1
|
Carretta C, Parenti S, Bertesi M, Rontauroli S, Badii F, Tavernari L, Genovese E, Malerba M, Papa E, Sperduti S, Enzo E, Mirabile M, Pedrazzi F, Neroni A, Tombari C, Mora B, Maffioli M, Mondini M, Brociner M, Maccaferri M, Tenedini E, Martinelli S, Bartalucci N, Bianchi E, Casarini L, Potenza L, Luppi M, Tagliafico E, Guglielmelli P, Simoni M, Passamonti F, Norfo R, Vannucchi AM, Manfredini R. Chromosome 9p trisomy increases stem cells clonogenic potential and fosters T-cell exhaustion in JAK2-mutant myeloproliferative neoplasms. Leukemia 2024; 38:2171-2182. [PMID: 39179669 PMCID: PMC11436358 DOI: 10.1038/s41375-024-02373-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024]
Abstract
JAK2V617F is the most recurrent genetic mutation in Philadelphia-negative chronic Myeloproliferative Neoplasms (MPNs). Since the JAK2 locus is located on Chromosome 9, we hypothesized that Chromosome 9 copy number abnormalities may be a disease modifier in JAK2V617F-mutant MPN patients. In this study, we identified a subset of MPN patients with partial or complete Chromosome 9 trisomy (+9p patients), who differ from JAK2V617F-homozygous MPN patients as they carry three JAK2 alleles as well as three copies of all neighboring gene loci, including CD274, encoding immunosuppressive Programmed death-ligand 1 (PD-L1) protein. Investigation of the clonal hierarchy revealed that the JAK2V617F occurs first, followed by +9p. Functionally, CD34+ cells from +9p MPN patients demonstrated increased clonogenicity, generating a greater number of primitive colonies, due to high OCT4 and NANOG expression, with knock-down of these genes leading to a genotype-specific decrease in colony numbers. Moreover, our analysis revealed increased PD-L1 surface expression in malignant monocytes from +9p patients, while analysis of the T cell compartment unveiled elevated levels of exhausted cytotoxic T cells. Overall, here we identify a distinct novel subgroup of MPN patients, who feature a synergistic interplay between +9p and JAK2V617F that shapes immune escape characteristics and increased stemness in CD34+ cells.
Collapse
Affiliation(s)
- Chiara Carretta
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sandra Parenti
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Bertesi
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sebastiano Rontauroli
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Filippo Badii
- Department of Cancer Biology, Thomas Jefferson University and Sidney Kimmel Cancer Center, Philadelphia, PA, USA
| | - Lara Tavernari
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Genovese
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marica Malerba
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Papa
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Samantha Sperduti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Enzo
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Margherita Mirabile
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Pedrazzi
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anita Neroni
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Camilla Tombari
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Margherita Maffioli
- S.C. Ematologia, Ospedale di Circolo e Fondazione Macchi-ASST Sette Laghi, Varese, Italy
| | - Marco Mondini
- S.C. Ematologia, Ospedale di Circolo e Fondazione Macchi-ASST Sette Laghi, Varese, Italy
| | - Marco Brociner
- S.C. Ematologia, Ospedale di Circolo e Fondazione Macchi-ASST Sette Laghi, Varese, Italy
| | | | - Elena Tenedini
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Martinelli
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Niccolò Bartalucci
- CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence, AOU Careggi, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elisa Bianchi
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Leonardo Potenza
- Hematology Unit, Modena University Hospital, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Mario Luppi
- Hematology Unit, Modena University Hospital, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Paola Guglielmelli
- CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence, AOU Careggi, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Ruggiero Norfo
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Maria Vannucchi
- CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence, AOU Careggi, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Rossella Manfredini
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy.
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
2
|
Vadeikienė R, Jakštys B, Laukaitienė D, Šatkauskas S, Juozaitytė E, Ugenskienė R. The Role of Mutated Calreticulin in the Pathogenesis of BCR-ABL1-Negative Myeloproliferative Neoplasms. Int J Mol Sci 2024; 25:9873. [PMID: 39337361 PMCID: PMC11432199 DOI: 10.3390/ijms25189873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs) are characterized by increased proliferation of myeloid lineages in the bone marrow. Calreticulin (CALR) 52 bp deletion and CALR 5 bp insertion have been identified in essential thrombocythemia (ET) and primary myelofibrosis (PMF). There is not much data on the crosstalk between mutated CALR and MPN-related signaling pathways, such as JAK/STAT, PI3K/Akt/mTOR, and Hedgehog. Calreticulin, a multifunctional protein, takes part in many cellular processes. Nevertheless, there is little data on how mutated CALR affects the oxidative stress response and oxidative stress-induced DNA damage, apoptosis, and cell cycle progression. We aimed to investigate the role of the CALR 52 bp deletion and 5 bp insertion in the pathogenesis of MPN, including signaling pathway activation and functional analysis in CALR-mutated cells. Our data indicate that the JAK/STAT and PI3K/Akt/mTOR pathways are activated in CALR-mutated cells, and this activation does not necessarily depend on the CALR and MPL interaction. Moreover, it was found that CALR mutations impair calreticulin function, leading to reduced responses to oxidative stress and DNA damage. It was revealed that the accumulation of G2/M-CALR-mutated cells indicates that oxidative stress-induced DNA damage is difficult to repair. Taken together, this study contributes to a deeper understanding of the specific molecular mechanisms underlying CALR-mutated MPNs.
