1
|
Hou X, Chen Y, Carrillo ND, Cryns VL, Anderson RA, Sun J, Wang S, Chen M. Phosphoinositide signaling at the cytoskeleton in the regulation of cell dynamics. Cell Death Dis 2025; 16:296. [PMID: 40229242 PMCID: PMC11997203 DOI: 10.1038/s41419-025-07616-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
The cytoskeleton, composed of microfilaments, intermediate filaments, and microtubules, provides the structural basis for cellular functions such as motility and adhesion. Equally crucial, phosphoinositide (PIPn) signaling is a critical regulator of these processes and other biological activities, though its precise impact on cytoskeletal dynamics has yet to be systematically investigated. This review explores the complex interplay between PIPn signaling and the cytoskeleton, detailing how PIPn modulates the dynamics of actin, intermediate filaments, and microtubules to shape cellular behavior. Dysregulation of PIPn signaling is implicated in various diseases, including cancer, highlighting promising therapeutic opportunities through targeted modulation of these pathways. Future research should aim to elucidate the intricate molecular interactions and broader cellular responses to PIPn signaling perturbations, particularly in disease contexts, to devise effective strategies for restoring cytoskeletal integrity.
Collapse
Affiliation(s)
- Xiaoting Hou
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yu Chen
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Noah D Carrillo
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Vincent L Cryns
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard A Anderson
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Jichao Sun
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Department of Critical Care Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
| | - Songlin Wang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.
| | - Mo Chen
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
2
|
Salinas Y, Chauhan SC, Bandyopadhyay D. Small-Molecule Mitotic Inhibitors as Anticancer Agents: Discovery, Classification, Mechanisms of Action, and Clinical Trials. Int J Mol Sci 2025; 26:3279. [PMID: 40244152 PMCID: PMC11989755 DOI: 10.3390/ijms26073279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Despite decades of research, cancer continues to be a disease of great concern to millions of people around the world. It has been responsible for a total of 609,820 deaths in the U.S. alone in 2023. Over the years, many drugs have been developed to remove or reduce the disease's impact, all with varying mechanisms of action and side effects. One class of these drugs is small-molecule mitotic inhibitors. These drugs inhibit cancer cell mitosis or self-replication, impeding cell proliferation and eventually leading to cell death. In this paper, small-molecule mitotic inhibitors are discussed and classified through their discovery, underlying chemistry, and mechanism(s) of action. The binding/inhibition of microtubule-related proteins, DNA damage through the inhibition of Checkpoint Kinase 1 protein, and the inhibition of mitotic kinase proteins are discussed in terms of their anticancer activity to provide an overview of a variety of mitotic inhibitors currently commercially available or under investigation, including those in ongoing clinical trial. Clinical trials for anti-mitotic agents are discussed to track research progress, gauge current understanding, and identify possible future prospects. Additionally, antibody-drug conjugates that use mitotic inhibitors as cytotoxic payloads are discussed as possible ways of administering effective anticancer treatments with minimal toxicity.
Collapse
Affiliation(s)
- Yazmin Salinas
- School of Integrative Biological and Chemical Sciences, The University of Texas Rio Grande Valley, 1201 West University Drive, Edinburg, TX 78539, USA;
| | - Subhash C. Chauhan
- Division of Cancer Immunology and Microbiology, Medicine, and Oncology Integrated Service Unit, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA;
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX 78504, USA
| | - Debasish Bandyopadhyay
- School of Integrative Biological and Chemical Sciences, The University of Texas Rio Grande Valley, 1201 West University Drive, Edinburg, TX 78539, USA;
- School of Earth, Environmental, and Marine Sciences (SEEMS), The University of Texas Rio Grande Valley, 1201 West University Drive, Edinburg, TX 78539, USA
| |
Collapse
|
3
|
Ringlander J, Rydell GE, Kann M. From the Cytoplasm into the Nucleus-Hepatitis B Virus Travel and Genome Repair. Microorganisms 2025; 13:157. [PMID: 39858925 PMCID: PMC11767736 DOI: 10.3390/microorganisms13010157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Hepatitis B virus (HBV) is a major global health concern, affecting millions of people worldwide. HBV is part of the hepadnaviridae family and one of the primary causes of acute and chronic liver infections, leading to conditions such as cirrhosis and hepatocellular carcinoma (HCC). Understanding the intracellular transport and genome repair mechanisms of HBV is crucial for developing new drugs, which-in combination with immune modulators-may contribute to potential cures. This review will explore the current knowledge of HBV intracytoplasmic and nuclear transport, as well as genome repair processes, while drawing comparisons to other viruses with nuclear replication.
Collapse
Affiliation(s)
- Johan Ringlander
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden; (J.R.); (G.E.R.)
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, 41346 Gothenburg, Sweden
| | - Gustaf E. Rydell
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden; (J.R.); (G.E.R.)
| | - Michael Kann
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden; (J.R.); (G.E.R.)
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, 41346 Gothenburg, Sweden
| |
Collapse
|
4
|
Laflamme N, Triassi V, Martineau L, Toffa DH, Létourneau-Guillon L, Laplante A, Cossette P, Samarut É, Tétreault M, Nguyen DK. X-Linked Bilateral Polymicrogyria With Epilepsy and Intellectual Disability Associated With a Novel KIF4A Variant. Am J Med Genet A 2025; 197:e63860. [PMID: 39268972 DOI: 10.1002/ajmg.a.63860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 07/19/2024] [Accepted: 08/18/2024] [Indexed: 09/15/2024]
Abstract
We studied three brothers and a maternal half-brother featuring global developmental delay, mild to moderate intellectual disability, epilepsy, microcephaly, and strabismus. All had bilateral perisylvian and perirolandic polymicrogyria, while some also had malformations of the hippocampus (malrotation and dysplasia), cerebellum (heterotopias and asymmetric aplasia), corpus callosum dysgenesis, and brainstem asymmetric dysplasia. Exome sequencing showed that all four patients had a novel variant (c.1597C>T:p.Leu533Phe) on the KIF4A gene on chromosome X. We discuss how this variant is possibly pathogenic and could explain the reported phenotype.
Collapse
Affiliation(s)
- Naomi Laflamme
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Valérie Triassi
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
- Bioinformatics Program, Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Laurence Martineau
- Neurology Division, Centre Hospitalier Universitaire de Québec, Université Laval, Quebec City, Quebec, Canada
| | - Dènahin Hinnoutondji Toffa
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada
| | | | - Annie Laplante
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Patrick Cossette
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada
- Division of Radiology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Éric Samarut
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada
| | - Martine Tétreault
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada
| | - Dang Khoa Nguyen
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada
- Neurology Division, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| |
Collapse
|
5
|
McMaster B, Thorpe CJ, Rossjohn J, Deane CM, Koohy H. Quantifying conformational changes in the TCR:pMHC-I binding interface. Front Immunol 2024; 15:1491656. [PMID: 39687625 PMCID: PMC11646856 DOI: 10.3389/fimmu.2024.1491656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/24/2024] [Indexed: 12/18/2024] Open
Abstract
Background T cells form one of the key pillars of adaptive immunity. Using their surface bound T cell antigen receptors (TCRs), these cells screen millions of antigens presented by major histocompatibility complex (MHC) or MHC-like molecules. In other protein families, the dynamics of protein-protein interactions have important implications for protein function. Case studies of TCR:class I peptide-MHCs (pMHC-Is) structures have reported mixed results on whether the binding interfaces undergo conformational change during engagement and no robust statistical quantification has been done to generalise these results. Thus, it remains an open question of whether movement occurs in the binding interface that enables the recognition and activation of T cells. Methods In this work, we quantify the conformational changes in the TCR:pMHC-I binding interface by creating a dataset of 391 structures, comprising 22 TCRs, 19 MHC alleles, and 79 peptide structures in both unbound (apo) and bound (holo) conformations. Results In support of some case studies, we demonstrate that all complementarity determining region (CDR) loops move to a certain extent but only CDR3α and CDR3β loops modify their shape when binding pMHC-Is. We also map the contacts between TCRs and pMHC-Is, generating a novel fingerprint of TCRs on MHC molecules and show that the CDR3α tends to bind the N-terminus of the peptide and the CDR3β tends to bind the C-terminus of the peptide. Finally, we show that the presented peptides can undergo conformational changes when engaged by TCRs, as has been reported in past literature, but novelly show these changes depend on how the peptides are anchored in the MHC binding groove. Conclusions Our work has implications in understanding the behaviour of TCR:pMHC-I interactions and providing insights that can be used for modelling Tcell antigen specificity, an ongoing grand challenge in immunology.
Collapse
Affiliation(s)
- Benjamin McMaster
- Koohy Lab, Medical Research Council Translational Immune Discovery Unit (MRC TIDU), Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Christopher J. Thorpe
- Open Targets, Wellcome Genome Campus, Hinxton, United Kingdom
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, United Kingdom
| | - Jamie Rossjohn
- Rossjohn Lab, Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Rossjohn Lab, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Charlotte M. Deane
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Hashem Koohy
- Koohy Lab, Medical Research Council Translational Immune Discovery Unit (MRC TIDU), Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Zhou L, Xiong Y, Dwivedy A, Zheng M, Cooper L, Shepherd S, Song T, Hong W, Le LTP, Chen X, Umrao S, Rong L, Wang T, Cunningham BT, Wang X. Bioinspired designer DNA NanoGripper for virus sensing and potential inhibition. Sci Robot 2024; 9:eadi2084. [PMID: 39602515 PMCID: PMC11750070 DOI: 10.1126/scirobotics.adi2084] [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: 04/11/2023] [Accepted: 10/25/2024] [Indexed: 11/29/2024]
Abstract
DNA has shown great biocompatibility, programmable mechanical properties, and precise structural addressability at the nanometer scale, rendering it a material for constructing versatile nanorobots for biomedical applications. Here, we present the design principle, synthesis, and characterization of a DNA nanorobotic hand, called DNA NanoGripper, that contains a palm and four bendable fingers as inspired by naturally evolved human hands, bird claws, and bacteriophages. Each NanoGripper finger consists of three phalanges connected by three rotatable joints that are bendable in response to the binding of other entities. NanoGripper functions are enabled and driven by the interactions between moieties attached to the fingers and their binding partners. We demonstrate that the NanoGripper can be engineered to effectively interact with and capture nanometer-scale objects, including gold nanoparticles, gold NanoUrchins, and SARS-CoV-2 virions. With multiple DNA aptamer nanoswitches programmed to generate a fluorescent signal that is enhanced on a photonic crystal platform, the NanoGripper functions as a highly sensitive biosensor that selectively detects intact SARS-CoV-2 virions in human saliva with a limit of detection of ~100 copies per milliliter, providing a sensitivity equal to that of reverse transcription quantitative polymerase chain reaction (RT-qPCR). Quantified by flow cytometry assays, we demonstrated that the NanoGripper-aptamer complex can effectively block viral entry into the host cells, suggesting its potential for inhibiting virus infections. The design, synthesis, and characterization of a sophisticated nanomachine that can be tailored for specific applications highlight a promising pathway toward feasible and efficient solutions to the detection and potential inhibition of virus infections.
Collapse
Affiliation(s)
- Lifeng Zhou
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yanyu Xiong
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Abhisek Dwivedy
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Mengxi Zheng
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Laura Cooper
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Skye Shepherd
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Tingjie Song
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Wei Hong
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Linh T. P. Le
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- VinUni-Illinois Smart Health Center, VinUniversity, Hanoi, Vietnam
| | - Xin Chen
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Saurabh Umrao
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Tong Wang
- Advanced Science Research Center at Graduate Center, City University of New York, New York, NY 10031, USA
| | - Brian T. Cunningham
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Xing Wang
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
7
|
Ning R, Li C, Fan T, Ji T, Xu W. Metabolite and Transcriptomic Changes Reveal the Cold Stratification Process in Sinopodophyllum hexandrum Seeds. PLANTS (BASEL, SWITZERLAND) 2024; 13:2693. [PMID: 39409563 PMCID: PMC11479046 DOI: 10.3390/plants13192693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024]
Abstract
Sinopodophyllum hexandrum (Royle) Ying, an endangered perennial medicinal herb, exhibits morpho-physiological dormancy in its seeds, requiring cold stratification for germination. However, the precise molecular mechanisms underlying this transition from dormancy to germination remain unclear. This study integrates transcriptome and plant hormone-targeted metabolomics techniques to unravel these intricate molecular regulatory mechanisms during cold stratification in S. hexandrum seeds. Significant alterations in the physicochemical properties (starch, soluble sugars, soluble proteins) and enzyme activities (PK, SDH, G-6-PDH) within the seeds occur during stratification. To characterize and monitor the formation and transformation of plant hormones throughout this process, extracts from S. hexandrum seeds at five stratification stages of 0 days (S0), 30 days (S1), 60 days (S2), 90 days (S3), and 120 days (S4) were analyzed using UPLC-MS/MS, revealing a total of 37 differential metabolites belonging to seven major classes of plant hormones. To investigate the biosynthetic and conversion processes of plant hormones related to seed dormancy and germination, the transcriptome of S. hexandrum seeds was monitored via RNA-seq, revealing 65,372 differentially expressed genes associated with plant hormone synthesis and signaling. Notably, cytokinins (CKs) and gibberellins (GAs) exhibited synergistic effects, while abscisic acid (ABA) displayed antagonistic effects. Furthermore, key hub genes were identified through integrated network analysis. In this rigorous scientific study, we systematically elucidate the intricate dynamic molecular regulatory mechanisms that govern the transition from dormancy to germination in S. hexandrum seeds during stratification. By meticulously examining these mechanisms, we establish a solid foundation of knowledge that serves as a scientific basis for facilitating large-scale breeding programs and advancing the artificial cultivation of this highly valued medicinal plant.
Collapse
Affiliation(s)
- Rongchun Ning
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China; (R.N.)
- Qinghai Key Laboratory of Qinghai-Tibet Plateau Biological Resources, Xining 810008, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Caixia Li
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China; (R.N.)
| | - Tingting Fan
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China; (R.N.)
- Qinghai Key Laboratory of Qinghai-Tibet Plateau Biological Resources, Xining 810008, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingting Ji
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China; (R.N.)
| | - Wenhua Xu
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China; (R.N.)
