1
|
Wu F, Mu WC, Markov NT, Fuentealba M, Halaweh H, Senchyna F, Manwaring-Mueller MN, Winer DA, Furman D. Immunological biomarkers of aging. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:889-902. [PMID: 40443365 PMCID: PMC12123219 DOI: 10.1093/jimmun/vkae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/22/2024] [Indexed: 06/02/2025]
Abstract
The immune system has long been recognized for its critical role in the elimination of pathogens and the development of autoimmune diseases, but recent evidence demonstrates that it also contributes to noncommunicable diseases associated with biological aging processes, such as cancer, cardiovascular disease, neurodegeneration, and frailty. This review examines immunological biomarkers of aging, focusing on how the immune system evolves with age and its impact on health and disease. It discusses the historical development of immunological assessments, technological advancements, and the creation of novel biomarkers and models to study immune aging. We also explore the clinical implications of immune aging, such as increased susceptibility to infectious diseases, poor vaccine responses, and a higher incidence of noncommunicable diseases. In summary, we provide a comprehensive overview of current research, highlight the clinical relevance of immune aging, and identify gaps in knowledge that require further investigation.
Collapse
Affiliation(s)
- Fei Wu
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Wei-Chieh Mu
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Nikola T Markov
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Matias Fuentealba
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Heather Halaweh
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Fiona Senchyna
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | | | - Daniel A Winer
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
- Buck Institute for Research on Aging, Novato, CA, United States
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - David Furman
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, United States
- Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
2
|
Abubakar M, Fan S, Klein A, Pfeiffer RM, Lawrence S, Mutreja K, Kimes TM, Richert-Boe K, Figueroa JD, Gierach GL, Duggan MA, Rohan TE. Spatially Resolved Single-Cell Morphometry of Benign Breast Disease Biopsy Images Uncovers Quantitative Cytomorphometric Features Predictive of Subsequent Invasive Breast Cancer Risk. Mod Pathol 2025; 38:100767. [PMID: 40210131 DOI: 10.1016/j.modpat.2025.100767] [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: 11/18/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/12/2025]
Abstract
Currently, benign breast disease (BBD) pathologic classification and invasive breast cancer (BC) risk assessment are based on qualitative epithelial changes, with limited utility for BC risk stratification for women with lower-risk category BBD (ie, nonproliferative disease [NPD] and proliferative disease without atypia [PDWA]). Here, machine learning-based single-cell morphometry was used to characterize quantitative changes in epithelial nuclear morphology that reflect functional/structural decline (ie, increasing nuclear size, assessed as epithelial nuclear area and nuclear perimeter), altered DNA chromatin content (ie, increasing nuclear chromasia), and increased cellular crowding/proliferation (ie, increasing nuclear contour irregularity). Cytomorphologic changes reflecting chronic stromal inflammation were assessed using stromal cellular density. Data and pathology materials were obtained from a case-control study (n = 972) nested within a cohort of 15,395 women diagnosed with BBD at Kaiser Permanente Northwest (1971-2012). Odds ratios (ORs) and 95% confidence intervals (CIs) for associations of cytomorphometric features with risk of subsequent BC were assessed using multivariable logistic regression. More than 55 million epithelial and 37 million stromal cells were profiled across 972 BBD images. Cytomorphometric features were individually predictive of subsequent BC risk, independently of BBD histologic classification. However, cytomorphometric features of epithelial functional/structural decline were statistically significantly predictive of low-grade but not high-grade BC following PDWA (OR for low-grade BC per 1-SD increase in nuclear area and nuclear perimeter, 2.10; 95% CI, 1.26-3.49, and 2.22; 95% CI, 1.30-3.78, respectively), whereas stromal inflammation was predictive of high-grade but not low-grade BC following NPD (OR for high-grade BC per 1-SD increase in stromal cellular density, 1.53; 95% CI, 1.13-2.08). Associations of nuclear chromasia and nuclear contour irregularity with subsequent tumor grade were context specific, with both features predicting low-grade BC risk following PDWA (OR per 1-SD, 1.58; 95% CI, 1.06-2.35, and 2.21; 95% CI, 1.25-3.91, for nuclear chromasia and nuclear contour irregularity, respectively) and high-grade BC following NPD (OR per 1-SD, 1.47; 95% CI, 1.11-1.96, and 1.29; 95% CI, 1.00-1.70, for nuclear chromasia and nuclear contour irregularity, respectively). The results indicate that cytomorphometric features on BBD hematoxylin-eosin-stained images might help to refine BC risk estimation and potentially inform BC risk reduction strategies for BBD patients, particularly those currently designated as low risk.
Collapse
Affiliation(s)
- Mustapha Abubakar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Rockville, Maryland.
| | - Shaoqi Fan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Rockville, Maryland
| | - Alyssa Klein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Rockville, Maryland
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Rockville, Maryland
| | - Scott Lawrence
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick, Maryland
| | - Karun Mutreja
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick, Maryland
| | - Teresa M Kimes
- Kaiser Permanente Center for Health Research, Portland, Oregon
| | | | - Jonine D Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Rockville, Maryland
| | - Gretchen L Gierach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Rockville, Maryland
| | - Maire A Duggan
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
3
|
Davies P, Cavallaro M, Hebenstreit D. Single-Calibration Cell Size Measurement With Flow Cytometry. Cytometry A 2025; 107:263-270. [PMID: 40071841 DOI: 10.1002/cyto.a.24924] [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: 08/09/2024] [Revised: 12/06/2024] [Accepted: 02/27/2025] [Indexed: 04/30/2025]
Abstract
Measuring the size of individual cells in high-throughput experiments is often important in biomedical research and applications. Nevertheless, popular tools for high-throughput single-cell biology, such as flow cytometers, only offer proxies of a cell's size, typically reported in arbitrary scales and often subject to changes in the instrument's settings as selected by multiple users. In this paper, we demonstrate that it is possible to calibrate flowcytometry laser scatter signals with accurate measures of cell diameter from separate devices and that the calibration can be conserved upon changes in the laser settings. We demonstrate our approach based on flow cytometric sorting of cells of a mammalian cell line according to a selection of scatter parameters, followed by cell size determination with a Coulter counter. A straightforward procedure is presented that relates the flow cytometric scatter parameters to the absolute size measurements using linear models, along with a linear transformation that converts between different instrument settings on the flow cytometer. Our method makes it possible to record on a flow cytometer a cell's size in absolute units and correlate it with other features that are recorded in parallel in the fluorescence detection channels.
Collapse
Affiliation(s)
- Philip Davies
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Massimo Cavallaro
- School of Life Sciences, University of Warwick, Coventry, UK
- School of Computing and Mathematical Sciences, University of Leicester, Leicester, UK
| | | |
Collapse
|
4
|
Tan C, Lanz MC, Swaffer M, Skotheim J, Chang F. Intracellular diffusion in the cytoplasm increases with cell size in fission yeast. Mol Biol Cell 2025; 36:ar51. [PMID: 39969966 PMCID: PMC12005113 DOI: 10.1091/mbc.e24-11-0488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Diffusion in the cytoplasm can greatly impact cellular processes, yet regulation of macromolecular diffusion remains poorly understood. There is increasing evidence that cell size affects the density and macromolecular composition of the cytoplasm. Here, we studied whether cell size affects diffusion at the scale of macromolecules tens of microns in diameter. We analyzed the diffusive motions of intracellular genetically-encoded multimeric 40 nm nanoparticles (cytGEMs) in the cytoplasm of the fission yeast Schizosaccharomyces pombe. Using cell size mutants, we showed that cytGEMs diffusion coefficients decreased in smaller cells and increased in larger cells. This increase in diffusion in large cells may be due to a decrease in the DNA-to-cytoplasm ratio, as diffusion was not affected in large multinucleate cytokinesis mutant cells. In investigating the underlying causes of altered cytGEMs diffusion, we found that the proteomes of large and small cells exhibited size-specific changes, including the subscaling of ribosomal proteins in large cells. Comparison with a similar dataset from human cells revealed that features of size-dependent proteome remodeling were conserved. These studies demonstrate that cell size is an important parameter in determining the biophysical properties and the composition of the cytoplasm.
Collapse
Affiliation(s)
- Catherine Tan
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
| | - Michael C. Lanz
- Department of Biology, Stanford University, Stanford, CA 94305
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA 94158
| | - Matthew Swaffer
- Department of Biology, Stanford University, Stanford, CA 94305
| | - Jan Skotheim
- Department of Biology, Stanford University, Stanford, CA 94305
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA 94158
| | - Fred Chang
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
5
|
Hsueh YJ, Chen HC, Pan YY, Hsiao FC, Yang SJ, Liu MC, Lai WY, Li G, Hui-Kang Ma D, James Meir YJ. The hiPSC-derived corneal endothelial progenitor-like cell recovers the rabbit model of corneal endothelial dystrophy. J Adv Res 2025; 70:355-369. [PMID: 38729560 PMCID: PMC11976410 DOI: 10.1016/j.jare.2024.05.008] [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: 11/18/2023] [Revised: 04/24/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
INTRODUCTION Corneal endothelial dysfunction results in cornea opacity, damaging sightedness, and affecting quality of life. A corneal transplant is the current effective intervention. Due to the scarcity of donated cornea, such an unmet medical need requires a novel therapeutic modality. OBJECTIVES Customizing patients' corneal endothelial progenitor cells with proliferative activity and lineage restriction properties shall offer sufficient therapeutic cells for corneal endothelial dystrophy. METHODS The customized induced human corneal endothelial progenitor-like cell (iHCEPLC) was obtained through cell fate conversions starting from PBMC (peripheral blood mononuclear cell), hiPSC (human induced pluripotent stem cell), and hNCC (human neural crest cell), while it finally reached the iHCEPLC state via a series of induction. Several molecular diagnoses were applied to depict its progenitor state, including RNAseq, FlowCytometer, immunostainings, and rtPCR. Significantly, it can be induced to gain differentiation maturity through contact inhibition. In addition, a BAK-mediated rabbit model of corneal endothelial dystrophy was established in the present study to test the therapeutic effectiveness of the iHCEPLC. RESULTS After inducing cell fate conversion, the specific HCEC markers were detected by rtPCR and immunostaining in iHCEPLC. Further, RNAseq was applied to distinguish its progenitor-like cell fate from primary human corneal endothelial cells (HECE). FlowCytometry profiled the heterogeneity subpopulation, consistently displaying a subtle difference from primary HCEC. A terminal differentiation can be induced in iHCEPLC, addressing its progenitor-like fate. iHCEPLC can restore the BAK-based rabbit model of corneal endothelial dystrophy. Immunohistochemistry verified that such acuity restoration of the BAK-treated cornea is due to the introduced iHCEPLC, and such therapeutic effectiveness is observed in the long term. CONCLUSION Here, we demonstrated that customized iHCEPLC has long-term therapeutic efficacy. As a progenitor cell, our iHCEPLC has a restricted cell lineage nature and can proliferate in vitro, supporting sufficient therapeutic candidate cells. Due to the immune-privileged nature of the cornea, our iHCEPLC proves the principle of therapeutical feasibility in both autogenic and allogeneic modalities.
Collapse
Affiliation(s)
- Yi-Jen Hsueh
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan; Dept. of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Hung-Chi Chen
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan; Dept. of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan; Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yu-Yun Pan
- Dept. of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Fang-Chi Hsiao
- Dept. of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Shun-Jie Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Mei-Chun Liu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Wei-Yu Lai
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Guigang Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei Province 430030, China
| | - David Hui-Kang Ma
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan; Dept. of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan; Department of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yaa-Jyuhn James Meir
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan; Dept. of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan; Dept. of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan.
| |
Collapse
|
6
|
Cavanaugh M, Willits RK. Mechanotransductive N-cadherin binding induces differentiation in human neural stem cells. MECHANOBIOLOGY IN MEDICINE 2025; 3:100099. [PMID: 40396132 PMCID: PMC12082155 DOI: 10.1016/j.mbm.2024.100099] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 05/22/2025]
Abstract
The neural stem cell niche is a complex microenvironment that includes cellular factors, secreted factors, and physical factors that impact stem cell behavior and development. Cellular interactions through cadherins, cell-cell binding proteins, have implications in embryonic development and mesenchymal stem cell differentiation. However, little is known about the influence of cadherins within the neural stem cell microenvironment and their effect on human stem cell maintenance and differentiation. Therefore, the purpose of this study was to develop synthetic substrates to examine the effect of cadherin mechanotransduction on human neural stem cells. Glass substrates were fabricated using silane, protein A, and recombinant N-cadherin; we used these substrates to examine the effect of N-cadherin binding on neural stem cell proliferation, cytoskeletal structure and morphology, Yes-associated protein-1 (YAP) translocation, and differentiation. Bound exogenous N-cadherin induced concentration-dependent increases in adherens junction formation, YAP translocation, and early expression of neurogenic differentiation markers. Strong F-actin ring structures were initiated by homophilic N-cadherin binding, eliciting neuronal differentiation of cells within 96 h without added soluble differentiation factors. Our findings show that active N-cadherin binding plays an important role for differentiation of human iPS-derived neural stem cells towards neurons, providing a new tool to differentiate cells in vitro.