Collapse
Affiliation(s)
- Roberta Vadeikienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Baltramiejus Jakštys
- Research on Delivery of Medicine and Genes Cluster, Faculty of Natural Sciences, Vytautas Magnus University, LT-44001 Kaunas, Lithuania
| | - Danguolė Laukaitienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Saulius Šatkauskas
- Research on Delivery of Medicine and Genes Cluster, Faculty of Natural Sciences, Vytautas Magnus University, LT-44001 Kaunas, Lithuania
| | - Elona Juozaitytė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Rasa Ugenskienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| |
Collapse
|
3
|
Zheng G, Fang Z, Lin Z, Guan G. Miltirone induces GSDME-dependent pyroptosis in colorectal cancer by activating caspase 3. Heliyon 2024; 10:e36603. [PMID: 39262975 PMCID: PMC11388397 DOI: 10.1016/j.heliyon.2024.e36603] [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: 05/13/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Colorectal cancer (CRC) is a common and malignant tumor, ranking as the third most common cancer in men and the second most common cancer in women. Pyroptosis, a recently described programmed cell death mechanism mediated by the GSDM family, has emerged as an immunogenic mechanism for chemotherapy drugs in tumor treatment. In this study, we discovered that Miltirone has the ability to reduce the viability of CRC cells (SW620 and HCT116) and cause the proteolytic cleavage of gasdermin E (GSDME) in CRC cells. It was also observed that inhibiting GSDME prevented pyroptotic cell death induced by Miltirone in SW620 and HCT116 cells. Furthermore, the main active component of Miltirone was found to effectively bind with caspase 3. SiRNA-mediated caspase 3 silencing and specific caspase 3 inhibitor Z-DEVD-FMK were shown to weaken Miltirone-induced GSDME-dependent cell death. The findings of the study suggest that Miltirone has the potential to inhibit the growth of CRC tumors in vivo by inducing pyroptotic cell death. This indicates that Miltirone could be a viable therapeutic agent for the treatment of CRC through GSDME-dependent pyroptosis. These results offer a promising new option for the clinical treatment of CRC.
Collapse
Affiliation(s)
- Guangwei Zheng
- Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Emergency Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Zhipeng Fang
- Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Emergency Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Zhenlv Lin
- Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Emergency Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Guoxian Guan
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Colorectal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| |
Collapse
|
4
|
Găman MA, Mambet C, Neagu AI, Bleotu C, Gurban P, Necula L, Botezatu A, Ataman M, Diaconu CC, Ionescu BO, Ghiaur AE, Tatic A, Coriu D, Găman AM, Diaconu CC. Assessment of Total Antioxidant Capacity, 8-Hydroxy-2'-deoxy-guanosine, the Genetic Landscape, and Their Associations in BCR::ABL-1-Negative Chronic and Blast Phase Myeloproliferative Neoplasms. Int J Mol Sci 2024; 25:6652. [PMID: 38928358 PMCID: PMC11203765 DOI: 10.3390/ijms25126652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/12/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs), namely, polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are clonal stem cell disorders defined by an excessive production of functionally mature and terminally differentiated myeloid cells. MPNs can transform into secondary acute myeloid leukemia (sAML/blast phase MPN) and are linked to alterations in the redox balance, i.e., elevated concentrations of reactive oxygen species and markers of oxidative stress (OS), and changes in antioxidant systems. We evaluated OS in 117 chronic phase MPNs and 21 sAML cases versus controls by measuring total antioxidant capacity (TAC) and 8-hydroxy-2'-deoxy-guanosine (8-OHdG) concentrations. TAC was higher in MPNs than controls (p = 0.03), particularly in ET (p = 0.04) and PMF (p = 0.01). MPL W515L-positive MPNs had higher TAC than controls (p = 0.002) and triple-negative MPNs (p = 0.01). PMF patients who had treatment expressed lower TAC than therapy-free subjects (p = 0.03). 8-OHdG concentrations were similar between controls and MPNs, controls and sAML, and MPNs and sAML. We noted associations between TAC and MPNs (OR = 1.82; p = 0.05), i.e., ET (OR = 2.36; p = 0.03) and PMF (OR = 2.11; p = 0.03), but not sAML. 8-OHdG concentrations were not associated with MPNs (OR = 1.73; p = 0.62) or sAML (OR = 1.89; p = 0.49). In conclusion, we detected redox imbalances in MPNs based on disease subtype, driver mutations, and treatment history.
