1
|
Ficarro SB, Kothiwal D, Bae HJ, Tavares I, Giordano G, Buratowski S, Marto JA. Leveraging HILIC/ERLIC separations for online nanoscale LC-MS/MS analysis of phosphopeptide isoforms from RNA polymerase II C-terminal domain. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1257:124560. [PMID: 40158465 DOI: 10.1016/j.jchromb.2025.124560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/06/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025]
Abstract
The eukaryotic RNA polymerase II (Pol II) multi-protein complex transcribes mRNA and coordinates several steps of co-transcriptional mRNA processing and chromatin modification. The largest Pol II subunit, Rpb1, has a C-terminal domain (CTD) comprising dozens of repeated heptad sequences (Tyr1-Ser2-Pro3-Thr4-Ser5-Pro6-Ser7), each containing five phospho-accepting amino acids. The CTD heptads are dynamically phosphorylated, creating specific patterns correlated with steps of transcription initiation, elongation, and termination. This CTD phosphorylation 'code' choreographs dynamic recruitment of important co-regulatory proteins during gene transcription. Genetic tools were used to engineer protease cleavage sites across the CTD (msCTD), creating tryptic peptides with unique sequences amenable to mass spectrometry analysis. However, phosphorylation isoforms within each msCTD sequence are difficult to resolve by standard reversed phase chromatography typically used for LC-MS/MS applications. Here, we use a panel of synthetic CTD phosphopeptides to explore the potential of hydrophilic interaction and electrostatic repulsion hydrophilic interaction (HILIC and ERLIC) chromatography as alternatives to reversed phase separation for CTD phosphopeptide analysis. Our results demonstrate that ERLIC provides improved performance for separation of singly- and doubly-phosphorylated CTD peptides for sequence analysis by LC-MS/MS. Analysis of native yeast msCTD confirms that phosphorylation on Ser5 and Ser2 represents the major endogenous phosphoisoforms. We expect this methodology will be especially useful in the investigation of pathways where multiple protein phosphorylation events converge in close proximity.
Collapse
Affiliation(s)
- Scott B Ficarro
- Department of Cancer Biology and Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Deepash Kothiwal
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Hyun Jin Bae
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Isidoro Tavares
- Department of Cancer Biology and Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Gabriela Giordano
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen Buratowski
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| | - Jarrod A Marto
- Department of Cancer Biology and Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
2
|
Tamura T, Umekawa E, Mori M, Otsuki M, Shibagaki Y, Hattori S, Sugawara T, Saito K, Ohta Y. Septin2 regulates ARHGAP25-mediated suppression of lamellipodia formation and cell spreading. FEBS Lett 2025. [PMID: 40205813 DOI: 10.1002/1873-3468.70041] [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: 12/05/2024] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025]
Abstract
Rho family small GTPases are key regulators of the actin cytoskeletal organization that controls cell morphology, but the regulatory mechanism of Rho small GTPase activity is not fully understood. Here we identified septin2, a component of the septin cytoskeleton, as an interacting protein of ARHGAP25, a GTPase-activating protein for Rho small GTPase Rac, in mammalian cells. ARHGAP25 colocalized with septin2 at lamellipodia, which are actin filament-rich protrusions. Overexpression of ARHGAP25 suppressed Rac-dependent lamellipodia formation and cell spreading, and ARHGAP25-mediated suppression was restored by depletion of septin2. Forced expression of septin2 enhanced ARHGAP25-mediated suppression of cell spreading, and septin2-enhanced suppression was restored by the depletion of ARHGAP25. These results suggest that septin2 controls cell morphology by regulating the function of ARHGAP25.
Collapse
Affiliation(s)
- Tomoe Tamura
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| | - Emi Umekawa
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| | - Mamiko Mori
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| | - Mayuko Otsuki
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| | - Yoshio Shibagaki
- Division of Biochemistry, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | - Seisuke Hattori
- Division of Biochemistry, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | - Takeyuki Sugawara
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Koji Saito
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| | - Yasutaka Ohta
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| |
Collapse
|
3
|
Orcutt-Jahns B, Lima Junior JR, Lin E, Rockne RC, Matache A, Branciamore S, Hung E, Rodin AS, Lee PP, Meyer AS. Systems profiling reveals recurrently dysregulated cytokine signaling responses in ER+ breast cancer patients' blood. NPJ Syst Biol Appl 2024; 10:118. [PMID: 39389979 PMCID: PMC11467214 DOI: 10.1038/s41540-024-00447-0] [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: 04/17/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Cytokines operate in concert to maintain immune homeostasis and coordinate immune responses. In cases of ER+ breast cancer, peripheral immune cells exhibit altered responses to several cytokines, and these alterations are correlated strongly with patient outcomes. To develop a systems-level understanding of this dysregulation, we measured a panel of cytokine responses and receptor abundances in the peripheral blood of healthy controls and ER+ breast cancer patients across immune cell types. Using tensor factorization to model this multidimensional data, we found that breast cancer patients exhibited widespread alterations in response, including drastically reduced response to IL-10 and heightened basal levels of pSmad2/3 and pSTAT4. ER+ patients also featured upregulation of PD-L1, IL6Rα, and IL2Rα, among other receptors. Despite this, alterations in response to cytokines were not explained by changes in receptor abundances. Thus, tensor factorization helped to reveal a coordinated reprogramming of the immune system that was consistent across our cohort.
