1
|
Li D, Zheng P, Huang S. SLC12A9 is an immunological and prognostic biomarker for glioma. Gene 2025; 937:149136. [PMID: 39622394 DOI: 10.1016/j.gene.2024.149136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Glioma is one of the most common malignant brain tumors. It has a high rate of progression and a poor prognosis, and effective biomarkers still need to be identified. The solute carrier family 12 (SLC12) family has been reported to be involved in various physiological and pathological processes, but their functional roles in glioma remain unclear. METHODS Using public datasets, we studied the mutation and expression level of SLC12 family genes in glioma and identified the significantly differentially expressed member solute carrier family 12 member 9 (SLC12A9). We further predicted the prognostic role of SLC12A9 in glioma by using the Kaplan-Meier method and Cox regression analysis. Then, we performed biological functional enrichment analysis. We focused on the relationships between SLC12A9 expression and immune infiltration in glioma. Meanwhile, we conducted in vitro experiments to evaluate the effect of SLC12A9 expression on glioma cells. RESULTS Among the members of the SLC12 family, SLC12A9 had the highest mutation rate in glioma, with gene amplification as the major mutation type, and its expression was significantly upregulated in glioma. Higher SLC12A9 expression was significantly associated with older age, higher grade, wild-type isocitrate dehydrogenase (IDH), and a worse prognosis. The functional enrichment analysis indicated that SLC12A9 is mainly related to ion channel annotation. Gene set enrichment analysis (GSEA) revealed that SLC12A9 was mainly related to the DNA replication pathway. Furthermore, we found that SLC12A9 correlated with tumor-infiltrating immune cells and immune checkpoints. Thus, SLC12A9 may be involved in regulating the immune response of glioma. Finally, our in vitro experiments revealed that silencing SLC12A9 dramatically inhibited glioma cell growth and migration. CONCLUSIONS We showed that SLC12A9 may be a new predictive biomarker for glioma diagnosis, prognosis, and immunotherapy response, offering helpful guidelines to advance glioma treatment.
Collapse
Affiliation(s)
- Danting Li
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China; School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Peilin Zheng
- Department of General Practice, People's Hospital of Longhua, Shenzhen 518109, Guangdong, China.
| | - Shoujun Huang
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
2
|
Jeon S, Jeon Y, Lim JY, Kim Y, Cha B, Kim W. Emerging regulatory mechanisms and functions of biomolecular condensates: implications for therapeutic targets. Signal Transduct Target Ther 2025; 10:4. [PMID: 39757214 DOI: 10.1038/s41392-024-02070-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/01/2024] [Accepted: 11/06/2024] [Indexed: 01/07/2025] Open
Abstract
Cells orchestrate their processes through complex interactions, precisely organizing biomolecules in space and time. Recent discoveries have highlighted the crucial role of biomolecular condensates-membrane-less assemblies formed through the condensation of proteins, nucleic acids, and other molecules-in driving efficient and dynamic cellular processes. These condensates are integral to various physiological functions, such as gene expression and intracellular signal transduction, enabling rapid and finely tuned cellular responses. Their ability to regulate cellular signaling pathways is particularly significant, as it requires a careful balance between flexibility and precision. Disruption of this balance can lead to pathological conditions, including neurodegenerative diseases, cancer, and viral infections. Consequently, biomolecular condensates have emerged as promising therapeutic targets, with the potential to offer novel approaches to disease treatment. In this review, we present the recent insights into the regulatory mechanisms by which biomolecular condensates influence intracellular signaling pathways, their roles in health and disease, and potential strategies for modulating condensate dynamics as a therapeutic approach. Understanding these emerging principles may provide valuable directions for developing effective treatments targeting the aberrant behavior of biomolecular condensates in various diseases.
Collapse
Affiliation(s)
- Soyoung Jeon
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Yeram Jeon
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Ji-Youn Lim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Yujeong Kim
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Boksik Cha
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea.
| | - Wantae Kim
- Department of Life Science, University of Seoul, Seoul, South Korea.
| |
Collapse
|
3
|
Huang X, Ma Z, He D, Han X, Liu X, Dong Q, Tan C, Yu B, Sun T, Nordenskiöld L, Lu L, Miao Y, Hou X. Molecular condensation of the CO/NF-YB/NF-YC/FT complex gates floral transition in Arabidopsis. EMBO J 2025; 44:225-250. [PMID: 39567828 PMCID: PMC11696179 DOI: 10.1038/s44318-024-00293-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024] Open
Abstract
The plant master photoperiodic regulator CONSTANS (CO) interacts with Nuclear Factor-Y subunits B2 (NF-YB2) and C9 (NF-YC9) and transcriptionally activates the florigen gene FLOWERING LOCUS T (FT), regulating floral transition. However, the molecular mechanism of the functional four-component complex assembly in the nucleus remains elusive. We report that co-phase separation of CO with NF-YB2/NF-YC9/FT precisely controls heterogeneous CO assembly and FT transcriptional activation. In response to light signals, CO proteins form functional percolation clusters from a diffuse distribution in a B-box-motif-dependent manner. Multivalent coassembly with NF-YC9 and NF-YB2 prevents inhibitory condensate formation and is necessary to maintain proper CO assembly and material properties. The intrinsically disordered region (IDR) of NF-YC9, containing a polyglutamine motif, fine-tunes the functional properties of CO/NF-YB/NF-YC condensates. Specific FT promoter recognition with polyelectrolyte partitioning also enables the fluidic functional properties of CO/NF-YB/NF-YC/FT condensates. Our findings offer novel insights into the tunable macromolecular condensation of the CO/NF-YB/NF-YC/FT complex in controlling flowering in the photoperiod control.
Collapse
Affiliation(s)
- Xiang Huang
- Guangdong Provincial Key Laboratory of Applied Botany & State Key Laboratory of Plant Diversity and Specialty Crops, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Zhiming Ma
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Danxia He
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Xiao Han
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Xu Liu
- Guangdong Provincial Key Laboratory of Applied Botany & State Key Laboratory of Plant Diversity and Specialty Crops, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Qiong Dong
- Guangdong Provincial Key Laboratory of Applied Botany & State Key Laboratory of Plant Diversity and Specialty Crops, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Cuirong Tan
- Guangdong Provincial Key Laboratory of Applied Botany & State Key Laboratory of Plant Diversity and Specialty Crops, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Bin Yu
- Guangdong Provincial Key Laboratory of Applied Botany & State Key Laboratory of Plant Diversity and Specialty Crops, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Tiedong Sun
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Lars Nordenskiöld
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Lanyuan Lu
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Yansong Miao
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore, 636921, Singapore.
| | - Xingliang Hou
- Guangdong Provincial Key Laboratory of Applied Botany & State Key Laboratory of Plant Diversity and Specialty Crops, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China.
- University of the Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
4
|
Ni Q, Ge Z, Li Y, Shatkin G, Fu J, Sen A, Bera K, Yang Y, Wang Y, Wu Y, Nogueira Vasconcelos AC, Yan Y, Lin D, Feinberg AP, Konstantopoulos K, Sun SX. Cytoskeletal activation of NHE1 regulates mechanosensitive cell volume adaptation and proliferation. Cell Rep 2024; 43:114992. [PMID: 39579355 DOI: 10.1016/j.celrep.2024.114992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/24/2024] [Accepted: 11/04/2024] [Indexed: 11/25/2024] Open
Abstract
Mammalian cells rapidly respond to environmental changes by altering transmembrane water and ion fluxes, changing cell volume. Contractile forces generated by actomyosin have been proposed to mechanically regulate cell volume. However, our findings reveal a different mechanism in adherent cells, where elevated actomyosin activity increases cell volume in normal-like cells (NIH 3T3 and others) through interaction with the sodium-hydrogen exchanger isoform 1 (NHE1). This leads to a slow secondary volume increase (SVI) following the initial regulatory volume decrease during hypotonic shock. The active cell response is further confirmed by intracellular alkalinization during mechanical stretch. Moreover, cytoskeletal activation of NHE1 during SVI deforms the nucleus, causing immediate transcriptomic changes and ERK-dependent growth inhibition. Notably, SVI and its associated changes are absent in many cancer cell lines or cells on compliant substrates with reduced actomyosin activity. Thus, actomyosin acts as a sensory element rather than a force generator during adaptation to environmental challenges.
Collapse
Affiliation(s)
- Qin Ni
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhuoxu Ge
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yizeng Li
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
| | - Gabriel Shatkin
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jinyu Fu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Physics, Johns Hopkins University, Baltimore, MD, USA
| | - Anindya Sen
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Kaustav Bera
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yuhan Yang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yichen Wang
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yufei Wu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ana Carina Nogueira Vasconcelos
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yuqing Yan
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Dingchang Lin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew P Feinberg
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Konstantinos Konstantopoulos
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sean X Sun
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
5
|
Fu J, Ni Q, Wu Y, Gupta A, Ge Z, Yang H, Afrida Y, Barman I, Sun S. Cells Prioritize the Regulation of Cell Mass Density. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627803. [PMID: 39713365 PMCID: PMC11661194 DOI: 10.1101/2024.12.10.627803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
A cell's global physical state is characterized by its volume and dry mass. The ratio of cell mass to volume is the cell mass density (CMD), which is also a measure of macromolecular crowding and concentrations of all proteins. Using the Fluorescence eXclusion method (FXm) and Quantitative Phase Microscopy (QPM), we investigate CMD dynamics after exposure to sudden media osmolarity change. We find that while the cell volume and mass exhibit complex behavior after osmotic shock, CMD follows a straightforward monotonic recovery in 48 hours. The recovery is cell-cycle independent and relies on a coordinated adjustment of protein synthesis and volume growth rates. Surprisingly, we find that the protein synthesis rate decreases when CMD increases. This result is explained by CMD-dependent nucleoplasm-cytoplasm transport, which serves as negative regulatory feedback on CMD. The Na+/H+ exchanger NHE plays a role in regulating CMD by affecting both protein synthesis and volume change. Taken together, we reveal that cells possess a robust control system that actively regulates CMD during environmental change.
Collapse
|
6
|
Tikhomirova MA, Kuzmenko OL, Arifulin EA, Shirokova OM, Musinova YR, Sheval EV. The Nucleocapsid (N) Proteins of Different Human Coronaviruses Demonstrate a Variable Capacity to Induce the Formation of Cytoplasmic Condensates. Int J Mol Sci 2024; 25:13162. [PMID: 39684875 DOI: 10.3390/ijms252313162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
To date, seven human coronaviruses (HCoVs) have been identified. Four of these viruses typically manifest as a mild respiratory disease, whereas the remaining three can cause severe conditions that often result in death. The reasons for these differences remain poorly understood, but they may be related to the properties of individual viral proteins. The nucleocapsid (N) protein plays a crucial role in the packaging of viral genomic RNA and the modification of host cells during infection, in part due to its capacity to form dynamic biological condensates via liquid-liquid phase separation (LLPS). In this study, we investigated the capacity of N proteins derived from all HCoVs to form condensates when transiently expressed in cultured human cells. Some of the transfected cells were observed to contain cytoplasmic granules in which most of the N proteins were accumulated. Notably, the N proteins of SARS-CoV and SARS-CoV-2 showed a significantly reduced tendency to form cytoplasmic condensates. The condensate formation was not a consequence of overexpression of N proteins, as is typical for LLPS-inducing proteins. These condensates contained components of stress granules (SGs), indicating that the expression of N proteins caused the formation of SGs, which integrate N proteins. Thus, the N proteins of two closely related viruses, SARS-CoV and SARS-CoV-2, have the capacity to antagonize SG induction and/or assembly, in contrast to all other known HCoVs.
Collapse
Affiliation(s)
- Maria A Tikhomirova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Oleg L Kuzmenko
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Eugene A Arifulin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Olesya M Shirokova
- Central Scientific Research Laboratory, Institute of Fundamental Medicine, Privolzhsky Research Medical University, 603950 Nizhny Novgorod, Russia
| | - Yana R Musinova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Eugene V Sheval
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
7
|
Yang W, Wang Y, Liu G, Wang Y, Wu C. TPM4 condensates glycolytic enzymes and facilitates actin reorganization under hyperosmotic stress. Cell Discov 2024; 10:120. [PMID: 39622827 PMCID: PMC11612400 DOI: 10.1038/s41421-024-00744-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/20/2024] [Indexed: 12/06/2024] Open
Abstract
Actin homeostasis is fundamental for cell structure and consumes a large portion of cellular ATP. It has been documented in the literature that certain glycolytic enzymes can interact with actin, indicating an intricate interplay between the cytoskeleton and cellular metabolism. Here we report that hyperosmotic stress triggers actin severing and subsequent phase separation of the actin-binding protein tropomyosin 4 (TPM4). TPM4 condensates recruit glycolytic enzymes such as HK2, PFKM, and PKM2, while wetting actin filaments. Notably, the condensates of TPM4 and glycolytic enzymes are enriched of NADH and ATP, suggestive of their functional importance in cell metabolism. At cellular level, actin filament assembly is enhanced upon hyperosmotic stress and TPM4 condensation, while depletion of TPM4 impairs osmolarity-induced actin reorganization. At tissue level, colocalized condensates of TPM4 and glycolytic enzymes are observed in renal tissues subjected to hyperosmotic stress. Together, our findings suggest that stress-induced actin perturbation may act on TPM4 to organize glycolytic hubs that tether energy production to cytoskeletal reorganization.