Collapse
Affiliation(s)
- Mihnea-Alexandru Găman
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 010221 Bucharest, Romania; (M.-A.G.); (C.M.); (C.C.D.); (A.T.); (D.C.)
- Department of Hematology, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania; (B.O.I.); (A.E.G.)
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.I.N.); (C.B.); (P.G.); (L.N.); (A.B.); (M.A.); (C.C.D.)
| | - Cristina Mambet
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 010221 Bucharest, Romania; (M.-A.G.); (C.M.); (C.C.D.); (A.T.); (D.C.)
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.I.N.); (C.B.); (P.G.); (L.N.); (A.B.); (M.A.); (C.C.D.)
| | - Ana Iulia Neagu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.I.N.); (C.B.); (P.G.); (L.N.); (A.B.); (M.A.); (C.C.D.)
| | - Coralia Bleotu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.I.N.); (C.B.); (P.G.); (L.N.); (A.B.); (M.A.); (C.C.D.)
| | - Petruta Gurban
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.I.N.); (C.B.); (P.G.); (L.N.); (A.B.); (M.A.); (C.C.D.)
| | - Laura Necula
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.I.N.); (C.B.); (P.G.); (L.N.); (A.B.); (M.A.); (C.C.D.)
| | - Anca Botezatu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.I.N.); (C.B.); (P.G.); (L.N.); (A.B.); (M.A.); (C.C.D.)
| | - Marius Ataman
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.I.N.); (C.B.); (P.G.); (L.N.); (A.B.); (M.A.); (C.C.D.)
| | - Camelia Cristina Diaconu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 010221 Bucharest, Romania; (M.-A.G.); (C.M.); (C.C.D.); (A.T.); (D.C.)
| | - Bogdan Octavian Ionescu
- Department of Hematology, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania; (B.O.I.); (A.E.G.)
| | - Alexandra Elena Ghiaur
- Department of Hematology, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania; (B.O.I.); (A.E.G.)
| | - Aurelia Tatic
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 010221 Bucharest, Romania; (M.-A.G.); (C.M.); (C.C.D.); (A.T.); (D.C.)
- Department of Hematology, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania; (B.O.I.); (A.E.G.)
| | - Daniel Coriu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 010221 Bucharest, Romania; (M.-A.G.); (C.M.); (C.C.D.); (A.T.); (D.C.)
- Department of Hematology, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania; (B.O.I.); (A.E.G.)
| | - Amelia Maria Găman
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Clinic of Hematology, Filantropia City Hospital, 200143 Craiova, Romania
| | - Carmen Cristina Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.I.N.); (C.B.); (P.G.); (L.N.); (A.B.); (M.A.); (C.C.D.)