Collapse
Affiliation(s)
- Brian Orcutt-Jahns
- Department of Bioengineering, University of California, Los Angeles (UCLA), CA, USA
| | | | - Emily Lin
- Department of Bioengineering, University of California, Los Angeles (UCLA), CA, USA
| | - Russell C Rockne
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Adina Matache
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Sergio Branciamore
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Ethan Hung
- Department of Bioengineering, University of California, Los Angeles (UCLA), CA, USA
| | - Andrei S Rodin
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Aaron S Meyer
- Department of Bioengineering, University of California, Los Angeles (UCLA), CA, USA.
- Jonsson Comprehensive Cancer Center, Los Angeles (UCLA), CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles (UCLA), CA, USA.
| |
Collapse
|
4
|
Ficarro SB, Kothiwal D, Bae HJ, Tavares I, Giordano G, Buratowski S, Marto JA. Leveraging HILIC/ERLIC Separations for Online Nanoscale LC-MS/MS Analysis of Phosphopeptide Isoforms from RNA Polymerase II C-terminal Domain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617299. [PMID: 39416017 PMCID: PMC11482835 DOI: 10.1101/2024.10.08.617299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The eukaryotic RNA polymerase II (Pol II) multi-protein complex transcribes mRNA and coordinates several steps of co-transcriptional mRNA processing and chromatin modification. The largest Pol II subunit, Rpb1, has a C-terminal domain (CTD) comprising dozens of repeated heptad sequences (Tyr1-Ser2-Pro3-Thr4-Ser5-Pro6-Ser7), each containing five phospho-accepting amino acids. The CTD heptads are dynamically phosphorylated, creating specific patterns correlated with steps of transcription initiation, elongation, and termination. This CTD phosphorylation 'code' choreographs dynamic recruitment of important co-regulatory proteins during gene transcription. Genetic tools were used to engineer protease cleavage sites across the CTD (msCTD), creating tryptic peptides with unique sequences amenable to mass spectrometry analysis. However, phosphorylation isoforms within each msCTD sequence are difficult to resolve by standard reversed phase chromatography typically used for LC-MS/MS applications. Here, we use a panel of synthetic CTD phosphopeptides to explore the potential of hydrophilic interaction and electrostatic repulsion hydrophilic interaction (HILIC and ERLIC) chromatography as alternatives to reversed phase separation for CTD phosphopeptide analysis. Our results demonstrate that ERLIC provides improved performance for separation of singly- and doubly-phosphorylated CTD peptides for sequence analysis by LC-MS/MS. Analysis of native yeast msCTD confirms that phosphorylation on Ser5 and Ser2 represents the major endogenous phosphoisoforms. We expect this methodology will be especially useful in the investigation of pathways where multiple protein phosphorylation events converge in close proximity.
Collapse
|
5
|
Sasvári P, Pettkó-Szandtner A, Wisniewski É, Csépányi-Kömi R. Neutrophil-specific interactome of ARHGAP25 reveals novel partners and regulatory insights. Sci Rep 2024; 14:20106. [PMID: 39210013 PMCID: PMC11362597 DOI: 10.1038/s41598-024-71002-4] [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: 04/19/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
ARHGAP25, a crucial molecule in immunological processes, serves as a Rac-specific GTPase-activating protein. Its role in cell migration and phagocyte functions, affecting the outcome of complex immunological diseases such as rheumatoid arthritis, renders it a promising target for drug research. Despite its importance, our knowledge of its intracellular interactions is still limited. This study employed proteomic analysis of glutathione S-transferase (GST)-tag pulldowns and co-immunoprecipitation from neutrophilic granulocyte cell lysate, revealing 76 candidates for potential physical interactions that complement ARHGAP25's known profile. Notably, four small GTPases (RAC2, RHOG, ARF4, and RAB27A) exhibited high affinity for ARHGAP25. The ARHGAP25-RAC2 and ARHGAP25-RHOG interactions appeared to be affected by the activation state of the small GTPases, suggesting a GTP-GDP cycle-dependent interaction. In silico dimer prediction pinpointed ARHGAP25's GAP domain as a credible binding interface, suggesting its suitability for GTP hydrolysis. Additionally, a list of Fc receptor-related kinases, phosphatases, and three of the 14-3-3 members were identified as potential partners, with in silico predictions highlighting eight binding sites, presenting novel insight on a potential regulatory mechanism for ARHGAP25.