- Qinghai Key Laboratory of Qinghai-Tibet Plateau Biological Resources, Xining 810008, China
| |
Collapse
|
8
|
Zhang H, Wang X, Qu M, Yu H, Yin J, Liu X, Liu Y, Zhang B, Zhang Y, Wei Z, Yang F, Wang J, Shi C, Fan G, Sun J, Long L, Hutchins DA, Bowler C, Lin S, Wang D, Lin Q. Genome of Halimeda opuntia reveals differentiation of subgenomes and molecular bases of multinucleation and calcification in algae. Proc Natl Acad Sci U S A 2024; 121:e2403222121. [PMID: 39302967 PMCID: PMC11441479 DOI: 10.1073/pnas.2403222121] [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: 02/15/2024] [Accepted: 08/16/2024] [Indexed: 09/22/2024] Open
Abstract
Algae mostly occur either as unicellular (microalgae) or multicellular (macroalgae) species, both being uninucleate. There are important exceptions, however, as some unicellular algae are multinucleate and macroscopic, some of which inhabit tropical seas and contribute to biocalcification and coral reef robustness. The evolutionary mechanisms and ecological significance of multinucleation and associated traits (e.g., rapid wound healing) are poorly understood. Here, we report the genome of Halimeda opuntia, a giant multinucleate unicellular chlorophyte characterized by interutricular calcification. We achieve a high-quality genome assembly that shows segregation into four subgenomes, with evidence for polyploidization concomitant with historical sea level and climate changes. We further find myosin VIII missing in H. opuntia and three other unicellular multinucleate chlorophytes, suggesting a potential mechanism that may underpin multinucleation. Genome analysis provides clues about how the unicellular alga could survive fragmentation and regenerate, as well as potential signatures for extracellular calcification and the coupling of calcification with photosynthesis. In addition, proteomic alkalinity shifts were found to potentially confer plasticity of H. opuntia to ocean acidification (OA). Our study provides crucial genetic information necessary for understanding multinucleation, cell regeneration, plasticity to OA, and different modes of calcification in algae and other organisms, which has important implications in reef conservation and bioengineering.
Collapse
Affiliation(s)
- Hao Zhang
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
- Marine Biodiversity and Ecological Evolution Research Center, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen361102, China
| | - Xin Wang
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
- Marine Biodiversity and Ecological Evolution Research Center, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
| | - Meng Qu
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
- Marine Biodiversity and Ecological Evolution Research Center, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
| | - Haiyan Yu
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen361102, China
| | - Jianping Yin
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
| | | | - Yuhong Liu
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
| | - Bo Zhang
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
| | - Yanhong Zhang
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
| | - Zhangliang Wei
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
| | - Fangfang Yang
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
| | - Jingtian Wang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen361102, China
| | | | | | - Jun Sun
- College of Marine Science and Technology, China University of Geosciences, Wuhan430074, China
| | - Lijuan Long
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
| | - David A. Hutchins
- Department of Biological Sciences, Marine and Environmental Biology, University of Southern California, Los Angeles, CA90007
| | - Chris Bowler
- Institut de Biologie de l’Ecole Normale Supérieure, Ecole Normale Supérieure, CNRS, INSERM, Paris Sciences et Lettres Research University, Paris75005, France
| | - Senjie Lin
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen361102, China
- Department of Marine Sciences, University of Connecticut, Groton, CT06340
| | - Dazhi Wang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen361102, China
| | - Qiang Lin
- Chinese Academy of Sciences Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
- Marine Biodiversity and Ecological Evolution Research Center, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou510301, China
- University of Chinese Academy of Sciences, Beijing100049, China
| |
Collapse
|
9
|
Kreis NN, Moon HH, Wordeman L, Louwen F, Solbach C, Yuan J, Ritter A. KIF2C/MCAK a prognostic biomarker and its oncogenic potential in malignant progression, and prognosis of cancer patients: a systematic review and meta-analysis as biomarker. Crit Rev Clin Lab Sci 2024; 61:404-434. [PMID: 38344808 PMCID: PMC11815995 DOI: 10.1080/10408363.2024.2309933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/05/2023] [Accepted: 01/22/2024] [Indexed: 03/24/2024]
Abstract
KIF2C/MCAK (KIF2C) is the most well-characterized member of the kinesin-13 family, which is critical in the regulation of microtubule (MT) dynamics during mitosis, as well as interphase. This systematic review briefly describes the important structural elements of KIF2C, its regulation by multiple molecular mechanisms, and its broad cellular functions. Furthermore, it systematically summarizes its oncogenic potential in malignant progression and performs a meta-analysis of its prognostic value in cancer patients. KIF2C was shown to be involved in multiple crucial cellular processes including cell migration and invasion, DNA repair, senescence induction and immune modulation, which are all known to be critical during the development of malignant tumors. Indeed, an increasing number of publications indicate that KIF2C is aberrantly expressed in multiple cancer entities. Consequently, we have highlighted its involvement in at least five hallmarks of cancer, namely: genome instability, resisting cell death, activating invasion and metastasis, avoiding immune destruction and cellular senescence. This was followed by a systematic search of KIF2C/MCAK's expression in various malignant tumor entities and its correlation with clinicopathologic features. Available data were pooled into multiple weighted meta-analyses for the correlation between KIF2Chigh protein or gene expression and the overall survival in breast cancer, non-small cell lung cancer and hepatocellular carcinoma patients. Furthermore, high expression of KIF2C was correlated to disease-free survival of hepatocellular carcinoma. All meta-analyses showed poor prognosis for cancer patients with KIF2Chigh expression, associated with a decreased overall survival and reduced disease-free survival, indicating KIF2C's oncogenic potential in malignant progression and as a prognostic marker. This work delineated the promising research perspective of KIF2C with modern in vivo and in vitro technologies to further decipher the function of KIF2C in malignant tumor development and progression. This might help to establish KIF2C as a biomarker for the diagnosis or evaluation of at least three cancer entities.
Collapse
Affiliation(s)
- Nina-Naomi Kreis
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Ha Hyung Moon
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Linda Wordeman
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Frank Louwen
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Christine Solbach
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Juping Yuan
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Andreas Ritter
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| |
Collapse
|
10
|
Lucas J, Geisler M. Plant Kinesin Repertoires Expand with New Domain Architecture and Contract with the Loss of Flagella. J Mol Evol 2024; 92:381-401. [PMID: 38926179 DOI: 10.1007/s00239-024-10178-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
Kinesins are eukaryotic microtubule motor proteins subdivided into conserved families with distinct functional roles. While many kinesin families are widespread in eukaryotes, each organismal lineage maintains a unique kinesin repertoire composed of many families with distinct numbers of genes. Previous genomic surveys indicated that land plant kinesin repertoires differ markedly from other eukaryotes. To determine when repertoires diverged during plant evolution, we performed robust phylogenomic analyses of kinesins in 24 representative plants, two algae, two animals, and one yeast. These analyses show that kinesin repertoires expand and contract coincident with major shifts in the biology of algae and land plants. One kinesin family and five subfamilies, each defined by unique domain architectures, emerged in the green algae. Four of those kinesin groups expanded in ancestors of modern land plants, while six other kinesin groups were lost in the ancestors of pollen-bearing plants. Expansions of different kinesin families and subfamilies occurred in moss and angiosperm lineages. Other kinesin families remained stable and did not expand throughout plant evolution. Collectively these data support a radiation of kinesin domain architectures in algae followed by differential positive and negative selection on kinesins families and subfamilies in different lineages of land plants.
Collapse
Affiliation(s)
- Jessica Lucas
- Department of Biology, University of Wisconsin-Oshkosh, 800 Algoma Blvd, Oshkosh, WI, 54901, USA.
| | - Matt Geisler
- School of Biological Science, Southern Illinois University, Carbondale, IL, 54901, USA
| |
Collapse
|
11
|
Song Z, Zhang C, Song G, Wei H, Xu W, Pan H, Ding C, Xu M, Zhen Y. Unraveling the lncRNA-miRNA-mRNA Regulatory Network Involved in Poplar Coma Development through High-Throughput Sequencing. Int J Mol Sci 2024; 25:7403. [PMID: 39000510 PMCID: PMC11242837 DOI: 10.3390/ijms25137403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Poplar coma, the fluff-like appendages of seeds originating from the differentiated surface cells of the placenta and funicle, aids in the long-distance dispersal of seeds in the spring. However, it also poses hazards to human safety and causes pollution in the surrounding environment. Unraveling the regulatory mechanisms governing the initiation and development of coma is essential for addressing this issue comprehensively. In this study, strand-specific RNA-seq was conducted at three distinct stages of coma development, revealing 1888 lncRNAs and 52,810 mRNAs. The expression profiles of lncRNAs and mRNAs during coma development were analyzed. Subsequently, potential target genes of lncRNAs were predicted through co-localization and co-expression analyses. Integrating various types of sequencing data, lncRNA-miRNA-TF regulatory networks related to the initiation of coma were constructed. Utilizing identified differentially expressed genes encoding kinesin and actin, lncRNA-miRNA-mRNA regulatory networks associated with the construction and arrangement of the coma cytoskeleton were established. Additionally, relying on differentially expressed genes encoding cellulose synthase, sucrose synthase, and expansin, lncRNA-miRNA-mRNA regulatory networks related to coma cell wall synthesis and remodeling were developed. This study not only enhances the comprehension of lncRNA but also provides novel insights into the molecular mechanisms governing the initiation and development of poplar coma.
Collapse
Affiliation(s)
- Zihe Song
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Chenghao Zhang
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Guotao Song
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Hang Wei
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation of State Forestry Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing 100091, China
| | - Wenlin Xu
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Huixin Pan
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Changjun Ding
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation of State Forestry Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing 100091, China
| | - Meng Xu
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Yan Zhen
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
12
|
Abedini SS, Akhavantabasi S, Liang Y, Heng JIT, Alizadehsani R, Dehzangi I, Bauer DC, Alinejad-Rokny H. A critical review of the impact of candidate copy number variants on autism spectrum disorder. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 794:108509. [PMID: 38977176 DOI: 10.1016/j.mrrev.2024.108509] [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: 02/04/2023] [Revised: 04/14/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder (NDD) influenced by genetic, epigenetic, and environmental factors. Recent advancements in genomic analysis have shed light on numerous genes associated with ASD, highlighting the significant role of both common and rare genetic mutations, as well as copy number variations (CNVs), single nucleotide polymorphisms (SNPs) and unique de novo variants. These genetic variations disrupt neurodevelopmental pathways, contributing to the disorder's complexity. Notably, CNVs are present in 10 %-20 % of individuals with autism, with 3 %-7 % detectable through cytogenetic methods. While the role of submicroscopic CNVs in ASD has been recently studied, their association with genomic loci and genes has not been thoroughly explored. In this review, we focus on 47 CNV regions linked to ASD, encompassing 1632 genes, including protein-coding genes and long non-coding RNAs (lncRNAs), of which 659 show significant brain expression. Using a list of ASD-associated genes from SFARI, we detect 17 regions harboring at least one known ASD-related protein-coding gene. Of the remaining 30 regions, we identify 24 regions containing at least one protein-coding gene with brain-enriched expression and a nervous system phenotype in mouse mutants, and one lncRNA with both brain-enriched expression and upregulation in iPSC to neuron differentiation. This review not only expands our understanding of the genetic diversity associated with ASD but also underscores the potential of lncRNAs in contributing to its etiology. Additionally, the discovered CNVs will be a valuable resource for future diagnostic, therapeutic, and research endeavors aimed at prioritizing genetic variations in ASD.
Collapse
Affiliation(s)
- Seyedeh Sedigheh Abedini
- UNSW BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia; School of Biotechnology & Biomolecular Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Shiva Akhavantabasi
- Department of Molecular Biology and Genetics, Yeni Yuzyil University, Istanbul, Turkey; Ghiaseddin Jamshid Kashani University, Andisheh University Town, Danesh Blvd, 3441356611, Abyek, Qazvin, Iran
| | - Yuheng Liang
- UNSW BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Julian Ik-Tsen Heng
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6845, Australia
| | - Roohallah Alizadehsani
- Institute for Intelligent Systems Research and Innovation (IISRI), Deakin University, Victoria, Australia
| | - Iman Dehzangi
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA; Department of Computer Science, Rutgers University, Camden, NJ 08102, USA
| | - Denis C Bauer
- Transformational Bioinformatics, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Sydney, Australia; Applied BioSciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, Australia
| | - Hamid Alinejad-Rokny
- UNSW BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia; Tyree Institute of Health Engineering (IHealthE), UNSW Sydney, Sydney, NSW 2052, Australia.
| |
Collapse
|
13
|
Reyaz E, Tandon R, Beg MA, Dey R, Puri N, Salotra P, Nakhasi HL, Selvapandiyan A. Proteome profile of Leishmania donovani Centrin1 -/- parasite-infected human macrophage cell line and its implications in determining possible mechanisms of protective immunity. Microbes Infect 2024; 26:105340. [PMID: 38663721 DOI: 10.1016/j.micinf.2024.105340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024]
Abstract
Our developed cell division-specific 'centrin' gene deleted Leishmania donovani (LdCen1-/-) the causative parasite of the fatal visceral-leishmaniasis (VL), exhibits a selective growth arrest at the intracellular stage and is anticipated as a live attenuated vaccine candidate against VL. LdCen1-/- immunization in animals has shown increased IFN-γ secreting CD4+ and CD8+ T cells along with protection conferred by a protective proinflammatory immune response. A label-free proteomics approach has been employed to understand the physiology of infection and predict disease interceptors during Leishmania-host interactions. Proteomic modulation after infection of human macrophage cell lines suggested elevated annexin A6, implying involvement in various biological processes such as membrane repair, transport, actin dynamics, cell proliferation, survival, differentiation, and inflammation, thereby potentiating its immunological protective capacity. Additionally, S100A8 and S100A9 proteins, known for maintaining homeostatic balance in regulating the inflammatory response, have been upregulated after infection. The inhibitory clade of serpins, known to inhibit cysteine proteases (CPs), was upregulated in host cells after 48 h of infection. This is reflected in the diminished expression of CPs in the parasites during infection. Such proteome analysis confirms LdCen1-/- efficacy as a vaccine candidate and predicts potential markers in future vaccine development strategies against infectious diseases.