| |
Collapse
|
5
|
Haage TR, Charakopoulos E, Bhuria V, Baldauf CK, Korthals M, Handschuh J, Müller P, Li J, Harit K, Nishanth G, Frey S, Böttcher M, Fischer KD, Dudeck J, Dudeck A, Lipka DB, Schraven B, Green AR, Müller AJ, Mougiakakos D, Fischer T. Neutrophil-specific expression of JAK2-V617F or CALRmut induces distinct inflammatory profiles in myeloproliferative neoplasia. J Hematol Oncol 2024; 17:43. [PMID: 38853260 PMCID: PMC11163796 DOI: 10.1186/s13045-024-01562-5] [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: 03/22/2024] [Accepted: 05/29/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Neutrophils play a crucial role in inflammation and in the increased thrombotic risk in myeloproliferative neoplasms (MPNs). We have investigated how neutrophil-specific expression of JAK2-V617F or CALRdel re-programs the functions of neutrophils. METHODS Ly6G-Cre JAK2-V617F and Ly6G-Cre CALRdel mice were generated. MPN parameters as blood counts, splenomegaly and bone marrow histology were compared to wild-type mice. Megakaryocyte differentiation was investigated using lineage-negative bone marrow cells upon in vitro incubation with TPO/IL-1β. Cytokine concentrations in serum of mice were determined by Mouse Cytokine Array. IL-1α expression in various hematopoietic cell populations was determined by intracellular FACS analysis. RNA-seq to analyse gene expression of inflammatory cytokines was performed in isolated neutrophils from JAK2-V617F and CALR-mutated mice and patients. Bioenergetics of neutrophils were recorded on a Seahorse extracellular flux analyzer. Cell motility of neutrophils was monitored in vitro (time lapse microscopy), and in vivo (two-photon microscopy) upon creating an inflammatory environment. Cell adhesion to integrins, E-selectin and P-selection was investigated in-vitro. Statistical analysis was carried out using GraphPad Prism. Data are shown as mean ± SEM. Unpaired, two-tailed t-tests were applied. RESULTS Strikingly, neutrophil-specific expression of JAK2-V617F, but not CALRdel, was sufficient to induce pro-inflammatory cytokines including IL-1 in serum of mice. RNA-seq analysis in neutrophils from JAK2-V617F mice and patients revealed a distinct inflammatory chemokine signature which was not expressed in CALR-mutant neutrophils. In addition, IL-1 response genes were significantly enriched in neutrophils of JAK2-V617F patients as compared to CALR-mutant patients. Thus, JAK2-V617F positive neutrophils, but not CALR-mutant neutrophils, are pathogenic drivers of inflammation in MPN. In line with this, expression of JAK2-V617F or CALRdel elicited a significant difference in the metabolic phenotype of neutrophils, suggesting a stronger inflammatory activity of JAK2-V617F cells. Furthermore, JAK2-V617F, but not CALRdel, induced a VLA4 integrin-mediated adhesive phenotype in neutrophils. This resulted in reduced neutrophil migration in vitro and in an inflamed vessel. This mechanism may contribute to the increased thrombotic risk of JAK2-V617F patients compared to CALR-mutant individuals. CONCLUSIONS Taken together, our findings highlight genotype-specific differences in MPN-neutrophils that have implications for the differential pathophysiology of JAK2-V617F versus CALR-mutant disease.
Collapse
Affiliation(s)
- Tobias Ronny Haage
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Emmanouil Charakopoulos
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Vikas Bhuria
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
| | - Conny K Baldauf
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Mark Korthals
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Juliane Handschuh
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Müller
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Juan Li
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, GB, England
| | - Kunjan Harit
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Gopala Nishanth
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Stephanie Frey
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Martin Böttcher
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Jan Dudeck
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Dudeck
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniel B Lipka
- Section of Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
- Faculty of Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Burkhart Schraven
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
| | - Anthony R Green
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, GB, England
| | - Andreas J Müller
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Thomas Fischer
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany.
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
6
|
Holl K, Chatain N, Krapp S, Baumeister J, Maié T, Schmitz S, Scheufen A, Brock N, Koschmieder S, Moreno-Andrés D. Calreticulin and JAK2V617F driver mutations induce distinct mitotic defects in myeloproliferative neoplasms. Sci Rep 2024; 14:2810. [PMID: 38308077 PMCID: PMC10837458 DOI: 10.1038/s41598-024-53240-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/30/2024] [Indexed: 02/04/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs) encompass a diverse group of hematologic disorders driven by mutations in JAK2, CALR, or MPL. The prevailing working model explaining how these driver mutations induce different disease phenotypes is based on the decisive influence of the cellular microenvironment and the acquisition of additional mutations. Here, we report increased levels of chromatin segregation errors in hematopoietic cells stably expressing CALRdel52 or JAK2V617F mutations. Our investigations employing murine 32DMPL and human erythroleukemic TF-1MPL cells demonstrate a link between CALRdel52 or JAK2V617F expression and a compromised spindle assembly checkpoint (SAC), a phenomenon contributing to error-prone mitosis. This defective SAC is associated with imbalances in the recruitment of SAC factors to mitotic kinetochores upon CALRdel52 or JAK2V617F expression. We show that JAK2 mutant CD34 + MPN patient-derived cells exhibit reduced expression of the master mitotic regulators PLK1, aurora kinase B, and PP2A catalytic subunit. Furthermore, the expression profile of mitotic regulators in CD34 + patient-derived cells allows to faithfully distinguish patients from healthy controls, as well as to differentiate primary and secondary myelofibrosis from essential thrombocythemia and polycythemia vera. Altogether, our data suggest alterations in mitotic regulation as a potential driver in the pathogenesis in MPN.