Collapse
Affiliation(s)
- McKay Cavanaugh
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Rebecca Kuntz Willits
- Department of Bioengineering, Northeastern University, Boston, MA, USA
- Department of Chemical Engineering, Northeastern University, Boston MA, USA
| |
Collapse
|
7
|
Vandiver AR, Torres A, Sanden A, Nguyen TL, Gasilla J, Doan MT, Martirosian V, Hoang A, Wanagat J, Teitell MA. Increased mitochondrial mutation heteroplasmy induces aging phenotypes in pluripotent stem cells and their differentiated progeny. Aging Cell 2025; 24:e14402. [PMID: 39680477 PMCID: PMC11896400 DOI: 10.1111/acel.14402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 12/18/2024] Open
Abstract
The mitochondrial genome (mtDNA) is an important source of inherited extranuclear variation. Clonal increases in mtDNA mutation heteroplasmy have been implicated in aging and disease, although the impact of this shift on cell function is challenging to assess. Reprogramming to pluripotency affects mtDNA mutation heteroplasmy. We reprogrammed three human fibroblast lines with known heteroplasmy for deleterious mtDNA point or deletion mutations. Quantification of mutation heteroplasmy in the resulting 76 induced pluripotent stem cell (iPSC) clones yielded a bimodal distribution, creating three sets of clones with high levels or absent mutation heteroplasmy with matched nuclear genomes. iPSC clones with elevated deletion mutation heteroplasmy show altered growth dynamics, which persist in iPSC-derived progenitor cells. We identify transcriptomic and metabolic shifts consistent with increased investment in neutral lipid synthesis as well as increased epigenetic age in high mtDNA deletion mutation iPSC, consistent with changes occurring in cellular aging. Together, these data demonstrate that high mtDNA mutation heteroplasmy induces changes occurring in cellular aging.
Collapse
Affiliation(s)
- Amy R. Vandiver
- Division of Dermatology, Department of Medicine, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
- Veterans Administration Greater Los Angeles Healthcare SystemLos AngelesCaliforniaUSA
| | - Alejandro Torres
- Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| | - Amberly Sanden
- Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
- Department of BiologyCalifornia State University NorthridgeCaliforniaUSA
| | - Thang L. Nguyen
- Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| | - Jasmine Gasilla
- Division of Dermatology, Department of Medicine, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| | - Mary T. Doan
- Department of Molecular and Medical Pharmacology, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| | - Vahan Martirosian
- Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| | - Austin Hoang
- Division of Geriatrics, Department of Medicine, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| | - Jonathan Wanagat
- Veterans Administration Greater Los Angeles Healthcare SystemLos AngelesCaliforniaUSA
- Division of Geriatrics, Department of Medicine, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| | - Michael A. Teitell
- Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
- Molecular Biology InstituteUniversity of California at Los AngelesLos AngelesCaliforniaUSA
- Department of Bioengineering, California Nano Systems Institute, and Broad Center for Regenerative Medicine and Stem Cell ResearchUniversity of California at Los AngelesLos AngelesCaliforniaUSA
- Department of Pediatrics, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
- Jonsson Comprehensive Cancer Center, David Geffen School of MedicineUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
8
|
Fotopoulou F, Rodríguez-Correa E, Dussiau C, Milsom MD. Reconsidering the usual suspects in age-related hematologic disorders: is stem cell dysfunction a root cause of aging? Exp Hematol 2025; 143:104698. [PMID: 39725143 DOI: 10.1016/j.exphem.2024.104698] [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: 11/11/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Aging exerts a profound impact on the hematopoietic system, leading to increased susceptibility to infections, autoimmune diseases, chronic inflammation, anemia, thrombotic events, and hematologic malignancies. Within the field of experimental hematology, the functional decline of hematopoietic stem cells (HSCs) is often regarded as a primary driver of age-related hematologic conditions. However, aging is clearly a complex multifaceted process involving not only HSCs but also mature blood cells and their interactions with other tissues. This review reappraises an HSC-centric view of hematopoietic aging by exploring how the entire hematopoietic hierarchy, from stem cells to mature cells, contributes to age-related disorders. It highlights the decline of both innate and adaptive immunity, leading to increased susceptibility to infections and cancer, and the rise of autoimmunity as peripheral immune cells undergo aging-induced changes. It explores the concept of "inflammaging," where persistent, low-grade inflammation driven by old immune cells creates a cycle of tissue damage and disease. Additionally, this review delves into the roles of inflammation and homeostatic regulation in age-related conditions such as thrombotic events and anemia, arguing that these issues arise from broader dysfunctions rather than stemming from HSC functional attrition alone. In summary, this review highlights the importance of taking a holistic approach to studying hematopoietic aging and its related pathologies. By looking beyond just stem cells and considering the full spectrum of age-associated changes, one can better capture the complexity of aging and attempt to develop preventative or rejuvenation strategies that target multiple facets of this process.
Collapse
Affiliation(s)
- Foteini Fotopoulou
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Experimental Hematology Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Esther Rodríguez-Correa
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Experimental Hematology Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Charles Dussiau
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Experimental Hematology Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany; Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Michael D Milsom
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Experimental Hematology Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany.
| |
Collapse
|
9
|
Pang L, Tian C, Wang Q, Zhao Z, Pan B, Luo Z, Wu S, Li X, Fan J. An Integrating Microfluidic System for Concentration Gradient Generation of Exosomes and Exosome-Assisted Single-Cell-Derived Tumor-Sphere Formation. ACS Sens 2025; 10:678-688. [PMID: 39866075 DOI: 10.1021/acssensors.4c01542] [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: 01/28/2025]
Abstract
To enhance exploration on tumor stem-like cells (TSCs) without altering their cellular biological characteristics, researchers advocate for application of single-cell-derived tumor-spheres (STSs). TSCs are regulated by their surrounding microenvironment, making it crucial to simulate a tumor microenvironment to facilitate STS formation. Recently, exosomes that originated from the tumor microenvironment have emerged as a promising approach for mimicking the tumor microenvironment. In the tumor microenvironment, various associated cells (such as fibroblasts, endothelial cells, and immune cells) play crucial roles. Utilizing exosomes derived from these cells enabled us to simulate the tumor microenvironment and promote STS formation. Herein, we have developed an integrated microfluidic platform to generate serial concentration gradients and evaluate the effects of multiple exosomes on STS formation. To demonstrate the feasibility of our approach, we generated serial concentration gradients of exosomes derived from two different cell types (HUVEC and NIH/3T3 cells) and assessed their effects on STS formation. Subsequently, we investigated the drug resistance of STSs treated with free doxorubicin and doxorubicin-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles. Our findings revealed that the serial concentration gradients of mixed exosomes could be successfully generated, leading to an enhanced formation rate and size of STSs. Compared to exosomes derived from one cell type, the mixed exosomes exhibited superior promotion of STS formation. Additionally, nanomedicines demonstrated a reduction in the drug resistance of TSCs compared to free drug treatment, particularly in smaller and/or more deformable TSCs. This platform provides an innovative approach for STS formation enhancement and tumor microenvironment simulation.
Collapse
Affiliation(s)
- Long Pang
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Chang Tian
- Public Health School, Anhui University of Science & Technology, Huainan, Anhui 232001, China
| | - Qirui Wang
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
| | - Zhaohua Zhao
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
| | - Bofeng Pan
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
| | - Zichun Luo
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
| | - Shuqiang Wu
- Oncology Department of Xi'an Fengcheng Hospital, Xi'an 710021, China
| | - Xueping Li
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
| | - Jianglin Fan
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
10
|
Maria MKM, Abdel Moniem EM, Hanafy AK, Farag DBE, Radwan IA, Abbass MMS, El Moshy S, Rady D, Dörfer CE, Fawzy El-Sayed KM. Age-Related Oral and Para-Oral Tissue Disorders: The Evolving Therapeutic and Diagnostic Potential of Exosomes. Dent J (Basel) 2025; 13:106. [PMID: 40136734 PMCID: PMC11941486 DOI: 10.3390/dj13030106] [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: 01/02/2025] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
This review highlights the key molecular and cellular mechanisms contributing to aging, such as DNA damage, mitochondrial dysfunction, telomere shortening, protein dysfunction, and defective autophagy. These biological mechanisms are involved in various oral health conditions prevalent in the elderly, including periodontal disease, oral cancer, xerostomia, dental caries, and temporomandibular joint disorders. Exosomes generated by mesenchymal stem cells possess substantial therapeutic potential. These exosomes are nanosized extracellular vesicles derived from cells and are involved in essential intercellular communication and tissue homeostasis. The exosome-based therapies proved superior to traditional cell-based approaches, due to lower immunogenicity, ease of storage, and avoidance of complications associated with cell transplantation. Furthermore, the diagnostic potential of exosomes as non-invasive biomarkers for aging processes and age-related oral diseases offers insights into disease diagnosis, staging, and monitoring. Among the challenges and future perspectives of translating exosome research from preclinical studies to clinical applications is the need for standardized procedures to fully harness the therapeutic and diagnostic capabilities of exosomes.
Collapse
Affiliation(s)
- Mohamed Khaled Mohamed Maria
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
| | | | - Ahmed Khaled Hanafy
- Oral Biology Department, Faculty of Dentistry, Egyptian Russian University, Badr City 11829, Egypt;
| | - Dina B. E. Farag
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
| | - Israa Ahmed Radwan
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 12588, Egypt
| | - Marwa M. S. Abbass
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 12588, Egypt
| | - Sara El Moshy
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 12588, Egypt
| | - Dina Rady
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 12588, Egypt
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24118 Kiel, Germany;
| | - Karim M. Fawzy El-Sayed
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 12588, Egypt
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24118 Kiel, Germany;
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt
| |
Collapse
|
11
|
Joung J, Heo Y, Kim Y, Kim J, Choi H, Jeon T, Jang Y, Kim EJ, Lee SH, Suh JM, Elledge SJ, Kim MS, Kang C. Cell enlargement modulated by GATA4 and YAP instructs the senescence-associated secretory phenotype. Nat Commun 2025; 16:1696. [PMID: 39962062 PMCID: PMC11833096 DOI: 10.1038/s41467-025-56929-0] [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: 04/06/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Dynamic changes in cell size are associated with development and pathological conditions, including aging. Although cell enlargement is a prominent morphological feature of cellular senescence, its functional implications are unknown; moreover, how senescent cells maintain their enlargement state is less understood. Here we show that an extensive remodeling of actin cytoskeleton is necessary for establishing senescence-associated cell enlargement and pro-inflammatory senescence-associated secretory phenotype (SASP). This remodeling is attributed to a balancing act between the SASP regulator GATA4 and the mechanosensor YAP on the expression of the Rho family of GTPase RHOU. Genetic or pharmacological interventions that reduce cell enlargement attenuate SASP with minimal effect on senescence growth arrest. Mechanistically, actin cytoskeleton remodeling couples cell enlargement to the nuclear localization of GATA4 and NF-κB via the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex. RhoU protein accumulates in mouse adipose tissue under senescence-inducing conditions. Furthermore, RHOU expression correlates with SASP expression in adipose tissue during human aging. Thus, our study highlights an unexpected instructive role of cell enlargement in modulating the SASP and reveals a mechanical branch in the senescence regulatory network.
Collapse
Affiliation(s)
- Joae Joung
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Yekang Heo
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Yeonju Kim
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Jaejin Kim
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Haebeen Choi
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Taerang Jeon
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Yeji Jang
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Eun-Jung Kim
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Sang Heon Lee
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, South Korea
| | - Jae Myoung Suh
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, South Korea
| | - Stephen J Elledge
- Department of Genetics, Harvard Medical School and Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Mi-Sung Kim
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea.
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea.
| | - Chanhee Kang
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea.
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
12
|
Rattananinsruang P, Noonin C, Thongboonkerd V. Comparative analysis of various senescence inducers in proximal renal tubular cells. J Pharm Biomed Anal 2025; 254:116571. [PMID: 39579525 DOI: 10.1016/j.jpba.2024.116571] [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: 07/08/2024] [Revised: 11/06/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Senescence in renal cells has attracted wide attention as the critical factor promoting renal fibrosis and chronic kidney disease. Establishing a reliable cellular model is essential to study mechanisms underlying renal cell senescence. Herein, we compared various inducers to define the most suitable senescence inducer for HK-2 proximal tubular cells. These inducers included hydrogen peroxide (H2O2), high-temperature (HT), glucose, mannitol and hydroxyurea (HU). To screen for optimal concentration/level, the highest concentration/level of each inducer that did not increase cell death (to avoid severe toxicity) was selected for senescence induction and comparative analysis using the two most appropriate markers for HK-2 cell senescence as recently established. The data revealed that 0.4 mM, 43 °C, 80 mM, 80 mM and 100 μM were the optimal concentrations/levels of H2O2, HT, glucose, mannitol and HU, respectively. Comparative analysis using optimal concentration/level of each marker revealed that 0.4 mM H2O2, HT at 43 °C, 80 mM glucose and 80 mM mannitol were the weak senescence inducers. The most effective inducer for HK-2 senescence was 100 μM HU, which provided the greatest fold-changes of cell area and granularity when compared with other stimuli in a time-dependent manner. Based on these data comparing H2O2, HT, glucose, mannitol and HU at their optimal concentrations/levels, 100 μM HU seems to be most effective for senescence induction in HK-2 cells for in vitro study of proximal renal tubular cells.
