE, Koelle MR, Voisine C, Woods IG, Lo TW, Stern MJ, de la Cova CC. The Caenorhabditis elegans protein SOC-3 permits an alternative mode of signal transduction by the EGL-15 FGF receptor. Dev Biol 2024; 516:183-195. [PMID: 39173814 PMCID: PMC11488645 DOI: 10.1016/j.ydbio.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/02/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Fibroblast Growth Factors and their receptors (FGFRs) comprise a cell signaling module that can stimulate signaling by Ras and the kinases Raf, MEK, and ERK to regulate animal development and homeostatic functions. In Caenorhabditis elegans, the sole FGFR ortholog EGL-15 acts with the GRB2 ortholog SEM-5 to promote chemoattraction and migration by the sex myoblasts (SMs) and fluid homeostasis by the hypodermis (Hyp7). Cell-specific differences in EGL-15 signaling were suggested by the phenotypes caused by egl-15(n1457), an allele that removes a region of its C-terminal domain (CTD) known to bind SEM-5. To determine how mutations altered EGL-15 activity in the SMs and Hyp7, we used the kinase reporter ERK-KTR to measure activation of the ERK ortholog MPK-1. Consequences of egl-15(n1457) were cell-specific, resulting in loss of MPK-1 activity in the SMs and elevated activity in Hyp7. Previous studies of Hyp7 showed that loss of the CLR-1 phosphatase causes a fluid homeostasis defect termed "Clear" that is suppressed by reduction of EGL-15 signaling, a phenotype termed "Suppressor of Clear" (Soc). To identify mechanisms that permit EGL-15 signaling in Hyp7, we conducted a genetic screen for Soc mutants in the clr-1; egl-15(n1457) genotype. We report the identification of SOC-3, a protein with putative SEM-5-binding motifs and PH and PTB domains similar to DOK and IRS proteins. In combination with the egl-15(n1457) mutation, loss of either soc-3, the GAB1 ortholog soc-1, or the SHP2 ortholog ptp-2, reduced MPK-1 activation. We generated alleles of soc-3 to test the requirement for the SEM-5-binding motifs, finding that residue Tyr356 is required for function. We propose that EGL-15-mediated SM chemoattraction relies solely on the direct interaction between SEM-5 and the EGL-15 CTD. In Hyp7, EGL-15 signaling uses two mechanisms: the direct SEM-5 binding mechanism; and an alternative, CTD-independent mechanism involving SOC-3, SOC-1, and PTP-2. This work demonstrates that FGF signaling uses distinct, tissue-specific mechanisms in development, and identifies SOC-3 as a potential adaptor that facilitates Ras pathway activation by FGFR.
Collapse
Affiliation(s)
| | - Nicole B Wicker
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | | | - Mariya Stefinko
- Department of Biology, Northeastern Illinois University, Chicago, IL, 60625, USA
| | | | | | | | - Sainabou Jallow
- Department of Biology, Ithaca College, Ithaca, NY, 14850, USA
| | - Katelyn Flitcroft
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | | | - Omar A Payán Parra
- Department of Biology, Northeastern Illinois University, Chicago, IL, 60625, USA
| | - Yaakov E Stern
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | | | - Cindy Voisine
- Department of Biology, Northeastern Illinois University, Chicago, IL, 60625, USA
| | - Ian G Woods
- Department of Biology, Ithaca College, Ithaca, NY, 14850, USA
| | - Te-Wen Lo
- Department of Biology, Ithaca College, Ithaca, NY, 14850, USA
| | - Michael J Stern
- Department of Biology, Northeastern Illinois University, Chicago, IL, 60625, USA
| | - Claire C de la Cova
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA.
| |
Collapse
|
144
|
Latham ZD, Bermudez A, Hu JK, Lin NYC. Regulation of epithelial cell jamming transition by cytoskeleton and cell-cell interactions. BIOPHYSICS REVIEWS 2024; 5:041301. [PMID: 39416285 PMCID: PMC11479637 DOI: 10.1063/5.0220088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Multicellular systems, such as epithelial cell collectives, undergo transitions similar to those in inert physical systems like sand piles and foams. To remodel or maintain tissue organization during development or disease, these collectives transition between fluid-like and solid-like states, undergoing jamming or unjamming transitions. While these transitions share principles with physical systems, understanding their regulation and implications in cell biology is challenging. Although cell jamming and unjamming follow physics principles described by the jamming diagram, they are fundamentally biological processes. In this review, we explore how cellular processes and interactions regulate jamming and unjamming transitions. We begin with an overview of how these transitions control tissue remodeling in epithelial model systems and describe recent findings of the physical principles governing tissue solidification and fluidization. We then explore the mechanistic pathways that modulate the jamming phase diagram axes, focusing on the regulation of cell fluctuations and geometric compatibility. Drawing upon seminal works in cell biology, we discuss the roles of cytoskeleton and cell-cell adhesion in controlling cell motility and geometry. This comprehensive view illustrates the molecular control of cell jamming and unjamming, crucial for tissue remodeling in various biological contexts.