Collapse
Affiliation(s)
- Kristin Holl
- Institute of Biochemistry and Molecular Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center of Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Susanne Krapp
- Institute of Biochemistry and Molecular Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Julian Baumeister
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center of Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Tiago Maié
- Institute for Computational Genomics, Joint Research Center for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Sarah Schmitz
- Institute of Biochemistry and Molecular Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Anja Scheufen
- Institute of Biochemistry and Molecular Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Nathalie Brock
- Institute of Biochemistry and Molecular Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center of Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Daniel Moreno-Andrés
- Institute of Biochemistry and Molecular Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
7
|
Wildschut MHE, Mena J, Dördelmann C, van Oostrum M, Hale BD, Settelmeier J, Festl Y, Lysenko V, Schürch PM, Ring A, Severin Y, Bader MS, Pedrioli PGA, Goetze S, van Drogen A, Balabanov S, Skoda RC, Lopes M, Wollscheid B, Theocharides APA, Snijder B. Proteogenetic drug response profiling elucidates targetable vulnerabilities of myelofibrosis. Nat Commun 2023; 14:6414. [PMID: 37828014 PMCID: PMC10570306 DOI: 10.1038/s41467-023-42101-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Myelofibrosis is a hematopoietic stem cell disorder belonging to the myeloproliferative neoplasms. Myelofibrosis patients frequently carry driver mutations in either JAK2 or Calreticulin (CALR) and have limited therapeutic options. Here, we integrate ex vivo drug response and proteotype analyses across myelofibrosis patient cohorts to discover targetable vulnerabilities and associated therapeutic strategies. Drug sensitivities of mutated and progenitor cells were measured in patient blood using high-content imaging and single-cell deep learning-based analyses. Integration with matched molecular profiling revealed three targetable vulnerabilities. First, CALR mutations drive BET and HDAC inhibitor sensitivity, particularly in the absence of high Ras pathway protein levels. Second, an MCM complex-high proliferative signature corresponds to advanced disease and sensitivity to drugs targeting pro-survival signaling and DNA replication. Third, homozygous CALR mutations result in high endoplasmic reticulum (ER) stress, responding to ER stressors and unfolded protein response inhibition. Overall, our integrated analyses provide a molecularly motivated roadmap for individualized myelofibrosis patient treatment.
Collapse
Affiliation(s)
- Mattheus H E Wildschut
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Department of Medical Oncology and Hematology, Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Julien Mena
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Cyril Dördelmann
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Marc van Oostrum
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Benjamin D Hale
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jens Settelmeier
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Yasmin Festl
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Veronika Lysenko
- Department of Medical Oncology and Hematology, Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Patrick M Schürch
- Department of Medical Oncology and Hematology, Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Alexander Ring
- Department of Medical Oncology and Hematology, Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Yannik Severin
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Michael S Bader
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Patrick G A Pedrioli
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), Zurich, Switzerland
| | - Sandra Goetze
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), Zurich, Switzerland
| | - Audrey van Drogen
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), Zurich, Switzerland
| | - Stefan Balabanov
- Department of Medical Oncology and Hematology, Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Radek C Skoda
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Massimo Lopes
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Bernd Wollscheid
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Alexandre P A Theocharides
- Department of Medical Oncology and Hematology, Division of Hematology, University Hospital Zurich, Zurich, Switzerland.
| | - Berend Snijder
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
8
|
Yu G, Gao J, Hu W, Hu D, Wang W, Yang S, Gao J. ORM1 promotes tumor progression of kidney renal clear cell carcinoma (KIRC) through CALR-mediated apoptosis. Sci Rep 2023; 13:15687. [PMID: 37735575 PMCID: PMC10514263 DOI: 10.1038/s41598-023-42962-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) is the most prevalent type of kidney cancer and causes thousands of deaths each year. The prognosis for KIRC is poor. One critical factor is that the mechanism beneath KIRC is unclear. ORM1 is a reactant to acute inflammation. In this study, we demonstrated that methylation of ORM1 promoter was low and ORM1 was expressed significantly higher in KIRC. KIRC with higher ORM1 expression exhibited worse survival probability. Meanwhile, ORM1 was expressed higher in KIRC cell lines. When ORM1 was knocked down, cell proliferation ability was inhibited potently compared to the NC control. Cell migration as well as invasion ability were also suppressed dramatically. At molecular level, the expression of active caspase-3 and Bax was upregulated in ORM1-KD group while Bcl-2 downregulated. Moreover, CALR decreased following ORM1-KD and rescued expression of CALR increased Bcl-2 level but reduced the level of cleaved caspase-3 and Bax. Consistently, the apoptotic rate of 786-O and Caki-2 cells was upregulated in ORM1-KD but downregulated after CALR overexpression. The activity of caspase-3 was also regulated by ORM1-KD. In addition, the inhibition rate of sorafenib was enhanced in ORM1 KD group but reduced after overexpression of ORM1. Conclusively, ORM1 is clinically associated with progression of KIRC and regulates cell proliferation, migration, invasion, and apoptosis in KIRC. Moreover, ORM1 affects the efficiency of sorafenib in KIRC and regulates caspase-3 mediated cascades response through CALR molecule. This study provides us a new way to recognize the development and progression in KIRC.