Collapse
Affiliation(s)
- Enam Reyaz
- JH-Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Rati Tandon
- JH-Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Mirza Adil Beg
- JH-Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Niti Puri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Poonam Salotra
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi 110029, India
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD 20993, USA
| | - A Selvapandiyan
- JH-Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
14
|
Liu H, Welburn JPI. A circle of life: platelet and megakaryocyte cytoskeleton dynamics in health and disease. Open Biol 2024; 14:240041. [PMID: 38835242 DOI: 10.1098/rsob.240041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/24/2024] [Indexed: 06/06/2024] Open
Abstract
Platelets are blood cells derived from megakaryocytes that play a central role in regulating haemostasis and vascular integrity. The microtubule cytoskeleton of megakaryocytes undergoes a critical dynamic reorganization during cycles of endomitosis and platelet biogenesis. Quiescent platelets have a discoid shape maintained by a marginal band composed of microtubule bundles, which undergoes remarkable remodelling during platelet activation, driving shape change and platelet function. Disrupting or enhancing this process can cause platelet dysfunction such as bleeding disorders or thrombosis. However, little is known about the molecular mechanisms underlying the reorganization of the cytoskeleton in the platelet lineage. Recent studies indicate that the emergence of a unique platelet tubulin code and specific pathogenic tubulin mutations cause platelet defects and bleeding disorders. Frequently, these mutations exhibit dominant negative effects, offering valuable insights into both platelet disease mechanisms and the functioning of tubulins. This review will highlight our current understanding of the role of the microtubule cytoskeleton in the life and death of platelets, along with its relevance to platelet disorders.
Collapse
Affiliation(s)
- Haonan Liu
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Julie P I Welburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| |
Collapse
|
15
|
Ni BB, Liu H, Wang ZS, Zhang GY, Sang ZY, Liu JJ, He CY, Zhang JG. A chromosome-scale genome of Rhus chinensis Mill. provides new insights into plant-insect interaction and gallotannins biosynthesis. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2024; 118:766-786. [PMID: 38271098 DOI: 10.1111/tpj.16631] [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: 07/21/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/27/2024]
Abstract
Rhus chinensis Mill., an economically valuable Anacardiaceae species, is parasitized by the galling aphid Schlechtendalia chinensis, resulting in the formation of the Chinese gallnut (CG). Here, we report a chromosomal-level genome assembly of R. chinensis, with a total size of 389.40 Mb and scaffold N50 of 23.02 Mb. Comparative genomic and transcriptome analysis revealed that the enhanced structure of CG and nutritional metabolism contribute to improving the adaptability of R. chinensis to S. chinensis by supporting CG and galling aphid growth. CG was observed to be abundant in hydrolysable tannins (HT), particularly gallotannin and its isomers. Tandem repeat clusters of dehydroquinate dehydratase/shikimate dehydrogenase (DQD/SDH) and serine carboxypeptidase-like (SCPL) and their homologs involved in HT production were determined as specific to HT-rich species. The functional differentiation of DQD/SDH tandem duplicate genes and the significant contraction in the phenylalanine ammonia-lyase (PAL) gene family contributed to the accumulation of gallic acid and HT while minimizing the production of shikimic acid, flavonoids, and condensed tannins in CG. Furthermore, we identified one UDP glucosyltransferase (UGT84A), three carboxylesterase (CXE), and six SCPL genes from conserved tandem repeat clusters that are involved in gallotannin biosynthesis and hydrolysis in CG. We then constructed a regulatory network of these genes based on co-expression and transcription factor motif analysis. Our findings provide a genomic resource for the exploration of the underlying mechanisms of plant-galling insect interaction and highlight the importance of the functional divergence of tandem duplicate genes in the accumulation of secondary metabolites.
Collapse
Affiliation(s)
- Bing-Bing Ni
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation, National Forestry and Grassland Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing, 100091, China
- Collaborative Innovation Center of Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China
| | - Hong Liu
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation, National Forestry and Grassland Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing, 100091, China
| | - Zhao-Shan Wang
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation, National Forestry and Grassland Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing, 100091, China
| | - Guo-Yun Zhang
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation, National Forestry and Grassland Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing, 100091, China
| | - Zi-Yang Sang
- Forest Enterprise of Wufeng County in Hubei Province, Wufeng, 443400, Hubei, China
| | - Juan-Juan Liu
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation, National Forestry and Grassland Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing, 100091, China
| | - Cai-Yun He
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation, National Forestry and Grassland Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing, 100091, China
- Collaborative Innovation Center of Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China
| | - Jian-Guo Zhang
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation, National Forestry and Grassland Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing, 100091, China
- Collaborative Innovation Center of Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China
| |
Collapse
|
16
|
Nambala P, Noyes H, Namulondo J, Nyangiri O, Alibu VP, Nerima B, MacLeod A, Matovu E, Musaya J, Mulindwa J. Transcriptome profiles of Trypanosoma brucei rhodesiense in Malawi reveal focus specific gene expression profiles associated with pathology. PLoS Negl Trop Dis 2024; 18:e0011516. [PMID: 38701067 PMCID: PMC11095692 DOI: 10.1371/journal.pntd.0011516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 05/15/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Sleeping sickness caused by Trypanosoma brucei rhodesiense is a fatal disease and endemic in Southern and Eastern Africa. There is an urgent need to develop novel diagnostic and control tools to achieve elimination of rhodesiense sleeping sickness which might be achieved through a better understanding of trypanosome gene expression and genetics using endemic isolates. Here, we describe transcriptome profiles and population structure of endemic T. b. rhodesiense isolates in human blood in Malawi. METHODOLOGY Blood samples of r-HAT cases from Nkhotakota and Rumphi foci were collected in PaxGene tubes for RNA extraction before initiation of r-HAT treatment. 100 million reads were obtained per sample, reads were initially mapped to the human genome reference GRCh38 using HiSat2 and then the unmapped reads were mapped against Trypanosoma brucei reference transcriptome (TriTrypDB54_TbruceiTREU927) using HiSat2. Differential gene expression analysis was done using the DeSeq2 package in R. SNP calling from reads that were mapped to the T. brucei genome was done using GATK in order to identify T.b. rhodesiense population structure. RESULTS 24 samples were collected from r-HAT cases of which 8 were from Rumphi and 16 from Nkhotakota foci. The isolates from Nkhotakota were enriched with transcripts for cell cycle arrest and stumpy form markers, whereas isolates in Rumphi focus were enriched with transcripts for folate biosynthesis and antigenic variation pathways. These parasite focus-specific transcriptome profiles are consistent with the more virulent disease observed in Rumphi and a less symptomatic disease in Nkhotakota associated with the non-dividing stumpy form. Interestingly, the Malawi T.b. rhodesiense isolates expressed genes enriched for reduced cell proliferation compared to the Uganda T.b. rhodesiense isolates. PCA analysis using SNPs called from the RNAseq data showed that T. b. rhodesiense parasites from Nkhotakota are genetically distinct from those collected in Rumphi. CONCLUSION Our results suggest that the differences in disease presentation in the two foci is mainly driven by genetic differences in the parasites in the two major endemic foci of Rumphi and Nkhotakota rather than differences in the environment or host response.
Collapse
Affiliation(s)
- Peter Nambala
- Department of Biochemistry and Sports Sciences, College of Natural Sciences, Makerere University, Kampala, Uganda
- Kamuzu University of Health Sciences, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Harry Noyes
- Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Joyce Namulondo
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Oscar Nyangiri
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Vincent Pius Alibu
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Barbara Nerima
- Department of Biochemistry and Sports Sciences, College of Natural Sciences, Makerere University, Kampala, Uganda
| | - Annette MacLeod
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
| | - Enock Matovu
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Janelisa Musaya
- Kamuzu University of Health Sciences, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Julius Mulindwa
- Department of Biochemistry and Sports Sciences, College of Natural Sciences, Makerere University, Kampala, Uganda
| | | |
Collapse
|
17
|
Zhu H, Xu J, Yu K, Wu J, Xu H, Wang S, Wen T. Genome-wide identification of the key kinesin genes during fiber and boll development in upland cotton (Gossypium hirsutum L.). Mol Genet Genomics 2024; 299:38. [PMID: 38517563 DOI: 10.1007/s00438-024-02093-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/11/2023] [Indexed: 03/24/2024]
Abstract
Kinesin is a kind of motor protein, which interacts with microtubule filaments and regulates cellulose synthesis. Cotton fiber is a natural model for studying the cellular development and cellulose synthesis. Therefore, a systematic research of kinesin gene family in cotton (Gossypium spp.) will be beneficial for both understanding the function of kinesin protein and assisting the fiber improvement. Here, we aimed to identify the key kinesin genes present in cotton by combining genome-wide expression profile data, association mapping, and public quantitative trait loci (QTLs) in upland cotton (G. hirsutum L.). Results showed that 159 kinesin genes, including 15 genes of the kinesin-13 gene subfamily, were identified in upland cotton; of which 157 kinesin genes can be traced back to the diploid ancestors, G. raimondii and G. arboreum. Using a combined analysis of public QTLs and genome-wide expression profile information, there were 29 QTLs co-localized together with 28 kinesin genes in upland cotton, including 10 kinesin-13 subfamily genes. Genome-wide expression profile data indicated that, among the 28 co-localized genes, seven kinesin genes were predominantly expressed in fibers or ovules. By association mapping analysis, 30 kinesin genes were significantly associated with three fiber traits, among which a kinesin-13 gene, Ghir_A11G028430, was found to be associated with both cotton boll length and lint weight, and one kinesin-7 gene, Ghir_D04G017880 (Gh_Kinesin7), was significantly associated with fiber strength. In addition, two missense mutations were identified in the motor domain of the Gh_Kinesin7 protein. Overall, the kinesin gene family seemingly plays an important role in cotton fiber and boll development. The exploited kinesin genes will be beneficial for the genetic improvement of fiber quality and yield.
Collapse
Affiliation(s)
- Hong Zhu
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Jianzhong Xu
- Stock seed farm of Gao'an, Yichun, 330800, Jiangxi, China
| | - Kanbing Yu
- Xishuangbanna Institute of Agricultural Science, Xishuangbanna Autonomous Prefecture, Yunnan, 666100, China
| | - Jianfei Wu
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Huifang Xu
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Shubin Wang
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Tianwang Wen
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China.
| |
Collapse
|
18
|
Roy A, Pathak Z, Kumar H. Investigating LIM (Lin-11, Isl-1, and Mec-3) Kinases and Their Correlation with Pathological Events and Microtubule Dynamics in an Experimental Model of Spinal Cord Injury in Rats. ACS Pharmacol Transl Sci 2024; 7:667-679. [PMID: 38481685 PMCID: PMC10928889 DOI: 10.1021/acsptsci.3c00272] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2025]
Abstract
The spinal cord injury (SCI) and the neurodegenerative processes accompanying it follow an intricate pathway with very limited options for treatment strategies until now. Microtubules, essential for the growth and maintenance of neurons, are mostly disorganized and destabilized due to neurodegeneration. Regeneration or plasticity is restricted to the adult central nervous system (CNS) due to several intrinsic and extrinsic mechanisms. Some fundamental or injury-induced expressions of specific molecules can be inhibited or antagonized pharmacologically to protect neurons to a certain extent after neurodegeneration. Accordingly, these molecules offer an excellent target as a therapeutic approach to promote neuroprotection. LIM kinases (LIMKs) are one of these molecules that phosphorylates members of the actin-depolymerizing factor (ADF)/cofilin family of actin-binding and filament-severing proteins. The individual role of LIMKs has not yet been studied in the pathology of SCI. In this study, we targeted LIMK and checked its role in microtubule destabilization in vitro. LIMK1 was found to be upregulated after microtubule depolymerization and inhibition of LIMK with specific inhibitor-protected neurons. Then, we checked the expressions of individual LIMKs throughout different time points across SCI in a rat contusion model, correlating with established pathophysiological markers. The phosphorylated form of LIMK1 was found to be elevated at chronic time points after injury, where scar formation and diminution of neurons prevail. Finally, we targeted the LIMK pathway with its specific inhibitor BMS-5, which showed neuroprotection after SCI. Overall, our results provided a concept concerning how a small-molecule inhibitor of LIMK may offer a strategy to treat SCI-associated neurodegeneration.
Collapse
Affiliation(s)
- Abhishek Roy
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Zarna Pathak
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
19
|
Chatterjee S, Ganguly A, Bhattacharyya D. Reprogramming nucleolar size by genetic perturbation of the extranuclear Rab GTPases Ypt6 and Ypt32. FEBS Lett 2024; 598:283-301. [PMID: 37994551 DOI: 10.1002/1873-3468.14776] [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: 09/03/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 11/24/2023]
Abstract
Reprogramming organelle size has been proposed as a potential therapeutic approach. However, there have been few reports of nucleolar size reprogramming. We addressed this question in Saccharomyces cerevisiae by studying mutants having opposite effects on the nucleolar size. Mutations in genes involved in nuclear functions (KAR3, CIN8, and PRP45) led to enlarged nuclei/nucleoli, whereas mutations in secretory pathway family genes, namely the Rab-GTPases YPT6 and YPT32, reduced nucleolar size. When combined with mutations leading to enlarged nuclei/nucleoli, the YPT6 or YPT32 mutants can effectively reprogram the nuclear/nucleolar size almost back to normal. Our results further indicate that null mutation of YPT6 causes secretory stress that indirectly influences nuclear localization of Maf1, the negative regulator of RNA Polymerase III, which might reduce the nucleolar size by inhibiting nucleolar transcript enrichment.
Collapse
Affiliation(s)
- Shreosi Chatterjee
- Department of Cell and Tumor Biology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, Maharashtra, India
| | - Abira Ganguly
- Department of Cell and Tumor Biology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, Maharashtra, India
| | - Dibyendu Bhattacharyya
- Department of Cell and Tumor Biology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, Maharashtra, India
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
20
|
Nambiar A, Manjithaya R. Driving autophagy - the role of molecular motors. J Cell Sci 2024; 137:jcs260481. [PMID: 38329417 DOI: 10.1242/jcs.260481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Most of the vesicular transport pathways inside the cell are facilitated by molecular motors that move along cytoskeletal networks. Autophagy is a well-explored catabolic pathway that is initiated by the formation of an isolation membrane known as the phagophore, which expands to form a double-membraned structure that captures its cargo and eventually moves towards the lysosomes for fusion. Molecular motors and cytoskeletal elements have been suggested to participate at different stages of the process as the autophagic vesicles move along cytoskeletal tracks. Dynein and kinesins govern autophagosome trafficking on microtubules through the sequential recruitment of their effector proteins, post-translational modifications and interactions with LC3-interacting regions (LIRs). In contrast, myosins are actin-based motors that participate in various stages of the autophagic flux, as well as in selective autophagy pathways. However, several outstanding questions remain with regard to how the dominance of a particular motor protein over another is controlled, and to the molecular mechanisms that underlie specific disease variants in motor proteins. In this Review, we aim to provide an overview of the role of molecular motors in autophagic flux, as well as highlight their dysregulation in diseases, such as neurodegenerative disorders and pathogenic infections, and ageing.