Collapse
Affiliation(s)
- Wenzhong Yang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Yuan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Geyao Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Yan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- International Cancer Institute, Peking University, Beijing, China.
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China.
| |
Collapse
|
8
|
Zhou B, Min B, Liu W, Li Y, Zhu F, Huang J, Fang J, Chen Q, Wu D. Construction of a five-gene-based prognostic model for relapsed/refractory acute lymphoblastic leukemia. Hematology 2024; 29:2412952. [PMID: 39453390 DOI: 10.1080/16078454.2024.2412952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Relapsed/refractory acute lymphoblastic leukemia (R/R ALL) continues to be a major cause of mortality in children worldwide, with around 15% of ALL patients experiencing relapse and approximately 10% eventually dying from the disease. Early identification of R/R ALL in children has posed a longstanding clinical challenge. METHOD Genetic analysis of survival outcomes in pediatric patients with ALL from the TARGET-ALL dataset revealed five risk score factors identified through the intersection of differential genes (relapse/non-relapse) from the GSE17703 and GSE6092 databases. A risk score equation was formulated using these factors and validated against prognostic data from 46 ALL cases at our institution. Patients from multiple datasets were stratified into high and low-score groups based on this equation. Protein-protein interaction networks (PPI) were then constructed using the intersecting differential genes from all three datasets to identify hub nodes and predict interacting transcription factors. Additionally, genes related to cell pyroptosis with varying expression across these datasets were screened, and a multifactorial ROC curve (incorporating risk score and differential expression of pyroptosis-related genes) was generated. Furthermore, relationships among variables in the predictive model were depicted using a nomogram, and model efficacy was assessed through decision curve analysis (DCA). RESULTS By analyzing the TARGET-ALL, GSE17703, and GSE6092 databases, we developed a prognostic risk assessment model for pediatric ALL incorporating BAG2, EPHA4, FBXO9, SNX10, and WNK1. Validation of this model was conducted using data from 46 pediatric ALL cases obtained from our institution. Following the identification of 27 differentially expressed genes, we constructed a PPI and identified the top 10 hub genes (PTPRC, BTK, LCK, PRKCQ, CD3D, CD27, CD3G, BLNK, RASGRP1, VPREB1). Using this network, we predicted the top 5 transcription factors (HOXB4, MYC, SOX2, E2F1, NANOG). ROC and DCA were conducted on pyroptosis-related genes exhibiting differential expression and risk scores. Subsequently, a nomogram was generated, demonstrating the effectiveness of the risk score in predicting prognosis for pediatric ALL patients. CONCLUSIONS We have developed a risk prediction model for pediatric R/R ALL utilizing the genes BAG2, EPHA4, FBXO9, SNX10, and WNK1. This model provides a scientific foundation for early identification of R/R ALL in children.
Collapse
Affiliation(s)
- Bi Zhou
- Department of Pediatric, Suzhou Hospital of AnHui Medical University, Suzhou City, People's Republic of China
| | - BoJie Min
- Department of Pediatrics, the First Affiliated Hospital of AnHui Medical University, Hefei City, People's Republic of China
| | - WenYuan Liu
- Department of Pediatrics, The Second Affiliated Hospital of AnHui Medical University, Hefei City, People's Republic of China
| | - Ying Li
- Department of Pediatric, Suzhou Hospital of AnHui Medical University, Suzhou City, People's Republic of China
| | - Feng Zhu
- Department of Pediatric, Suzhou Hospital of AnHui Medical University, Suzhou City, People's Republic of China
| | - Jin Huang
- Department of Pediatric, Suzhou Hospital of AnHui Medical University, Suzhou City, People's Republic of China
| | - Jing Fang
- Graduate School, Bengbu Medical College, Bengbu City, People's Republic of China
| | - Qin Chen
- Department of Nursing, Suzhou Hospital of AnHui Medical University, Suzhou City, People's Republic of China
| | - De Wu
- Department of Pediatrics, the First Affiliated Hospital of AnHui Medical University, Hefei City, People's Republic of China
| |
Collapse
|
9
|
Zook E, Pan YE, Wipplinger A, Kerschbaum HH, Clements RJ, Ritter M, Stauber T, Model MA. Delayed vacuolation in mammalian cells caused by hypotonicity and ion loss. Sci Rep 2024; 14:29354. [PMID: 39592718 PMCID: PMC11599563 DOI: 10.1038/s41598-024-79815-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Prolonged exposure of mammalian cells to hypotonic environments stimulates the development of sometimes large and numerous vacuoles of unknown origin. Here, we investigate the nature and formation of these vacuoles, which we term LateVacs. Vacuolation starts after osmotic cell swelling has subsided and continues for many hours thereafter. Most of the vacuoles are positive for the lysosomal marker LAMP-1 but not for the autophagosomal marker LC3. Vacuoles do not appear to have acidic pH, as they exclude LysoTracker and acridine orange; inhibiting the V-ATPase with bafilomycin A1 has no effect on their formation. No LateVacs were formed in cells with a knockout of the essential LRRC8A subunit of the volume-regulated anion channel (VRAC). Since the main feature of cells recovered from hypotonic swelling should be reduced chloride concentration, we tested if chloride depletion can act as a signal for vacuolation. Indeed, four different low-chloride buffers resulted in the development of similar vacuoles. Moreover, vacuolation was suppressed in WNK1/WNK3 double knockouts or by the inhibition of WNK kinase, which is activated by low chloride; in hypotonic media, the WNK inhibitor had a similar effect. However, exposing cells to a low-sodium, high-potassium medium also resulted in vacuoles, which were insensitive to WNK. We conclude that vacuole development can be triggered either by the loss of chloride or by the loss of sodium.
Collapse
Affiliation(s)
- Emily Zook
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Yingzhou Edward Pan
- Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Anna Wipplinger
- Center for Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology, Paracelsus Medical University, Salzburg, Austria
| | - Hubert H Kerschbaum
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Robert J Clements
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Markus Ritter
- Center for Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology, Paracelsus Medical University, Salzburg, Austria
| | - Tobias Stauber
- Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany.
| | - Michael A Model
- Department of Biological Sciences, Kent State University, Kent, OH, USA.
| |
Collapse
|
10
|
Sharma Y, Lo R, Tomilin VN, Ha K, Deremo H, Pareek AV, Dong W, Liao X, Lebedeva S, Charu V, Kambham N, Mutig K, Pochynyuk O, Bhalla V. ClC-Kb pore mutation disrupts glycosylation and triggers distal tubular remodeling. JCI Insight 2024; 9:e175998. [PMID: 39405114 PMCID: PMC11601903 DOI: 10.1172/jci.insight.175998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/08/2024] [Indexed: 11/29/2024] Open
Abstract
Mutations in the CLCNKB gene (1p36), encoding the basolateral chloride channel ClC-Kb, cause type 3 Bartter syndrome. We identified a family with a mixed Bartter/Gitelman phenotype and early-onset kidney failure and by employing a candidate gene approach, identified what we believe is a novel homozygous mutation (CLCNKB c.499G>T [p.Gly167Cys]) in exon 6 of CLCNKB in the index patient. We then validated these results with Sanger and whole-exome sequencing. Compared with wild-type ClC-Kb, the Gly167Cys mutant conducted less current and exhibited impaired complex N-linked glycosylation in vitro. We demonstrated that loss of Gly-167, rather than gain of a mutant Cys, impairs complex glycosylation, but that surface expression remains intact. Moreover, Asn-364 was necessary for channel function and complex glycosylation. Morphologic evaluation of human kidney biopsies revealed typical basolateral localization of mutant Gly167Cys ClC-Kb in cortical distal tubular epithelia. However, we detected attenuated expression of distal sodium transport proteins, changes in abundance of distal tubule segments, and hypokalemia-associated intracellular condensates from the index patient compared with control nephrectomy specimens. The present data establish what we believe are novel regulatory mechanisms of ClC-Kb activity and demonstrate nephron remodeling in humans, caused by mutant ClC-Kb, with implications for renal electrolyte handling, blood pressure control, and kidney disease.
Collapse
Affiliation(s)
- Yogita Sharma
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Robin Lo
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Viktor N. Tomilin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kotdaji Ha
- Department of Physiology, UCSF, San Francisco, California, USA
| | - Holly Deremo
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Aishwarya V. Pareek
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Wuxing Dong
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Xiaohui Liao
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Svetlana Lebedeva
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vivek Charu
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Neeraja Kambham
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Kerim Mutig
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Translational Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
11
|
Li Y, Liu Y, Yu XY, Xu Y, Pan X, Sun Y, Wang Y, Song YH, Shen Z. Membraneless organelles in health and disease: exploring the molecular basis, physiological roles and pathological implications. Signal Transduct Target Ther 2024; 9:305. [PMID: 39551864 PMCID: PMC11570651 DOI: 10.1038/s41392-024-02013-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/22/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024] Open
Abstract
Once considered unconventional cellular structures, membraneless organelles (MLOs), cellular substructures involved in biological processes or pathways under physiological conditions, have emerged as central players in cellular dynamics and function. MLOs can be formed through liquid-liquid phase separation (LLPS), resulting in the creation of condensates. From neurodegenerative disorders, cardiovascular diseases, aging, and metabolism to cancer, the influence of MLOs on human health and disease extends widely. This review discusses the underlying mechanisms of LLPS, the biophysical properties that drive MLO formation, and their implications for cellular function. We highlight recent advances in understanding how the physicochemical environment, molecular interactions, and post-translational modifications regulate LLPS and MLO dynamics. This review offers an overview of the discovery and current understanding of MLOs and biomolecular condensate in physiological conditions and diseases. This article aims to deliver the latest insights on MLOs and LLPS by analyzing current research, highlighting their critical role in cellular organization. The discussion also covers the role of membrane-associated condensates in cell signaling, including those involving T-cell receptors, stress granules linked to lysosomes, and biomolecular condensates within the Golgi apparatus. Additionally, the potential of targeting LLPS in clinical settings is explored, highlighting promising avenues for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Yangxin Li
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
| | - Yuzhe Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, 130041, P. R. China
| | - Xi-Yong Yu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Yan Xu
- Department of General Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P. R. China
| | - Xiangbin Pan
- Department of Structural Heart Disease, National Center for Cardiovascular Disease, China & Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State key laboratory of cardiovascular disease, Beijing, 100037, P. R. China
| | - Yi Sun
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, 650102, P. R. China
| | - Yanli Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Yao-Hua Song
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P.R. China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
| |
Collapse
|
12
|
Zhao JZ, Xia J, Brangwynne CP. Chromatin compaction during confined cell migration induces and reshapes nuclear condensates. Nat Commun 2024; 15:9964. [PMID: 39557835 PMCID: PMC11574006 DOI: 10.1038/s41467-024-54120-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 11/01/2024] [Indexed: 11/20/2024] Open
Abstract
Cell migration through small constrictions during cancer metastasis requires significant deformation of the nucleus, with associated mechanical stress on the nuclear lamina and chromatin. However, how mechanical deformation impacts various subnuclear structures, including protein and nucleic acid-rich biomolecular condensates, is largely unknown. Here, we find that cell migration through confined spaces gives rise to mechanical deformations of the chromatin network, which cause embedded nuclear condensates, including nucleoli and nuclear speckles, to deform and coalesce. Chromatin deformations exhibit differential behavior in the advancing vs. trailing region of the nucleus, with the trailing half being more permissive for de novo condensate formation. We show that this results from increased chromatin heterogeneity, which gives rise to a shift in the binodal phase boundary. Taken together, our findings show how chromatin deformation impacts condensate assembly and properties, which can potentially contribute to cellular mechanosensing.
Collapse
Affiliation(s)
- Jessica Z Zhao
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Jing Xia
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Clifford P Brangwynne
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA.
- Princeton Materials Institute, Princeton University, Princeton, NJ, USA.
- Howard Hughes Medical Institute, Princeton University, Princeton, NJ, USA.
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
13
|
Wang Z, Yang Q, Zhang D, Lu Y, Wang Y, Pan Y, Qiu Y, Men Y, Yan W, Xiao Z, Sun R, Li W, Huang H, Guo H. A cytoplasmic osmosensing mechanism mediated by molecular crowding-sensitive DCP5. Science 2024; 386:eadk9067. [PMID: 39480925 DOI: 10.1126/science.adk9067] [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: 09/20/2023] [Revised: 06/10/2024] [Accepted: 09/09/2024] [Indexed: 11/02/2024]
Abstract
Plants are frequently challenged by osmotic stresses. How plant cells sense environmental osmolarity changes is not fully understood. We report that Arabidopsis Decapping 5 (DCP5) functions as a multifunctional cytoplasmic osmosensor that senses and responds to extracellular hyperosmolarity. DCP5 harbors a plant-specific intramolecular crowding sensor (ICS) that undergoes conformational change and drives phase separation in response to osmotically intensified molecular crowding. Upon hyperosmolarity exposure, DCP5 rapidly and reversibly assembles to DCP5-enriched osmotic stress granules (DOSGs), which sequestrate plenty of mRNA and regulatory proteins, and thus adaptively reprograms both the translatome and transcriptome to facilitate plant osmotic stress adaptation. Our findings uncover a cytoplasmic osmosensing mechanism mediated by DCP5 with plant-specific molecular crowding sensitivity and suggest a stress sensory function for hyperosmotically induced stress granules.