Collapse
Affiliation(s)
- Zoe D. Latham
- Bioengineering Department, UCLA, Los Angeles, California 90095, USA
| | | | - Jimmy K. Hu
- Authors to whom correspondence should be addressed: and
| | | |
Collapse
|
145
|
Martins SG, Ribeiro V, Melo C, Paulino-Cavaco C, Antonini D, Dayalan Naidu S, Murtinheira F, Fonseca I, Saget B, Pita M, Fernandes DR, Gameiro Dos Santos P, Rodrigues G, Zilhão R, Herrera F, Dinkova-Kostova AT, Carlos AR, Thorsteinsdóttir S. Laminin-α2 chain deficiency in skeletal muscle causes dysregulation of multiple cellular mechanisms. Life Sci Alliance 2024; 7:e202402829. [PMID: 39379105 PMCID: PMC11463332 DOI: 10.26508/lsa.202402829] [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: 05/17/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024] Open
Abstract
LAMA2, coding for the laminin-α2 chain, is a crucial ECM component, particularly abundant in skeletal muscle. Mutations in LAMA2 trigger the often-lethal LAMA2-congenital muscular dystrophy (LAMA2-CMD). Various phenotypes have been linked to LAMA2-CMD; nevertheless, the precise mechanisms that malfunction during disease onset in utero remain unknown. We generated Lama2-deficient C2C12 cells and found that Lama2-deficient myoblasts display proliferation, differentiation, and fusion defects, DNA damage, oxidative stress, and mitochondrial dysfunction. Moreover, fetal myoblasts isolated from the dy W mouse model of LAMA2-CMD display impaired differentiation and fusion in vitro. We also showed that disease onset during fetal development is characterized by a significant down-regulation of gene expression in muscle fibers, causing pronounced effects on cytoskeletal organization, muscle differentiation, and altered DNA repair and oxidative stress responses. Together, our findings provide unique insights into the critical importance of the laminin-α2 chain for muscle differentiation and muscle cell homeostasis.
Collapse
Affiliation(s)
- Susana G Martins
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Vanessa Ribeiro
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Melo
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia Paulino-Cavaco
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Dario Antonini
- Department of Biology, University of Naples "Federico II", Naples, Italy
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Fernanda Murtinheira
- https://ror.org/01c27hj86 Biosystems and Integrative Sciences Institute (BioISI), Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Inês Fonseca
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Bérénice Saget
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Mafalda Pita
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Diogo R Fernandes
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro Gameiro Dos Santos
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Gabriela Rodrigues
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Rita Zilhão
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Vegetal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Federico Herrera
- https://ror.org/01c27hj86 Biosystems and Integrative Sciences Institute (BioISI), Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Ana Rita Carlos
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Sólveig Thorsteinsdóttir
- https://ror.org/01c27hj86 Centre for Ecology, Evolution and Environmental Changes (CE3C) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- https://ror.org/01c27hj86 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
146
|
Lv Y, Ji L, Dai H, Qiu S, Wang Y, Teng C, Yu B, Mi D, Yao C. Identification of key regulatory genes involved in myelination after spinal cord injury by GSEA analysis. Exp Neurol 2024; 382:114966. [PMID: 39326824 DOI: 10.1016/j.expneurol.2024.114966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/31/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Multilayer dense myelin tissue provides insulating space and nutritional support for axons in healthy spinal cord tissue. Oligodendrocyte precursor cells (OPCs) are the main glial cells that complement myelin loss in the central nervous system and play an important role in the repair of spinal cord injury (SCI). However, the regulation of axonal remyelination after SCI is still insufficient. In this study, we focused on the changes in genes related to myelin repair after rat hemisection SCI by gene set enrichment analysis (GSEA). Key genes proteolipid protein 1 (Plp1), hexosaminidase subunit alpha (Hexa), and hexosaminidase subunit beta (Hexb) during remyelination after SCI were found. Through quantitative real-time polymerase chain reaction (qPCR) experiments, we confirmed that within 28 days after rat hemisection SCI, the mRNA expression of gene Plp1 gradually decreased, while the expressions of gene Hexa and Hexb gradually increased, which was consistent with RNA sequencing results. In vitro, we performed EdU proliferation assays on OPC cell line OLN-93 and primary rat OPCs. We found that interference of Plp1 promoted OPC proliferation, while interference of Hexa and Hexb inhibited OPC proliferation. In addition, we performed in vitro differentiation experiments on primary rat OPCs. By measuring myelin sheath branch outgrowth and the fluorescence intensity of the mature myelin sheath marker myelin basic protein (MBP), we found that interference of Hexa or Hexb promoted OPC differentiation and maturation, but interference of Plp1 inhibited this process. Finally, we injected Hexb siRNA in vivo and found that interfering Hexb could improve motor movements and myelin regeneration after SCI in rats. Our results provide new target genes that can selectively regulate the proliferation and differentiation of endogenous OPCs, providing new ideas for promoting remyelination and functional recovery after SCI.