Collapse
Affiliation(s)
- Akshaya Nambiar
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
| |
Collapse
|
21
|
Zhu H, Xu J, Yu K, Wu J, Xu H, Wang S, Wen T. Genome-wide identification of the key Kinesin genes during fiber and boll development in upland cotton (Gossypium hirsutum L). Mol Genet Genomics 2024; 299:2. [PMID: 38200363 DOI: 10.1007/s00438-023-02087-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/11/2023] [Indexed: 01/12/2024]
Abstract
Kinesin is a kind of motor protein, which interacts with microtubule filaments and regulates cellulose synthesis. Cotton fiber is a natural model for studying the cellular development and cellulose synthesis. Therefore, a systematic research of Kinesin gene family in cotton (Gossypium spp.) will be beneficial for both understanding the function of Kinesin protein and assisting the fiber improvement. Here, we aimed to identify the key Kinesin genes present in cotton by combining genome-wide expression profile data, association mapping, and public quantitative trait loci (QTLs) in upland cotton (Gossypium hirsutum L.). Results showed that 159 Kinesin genes, including 15 genes of the Kinesin-13 gene subfamily, were identified in upland cotton; of which 157 Kinesin genes can be traced back to the diploid ancestors, G. raimondii and G. arboreum. Using a combined analysis of public QTLs and genome-wide expression profile information, there were 29 QTLs co-localized together with 28 Kinesin genes in upland cotton, including 10 Kinesin-13 subfamily genes. Genome-wide expression profile data indicated that, among the 28 co-localized genes, seven Kinesin genes were predominantly expressed in fibers or ovules. By association mapping analysis, 30 Kinesin genes were significantly associated with three fiber traits, among which a Kinesin-13 gene, Ghir_A11G028430, was found to be associated with both cotton boll length and lint weight, and one Kinesin-7 gene, Ghir_D04G017880 (Gh_Kinesin7), was significantly associated with fiber strength. In addition, two missense mutations were identified in the motor domain of the Gh_Kinesin7 protein. Overall, the Kinesin gene family seemingly plays an important role in cotton fiber and boll development. The exploited Kinesin genes will be beneficial for the genetic improvement of fiber quality and yield.
Collapse
Affiliation(s)
- Hong Zhu
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Jianzhong Xu
- Agriculture and Rural Affairs Bureau of Gao'an, Yichun, 330800, Jiangxi, China
| | - Kanbing Yu
- Xishuangbanna Institute of Agricultural Science, Xishuangbanna Autonomous Prefecture, 666100, Yunnan, China
| | - Jianfei Wu
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Huifang Xu
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Shubin Wang
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Tianwang Wen
- Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China.
| |
Collapse
|
22
|
Koenig AM, Liu B, Hu J. Visualizing the dynamics of plant energy organelles. Biochem Soc Trans 2023; 51:2029-2040. [PMID: 37975429 PMCID: PMC10754284 DOI: 10.1042/bst20221093] [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: 09/28/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Plant organelles predominantly rely on the actin cytoskeleton and the myosin motors for long-distance trafficking, while using microtubules and the kinesin motors mostly for short-range movement. The distribution and motility of organelles in the plant cell are fundamentally important to robust plant growth and defense. Chloroplasts, mitochondria, and peroxisomes are essential organelles in plants that function independently and coordinately during energy metabolism and other key metabolic processes. In response to developmental and environmental stimuli, these energy organelles modulate their metabolism, morphology, abundance, distribution and motility in the cell to meet the need of the plant. Consistent with their metabolic links in processes like photorespiration and fatty acid mobilization is the frequently observed inter-organellar physical interaction, sometimes through organelle membranous protrusions. The development of various organelle-specific fluorescent protein tags has allowed the simultaneous visualization of organelle movement in living plant cells by confocal microscopy. These energy organelles display an array of morphology and movement patterns and redistribute within the cell in response to changes such as varying light conditions, temperature fluctuations, ROS-inducible treatments, and during pollen tube development and immune response, independently or in association with one another. Although there are more reports on the mechanism of chloroplast movement than that of peroxisomes and mitochondria, our knowledge of how and why these three energy organelles move and distribute in the plant cell is still scarce at the functional and mechanistic level. It is critical to identify factors that control organelle motility coupled with plant growth, development, and stress response.
Collapse
Affiliation(s)
- Amanda M. Koenig
- Michigan State University-Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, MI, U.S.A
| | - Bo Liu
- Department of Plant Biology, University of California, Davis, CA, U.S.A
| | - Jianping Hu
- Michigan State University-Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, MI, U.S.A
| |
Collapse
|
23
|
Suárez-Baron H, Alzate JF, Ambrose BA, Pelaz S, González F, Pabón-Mora N. Comparative morphoanatomy and transcriptomic analyses reveal key factors controlling floral trichome development in Aristolochia (Aristolochiaceae). JOURNAL OF EXPERIMENTAL BOTANY 2023; 74:6588-6607. [PMID: 37656729 DOI: 10.1093/jxb/erad345] [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: 03/21/2023] [Accepted: 08/30/2023] [Indexed: 09/03/2023]
Abstract
Trichomes are specialized epidermal cells in aerial plant parts. Trichome development proceeds in three stages, determination of cell fate, specification, and morphogenesis. Most genes responsible for these processes have been identified in the unicellular branched leaf trichomes from the model Arabidopsis thaliana. Less is known about the molecular basis of multicellular trichome formation across flowering plants, especially those formed in floral organs of early diverging angiosperms. Here, we aim to identify the genetic regulatory network (GRN) underlying multicellular trichome development in the kettle-shaped trap flowers of Aristolochia (Aristolochiaceae). We selected two taxa for comparison, A. fimbriata, with trichomes inside the perianth, which play critical roles in pollination, and A. macrophylla, lacking specialized trichomes in the perianth. A detailed morphoanatomical characterization of floral epidermis is presented for the two species. We compared transcriptomic profiling at two different developmental stages in the different perianth portions (limb, tube, and utricle) of the two species. Moreover, we present a comprehensive expression map for positive regulators and repressors of trichome development, as well as cell cycle regulators. Our data point to extensive modifications in gene composition, expression, and putative roles in all functional categories when compared with model species. We also record novel differentially expressed genes (DEGs) linked to epidermis patterning and trichome development. We thus propose the first hypothetical genetic regulatory network (GRN) underlying floral multicellular trichome development in Aristolochia, and pinpoint key factors responsible for the presence and specialization of floral trichomes in phylogenetically distant species of the genus.
Collapse
Affiliation(s)
- Harold Suárez-Baron
- Department of Natural Sciences and Mathematics, Pontificia Universidad Javeriana Cali, Cali, Colombia
- Instituto de Biología, Universidad de Antioquia, Medellín, Colombia
| | - Juan F Alzate
- Centro Nacional de Secuenciación Genómica (CNSG), Sede de Investigación Universitaria, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | | | - Soraya Pelaz
- Centre for Research in Agricultural Genomics, CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Barcelona, Spain
- ICREA (Institució Catalana de Recerca i Estudis Avançats), Barcelona, Spain
| | - Favio González
- Universidad Nacional de Colombia, Sede Bogotá Facultad de Ciencias, Instituto de Ciencias Naturales, Bogotá, Colombia
| | | |
Collapse
|
24
|
Khoso MA, Zhang H, Khoso MH, Poudel TR, Wagan S, Papiashvili T, Saha S, Ali A, Murtaza G, Manghwar H, Liu F. Synergism of vesicle trafficking and cytoskeleton during regulation of plant growth and development: A mechanistic outlook. Heliyon 2023; 9:e21976. [PMID: 38034654 PMCID: PMC10682163 DOI: 10.1016/j.heliyon.2023.e21976] [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: 06/07/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
The cytoskeleton is a fundamental component found in all eukaryotic organisms, serving as a critical factor in various essential cyto-biological mechanisms, particularly in the locomotion and morphological transformations of plant cells. The cytoskeleton is comprised of three main components: microtubules (MT), microfilaments (MF), and intermediate filaments (IF). The cytoskeleton plays a crucial role in the process of cell wall formation and remodeling throughout the growth and development of cells. It is a highly organized and regulated network composed of filamentous components. In the basic processes of intracellular transport, such as mitosis, cytokinesis, and cell polarity, the plant cytoskeleton plays a crucial role according to recent studies. The major flaws in the organization of the cytoskeletal framework are at the root of the aberrant organogenesis currently observed in plant mutants. The regulation of protein compartmentalization and abundance within cells is predominantly governed by the process of vesicle/membrane transport, which plays a crucial role in several signaling cascades.The regulation of membrane transport in eukaryotic cells is governed by a diverse array of proteins. Recent developments in genomics have provided new tools to study the evolutionary relationships between membrane proteins in different plant species. It is known that members of the GTPases, COP, SNAREs, Rabs, tethering factors, and PIN families play essential roles in vesicle transport between plant, animal, and microbial species. This Review presents the latest research on the plant cytoskeleton, focusing on recent developments related to the cytoskeleton and summarizing the role of various proteins in vesicle transport. In addition, the report predicts future research direction of plant cytoskeleton and vesicle trafficking, potential research priorities, and provides researchers with specific pointers to further investigate the significant link between cytoskeleton and vesicle trafficking.
Collapse
Affiliation(s)
- Muneer Ahmed Khoso
- Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332000, China
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Department of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Hailong Zhang
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Department of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Mir Hassan Khoso
- Department of Biochemistry, Shaheed Mohtarma Benazir Bhutto Medical University Larkana, Pakistan
| | - Tika Ram Poudel
- Feline Research Center of National Forestry and Grassland Administration, College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Sindho Wagan
- Laboratory of Pest Physiology Biochemistry and Molecular Toxicology Department of Forest Protection Northeast Forestry University Harbin 150040, China
| | - Tamar Papiashvili
- School of Economics and Management Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Sudipta Saha
- School of Forestry, Department of Silviculture, Northeast Forestry University, Harbin 150040, China
| | - Abid Ali
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Department of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Ghulam Murtaza
- Department of Biochemistry and Molecular Biology Harbin Medical University China, China
| | - Hakim Manghwar
- Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332000, China
| | - Fen Liu
- Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332000, China
| |
Collapse
|
25
|
Marom R, Zhang B, Washington ME, Song IW, Burrage LC, Rossi VC, Berrier AS, Lindsey A, Lesinski J, Nonet ML, Chen J, Baldridge D, Silverman GA, Sutton VR, Rosenfeld JA, Tran AA, Hicks MJ, Murdock DR, Dai H, Weis M, Jhangiani SN, Muzny DM, Gibbs RA, Caswell R, Pottinger C, Cilliers D, Stals K, Eyre D, Krakow D, Schedl T, Pak SC, Lee BH. Dominant negative variants in KIF5B cause osteogenesis imperfecta via down regulation of mTOR signaling. PLoS Genet 2023; 19:e1011005. [PMID: 37934770 PMCID: PMC10656020 DOI: 10.1371/journal.pgen.1011005] [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: 05/29/2023] [Revised: 11/17/2023] [Accepted: 10/03/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Kinesin motor proteins transport intracellular cargo, including mRNA, proteins, and organelles. Pathogenic variants in kinesin-related genes have been implicated in neurodevelopmental disorders and skeletal dysplasias. We identified de novo, heterozygous variants in KIF5B, encoding a kinesin-1 subunit, in four individuals with osteogenesis imperfecta. The variants cluster within the highly conserved kinesin motor domain and are predicted to interfere with nucleotide binding, although the mechanistic consequences on cell signaling and function are unknown. METHODS To understand the in vivo genetic mechanism of KIF5B variants, we modeled the p.Thr87Ile variant that was found in two patients in the C. elegans ortholog, unc-116, at the corresponding position (Thr90Ile) by CRISPR/Cas9 editing and performed functional analysis. Next, we studied the cellular and molecular consequences of the recurrent p.Thr87Ile variant by microscopy, RNA and protein analysis in NIH3T3 cells, primary human fibroblasts and bone biopsy. RESULTS C. elegans heterozygous for the unc-116 Thr90Ile variant displayed abnormal body length and motility phenotypes that were suppressed by additional copies of the wild type allele, consistent with a dominant negative mechanism. Time-lapse imaging of GFP-tagged mitochondria showed defective mitochondria transport in unc-116 Thr90Ile neurons providing strong evidence for disrupted kinesin motor function. Microscopy studies in human cells showed dilated endoplasmic reticulum, multiple intracellular vacuoles, and abnormal distribution of the Golgi complex, supporting an intracellular trafficking defect. RNA sequencing, proteomic analysis, and bone immunohistochemistry demonstrated down regulation of the mTOR signaling pathway that was partially rescued with leucine supplementation in patient cells. CONCLUSION We report dominant negative variants in the KIF5B kinesin motor domain in individuals with osteogenesis imperfecta. This study expands the spectrum of kinesin-related disorders and identifies dysregulated signaling targets for KIF5B in skeletal development.