Collapse
Affiliation(s)
- Piyaporn Rattananinsruang
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chadanat Noonin
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
13
|
Barve A, Cornwell A, Sriram P, Kopyov A, Dabas P, Kooienga E, Kelley Z, Johnson J, Myers JA, Obeng EA, Kang G, Olufadi Y, Cain T, Talbot L, Spence D, Cortes M, Miller S, Loeffler D, Sharma A, McKinney-Freeman S. Senolytics restore hematopoietic stem cell function in sickle cell disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.08.636742. [PMID: 39975016 PMCID: PMC11839116 DOI: 10.1101/2025.02.08.636742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Sickle Cell Disease (SCD) is a blood disorder affecting millions worldwide. Emerging evidence reveals that SCD pathophysiology increases risk of myeloid malignancies and hematopoietic stem cell (HSC) dysfunction, possibly due to pathological stress on bone marrow. To investigate this further, we interrogated mice and individuals with SCD and observed extended cell cycle times, oxidative stress, DNA damage, senescence, and dysregulation of molecular programs associated with these processes in bone marrow hematopoietic stem and progenitor cells (HSPCs). Human SCD HSPCs displayed poor hematopoietic potential ex vivo . SCD mice displayed a dramatic loss of transplantable bone marrow HSPCs, which was reversed upon treatment of SCD mice with the senolytic agent, ABT-263 (navitoclax). Thus, senolytics restore bone marrow function during SCD in mice and represent a novel strategy to improve bone marrow health in individuals with SCD and improve the safety of potentially curative gene therapies that utilize autologous HSPCs from individuals with SCD.
Collapse
|
14
|
MOON HAEUN, DU JINHONG, LEI JING, ROEDER KATHRYN. AUGMENTED DOUBLY ROBUST POST-IMPUTATION INFERENCE FOR PROTEOMIC DATA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.23.586387. [PMID: 39868108 PMCID: PMC11761724 DOI: 10.1101/2024.03.23.586387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Quantitative measurements produced by mass spectrometry proteomics experiments offer a direct way to explore the role of proteins in molecular mechanisms. However, analysis of such data is challenging due to the large proportion of missing values. A common strategy to address this issue is to utilize an imputed dataset, which often introduces systematic bias into downstream analyses if the imputation errors are ignored. In this paper, we propose a statistical framework inspired by doubly robust estimators that offers valid and efficient inference for proteomic data. Our framework combines powerful machine learning tools, such as variational autoencoders, to augment the imputation quality with high-dimensional peptide data, and a parametric model to estimate the propensity score for debiasing imputed outcomes. Our estimator is compatible with the double machine learning framework and has provable properties. Simulation studies verify its empirical superiority over other existing procedures. In application to both single-cell proteomic data and bulk-cell Alzheimer's Disease data our method utilizes the imputed data to gain additional, meaningful discoveries and yet maintains good control of false positives.
Collapse
Affiliation(s)
- HAEUN MOON
- Department of Statistics, Seoul National University
| | - JIN-HONG DU
- Department of Statistics and Data Science, Carnegie Mellon University
| | - JING LEI
- Department of Statistics and Data Science, Carnegie Mellon University
| | - KATHRYN ROEDER
- Department of Statistics and Data Science, Carnegie Mellon University
| |
Collapse
|
15
|
Campos J, Sampaio-Marques B, Santos D, Barata-Antunes S, Ribeiro M, Serra SC, Pinho TS, Canto-Gomes J, Marote A, Cortez M, Silva NA, Michael-Titus AT, Salgado AJ. Lipid Priming of Adipose Mesenchymal Stromal Cells with Docosahexaenoic Acid: Impact on Cell Differentiation, Senescence and the Secretome Neuroregulatory Profile. Tissue Eng Regen Med 2025; 22:113-128. [PMID: 39495459 PMCID: PMC11711600 DOI: 10.1007/s13770-024-00679-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/23/2024] [Accepted: 10/02/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Priming strategies that improve the functionality of MSCs may be required to address issues limiting successful clinical translation of MSC therapies. For conditions requiring high trophic support such as brain and spinal cord injuries, priming MSCs to produce higher levels of trophic factors may be instrumental to facilitate translation of current MSC therapies. We developed and tested a novel molecular priming paradigm using docosahexaenoic acid (DHA) to prime adipose tissue-derived mesenchymal stromal cells (ASCs) to enhance the secretome neuroregulatory potential. METHODS Comprehensive dose-response and time-course assays were carried to determine an optimal priming protocol. Secretome total protein measurements were taken in association with cell viability, density and morphometric assessments. Cell identity and differentiation capacity were studied by flow cytometry and lineage-specific markers. Cell growth was assessed by trypan-blue exclusion and senescence was probed over time using SA-β-gal, morphometry and gene expression. Secretomes were tested for their ability to support differentiation and neurite outgrowth of human neural progenitor cells (hNPCs). Neuroregulatory proteins in the secretome were identified using multiplex membrane arrays. RESULTS Priming with 40 µM DHA for 72 h significantly enhanced the biosynthetic capacity of ASCs, producing a secretome with higher protein levels and increased metabolic viability. DHA priming enhanced ASCs adipogenic differentiation and adapted their responses to replicative senescence induction. Furthermore, priming increased concentrations of neurotrophic factors in the secretome promoting neurite outgrowth and modulating the differentiation of hNPCs. CONCLUSIONS These results provide proof-of-concept evidence that DHA priming is a viable strategy to improve the neuroregulatory profile of ASCs.
Collapse
Affiliation(s)
- Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Diogo Santos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sandra Barata-Antunes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Miguel Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sofia C Serra
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Tiffany S Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João Canto-Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Marote
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Margarida Cortez
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Adina T Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
16
|
Ho KYL, Ou AYJ, Samuelson N, Tanentzapf G. Novel features of Drosophila hematopoiesis uncovered by long-term live imaging. Dev Biol 2025; 517:286-300. [PMID: 39536928 DOI: 10.1016/j.ydbio.2024.10.004] [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: 05/25/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Stem cells are subject to continuous regulation to ensure that the correct balance between stem cell differentiation and self-renewal is maintained. The dynamic and ongoing nature of stem cell regulation, as well as the complex signaling microenvironment in which stem cells are typically found, means that studying them in their endogenous environment in real time has multiple advantages over static fixed-sample approaches. We recently described a method for long-term, ex-vivo, live imaging of the blood progenitors in the Drosophila larval hematopoietic organ, the Lymph Gland (LG). This methodology has allowed us to analyze multiple aspects of fly hematopoiesis, in real time, in a manner that could not be carried out previously. Here, we describe novel insights derived from our quantitative live imaging approach. These insights include: the identification of extensive filopodia in the progenitors and description of their morphology and dynamics; visualization and quantitative analysis of JAK/STAT signaling in progenitors by the simultaneous tracking of thousands of vesicles containing internalized Domeless receptors; quantitative analysis of the location, morphology, and dynamics of mitochondria in blood progenitors; long-term tracking of patterns of cell division and migration of mature blood cell in the LG; long-term tracking of multiple cell behaviors in the distal committed progenitors; analysis of Ca2+ signaling of blood progenitors in the secondary lobes of the LG. Together, these observations illustrate the power of imaging fly hematopoiesis in real time and identify many previously undescribed processes and behaviors in the LG that are likely to play important roles in the regulation of progenitor differentiation and self-renewal.
Collapse
Affiliation(s)
- Kevin Y L Ho
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Annie Y J Ou
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada; School of Kinesiology, University of British Columbia, Vancouver, V6T 1Z1, Canada; Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Nicholas Samuelson
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
17
|
Lanz MC, Zhang S, Swaffer MP, Ziv I, Götz LH, Kim J, McCarthy F, Jarosz DF, Elias JE, Skotheim JM. Genome dilution by cell growth drives starvation-like proteome remodeling in mammalian and yeast cells. Nat Struct Mol Biol 2024; 31:1859-1871. [PMID: 39048803 DOI: 10.1038/s41594-024-01353-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 06/12/2024] [Indexed: 07/27/2024]
Abstract
Cell size is tightly controlled in healthy tissues and single-celled organisms, but it remains unclear how cell size influences physiology. Increasing cell size was recently shown to remodel the proteomes of cultured human cells, demonstrating that large and small cells of the same type can be compositionally different. In the present study, we utilize the natural heterogeneity of hepatocyte ploidy and yeast genetics to establish that the ploidy-to-cell size ratio is a highly conserved determinant of proteome composition. In both mammalian and yeast cells, genome dilution by cell growth elicits a starvation-like phenotype, suggesting that growth in large cells is restricted by genome concentration in a manner that mimics a limiting nutrient. Moreover, genome dilution explains some proteomic changes ascribed to yeast aging. Overall, our data indicate that genome concentration drives changes in cell composition independently of external environmental cues.
Collapse
Affiliation(s)
- Michael C Lanz
- Department of Biology, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA.
| | - Shuyuan Zhang
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Inbal Ziv
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | | | - Jacob Kim
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Frank McCarthy
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA
| | - Daniel F Jarosz
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
| | - Joshua E Elias
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA
| | - Jan M Skotheim
- Department of Biology, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA.
| |
Collapse
|
18
|
Rizzo HE, Zhang AL, Gardel ML. Mechanochemical control systems regulating animal cell size. Curr Opin Cell Biol 2024; 91:102443. [PMID: 39504614 DOI: 10.1016/j.ceb.2024.102443] [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: 07/08/2024] [Revised: 09/27/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024]
Abstract
Cell size regulation arises from physical manifestations of cell proliferation and metabolic pathways. On one hand, coordination between these systems yields a constant cell size over generations to maintain cell size homeostasis. However, active regulation of cell size is crucial to physiology and to establish broad variation of cell sizes within an individual organism, and is accomplished via physical and biochemical pathways modulated by myriad intrinsic and extrinsic cues. In this review, we explore recent data elucidating the mechanobiological regulation of the volume of animal cells and its coordination with metabolic and proliferative pathways.
Collapse
Affiliation(s)
- Heather E Rizzo
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Andy L Zhang
- Department of Physics, The University of Chicago, Chicago, IL 60637, USA
| | - Margaret L Gardel
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA; Department of Physics, The University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60613, USA; CZ Biohub Chicago, LLC, Chicago, IL 60642, USA.
| |
Collapse
|
19
|
Kleeblatt E, Lazki-Hagenbach P, Nabet E, Cohen R, Bahri R, Rogers N, Langton A, Bulfone-Paus S, Frenkel D, Sagi-Eisenberg R. p16 Ink4a-induced senescence in cultured mast cells as a model for ageing reveals significant morphological and functional changes. Immun Ageing 2024; 21:77. [PMID: 39529115 PMCID: PMC11552350 DOI: 10.1186/s12979-024-00478-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Mast cells (MCs) are tissue resident cells of the immune system, mainly known for their role in allergy. However, mounting evidence indicates their involvement in the pathology of age-related diseases, such as Alzheimer's disease, Parkinson's disease, and cancer. MC numbers increase in aged tissues, but how ageing affects MCs is poorly understood. RESULTS We show that MC ageing is associated with the increased expression of the cell cycle inhibitor p16 Ink4a, a marker and inducer of cellular senescence. Relying on this observation and the tight association of ageing with senescence, we developed a model of inducible senescence based on doxycycline-induced expression of p16Ink4a in cultured bone marrow derived MCs (BMMCs). Using this model, we show that senescent MCs upregulate IL-1β, TNF-α and VEGF-A. We also demonstrate that senescence causes marked morphological changes that impact MC function. Senescent MCs are larger, contain a larger number of secretory granules (SGs) and have less membrane protrusions. Particularly striking are the changes in their SGs, reflected in a significant reduction in the number of electron dense SGs with a concomitant increase in lucent SGs containing intraluminal vesicles. The changes in SG morphology are accompanied by changes in MC degranulation, including a significant increase in receptor-triggered release of CD63-positive extracellular vesicles (EVs) and the exteriorisation of proteoglycans, as opposed to a gradual inhibition of the release of β-hexosaminidase. CONCLUSIONS The inducible expression of p16Ink4a imposes MC senescence, providing a model for tracking the autonomous changes that occur in MCs during ageing. These changes include both morphological and functional alterations. In particular, the increased release of small EVs by senescent MCs suggests an enhanced ability to modulate neighbouring cells.