Collapse
Affiliation(s)
- Péter Sasvári
- Department of Physiology, Semmelweis University, Tűzoltó u. 37-47., Budapest, 1085, Hungary
| | - Aladár Pettkó-Szandtner
- Proteomics Research Group, Core Facility, HUN-REN Biological Research Centre, Szeged, 6726, Hungary
| | - Éva Wisniewski
- Department of Physiology, Semmelweis University, Tűzoltó u. 37-47., Budapest, 1085, Hungary
| | - Roland Csépányi-Kömi
- Department of Physiology, Semmelweis University, Tűzoltó u. 37-47., Budapest, 1085, Hungary.
| |
Collapse
|
6
|
Orcutt-Jahns B, Junior JRL, Rockne RC, Matache A, Branciamore S, Hung E, Rodin AS, Lee PP, Meyer AS. Systems profiling reveals recurrently dysregulated cytokine signaling responses in ER+ breast cancer patients' blood. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.31.564987. [PMID: 37961682 PMCID: PMC10635026 DOI: 10.1101/2023.10.31.564987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Cytokines mediate cell-to-cell communication across the immune system and therefore are critical to immunosurveillance in cancer and other diseases. Several cytokines show dysregulated abundance or signaling responses in breast cancer, associated with the disease and differences in survival and progression. Cytokines operate in a coordinated manner to affect immune surveillance and regulate one another, necessitating a systems approach for a complete picture of this dysregulation. Here, we profiled cytokine signaling responses of peripheral immune cells from breast cancer patients as compared to healthy controls in a multidimensional manner across ligands, cell populations, and responsive pathways. We find alterations in cytokine responsiveness across pathways and cell types that are best defined by integrated signatures across dimensions. Alterations in the abundance of a cytokine's cognate receptor do not explain differences in responsiveness. Rather, alterations in baseline signaling and receptor abundance suggesting immune cell reprogramming are associated with altered responses. These integrated features suggest a global reprogramming of immune cell communication in breast cancer.
Collapse
Affiliation(s)
- Brian Orcutt-Jahns
- Department of Bioengineering, University of California, Los Angeles (UCLA), USA
| | | | - Russell C. Rockne
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Adina Matache
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Sergio Branciamore
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Ethan Hung
- Department of Bioengineering, University of California, Los Angeles (UCLA), USA
| | - Andrei S. Rodin
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Peter P. Lee
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Aaron S. Meyer
- Department of Bioengineering, University of California, Los Angeles (UCLA), USA
- Jonsson Comprehensive Cancer Center, UCLA, United States of America
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, USA
| |
Collapse
|
7
|
Czárán D, Sasvári P, Horváth ÁI, Ella K, Sűdy ÁR, Borbély É, Rusznák K, Czéh B, Mócsai A, Helyes Z, Csépányi-Kömi R. Lacking ARHGAP25 mitigates the symptoms of autoantibody-induced arthritis in mice. Front Immunol 2023; 14:1182278. [PMID: 37234175 PMCID: PMC10208528 DOI: 10.3389/fimmu.2023.1182278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Objective Despite intensive research on rheumatoid arthritis, the pathomechanism of the disease is still not fully understood and the treatment has not been completely resolved. Previously we demonstrated that the GTPase-activating protein, ARHGAP25 has a crucial role in the regulation of basic phagocyte functions. Here we investigate the role of ARHGAP25 in the complex inflammatory process of autoantibody-induced arthritis. Methods Wild-type and ARHGAP25 deficient (KO) mice on a C57BL/6 background, as well as bone marrow chimeric mice, were treated i.p. with the K/BxN arthritogenic or control serum, and the severity of inflammation and pain-related behavior was measured. Histology was prepared, leukocyte infiltration, cytokine production, myeloperoxidase activity, and superoxide production were determined, and comprehensive western blot analysis was conducted. Results In the absence of ARHGAP25, the severity of inflammation, joint destruction, and mechanical hyperalgesia significantly decreased, similarly to phagocyte infiltration, IL-1β, and MIP-2 levels in the tibiotarsal joint, whereas superoxide production or myeloperoxidase activity was unchanged. We observed a significantly mitigated phenotype in KO bone marrow chimeras as well. In addition, fibroblast-like synoviocytes showed comparable expression of ARHGAP25 to neutrophils. Significantly reduced ERK1/2, MAPK, and I-κB protein signals were detected in the arthritic KO mouse ankles. Conclusion Our findings suggest that ARHGAP25 has a key role in the pathomechanism of autoantibody-induced arthritis in which it regulates inflammation via the I-κB/NF-κB/IL-1β axis with the involvement of both immune cells and fibroblast-like synoviocytes.