Collapse
Affiliation(s)
- Ronit Marom
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital, Houston, Texas, United States of America
| | - Bo Zhang
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Megan E. Washington
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - I-Wen Song
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lindsay C. Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital, Houston, Texas, United States of America
| | - Vittoria C. Rossi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital, Houston, Texas, United States of America
| | - Ava S. Berrier
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Anika Lindsey
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Jacob Lesinski
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Michael L. Nonet
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Jian Chen
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Dustin Baldridge
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Gary A. Silverman
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - V. Reid Sutton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital, Houston, Texas, United States of America
| | - Jill A. Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alyssa A. Tran
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - M. John Hicks
- Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - David R. Murdock
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hongzheng Dai
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - MaryAnn Weis
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Shalini N. Jhangiani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Donna M. Muzny
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Richard A. Gibbs
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Richard Caswell
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | - Carrie Pottinger
- All Wales Medical Genomics Service, Wrexham Maelor Hospital, Wrexham, UK
| | - Deirdre Cilliers
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Karen Stals
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | | | - David Eyre
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Deborah Krakow
- Human Genetics, Obstetrics & Gynecology, Orthopedic Surgery, University of California, Los Angeles, California, United States of America
| | - Tim Schedl
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Stephen C. Pak
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Brendan H. Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital, Houston, Texas, United States of America
| |
Collapse
|
26
|
Lin JJ, Lin CL, Chen CC, Lin YH, Cho DY, Chen X, Chen DC, Chen HY. Unlocking Colchicine's Untapped Potential: A Paradigm Shift in Hepatocellular Carcinoma Prevention. Cancers (Basel) 2023; 15:5031. [PMID: 37894398 PMCID: PMC10605746 DOI: 10.3390/cancers15205031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Background: Liver cancer and notably hepatocellular carcinoma (HCC), results in significantly high mortality rates worldwide. Chronic hepatitis and fatty liver, recognized precursors, underscore the imperative need for effective preventive strategies. This study explores colchicine, traditionally acknowledged for its anti-inflammatory properties and investigates its potential in liver cancer prevention. Methods: Utilizing the iHi Data Platform of China Medical University Hospital, Taiwan, this study analyzed two decades of medical data, incorporating 10,353 patients each in the Colchicine and Non-Colchicine cohorts, to investigate the association between colchicine use and liver cancer risk. Results: The study identified that colchicine users exhibited a 19% reduction in liver cancer risk, with a multivariable-adjusted odds ratio of 0.81 after accounting for confounding variables. Additionally, the influence of gender and comorbidities like diabetes mellitus on liver cancer risk was identified, corroborating the existing literature. A notable finding was that the prolonged use of colchicine was associated with improved outcomes, indicating a potential dose-response relationship. Conclusions: This study proposes a potential new role for colchicine in liver cancer prevention, extending beyond its established anti-inflammatory applications. While the findings are promising, further research is essential to validate these results. This research may serve as a foundation for future studies, aiming to further explore colchicine's role via clinical trials and in-depth investigations, potentially impacting preventive strategies for liver cancer.
Collapse
Affiliation(s)
- Jung-Ju Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan;
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung 404, Taiwan;
| | - Chun-Chung Chen
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; (C.-C.C.); (Y.-H.L.); (D.-Y.C.); (X.C.)
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 404, Taiwan
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 404, Taiwan
- Neuroscience and Brain Disease Center, China Medical University, Taichung 404, Taiwan
| | - Yu-Hsiang Lin
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; (C.-C.C.); (Y.-H.L.); (D.-Y.C.); (X.C.)
| | - Der-Yang Cho
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; (C.-C.C.); (Y.-H.L.); (D.-Y.C.); (X.C.)
| | - XianXiu Chen
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; (C.-C.C.); (Y.-H.L.); (D.-Y.C.); (X.C.)
- Neuroscience and Brain Disease Center, China Medical University, Taichung 404, Taiwan
| | - Der-Cherng Chen
- Department of Neurosurgery, China Medical University Hospital, Taichung 404, Taiwan; (C.-C.C.); (Y.-H.L.); (D.-Y.C.); (X.C.)
| | - Hung-Yao Chen
- School of Medicine, China Medical University, Taichung 404, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|
27
|
Zhao X, Chen T, Fu B, Fu Z, Xu K, Zhou W. Mutations obstructing ATP's emplacement in KIF2A nucleotide-binding pocket causes parenchymal malformations, motor developmental delay, with intellectual disability. Mol Genet Genomic Med 2023; 11:e2225. [PMID: 37331001 PMCID: PMC10568378 DOI: 10.1002/mgg3.2225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND KIF2A-related tubulinopathy (MIM: #615411) is a very rare disorder that was clinically characterized as microcephaly, epilepsy, motor developmental disorder (MDD), and various malformations of cortical development, but intellectual disability (ID) or global developmental delay (GDD) was rarely reported in the patients. METHODS Quad whole-exome sequencing (WES) was performed on the proband, the older brother, and their parents. Sanger sequencing was used to verify the candidate gene variant. RESULTS The proband, a 23-month-old boy, was previously diagnosed with GDD, and his brother, aged nine years, had ID; both were born to a healthy couple. Quad-WES detected a novel heterozygous KIF2A variant, c.1318G>A (p.G440R), in both the brothers but not in the parents. In-silico analysis revealed that the variants G440R and G318R (which were previously reported in the only reported patient with GDD) lead to markedly enlarged side chains and hinder ATP's emplacement in the NBD pocket. CONCLUSIONS The type of KIF2A variants that sterically hinder ATP emplacing in KIF2A NBD pocket may be associated with the intellectual disability phenotype; however, further studies are needed. Findings in this case also suggest a rare parental germline mosaicism of KIF2A G440R.
Collapse
Affiliation(s)
- Xiuying Zhao
- Department of Pediatricsthe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- Department of Children's RehabilitationHainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University)HaikouChina
| | - Tao Chen
- Department of NeurologyHainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University)HaikouChina
| | - Binsha Fu
- Department of Children's RehabilitationHainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University)HaikouChina
| | - Zhifu Fu
- Department of Children's RehabilitationHainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University)HaikouChina
| | - Kaishou Xu
- Department of RehabilitationGuangzhou Women and Children's Medical Center/National Children's Medical Center for South Central RegionGuangzhouChina
| | - Wei Zhou
- Department of Pediatricsthe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- Neonatal Intensive Care UnitGuangzhou Women and Children's Medical Center/National Children's Medical Center for South Central RegionGuangzhouChina
| |
Collapse
|
28
|
Sumbal S, Ali A, Nasser Binjawhar D, Ullah Z, Eldin SM, Iqbal R, Sher H, Ali I. Comparative Effects of Hydropriming and Iron Priming on Germination and Seedling Morphophysiological Attributes of Stay-Green Wheat. ACS OMEGA 2023; 8:23078-23088. [PMID: 37396271 PMCID: PMC10308549 DOI: 10.1021/acsomega.3c02359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/11/2023] [Indexed: 07/04/2023]
Abstract
Seed priming is considered to play an essential role in the overall improvement of agricultural crops. The current research work was carried out in order to investigate the comparative effects of hydropriming and iron priming on the germination behavior and morphophysiological attributes of wheat seedlings. The experimental materials consisted of three wheat genotypes including a synthetically derived wheat line (SD-194), stay-green wheat genotype (Chirya-7), and conventional wheat variety (Chakwal-50). The treatments included hydro (distilled and tap water)- and iron priming (10 and 50 mM) of wheat seeds for 12 h duration. Results indicated that both priming treatment and wheat genotypes exhibited highly different germination and seedling characteristics. These included germination percentage, root volume, root surface, root length, relative water content, chlorophyll content, membrane stability index, and chlorophyll fluorescence attributes. Furthermore, the synthetically derived line (SD-194) was the most promising in majority of the studied attributes by exhibiting a high germination index (2.21%), root fresh weight (7.76%), shoot dry weight (3.36%), relative water content (19.9%), chlorophyll content (7.58%), and photochemical quenching coefficient (2.58%) when compared with stay-green wheat (Chirya-7). The study also found that hydropriming with tap water and priming wheat seeds with low concentrations of iron yielded better results when a comparison was made with wheat seeds primed at high concentrations of iron. Therefore, wheat seed priming with tap water and iron solution for 12 h is recommended for optimum wheat improvement. Furthermore, current findings suggest that seed priming may have the prospect of an innovative and user-friendly approach for wheat biofortification with the aim of enhanced iron acquisition and accumulation in grains.
Collapse
Affiliation(s)
- Sumbal Sumbal
- Center
for Plant Sciences and Biodiversity, University
of Swat, Charbagh 19120, Pakistan
| | - Ahmad Ali
- Center
for Plant Sciences and Biodiversity, University
of Swat, Charbagh 19120, Pakistan
| | - Dalal Nasser Binjawhar
- Department
of Chemistry, College of Science, Princess
Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Zahid Ullah
- Center
for Plant Sciences and Biodiversity, University
of Swat, Charbagh 19120, Pakistan
| | - Sayed M. Eldin
- Center
of Research, Faculty of Engineering, Future
University in Egypt, New Cairo 18939, Egypt
| | - Rashid Iqbal
- Department
of Agronomy, Faculty of Agriculture and Environment, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Hassan Sher
- Center
for Plant Sciences and Biodiversity, University
of Swat, Charbagh 19120, Pakistan
| | - Iftikhar Ali
- Center
for Plant Sciences and Biodiversity, University
of Swat, Charbagh 19120, Pakistan
- Department
of Genetics and Development, Columbia University
Irving Medical Center, New York, New York 10032, United States
- School of
Life Sciences & Center of Novel Biomaterials, The Chinese University of Hong Kong, Hong Kong, SAR, China
| |
Collapse
|
29
|
Ilyas M, Ali I, Nasser Binjawhar D, Ullah S, Eldin SM, Ali B, Iqbal R, Bokhari SHA, Mahmood T. Molecular Characterization of Germin-like Protein Genes in Zea mays ( ZmGLPs) Using Various In Silico Approaches. ACS OMEGA 2023; 8:16327-16344. [PMID: 37179620 PMCID: PMC10173433 DOI: 10.1021/acsomega.3c01104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/10/2023] [Indexed: 05/15/2023]
Abstract
Germin (GER) and germin-like proteins (GLPs) play an important role in various plant processes. Zea mays contains 26 germin-like protein genes (ZmGLPs) located on chromosomes 2, 4, and 10; most of which are functionally unexplored. The present study aimed to characterize all ZmGLPs using the latest computational tools. All of them were studied at a physicochemical, subcellular, structural, and functional level, and their expression was predicted in plant development, against biotic and abiotic stresses using various in silico approaches. Overall, ZmGLPs showed greater similarity in their physicochemical properties, domain architecture, and structure, mostly localized in the cytoplasmic or extracellular regions. Phylogenetically, they have a narrow genetic background with a recent history of gene duplication events on chromosome 4. Functional analysis revealed novel enzymatic activities of phosphoglycolate phosphatase, adenosylhomocysteinase, phosphoglycolate phosphatase-like, osmotin/thaumatin-like, and acetohydroxy acid isomeroreductase largely mediated by disulfide bonding. Expression analysis revealed their crucial role in the root, root tips, crown root, elongation and maturation zones, radicle, and cortex with the highest expression being observed during germination and at the maturity levels. Further, ZmGLPs showed strong expression against biotic (Aspergillus flavus, Colletotrichum graminicola, Cercospora zeina, Fusarium verticillioides, and Fusarium virguliforme) while limited expression was noted against abiotic stresses. Concisely, our results provide a platform for additional functional exploration of the ZmGLP genes against various environmental stresses.
Collapse
Affiliation(s)
- Muhammad Ilyas
- Department
of Botany, Kohsar University Murree, Murree 19679, Punjab, Pakistan
| | - Iftikhar Ali
- Centre
for Plant Science and Biodiversity, University
of Swat, Charbagh 19120, Pakistan
- Department
of Genetics and Development, Columbia University
Irving Medical Center, New York, New York 10032, United States
| | - Dalal Nasser Binjawhar
- Department
of Chemistry, College of Science, Princess
Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Sami Ullah
- Department
of Forestry & Range Management, Kohsar
University Murree, Murree 19679, Pakistan
| | - Sayed M Eldin
- Center
of
Research, Faculty of Engineering, Future
University in Egypt, New Cairo 11835, Egypt
| | - Baber Ali
- Department
of Plant Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Rashid Iqbal
- Department
of Agronomy, Faculty of Agriculture and Environment, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Syed Habib Ali Bokhari
- Department
of Biosciences, CUI, Islamabad, Pakistan; Faculty of Biomedical and
Life Sciences, Kohsar University Murree, Murree 19679, Pakistan
| | - Tariq Mahmood
- Department
of Plant Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| |
Collapse
|
30
|
Zhou L, Xiong Y, Cooper L, Shepherd S, Song T, Dwivedy A, Rong L, Wang T, Cunningham BT, Wang X. Designer DNA NanoGripper. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538490. [PMID: 37162861 PMCID: PMC10168355 DOI: 10.1101/2023.04.26.538490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
DNA has shown great biocompatibility, programmable mechanical properties, and structural addressability at the nanometer scale, making it a versatile material for building high precision nanorobotics for biomedical applications. Herein, we present design principle, synthesis, and characterization of a DNA nanorobotic hand, called the "NanoGripper", that contains a palm and four bendable fingers as inspired by human hands, bird claws, and bacteriophages evolved in nature. Each NanoGripper finger has three phalanges connected by two flexible and rotatable joints that are bendable in response to binding to other entities. Functions of the NanoGripper have been enabled and driven by the interactions between moieties attached to the fingers and their binding partners. We showcase that the NanoGripper can be engineered to interact with and capture various objects with different dimensions, including gold nanoparticles, gold NanoUrchins, and SARS-CoV-2 virions. When carrying multiple DNA aptamer nanoswitches programmed to generate fluorescent signal enhanced on a photonic crystal platform, the NanoGripper functions as a sensitive viral biosensor that detects intact SARS-CoV-2 virions in human saliva with a limit of detection of ~ 100 copies/mL, providing RT-PCR equivalent sensitivity. Additionally, we use confocal microscopy to visualize how the NanoGripper-aptamer complex can effectively block viral entry into the host cells, indicating the viral inhibition. In summary, we report the design, synthesis, and characterization of a complex nanomachine that can be readily tailored for specific applications. The study highlights a path toward novel, feasible, and efficient solutions for the diagnosis and therapy of other diseases such as HIV and influenza.