Collapse
Affiliation(s)
- Elisabeth Kleeblatt
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Pia Lazki-Hagenbach
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ellon Nabet
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Reli Cohen
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Rajia Bahri
- Lydia Becker Institute of Immunology and Inflammation, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Nicholas Rogers
- Department of Environmental Studies, School of Mechanical Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Abigail Langton
- Lydia Becker Institute of Immunology and Inflammation, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Ronit Sagi-Eisenberg
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, 69978, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
20
|
Fritz García JHG, Keller Valsecchi CI, Basilicata MF. Sex as a biological variable in ageing: insights and perspectives on the molecular and cellular hallmarks. Open Biol 2024; 14:240177. [PMID: 39471841 PMCID: PMC11521605 DOI: 10.1098/rsob.240177] [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: 06/25/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 11/01/2024] Open
Abstract
Sex-specific differences in lifespan and ageing are observed in various species. In humans, women generally live longer but are frailer and suffer from different age-related diseases compared to men. The hallmarks of ageing, such as genomic instability, telomere attrition or loss of proteostasis, exhibit sex-specific patterns. Sex chromosomes and sex hormones, as well as the epigenetic regulation of the inactive X chromosome, have been shown to affect lifespan and age-related diseases. Here we review the current knowledge on the biological basis of sex-biased ageing. While our review is focused on humans, we also discuss examples of model organisms such as the mouse, fruit fly or the killifish. Understanding these molecular differences is crucial as the elderly population is expected to double worldwide by 2050, making sex-specific approaches in the diagnosis, treatment, therapeutic development and prevention of age-related diseases a pressing need.
Collapse
Affiliation(s)
| | | | - M. Felicia Basilicata
- Institute of Molecular Biology (IMB), Mainz, Germany
- University Medical Center (UMC), Mainz, Germany
| |
Collapse
|
21
|
Chadha Y, Khurana A, Schmoller KM. Eukaryotic cell size regulation and its implications for cellular function and dysfunction. Physiol Rev 2024; 104:1679-1717. [PMID: 38900644 PMCID: PMC11495193 DOI: 10.1152/physrev.00046.2023] [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: 12/26/2023] [Revised: 05/24/2024] [Accepted: 06/19/2024] [Indexed: 06/22/2024] Open
Abstract
Depending on cell type, environmental inputs, and disease, the cells in the human body can have widely different sizes. In recent years, it has become clear that cell size is a major regulator of cell function. However, we are only beginning to understand how the optimization of cell function determines a given cell's optimal size. Here, we review currently known size control strategies of eukaryotic cells and the intricate link of cell size to intracellular biomolecular scaling, organelle homeostasis, and cell cycle progression. We detail the cell size-dependent regulation of early development and the impact of cell size on cell differentiation. Given the importance of cell size for normal cellular physiology, cell size control must account for changing environmental conditions. We describe how cells sense environmental stimuli, such as nutrient availability, and accordingly adapt their size by regulating cell growth and cell cycle progression. Moreover, we discuss the correlation of pathological states with misregulation of cell size and how for a long time this was considered a downstream consequence of cellular dysfunction. We review newer studies that reveal a reversed causality, with misregulated cell size leading to pathophysiological phenotypes such as senescence and aging. In summary, we highlight the important roles of cell size in cellular function and dysfunction, which could have major implications for both diagnostics and treatment in the clinic.
Collapse
Affiliation(s)
- Yagya Chadha
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Arohi Khurana
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Kurt M Schmoller
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
22
|
Kumar S, Vassallo JD, Nattamai KJ, Hassan A, Vollmer A, Karns R, Sacma M, Nemkov T, D'Alessandro A, Geiger H. Rejuvenation of the reconstitution potential and reversal of myeloid bias of aged HSCs upon pH treatment. Aging Cell 2024; 23:e14324. [PMID: 39236298 PMCID: PMC11464122 DOI: 10.1111/acel.14324] [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: 02/18/2024] [Revised: 07/18/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Aged hematopoietic stem cells (HSCs) show reduced reconstitution potential, limiting their use in transplantation settings in the clinic. We demonstrate here that exposure of aged HSCs ex vivo to a pH of 6.9 instead of the commonly used pH of 7.4 results in enhanced HSCs potential that is consistent with rejuvenation, including attenuation of the myeloid bias of aged HSC and restoration of a youthful frequency of epigenetic polarity. Rejuvenation of aged HSCs by pH 6.9 is, at least in part, due to alterations in the polyamine/methionine pathway within pH 6.9 HSCs, and consequently, attenuation of the production of spermidine also attenuated aging of HSCs. Exposure of aged HSCs to pH 6.9, or pharmacological targeting of the polyamine pathway, might thus extend the use of HSCs from aged donors for therapeutic applications.
Collapse
Affiliation(s)
- Sachin Kumar
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Research FoundationCincinnatiOhioUSA
- Pharmacology DivisionCSIR‐Central Drug Research InstituteLucknowIndia
| | - Jeffrey D. Vassallo
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Research FoundationCincinnatiOhioUSA
| | - Kalpana J. Nattamai
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Research FoundationCincinnatiOhioUSA
| | - Aishlin Hassan
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Research FoundationCincinnatiOhioUSA
| | | | - Rebekah Karns
- Division of Gastroenterology, Hepatology and NutritionCincinnati Children's Hospital Medical Center and University of CincinnatiCincinnatiOhioUSA
| | - Mehmet Sacma
- Institute of Molecular MedicineUlm UniversityUlmGermany
| | - Travis Nemkov
- University of Colorado Denver—Anschutz Medical CampusAuroraColoradoUSA
| | | | - Hartmut Geiger
- Institute of Molecular MedicineUlm UniversityUlmGermany
- Aging Research CenterUlm UniversityUlmGermany
| |
Collapse
|
23
|
Wiatrak B, Rayad S, Gębarowski T, Hadzik J, Styczyńska M, Gedrange T, Dobrzyński M, Barg E, Dominiak M. Comparative Analysis of Heavy Metal Content in Impacted Third Molars from Industrial and Non-Industrial Areas and Its Effect on the Isolation, Culture, and Proliferation of Dental Stem Cells (DSCs). J Clin Med 2024; 13:5465. [PMID: 39336954 PMCID: PMC11432618 DOI: 10.3390/jcm13185465] [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: 08/03/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Background: This study investigates the impact of environmental pollution on the quality and viability of dental stem cells (DSCs) from impacted third molars. By comparing DSCs from patients in industrial areas with high air pollution and those from non-industrial regions, the research assesses the adverse effects of heavy metals on stem cell proliferation. Methods: Impacted lower third molars were collected from 28 patients-10 from industrial and 18 from non-industrial areas. Patients were divided into two age groups: 18-27 years and 28-38 years old. Dental pulp was extracted under sterile conditions, and DSCs were isolated and cultured. Heavy metal concentrations in dental tissues were measured using atomic absorption/emission spectrometry. Results: The study found significantly higher concentrations of copper and lead in the dental tissues of patients in industrial areas. Cell viability was lower in samples from these areas, with a statistically significant difference in average doubling time and the number of cells obtained after the first passage. There was no significant impact of gender on heavy metal content, except for higher iron levels in men. Conclusions: Exposure to industrial pollutants negatively affects the viability and proliferation of DSCs, but there are no differences in differentiation in the osteogenic medium regarding cell mineralization. These studies highlight the importance of environmental factors for oral health, suggesting that residents of polluted areas may face greater difficulties in dental and regenerative treatments. Further research is needed to develop strategies to mitigate the effects and improve clinical outcomes for affected populations.
Collapse
Affiliation(s)
- Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland;
| | - Sadri Rayad
- Academic Dental Polyclinic of Dental Center, Technology Transfer Ltd., Krakowska 26, 50-425 Wroclaw, Poland;
| | - Tomasz Gębarowski
- Department of Biostructure and Animal Physiology, The Wroclaw University of Environmental and Life Sciences, Kożuchowska 1/3, 51-631 Wroclaw, Poland
| | - Jakub Hadzik
- Department of Dental Surgery, Wroclaw Medical University, Krakowska 26, 50-425 Wroclaw, Poland; (J.H.); (T.G.); (M.D.)
| | - Marzena Styczyńska
- Department of Human Nutrition, Wroclaw University of Environmental and Life Sciences, Chelmonskiego 37/41, 51-630 Wroclaw, Poland;
| | - Tomasz Gedrange
- Department of Dental Surgery, Wroclaw Medical University, Krakowska 26, 50-425 Wroclaw, Poland; (J.H.); (T.G.); (M.D.)
| | - Maciej Dobrzyński
- Department of Pediatric Dentistry and Preclinical Dentistry, Wroclaw Medical University, Krakowska 26, 50-425 Wroclaw, Poland;
| | - Ewa Barg
- Department of Basic Medical Sciences, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Marzena Dominiak
- Department of Dental Surgery, Wroclaw Medical University, Krakowska 26, 50-425 Wroclaw, Poland; (J.H.); (T.G.); (M.D.)
| |
Collapse
|
24
|
Das P, Murthy S, Abbas E, White K, Arya R. The Hox Gene, abdominal A controls timely mitotic entry of neural stem cell and their growth during CNS development in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611161. [PMID: 39282366 PMCID: PMC11398374 DOI: 10.1101/2024.09.04.611161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
The size of a cell is important for its function and physiology. Interestingly, size variation can be easily observed in clonally derived embryonic and hematopoietic stem cells. Here, we investigated the regulation of stem cell growth and its association with cell fate. We observed heterogeneous sizes of neuroblasts or neural stem cells (NSCs) in the Drosophila ventral nerve cord (VNC). Specifically, thoracic NSCs were larger than those in the abdominal region of the VNC. Our research uncovered a significant role of the Hox gene abdominal A (abdA) in the regulation of abdominal NSC growth. Developmental expression of AbdA retards their growth and delays mitotic entry compared to thoracic NSCs. The targeted loss of abdA enhanced their growth and caused an earlier entry into mitosis with a faster cycling rate. Furthermore, ectopic expression of abdA reduced the size of thoracic NSCs and delayed their entry into mitosis. We suggest that abdA plays an instructive role in regulating NSC size and exit from quiescence. This study demonstrates for the first time the involvement of abdA in NSC fate determination by regulating their growth, entry into mitosis and proliferation rate, and thus their potential to make appropriate number of progeny for CNS patterning.
Collapse
Affiliation(s)
- Papri Das
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi-221005
| | | | - Eshan Abbas
- ADP Road, Christianpatty, Nagaon, Assam- 782003, India
| | - Kristin White
- MGH/Harvard Medical School,CBRC, Bldg 149, 13th St, Charlestown, MA 02129
| | - Richa Arya
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi-221005
| |
Collapse
|
25
|
Jiang L, Yan C, Yi Y, Zhu L, Liu Z, Zhang D, Jiang W. Cell size regulates human endoderm specification through actomyosin-dependent AMOT-YAP signaling. Stem Cell Reports 2024; 19:1137-1155. [PMID: 39094563 PMCID: PMC11368700 DOI: 10.1016/j.stemcr.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Cell size is a crucial physical property that significantly impacts cellular physiology and function. However, the influence of cell size on stem cell specification remains largely unknown. Here, we investigated the dynamic changes in cell size during the differentiation of human pluripotent stem cells into definitive endoderm (DE). Interestingly, cell size exhibited a gradual decrease as DE differentiation progressed with higher stiffness. Furthermore, the application of hypertonic pressure or chemical to accelerate the reduction in cell size significantly and specifically enhanced DE differentiation. By functionally intervening in mechanosensitive elements, we have identified actomyosin activity as a crucial mediator of both DE differentiation and cell size reduction. Mechanistically, the reduction in cell size induces actomyosin-dependent angiomotin (AMOT) nuclear translocation, which suppresses Yes-associated protein (YAP) activity and thus facilitates DE differentiation. Together, our study has established a novel connection between cell size diminution and DE differentiation, which is mediated by AMOT nuclear translocation. Additionally, our findings suggest that the application of osmotic pressure can effectively promote human endodermal lineage differentiation.
Collapse
Affiliation(s)
- Lai Jiang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China; Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Chenchao Yan
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Ying Yi
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Lihang Zhu
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Zheng Liu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China.