Collapse
Affiliation(s)
- Domonkos Czárán
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Péter Sasvári
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Ádám István Horváth
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| | - Krisztina Ella
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Ágnes Réka Sűdy
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Éva Borbély
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| | - Kitti Rusznák
- Histology and Light Microscopy Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Boldizsár Czéh
- Histology and Light Microscopy Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Attila Mócsai
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Chronic Pain Research Group, Eötvös Loránd Research Network, University of Pécs, Pécs, Hungary
- PharmInVivo Ltd., Pécs, Hungary
| | | |
Collapse
|
8
|
Wisniewski É, Czárán D, Kovács F, Bahurek E, Németh A, Sasvári P, Szanda G, Pettkó-Szandtner A, Klement E, Ligeti E, Csépányi-Kömi R. A novel BRET-Based GAP assay reveals phosphorylation-dependent regulation of the RAC-specific GTPase activating protein ARHGAP25. FASEB J 2022; 36:e22584. [PMID: 36190314 DOI: 10.1096/fj.202200689r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/07/2022] [Accepted: 09/20/2022] [Indexed: 11/11/2022]
Abstract
ARHGAP25, a RAC-specific GTPase activating protein (GAP), is an essential regulator of phagocyte effector functions such as phagocytosis, superoxide production, and transendothelial migration. Furthermore, its complex role in tumor behavior has recently been recognized. We previously demonstrated that phosphorylation of serine 363 in ARHGAP25 regulates hematopoietic stem cells and progenitor cells in mouse bone marrow. However, the significance of other potential phosphorylation sites of ARHGAP25 remained unknown. Now, we developed a novel, real-time bioluminescence resonance energy transfer (BRET) assay to monitor the GAP activity of ARHGAP25 in vitro. Using this approach, we revealed that phosphorylation of S363 and S488, but not that of S379-380, controls ARHGAP25's RACGAP activity. On the other hand, we found in granulocyte-differentiated human PLB-985 cells that superoxide production and actin depolymerization are regulated by residues S363 and S379-380. The present data demonstrate the value of our BRET-GAP assay and show that different phosphorylation patterns regulate ARHGAP25's GAP activity and its effect on superoxide production and phagocytosis.
Collapse
Affiliation(s)
- Éva Wisniewski
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Domonkos Czárán
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Fanni Kovács
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Enikő Bahurek
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Afrodité Németh
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Péter Sasvári
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Gergő Szanda
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | | - Eva Klement
- Laboratory of Proteomics Research, Biological Research Centre, Szeged, Hungary.,Single Cell Omics ACF, Hungarian Centre of Excellence for Molecular Medicine, Szeged, Hungary
| | - Erzsébet Ligeti
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | |
Collapse
|
9
|
Mitochondrial oxidative phosphorylation is dispensable for survival of CD34+ chronic myeloid leukemia stem and progenitor cells. Cell Death Dis 2022; 13:384. [PMID: 35444236 PMCID: PMC9021200 DOI: 10.1038/s41419-022-04842-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 12/24/2022]
Abstract
AbstractChronic myeloid leukemia (CML) are initiated and sustained by self-renewing malignant CD34+ stem cells. Extensive efforts have been made to reveal the metabolic signature of the leukemia stem/progenitor cells in genomic, transcriptomic, and metabolomic studies. However, very little proteomic investigation has been conducted and the mechanism regarding at what level the metabolic program was rewired remains poorly understood. Here, using label-free quantitative proteomic profiling, we compared the signature of CD34+ stem/progenitor cells collected from CML individuals with that of healthy donors and observed significant changes in the abundance of enzymes associated with aerobic central carbonate metabolic pathways. Specifically, CML stem/progenitor cells expressed increased tricarboxylic acid cycle (TCA) with decreased glycolytic proteins, accompanying by increased oxidative phosphorylation (OXPHOS) and decreased glycolysis activity. Administration of the well-known OXPHOS inhibitor metformin eradicated CML stem/progenitor cells and re-sensitized CD34+ CML cells to imatinib in vitro and in patient-derived tumor xenograft murine model. However, different from normal CD34+ cells, the abundance and activity of OXPHOS protein were both unexpectedly elevated with endoplasmic reticulum stress induced by metformin in CML CD34+ cells. The four major aberrantly expressed protein sets, in contrast, were downregulated by metformin in CML CD34+ cells. These data challenged the dependency of OXPHOS for CML CD34+ cell survival and underlined the novel mechanism of metformin. More importantly, it suggested a strong rationale for the use of tyrosine kinase inhibitors in combination with metformin in treating CML.