Collapse
Affiliation(s)
- Lifeng Zhou
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yanyu Xiong
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Laura Cooper
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Skye Shepherd
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Tingjie Song
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Abhisek Dwivedy
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Tong Wang
- Advanced Science Research Center at Graduate Center, City University of New York, New York, NY 10031, USA
| | - Brian T. Cunningham
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Xing Wang
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Holonyak Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
31
|
eldesouky M, Abd-elazim A, Elhakim H, Fayed H. Impact of KIF6 Trp719Arg gene variant on Coronary Artery Disease Development.. [DOI: 10.21203/rs.3.rs-2705882/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Abstract
Coronary artery disease (CAD) is a multifactorial disease resulting from the interaction of genetic varia-tion and environmental traditional risk factors (TRFs), including diabetes mellitus, smoking, dyslipidemia, and hypertension. KIF6 Trp719Arg (rs20455; A>G) is an interesting gene variant reported as one of the most important risk factors for CAD in different populations. The study enrolled 150 participants belong-ing to the National Heart Institute (NHI) catheterization unit in Egypt, who were grouped into three main study groups regarding the presence of different TRFs. Biochemical investigations and clinical data were assessed and recorded. Analysis for KIF6 Trp719Arg polymorphism (rs20455; A>G) was performed for all participants using the TaqMan genotyping real-time PCR assay (rs20455). The study demonstrated that diabetes mellitus, hypertension, dyslipidemia, and smoking were highly statistically significant among CAD with TRF and non-CAD with TRF patients with p-values of 0.009*, 0.003*, 0.046*, and 0.001**, re-spectively. The family history of premature CAD represents a high percentage of CAD without TRF pa-tients compared to the other groups with a statistical difference of p-value= 0.004*. A high prevalence of AG+GG genotypes among the different groups was obtained, representing 66.0% of CAD with TRF, 76.0% of CAD without TRF, and 60% of non-CAD with TRF patients. The present study elucidated the impact of KIF6 Trp719Arg as a dependent risk factor for CAD, as it could have a significant role in CAD develop-ment when it interacts with one or more of the other traditional risk factors.
Collapse
|
32
|
Liu L, Li M, Zhang J, Xu D, Guo Y, Zhang H, Cang S. KIF14 mediates cabazitaxel-docetaxel cross-resistance in advanced prostate cancer by promoting AKT phosphorylation. Arch Biochem Biophys 2023; 737:109551. [PMID: 36822388 DOI: 10.1016/j.abb.2023.109551] [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: 09/20/2022] [Revised: 02/01/2023] [Accepted: 02/19/2023] [Indexed: 02/23/2023]
Abstract
Docetaxel is a first-line chemotherapy drug for castration-resistant prostate cancer (CRPC); yet, some CRPC patients develop docetaxel drug resistance. Cabazitaxel is approved in the post-docetaxel treatment setting. However, recent studies suggested cross-resistance between the development of drug resistance and current treatments. In this study, we used docetaxel-resistant cell lines DU145/DTX50 and PC-3/DTX30 to measure the responses to cabazitaxel. Our findings demonstrated that docetaxel resistance could lead to cross-resistance to cabazitaxel. After docetaxel-resistant cells were treated with cabazitaxel, transcriptome analysis was performed, and the results were analyzed in combination with survival analysis and correlation analysis with Gleason score to screen the cross-resistance genes. The continuously increased expression of kinesin family member 14 (KIF14) was identified as the main cause of cross-resistance to cabazitaxel in docetaxel-resistant cells. Silencing the expression of KIF14 could restore the sensitivity of resistant PCa cells to docetaxel and cabazitaxel, attenuate proliferation and promote apoptosis of the resistant PCa cells. Notably, the depressed expression of KIF14 inhibited the phosphorylation of Akt located downstream. In summary, KIF14 mediates the cross-resistance between docetaxel and cabazitaxel, and targeting KIF14 could be an effective measurement for reversing docetaxel or cabazitaxel chemotherapy failure or enhancing the anti-tumor effects of docetaxel or cabazitaxel.
Collapse
Affiliation(s)
- Lina Liu
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450053, Henan, China
| | - Mengyuan Li
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450053, Henan, China
| | - Junshuo Zhang
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450053, Henan, China
| | - Dengfei Xu
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450053, Henan, China
| | - Yan Guo
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450053, Henan, China
| | - Hui Zhang
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450053, Henan, China
| | - Shundong Cang
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450053, Henan, China.
| |
Collapse
|
33
|
Contrasting Metabolisms in Green and White Leaf Sectors of Variegated Pelargonium zonale—An Integrative Transcriptomic and Metabolomic Study. Int J Mol Sci 2023; 24:ijms24065288. [PMID: 36982362 PMCID: PMC10048803 DOI: 10.3390/ijms24065288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
The photosynthetically active green leaf (GL) and non-active white leaf (WL) tissues of variegated Pelargonium zonale provide an excellent model system for studying processes associated with photosynthesis and sink-source interactions, enabling the same microenvironmental conditions. By combining differential transcriptomics and metabolomics, we identified the main differences between these two metabolically contrasting tissues. Genes related to photosynthesis and associated pigments, the Calvin–Benson cycle, fermentation, and glycolysis were strongly repressed in WL. On the other hand, genes related to nitrogen and protein metabolism, defence, cytoskeletal components (motor proteins), cell division, DNA replication, repair and recombination, chromatin remodelling, and histone modifications were upregulated in WL. A content of soluble sugars, TCA intermediates, ascorbate, and hydroxybenzoic acids was lower, while the concentration of free amino acids (AAs), hydroxycinnamic acids, and several quercetin and kaempferol glycosides was higher in WL than in GL. Therefore, WL presents a carbon sink and depends on photosynthetic and energy-generating processes in GL. Furthermore, the upregulated nitrogen metabolism in WL compensates for the insufficient energy from carbon metabolism by providing alternative respiratory substrates. At the same time, WL serves as nitrogen storage. Overall, our study provides a new genetic data resource for the use of this excellent model system and for ornamental pelargonium breeding and contributes to uncovering molecular mechanisms underlying variegation and its adaptive ecological value.
Collapse
|
34
|
Zhang S, Tu Q, Qian X, Wang J, Ma C, Yang L, Liu Y, Wu R, Liu M. Deficiency of Kif15 gene inhibits tumor growth due to host CD8 +T lymphocytes increase. Biochem Biophys Res Commun 2023; 655:110-117. [PMID: 36934586 DOI: 10.1016/j.bbrc.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023]
Abstract
Kif15, also name kinesin-12, is a microtubule (MT) associate protein, which functions as a regulator of MT-dependent transport or spindle organization. Previous studies reported Kif15 increases in many tumors, however the effect of host Kif15 gene lack on tumor growth is not investigated. In this study, CRISPR/Cas9 mediated Kif15 gene knockout (Kif15-/-) mice were established and HE (Hematoxylin-Eosin) assay revealed no significant differences of morphology in most adult tissues (heart, liver, lung, kidney, and brain) except a retarded development of spleen in adult Kif15-/- mice. RNA sequence analysis of adult spleen tissues of Kif15-/- and Kif15+/+ mice was performed, and the results revealed that a total of 438 mRNAs were significantly differentially expressed in Kif15 knockout spleen, showing the top biological process was immune system process. FCM (Flow Cytometry) assay showed the percentage of CD8+ T lymphocytes notably increased in spleens of 9 w and 12 w old Kif15-/- mice. The CD8+ T lymphocytes are cytotoxic effector cells fighting against tumor. We thus detected the tumor growth in Kif15-/- mice using the melanoma cells inoculated subcutaneously. The tumor size significantly reduced in Kif15-/- mice. We finally detected whether Kif15 dysfunction affects the phagocytic function of macrophages on tumor cells, and the result showed Kif15 inhibitor treated macrophages significantly promoted the phagocytosis in vitro. In summary, this study revealed that the tumor-bearing mice of Kif15 gene deficiency notably inhibited tumor growth due to innate immune activation, which was the first report of the relation of Kif15 on the immunoreactivity.
Collapse
Affiliation(s)
- Siming Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China; Cancer Research Center Nantong, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Qifeng Tu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Xiaowei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Junpei Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Chao Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Liu Yang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Ronghua Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China.
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China.
| |
Collapse
|
35
|
Zhang J, An L, Zhao R, Shi R, Zhou X, Wei S, Zhang Q, Zhang T, Feng D, Yu Z, Wang H. KIF4A promotes genomic stability and progression of endometrial cancer through regulation of TPX2 protein degradation. Mol Carcinog 2023; 62:303-318. [PMID: 36468837 DOI: 10.1002/mc.23487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/07/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022]
Abstract
Kinesin family member 4A (KIF4A) belongs to the kinesin superfamily proteins, which are closely associated with mitophagy. Nonetheless, the role of KIF4A in endometrial cancer (EC) remains poorly characterized. The present study showed that KIF4A not only was upregulated but also predicted poor prognosis in patients with EC. KIF4A knockdown in EC cells resulted in attenuated proliferative capacity in vitro and in vivo. Transcriptome sequencing and gene function analysis revealed that KIF4A contributed to the maintenance of EC cells' genomic stability and that KIF4A knockdown induced the DNA damage response, cell cycle arrest, and apoptosis. Mechanistically, KIF4A interacted with TPX2 (a protein involved in DNA damage repair to cope with the replication pressure) to enhance its stability via inhibition of TPX2 ubiquitination and eventually ensured the genomic stability of EC cells during mitosis. Taken together, our results indicated that KIF4A functions as a tumor oncogene that facilitates EC progression via the maintenance of genomic stability. Therefore, targeting the KIF4A/TPX2 axis may provide new concepts and strategies for the treatment of patients with EC.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lanfen An
- Division of Life Science and Medicine, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Rong Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rui Shi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xing Zhou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sitian Wei
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tangansu Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dilu Feng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhicheng Yu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Clinical Research Center of Cancer Immunotherapy, Wuhan, Hubei, China
| |
Collapse
|
36
|
Cason SE, Fenton AR, Holzbaur ELF. Employing Live-Cell Imaging to Study Motor-Mediated Transport. Methods Mol Biol 2023; 2623:45-59. [PMID: 36602678 DOI: 10.1007/978-1-0716-2958-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Microtubule-based transport is a highly regulated process, requiring kinesin and/or dynein motors, a multitude of motor-associated regulatory proteins including activating adaptors and scaffolding proteins, and microtubule tracks that also provide regulatory cues. While in vitro studies are invaluable, fully replicating the physiological conditions under which motility occurs in cells is not yet possible. Here, we describe two methods that can be employed to study motor-based transport and motor regulation in a cellular context. Live-cell imaging of organelle transport in neurons leverages the uniform polarity of microtubules in axons to better understand the factors regulating microtubule-based motility. Peroxisome recruitment assays allow users to examine the net effect of motors and motor-regulatory proteins on organelle distribution. Together, these methods open the door to motility experiments that more fully interrogate the complex cellular environment.
Collapse
Affiliation(s)
- Sydney E Cason
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Adam R Fenton
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Chatterjee D, Beaulieu JM. Inhibition of glycogen synthase kinase 3 by lithium, a mechanism in search of specificity. Front Mol Neurosci 2022; 15:1028963. [PMID: 36504683 PMCID: PMC9731798 DOI: 10.3389/fnmol.2022.1028963] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/24/2022] [Indexed: 11/25/2022] Open
Abstract
Inhibition of Glycogen synthase kinase 3 (GSK3) is a popular explanation for the effects of lithium ions on mood regulation in bipolar disorder and other mental illnesses, including major depression, cyclothymia, and schizophrenia. Contribution of GSK3 is supported by evidence obtained from animal and patient derived model systems. However, the two GSK3 enzymes, GSK3α and GSK3β, have more than 100 validated substrates. They are thus central hubs for major biological functions, such as dopamine-glutamate neurotransmission, synaptic plasticity (Hebbian and homeostatic), inflammation, circadian regulation, protein synthesis, metabolism, inflammation, and mitochondrial functions. The intricate contributions of GSK3 to several biological processes make it difficult to identify specific mechanisms of mood stabilization for therapeutic development. Identification of GSK3 substrates involved in lithium therapeutic action is thus critical. We provide an overview of GSK3 biological functions and substrates for which there is evidence for a contribution to lithium effects. A particular focus is given to four of these: the transcription factor cAMP response element-binding protein (CREB), the RNA-binding protein FXR1, kinesin subunits, and the cytoskeletal regulator CRMP2. An overview of how co-regulation of these substrates may result in shared outcomes is also presented. Better understanding of how inhibition of GSK3 contributes to the therapeutic effects of lithium should allow for identification of more specific targets for future drug development. It may also provide a framework for the understanding of how lithium effects overlap with those of other drugs such as ketamine and antipsychotics, which also inhibit brain GSK3.
Collapse
Affiliation(s)
| | - Jean Martin Beaulieu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
38
|
Christensen JR, Reck-Peterson SL. Hitchhiking Across Kingdoms: Cotransport of Cargos in Fungal, Animal, and Plant Cells. Annu Rev Cell Dev Biol 2022; 38:155-178. [PMID: 35905769 PMCID: PMC10967659 DOI: 10.1146/annurev-cellbio-120420-104341] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Eukaryotic cells across the tree of life organize their subcellular components via intracellular transport mechanisms. In canonical transport, myosin, kinesin, and dynein motor proteins interact with cargos via adaptor proteins and move along filamentous actin or microtubule tracks. In contrast to this canonical mode, hitchhiking is a newly discovered mode of intracellular transport in which a cargo attaches itself to an already-motile cargo rather than directly associating with a motor protein itself. Many cargos including messenger RNAs, protein complexes, and organelles hitchhike on membrane-bound cargos. Hitchhiking-like behaviors have been shown to impact cellular processes including local protein translation, long-distance signaling, and organelle network reorganization. Here, we review instances of cargo hitchhiking in fungal, animal, and plant cells and discuss the potential cellular and evolutionary importance of hitchhiking in these different contexts.