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| |
Collapse
|
26
|
Rybiczka-Tešulov M, Garritsen O, Venø MT, Wieg L, Dijk RV, Rahimi K, Gomes-Duarte A, Wit MD, van de Haar LL, Michels L, van Kronenburg NCH, van der Meer C, Kjems J, Vangoor VR, Pasterkamp RJ. Circular RNAs regulate neuron size and migration of midbrain dopamine neurons during development. Nat Commun 2024; 15:6773. [PMID: 39117691 PMCID: PMC11310423 DOI: 10.1038/s41467-024-51041-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Midbrain dopamine (mDA) neurons play an essential role in cognitive and motor behaviours and are linked to different brain disorders. However, the molecular mechanisms underlying their development, and in particular the role of non-coding RNAs (ncRNAs), remain incompletely understood. Here, we establish the transcriptomic landscape and alternative splicing patterns of circular RNAs (circRNAs) at key developmental timepoints in mouse mDA neurons in vivo using fluorescence-activated cell sorting followed by short- and long-read RNA sequencing. In situ hybridisation shows expression of several circRNAs during early mDA neuron development and post-transcriptional silencing unveils roles for different circRNAs in regulating mDA neuron morphology. Finally, in utero electroporation and time-lapse imaging implicate circRmst, a circRNA with widespread morphological effects, in the migration of developing mDA neurons in vivo. Together, these data for the first time suggest a functional role for circRNAs in developing mDA neurons and characterise poorly defined aspects of mDA neuron development.
Collapse
Affiliation(s)
- Mateja Rybiczka-Tešulov
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Oxana Garritsen
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Morten T Venø
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus C, Denmark
- Omiics ApS, Aarhus N, Denmark
| | - Laura Wieg
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Roland van Dijk
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- VectorY Therapeutics, Matrix Innovation Center VI, Amsterdam, The Netherlands
| | - Karim Rahimi
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus C, Denmark
- Department of Genetics, Blavatnik Institute, Harvard Medical School, MA, Boston, USA
| | - Andreia Gomes-Duarte
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- VectorY Therapeutics, Matrix Innovation Center VI, Amsterdam, The Netherlands
| | - Marina de Wit
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lieke L van de Haar
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Berlin Institute for Medical Systems Biology, Max Delbrück Center, Berlin, Germany
| | - Lars Michels
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- VectorY Therapeutics, Matrix Innovation Center VI, Amsterdam, The Netherlands
| | - Nicky C H van Kronenburg
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christiaan van der Meer
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus C, Denmark
| | - Vamshidhar R Vangoor
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
27
|
Yin K, Büttner M, Deligiannis IK, Strzelecki M, Zhang L, Talavera-López C, Theis F, Odom DT, Martinez-Jimenez CP. Polyploidisation pleiotropically buffers ageing in hepatocytes. J Hepatol 2024; 81:289-302. [PMID: 38583492 DOI: 10.1016/j.jhep.2024.03.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND & AIMS Polyploidy in hepatocytes has been proposed as a genetic mechanism to buffer against transcriptional dysregulation. Here, we aim to demonstrate the role of polyploidy in modulating gene regulatory networks in hepatocytes during ageing. METHODS We performed single-nucleus RNA sequencing in hepatocyte nuclei of different ploidy levels isolated from young and old wild-type mice. Changes in the gene expression and regulatory network were compared to three independent strains that were haploinsufficient for HNF4A, CEBPA or CTCF, representing non-deleterious perturbations. Phenotypic characteristics of the liver section were additionally evaluated histologically, whereas the genomic allele composition of hepatocytes was analysed by BaseScope. RESULTS We observed that ageing in wild-type mice results in nuclei polyploidy and a marked increase in steatosis. Haploinsufficiency of liver-specific master regulators (HFN4A or CEBPA) results in the enrichment of hepatocytes with tetraploid nuclei at a young age, affecting the genomic regulatory network, and dramatically suppressing ageing-related steatosis tissue wide. Notably, these phenotypes are not the result of subtle disruption to liver-specific transcriptional networks, since haploinsufficiency in the CTCF insulator protein resulted in the same phenotype. Further quantification of genotypes of tetraploid hepatocytes in young and old HFN4A-haploinsufficient mice revealed that during ageing, tetraploid hepatocytes lead to the selection of wild-type alleles, restoring non-deleterious genetic perturbations. CONCLUSIONS Our results suggest a model whereby polyploidisation leads to fundamentally different cell states. Polyploid conversion enables pleiotropic buffering against age-related decline via non-random allelic segregation to restore a wild-type genome. IMPACT AND IMPLICATIONS The functional role of hepatocyte polyploidisation during ageing is poorly understood. Using single-nucleus RNA sequencing and BaseScope approaches, we have studied ploidy dynamics during ageing in murine livers with non-deleterious genetic perturbations. We have identified that hepatocytes present different cellular states and the ability to buffer ageing-associated dysfunctions. Tetraploid nuclei exhibit robust transcriptional networks and are better adapted to genomically overcome perturbations. Novel therapeutic interventions aimed at attenuating age-related changes in tissue function could be exploited by manipulation of ploidy dynamics during chronic liver conditions.
Collapse
Affiliation(s)
- Kelvin Yin
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany
| | - Maren Büttner
- Institute of Computational Biology, Computational Health Department, Helmholtz Munich, Neuherberg, Germany
| | | | | | - Liwei Zhang
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany
| | - Carlos Talavera-López
- Division of Infectious Diseases and Tropical Medicine, Ludwig-Maximilian-Universität Klinikum, Germany
| | - Fabian Theis
- Institute of Computational Biology, Computational Health Department, Helmholtz Munich, Neuherberg, Germany; Technical University of Munich, Department of Mathematics, 85748 Garching. Munich, Germany; German Cancer Research Centre, Heidelberg, Germany.
| | - Duncan T Odom
- German Cancer Research Center, Division of Regulatory Genomics and Cancer Evolution (B270), Heidelberg, Germany; Cancer Research UK Cambridge Institute, University of Cambridge, CB20RE, United Kingdom.
| | - Celia P Martinez-Jimenez
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany; TUM School of Medicine, Technical University of Munich, Munich, Germany; Institute of Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, Spain.
| |
Collapse
|
28
|
Manohar S, Neurohr GE. Too big not to fail: emerging evidence for size-induced senescence. FEBS J 2024; 291:2291-2305. [PMID: 37986656 DOI: 10.1111/febs.16983] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/02/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023]
Abstract
Cellular senescence refers to a permanent and stable state of cell cycle exit. This process plays an important role in many cellular functions, including tumor suppression. It was first noted that senescence is associated with increased cell size in the early 1960s; however, how this contributes to permanent cell cycle exit was poorly understood until recently. In this review, we discuss new findings that identify increased cell size as not only a consequence but also a cause of permanent cell cycle exit. We highlight recent insights into how increased cell size alters normal cellular physiology and creates homeostatic imbalances that contribute to senescence induction. Finally, we focus on the potential clinical implications of these findings in the context of cell cycle arrest-causing cancer therapeutics and speculate on how tumor cell size changes may impact outcomes in patients treated with these drugs.
Collapse
Affiliation(s)
- Sandhya Manohar
- Department of Biology, Institute for Biochemistry, ETH Zürich, Switzerland
| | - Gabriel E Neurohr
- Department of Biology, Institute for Biochemistry, ETH Zürich, Switzerland
| |
Collapse
|
29
|
Verma S, Lin X, Coulson-Thomas VJ. The Potential Reversible Transition between Stem Cells and Transient-Amplifying Cells: The Limbal Epithelial Stem Cell Perspective. Cells 2024; 13:748. [PMID: 38727284 PMCID: PMC11083486 DOI: 10.3390/cells13090748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Stem cells (SCs) undergo asymmetric division, producing transit-amplifying cells (TACs) with increased proliferative potential that move into tissues and ultimately differentiate into a specialized cell type. Thus, TACs represent an intermediary state between stem cells and differentiated cells. In the cornea, a population of stem cells resides in the limbal region, named the limbal epithelial stem cells (LESCs). As LESCs proliferate, they generate TACs that move centripetally into the cornea and differentiate into corneal epithelial cells. Upon limbal injury, research suggests a population of progenitor-like cells that exists within the cornea can move centrifugally into the limbus, where they dedifferentiate into LESCs. Herein, we summarize recent advances made in understanding the mechanism that governs the differentiation of LESCs into TACs, and thereafter, into corneal epithelial cells. We also outline the evidence in support of the existence of progenitor-like cells in the cornea and whether TACs could represent a population of cells with progenitor-like capabilities within the cornea. Furthermore, to gain further insights into the dynamics of TACs in the cornea, we outline the most recent findings in other organ systems that support the hypothesis that TACs can dedifferentiate into SCs.
Collapse
Affiliation(s)
- Sudhir Verma
- College of Optometry, University of Houston, 4901 Calhoun Road, Houston, TX 77204, USA;
- Deen Dayal Upadhyaya College, University of Delhi, Delhi 110078, India
| | - Xiao Lin
- College of Optometry, University of Houston, 4901 Calhoun Road, Houston, TX 77204, USA;
| | | |
Collapse
|
30
|
Xie S, Zhang S, de Medeiros G, Liberali P, Skotheim JM. The G1/S transition in mammalian stem cells in vivo is autonomously regulated by cell size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588781. [PMID: 38645246 PMCID: PMC11030448 DOI: 10.1101/2024.04.09.588781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Cell growth and division must be coordinated to maintain a stable cell size, but how this coordination is implemented in multicellular tissues remains unclear. In unicellular eukaryotes, autonomous cell size control mechanisms couple cell growth and division with little extracellular input. However, in multicellular tissues we do not know if autonomous cell size control mechanisms operate the same way or whether cell growth and cell cycle progression are separately controlled by cell-extrinsic signals. Here, we address this question by tracking single epidermal stem cells growing in adult mice. We find that a cell-autonomous size control mechanism, dependent on the RB pathway, sets the timing of S phase entry based on the cell's current size. Cell-extrinsic variations in the cellular microenvironment affect cell growth rates but not this autonomous coupling. Our work reassesses long-standing models of cell cycle regulation within complex metazoan tissues and identifies cell-autonomous size control as a critical mechanism regulating cell divisions in vivo and thereby a major contributor to stem cell heterogeneity.
Collapse
|
31
|
Watanuki S, Kobayashi H, Sugiura Y, Yamamoto M, Karigane D, Shiroshita K, Sorimachi Y, Fujita S, Morikawa T, Koide S, Oshima M, Nishiyama A, Murakami K, Haraguchi M, Tamaki S, Yamamoto T, Yabushita T, Tanaka Y, Nagamatsu G, Honda H, Okamoto S, Goda N, Tamura T, Nakamura-Ishizu A, Suematsu M, Iwama A, Suda T, Takubo K. Context-dependent modification of PFKFB3 in hematopoietic stem cells promotes anaerobic glycolysis and ensures stress hematopoiesis. eLife 2024; 12:RP87674. [PMID: 38573813 PMCID: PMC10994660 DOI: 10.7554/elife.87674] [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: 04/06/2024] Open
Abstract
Metabolic pathways are plastic and rapidly change in response to stress or perturbation. Current metabolic profiling techniques require lysis of many cells, complicating the tracking of metabolic changes over time after stress in rare cells such as hematopoietic stem cells (HSCs). Here, we aimed to identify the key metabolic enzymes that define differences in glycolytic metabolism between steady-state and stress conditions in murine HSCs and elucidate their regulatory mechanisms. Through quantitative 13C metabolic flux analysis of glucose metabolism using high-sensitivity glucose tracing and mathematical modeling, we found that HSCs activate the glycolytic rate-limiting enzyme phosphofructokinase (PFK) during proliferation and oxidative phosphorylation (OXPHOS) inhibition. Real-time measurement of ATP levels in single HSCs demonstrated that proliferative stress or OXPHOS inhibition led to accelerated glycolysis via increased activity of PFKFB3, the enzyme regulating an allosteric PFK activator, within seconds to meet ATP requirements. Furthermore, varying stresses differentially activated PFKFB3 via PRMT1-dependent methylation during proliferative stress and via AMPK-dependent phosphorylation during OXPHOS inhibition. Overexpression of Pfkfb3 induced HSC proliferation and promoted differentiated cell production, whereas inhibition or loss of Pfkfb3 suppressed them. This study reveals the flexible and multilayered regulation of HSC glycolytic metabolism to sustain hematopoiesis under stress and provides techniques to better understand the physiological metabolism of rare hematopoietic cells.