Collapse
Affiliation(s)
- Yehua Lv
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Lingyun Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Hui Dai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Shanru Qiu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Yu Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Cheng Teng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Daguo Mi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China.
| | - Chun Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
147
|
Sevriev B, Dimitrova S, Kehayova G, Dragomanova S. Trehalose: Neuroprotective Effects and Mechanisms-An Updated Review. NEUROSCI 2024; 5:429-444. [PMID: 39484301 PMCID: PMC11503274 DOI: 10.3390/neurosci5040032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
Trehalose is a naturally occurring disaccharide that has recently gained significant attention for its neuroprotective properties in various models of neurodegeneration. This review provides an overview of available experimental data on the beneficial properties of trehalose for central nervous system pathological conditions. Trehalose's impact on neuronal cell survival and function was also examined. As a result, we identified that trehalose's neuroprotection includes autophagy modulation as well as its capability to stabilize proteins and inhibit the formation of misfolded ones. Moreover, trehalose mitigates oxidative stress-induced neuronal damage by stabilizing cellular membranes and modulating mitochondrial function. Furthermore, trehalose attenuates excitotoxicity-induced neuroinflammation by suppressing pro-inflammatory cytokine release and inhibiting inflammasome activation. A possible connection of trehalose with the gut-brain axis was also examined. These findings highlight the potential therapeutic effects of trehalose in neurodegenerative diseases. According to the conclusions drawn from this study, trehalose is a promising neuroprotective agent as a result of its distinct mechanism of action, which makes this compound a candidate for further research and the development of therapeutic strategies to combat neuronal damage and promote neuroprotection in various neurological diseases.
Collapse
Affiliation(s)
- Borislav Sevriev
- Faculty of Pharmacy, Medical University of Varna "Prof. Dr. Paraskev Stoyanov", 9000 Varna, Bulgaria;
| | - Simeonka Dimitrova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Varna "Prof. Dr. Paraskev Stoyanov", 9000 Varna, Bulgaria; (S.D.); (G.K.)
| | - Gabriela Kehayova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Varna "Prof. Dr. Paraskev Stoyanov", 9000 Varna, Bulgaria; (S.D.); (G.K.)
| | - Stela Dragomanova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Varna "Prof. Dr. Paraskev Stoyanov", 9000 Varna, Bulgaria; (S.D.); (G.K.)
| |
Collapse
|
148
|
Guo P, Yu Y, Kang H, Liu Y, Zhu D, Sun C, Xing Z, Tang Z, Chen K, Tan A. GASZ is indispensable for gametogenesis in the silkworm, Bombyx mori. INSECT MOLECULAR BIOLOGY 2024; 33:626-637. [PMID: 38728119 DOI: 10.1111/imb.12921] [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: 01/20/2024] [Accepted: 04/28/2024] [Indexed: 05/12/2024]
Abstract
The prominent role of the P-element induced wimpy testis (PIWI)-interacting RNA (piRNA) pathway in animals is to silence transposable elements and maintain genome stability, ensuring proper gametogenesis in gonads. GASZ (Germ cell protein with Ankyrin repeats, Sterile alpha motif, and leucine Zipper) is an evolutionarily conserved protein located on the outer mitochondrial membrane of germ cells and plays vital roles in the piRNA pathway and spermatogenesis in mammals. In the model insect Drosophila melanogaster, GASZ is essential for piRNA biogenesis and oogenesis, whereas its biological functions in non-drosophilid insects are still unknown. Here, we describe a comprehensive investigation of GASZ functions in the silkworm, Bombyx mori, a lepidopteran model insect, by using a binary transgenic CRISPR/Cas9 system. The BmGASZ mutation did not affect growth and development, but led to sterility in both males and females. Eupyrene sperm bundles of mutant males exhibited developmental defects, while the apyrene sperm bundles were normal, which were further confirmed through double copulation experiments with sex-lethal mutants, which males possess functional eupyrene sperm and abnormal apyrene sperm. In female mutant moths, ovarioles were severely degenerated and the eggs in ovarioles were deformed compared with that of wild type (WT). Further RNA-seq and RT-qPCR analysis revealed that amounts of piRNAs and transposon expression were dysregulated in gonads of mutants. In summary, this study has demonstrated vital roles of BmGASZ in gametogenesis through regulating the piRNA pathway in B. mori.