Collapse
Affiliation(s)
- Zhenyu Wang
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Qiuhua Yang
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Dan Zhang
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yuanyi Lu
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yichuan Wang
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yajie Pan
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yuping Qiu
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yongfan Men
- Research Laboratory of Biomedical Optics and Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Wei Yan
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zhina Xiao
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Ruixue Sun
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Wenyang Li
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Hongda Huang
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Hongwei Guo
- New Cornerstone Science Laboratory, Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
14
|
Wu A, Zhang Y, Bock F, Arroyo JP, Delpire E, Zhang MZ, Harris RC, Terker AS. Macrophage SPAK deletion limits a low potassium-induced kidney inflammatory program. Am J Physiol Renal Physiol 2024; 327:F899-F909. [PMID: 39298551 PMCID: PMC11563591 DOI: 10.1152/ajprenal.00175.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/28/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024] Open
Abstract
Inadequate dietary potassium (K+) consumption is a significant contributor to poor cardiovascular outcomes. A diet with reduced K+ content has been shown to cause salt-sensitive increases in blood pressure. More recently, we have also shown that reductions in blood K+ can cause direct kidney injury, independent of dietary sodium (Na+) content. Here, we investigated the role of the kinase Ste20p-related proline-alanine-rich kinase (SPAK) in this kidney injury response. We observed that global SPAK deletion protected the kidney from the damaging effects of a diet high in Na+ and low in K+. We hypothesized that kidney macrophages were contributing to the injury response and that macrophage-expressed SPAK is essential in this process. We observed SPAK protein expression in isolated macrophages in vitro. Culture in K+-deficient medium increased SPAK phosphorylation and caused SPAK to localize to cytosolic puncta, reminiscent of with-no-lysine kinase (WNK) bodies identified along the distal nephron epithelium. WNK1 also adopted a punctate staining pattern under low K+ conditions, and SPAK phosphorylation was prevented by treatment with the WNK inhibitor WNK463. Macrophage-specific SPAK deletion in vivo protected against the low K+-mediated renal inflammatory and fibrotic responses. Our results highlight an important role for macrophages and macrophage-expressed SPAK in the propagation of kidney damage that occurs in response to reduced dietary K+ consumption.NEW & NOTEWORTHY Global Ste20p-related proline alanine-rich kinase (SPAK) deletion protects against harmful kidney effects of dietary K+ deficiency. Exposure to low K+ conditions increases SPAK phosphorylation and induces SPAK to adopt a punctate staining pattern. Macrophage-specific deletion of SPAK confers protection to low K+-induced kidney injury in vivo. Macrophage-expressed SPAK plays a key role in the development of kidney injury in response to a low K+ diet.
Collapse
Grants
- DP5OD033412 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- DK135931 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- DK093501 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- DK134879 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- DK51265 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
Collapse
Affiliation(s)
- Aihua Wu
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, United States
| | - Yahua Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, United States
| | - Fabian Bock
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, United States
| | - Juan Pablo Arroyo
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, United States
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Ming-Zhi Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, United States
| | - Raymond C Harris
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, United States
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, United States
| | - Andrew S Terker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, United States
| |
Collapse
|
15
|
Gao J, Jing J, Shang G, Chen C, Duan M, Yu W, Wang K, Luo J, Song M, Chen K, Chen C, Zhang T, Ding D. TDRD1 phase separation drives intermitochondrial cement assembly to promote piRNA biogenesis and fertility. Dev Cell 2024; 59:2704-2718.e6. [PMID: 39029469 DOI: 10.1016/j.devcel.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/22/2024] [Accepted: 06/20/2024] [Indexed: 07/21/2024]
Abstract
The intermitochondrial cement (IMC) is a prominent germ granule that locates among clustered mitochondria in mammalian germ cells. Serving as a key platform for Piwi-interacting RNA (piRNA) biogenesis; however, how the IMC assembles among mitochondria remains elusive. Here, we identify that Tudor domain-containing 1 (TDRD1) triggers IMC assembly via phase separation. TDRD1 phase separation is driven by the cooperation of its tetramerized coiled-coil domain and dimethylarginine-binding Tudor domains but is independent of its intrinsically disordered region. TDRD1 is recruited to mitochondria by MILI and sequentially enhances mitochondrial clustering and triggers IMC assembly via phase separation to promote piRNA processing. TDRD1 phase separation deficiency in mice disrupts IMC assembly and piRNA biogenesis, leading to transposon de-repression and spermatogenic arrest. Moreover, TDRD1 phase separation is conserved in vertebrates but not in invertebrates. Collectively, our findings demonstrate a role of phase separation in germ granule formation and establish a link between membrane-bound organelles and membrane-less organelles.
Collapse
Affiliation(s)
- Jie Gao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jiongjie Jing
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200127, China
| | - Guanyi Shang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Canmei Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Maoping Duan
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Wenyang Yu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Ke Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jie Luo
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Manxiu Song
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Kun Chen
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200127, China
| | - Chen Chen
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Tuo Zhang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Deqiang Ding
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
16
|
Sabari BR, Hyman AA, Hnisz D. Functional specificity in biomolecular condensates revealed by genetic complementation. Nat Rev Genet 2024:10.1038/s41576-024-00780-4. [PMID: 39433596 DOI: 10.1038/s41576-024-00780-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2024] [Indexed: 10/23/2024]
Abstract
Biomolecular condensates are thought to create subcellular microenvironments that regulate specific biochemical activities. Extensive in vitro work has helped link condensate formation to a wide range of cellular processes, including gene expression, nuclear transport, signalling and stress responses. However, testing the relationship between condensate formation and function in cells is more challenging. In particular, the extent to which the cellular functions of condensates depend on the nature of the molecular interactions through which the condensates form is a major outstanding question. Here, we review results from recent genetic complementation experiments in cells, and highlight how genetic complementation provides important insights into cellular functions and functional specificity of biomolecular condensates. Combined with observations from human genetic disease, these experiments suggest that diverse condensate-promoting regions within cellular proteins confer different condensate compositions, biophysical properties and functions.
Collapse
Affiliation(s)
- Benjamin R Sabari
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Anthony A Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Denes Hnisz
- Max Planck Institute for Molecular Genetics, Berlin, Germany.
| |
Collapse
|
17
|
Dai Y, Zhou Z, Yu W, Ma Y, Kim K, Rivera N, Mohammed J, Lantelme E, Hsu-Kim H, Chilkoti A, You L. Biomolecular condensates regulate cellular electrochemical equilibria. Cell 2024; 187:5951-5966.e18. [PMID: 39260373 PMCID: PMC11490381 DOI: 10.1016/j.cell.2024.08.018] [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: 10/20/2023] [Revised: 05/22/2024] [Accepted: 08/09/2024] [Indexed: 09/13/2024]
Abstract
Control of the electrochemical environment in living cells is typically attributed to ion channels. Here, we show that the formation of biomolecular condensates can modulate the electrochemical environment in bacterial cells, which affects cellular processes globally. Condensate formation generates an electric potential gradient, which directly affects the electrochemical properties of a cell, including cytoplasmic pH and membrane potential. Condensate formation also amplifies cell-cell variability of their electrochemical properties due to passive environmental effect. The modulation of the electrochemical equilibria further controls cell-environment interactions, thus directly influencing bacterial survival under antibiotic stress. The condensate-mediated shift in intracellular electrochemical equilibria drives a change of the global gene expression profile. Our work reveals the biochemical functions of condensates, which extend beyond the functions of biomolecules driving and participating in condensate formation, and uncovers a role of condensates in regulating global cellular physiology.
Collapse
Affiliation(s)
- Yifan Dai
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Zhengqing Zhou
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Wen Yu
- Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Yuefeng Ma
- Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Kyeri Kim
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Nelson Rivera
- Department of Civil and Environmental Engineering, Duke University, Durham, NC 27708, USA
| | - Javid Mohammed
- Department of Immunology, Duke University, Durham, NC 27705, USA
| | - Erica Lantelme
- Department of Pathology and Immunology, Washington University in St. Louis, Saint Louis, MO 63110, USA
| | - Heileen Hsu-Kim
- Department of Civil and Environmental Engineering, Duke University, Durham, NC 27708, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Department of Immunology, Duke University, Durham, NC 27705, USA.
| | - Lingchong You
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Department of Immunology, Duke University, Durham, NC 27705, USA; Center for Quantitative Biodesign, Duke University, Durham, NC 27708, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
18
|
Navarro AM, Alonso M, Martínez-Pérez E, Lazar T, Gibson TJ, Iserte JA, Tompa P, Marino-Buslje C. Unveiling the Complexity of cis-Regulation Mechanisms in Kinases: A Comprehensive Analysis. Proteins 2024. [PMID: 39366918 DOI: 10.1002/prot.26751] [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: 04/26/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 10/06/2024]
Abstract
Protein cis-regulatory elements (CREs) are regions that modulate the activity of a protein through intramolecular interactions. Kinases, pivotal enzymes in numerous biological processes, often undergo regulatory control via inhibitory interactions in cis. This study delves into the mechanisms of cis regulation in kinases mediated by CREs, employing a combined structural and sequence analysis. To accomplish this, we curated an extensive dataset of kinases featuring annotated CREs, organized into homolog families through multiple sequence alignments. Key molecular attributes, including disorder and secondary structure content, active and ATP-binding sites, post-translational modifications, and disease-associated mutations, were systematically mapped onto all sequences. Additionally, we explored the potential for conformational changes between active and inactive states. Finally, we explored the presence of these kinases within membraneless organelles and elucidated their functional roles therein. CREs display a continuum of structures, ranging from short disordered stretches to fully folded domains. The adaptability demonstrated by CREs in achieving the common goal of kinase inhibition spans from direct autoinhibitory interaction with the active site within the kinase domain, to CREs binding to an alternative site, inducing allosteric regulation revealing distinct types of inhibitory mechanisms, which we exemplify by archetypical representative systems. While this study provides a systematic approach to comprehend kinase CREs, further experimental investigations are imperative to unravel the complexity within distinct kinase families. The insights gleaned from this research lay the foundation for future studies aiming to decipher the molecular basis of kinase dysregulation, and explore potential therapeutic interventions.
Collapse
Affiliation(s)
- Alvaro M Navarro
- Structural Bioinformatics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Macarena Alonso
- Structural Bioinformatics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina
| | | | - Tamas Lazar
- VIB-VUB Center for Structural Biology, Flanders Institute for Biotechnology (VIB), Brussels, Belgium
- Structural Biology Brussels, Department of Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
| | - Toby J Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Javier A Iserte
- Structural Bioinformatics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Peter Tompa
- VIB-VUB Center for Structural Biology, Flanders Institute for Biotechnology (VIB), Brussels, Belgium
- Structural Biology Brussels, Department of Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
- Research Centre for Natural Sciences, Hungarian Research Network, Institute of Enzymology, Budapest, Hungary
| | | |
Collapse
|
19
|
Posey AE, Bremer A, Erkamp NA, Pant A, Knowles TPJ, Dai Y, Mittag T, Pappu RV. Biomolecular Condensates are Characterized by Interphase Electric Potentials. J Am Chem Soc 2024. [PMID: 39356108 DOI: 10.1021/jacs.4c08946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Biomolecular condensates form via processes that combine phase separation and reversible associations of multivalent macromolecules. Condensates can be two- or multiphase systems defined by coexisting dense and dilute phases. Here, we show that solution ions partition asymmetrically across coexisting phases defined by condensates formed by intrinsically disordered proteins or homopolymeric RNA molecules. Our findings were enabled by direct measurements of the activities of cations and anions within coexisting phases of protein and RNA condensates. Asymmetries in ion partitioning between coexisting phases vary with protein sequence, macromolecular composition, salt concentration, and ion type. The Donnan equilibrium set up by the asymmetrical partitioning of solution ions generates interphase electric potentials known as Donnan and Nernst potentials. Our measurements show that the interphase potentials of condensates are of the same order of magnitude as membrane potentials of membrane-bound organelles. Interphase potentials quantify the degree to which microenvironments of coexisting phases are different from one another. Importantly, and based on condensate-specific interphase electric potentials, we reason that condensates are akin to capacitors that store charge. Interphase potentials should lead to electric double layers at condensate interfaces, thereby explaining recent observations of condensate interfaces being electrochemically active.