Collapse
Affiliation(s)
- Jenna R Christensen
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA; ,
| | - Samara L Reck-Peterson
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA; ,
- Department of Biological Sciences, Cell and Developmental Biology Section, University of California, San Diego, La Jolla, California, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
39
|
Pasten MC, Carballo J, Gallardo J, Zappacosta D, Selva JP, Rodrigo JM, Echenique V, Garbus I. A combined transcriptome - miRNAome approach revealed that a kinesin gene is differentially targeted by a novel miRNA in an apomictic genotype of Eragrostis curvula. FRONTIERS IN PLANT SCIENCE 2022; 13:1012682. [PMID: 36247597 PMCID: PMC9563718 DOI: 10.3389/fpls.2022.1012682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/14/2022] [Indexed: 06/06/2023]
Abstract
Weeping lovegrass (Eragrostis curvula [Shrad.] Nees) is a perennial grass typically established in semi-arid regions, with good adaptability to dry conditions and sandy soils. This polymorphic complex includes both sexual and apomictic cytotypes, with different ploidy levels (2x-8x). Diploids are known to be sexual, while most polyploids are facultative apomicts, and full apomicts have also been reported. Plant breeding studies throughout the years have focused on achieving the introgression of apomixis into species of agricultural relevance, but, given the complexity of the trait, a deeper understanding of the molecular basis of regulatory mechanisms of apomixis is still required. Apomixis is thought to be associated with silencing or disruption of the sexual pathway, and studies have shown it is influenced by epigenetic mechanisms. In a previous study, we explored the role of miRNA-mRNA interactions using two contrasting E. curvula phenotypes. Here, the sexual OTA-S, the facultative Don Walter and the obligate apomictic Tanganyika cDNA and sRNA libraries were inquired, searching for miRNA discovery and miRNA expression regulation of genes related to the reproductive mode. This allowed for the characterization of seven miRNAs and the validation of their miRNA-target interactions. Interestingly, a kinesin gene was found to be repressed in the apomictic cultivar Tanganyika, targeted by a novel miRNA that was found to be overexpressed in this genotype, suggestive of an involvement in the reproductive mode expression. Our work provided additional evidence of the contribution of the epigenetic regulation of the apomictic pathway.
Collapse
Affiliation(s)
- María Cielo Pasten
- Centro de Recursos Naturales Renovables de la Zona Semiárida (CERZOS), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| | - José Carballo
- Centro de Recursos Naturales Renovables de la Zona Semiárida (CERZOS), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| | - Jimena Gallardo
- Centro de Recursos Naturales Renovables de la Zona Semiárida (CERZOS), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
- Departamento de Agronomía, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Diego Zappacosta
- Centro de Recursos Naturales Renovables de la Zona Semiárida (CERZOS), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
- Departamento de Agronomía, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Juan Pablo Selva
- Centro de Recursos Naturales Renovables de la Zona Semiárida (CERZOS), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Juan Manuel Rodrigo
- Centro de Recursos Naturales Renovables de la Zona Semiárida (CERZOS), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
- Departamento de Agronomía, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Viviana Echenique
- Centro de Recursos Naturales Renovables de la Zona Semiárida (CERZOS), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
- Departamento de Agronomía, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Ingrid Garbus
- Centro de Recursos Naturales Renovables de la Zona Semiárida (CERZOS), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| |
Collapse
|
40
|
Wang W, Todorov P, Pei C, Wang M, Isachenko E, Rahimi G, Mallmann P, Isachenko V. Epigenetic Alterations in Cryopreserved Human Spermatozoa: Suspected Potential Functional Defects. Cells 2022; 11:cells11132110. [PMID: 35805194 PMCID: PMC9266127 DOI: 10.3390/cells11132110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Gene set enrichment analysis (GSEA) was conducted on raw data, and alternative splicing (AS) events were found after mRNA sequencing of human spermatozoa. In this study, we aimed to compare unknown micro-epigenetics alternations in fresh and cryopreserved spermatozoa to evaluate the effectivity of cryopreservation protocols. Methods: Spermatozoa were divided into three groups: fresh spermatozoa (group 1), cryoprotectant-free vitrified spermatozoa (group 2), and conventionally frozen spermatozoa (group 3). Nine RNA samples (three replicates in each group) were detected and were used for library preparation with an Illumina compatible kit and sequencing by the Illumina platform. Results: Three Gene Ontology (GO) terms were found to be enriched in vitrified spermatozoa compared with fresh spermatozoa: mitochondrial tRNA aminoacylation, ATP-dependent microtubule motor activity, and male meiotic nuclear division. In alternative splicing analysis, a number of unknown AS events were found, including functional gene exon skipping (SE), alternative 5′ splice sites (A5SS), alternative 3′ splice sites (A3SS), mutually exclusive exon (MXE), and retained intron (RI). Conclusions: Cryopreservation of spermatozoa from some patients can agitate epigenetic instability, including increased alternative splicing events and changes in crucial mitochondrial functional activities. For fertilization of oocytes, for such patients, it is recommended to use fresh spermatozoa whenever possible; cryopreservation of sperm is recommended to be used only in uncontested situations.
Collapse
Affiliation(s)
- Wanxue Wang
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (W.W.); (C.P.); (M.W.); (E.I.); (G.R.); (P.M.)
| | - Plamen Todorov
- Institute of Biology and Immunology of Reproduction of Bulgarian Academy of Sciences, Tsarigradsko highway 73A, 1113 Sofia, Bulgaria;
| | - Cheng Pei
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (W.W.); (C.P.); (M.W.); (E.I.); (G.R.); (P.M.)
| | - Mengying Wang
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (W.W.); (C.P.); (M.W.); (E.I.); (G.R.); (P.M.)
| | - Evgenia Isachenko
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (W.W.); (C.P.); (M.W.); (E.I.); (G.R.); (P.M.)
| | - Gohar Rahimi
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (W.W.); (C.P.); (M.W.); (E.I.); (G.R.); (P.M.)
| | - Peter Mallmann
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (W.W.); (C.P.); (M.W.); (E.I.); (G.R.); (P.M.)
| | - Vladimir Isachenko
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (W.W.); (C.P.); (M.W.); (E.I.); (G.R.); (P.M.)
- Correspondence:
| |
Collapse
|
41
|
Cardoso AL, Venturelli NB, da Cruz I, de Sá Patroni FM, de Moraes D, de Oliveira RA, Benavente R, Martins C. Meiotic behavior, transmission and active genes of B chromosomes in the cichlid Astatotilapia latifasciata: new clues about nature, evolution and maintenance of accessory elements. Mol Genet Genomics 2022; 297:1151-1167. [PMID: 35704117 DOI: 10.1007/s00438-022-01911-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 05/24/2022] [Indexed: 12/23/2022]
Abstract
Supernumerary B chromosomes (Bs) are dispensable genetic elements widespread in eukaryotes and are poorly understood mainly in relation to mechanisms of maintenance and transmission. The cichlid Astatotilapia latifasciata can harbor Bs in a range of 0 (named B -) and 1-2 (named B +). The B in A. latifasciata is rich in several classes of repetitive DNA sequences, contains protein coding genes, and affects hosts in diverse ways, including sex-biased effects. To advance in the knowledge about the mechanisms of maintenance and transmission of B chromosomes in A. latifasciata, here, we studied the meiotic behavior in males and transmission rates of A. latifasciata B chromosome. We also analyzed structurally and functionally the predicted B chromosome copies of the cell cycle genes separin-like, tubb1-like and kif11-like. We identified in the meiotic structure relative to the B chromosome the presence of proteins associated with Synaptonemal Complex organization (SMC3, SYCP1 and SYCP3) and found that the B performs self-pairing. These data suggest that isochromosome formation was a step during B chromosome evolution and this element is in a stage of diversification of the two arms keeping the self-pairing behavior to protect the A chromosome complement of negative effects of recombination. Moreover, we observed no occurrence of B-drive and confirmed the presence of cell cycle genes copies in the B chromosome and their transcription in encephalon, muscle and gonads, which can indicates beneficial effects to hosts and contribute to B maintenance.
Collapse
Affiliation(s)
- Adauto Lima Cardoso
- Department of Structural and Functional Biology, Institute of Biosciences at Botucatu, Sao Paulo State University, UNESP, Botucatu, SP, 18618-689, Brazil
| | - Natália Bortholazzi Venturelli
- Department of Structural and Functional Biology, Institute of Biosciences at Botucatu, Sao Paulo State University, UNESP, Botucatu, SP, 18618-689, Brazil
| | - Irene da Cruz
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Fábio Malta de Sá Patroni
- Department of Structural and Functional Biology, Institute of Biosciences at Botucatu, Sao Paulo State University, UNESP, Botucatu, SP, 18618-689, Brazil
| | - Diogo de Moraes
- Department of Structural and Functional Biology, Institute of Biosciences at Botucatu, Sao Paulo State University, UNESP, Botucatu, SP, 18618-689, Brazil
| | - Rogério Antonio de Oliveira
- Department of Biostatistics, Plant Biology, Parasitology and Zoology, Institute of Biosciences at Botucatu, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Ricardo Benavente
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Cesar Martins
- Department of Structural and Functional Biology, Institute of Biosciences at Botucatu, Sao Paulo State University, UNESP, Botucatu, SP, 18618-689, Brazil.
| |
Collapse
|
42
|
Seo D, Gammon DB. Manipulation of Host Microtubule Networks by Viral Microtubule-Associated Proteins. Viruses 2022; 14:v14050979. [PMID: 35632720 PMCID: PMC9147350 DOI: 10.3390/v14050979] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/19/2022] Open
Abstract
Diverse DNA and RNA viruses utilize cytoskeletal networks to efficiently enter, replicate, and exit the host cell, while evading host immune responses. It is well established that the microtubule (MT) network is commonly hijacked by viruses to traffic to sites of replication after entry and to promote egress from the cell. However, mounting evidence suggests that the MT network is also a key regulator of host immune responses to infection. At the same time, viruses have acquired mechanisms to manipulate and/or usurp MT networks to evade these immune responses. Central to most interactions of viruses with the MT network are virally encoded microtubule-associated proteins (MAPs) that bind to MTs directly or indirectly. These MAPs associate with MTs and other viral or cellular MAPs to regulate various aspects of the MT network, including MT dynamics, MT-dependent transport via motor proteins such as kinesins and dyneins, and MT-dependent regulation of innate immune responses. In this review, we examine how viral MAP interactions with the MT network facilitate viral replication and immune evasion.
Collapse
|
43
|
Benowitz KM, Allan CW, Degain BA, Li X, Fabrick JA, Tabashnik BE, Carrière Y, Matzkin LM. Novel genetic basis of resistance to Bt toxin Cry1Ac in Helicoverpa zea. Genetics 2022; 221:iyac037. [PMID: 35234875 PMCID: PMC9071530 DOI: 10.1093/genetics/iyac037] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/25/2022] [Indexed: 11/14/2022] Open
Abstract
Crops genetically engineered to produce insecticidal proteins from the bacterium Bacillus thuringiensis have advanced pest management, but their benefits are diminished when pests evolve resistance. Elucidating the genetic basis of pest resistance to Bacillus thuringiensis toxins can improve resistance monitoring, resistance management, and the design of new insecticides. Here, we investigated the genetic basis of resistance to Bacillus thuringiensis toxin Cry1Ac in the lepidopteran Helicoverpa zea, one of the most damaging crop pests in the United States. To facilitate this research, we built the first chromosome-level genome assembly for this species, which has 31 chromosomes containing 375 Mb and 15,482 predicted proteins. Using a genome-wide association study, fine-scale mapping, and RNA-seq, we identified a 250-kb quantitative trait locus on chromosome 13 that was strongly associated with resistance in a strain of Helicoverpa zea that had been selected for resistance in the field and lab. The mutation in this quantitative trait locus contributed to but was not sufficient for resistance, which implies alleles in more than one gene contributed to resistance. This quantitative trait locus contains no genes with a previously reported role in resistance or susceptibility to Bacillus thuringiensis toxins. However, in resistant insects, this quantitative trait locus has a premature stop codon in a kinesin gene, which is a primary candidate as a mutation contributing to resistance. We found no changes in gene sequence or expression consistently associated with resistance for 11 genes previously implicated in lepidopteran resistance to Cry1Ac. Thus, the results reveal a novel and polygenic basis of resistance.
Collapse
Affiliation(s)
- Kyle M Benowitz
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA
- Department of Biology, Austin Peay State University, Clarksville, TN 37040, USA
| | - Carson W Allan
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA
| | - Benjamin A Degain
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA
| | - Xianchun Li
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA
| | - Jeffrey A Fabrick
- U.S. Department of Agriculture, Agricultural Research Service, U.S. Arid Land Agricultural Research Center, Maricopa, AZ 85138, USA
| | - Bruce E Tabashnik
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA
| | - Yves Carrière
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA
| | - Luciano M Matzkin
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ 85721, USA
- Bio5 Institute, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
44
|
Tati S, Alisaraie L. Recruitment of dynein and kinesin to viral particles. FASEB J 2022; 36:e22311. [DOI: 10.1096/fj.202101900rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/14/2022] [Accepted: 03/29/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Sayi’Mone Tati
- School of Pharmacy Memorial University of Newfoundland St. John’s Newfoundland Canada
| | - Laleh Alisaraie
- School of Pharmacy Memorial University of Newfoundland St. John’s Newfoundland Canada
| |
Collapse
|
45
|
Cocola C, Magnaghi V, Abeni E, Pelucchi P, Martino V, Vilardo L, Piscitelli E, Consiglio A, Grillo G, Mosca E, Gualtierotti R, Mazzaccaro D, La Sala G, Di Pietro C, Palizban M, Liuni S, DePedro G, Morara S, Nano G, Kehler J, Greve B, Noghero A, Marazziti D, Bussolino F, Bellipanni G, D'Agnano I, Götte M, Zucchi I, Reinbold R. Transmembrane Protein TMEM230, a Target of Glioblastoma Therapy. Front Cell Neurosci 2021; 15:703431. [PMID: 34867197 PMCID: PMC8636015 DOI: 10.3389/fncel.2021.703431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Abstract
Glioblastomas (GBM) are the most aggressive tumors originating in the brain. Histopathologic features include circuitous, disorganized, and highly permeable blood vessels with intermittent blood flow. These features contribute to the inability to direct therapeutic agents to tumor cells. Known targets for anti-angiogenic therapies provide minimal or no effect in overall survival of 12–15 months following diagnosis. Identification of novel targets therefore remains an important goal for effective treatment of highly vascularized tumors such as GBM. We previously demonstrated in zebrafish that a balanced level of expression of the transmembrane protein TMEM230/C20ORF30 was required to maintain normal blood vessel structural integrity and promote proper vessel network formation. To investigate whether TMEM230 has a role in the pathogenesis of GBM, we analyzed its prognostic value in patient tumor gene expression datasets and performed cell functional analysis. TMEM230 was found necessary for growth of U87-MG cells, a model of human GBM. Downregulation of TMEM230 resulted in loss of U87 migration, substratum adhesion, and re-passaging capacity. Conditioned media from U87 expressing endogenous TMEM230 induced sprouting and tubule-like structure formation of HUVECs. Moreover, TMEM230 promoted vascular mimicry-like behavior of U87 cells. Gene expression analysis of 702 patients identified that TMEM230 expression levels distinguished high from low grade gliomas. Transcriptomic analysis of patients with gliomas revealed molecular pathways consistent with properties observed in U87 cell assays. Within low grade gliomas, elevated TMEM230 expression levels correlated with reduced overall survival independent from tumor subtype. Highest level of TMEM230 correlated with glioblastoma and ATP-dependent microtubule kinesin motor activity, providing a direction for future therapeutic intervention. Our studies support that TMEM230 has both glial tumor and endothelial cell intracellular and extracellular functions. Elevated levels of TMEM230 promote glial tumor cell migration, extracellular scaffold remodeling, and hypervascularization and abnormal formation of blood vessels. Downregulation of TMEM230 expression may inhibit both low grade glioma and glioblastoma tumor progression and promote normalization of abnormally formed blood vessels. TMEM230 therefore is both a promising anticancer and antiangiogenic therapeutic target for inhibiting GBM tumor cells and tumor-driven angiogenesis.