Collapse
|
10
|
Ficarro SB, Max Alexander W, Tavares I, Marto JA. Open source fraction collector/MALDI spotter for proteomics. HARDWAREX 2022; 11:e00305. [PMID: 35518277 PMCID: PMC9062586 DOI: 10.1016/j.ohx.2022.e00305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/07/2022] [Accepted: 04/09/2022] [Indexed: 06/14/2023]
Abstract
We describe a complete open-source hardware/software solution for high performance thermostatted peptide fraction collection to support mass spectrometry experiments with complex proteomes. The instrument is easy to assemble using parts readily available through retail channels at a fraction of the cost compared to typical commercial systems. Control software is written in Python allowing for rapid customization. We demonstrate several useful applications, including the automated deposition of LC separated peptides for matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) as well as collection and concatenation of peptide fractions from nanoflow HPLC separations.
Collapse
Affiliation(s)
- Scott B. Ficarro
- Department of Cancer Biology, Blais Proteomics Center, Dana-Farber Cancer Institute, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, 360 Longwood Avenue, LC 2208, Boston, MA 02215-5450, USA
| | - William Max Alexander
- Department of Cancer Biology, Blais Proteomics Center, Dana-Farber Cancer Institute, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, 360 Longwood Avenue, LC 2208, Boston, MA 02215-5450, USA
| | - Isidoro Tavares
- Department of Cancer Biology, Blais Proteomics Center, Dana-Farber Cancer Institute, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, 360 Longwood Avenue, LC 2208, Boston, MA 02215-5450, USA
| | - Jarrod A. Marto
- Department of Cancer Biology, Blais Proteomics Center, Dana-Farber Cancer Institute, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, 360 Longwood Avenue, LC 2208, Boston, MA 02215-5450, USA
| |
Collapse
|
11
|
The role of ARHGAP9: clinical implication and potential function in acute myeloid leukemia. J Transl Med 2021; 19:65. [PMID: 33579308 PMCID: PMC7881617 DOI: 10.1186/s12967-021-02733-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/01/2021] [Indexed: 12/16/2022] Open
Abstract
Background Rho GTPase activating protein 9 (ARHGAP9) is expressed in various types of cancers and can inactivate Rho GTPases that mainly regulate cytoskeletal dynamics. However, the exact role of ARHGAP9 in acute myeloid leukemia (AML) has yet to be clarified. Methods We compared the transcriptional expression, prognosis, differentially expressed genes, functional enrichment, and hub genes in AML patients on the basis of the data published in the following databases: UALCAN, GEPIA, Gene Expression Omnibus, the Human Protein Atlas, Cancer Cell Line Encyclopedia, LinkedOmics, Metascape, and String. Data from the Cancer Genome Atlas database was used to evaluate the correlations between ARHGAP9 expression and various clinicopathological parameters, as well as the significantly different genes associated with ARHGAP9 expression. Results We found that ARHGAP9 expression was higher in the tissues and cell lines extracted from patients with AML than corresponding control tissues and other cancer types. ARHGAP9 overexpression was associated with decreased overall survival (OS) in AML. Compared with the ARHGAP9low group, the ARHGAP9high group, which received only chemotherapy, showed significantly worse OS and event-free survival (EFS); however, no significant difference was observed after treatment with autologous or allogeneic hematopoietic stem cell transplantation (auto/allo-HSCT). The ARHGAP9high patients undergoing auto/allo-HSCT also had a significantly better prognosis with respect to OS and EFS than those receiving only chemotherapy. Most overlapping genes of the significantly different genes and co-expression genes exhibited enriched immune functions, suggesting the immune regulation potential of ARHGAP9 in AML. A total of 32 hub genes were identified from the differentially expressed genes, within which the KIF20A had a significant prognostic value for AML. Conclusions ARHGAP9 overexpression was associated with poor OS in AML patients and can be used as a prognostic biomarker. AML patients with ARHGAP9 overexpression can benefit from auto/allo-HSCT rather than chemotherapy.
Collapse
|
12
|
Quantitative proteomics reveals specific metabolic features of acute myeloid leukemia stem cells. Blood 2020; 136:1507-1519. [DOI: 10.1182/blood.2019003654] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Acute myeloid leukemia is characterized by the accumulation of clonal myeloid blast cells unable to differentiate into mature leukocytes. Chemotherapy induces remission in the majority of patients, but relapse rates are high and lead to poor clinical outcomes. Because this is primarily caused by chemotherapy-resistant leukemic stem cells (LSCs), it is essential to eradicate LSCs to improve patient survival. LSCs have predominantly been studied at the transcript level, thus information about posttranscriptionally regulated genes and associated networks is lacking. Here, we extend our previous report on LSC proteomes to healthy age-matched hematopoietic stem and progenitor cells (HSPCs) and correlate the proteomes to the corresponding transcriptomes. By comparing LSCs to leukemic blasts and healthy HSPCs, we validate candidate LSC markers and highlight novel and potentially targetable proteins that are absent or only lowly expressed in HSPCs. In addition, our data provide strong evidence that LSCs harbor a characteristic energy metabolism, adhesion molecule composition, as well as RNA-processing properties. Furthermore, correlating proteome and transcript data of the same individual samples highlights the strength of proteome analyses, which are particularly potent in detecting alterations in metabolic pathways. In summary, our study provides a comprehensive proteomic and transcriptomic characterization of functionally validated LSCs, blasts, and healthy HSPCs, representing a valuable resource helping to design LSC-directed therapies.