Collapse
Affiliation(s)
- Ammon E Posey
- Department of Biomedical Engineering, Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130-4899, United States
| | - Anne Bremer
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38103, United States
| | - Nadia A Erkamp
- Department of Biomedical Engineering, Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130-4899, United States
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Avnika Pant
- Department of Biomedical Engineering, Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130-4899, United States
| | - Tuomas P J Knowles
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
- Cavendish Laboratory, Department of Physics, University of Cambridge, J. J. Thomson Ave, Cambridge CB3 0HE, U.K
| | - Yifan Dai
- Department of Biomedical Engineering, Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130-4899, United States
| | - Tanja Mittag
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38103, United States
| | - Rohit V Pappu
- Department of Biomedical Engineering, Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130-4899, United States
| |
Collapse
|
20
|
Rodan AR. With No Lysine (K) Kinases and Sodium Transporter Function in Solute Exchange with Implications for BP Regulation as Elucidated through Drosophila. KIDNEY360 2024; 5:1553-1562. [PMID: 39186374 PMCID: PMC11556937 DOI: 10.34067/kid.0000000000000564] [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: 06/03/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Like other multicellular organisms, the fruit fly Drosophila melanogaster must maintain homeostasis of the internal milieu, including the maintenance of constant ion and water concentrations. In mammals, the with no lysine (K) (WNK)-Ste20-proline/alanine rich kinase/oxidative stress response 1 kinase cascade is an important regulator of epithelial ion transport in the kidney. This pathway regulates SLC12 family cotransporters, including sodium-potassium-2-chloride, sodium chloride, and potassium chloride cotransporters. The WNK-Ste20-proline/alanine rich kinase/oxidative stress response 1 kinase cascade also regulates epithelial ion transport via regulation of the Drosophila sodium-potassium-2-chloride cotransporter in the Malpighian tubule, the renal epithelium of the fly. Studies in Drosophila have contributed to the understanding of multiple regulators of WNK pathway signaling, including intracellular chloride and potassium, the scaffold protein Mo25, hypertonic stress, hydrostatic pressure, and macromolecular crowding. These will be discussed together, with implications for mammalian kidney function and BP control.
Collapse
Affiliation(s)
- Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah; Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, Utah; Department of Human Genetics, University of Utah, Salt Lake City, Utah; and Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| |
Collapse
|
21
|
Biswas P, Roy P, Jana S, Ray D, Das J, Chaudhuri B, Basunia RR, Sinha B, Sinha DK. Exploring the role of macromolecular crowding and TNFR1 in cell volume control. eLife 2024; 13:e92719. [PMID: 39297502 PMCID: PMC11581439 DOI: 10.7554/elife.92719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 09/18/2024] [Indexed: 11/22/2024] Open
Abstract
The excessive cosolute densities in the intracellular fluid create a physicochemical condition called macromolecular crowding (MMC). Intracellular MMC entropically maintains the biochemical thermodynamic equilibria by favoring associative reactions while hindering transport processes. Rapid cell volume shrinkage during extracellular hypertonicity elevates the MMC and disrupts the equilibria, potentially ushering cell death. Consequently, cells actively counter the hypertonic stress through regulatory volume increase (RVI) and restore the MMC homeostasis. Here, we establish fluorescence anisotropy of EGFP as a reliable tool for studying cellular MMC and explore the spatiotemporal dynamics of MMC during cell volume instabilities under multiple conditions. Our studies reveal that the actin cytoskeleton enforces spatially varying MMC levels inside adhered cells. Within cell populations, MMC is uncorrelated with nuclear DNA content but anti-correlated with the cell spread area. Although different cell lines have statistically similar MMC distributions, their responses to extracellular hypertonicity vary. The intensity of the extracellular hypertonicity determines a cell's ability for RVI, which correlates with nuclear factor kappa beta (NFkB) activation. Pharmacological inhibition and knockdown experiments reveal that tumor necrosis factor receptor 1 (TNFR1) initiates the hypertonicity-induced NFkB signaling and RVI. At severe hypertonicities, the elevated MMC amplifies cytoplasmic microviscosity and hinders receptor interacting protein kinase 1 (RIPK1) recruitment at the TNFR1 complex, incapacitating the TNFR1-NFkB signaling and consequently, RVI. Together, our studies unveil the involvement of TNFR1-NFkB signaling in modulating RVI and demonstrate the pivotal role of MMC in determining cellular osmoadaptability.
Collapse
Affiliation(s)
- Parijat Biswas
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Priyanka Roy
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Subhamoy Jana
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Dipanjan Ray
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Jibitesh Das
- Department of Biological Sciences, Indian Institute of Science Education and Research KolkataKolkataIndia
| | - Bipasa Chaudhuri
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Ridita Ray Basunia
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| | - Bidisha Sinha
- Department of Biological Sciences, Indian Institute of Science Education and Research KolkataKolkataIndia
| | - Deepak Kumar Sinha
- School of Biological Sciences, Indian Association for the Cultivation of ScienceKolkataIndia
| |
Collapse
|
22
|
Kim JS, Kehrl JH. Inhibition of WNK Kinases in NK Cells Disrupts Cellular Osmoregulation and Control of Tumor Metastasis. J Innate Immun 2024; 16:451-469. [PMID: 39265537 PMCID: PMC11521464 DOI: 10.1159/000540744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 08/01/2024] [Indexed: 09/14/2024] Open
Abstract
INTRODUCTION The serine/threonine with-no-lysine (WNK) kinase family function in blood pressure control, electrolyte homeostasis, and cellular osmoregulation. These kinases and their downstream effectors are considered promising therapeutic targets in hypertension and stroke. However, the role of WNK kinases in immune cells remains poorly understood. METHODS Using the small-molecule WNK kinase inhibitors WNK463 and WNK-IN-11, we investigated how WNK kinase inhibition affects natural killer (NK) cell physiology. RESULTS WNK kinase inhibition with WNK463 or WNK-IN-11 significantly decreased IL-2-activated NK cell volume, motility, and cytolytic activity. Treatment of NK cells with these inhibitors induced autophagy by activating AMPK and inhibiting mTOR signaling. Moreover, WNK kinase inhibition increased phosphorylation of Akt and c-Myc by misaligning activity of activating kinases and inhibitory phosphatases. Treatment of tumor-bearing mice with WNK463 impaired tumor metastasis control by adoptively transferred NK cells. CONCLUSION The catalytic activity of WNK kinases has a critical role of multiple aspects of NK cell physiology and their pharmacologic inhibition negatively impacts NK cell function.
Collapse
Affiliation(s)
- Ji Sung Kim
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John H Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
23
|
Boyd-Shiwarski CR, Shiwarski DJ, Subramanya AR. A New Phase for WNK Kinase Signaling Complexes as Biomolecular Condensates. Physiology (Bethesda) 2024; 39:0. [PMID: 38624245 PMCID: PMC11460533 DOI: 10.1152/physiol.00013.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
The purpose of this review is to highlight transformative advances that have been made in the field of biomolecular condensates, with special emphasis on condensate material properties, physiology, and kinases, using the With-No-Lysine (WNK) kinases as a prototypical example. To convey how WNK kinases illustrate important concepts for biomolecular condensates, we start with a brief history, focus on defining features of biomolecular condensates, and delve into some examples of how condensates are implicated in cellular physiology (and pathophysiology). We then highlight how WNK kinases, through the action of "WNK droplets" that ubiquitously regulate intracellular volume and kidney-specific "WNK bodies" that are implicated in distal tubule salt reabsorption and potassium homeostasis, exemplify many of the defining features of condensates. Finally, this review addresses the controversies within this emerging field and questions to address.
Collapse
Affiliation(s)
- Cary R Boyd-Shiwarski
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Daniel J Shiwarski
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Arohan R Subramanya
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
24
|
Li H, Wan L, Liu M, Ma E, Huang L, Yang Y, Li Q, Fang Y, Li J, Han B, Zhang C, Sun L, Hou X, Li H, Sun M, Qian S, Duan X, Zhao R, Yang X, Chen Y, Wu S, Zhang X, Zhang Y, Cheng G, Chen G, Gao Q, Xu J, Hou L, Wei C, Zhong H. SARS-CoV-2 spike-induced syncytia are senescent and contribute to exacerbated heart failure. PLoS Pathog 2024; 20:e1012291. [PMID: 39102426 PMCID: PMC11326701 DOI: 10.1371/journal.ppat.1012291] [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: 10/18/2023] [Revised: 08/15/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024] Open
Abstract
SARS-CoV-2 spike protein (SARS-2-S) induced cell-cell fusion in uninfected cells may occur in long COVID-19 syndrome, as circulating SARS-2-S or extracellular vesicles containing SARS-2-S (S-EVs) were found to be prevalent in post-acute sequelae of COVID-19 (PASC) for up to 12 months after diagnosis. Although isolated recombinant SARS-2-S protein has been shown to increase the SASP in senescent ACE2-expressing cells, the direct linkage of SARS-2-S syncytia with senescence in the absence of virus infection and the degree to which SARS-2-S syncytia affect pathology in the setting of cardiac dysfunction are unknown. Here, we found that the senescent outcome of SARS-2-S induced syncytia exacerbated heart failure progression. We first demonstrated that syncytium formation in cells expressing SARS-2-S delivered by DNA plasmid or LNP-mRNA exhibits a senescence-like phenotype. Extracellular vesicles containing SARS-2-S (S-EVs) also confer a potent ability to form senescent syncytia without de novo synthesis of SARS-2-S. However, it is important to note that currently approved COVID-19 mRNA vaccines do not induce syncytium formation or cellular senescence. Mechanistically, SARS-2-S syncytia provoke the formation of functional MAVS aggregates, which regulate the senescence fate of SARS-2-S syncytia by TNFα. We further demonstrate that senescent SARS-2-S syncytia exhibit shrinked morphology, leading to the activation of WNK1 and impaired cardiac metabolism. In pre-existing heart failure mice, the WNK1 inhibitor WNK463, anti-syncytial drug niclosamide, and senolytic dasatinib protect the heart from exacerbated heart failure triggered by SARS-2-S. Our findings thus suggest a potential mechanism for COVID-19-mediated cardiac pathology and recommend the application of WNK1 inhibitor for therapy especially in individuals with post-acute sequelae of COVID-19.
Collapse
Affiliation(s)
- Huilong Li
- Beijing Institute of Biotechnology, Beijing, China
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Luming Wan
- Beijing Institute of Biotechnology, Beijing, China
| | - Muyi Liu
- Beijing Institute of Biotechnology, Beijing, China
| | - Enhao Ma
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Linfei Huang
- Beijing Institute of Biotechnology, Beijing, China
| | - Yilong Yang
- Beijing Institute of Biotechnology, Beijing, China
| | - Qihong Li
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yi Fang
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jingfei Li
- Beijing Institute of Biotechnology, Beijing, China
| | - Bingqing Han
- Beijing Institute of Biotechnology, Beijing, China
| | - Chang Zhang
- Beijing Institute of Biotechnology, Beijing, China
| | - Lijuan Sun
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Xufeng Hou
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Haiyang Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mingyu Sun
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Sichong Qian
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xuejing Duan
- Department of Pathology, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, China
| | - Ruzhou Zhao
- Beijing Institute of Biotechnology, Beijing, China
| | - Xiaopan Yang
- Beijing Institute of Biotechnology, Beijing, China
| | - Yi Chen
- Beijing Institute of Biotechnology, Beijing, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Xuhui Zhang
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | | | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Gengye Chen
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qi Gao
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Junjie Xu
- Beijing Institute of Biotechnology, Beijing, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Congwen Wei
- Beijing Institute of Biotechnology, Beijing, China
| | - Hui Zhong
- Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
25
|
Yang F, Ding M, Song X, Chen F, Yang T, Wang C, Hu C, Hu Q, Yao Y, Du S, Yao PY, Xia P, Adams Jr G, Fu C, Xiang S, Liu D, Wang Z, Yuan K, Liu X. Organization of microtubule plus-end dynamics by phase separation in mitosis. J Mol Cell Biol 2024; 16:mjae006. [PMID: 38323478 PMCID: PMC11337005 DOI: 10.1093/jmcb/mjae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 08/17/2023] [Accepted: 02/05/2024] [Indexed: 02/08/2024] Open
Abstract
In eukaryotes, microtubule polymers are essential for cellular plasticity and fate decisions. End-binding (EB) proteins serve as scaffolds for orchestrating microtubule polymer dynamics and are essential for cellular dynamics and chromosome segregation in mitosis. Here, we show that EB1 forms molecular condensates with TIP150 and MCAK through liquid-liquid phase separation to compartmentalize the kinetochore-microtubule plus-end machinery, ensuring accurate kinetochore-microtubule interactions during chromosome segregation in mitosis. Perturbation of EB1-TIP150 polymer formation by a competing peptide prevents phase separation of the EB1-mediated complex and chromosome alignment at the metaphase equator in both cultured cells and Drosophila embryos. Lys220 of EB1 is dynamically acetylated by p300/CBP-associated factor in early mitosis, and persistent acetylation at Lys220 attenuates phase separation of the EB1-mediated complex, dissolves droplets in vitro, and harnesses accurate chromosome segregation. Our data suggest a novel framework for understanding the organization and regulation of eukaryotic spindle for accurate chromosome segregation in mitosis.