Collapse
Affiliation(s)
- Shintaro Watanuki
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and MedicineTokyoJapan
- Division of Hematology, Department of Medicine, Keio University School of MedicineTokyoJapan
| | - Hiroshi Kobayashi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and MedicineTokyoJapan
- Department of Cell Fate Biology and Stem Cell Medicine, Tohoku University Graduate School of MedicineSendaiJapan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of MedicineTokyoJapan
- Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of MedicineKyotoJapan
| | - Masamichi Yamamoto
- Department of Research Promotion and Management, National Cerebral and Cardiovascular CenterOsakaJapan
| | - Daiki Karigane
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and MedicineTokyoJapan
- Division of Hematology, Department of Medicine, Keio University School of MedicineTokyoJapan
| | - Kohei Shiroshita
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and MedicineTokyoJapan
- Division of Hematology, Department of Medicine, Keio University School of MedicineTokyoJapan
| | - Yuriko Sorimachi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and MedicineTokyoJapan
- Department of Life Sciences and Medical BioScience, Waseda University School of Advanced Science and EngineeringTokyoJapan
| | - Shinya Fujita
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and MedicineTokyoJapan
- Division of Hematology, Department of Medicine, Keio University School of MedicineTokyoJapan
| | - Takayuki Morikawa
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and MedicineTokyoJapan
| | - Shuhei Koide
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, University of TokyoTokyoJapan
| | - Motohiko Oshima
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, University of TokyoTokyoJapan
| | - Akira Nishiyama
- Department of Immunology, Yokohama City University Graduate School of MedicineKanagawaJapan
| | - Koichi Murakami
- Department of Immunology, Yokohama City University Graduate School of MedicineKanagawaJapan
- Advanced Medical Research Center, Yokohama City UniversityKanagawaJapan
| | - Miho Haraguchi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and MedicineTokyoJapan
| | - Shinpei Tamaki
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and MedicineTokyoJapan
| | - Takehiro Yamamoto
- Department of Biochemistry, Keio University School of MedicineTokyoJapan
| | - Tomohiro Yabushita
- Division of Cellular Therapy, The Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yosuke Tanaka
- International Research Center for Medical Sciences, Kumamoto UniversityKumamotoJapan
| | - Go Nagamatsu
- Center for Advanced Assisted Reproductive Technologies, University of YamanashiYamanashiJapan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology AgencySaitamaJapan
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women's Medical UniversityTokyoJapan
| | - Shinichiro Okamoto
- Division of Hematology, Department of Medicine, Keio University School of MedicineTokyoJapan
| | - Nobuhito Goda
- Department of Life Sciences and Medical BioScience, Waseda University School of Advanced Science and EngineeringTokyoJapan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of MedicineKanagawaJapan
- Advanced Medical Research Center, Yokohama City UniversityKanagawaJapan
| | - Ayako Nakamura-Ishizu
- Department of Microscopic and Developmental Anatomy, Tokyo Women's Medical UniversityTokyoJapan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of MedicineTokyoJapan
- Live Imaging Center, Central Institute for Experimental AnimalsKanagawaJapan
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, University of TokyoTokyoJapan
| | - Toshio Suda
- International Research Center for Medical Sciences, Kumamoto UniversityKumamotoJapan
- Cancer Science Institute of Singapore, National University of SingaporeSingaporeSingapore
| | - Keiyo Takubo
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and MedicineTokyoJapan
- Department of Cell Fate Biology and Stem Cell Medicine, Tohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
32
|
Pinto SC, Stojilković B, Zhang X, Sablowski R. Plant cell size: Links to cell cycle, differentiation and ploidy. CURRENT OPINION IN PLANT BIOLOGY 2024; 78:102527. [PMID: 38484440 DOI: 10.1016/j.pbi.2024.102527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 04/07/2024]
Abstract
Cell size affects many processes, including exchange of nutrients and external signals, cell division and tissue mechanics. Across eukaryotes, cells have evolved mechanisms that assess their own size to inform processes such as cell cycle progression or gene expression. Here, we review recent progress in understanding plant cell size regulation and its implications, relating these findings to work in other eukaryotes. Highlights include use of DNA contents as reference point to control the cell cycle in shoot meristems, a size-dependent cell fate decision during stomatal development and insights into the interconnection between ploidy, cell size and cell wall mechanics.
Collapse
Affiliation(s)
- Sara C Pinto
- Cell and Developmental Biology, John Innes Centre, Norwich, UK
| | | | - Xinyu Zhang
- Cell and Developmental Biology, John Innes Centre, Norwich, UK
| | | |
Collapse
|
33
|
Bonora M, Morganti C, van Gastel N, Ito K, Calura E, Zanolla I, Ferroni L, Zhang Y, Jung Y, Sales G, Martini P, Nakamura T, Lasorsa FM, Finkel T, Lin CP, Zavan B, Pinton P, Georgakoudi I, Romualdi C, Scadden DT, Ito K. A mitochondrial NADPH-cholesterol axis regulates extracellular vesicle biogenesis to support hematopoietic stem cell fate. Cell Stem Cell 2024; 31:359-377.e10. [PMID: 38458178 PMCID: PMC10957094 DOI: 10.1016/j.stem.2024.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 11/16/2023] [Accepted: 02/08/2024] [Indexed: 03/10/2024]
Abstract
Mitochondrial fatty acid oxidation (FAO) is essential for hematopoietic stem cell (HSC) self-renewal; however, the mechanism by which mitochondrial metabolism controls HSC fate remains unknown. Here, we show that within the hematopoietic lineage, HSCs have the largest mitochondrial NADPH pools, which are required for proper HSC cell fate and homeostasis. Bioinformatic analysis of the HSC transcriptome, biochemical assays, and genetic inactivation of FAO all indicate that FAO-generated NADPH fuels cholesterol synthesis in HSCs. Interference with FAO disturbs the segregation of mitochondrial NADPH toward corresponding daughter cells upon single HSC division. Importantly, we have found that the FAO-NADPH-cholesterol axis drives extracellular vesicle (EV) biogenesis and release in HSCs, while inhibition of EV signaling impairs HSC self-renewal. These data reveal the existence of a mitochondrial NADPH-cholesterol axis for EV biogenesis that is required for hematopoietic homeostasis and highlight the non-stochastic nature of HSC fate determination.
Collapse
Affiliation(s)
- Massimo Bonora
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA
| | - Claudia Morganti
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA
| | - Nick van Gastel
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA; de Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | - Kyoko Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA
| | - Enrica Calura
- Department of Biology, University of Padova, 35121 Padua, Italy
| | - Ilaria Zanolla
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Letizia Ferroni
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy
| | - Yang Zhang
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Yookyung Jung
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA; Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Gabriele Sales
- Department of Biology, University of Padova, 35121 Padua, Italy
| | - Paolo Martini
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Takahisa Nakamura
- Divisions of Endocrinology and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Department of Metabolic Bioregulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Francesco Massimo Lasorsa
- Department of Biosciences Biotechnologies and Environment University of Bari and Institute of Biomembranes Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70125 Bari, Italy
| | - Toren Finkel
- Aging Institute and Department of Medicine, University of Pittsburgh School of Medicine/University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Barbara Zavan
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; Translational Medicine Department, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Chiara Romualdi
- Department of Biology, University of Padova, 35121 Padua, Italy
| | - David T Scadden
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA; Montefiore Einstein Comprehensive Cancer Center and Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
34
|
Foy R, Lew KX, Saurin AT. The search for CDK4/6 inhibitor biomarkers has been hampered by inappropriate proliferation assays. NPJ Breast Cancer 2024; 10:19. [PMID: 38438376 PMCID: PMC10912267 DOI: 10.1038/s41523-024-00624-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 02/16/2024] [Indexed: 03/06/2024] Open
Abstract
CDK4/6 inhibitors are effective at treating advanced HR+ /HER2- breast cancer, however biomarkers that can predict response are urgently needed. We demonstrate here that previous large-scale screens designed to identify which tumour types or genotypes are most sensitive to CDK4/6 inhibitors have misrepresented the responsive cell lines because of a reliance on metabolic proliferation assays. CDK4/6-inhibited cells arrest in G1 but continue to grow in size, thereby producing more mitochondria. We show that this growth obscures the arrest using ATP-based proliferation assays but not if DNA-based assays are used instead. Furthermore, lymphoma lines, previously identified as the most sensitive, simply appear to respond the best using ATP-based assays because they fail to overgrow during the G1 arrest. Similarly, the CDK4/6 inhibitor abemaciclib appears to inhibit proliferation better than palbociclib because it also restricts cellular overgrowth through off-target effects. DepMap analysis of screening data using reliable assay types, demonstrates that palbociclib-sensitive cell types are also sensitive to Cyclin D1, CDK4 and CDK6 knockout/knockdown, whereas the palbociclib-resistant lines are sensitive to Cyclin E1, CDK2 and SKP2 knockout/knockdown. Potential biomarkers of palbociclib-sensitive cells are increased expression of CCND1 and RB1, and reduced expression of CCNE1 and CDKN2A. Probing DepMap with similar data from metabolic assays fails to reveal these associations. Together, this demonstrates why CDK4/6 inhibitors, and any other anti-cancer drugs that arrest the cell cycle but permit continued cell growth, must now be re-screened against a wide-range of cell types using an appropriate proliferation assay. This would help to better inform clinical trials and to identify much needed biomarkers of response.
Collapse
Affiliation(s)
- Reece Foy
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Kah Xin Lew
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Adrian T Saurin
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
35
|
Mattila J, Viitanen A, Fabris G, Strutynska T, Korzelius J, Hietakangas V. Stem cell mTOR signaling directs region-specific cell fate decisions during intestinal nutrient adaptation. SCIENCE ADVANCES 2024; 10:eadi2671. [PMID: 38335286 PMCID: PMC10857434 DOI: 10.1126/sciadv.adi2671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024]
Abstract
The adult intestine is a regionalized organ, whose size and cellular composition are adjusted in response to nutrient status. This involves dynamic regulation of intestinal stem cell (ISC) proliferation and differentiation. How nutrient signaling controls cell fate decisions to drive regional changes in cell-type composition remains unclear. Here, we show that intestinal nutrient adaptation involves region-specific control of cell size, cell number, and differentiation. We uncovered that activation of mTOR complex 1 (mTORC1) increases ISC size in a region-specific manner. mTORC1 activity promotes Delta expression to direct cell fate toward the absorptive enteroblast lineage while inhibiting secretory enteroendocrine cell differentiation. In aged flies, the ISC mTORC1 signaling is deregulated, being constitutively high and unresponsive to diet, which can be mitigated through lifelong intermittent fasting. In conclusion, mTORC1 signaling contributes to the ISC fate decision, enabling regional control of intestinal cell differentiation in response to nutrition.
Collapse
Affiliation(s)
- Jaakko Mattila
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
| | - Arto Viitanen
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki 00790, Finland
| | - Gaia Fabris
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki 00790, Finland
| | - Tetiana Strutynska
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
| | - Jerome Korzelius
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
| | - Ville Hietakangas
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki 00790, Finland
| |
Collapse
|
36
|
Kot K, Kot Y, Kurbanov R, Andriiash H, Tigunova O, Blume Y, Shulga S. The effect of human PBMCs immobilization on their Аβ42 aggregates-dependent proinflammatory state on a cellular model of Alzheimer's disease. Front Neurosci 2024; 18:1325287. [PMID: 38406587 PMCID: PMC10884286 DOI: 10.3389/fnins.2024.1325287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
The leading pathological mechanisms of Alzheimer's disease are amyloidosis and inflammation. The presented work was aimed to study the effect of human peripheral blood mononuclear cells (hPBMcs) cells-matrix adhesion on their pro-inflammatory state in vitro. Although direct interaction of Аβ42 to PBMC is not a cellular model of Alzheimer's disease, PBMCs may serve as test cells to detect Аβ42-dependent molecular effects in monitoring disease progression. Peripheral blood mononuclear cells (PBMCs) are used to assess changes in cytokines released in response to diseases or Alzheimer's disease-specific cytotoxic molecules such as Aβ42. The effect of recombinant amyloid β-peptide rАβ42 on the concentration of endogenous amyloid β-peptide Aβ40 and pro-inflammatory cytokines TNFα and IL-1β in human peripheral blood mononuclear cells that were cultured in suspension and immobilized in alginate microcarriers for 24 h were investigated. The localization and accumulation of Aβ40 and rAβ42 peptides in cells, as well as quantitative determination of the concentration of Aβ40 peptide, TNFα and IL-1β cytokines, was performed by intravital fluorescence imaging. The results were qualitatively similar for both cell models. It was determined that the content of TNFα and Aβ40 in the absence of rAβ42 in the incubation medium did not change for 24 h after incubation, and the content of IL-1β was lower compared to the cells that were not incubated. Incubation of cells in vitro with exogenous rAβ42 led to an increase in the intracellular content of TNFα and Aβ40, and no accumulation of IL-1β in cells was observed. The accumulation of Aβ40 in the cytoplasm was accompanied by the aggregation of rAβ42 on the outer surface of the cell plasma membrane. It was shown that the basic levels of indicators and the intensity of the response of immobilized cells to an exogenous stimulus were significantly greater than those of cells in suspension. To explore whether non-neuronal cells effects in alginate microcarriers were cell-matrix adhesion mediated, we tested the effect of blocking β1 integrins on proamyloidogenic and proinflammation cellular state. Immobilization within alginate hydrogels after incubation with the β1 integrins blocking antibodies showed a remarkable inhibition of TNFα and Aβ40 accumulation in rAβ42-treated cells. It can be concluded that activation of signal transduction and synthesizing activity of a portion of mononuclear cells of human peripheral blood is possible (can significantly increase) in the presence of cell-matrix adhesion.