Collapse
Affiliation(s)
- Peilin Guo
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Ye Yu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Hongxia Kang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Yutong Liu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Dalin Zhu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Chenxin Sun
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Zhiping Xing
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Ziyue Tang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Kai Chen
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Anjiang Tan
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| |
Collapse
|
149
|
Ren N, Zhang G, Yang X, Chen J, Ni L, Jiang M. MAPKKK28 functions upstream of the MKK1-MPK1 cascade to regulate abscisic acid responses in rice. PLANT, CELL & ENVIRONMENT 2024; 47:5140-5157. [PMID: 39166350 DOI: 10.1111/pce.15095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/15/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024]
Abstract
The mitogen-activated protein kinase (MAPK) cascade (MAPKKK-MAPKK-MAPK) plays a critical role in biotic and abiotic stress responses and abscisic acid (ABA) signalling. A previous study has shown that the ABA-activated MKK1-MPK1 cascade is essential in regulating ABA response and stress tolerance in rice. However, the specific MAPKKK upstream of the MKK1-MPK1 cascade in ABA signalling remains unknown. Here, we identified that MAPKKK28, a previously uncharacterized member of the rice MEKK family, is involved in regulating ABA responses, including seed germination, root growth, stomatal closure, and the tolerance to oxidative stress and osmotic stress. We found that MAPKKK28 directly interacts with and phosphorylates MKK1. Further analysis indicated that the activation of both MKK1 and MPK1 depends on MAPKKK28 in ABA signalling. Genetic analysis revealed that MAPKKK28 functions upstream of the MKK1-MPK1 cascade to positively regulate ABA responses and enhance tolerance to oxidative and osmotic stress. These results not only reveal a new complete MAPK cascade in plants but also uncover its importance in ABA signalling.
Collapse
Affiliation(s)
- Ning Ren
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Gang Zhang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
| | - Xiaokun Yang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Jing Chen
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Lan Ni
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Mingyi Jiang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
150
|
Schultz A, Albertos-Arranz H, Sáez XS, Morgan J, Darland DC, Gonzalez-Duarte A, Kaufmann H, Mendoza-Santiesteban CE, Cuenca N, Lefcort F. Neuronal and glial cell alterations involved in the retinal degeneration of the familial dysautonomia optic neuropathy. Glia 2024; 72:2268-2294. [PMID: 39228100 DOI: 10.1002/glia.24612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 07/27/2024] [Accepted: 08/19/2024] [Indexed: 09/05/2024]
Abstract
Familial dysautonomia (FD) is a rare genetic neurodevelopmental and neurodegenerative disorder. In addition to the autonomic and peripheral sensory neuropathies that challenge patient survival, one of the most debilitating symptoms affecting patients' quality of life is progressive blindness resulting from the steady loss of retinal ganglion cells (RGCs). Within the FD community, there is a concerted effort to develop treatments to prevent the loss of RGCs. However, the mechanisms underlying the death of RGCs are not well understood. To study the mechanisms underlying RGC death, Pax6-cre;Elp1loxp/loxp male and female mice and postmortem retinal tissue from an FD patient were used to explore the neuronal and non-neuronal cellular pathology associated with the FD optic neuropathy. Neurons, astrocytes, microglia, Müller glia, and endothelial cells were investigated using a combination of histological analyses. We identified a novel disruption of cellular homeostasis and gliosis in the FD retina. Beginning shortly after birth and progressing with age, the FD retina is marked by astrogliosis and perturbations in microglia, which coincide with vascular remodeling. These changes begin before the onset of RGC death, suggesting alterations in the retinal neurovascular unit may contribute to and exacerbate RGC death. We reveal for the first time that the FD retina pathology includes reactive gliosis, increased microglial recruitment to the ganglion cell layer (GCL), disruptions in the deep and superficial vascular plexuses, and alterations in signaling pathways. These studies implicate the neurovascular unit as a disease-modifying target for therapeutic interventions in FD.
Collapse
Affiliation(s)
- Anastasia Schultz
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Henar Albertos-Arranz
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Xavier Sánchez Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Jamie Morgan
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Diane C Darland
- Department of Biology, University of North Dakota, Grand Forks, North Dakota, USA
| | | | - Horacio Kaufmann
- Department of Neurology, NYU Langone Health, New York, New York, USA
| | - Carlos E Mendoza-Santiesteban
- Department of Neurology, NYU Langone Health, New York, New York, USA
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Frances Lefcort
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| |
Collapse
|