Collapse
Affiliation(s)
- Fengrui Yang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
- Keck Center for Organoids Plasticity, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Mingrui Ding
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
| | - Xiaoyu Song
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
- Keck Center for Organoids Plasticity, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Fang Chen
- Hunan Key Laboratory of Molecular Precision Medicine, Central South University, Changsha 410083, China
| | - Tongtong Yang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| | - Chunyue Wang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| | - Chengcheng Hu
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| | - Qing Hu
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| | - Yihan Yao
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
| | - Shihao Du
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| | - Phil Y Yao
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
- Keck Center for Organoids Plasticity, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Peng Xia
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| | - Gregory Adams Jr
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
- Keck Center for Organoids Plasticity, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Chuanhai Fu
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
- Keck Center for Organoids Plasticity, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Shengqi Xiang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
| | - Dan Liu
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
| | - Zhikai Wang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
- Keck Center for Organoids Plasticity, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Kai Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Central South University, Changsha 410083, China
| | - Xing Liu
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Cross-disciplinary Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| |
Collapse
|
26
|
Xiao YX, Lee SY, Aguilera-Uribe M, Samson R, Au A, Khanna Y, Liu Z, Cheng R, Aulakh K, Wei J, Farias AG, Reilly T, Birkadze S, Habsid A, Brown KR, Chan K, Mero P, Huang JQ, Billmann M, Rahman M, Myers C, Andrews BJ, Youn JY, Yip CM, Rotin D, Derry WB, Forman-Kay JD, Moses AM, Pritišanac I, Gingras AC, Moffat J. The TSC22D, WNK, and NRBP gene families exhibit functional buffering and evolved with Metazoa for cell volume regulation. Cell Rep 2024; 43:114417. [PMID: 38980795 DOI: 10.1016/j.celrep.2024.114417] [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: 02/22/2024] [Revised: 05/08/2024] [Accepted: 06/13/2024] [Indexed: 07/11/2024] Open
Abstract
The ability to sense and respond to osmotic fluctuations is critical for the maintenance of cellular integrity. We used gene co-essentiality analysis to identify an unappreciated relationship between TSC22D2, WNK1, and NRBP1 in regulating cell volume homeostasis. All of these genes have paralogs and are functionally buffered for osmo-sensing and cell volume control. Within seconds of hyperosmotic stress, TSC22D, WNK, and NRBP family members physically associate into biomolecular condensates, a process that is dependent on intrinsically disordered regions (IDRs). A close examination of these protein families across metazoans revealed that TSC22D genes evolved alongside a domain in NRBPs that specifically binds to TSC22D proteins, which we have termed NbrT (NRBP binding region with TSC22D), and this co-evolution is accompanied by rapid IDR length expansion in WNK-family kinases. Our study reveals that TSC22D, WNK, and NRBP genes evolved in metazoans to co-regulate rapid cell volume changes in response to osmolarity.
Collapse
Affiliation(s)
- Yu-Xi Xiao
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Seon Yong Lee
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Magali Aguilera-Uribe
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Reuben Samson
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, ON, Canada
| | - Aaron Au
- Institute for Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Yukti Khanna
- Otto-Loewi Research Center, Division of Medicinal Chemistry, Medical University of Graz, Neue Stiftingtalstrabe 6, 8010, Graz, Austria
| | - Zetao Liu
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Ran Cheng
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Kamaldeep Aulakh
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jiarun Wei
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Adrian Granda Farias
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Taylor Reilly
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Saba Birkadze
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Andrea Habsid
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Kevin R Brown
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Katherine Chan
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Patricia Mero
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jie Qi Huang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Maximilian Billmann
- Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Mahfuzur Rahman
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Chad Myers
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Brenda J Andrews
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Ji-Young Youn
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Christopher M Yip
- Institute for Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Daniela Rotin
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - W Brent Derry
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Julie D Forman-Kay
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada; Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Alan M Moses
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Iva Pritišanac
- Otto-Loewi Research Center, Division of Medicinal Chemistry, Medical University of Graz, Neue Stiftingtalstrabe 6, 8010, Graz, Austria
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, ON, Canada
| | - Jason Moffat
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Institute for Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
27
|
Yin T, He L, Du Y, Liu J, Peng L, Yang M, Sun S, Liu J, Li J, Cao J, Zhu H, Wang S. Macrophage WNK1 senses intracellular hypo-chlorine to regulate vulnerability to sepsis attack during hypochloremia. Int Immunopharmacol 2024; 139:112721. [PMID: 39033662 DOI: 10.1016/j.intimp.2024.112721] [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: 05/12/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Sepsis is one of the leading causes of death in critical patients worldwide and its occurrence is related to the excessive activation of macrophages. Chloride loss worsens the prognosis of patients with sepsis but the underlying mechanism is currently unclear. In this study, we founded that macrophages deficient in intracellular Cl- secrete more inflammatory cytokines such as IL-1β, IL-6 and TNF-α compared with control group. The intracellular chloride level decreased in WNK1 deficiency or activity inhibited macrophages with more severe inflammatory response after LPS treatment. Remimazolam, as classic GABAa receptor agonist, alleviates excessive inflammation cascade by promoting macrophage chloride influx during sepsis progression. Collectively, this study proves that macrophage WNK1 acts as a negative regulator of inflammatory response by sensing chloride to maintain intracellular chloride balance during sepsis coupled with hypochloremia.
Collapse
Affiliation(s)
- Tianyue Yin
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui 230001, China
| | - Lingwei He
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui 230001, China
| | - Yuhao Du
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Department of Anesthesiology, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
| | - Jiayuan Liu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui 230001, China
| | - Li Peng
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui 230001, China
| | - Mengmeng Yang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui 230001, China; Department of Anesthesiology, Anhui Provincial Hospital, Wannan Medical College, Hefei, Anhui 230001, China
| | - Shuaijie Sun
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui 230001, China; Department of Anesthesiology, Anhui Provincial Hospital, Wannan Medical College, Hefei, Anhui 230001, China
| | - Jingya Liu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Jun Li
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Jiangbing Cao
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui 230001, China; Department of Anesthesiology, Anhui Provincial Hospital, Wannan Medical College, Hefei, Anhui 230001, China
| | - Hongrui Zhu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Sheng Wang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
28
|
Zhou HX, Kota D, Qin S, Prasad R. Fundamental Aspects of Phase-Separated Biomolecular Condensates. Chem Rev 2024; 124:8550-8595. [PMID: 38885177 PMCID: PMC11260227 DOI: 10.1021/acs.chemrev.4c00138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Biomolecular condensates, formed through phase separation, are upending our understanding in much of molecular, cell, and developmental biology. There is an urgent need to elucidate the physicochemical foundations of the behaviors and properties of biomolecular condensates. Here we aim to fill this need by writing a comprehensive, critical, and accessible review on the fundamental aspects of phase-separated biomolecular condensates. We introduce the relevant theoretical background, present the theoretical basis for the computation and experimental measurement of condensate properties, and give mechanistic interpretations of condensate behaviors and properties in terms of interactions at the molecular and residue levels.
Collapse
Affiliation(s)
- Huan-Xiang Zhou
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, USA
- Department of Physics, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Divya Kota
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Sanbo Qin
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Ramesh Prasad
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
29
|
Posey AE, Bremer A, Erkamp NA, Pant A, Knowles TPJ, Dai Y, Mittag T, Pappu RV. Biomolecular condensates are characterized by interphase electric potentials. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601783. [PMID: 39005320 PMCID: PMC11245003 DOI: 10.1101/2024.07.02.601783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Biomolecular condensates form via processes that combine phase separation and reversible associations of multivalent macromolecules. Condensates can be two- or multi-phase systems defined by coexisting dense and dilute phases. Here, we show that solution ions can partition asymmetrically across coexisting phases defined by condensates formed by intrinsically disordered proteins or homopolymeric RNA molecules. Our findings were enabled by direct measurements of the activities of cations and anions within coexisting phases of protein and RNA condensates. Asymmetries in ion partitioning between coexisting phases vary with protein sequence, condensate type, salt concentration, and ion type. The Donnan equilibrium set up by asymmetrical partitioning of solution ions generates interphase electric potentials known as Donnan and Nernst potentials. Our measurements show that the interphase potentials of condensates are of the same order of magnitude as membrane potentials of membrane-bound organelles. Interphase potentials quantify the degree to which microenvironments of coexisting phases are different from one another. Importantly, and based on condensate-specific interphase electric potentials, which are membrane-like potentials of membraneless bodies, we reason that condensates are mesoscale capacitors that store charge. Interphase potentials lead to electric double layers at condensate interfaces. This helps explain recent observations of condensate interfaces being electrochemically active.
Collapse
|
30
|
Xie J, Huck WTS, Bao M. Unveiling the Intricate Connection: Cell Volume as a Key Regulator of Mechanotransduction. Annu Rev Biophys 2024; 53:299-317. [PMID: 38424091 DOI: 10.1146/annurev-biophys-030822-035656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The volumes of living cells undergo dynamic changes to maintain the cells' structural and functional integrity in many physiological processes. Minor fluctuations in cell volume can serve as intrinsic signals that play a crucial role in cell fate determination during mechanotransduction. In this review, we discuss the variability of cell volume and its role in vivo, along with an overview of the mechanisms governing cell volume regulation. Additionally, we provide insights into the current approaches used to control cell volume in vitro. Furthermore, we summarize the biological implications of cell volume regulation and discuss recent advances in understanding the fundamental relationship between cell volume and mechanotransduction. Finally, we delve into the potential underlying mechanisms, including intracellular macromolecular crowding and cellular mechanics, that govern the global regulation of cell fate in response to changes in cell volume. By exploring the intricate interplay between cell volume and mechanotransduction, we underscore the importance of considering cell volume as a fundamental signaling cue to unravel the basic principles of mechanotransduction. Additionally, we propose future research directions that can extend our current understanding of cell volume in mechanotransduction. Overall, this review highlights the significance of considering cell volume as a fundamental signal in understanding the basic principles in mechanotransduction and points out the possibility of controlling cell volume to control cell fate, mitigate disease-related damage, and facilitate the healing of damaged tissues.
Collapse
Affiliation(s)
- Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands;
| | - Min Bao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China;
| |
Collapse
|
31
|
Taylor CA, Jung JU, Kankanamalage SG, Li J, Grzemska M, Jaykumar AB, Earnest S, Stippec S, Saha P, Sauceda E, Cobb MH. Predictive and Experimental Motif Interaction Analysis Identifies Functions of the WNK-OSR1/SPAK Pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600905. [PMID: 38979344 PMCID: PMC11230372 DOI: 10.1101/2024.06.26.600905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The WNK-OSR1/SPAK protein kinase signaling pathway regulates ion homeostasis and cell volume, but its other functions are poorly understood. To uncover undefined signaling functions of the pathway we analyzed the binding specificity of the conserved C-terminal (CCT) domains of OSR1 and SPAK to find all possible interaction motifs in human proteins. These kinases bind the core consensus sequences R-F-x-V/I and R-x-F-x-V/I. Motifs were ranked based on sequence, conservation, cellular localization, and solvent accessibility. Out of nearly 3,700 motifs identified, 90% of previously published motifs were within the top 2% of those predicted. Selected candidates (TSC22D1, CAVIN1, ATG9A, NOS3, ARHGEF5) were tested. Upstream kinases WNKs 1-4 and their close relatives, the pseudokinases NRBP1/2, contain CCT-like domains as well. We identified additional distinct motif variants lacking the conserved arginine previously thought to be required, and found that the NRBP1 CCT-like domain binds TSC22D1 via the same motif as OSR1 and SPAK. Our results further highlight the rich and diverse functionality of CCT and CCT-like domains in connecting WNK signaling to cellular processes.
Collapse
|
32
|
Sakai K, Kondo Y, Goto Y, Aoki K. Cytoplasmic fluidization contributes to breaking spore dormancy in fission yeast. Proc Natl Acad Sci U S A 2024; 121:e2405553121. [PMID: 38889144 PMCID: PMC11214080 DOI: 10.1073/pnas.2405553121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/09/2024] [Indexed: 06/20/2024] Open
Abstract
The cytoplasm is a complex, crowded environment that influences myriad cellular processes including protein folding and metabolic reactions. Recent studies have suggested that changes in the biophysical properties of the cytoplasm play a key role in cellular homeostasis and adaptation. However, it still remains unclear how cells control their cytoplasmic properties in response to environmental cues. Here, we used fission yeast spores as a model system of dormant cells to elucidate the mechanisms underlying regulation of the cytoplasmic properties. By tracking fluorescent tracer particles, we found that particle mobility decreased in spores compared to vegetative cells and rapidly increased at the onset of dormancy breaking upon glucose addition. This cytoplasmic fluidization depended on glucose-sensing via the cyclic adenosine monophosphate-protein kinase A pathway. PKA activation led to trehalose degradation through trehalase Ntp1, thereby increasing particle mobility as the amount of trehalose decreased. In contrast, the rapid cytoplasmic fluidization did not require de novo protein synthesis, cytoskeletal dynamics, or cell volume increase. Furthermore, the measurement of diffusion coefficients with tracer particles of different sizes suggests that the spore cytoplasm impedes the movement of larger protein complexes (40 to 150 nm) such as ribosomes, while allowing free diffusion of smaller molecules (~3 nm) such as second messengers and signaling proteins. Our experiments have thus uncovered a series of signaling events that enable cells to quickly fluidize the cytoplasm at the onset of dormancy breaking.