Collapse
|
13
|
Lindner SE, Egelston CA, Huard SM, Lee PP, Wang LD. Arhgap25 Deficiency Leads to Decreased Numbers of Peripheral Blood B Cells and Defective Germinal Center Reactions. Immunohorizons 2020; 4:274-281. [PMID: 32434881 DOI: 10.4049/immunohorizons.2000021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/05/2020] [Indexed: 02/03/2023] Open
Abstract
Rho family GTPases are critical for normal B cell development and function, and their activity is regulated by a large and complex network of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). However, the role of GAPs in B cell development is poorly understood. In this study, we show that the novel Rac-GAP ARHGAP25 is important for B cell development in mice in a CXCR4-dependent manner. We show that Arhgap25 deficiency in mice leads to a significant decrease in peripheral blood B cell numbers as well as defects in mature B cell differentiation. Arhgap25-/- B cells respond to Ag stimulation in vitro and in vivo but have impaired germinal center formation and decreased IgG1 class switching. Additionally, Arhgap25-/- B cells show evidence of increased baseline motility and augmented chemotaxis to CXCL12. Taken together, these studies demonstrate an important role for Arhgap25 in peripheral B cell development and Ag response.
Collapse
Affiliation(s)
- Silke E Lindner
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and
| | - Colt A Egelston
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and
| | - Stephanie M Huard
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and
| | - Leo D Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and .,Department of Pediatrics, City of Hope National Medical Center, Duarte, CA 91010
| |
Collapse
|
14
|
Szade A, Szade K, Nowak WN, Bukowska-Strakova K, Muchova L, Gońka M, Żukowska M, Cieśla M, Kachamakova-Trojanowska N, Rams-Baron M, Ratuszna A, Dulak J, Józkowicz A. Cobalt protoporphyrin IX increases endogenous G-CSF and mobilizes HSC and granulocytes to the blood. EMBO Mol Med 2019; 11:e09571. [PMID: 31709729 PMCID: PMC6895613 DOI: 10.15252/emmm.201809571] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/09/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023] Open
Abstract
Granulocyte colony‐stimulating factor (G‐CSF) is used in clinical practice to mobilize cells from the bone marrow to the blood; however, it is not always effective. We show that cobalt protoporphyrin IX (CoPP) increases plasma concentrations of G‐CSF, IL‐6, and MCP‐1 in mice, triggering the mobilization of granulocytes and hematopoietic stem and progenitor cells (HSPC). Compared with recombinant G‐CSF, CoPP mobilizes higher number of HSPC and mature granulocytes. In contrast to G‐CSF, CoPP does not increase the number of circulating T cells. Transplantation of CoPP‐mobilized peripheral blood mononuclear cells (PBMC) results in higher chimerism and faster hematopoietic reconstitution than transplantation of PBMC mobilized by G‐CSF. Although CoPP is used to activate Nrf2/HO‐1 axis, the observed effects are Nrf2/HO‐1 independent. Concluding, CoPP increases expression of mobilization‐related cytokines and has superior mobilizing efficiency compared with recombinant G‐CSF. This observation could lead to the development of new strategies for the treatment of neutropenia and HSPC transplantation.
Collapse
Affiliation(s)
- Agata Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Witold N Nowak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.,Department of Clinical Immunology and Transplantology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Lucie Muchova
- Fourth Department of Internal Medicine and Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Monika Gońka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Monika Żukowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maciej Cieśla
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Neli Kachamakova-Trojanowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.,Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Marzena Rams-Baron
- A. Chelkowski Institute of Physics, University of Silesia, Chorzow, Poland.,Silesian Center for Education and Interdisciplinary Research, Chorzow, Poland
| | - Alicja Ratuszna
- A. Chelkowski Institute of Physics, University of Silesia, Chorzow, Poland.,Silesian Center for Education and Interdisciplinary Research, Chorzow, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.,Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
15
|
de Kruijf EJFM, Fibbe WE, van Pel M. Cytokine-induced hematopoietic stem and progenitor cell mobilization: unraveling interactions between stem cells and their niche. Ann N Y Acad Sci 2019; 1466:24-38. [PMID: 31006885 PMCID: PMC7217176 DOI: 10.1111/nyas.14059] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/15/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023]
Abstract
Peripheral blood hematopoietic stem and progenitor cells (HSPCs), mobilized by granulocyte colony‐stimulating factor, are widely used as a source for both autologous and allogeneic stem cell transplantation. The use of mobilized HSPCs has several advantages over traditional bone marrow–derived HSPCs, including a less invasive harvesting process for the donor, higher HSPC yields, and faster hematopoietic reconstitution in the recipient. For years, the mechanisms by which cytokines and other agents mobilize HSPCs from the bone marrow were not fully understood. The field of stem cell mobilization research has advanced significantly over the past decade, with major breakthroughs in the elucidation of the complex mechanisms that underlie stem cell mobilization. In this review, we provide an overview of the events that underlie HSPC mobilization and address the relevant cellular and molecular components of the bone marrow niche. Furthermore, current and future mobilizing agents will be discussed.