Collapse
Affiliation(s)
- Gang Yu
- Department of Nephrology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, 200233, China
| | - Juan Gao
- Physical Examination Center, Shanghai Lung Hospital Affiliated to Tongji University, Shanghai, 200433, China
| | - Weifeng Hu
- Department of Nephrology, PLA Naval Medical Center, Shanghai, 200052, China
| | - Dayong Hu
- Department of Nephrology, The Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Weibing Wang
- Department of Epidemiology, School of Public Health of Fudan University, Shanghai, 200032, China
| | - Shiquan Yang
- Department of General Practice, Xujiahui Community Healthcare Center of Xuhui District of Shanghai, West Guangyuan Road No.349, Shanghai, 200030, China.
| | - Jing Gao
- Department of General Practice, Xujiahui Community Healthcare Center of Xuhui District of Shanghai, West Guangyuan Road No.349, Shanghai, 200030, China.
| |
Collapse
|
9
|
Palacon MP, Ferrisse TM, Barbeiro CDO, Massucato EMS, Bufalino A. Calreticulin Expression in Human Carcinomas: A Systematic Review and Meta-Analysis. Asian Pac J Cancer Prev 2023; 24:2929-2940. [PMID: 37774043 PMCID: PMC10762747 DOI: 10.31557/apjcp.2023.24.9.2929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
OBJECTIVE The present study performed a systematic review and meta-analysis of observational studies on whether calreticulin levels could represent a prognostic factor in carcinoma patients. Calreticulin (CRT) is a multifunctional protein in the endoplasmic reticulum that can play distinct roles in different cancers. METHODS The search was performed in PubMed, Scopus, the Cochrane Library, Web of Science, Lilacs, Science Direct, Embase, Bireme, and SciELO databases. After a full-text evaluation, only 14 articles remained. The RoBANS tool assessed the risk of bias. The meta-analysis was performed with R software, and the odds ratio (OR) was the effect measure. The random effects model was chosen, and the quality of evidence was evaluated according to GRADE. RESULT The most frequent carcinomas were in the breasts and the colon. CRT expression varied according to carcinoma origin and type, but these diseases had a prevalence of high CRT levels, indicating tumor progression. The high CRT levels were associated with lymph node metastasis (OR = 3.06 [1.71; 5.48]/p = 0.0002/I2 = 0%). All included articles had a blinding bias. CONCLUSION High CRT levels may represent a prognostic factor for metastatic lymph nodes in carcinoma patients.
Collapse
Affiliation(s)
- Mariana Paravani Palacon
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Tulio Morandin Ferrisse
- Department of Dental Prothesis, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Camila de Oliveira Barbeiro
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Elaine Maria Sgavioli Massucato
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Andreia Bufalino
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| |
Collapse
|
10
|
Li G, Yu X, Portela Fontoura AB, Javaid A, de la Maza-Escolà VS, Salandy NS, Fubini SL, Grilli E, McFadden JW, Duan JE. Transcriptomic regulations of heat stress response in the liver of lactating dairy cows. BMC Genomics 2023; 24:410. [PMID: 37474909 PMCID: PMC10360291 DOI: 10.1186/s12864-023-09484-1] [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: 04/07/2023] [Accepted: 06/24/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND The global dairy industry is currently facing the challenge of heat stress (HS). Despite the implementation of various measures to mitigate the negative impact of HS on milk production, the cellular response of dairy cows to HS is still not well understood. Our study aims to analyze transcriptomic dynamics and functional changes in the liver of cows subjected to heat stress (HS). To achieve this, a total of 9 Holstein dairy cows were randomly selected from three environmental conditions - heat stress (HS), pair-fed (PF), and thermoneutral (TN) groups - and liver biopsies were obtained for transcriptome analysis. RESULTS Both the dry matter intake (DMI) and milk yield of cows in the HS group exhibited significant reduction compared to the TN group. Through liver transcriptomic analysis, 483 differentially expressed genes (DEGs) were identified among three experimental groups. Especially, we found all the protein coding genes in mitochondria were significantly downregulated under HS and 6 heat shock proteins were significant upregulated after HS exposure, indicating HS may affect mitochondria integrity and jeopardize the metabolic homeostasis in liver. Furthermore, Gene ontology (GO) enrichment of DEGs revealed that the protein folding pathway was upregulated while oxidative phosphorylation was downregulated in the HS group, corresponding to impaired energy production caused by mitochondria dysfunction. CONCLUSIONS The liver transcriptome analysis generated a comprehensive gene expression regulation network upon HS in lactating dairy cows. Overall, this study provides novel insights into molecular and metabolic changes of cows conditioned under HS. The key genes and pathways identified in this study provided further understanding of transcriptome regulation of HS response and could serve as vital references to mitigate the HS effects on dairy cow health and productivity.