Collapse
Affiliation(s)
- Keiichiro Sakai
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Myodaiji-cho, Okazaki, Aichi444-8787, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Myodaiji-cho, Okazaki, Aichi444-8787, Japan
| | - Yohei Kondo
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Myodaiji-cho, Okazaki, Aichi444-8787, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Myodaiji-cho, Okazaki, Aichi444-8787, Japan
- Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Myodaiji-cho, Okazaki, Aichi444-8787, Japan
- Division of Integrated Life Science, Department of Gene Mechanisms, Laboratory of Cell Cycle Regulation, Graduate School of Biostudies, Kyoto University, Kyoto606-8315, Japan
- Center for Living Systems Information Science, Graduate School of Biostudies, Kyoto University, Kyoto606-8315, Japan
| | - Yuhei Goto
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Myodaiji-cho, Okazaki, Aichi444-8787, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Myodaiji-cho, Okazaki, Aichi444-8787, Japan
- Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Myodaiji-cho, Okazaki, Aichi444-8787, Japan
- Division of Integrated Life Science, Department of Gene Mechanisms, Laboratory of Cell Cycle Regulation, Graduate School of Biostudies, Kyoto University, Kyoto606-8315, Japan
- Center for Living Systems Information Science, Graduate School of Biostudies, Kyoto University, Kyoto606-8315, Japan
| | - Kazuhiro Aoki
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Myodaiji-cho, Okazaki, Aichi444-8787, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Myodaiji-cho, Okazaki, Aichi444-8787, Japan
- Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Myodaiji-cho, Okazaki, Aichi444-8787, Japan
- Division of Integrated Life Science, Department of Gene Mechanisms, Laboratory of Cell Cycle Regulation, Graduate School of Biostudies, Kyoto University, Kyoto606-8315, Japan
- Center for Living Systems Information Science, Graduate School of Biostudies, Kyoto University, Kyoto606-8315, Japan
| |
Collapse
|
33
|
Zhang Y, Bock F, Ferdaus M, Arroyo JP, L Rose K, Patel P, Denton JS, Delpire E, Weinstein AM, Zhang MZ, Harris RC, Terker AS. Low potassium activation of proximal mTOR/AKT signaling is mediated by Kir4.2. Nat Commun 2024; 15:5144. [PMID: 38886379 PMCID: PMC11183202 DOI: 10.1038/s41467-024-49562-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
The renal epithelium is sensitive to changes in blood potassium (K+). We identify the basolateral K+ channel, Kir4.2, as a mediator of the proximal tubule response to K+ deficiency. Mice lacking Kir4.2 have a compensated baseline phenotype whereby they increase their distal transport burden to maintain homeostasis. Upon dietary K+ depletion, knockout animals decompensate as evidenced by increased urinary K+ excretion and development of a proximal renal tubular acidosis. Potassium wasting is not proximal in origin but is caused by higher ENaC activity and depends upon increased distal sodium delivery. Three-dimensional imaging reveals Kir4.2 knockouts fail to undergo proximal tubule expansion, while the distal convoluted tubule response is exaggerated. AKT signaling mediates the dietary K+ response, which is blunted in Kir4.2 knockouts. Lastly, we demonstrate in isolated tubules that AKT phosphorylation in response to low K+ depends upon mTORC2 activation by secondary changes in Cl- transport. Data support a proximal role for cell Cl- which, as it does along the distal nephron, responds to K+ changes to activate kinase signaling.
Collapse
Affiliation(s)
- Yahua Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Fabian Bock
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Mohammed Ferdaus
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Juan Pablo Arroyo
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Kristie L Rose
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Purvi Patel
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weil Medical College, New York, NY, USA
| | - Ming-Zhi Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Raymond C Harris
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Andrew S Terker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA.
| |
Collapse
|
34
|
Chen F, Li X, Guo W, Wang Y, Guo M, Shum HC. Size Scaling of Condensates in Multicomponent Phase Separation. J Am Chem Soc 2024; 146:16000-16009. [PMID: 38809420 DOI: 10.1021/jacs.4c02906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Constant proportionalities between cells and their intracellular organelles have been widely observed in various types of cells, known as intracellular size scaling. However, the mechanism underlying the size scaling and its modulation by environmental factors in multicomponent systems remain poorly understood. Here, we study the size scaling of membrane-less condensates using microdroplet-encapsulated minimalistic condensates formed by droplet microfluidics and mean-field theory. We demonstrate that the size scaling of condensates is an inherent characteristic of liquid-liquid phase separation. This concept is supported by experiments showing the occurrence of size scaling phenomena in various condensate systems and a generic lever rule acquired from mean-field theory. Moreover, it is found that the condensate-to-microdroplet scaling ratio can be affected by the solute and salt concentrations, with good agreement between experiments and predictions by theory. Notably, we identify a noise buffering mechanism whereby condensates composed of large macromolecules effectively maintain constant volumes and counteract concentration fluctuations of small molecules. This mechanism is achieved through the dynamic rearrangement of small molecules in and out of membrane-free interfaces. Our work provides crucial insights into understanding mechanistic principles that govern the size of cells and intracellular organelles as well as associated biological functions.
Collapse
Affiliation(s)
- Feipeng Chen
- Department of Mechanical Engineering, the University of Hong Kong, Pokfulam Road, Hong Kong (SAR) 999077, China
| | - Xiufeng Li
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong (SAR) 999077, China
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Wei Guo
- Department of Mechanical Engineering, the University of Hong Kong, Pokfulam Road, Hong Kong (SAR) 999077, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong (SAR) 999077, China
| | - Yuchao Wang
- Department of Mechanical Engineering, the University of Hong Kong, Pokfulam Road, Hong Kong (SAR) 999077, China
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Ho Cheung Shum
- Department of Mechanical Engineering, the University of Hong Kong, Pokfulam Road, Hong Kong (SAR) 999077, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong (SAR) 999077, China
| |
Collapse
|
35
|
Kok M, Brodsky JL. The biogenesis of potassium transporters: implications of disease-associated mutations. Crit Rev Biochem Mol Biol 2024; 59:154-198. [PMID: 38946646 PMCID: PMC11444911 DOI: 10.1080/10409238.2024.2369986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
The concentration of intracellular and extracellular potassium is tightly regulated due to the action of various ion transporters, channels, and pumps, which reside primarily in the kidney. Yet, potassium transporters and cotransporters play vital roles in all organs and cell types. Perhaps not surprisingly, defects in the biogenesis, function, and/or regulation of these proteins are linked to range of catastrophic human diseases, but to date, few drugs have been approved to treat these maladies. In this review, we discuss the structure, function, and activity of a group of potassium-chloride cotransporters, the KCCs, as well as the related sodium-potassium-chloride cotransporters, the NKCCs. Diseases associated with each of the four KCCs and two NKCCs are also discussed. Particular emphasis is placed on how these complex membrane proteins fold and mature in the endoplasmic reticulum, how non-native forms of the cotransporters are destroyed in the cell, and which cellular factors oversee their maturation and transport to the cell surface. When known, we also outline how the levels and activities of each cotransporter are regulated. Open questions in the field and avenues for future investigations are further outlined.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
36
|
Meneses-Reyes GI, Rodriguez-Bustos DL, Cuevas-Velazquez CL. Macromolecular crowding sensing during osmotic stress in plants. Trends Biochem Sci 2024; 49:480-493. [PMID: 38514274 DOI: 10.1016/j.tibs.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 03/23/2024]
Abstract
Osmotic stress conditions occur at multiple stages of plant life. Changes in water availability caused by osmotic stress induce alterations in the mechanical properties of the plasma membrane, its interaction with the cell wall, and the concentration of macromolecules in the cytoplasm. We summarize the reported players involved in the sensing mechanisms of osmotic stress in plants. We discuss how changes in macromolecular crowding are perceived intracellularly by intrinsically disordered regions (IDRs) in proteins. Finally, we review methods for dynamically monitoring macromolecular crowding in living cells and discuss why their implementation is required for the discovery of new plant osmosensors. Elucidating the osmosensing mechanisms will be essential for designing strategies to improve plant productivity in the face of climate change.
Collapse
Affiliation(s)
- G I Meneses-Reyes
- Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - D L Rodriguez-Bustos
- Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - C L Cuevas-Velazquez
- Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| |
Collapse
|
37
|
He Y, Yang X, Xia X, Wang Y, Dong Y, Wu L, Jiang P, Zhang X, Jiang C, Ma H, Ma W, Liu C, Whitford R, Tucker MR, Zhang Z, Li G. A phase-separated protein hub modulates resistance to Fusarium head blight in wheat. Cell Host Microbe 2024; 32:710-726.e10. [PMID: 38657607 DOI: 10.1016/j.chom.2024.04.002] [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: 07/12/2022] [Revised: 06/05/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
Fusarium head blight (FHB) is a devastating wheat disease. Fhb1, the most widely applied genetic locus for FHB resistance, is conferred by TaHRC of an unknown mode of action. Here, we show that TaHRC alleles distinctly drive liquid-liquid phase separation (LLPS) within a proteinaceous complex, determining FHB susceptibility or resistance. TaHRC-S (susceptible) exhibits stronger LLPS ability than TaHRC-R (resistant), and this distinction is further intensified by fungal mycotoxin deoxynivalenol, leading to opposing FHB symptoms. TaHRC recruits a protein class with intrinsic LLPS potentials, referred to as an "HRC-containing hub." TaHRC-S drives condensation of hub components, while TaHRC-R comparatively suppresses hub condensate formation. The function of TaSR45a splicing factor, a hub member, depends on TaHRC-driven condensate state, which in turn differentially directs alternative splicing, switching between susceptibility and resistance to wheat FHB. These findings reveal a mechanism for FHB spread within a spike and shed light on the roles of complex condensates in controlling plant disease.
Collapse
Affiliation(s)
- Yi He
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China; CIMMYT-JAAS Joint Center for Wheat Diseases, The Research Center of Wheat Scab, Zhongshan Biological Breeding Laboratory, Key Laboratory of Germplasm Innovation in Downstream of Huaihe River (Nanjing), Ministry of Agriculture and Rural Affairs, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Xiujuan Yang
- Waite Research Institute, School of Agriculture, Food and Wine, The University of Adelaide, Urrbrae, SA 5064, Australia
| | - Xiaobo Xia
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuhua Wang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Yifan Dong
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Lei Wu
- CIMMYT-JAAS Joint Center for Wheat Diseases, The Research Center of Wheat Scab, Zhongshan Biological Breeding Laboratory, Key Laboratory of Germplasm Innovation in Downstream of Huaihe River (Nanjing), Ministry of Agriculture and Rural Affairs, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Peng Jiang
- CIMMYT-JAAS Joint Center for Wheat Diseases, The Research Center of Wheat Scab, Zhongshan Biological Breeding Laboratory, Key Laboratory of Germplasm Innovation in Downstream of Huaihe River (Nanjing), Ministry of Agriculture and Rural Affairs, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Xu Zhang
- CIMMYT-JAAS Joint Center for Wheat Diseases, The Research Center of Wheat Scab, Zhongshan Biological Breeding Laboratory, Key Laboratory of Germplasm Innovation in Downstream of Huaihe River (Nanjing), Ministry of Agriculture and Rural Affairs, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Cong Jiang
- College of Plant Protection, Northwest A&F University, Yangling 712100, China
| | - Hongxiang Ma
- College of Agriculture, Yangzhou University, Yangzhou 225009, China
| | - Wujun Ma
- College of Agronomy, Qingdao Agricultural University, Qingdao 266000, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Ryan Whitford
- Centre for Crop and Food Innovation (CCFI), State Agricultural Biotechnology Centre (SABC), Food Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Matthew R Tucker
- Waite Research Institute, School of Agriculture, Food and Wine, The University of Adelaide, Urrbrae, SA 5064, Australia
| | - Zhengguang Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Gang Li
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
38
|
Li Y, Feng Y, He Q, Ni Z, Hu X, Feng X, Ni M. The predictive accuracy of machine learning for the risk of death in HIV patients: a systematic review and meta-analysis. BMC Infect Dis 2024; 24:474. [PMID: 38711068 PMCID: PMC11075245 DOI: 10.1186/s12879-024-09368-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/30/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Early prediction of mortality in individuals with HIV (PWH) has perpetually posed a formidable challenge. With the widespread integration of machine learning into clinical practice, some researchers endeavor to formulate models predicting the mortality risk for PWH. Nevertheless, the diverse timeframes of mortality among PWH and the potential multitude of modeling variables have cast doubt on the efficacy of the current predictive model for HIV-related deaths. To address this, we undertook a systematic review and meta-analysis, aiming to comprehensively assess the utilization of machine learning in the early prediction of HIV-related deaths and furnish evidence-based support for the advancement of artificial intelligence in this domain. METHODS We systematically combed through the PubMed, Cochrane, Embase, and Web of Science databases on November 25, 2023. To evaluate the bias risk in the original studies included, we employed the Predictive Model Bias Risk Assessment Tool (PROBAST). During the meta-analysis, we conducted subgroup analysis based on survival and non-survival models. Additionally, we utilized meta-regression to explore the influence of death time on the predictive value of the model for HIV-related deaths. RESULTS After our comprehensive review, we analyzed a total of 24 pieces of literature, encompassing data from 401,389 individuals diagnosed with HIV. Within this dataset, 23 articles specifically delved into deaths during long-term follow-ups outside hospital settings. The machine learning models applied for predicting these deaths comprised survival models (COX regression) and other non-survival models. The outcomes of the meta-analysis unveiled that within the training set, the c-index for predicting deaths among people with HIV (PWH) using predictive models stands at 0.83 (95% CI: 0.75-0.91). In the validation set, the c-index is slightly lower at 0.81 (95% CI: 0.78-0.85). Notably, the meta-regression analysis demonstrated that neither follow-up time nor the occurrence of death events significantly impacted the performance of the machine learning models. CONCLUSIONS The study suggests that machine learning is a viable approach for developing non-time-based predictions regarding HIV deaths. Nevertheless, the limited inclusion of original studies necessitates additional multicenter studies for thorough validation.