Collapse
Affiliation(s)
- Kateryna Kot
- Biochemistry Department, V. N. Karazin Kharkiv National University of Ministry of Education and Science of Ukraine, Kharkiv, Ukraine
| | - Yurii Kot
- Biochemistry Department, V. N. Karazin Kharkiv National University of Ministry of Education and Science of Ukraine, Kharkiv, Ukraine
| | - Rustam Kurbanov
- Biochemistry Department, V. N. Karazin Kharkiv National University of Ministry of Education and Science of Ukraine, Kharkiv, Ukraine
| | - Hanna Andriiash
- Department of Genomics and Molecular Biotechnology, Institute of Food Biotechnology and Genomics National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Olena Tigunova
- Department of Genomics and Molecular Biotechnology, Institute of Food Biotechnology and Genomics National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Yaroslav Blume
- Department of Genomics and Molecular Biotechnology, Institute of Food Biotechnology and Genomics National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Sergiy Shulga
- Department of Genomics and Molecular Biotechnology, Institute of Food Biotechnology and Genomics National Academy of Science of Ukraine, Kyiv, Ukraine
| |
Collapse
|
37
|
Diehl FF, Sapp KM, Vander Heiden MG. The bidirectional relationship between metabolism and cell cycle control. Trends Cell Biol 2024; 34:136-149. [PMID: 37385879 DOI: 10.1016/j.tcb.2023.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023]
Abstract
The relationship between metabolism and cell cycle progression is complex and bidirectional. Cells must rewire metabolism to meet changing biosynthetic demands across cell cycle phases. In turn, metabolism can influence cell cycle progression through direct regulation of cell cycle proteins, through nutrient-sensing signaling pathways, and through its impact on cell growth, which is linked to cell division. Furthermore, metabolism is a key player in mediating quiescence-proliferation transitions in physiologically important cell types, such as stem cells. How metabolism impacts cell cycle progression, exit, and re-entry, as well as how these processes impact metabolism, is not fully understood. Recent advances uncovering mechanistic links between cell cycle regulators and metabolic processes demonstrate a complex relationship between metabolism and cell cycle control, with many questions remaining.
Collapse
Affiliation(s)
- Frances F Diehl
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Koch Institute for Integrative Cancer Research and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kiera M Sapp
- Koch Institute for Integrative Cancer Research and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
38
|
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
|
39
|
Borsa M, Obba S, Richter FC, Zhang H, Riffelmacher T, Carrelha J, Alsaleh G, Jacobsen SEW, Simon AK. Autophagy preserves hematopoietic stem cells by restraining MTORC1-mediated cellular anabolism. Autophagy 2024; 20:45-57. [PMID: 37614038 PMCID: PMC10761185 DOI: 10.1080/15548627.2023.2247310] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
Adult stem cells are long-lived and quiescent with unique metabolic requirements. Macroautophagy/autophagy is a fundamental survival mechanism that allows cells to adapt to metabolic changes by degrading and recycling intracellular components. Here we address why autophagy depletion leads to a drastic loss of the stem cell compartment. Using inducible deletion of autophagy specifically in adult hematopoietic stem cells (HSCs) and in mice chimeric for autophagy-deficient and normal HSCs, we demonstrate that the stem cell loss is cell-intrinsic. Mechanistically, autophagy-deficient HSCs showed higher expression of several amino acid transporters (AAT) when compared to autophagy-competent cells, resulting in increased amino acid (AA) uptake. This was followed by sustained MTOR (mechanistic target of rapamycin) activation, with enlarged cell size, glucose uptake and translation, which is detrimental to the quiescent HSCs. MTOR inhibition by rapamycin treatment in vivo was able to rescue autophagy-deficient HSC loss and bone marrow failure and resulted in better reconstitution after transplantation. Our results suggest that targeting MTOR may improve aged stem cell function, promote reprogramming and stem cell transplantation.List of abbreviations: 5FU: fluoracil; AA: amino acids; AKT/PKB: thymoma viral proto-oncogene 1; ATF4: activating transcription factor 4; BafA: bafilomycin A1; BM: bone marrow; EIF2: eukaryotic initiation factor 2; EIF4EBP1/4EBP1: eukaryotic translation initiation factor 4E binding protein 1; KIT/CD117/c-Kit: KIT proto-oncogene receptor tyrosine kinase; HSCs: hematopoietic stem cells; HSPCs: hematopoietic stem and progenitor cells; Kyn: kynurenine; LSK: lineage- (Lin-), LY6A/Sca-1+, KIT/c-Kit/CD117+; LY6A/Sca-1: lymphocyte antigen 6 family member A; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; MTORC2: MTOR complex 2; OPP: O-propargyl-puromycin; PI3K: phosphoinositide 3-kinase; poly(I:C): polyinosinic:polycytidylic acid; RPS6/S6: ribosomal protein S6; tam: tamoxifen; TCA: tricarboxylic acid; TFEB: transcription factor EB; PTPRC/CD45: Protein Tyrosine Phosphatase Receptor Type C, CD45 antigen.
Collapse
Affiliation(s)
- Mariana Borsa
- Kennedy Institute of Rheumatology NDORMS, University of Oxford, Oxford, UK
| | - Sandrine Obba
- Kennedy Institute of Rheumatology NDORMS, University of Oxford, Oxford, UK
| | - Felix C. Richter
- Kennedy Institute of Rheumatology NDORMS, University of Oxford, Oxford, UK
| | - Hanlin Zhang
- Kennedy Institute of Rheumatology NDORMS, University of Oxford, Oxford, UK
| | | | - Joana Carrelha
- MRC Molecular Haematology Unit, MRC WIMM, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ghada Alsaleh
- Kennedy Institute of Rheumatology NDORMS, University of Oxford, Oxford, UK
| | - Sten Eirik W. Jacobsen
- MRC Molecular Haematology Unit, MRC WIMM, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- H7 Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anna Katharina Simon
- Kennedy Institute of Rheumatology NDORMS, University of Oxford, Oxford, UK
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
40
|
Sarachakov A, Varlamova A, Svekolkin V, Polyakova M, Valencia I, Unkenholz C, Pannellini T, Galkin I, Ovcharov P, Tabakov D, Postovalova E, Shin N, Sethi I, Bagaev A, Itkin T, Crane G, Kluk M, Geyer J, Inghirami G, Patel S. Spatial mapping of human hematopoiesis at single-cell resolution reveals aging-associated topographic remodeling. Blood 2023; 142:2282-2295. [PMID: 37774374 DOI: 10.1182/blood.2023021280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/01/2023] Open
Abstract
ABSTRACT The spatial anatomy of hematopoiesis in the bone marrow (BM) has been extensively studied in mice and other preclinical models, but technical challenges have precluded a commensurate exploration in humans. Institutional pathology archives contain thousands of paraffinized BM core biopsy tissue specimens, providing a rich resource for studying the intact human BM topography in a variety of physiologic states. Thus, we developed an end-to-end pipeline involving multiparameter whole tissue staining, in situ imaging at single-cell resolution, and artificial intelligence-based digital whole slide image analysis and then applied it to a cohort of disease-free samples to survey alterations in the hematopoietic topography associated with aging. Our data indicate heterogeneity in marrow adipose tissue (MAT) content within each age group and an inverse correlation between MAT content and proportions of early myeloid and erythroid precursors, irrespective of age. We identify consistent endosteal and perivascular positioning of hematopoietic stem and progenitor cells (HSPCs) with medullary localization of more differentiated elements and, importantly, uncover new evidence of aging-associated changes in cellular and vascular morphologies, microarchitectural alterations suggestive of foci with increased lymphocytes, and diminution of a potentially active megakaryocytic niche. Overall, our findings suggest that there is topographic remodeling of human hematopoiesis associated with aging. More generally, we demonstrate the potential to deeply unravel the spatial biology of normal and pathologic human BM states using intact archival tissue specimens.
Collapse
Affiliation(s)
| | | | | | | | - Itzel Valencia
- Multiparametric In Situ Imaging Laboratory, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Caitlin Unkenholz
- Multiparametric In Situ Imaging Laboratory, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Tania Pannellini
- Multiparametric In Situ Imaging Laboratory, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | | | | | | | | | | | | | | | - Tomer Itkin
- Division of Regenerative Medicine, Department of Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY
| | - Genevieve Crane
- Department of Laboratory Medicine, Cleveland Clinic, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland, OH
| | - Michael Kluk
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/NewYork-Presbyterian Hospital, New York, NY
| | - Julia Geyer
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/NewYork-Presbyterian Hospital, New York, NY
| | - Giorgio Inghirami
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/NewYork-Presbyterian Hospital, New York, NY
| | - Sanjay Patel
- Multiparametric In Situ Imaging Laboratory, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/NewYork-Presbyterian Hospital, New York, NY
| |
Collapse
|
41
|
Swaffer MP, Marinov GK, Zheng H, Fuentes Valenzuela L, Tsui CY, Jones AW, Greenwood J, Kundaje A, Greenleaf WJ, Reyes-Lamothe R, Skotheim JM. RNA polymerase II dynamics and mRNA stability feedback scale mRNA amounts with cell size. Cell 2023; 186:5254-5268.e26. [PMID: 37944513 DOI: 10.1016/j.cell.2023.10.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/16/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
A fundamental feature of cellular growth is that total protein and RNA amounts increase with cell size to keep concentrations approximately constant. A key component of this is that global transcription rates increase in larger cells. Here, we identify RNA polymerase II (RNAPII) as the limiting factor scaling mRNA transcription with cell size in budding yeast, as transcription is highly sensitive to the dosage of RNAPII but not to other components of the transcriptional machinery. Our experiments support a dynamic equilibrium model where global RNAPII transcription at a given size is set by the mass action recruitment kinetics of unengaged nucleoplasmic RNAPII to the genome. However, this only drives a sub-linear increase in transcription with size, which is then partially compensated for by a decrease in mRNA decay rates as cells enlarge. Thus, limiting RNAPII and feedback on mRNA stability work in concert to scale mRNA amounts with cell size.
Collapse
Affiliation(s)
| | - Georgi K Marinov
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Huan Zheng
- Department of Biology, McGill University, Montreal, QC H3G 0B1, Canada
| | | | - Crystal Yee Tsui
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | | | | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | | | | | - Jan M Skotheim
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
42
|
Wilson GA, Vuina K, Sava G, Huard C, Meneguello L, Coulombe-Huntington J, Bertomeu T, Maizels RJ, Lauring J, Kriston-Vizi J, Tyers M, Ali S, Bertoli C, de Bruin RAM. Active growth signaling promotes senescence and cancer cell sensitivity to CDK7 inhibition. Mol Cell 2023; 83:4078-4092.e6. [PMID: 37977119 DOI: 10.1016/j.molcel.2023.10.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 07/25/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023]
Abstract
Tumor growth is driven by continued cellular growth and proliferation. Cyclin-dependent kinase 7's (CDK7) role in activating mitotic CDKs and global gene expression makes it therefore an attractive target for cancer therapies. However, what makes cancer cells particularly sensitive to CDK7 inhibition (CDK7i) remains unclear. Here, we address this question. We show that CDK7i, by samuraciclib, induces a permanent cell-cycle exit, known as senescence, without promoting DNA damage signaling or cell death. A chemogenetic genome-wide CRISPR knockout screen identified that active mTOR (mammalian target of rapamycin) signaling promotes samuraciclib-induced senescence. mTOR inhibition decreases samuraciclib sensitivity, and increased mTOR-dependent growth signaling correlates with sensitivity in cancer cell lines. Reverting a growth-promoting mutation in PIK3CA to wild type decreases sensitivity to CDK7i. Our work establishes that enhanced growth alone promotes CDK7i sensitivity, providing an explanation for why some cancers are more sensitive to CDK inhibition than normally growing cells.
Collapse
Affiliation(s)
- Gemma A Wilson
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Karla Vuina
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Georgina Sava
- Division of Cancer, Department of Surgery & Cancer, Imperial College London, London, UK
| | - Caroline Huard
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Leticia Meneguello
- Laboratory for Molecular Cell Biology, University College London, London, UK; UCL Cancer Institute, University College London, London, UK
| | - Jasmin Coulombe-Huntington
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada; Department of Bioengineering, McGill University, Montréal, QC, Canada
| | - Thierry Bertomeu
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Rory J Maizels
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Josh Lauring
- Janssen Research and Development, the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Janos Kriston-Vizi
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada; Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Simak Ali
- Division of Cancer, Department of Surgery & Cancer, Imperial College London, London, UK
| | - Cosetta Bertoli
- Laboratory for Molecular Cell Biology, University College London, London, UK.
| | - Robertus A M de Bruin
- Laboratory for Molecular Cell Biology, University College London, London, UK; UCL Cancer Institute, University College London, London, UK.
| |
Collapse
|
43
|
Manohar S, Estrada ME, Uliana F, Vuina K, Alvarez PM, de Bruin RAM, Neurohr GE. Genome homeostasis defects drive enlarged cells into senescence. Mol Cell 2023; 83:4032-4046.e6. [PMID: 37977116 PMCID: PMC10659931 DOI: 10.1016/j.molcel.2023.10.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 06/30/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023]
Abstract
Cellular senescence refers to an irreversible state of cell-cycle arrest and plays important roles in aging and cancer biology. Because senescence is associated with increased cell size, we used reversible cell-cycle arrests combined with growth rate modulation to study how excessive growth affects proliferation. We find that enlarged cells upregulate p21, which limits cell-cycle progression. Cells that re-enter the cell cycle encounter replication stress that is well tolerated in physiologically sized cells but causes severe DNA damage in enlarged cells, ultimately resulting in mitotic failure and permanent cell-cycle withdrawal. We demonstrate that enlarged cells fail to recruit 53BP1 and other non-homologous end joining (NHEJ) machinery to DNA damage sites and fail to robustly initiate DNA damage-dependent p53 signaling, rendering them highly sensitive to genotoxic stress. We propose that an impaired DNA damage response primes enlarged cells for persistent replication-acquired damage, ultimately leading to cell division failure and permanent cell-cycle exit.