Collapse
Affiliation(s)
- Guangsheng Li
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, 14853, USA
| | - Xingtan Yu
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, 14853, USA
| | - Ananda B Portela Fontoura
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, 14853, USA
| | - Awais Javaid
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, 14853, USA
| | - Víctor Sáinz de la Maza-Escolà
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, 14853, USA
- Dipartamento di Scienze Mediche Veterinarie, Università di Bologna, Bologna, 40064, Italy
| | - Nia S Salandy
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, 14853, USA
- Department of Agriculture and Environmental Sciences, Tuskegee University, Tuskegee, 36088, USA
| | - Susan L Fubini
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, 14853, USA
| | - Ester Grilli
- Dipartamento di Scienze Mediche Veterinarie, Università di Bologna, Bologna, 40064, Italy
- VetAgro S.p.A, Reggio Emilia, 42124, Italy
| | - Joseph W McFadden
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, 14853, USA.
| | - Jingyue Ellie Duan
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, 14853, USA.
| |
Collapse
|
11
|
Bestepe F, Fritsche C, Lakhotiya K, Niosi CE, Ghanem GF, Martin GL, Pal-Ghosh R, Becker-Greene D, Weston J, Hollan I, Risnes I, Rynning SE, Solheim LH, Feinberg MW, Blanton RM, Icli B. Deficiency of miR-409-3p improves myocardial neovascularization and function through modulation of DNAJB9/p38 MAPK signaling. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:995-1009. [PMID: 37332476 PMCID: PMC10276151 DOI: 10.1016/j.omtn.2023.05.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/17/2023] [Indexed: 06/20/2023]
Abstract
Angiogenesis is critical for tissue repair following myocardial infarction (MI), which is exacerbated under insulin resistance or diabetes. MicroRNAs are regulators of angiogenesis. We examined the metabolic regulation of miR-409-3p in post-infarct angiogenesis. miR-409-3p was increased in patients with acute coronary syndrome (ACS) and in a mouse model of acute MI. In endothelial cells (ECs), miR-409-3p was induced by palmitate, while vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) decreased its expression. Overexpression of miR-409-3p decreased EC proliferation and migration in the presence of palmitate, whereas inhibition had the opposite effects. RNA sequencing (RNA-seq) profiling in ECs identified DNAJ homolog subfamily B member 9 (DNAJB9) as a target of miR-409-3p. Overexpression of miR-409-3p decreased DNAJB9 mRNA and protein expression by 47% and 31% respectively, while enriching DNAJB9 mRNA by 1.9-fold after Argonaute2 microribonucleoprotein immunoprecipitation. These effects were mediated through p38 mitogen-activated protein kinase (MAPK). Ischemia-reperfusion (I/R) injury in EC-specific miR-409-3p knockout (KO) mice (miR-409ECKO) fed a high-fat, high-sucrose diet increased isolectin B4 (53.3%), CD31 (56%), and DNAJB9 (41.5%). The left ventricular ejection fraction (EF) was improved by 28%, and the infarct area was decreased by 33.8% in miR-409ECKO compared with control mice. These findings support an important role of miR-409-3p in the angiogenic EC response to myocardial ischemia.