Collapse
Affiliation(s)
- Yuefei Li
- Public Health, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Ying Feng
- Urumqi Maternal and Child Health Hospital, Urumqi, Xinjiang, 830000, China
| | - Qian He
- Public Health, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Zhen Ni
- STD/HIV Prevention and Control Center, Xinjiang Uighur Autonomous Region Center for Disease Control and Prevention, No. 138 Jianquan 1st Street, Tianshan District, Urumqi, Xinjiang, 830002, China
| | - Xiaoyuan Hu
- STD/HIV Prevention and Control Center, Xinjiang Uighur Autonomous Region Center for Disease Control and Prevention, No. 138 Jianquan 1st Street, Tianshan District, Urumqi, Xinjiang, 830002, China
| | - Xinhuan Feng
- Clinical Laboratory, Second People's Hospital of Yining, Yining, Xinjiang, 835000, China
| | - Mingjian Ni
- STD/HIV Prevention and Control Center, Xinjiang Uighur Autonomous Region Center for Disease Control and Prevention, No. 138 Jianquan 1st Street, Tianshan District, Urumqi, Xinjiang, 830002, China.
| |
Collapse
|
39
|
Ni Q, Ge Z, Li Y, Shatkin G, Fu J, Bera K, Yang Y, Wang Y, Sen A, Wu Y, Vasconcelos ACN, Feinberg AP, Konstantopoulos K, Sun SX. Cytoskeletal activation of NHE1 regulates mechanosensitive cell volume adaptation and proliferation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.31.555808. [PMID: 37693593 PMCID: PMC10491192 DOI: 10.1101/2023.08.31.555808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Mammalian cells can rapidly respond to osmotic and hydrostatic pressure imbalances during an environmental change, generating large fluxes of water and ions that alter cell volume within minutes. While the role of ion pump and leak in cell volume regulation has been well-established, the potential contribution of the actomyosin cytoskeleton and its interplay with ion transporters is unclear. We discovered a cell volume regulation system that is controlled by cytoskeletal activation of ion transporters. After a hypotonic shock, normal-like cells (NIH-3T3, MCF-10A, and others) display a slow secondary volume increase (SVI) following the immediate regulatory volume decrease. We show that SVI is initiated by hypotonic stress induced Ca 2+ influx through stretch activated channel Piezo1, which subsequently triggers actomyosin remodeling. The actomyosin network further activates NHE1 through their synergistic linker ezrin, inducing SVI after the initial volume recovery. We find that SVI is absent in cancer cell lines such as HT1080 and MDA-MB-231, where volume regulation is dominated by intrinsic response of ion transporters. A similar cytoskeletal activation of NHE1 can also be achieved by mechanical stretching. On compliant substrates where cytoskeletal contractility is attenuated, SVI generation is abolished. Moreover, cytoskeletal activation of NHE1 during SVI triggers nuclear deformation, leading to a significant, immediate transcriptomic change in 3T3 cells, a phenomenon that is again absent in HT1080 cells. While hypotonic shock hinders ERK-dependent cell growth, cells deficient in SVI are unresponsive to such inhibitory effects. Overall, our findings reveal the critical role of Ca 2+ and actomyosin-mediated mechanosensation in the regulation of ion transport, cell volume, transcriptomics, and cell proliferation.
Collapse
|
40
|
Garrud TAC, Bell B, Mata-Daboin A, Peixoto-Neves D, Collier DM, Cordero-Morales JF, Jaggar JH. WNK kinase is a vasoactive chloride sensor in endothelial cells. Proc Natl Acad Sci U S A 2024; 121:e2322135121. [PMID: 38568964 PMCID: PMC11009681 DOI: 10.1073/pnas.2322135121] [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: 12/21/2023] [Accepted: 03/01/2024] [Indexed: 04/05/2024] Open
Abstract
Endothelial cells (ECs) line the wall of blood vessels and regulate arterial contractility to tune regional organ blood flow and systemic pressure. Chloride (Cl-) is the most abundant anion in ECs and the Cl- sensitive With-No-Lysine (WNK) kinase is expressed in this cell type. Whether intracellular Cl- signaling and WNK kinase regulate EC function to alter arterial contractility is unclear. Here, we tested the hypothesis that intracellular Cl- signaling in ECs regulates arterial contractility and examined the signaling mechanisms involved, including the participation of WNK kinase. Our data obtained using two-photon microscopy and cell-specific inducible knockout mice indicated that acetylcholine, a prototypical vasodilator, stimulated a rapid reduction in intracellular Cl- concentration ([Cl-]i) due to the activation of TMEM16A, a Cl- channel, in ECs of resistance-size arteries. TMEM16A channel-mediated Cl- signaling activated WNK kinase, which phosphorylated its substrate proteins SPAK and OSR1 in ECs. OSR1 potentiated transient receptor potential vanilloid 4 (TRPV4) currents in a kinase-dependent manner and required a conserved binding motif located in the channel C terminus. Intracellular Ca2+ signaling was measured in four dimensions in ECs using a high-speed lightsheet microscope. WNK kinase-dependent activation of TRPV4 channels increased local intracellular Ca2+ signaling in ECs and produced vasodilation. In summary, we show that TMEM16A channel activation reduces [Cl-]i, which activates WNK kinase in ECs. WNK kinase phosphorylates OSR1 which then stimulates TRPV4 channels to produce vasodilation. Thus, TMEM16A channels regulate intracellular Cl- signaling and WNK kinase activity in ECs to control arterial contractility.
Collapse
Affiliation(s)
- Tessa A. C. Garrud
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN38163
| | - Briar Bell
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN38163
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX77030
| | - Alejandro Mata-Daboin
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN38163
| | | | - Daniel M. Collier
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN38163
| | - Julio F. Cordero-Morales
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN38163
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX77030
| | - Jonathan H. Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN38163
| |
Collapse
|
41
|
Sun X, Zhou Y, Wang Z, Peng M, Wei X, Xie Y, Wen C, Liu J, Ye M. Biomolecular Condensates Decipher Molecular Codes of Cell Fate: From Biophysical Fundamentals to Therapeutic Practices. Int J Mol Sci 2024; 25:4127. [PMID: 38612940 PMCID: PMC11012904 DOI: 10.3390/ijms25074127] [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: 02/29/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
Cell fate is precisely modulated by complex but well-tuned molecular signaling networks, whose spatial and temporal dysregulation commonly leads to hazardous diseases. Biomolecular condensates (BCs), as a newly emerging type of biophysical assemblies, decipher the molecular codes bridging molecular behaviors, signaling axes, and clinical prognosis. Particularly, physical traits of BCs play an important role; however, a panoramic view from this perspective toward clinical practices remains lacking. In this review, we describe the most typical five physical traits of BCs, and comprehensively summarize their roles in molecular signaling axes and corresponding major determinants. Moreover, establishing the recent observed contribution of condensate physics on clinical therapeutics, we illustrate next-generation medical strategies by targeting condensate physics. Finally, the challenges and opportunities for future medical development along with the rapid scientific and technological advances are highlighted.
Collapse
Affiliation(s)
- Xing Sun
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China; (X.S.); (Y.Z.); (Z.W.); (M.P.); (X.W.)
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410000, China; (Y.X.); (C.W.)
| | - Yangyang Zhou
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China; (X.S.); (Y.Z.); (Z.W.); (M.P.); (X.W.)
| | - Zhiyan Wang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China; (X.S.); (Y.Z.); (Z.W.); (M.P.); (X.W.)
| | - Menglan Peng
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China; (X.S.); (Y.Z.); (Z.W.); (M.P.); (X.W.)
| | - Xianhua Wei
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China; (X.S.); (Y.Z.); (Z.W.); (M.P.); (X.W.)
| | - Yifang Xie
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410000, China; (Y.X.); (C.W.)
| | - Chengcai Wen
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410000, China; (Y.X.); (C.W.)
| | - Jing Liu
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410000, China; (Y.X.); (C.W.)
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China; (X.S.); (Y.Z.); (Z.W.); (M.P.); (X.W.)
| |
Collapse
|
42
|
Ferdaus MZ, Koumangoye RB, Welling PA, Delpire E. Kinase Scaffold Cab39 Is Necessary for Phospho-Activation of the Thiazide-Sensitive NCC. Hypertension 2024; 81:801-810. [PMID: 38258567 PMCID: PMC10954405 DOI: 10.1161/hypertensionaha.123.22464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/11/2024] [Indexed: 01/24/2024]
Abstract
BACKGROUND Potassium regulates the WNK (with no lysine kinase)-SPAK (STE20/SPS1-related proline/alanine-rich kinase) signaling axis, which in turn controls the phosphorylation and activation of the distal convoluted tubule thiazide-sensitive NCC (sodium-chloride cotransporter) for sodium-potassium balance. Although their roles in the kidney have not been investigated, it has been postulated that Cab39 (calcium-binding protein 39) or Cab39l (Cab39-like) is required for SPAK/OSR1 (oxidative stress response 1) activation. This study demonstrates how they control the WNK-SPAK/OSR1-NCC pathway. METHODS We created a global knockout of Cab39l and a tamoxifen-inducible, NCC-driven, Cab39 knockout. The 2 lines were crossed to generate Cab39-DKO (Cab39 double knockout) animals. Mice were studied under control and low-potassium diet, which activates WNK-SPAK/OSR1-NCC phosphorylation. Western blots were used to assess the expression and phosphorylation of proteins. Blood and urine electrolytes were measured to test for compromised NCC function. Immunofluorescence studies were conducted to localize SPAK and OSR1. RESULTS Both Cab39l and Cab39 are expressed in distal convoluted tubule, and only the elimination of both leads to a striking absence of NCC phosphorylation. Cab39-DKO mice exhibited a loss-of-NCC function, like in Gitelman syndrome. In contrast to the apical membrane colocalization of SPAK with NCC in wild-type mice, SPAK and OSR1 become confined to intracellular puncta in the Cab39-DKO mice. CONCLUSIONS In the absence of Cab39 proteins, NCC cannot be phosphorylated, resulting in a Gitelman-like phenotype. Cab39 proteins function to localize SPAK at the apical membrane with NCC, reminiscent of the Cab39 yeast homolog function, translocating kinases during cytokinesis.
Collapse
Affiliation(s)
- Mohammed Z Ferdaus
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN (M.Z.F, R.B.K., E.D.)
| | - Rainelli B Koumangoye
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN (M.Z.F, R.B.K., E.D.)
| | - Paul A Welling
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (P.A.W.)
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN (M.Z.F, R.B.K., E.D.)
| |
Collapse
|
43
|
Sehgal PB, Yuan H, Centone A, DiSenso-Browne SV. Oral Antiviral Defense: Saliva- and Beverage-like Hypotonicity Dynamically Regulate Formation of Membraneless Biomolecular Condensates of Antiviral Human MxA in Oral Epithelial Cells. Cells 2024; 13:590. [PMID: 38607029 PMCID: PMC11011872 DOI: 10.3390/cells13070590] [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: 02/14/2024] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
The oral mucosa represents a defensive barrier between the external environment and the rest of the body. Oral mucosal cells are constantly bathed in hypotonic saliva (normally one-third tonicity compared to plasma) and are repeatedly exposed to environmental stresses of tonicity, temperature, and pH by the drinks we imbibe (e.g., hypotonic: water, tea, and coffee; hypertonic: assorted fruit juices, and red wines). In the mouth, the broad-spectrum antiviral mediator MxA (a dynamin-family large GTPase) is constitutively expressed in healthy periodontal tissues and induced by Type III interferons (e.g., IFN-λ1/IL-29). Endogenously induced human MxA and exogenously expressed human GFP-MxA formed membraneless biomolecular condensates in the cytoplasm of oral carcinoma cells (OECM1 cell line). These condensates likely represent storage granules in equilibrium with antivirally active dispersed MxA. Remarkably, cytoplasmic MxA condensates were exquisitely sensitive sensors of hypotonicity-the condensates in oral epithelium disassembled within 1-2 min of exposure of cells to saliva-like one-third hypotonicity, and spontaneously reassembled in the next 4-7 min. Water, tea, and coffee enhanced this disassembly. Fluorescence changes in OECM1 cells preloaded with calcein-AM (a reporter of cytosolic "macromolecular crowding") confirmed that this process involved macromolecular uncrowding and subsequent recrowding secondary to changes in cell volume. However, hypertonicity had little effect on MxA condensates. The spontaneous reassembly of GFP-MxA condensates in oral epithelial cells, even under continuous saliva-like hypotonicity, was slowed by the protein-phosphatase-inhibitor cyclosporin A (CsA) and by the K-channel-blocker tetraethylammonium chloride (TEA); this is suggestive of the involvement of the volume-sensitive WNK kinase-protein phosphatase (PTP)-K-Cl cotransporter (KCC) pathway in the regulated volume decrease (RVD) during condensate reassembly in oral cells. The present study identifies a novel subcellular consequence of hypotonic stress in oral epithelial cells, in terms of the rapid and dynamic changes in the structure of one class of phase-separated biomolecular condensates in the cytoplasm-the antiviral MxA condensates. More generally, the data raise the possibility that hypotonicity-driven stresses likely affect other intracellular functions involving liquid-liquid phase separation (LLPS) in cells of the oral mucosa.