Collapse
Affiliation(s)
- Evert-Jan F M de Kruijf
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Willem E Fibbe
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Melissa van Pel
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
16
|
Csépányi-Kömi R, Pásztor M, Bartos B, Ligeti E. The neglected terminators: Rho family GAPs in neutrophils. Eur J Clin Invest 2018; 48 Suppl 2:e12993. [PMID: 29972685 DOI: 10.1111/eci.12993] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 07/02/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND GTPase-activating proteins (GAPs) accelerate the rate of hydrolysis of GTP bound to small GTPases, thereby limiting the prevalence and concentration of the active, GTP-bound form of these proteins. The large number of potential GAPs acting on members of the Rho family of small GTPases raises the question of specificity or redundancy. RESULTS In this review, we summarize experimental data obtained on the role of Rho family GAPs in neutrophils, highlight cases where more than one GAP is involved in a physiological function and show examples that GAPs can be involved not only in termination but also in initiation of cellular processes. We demonstrate that the expression-level regulation of GAPs may also occur in short-living cells such as neutrophils. Finally, we provide insight into the existence and structure of molecular complexes in which Rho family GAPs are involved. CONCLUSION GAPs play more complex and varied roles than being simple terminators of cellular processes.
Collapse
Affiliation(s)
| | - Máté Pásztor
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Balázs Bartos
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Erzsébet Ligeti
- Department of Physiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
17
|
Brown FC, Still E, Koche RP, Yim CY, Takao S, Cifani P, Reed C, Gunasekera S, Ficarro SB, Romanienko P, Mark W, McCarthy C, de Stanchina E, Gonen M, Seshan V, Bhola P, O'Donnell C, Spitzer B, Stutzke C, Lavallée VP, Hébert J, Krivtsov AV, Melnick A, Paietta EM, Tallman MS, Letai A, Sauvageau G, Pouliot G, Levine R, Marto JA, Armstrong SA, Kentsis A. MEF2C Phosphorylation Is Required for Chemotherapy Resistance in Acute Myeloid Leukemia. Cancer Discov 2018; 8:478-497. [PMID: 29431698 PMCID: PMC5882571 DOI: 10.1158/2159-8290.cd-17-1271] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/22/2018] [Accepted: 01/30/2018] [Indexed: 11/16/2022]
Abstract
In acute myeloid leukemia (AML), chemotherapy resistance remains prevalent and poorly understood. Using functional proteomics of patient AML specimens, we identified MEF2C S222 phosphorylation as a specific marker of primary chemoresistance. We found that Mef2cS222A/S222A knock-in mutant mice engineered to block MEF2C phosphorylation exhibited normal hematopoiesis, but were resistant to leukemogenesis induced by MLL-AF9 MEF2C phosphorylation was required for leukemia stem cell maintenance and induced by MARK kinases in cells. Treatment with the selective MARK/SIK inhibitor MRT199665 caused apoptosis and conferred chemosensitivity in MEF2C-activated human AML cell lines and primary patient specimens, but not those lacking MEF2C phosphorylation. These findings identify kinase-dependent dysregulation of transcription factor control as a determinant of therapy response in AML, with immediate potential for improved diagnosis and therapy for this disease.Significance: Functional proteomics identifies phosphorylation of MEF2C in the majority of primary chemotherapy-resistant AML. Kinase-dependent dysregulation of this transcription factor confers susceptibility to MARK/SIK kinase inhibition in preclinical models, substantiating its clinical investigation for improved diagnosis and therapy of AML. Cancer Discov; 8(4); 478-97. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 371.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Cell Line
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- MEF2 Transcription Factors/chemistry
- MEF2 Transcription Factors/metabolism
- Mice
- Mice, Transgenic
- Phosphorylation
- Protein Processing, Post-Translational
- Proteomics
Collapse
Affiliation(s)
- Fiona C Brown
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric Still
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard P Koche
- Center for Epigenetics Research, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christina Y Yim
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sumiko Takao
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paolo Cifani
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Casie Reed
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shehana Gunasekera
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott B Ficarro
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Peter Romanienko
- Mouse Genetics Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Willie Mark
- Mouse Genetics Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Craig McCarthy
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Venkatraman Seshan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Patrick Bhola
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Conor O'Donnell
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barbara Spitzer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Vincent-Philippe Lavallée
- The Leucegene Project at Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - Josée Hébert
- The Leucegene Project at Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Andrei V Krivtsov
- Center for Epigenetics Research, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ari Melnick
- Departments of Pediatrics, Pharmacology, and Physiology and Biophysics, Weill Cornell Medical College, Cornell University, New York, New York
| | - Elisabeth M Paietta
- Montefiore Medical Center-North Division, Albert Einstein College of Medicine, Bronx, New York, New York
| | - Martin S Tallman
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Guy Sauvageau
- The Leucegene Project at Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Gayle Pouliot
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ross Levine
- Center for Epigenetics Research, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center and Weill Medical College of Cornell University, New York, New York
| | - Jarrod A Marto
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Scott A Armstrong
- Center for Epigenetics Research, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alex Kentsis
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York.