Collapse
Affiliation(s)
- Cinzia Cocola
- Institute for Biomedical Technologies, National Research Council, Milan, Italy.,Consorzio Italbiotec, Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Edoardo Abeni
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Paride Pelucchi
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Valentina Martino
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Laura Vilardo
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Eleonora Piscitelli
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Arianna Consiglio
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Giorgio Grillo
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Ettore Mosca
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Roberta Gualtierotti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Mazzaccaro
- Operative Unit of Vascular Surgery, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Gina La Sala
- Institute of Biochemistry and Cell Biology, Italian National Research Council, Rome, Italy
| | - Chiara Di Pietro
- Institute of Biochemistry and Cell Biology, Italian National Research Council, Rome, Italy
| | - Mira Palizban
- Department of Gynecology and Obstetrics, University Hospital of Münster, Münster, Germany
| | - Sabino Liuni
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Giuseppina DePedro
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Giovanni Nano
- Operative Unit of Vascular Surgery, IRCCS Policlinico San Donato, San Donato Milanese, Italy.,Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - James Kehler
- National Institutes of Health, NIDDK, Laboratory of Cell and Molecular Biology, Bethesda, MD, United States
| | - Burkhard Greve
- Department of Radiation Therapy and Radiation Oncology, University Hospital of Münster, Münster, Germany
| | - Alessio Noghero
- Lovelace Biomedical Research Institute, Albuquerque, NM, United States.,Department of Oncology, University of Turin, Orbassano, Italy
| | - Daniela Marazziti
- Institute of Biochemistry and Cell Biology, Italian National Research Council, Rome, Italy
| | - Federico Bussolino
- Department of Oncology, University of Turin, Orbassano, Italy.,Laboratory of Vascular Oncology Candiolo Cancer Institute - IRCCS, Candiolo, Italy
| | - Gianfranco Bellipanni
- Department of Biology, Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United States
| | - Igea D'Agnano
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital of Münster, Münster, Germany
| | - Ileana Zucchi
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Rolland Reinbold
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| |
Collapse
|
46
|
Modeling Neurodevelopmental Disorders and Epilepsy Caused by Loss of Function of kif2a in Zebrafish. eNeuro 2021; 8:ENEURO.0055-21.2021. [PMID: 34404749 PMCID: PMC8425962 DOI: 10.1523/eneuro.0055-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 11/24/2022] Open
Abstract
In recent years there has been extensive research on malformations of cortical development (MCDs) that result in clinical features like developmental delay, intellectual disability, and drug-resistant epilepsy (DRE). Various studies highlighted the contribution of microtubule-associated genes (including tubulin and kinesin encoding genes) in MCD development. It has been reported that de novo mutations in KIF2A, a member of the kinesin-13 family, are linked to brain malformations and DRE. Although it is known that KIF2A functions by regulating microtubule depolymerization via an ATP-driven process, in vivo implications of KIF2A loss of function remain partly unclear. Here, we present a novel kif2a knock-out zebrafish model, showing hypoactivity, habituation deficits, pentylenetetrazole-induced seizure susceptibility and microcephaly, as well as neuronal cell proliferation defects and increased apoptosis. Interestingly, kif2a−/− larvae survived until adulthood and were fertile. Notably, our kif2a zebrafish knock-out model demonstrated many phenotypic similarities to KIF2A mouse models. This study provides valuable insights into the functional importance of kif2a in zebrafish and phenotypical hallmarks related to KIF2A mutations. Ultimately, this model could be used in a future search for more effective therapies that alleviate the clinical symptoms typically associated with MCDs.
Collapse
|
47
|
Hernandez-Toledano D, Vega L. The cytoskeleton as a non-cholinergic target of organophosphate compounds. Chem Biol Interact 2021; 346:109578. [PMID: 34265256 DOI: 10.1016/j.cbi.2021.109578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/19/2021] [Accepted: 07/12/2021] [Indexed: 12/29/2022]
Abstract
Current organophosphate (OP) toxicity research now considers potential non-cholinergic mechanisms for these compounds, since the inhibition of acetylcholinesterase (AChE) cannot completely explain all the adverse biological effects of OP. Thanks to the development of new strategies for OP detection, some potential molecular targets have been identified. Among these molecules are several cytoskeletal proteins, including actin, tubulin, intermediate filament proteins, and associated proteins, such as motor proteins, microtubule-associated proteins (MAPs), and cofilin. in vitro, ex vivo, and some in vivo reports have identified alterations in the cytoskeleton following OP exposure, including cell morphology defects, cells detachments, intracellular transport disruption, aberrant mitotic spindle formation, modification of cell motility, and reduced phagocytic capability, which implicate the cytoskeleton in OP toxicity. Here, we reviewed the evidence indicating the cytoskeletal targets of OP compounds, including their strategies, the potential effects of their alterations, and their possible participation in neurotoxicity, embryonic development, cell division, and immunotoxicity related to OP compounds exposure.
Collapse
Affiliation(s)
- David Hernandez-Toledano
- Department of Toxicology, Center for Research and Advanced Studies of the National Polytechnic Institute. Av. IPN 2508, San Pedro Zacatenco, C.P. 07360, Mexico City, Mexico
| | - Libia Vega
- Department of Toxicology, Center for Research and Advanced Studies of the National Polytechnic Institute. Av. IPN 2508, San Pedro Zacatenco, C.P. 07360, Mexico City, Mexico.
| |
Collapse
|
48
|
MicroRNA Omics Analysis of Camellia sinesis Pollen Tubes in Response to Low-Temperature and Nitric Oxide. Biomolecules 2021; 11:biom11070930. [PMID: 34201466 PMCID: PMC8301950 DOI: 10.3390/biom11070930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO) as a momentous signal molecule participates in plant reproductive development and responds to various abiotic stresses. Here, the inhibitory effects of the NO-dominated signal network on the pollen tube growth of Camellia sinensis under low temperature (LT) were studied by microRNA (miRNA) omics analysis. The results showed that 77 and 71 differentially expressed miRNAs (DEMs) were induced by LT and NO treatment, respectively. Gene ontology (GO) analysis showed that DEM target genes related to microtubules and actin were enriched uniquely under LT treatment, while DEM target genes related to redox process were enriched uniquely under NO treatment. In addition, the target genes of miRNA co-regulated by LT and NO are only located on the cell membrane and cell wall, and most of them are enriched in metal ion binding and/or transport and cell wall organization. Furthermore, DEM and its target genes related to metal ion binding/transport, redox process, actin, cell wall organization and carbohydrate metabolism were identified and quantified by functional analysis and qRT-PCR. In conclusion, miRNA omics analysis provides a complex signal network regulated by NO-mediated miRNA, which changes cell structure and component distribution by adjusting Ca2+ gradient, thus affecting the polar growth of the C. sinensis pollen tube tip under LT.
Collapse
|
49
|
Bonilla DA, Kreider RB, Stout JR, Forero DA, Kerksick CM, Roberts MD, Rawson ES. Metabolic Basis of Creatine in Health and Disease: A Bioinformatics-Assisted Review. Nutrients 2021; 13:nu13041238. [PMID: 33918657 PMCID: PMC8070484 DOI: 10.3390/nu13041238] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Creatine (Cr) is a ubiquitous molecule that is synthesized mainly in the liver, kidneys, and pancreas. Most of the Cr pool is found in tissues with high-energy demands. Cr enters target cells through a specific symporter called Na+/Cl−-dependent Cr transporter (CRT). Once within cells, creatine kinase (CK) catalyzes the reversible transphosphorylation reaction between [Mg2+:ATP4−]2− and Cr to produce phosphocreatine (PCr) and [Mg2+:ADP3−]−. We aimed to perform a comprehensive and bioinformatics-assisted review of the most recent research findings regarding Cr metabolism. Specifically, several public databases, repositories, and bioinformatics tools were utilized for this endeavor. Topics of biological complexity ranging from structural biology to cellular dynamics were addressed herein. In this sense, we sought to address certain pre-specified questions including: (i) What happens when creatine is transported into cells? (ii) How is the CK/PCr system involved in cellular bioenergetics? (iii) How is the CK/PCr system compartmentalized throughout the cell? (iv) What is the role of creatine amongst different tissues? and (v) What is the basis of creatine transport? Under the cellular allostasis paradigm, the CK/PCr system is physiologically essential for life (cell survival, growth, proliferation, differentiation, and migration/motility) by providing an evolutionary advantage for rapid, local, and temporal support of energy- and mechanical-dependent processes. Thus, we suggest the CK/PCr system acts as a dynamic biosensor based on chemo-mechanical energy transduction, which might explain why dysregulation in Cr metabolism contributes to a wide range of diseases besides the mitigating effect that Cr supplementation may have in some of these disease states.
Collapse
Affiliation(s)
- Diego A. Bonilla
- Research Division, Dynamical Business & Science Society–DBSS International SAS, Bogotá 110861, Colombia
- Research Group in Biochemistry and Molecular Biology, Universidad Distrital Francisco José de Caldas, Bogotá 110311, Colombia
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, Montería 230002, Colombia
- kDNA Genomics, Joxe Mari Korta Research Center, University of the Basque Country UPV/EHU, 20018 Donostia-San Sebastián, Spain
- Correspondence: ; Tel.: +57-320-335-2050
| | - Richard B. Kreider
- Exercise & Sport Nutrition Laboratory, Human Clinical Research Facility, Texas A&M University, College Station, TX 77843, USA;
| | - Jeffrey R. Stout
- Physiology of Work and Exercise Response (POWER) Laboratory, Institute of Exercise Physiology and Rehabilitation Science, University of Central Florida, Orlando, FL 32816, USA;
| | - Diego A. Forero
- Professional Program in Sport Training, School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá 111221, Colombia;
| | - Chad M. Kerksick
- Exercise and Performance Nutrition Laboratory, School of Health Sciences, Lindenwood University, Saint Charles, MO 63301, USA;
| | - Michael D. Roberts
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA;
- Edward via College of Osteopathic Medicine, Auburn, AL 36849, USA
| | - Eric S. Rawson
- Department of Health, Nutrition and Exercise Science, Messiah University, Mechanicsburg, PA 17055, USA;
| |
Collapse
|
50
|
Kamireddy K, Sonbarse PP, Mishra SK, Agrawal L, Chauhan PS, Lata C, Parvatam G. Proteomic approach to identify the differentially abundant proteins during flavour development in tuberous roots of Decalepis hamiltonii Wight & Arn. 3 Biotech 2021; 11:173. [PMID: 33927964 DOI: 10.1007/s13205-021-02714-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 03/03/2021] [Indexed: 01/14/2023] Open
Abstract
2-Hydroxy-4-Methoxy Benzaldehyde (2H4MB) is a structural isomer of vanillin produced in the tuberous roots of D. hamiltonii. Both vanillin and 2H4MB share the common phenylpropanoid pathway for their synthesis. Unlike vanillin, in which the biosynthetic pathway was well elucidated in V. planifolia, the 2H4MB biosynthetic pathway is not known in any of its plant sources. To find the key enzymes/proteins that promote 2H4MB biosynthesis, a comparative proteomic approach was adapted. In this case, two developmental stages of tuberous roots of D. hamiltonii were selected, where the flavour content was highly variable. The flavour content in the two stages was estimated using quantitative HPLC. The flavour content in the first and second stages of tuber development was 160 and 510 µgg-1, respectively. Two-dimensional electrophoresis (2-DE) was performed for these two stages of tubers; this was followed by PDquest analysis. A total of 180 protein spots were differentially abundant of which 57 spots were selected and subjected to MALDI-TOF-TOF analysis. The largest percentage of identified proteins was involved in stress and defence (27.9%), followed by proteins related to bioenergy and metabolism (23.2%), Cellular homeostasis proteins (18.6%), signaling proteins (11.6%), Plant growth and development proteins (9.3%). Holistically, we found the upregulation of methyltransferase, cell division responsive proteins, plant growth and development proteins which directly relate to flavour development and maturation. Similarly, stress-responsive and signaling proteins, vacuole proteins and ATPases were down-regulated with an increase in flavour content. In this study, we could not identify the specific 2H4MB metabolic pathway proteins, however, we could be able to study the changes in physiological and primary metabolic proteins with 2H4MB accumulation. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13205-021-02714-x.
Collapse
Affiliation(s)
- Kiran Kamireddy
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh India
- Plant Cell Biotechnology Department, CSIR - Central Food Technological Research Institute, Mysore, Karnataka India
| | - Priyanka Purushottam Sonbarse
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh India
- Plant Cell Biotechnology Department, CSIR - Central Food Technological Research Institute, Mysore, Karnataka India
| | - Shashank K Mishra
- CSIR-National Botanical Research Institute, Lucknow, Uttar Pradesh India
| | - Lalit Agrawal
- CSIR-National Botanical Research Institute, Lucknow, Uttar Pradesh India
| | - Puneet S Chauhan
- CSIR-National Botanical Research Institute, Lucknow, Uttar Pradesh India
| | - Charu Lata
- CSIR-National Botanical Research Institute, Lucknow, Uttar Pradesh India
| | - Giridhar Parvatam
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh India
- Plant Cell Biotechnology Department, CSIR - Central Food Technological Research Institute, Mysore, Karnataka India
| |
Collapse
|