Collapse
Affiliation(s)
- Pravin B. Sehgal
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA;
- Department of Medicine, New York Medical College, Valhalla, NY 10595, USA
| | - Huijuan Yuan
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA;
| | - Anthony Centone
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA;
| | | |
Collapse
|
44
|
Yu B, Chao DY, Zhao Y. How plants sense and respond to osmotic stress. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2024; 66:394-423. [PMID: 38329193 DOI: 10.1111/jipb.13622] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 02/09/2024]
Abstract
Drought is one of the most serious abiotic stresses to land plants. Plants sense and respond to drought stress to survive under water deficiency. Scientists have studied how plants sense drought stress, or osmotic stress caused by drought, ever since Charles Darwin, and gradually obtained clues about osmotic stress sensing and signaling in plants. Osmotic stress is a physical stimulus that triggers many physiological changes at the cellular level, including changes in turgor, cell wall stiffness and integrity, membrane tension, and cell fluid volume, and plants may sense some of these stimuli and trigger downstream responses. In this review, we emphasized water potential and movements in organisms, compared putative signal inputs in cell wall-containing and cell wall-free organisms, prospected how plants sense changes in turgor, membrane tension, and cell fluid volume under osmotic stress according to advances in plants, animals, yeasts, and bacteria, summarized multilevel biochemical and physiological signal outputs, such as plasma membrane nanodomain formation, membrane water permeability, root hydrotropism, root halotropism, Casparian strip and suberin lamellae, and finally proposed a hypothesis that osmotic stress responses are likely to be a cocktail of signaling mediated by multiple osmosensors. We also discussed the core scientific questions, provided perspective about the future directions in this field, and highlighted the importance of robust and smart root systems and efficient source-sink allocations for generating future high-yield stress-resistant crops and plants.
Collapse
Affiliation(s)
- Bo Yu
- Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Sciences, Shanghai, 200032, China
- Key Laboratory of Plant Carbon Capture, The Chinese Academy of Sciences, Shanghai, 200032, China
| | - Dai-Yin Chao
- National Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, The Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Zhao
- Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Sciences, Shanghai, 200032, China
- Key Laboratory of Plant Carbon Capture, The Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
45
|
Holehouse AS, Kragelund BB. The molecular basis for cellular function of intrinsically disordered protein regions. Nat Rev Mol Cell Biol 2024; 25:187-211. [PMID: 37957331 PMCID: PMC11459374 DOI: 10.1038/s41580-023-00673-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2023] [Indexed: 11/15/2023]
Abstract
Intrinsically disordered protein regions exist in a collection of dynamic interconverting conformations that lack a stable 3D structure. These regions are structurally heterogeneous, ubiquitous and found across all kingdoms of life. Despite the absence of a defined 3D structure, disordered regions are essential for cellular processes ranging from transcriptional control and cell signalling to subcellular organization. Through their conformational malleability and adaptability, disordered regions extend the repertoire of macromolecular interactions and are readily tunable by their structural and chemical context, making them ideal responders to regulatory cues. Recent work has led to major advances in understanding the link between protein sequence and conformational behaviour in disordered regions, yet the link between sequence and molecular function is less well defined. Here we consider the biochemical and biophysical foundations that underlie how and why disordered regions can engage in productive cellular functions, provide examples of emerging concepts and discuss how protein disorder contributes to intracellular information processing and regulation of cellular function.
Collapse
Affiliation(s)
- Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA.
- Center for Biomolecular Condensates, Washington University in St Louis, St Louis, MO, USA.
| | - Birthe B Kragelund
- REPIN, Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
46
|
Monterroso B, Margolin W, Boersma AJ, Rivas G, Poolman B, Zorrilla S. Macromolecular Crowding, Phase Separation, and Homeostasis in the Orchestration of Bacterial Cellular Functions. Chem Rev 2024; 124:1899-1949. [PMID: 38331392 PMCID: PMC10906006 DOI: 10.1021/acs.chemrev.3c00622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/01/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024]
Abstract
Macromolecular crowding affects the activity of proteins and functional macromolecular complexes in all cells, including bacteria. Crowding, together with physicochemical parameters such as pH, ionic strength, and the energy status, influences the structure of the cytoplasm and thereby indirectly macromolecular function. Notably, crowding also promotes the formation of biomolecular condensates by phase separation, initially identified in eukaryotic cells but more recently discovered to play key functions in bacteria. Bacterial cells require a variety of mechanisms to maintain physicochemical homeostasis, in particular in environments with fluctuating conditions, and the formation of biomolecular condensates is emerging as one such mechanism. In this work, we connect physicochemical homeostasis and macromolecular crowding with the formation and function of biomolecular condensates in the bacterial cell and compare the supramolecular structures found in bacteria with those of eukaryotic cells. We focus on the effects of crowding and phase separation on the control of bacterial chromosome replication, segregation, and cell division, and we discuss the contribution of biomolecular condensates to bacterial cell fitness and adaptation to environmental stress.
Collapse
Affiliation(s)
- Begoña Monterroso
- Department
of Structural and Chemical Biology, Centro de Investigaciones Biológicas
Margarita Salas, Consejo Superior de Investigaciones
Científicas (CSIC), 28040 Madrid, Spain
| | - William Margolin
- Department
of Microbiology and Molecular Genetics, McGovern Medical School, UTHealth-Houston, Houston, Texas 77030, United States
| | - Arnold J. Boersma
- Cellular
Protein Chemistry, Bijvoet Centre for Biomolecular Research, Faculty
of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Germán Rivas
- Department
of Structural and Chemical Biology, Centro de Investigaciones Biológicas
Margarita Salas, Consejo Superior de Investigaciones
Científicas (CSIC), 28040 Madrid, Spain
| | - Bert Poolman
- Department
of Biochemistry, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Silvia Zorrilla
- Department
of Structural and Chemical Biology, Centro de Investigaciones Biológicas
Margarita Salas, Consejo Superior de Investigaciones
Científicas (CSIC), 28040 Madrid, Spain
| |
Collapse
|
47
|
Sugiyama S, Suda K, Kono K. Cytoplasmic zoning by protein phase transition after membrane permeabilization. J Biochem 2024; 175:147-153. [PMID: 37972304 PMCID: PMC10873517 DOI: 10.1093/jb/mvad094] [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: 08/30/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023] Open
Abstract
Biological membranes, including plasma membrane (PM) and organelle membranes, restrict the flux of ions, molecules and organelles. However, the barrier function of biological membranes is frequently compromised by various perturbations, including physical membrane damage and protein- or chemical-induced pore formation. Recent evidence suggests that, upon PM damage, protein gelation and solid condensation are utilized to restrict ion/molecule/organelle flux across the damaged membranes by zoning the cytoplasm. In addition, membrane permeabilization dramatically alters intramembrane and extramembrane ion/molecule concentrations via the flux across the permeabilized membrane. The changes in ion/molecule concentration and their downstream pathways induce protein phase transition to form zones for biological processes or protein sequestration. Here, we review the mechanisms and functions of protein phase transition after biological membrane permeabilization.
Collapse
Affiliation(s)
- Shinju Sugiyama
- Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa, 904-0495, Japan
| | - Kojiro Suda
- Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa, 904-0495, Japan
| | - Keiko Kono
- Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa, 904-0495, Japan
| |
Collapse
|
48
|
Subramanya AR, Boyd-Shiwarski CR. Molecular Crowding: Physiologic Sensing and Control. Annu Rev Physiol 2024; 86:429-452. [PMID: 37931170 PMCID: PMC11472293 DOI: 10.1146/annurev-physiol-042222-025920] [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: 11/08/2023]
Abstract
The cytoplasm is densely packed with molecules that contribute to its nonideal behavior. Cytosolic crowding influences chemical reaction rates, intracellular water mobility, and macromolecular complex formation. Overcrowding is potentially catastrophic; to counteract this problem, cells have evolved acute and chronic homeostatic mechanisms that optimize cellular crowdedness. Here, we provide a physiology-focused overview of molecular crowding, highlighting contemporary advances in our understanding of its sensing and control. Long hypothesized as a form of crowding-induced microcompartmentation, phase separation allows cells to detect and respond to intracellular crowding through the action of biomolecular condensates, as indicated by recent studies. Growing evidence indicates that crowding is closely tied to cell size and fluid volume, homeostatic responses to physical compression and desiccation, tissue architecture, circadian rhythm, aging, transepithelial transport, and total body electrolyte and water balance. Thus, molecular crowding is a fundamental physiologic parameter that impacts diverse functions extending from molecule to organism.
Collapse
Affiliation(s)
- Arohan R Subramanya
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; ,
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - Cary R Boyd-Shiwarski
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; ,
- Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
49
|
Huang Y, Xu X, Lu Y, Sun Q, Zhang L, Shao J, Chen D, Chang Y, Sun X, Zhuo W, Zhou T. The phase separation of extracellular matrix protein matrilin-3 from cancer-associated fibroblasts contributes to gastric cancer invasion. FASEB J 2024; 38:e23406. [PMID: 38193601 DOI: 10.1096/fj.202301524r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/23/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024]
Abstract
Cancer-associated fibroblast (CAF) has emerged as a key contributor to the remodeling of tumor microenvironment through the expression and secretion of extracellular matrix (ECM) proteins, thereby promoting carcinogenesis. However, the precise contribution of ECM proteins from CAFs to gastric carcinogenesis remains poorly understood. In this study, we find that matrilin-3 (MATN3), an upregulated ECM protein associated with poorer prognosis in gastric cancer patients, originates from CAFs in gastric cancer tissues. Ectopic expression of MATN3 in CAFs significantly promotes the invasion of gastric cancer cells, which can be attenuated by neutralizing MATN3 with its antibody. Notably, a portion of MATN3 protein is found to form puncta in gastric cancer tissues ECM. MATN3 undergoes phase separation, which is mediated by its low complexity (LC) and coiled-coil (CC) domains. Moreover, overexpression of MATN3 deleted with either LC or CC in CAFs is unable to promote the invasion of gastric cancer cells, suggesting that LC or CC domain is required for the effect of CAF-secreted MATN3 in gastric cancer cell invasion. Additionally, orthotopic co-injection of gastric cancer cells and CAFs expressing MATN3, but not its ΔLC and ΔCC mutants, leads to enhanced gastric cancer cell invasion in mouse models. Collectively, our works suggest that MATN3 is secreted by CAFs and undergoes phase separation, which promotes gastric cancer invasion.
Collapse
Affiliation(s)
- Yuliang Huang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyang Xu
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunkun Lu
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Sun
- Center for RNA Medicine, International Institutes of Medicine and the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Lu Zhang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Shao
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Dingwei Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongxia Chang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxia Sun
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zhuo
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianhua Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
50
|
Marakhova II, Yurinskaya VE, Domnina AP. The Role of Intracellular Potassium in Cell Quiescence, Proliferation, and Death. Int J Mol Sci 2024; 25:884. [PMID: 38255956 PMCID: PMC10815214 DOI: 10.3390/ijms25020884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/28/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
This brief review explores the role of intracellular K+ during the transition of cells from quiescence to proliferation and the induction of apoptosis. We focus on the relationship between intracellular K+ and the growth and proliferation rates of different cells, including transformed cells in culture as well as human quiescent T cells and mesenchymal stem cells, and analyze the concomitant changes in K+ and water content in both proliferating and apoptotic cells. Evidence is discussed indicating that during the initiation of cell proliferation and apoptosis changes in the K+ content in cells occur in parallel with changes in water content and therefore do not lead to significant changes in the intracellular K+ concentration. We conclude that K+, as a dominant intracellular ion, is involved in the regulation of cell volume during the transit from quiescence, and the content of K+ and water in dividing cells is higher than in quiescent or differentiated cells, which can be considered to be a hallmark of cell proliferation and transformation.
Collapse
Affiliation(s)
- Irina I. Marakhova
- Department of Intracellular Signalling and Transport, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Avenue 4, 194064 Saint-Petersburg, Russia
| | - Valentina E. Yurinskaya
- Department of Molecular Cell Physiology, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Avenue 4, 194064 Saint-Petersburg, Russia
| | - Alisa P. Domnina
- Department of Intracellular Signalling and Transport, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Avenue 4, 194064 Saint-Petersburg, Russia
| |
Collapse
|