Collapse
Affiliation(s)
- Sandhya Manohar
- Institute for Biochemistry, Department of Biology, ETH Zürich 8093, Zürich, Zürich, Switzerland
| | - Marianna E Estrada
- Institute for Biochemistry, Department of Biology, ETH Zürich 8093, Zürich, Zürich, Switzerland
| | - Federico Uliana
- Institute for Biochemistry, Department of Biology, ETH Zürich 8093, Zürich, Zürich, Switzerland
| | - Karla Vuina
- Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Patricia Moyano Alvarez
- Institute for Biochemistry, Department of Biology, ETH Zürich 8093, Zürich, Zürich, Switzerland
| | - Robertus A M de Bruin
- Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; UCL Cancer Institute, University College London, London WC1E 6BT, UK
| | - Gabriel E Neurohr
- Institute for Biochemistry, Department of Biology, ETH Zürich 8093, Zürich, Zürich, Switzerland.
| |
Collapse
|
44
|
Khurana A, Chadha Y, Schmoller KM. Too big not to fail: Different paths lead to senescence of enlarged cells. Mol Cell 2023; 83:3946-3947. [PMID: 37977113 DOI: 10.1016/j.molcel.2023.10.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
In this issue of Molecular Cell, Crozier et al.,1 Foy et al.,2 Manohar et al.,3 and Wilson et al.4 show how excessive cell growth caused by a temporary G1 arrest leads to permanent cell cycle exit at different stages of the cell cycle.
Collapse
Affiliation(s)
- Arohi Khurana
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Yagya Chadha
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Kurt M Schmoller
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
45
|
Foy R, Crozier L, Pareri AU, Valverde JM, Park BH, Ly T, Saurin AT. Oncogenic signals prime cancer cells for toxic cell overgrowth during a G1 cell cycle arrest. Mol Cell 2023; 83:4047-4061.e6. [PMID: 37977117 DOI: 10.1016/j.molcel.2023.10.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 07/10/2023] [Accepted: 10/17/2023] [Indexed: 11/19/2023]
Abstract
CDK4/6 inhibitors are remarkable anti-cancer drugs that can arrest tumor cells in G1 and induce their senescence while causing only relatively mild toxicities in healthy tissues. How they achieve this mechanistically is unclear. We show here that tumor cells are specifically vulnerable to CDK4/6 inhibition because during the G1 arrest, oncogenic signals drive toxic cell overgrowth. This overgrowth causes permanent cell cycle withdrawal by either preventing progression from G1 or inducing genotoxic damage during the subsequent S-phase and mitosis. Inhibiting or reverting oncogenic signals that converge onto mTOR can rescue this excessive growth, DNA damage, and cell cycle exit in cancer cells. Conversely, inducing oncogenic signals in non-transformed cells can drive these toxic phenotypes and sensitize the cells to CDK4/6 inhibition. Together, this demonstrates that cell cycle arrest and oncogenic cell growth is a synthetic lethal combination that is exploited by CDK4/6 inhibitors to induce tumor-specific toxicity.
Collapse
Affiliation(s)
- Reece Foy
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Lisa Crozier
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Aanchal U Pareri
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Juan Manuel Valverde
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Ben Ho Park
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Tony Ly
- Molecular Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Adrian T Saurin
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee DD1 9SY, UK.
| |
Collapse
|
46
|
Crozier L, Foy R, Adib R, Kar A, Holt JA, Pareri AU, Valverde JM, Rivera R, Weston WA, Wilson R, Regnault C, Whitfield P, Badonyi M, Bennett LG, Vernon EG, Gamble A, Marsh JA, Staples CJ, Saurin AT, Barr AR, Ly T. CDK4/6 inhibitor-mediated cell overgrowth triggers osmotic and replication stress to promote senescence. Mol Cell 2023; 83:4062-4077.e5. [PMID: 37977118 DOI: 10.1016/j.molcel.2023.10.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 07/10/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023]
Abstract
Abnormal increases in cell size are associated with senescence and cell cycle exit. The mechanisms by which overgrowth primes cells to withdraw from the cell cycle remain unknown. We address this question using CDK4/6 inhibitors, which arrest cells in G0/G1 and are licensed to treat advanced HR+/HER2- breast cancer. We demonstrate that CDK4/6-inhibited cells overgrow during G0/G1, causing p38/p53/p21-dependent cell cycle withdrawal. Cell cycle withdrawal is triggered by biphasic p21 induction. The first p21 wave is caused by osmotic stress, leading to p38- and size-dependent accumulation of p21. CDK4/6 inhibitor washout results in some cells entering S-phase. Overgrown cells experience replication stress, resulting in a second p21 wave that promotes cell cycle withdrawal from G2 or the subsequent G1. We propose that the levels of p21 integrate signals from overgrowth-triggered stresses to determine cell fate. This model explains how hypertrophy can drive senescence and why CDK4/6 inhibitors have long-lasting effects in patients.
Collapse
Affiliation(s)
- Lisa Crozier
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, UK
| | - Reece Foy
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, UK
| | - Rozita Adib
- MRC Laboratory of Medical Sciences, London, UK
| | - Ananya Kar
- Molecular Cell and Developmental Biology, School of Life Sciences, Dundee, UK
| | | | - Aanchal U Pareri
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, UK
| | - Juan M Valverde
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, UK
| | - Rene Rivera
- Molecular Cell and Developmental Biology, School of Life Sciences, Dundee, UK
| | | | - Rona Wilson
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Clement Regnault
- Glasgow Polyomics College of Medical, Veterinary, and Life Sciences, University of Glasgow, UK
| | - Phil Whitfield
- Glasgow Polyomics College of Medical, Veterinary, and Life Sciences, University of Glasgow, UK
| | - Mihaly Badonyi
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK
| | - Laura G Bennett
- North West Cancer Research Institute, School of Medical and Health Sciences, Brambell Building, Deiniol Rd, Bangor LL57 2UW, UK
| | - Ellen G Vernon
- North West Cancer Research Institute, School of Medical and Health Sciences, Brambell Building, Deiniol Rd, Bangor LL57 2UW, UK
| | - Amelia Gamble
- North West Cancer Research Institute, School of Medical and Health Sciences, Brambell Building, Deiniol Rd, Bangor LL57 2UW, UK
| | - Joseph A Marsh
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK
| | - Christopher J Staples
- North West Cancer Research Institute, School of Medical and Health Sciences, Brambell Building, Deiniol Rd, Bangor LL57 2UW, UK
| | - Adrian T Saurin
- Cellular and Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Dundee, UK.
| | - Alexis R Barr
- MRC Laboratory of Medical Sciences, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Tony Ly
- Molecular Cell and Developmental Biology, School of Life Sciences, Dundee, UK; Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, UK; Glasgow Polyomics College of Medical, Veterinary, and Life Sciences, University of Glasgow, UK.
| |
Collapse
|
47
|
Mouton SN, Boersma AJ, Veenhoff LM. A physicochemical perspective on cellular ageing. Trends Biochem Sci 2023; 48:949-962. [PMID: 37716870 DOI: 10.1016/j.tibs.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/18/2023]
Abstract
Cellular ageing described at the molecular level is a multifactorial process that leads to a spectrum of ageing trajectories. There has been recent discussion about whether a decline in physicochemical homeostasis causes aberrant phase transitions, which are a driver of ageing. Indeed, the function of all biological macromolecules, regardless of their participation in biomolecular condensates, depends on parameters such as pH, crowding, and redox state. We expand on the physicochemical homeostasis hypothesis and summarise recent evidence that the intracellular milieu influences molecular processes involved in ageing.
Collapse
Affiliation(s)
- Sara N Mouton
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Arnold J Boersma
- Cellular Protein Chemistry, Bijvoet Centre for Biomolecular Research, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Liesbeth M Veenhoff
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
48
|
Lanz MC, Zhang S, Swaffer MP, Hernández Götz L, McCarty F, Ziv I, Jarosz DF, Elias JE, Skotheim JM. Genome dilution by cell growth drives starvation-like proteome remodeling in mammalian and yeast cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562558. [PMID: 37905015 PMCID: PMC10614910 DOI: 10.1101/2023.10.16.562558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cell size is tightly controlled in healthy tissues and single-celled organisms, but it remains unclear how size influences cell physiology. Increasing cell size was recently shown to remodel the proteomes of cultured human cells, demonstrating that large and small cells of the same type can be biochemically different. Here, we corroborate these results in mouse hepatocytes and extend our analysis using yeast. We find that size-dependent proteome changes are highly conserved and mostly independent of metabolic state. As eukaryotic cells grow larger, the dilution of the genome elicits a starvation-like proteome phenotype, suggesting that growth in large cells is limited by the genome in a manner analogous to a limiting nutrient. We also demonstrate that the proteomes of replicatively-aged yeast are primarily determined by their large size. Overall, our data suggest that genome concentration is a universal determinant of proteome content in growing cells.
Collapse
|
49
|
Yang Y, Yuan K, Liu Y, Wang Q, Lin Y, Yang S, Huang K, Kan T, Zhang Y, Xu M, Yu Z, Fan Q, Wang Y, Li H, Tang T. Constitutively activated AMPKα1 protects against skeletal aging in mice by promoting bone-derived IGF-1 secretion. Cell Prolif 2023; 56:e13476. [PMID: 37042047 PMCID: PMC10542616 DOI: 10.1111/cpr.13476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/13/2023] Open
Abstract
Senile osteoporosis is characterized by age-related bone loss and bone microarchitecture deterioration. However, little is known to date about the mechanism that maintains bone homeostasis during aging. In this study, we identify adenosine monophosphate-activated protein kinase alpha 1 (AMPKα1) as a critical factor regulating the senescence and lineage commitment of mesenchymal stem cells (MSCs). A phospho-mutant mouse model shows that constitutive AMPKα1 activation prevents age-related bone loss and promoted MSC osteogenic commitment with increased bone-derived insulin-like growth factor 1 (IGF-1) secretion. Mechanistically, upregulation of IGF-1 signalling by AMPKα1 depends on cAMP-response element binding protein (CREB)-mediated transcriptional regulation. Furthermore, the essential role of the AMPKα1/IGF-1/CREB axis in promoting aged MSC osteogenic potential is confirmed using three-dimensional (3D) culture systems. Taken together, these results can provide mechanistic insight into the protective effect of AMPKα1 against skeletal aging by promoting bone-derived IGF-1 secretion.
Collapse
Affiliation(s)
- Yiqi Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Kai Yuan
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qishan Wang
- School of PharmacyShanghai Jiao Tong UniversityShanghaiChina
| | - Yixuan Lin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shengbing Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Kai Huang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tianyou Kan
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuxin Zhang
- Department of Rehabilitation Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Mingming Xu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qiming Fan
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yugang Wang
- Department of Trauma Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Hanjun Li
- Clinical Stem Cell Research Center, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
50
|
Agarwal P, Cadart C, Fort L, Gahan J, Greenspan L, Juan T, Kameneva P, Miao Y. Pathway to Independence: the future of developmental biology. Development 2023; 150:dev202360. [PMID: 37812057 PMCID: PMC10705336 DOI: 10.1242/dev.202360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
In 2022, Development launched its Pathway to Independence (PI) Programme, aimed at supporting postdocs as they transition to their first independent position. We selected eight talented researchers as the first cohort of PI Fellows. In this article, each of our Fellows provides their perspective on the future of their field. Together, they paint an exciting picture of the current state of and open questions in developmental biology.
Collapse
Affiliation(s)
- Priti Agarwal
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Clotilde Cadart
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - Loic Fort
- Vanderbilt University School of Medicine, 465 21st Avenue South, U 3200 MRB III, Nashville, TN 37240-7935, USA
| | - James Gahan
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Leah Greenspan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Thomas Juan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - Polina Kameneva
- The Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Yuchuan Miao
- Department of Genetics, Harvard Medical School and Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|