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Departments of Pediatrics, Pharmacology, and Physiology and Biophysics, Weill Cornell Medical College, Cornell University, New York, New York
| |
Collapse
|
18
|
Abazova N, Krijgsveld J. Advances in stem cell proteomics. Curr Opin Genet Dev 2017; 46:149-155. [PMID: 28806595 DOI: 10.1016/j.gde.2017.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 12/15/2022]
Abstract
Stem cells are at the basis of organismal development, characterized by their potential to differentiate towards specific lineages upon receiving proper signals. To understand the molecular principles underlying gain and loss of pluripotency, proteomics plays an increasingly important role owing to technical developments in mass spectrometry and implementation of innovative biochemical approaches. Here we review how quantitative proteomics has been used to investigate protein expression, localization, interaction and modification in stem cells both in vitro and in vivo, thereby complementing other omics approaches to study fundamental properties of stem cell plasticity.
Collapse
Affiliation(s)
- Nade Abazova
- German Cancer Research Center (DKFZ), Heidelberg, Germany; Excellence Cluster CellNetworks, Heidelberg University, Heidelberg, Germany; European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Jeroen Krijgsveld
- German Cancer Research Center (DKFZ), Heidelberg, Germany; Excellence Cluster CellNetworks, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
19
|
How do stem cells mobilize? Bridging the GAP. Blood 2016; 128:1445-6. [PMID: 27633605 DOI: 10.1182/blood-2016-07-726414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
20
|
Csépányi-Kömi R, Wisniewski É, Bartos B, Lévai P, Németh T, Balázs B, Kurz ARM, Bierschenk S, Sperandio M, Ligeti E. Rac GTPase Activating Protein ARHGAP25 Regulates Leukocyte Transendothelial Migration in Mice. THE JOURNAL OF IMMUNOLOGY 2016; 197:2807-15. [PMID: 27566826 DOI: 10.4049/jimmunol.1502342] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 07/21/2016] [Indexed: 12/31/2022]
Abstract
ARHGAP25 is a Rac-specific GTPase-activating protein that is expressed primarily in hematopoietic cells. The involvement of ARHGAP25 in regulating the recruitment of leukocytes to inflammatory sites was investigated in genetically modified mice. Using intravital microscopy, we show that Arhgap25 deficiency affects all steps of leukocyte recruitment with a predominant enhancement of transendothelial migration of neutrophilic granulocytes. Increased transmigration of Arhgap25-deficient leukocytes is demonstrated in inflamed cremaster muscle venules, in a peritonitis model, and in an in vitro chemotaxis assay. Using bone marrow chimeric mice lacking ARHGAP25 in the hematopoietic compartment, we show that enhanced migration in the absence of ARHGAP25 is due to defective leukocyte function. In search for potential mechanisms of ARHGAP25-regulated migration of neutrophils, we detected an increase in the amount of active, GTP-bound Rac and Rac-dependent cytoskeletal changes in the absence of ARHGAP25, suggesting a critical role of ARHGAP25 in counterbalancing the Rac-activating effect of nucleotide exchange factors. Taken together, using Arhgap25-deficient mice, we identified ARHGAP25 as a relevant negative regulator of leukocyte transendothelial migration.
Collapse
Affiliation(s)
- Roland Csépányi-Kömi
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; and Walter-Brendel-Zentrum für Experimentelle Medizin, Ludwig-Maximilians Universität, 80539 Munich, Germany
| | - Éva Wisniewski
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; and
| | - Balázs Bartos
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; and
| | - Petra Lévai
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; and
| | - Tamás Németh
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; and
| | - Bernadett Balázs
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; and
| | - Angela R M Kurz
- Walter-Brendel-Zentrum für Experimentelle Medizin, Ludwig-Maximilians Universität, 80539 Munich, Germany
| | - Susanne Bierschenk
- Walter-Brendel-Zentrum für Experimentelle Medizin, Ludwig-Maximilians Universität, 80539 Munich, Germany
| | - Markus Sperandio
- Walter-Brendel-Zentrum für Experimentelle Medizin, Ludwig-Maximilians Universität, 80539 Munich, Germany
| | - Erzsébet Ligeti
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; and
| |
Collapse
|