Collapse
Affiliation(s)
- Furkan Bestepe
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Colette Fritsche
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Kartik Lakhotiya
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Carolyn E. Niosi
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - George F. Ghanem
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Gregory L. Martin
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Ruma Pal-Ghosh
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Dakota Becker-Greene
- Cardiovascular Division, Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - James Weston
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Ivana Hollan
- Department of Health Sciences, Norwegian University of Science and Technology, Gjøvik, Norway
| | - Ivar Risnes
- Department of Cardiac Surgery, LHL Hospital Gardermoen, Jessheim, Norway
| | - Stein Erik Rynning
- Department of Heart Diseases, Haukeland University Hospital, Bergen, Norway
| | | | - Mark W. Feinberg
- Cardiovascular Division, Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Robert M. Blanton
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Basak Icli
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| |
Collapse
|
12
|
Zarate SM, Huntington TE, Bagher P, Srinivasan R. Aging reduces calreticulin expression and alters spontaneous calcium signals in astrocytic endfeet of the mouse dorsolateral striatum. NPJ AGING 2023; 9:5. [PMID: 37002232 PMCID: PMC10066375 DOI: 10.1038/s41514-023-00102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023]
Abstract
Aging-related impairment of the blood brain barrier (BBB) and neurovascular unit (NVU) increases the risk for neurodegeneration. Among various cells that participate in BBB and NVU function, calcium signals in astrocytic endfeet are crucial for maintaining BBB and NVU integrity. To assess if aging is associated with altered calcium signals within astrocytic endfeet of the dorsolateral striatum (DLS), we expressed GCaMP6f in DLS astrocytes of young (3-4 months), middle-aged (12-15 months) and aging (20-30 months) mice. Compared to endfeet in young mice, DLS endfeet in aging mice demonstrated decreased calreticulin expression, and alterations to both spontaneous membrane-associated and mitochondrial calcium signals. While young mice required both extracellular and endoplasmic reticulum calcium sources for endfoot signals, middle-aged and aging mice showed heavy dependence on endoplasmic reticulum calcium. Thus, astrocytic endfeet show significant changes in calcium buffering and sources throughout the lifespan, which is important for understanding mechanisms by which aging impairs the BBB and NVU.
Collapse
Affiliation(s)
- Sara M Zarate
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
| | - Taylor E Huntington
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX, 77843, USA
| | - Pooneh Bagher
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rahul Srinivasan
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, 8447 Riverside Pkwy, Bryan, TX, 77807, USA.
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
13
|
Foßelteder J, Pabst G, Sconocchia T, Schlacher A, Auinger L, Kashofer K, Beham-Schmid C, Trajanoski S, Waskow C, Schöll W, Sill H, Zebisch A, Wölfler A, Thomas D, Reinisch A. Human gene-engineered calreticulin mutant stem cells recapitulate MPN hallmarks and identify targetable vulnerabilities. Leukemia 2023; 37:843-853. [PMID: 36813992 PMCID: PMC10079532 DOI: 10.1038/s41375-023-01848-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023]
Abstract
Calreticulin (CALR) mutations present the main oncogenic drivers in JAK2 wildtype (WT) myeloproliferative neoplasms (MPN), including essential thrombocythemia and myelofibrosis, where mutant (MUT) CALR is increasingly recognized as a suitable mutation-specific drug target. However, our current understanding of its mechanism-of-action is derived from mouse models or immortalized cell lines, where cross-species differences, ectopic over-expression and lack of disease penetrance are hampering translational research. Here, we describe the first human gene-engineered model of CALR MUT MPN using a CRISPR/Cas9 and adeno-associated viral vector-mediated knock-in strategy in primary human hematopoietic stem and progenitor cells (HSPCs) to establish a reproducible and trackable phenotype in vitro and in xenografted mice. Our humanized model recapitulates many disease hallmarks: thrombopoietin-independent megakaryopoiesis, myeloid-lineage skewing, splenomegaly, bone marrow fibrosis, and expansion of megakaryocyte-primed CD41+ progenitors. Strikingly, introduction of CALR mutations enforced early reprogramming of human HSPCs and the induction of an endoplasmic reticulum stress response. The observed compensatory upregulation of chaperones revealed novel mutation-specific vulnerabilities with preferential sensitivity of CALR mutant cells to inhibition of the BiP chaperone and the proteasome. Overall, our humanized model improves purely murine models and provides a readily usable basis for testing of novel therapeutic strategies in a human setting.
Collapse
Affiliation(s)
- Johannes Foßelteder
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Gabriel Pabst
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria.,Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria.,Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Tommaso Sconocchia
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Angelika Schlacher
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Lisa Auinger
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Karl Kashofer
- Diagnostic & Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Medical University of Graz, Graz, Austria
| | - Claudia Waskow
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany.,Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich-Schiller-University, Jena, Germany
| | - Wolfgang Schöll
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Heinz Sill
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Armin Zebisch
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria.,Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Albert Wölfler
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Daniel Thomas
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Andreas Reinisch
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria.
| |
Collapse
|
14
|
Macedo-da-Silva J, Rosa-Fernandes L, Gomes VDM, Santiago VF, Santos DM, Molnar CMS, Barboza BR, de Souza EE, Marques RF, Boscardin SB, Durigon EL, Marinho CRF, Wrenger C, Marie SKN, Palmisano G. Protein Arginylation Is Regulated during SARS-CoV-2 Infection. Viruses 2023; 15:v15020290. [PMID: 36851505 PMCID: PMC9964439 DOI: 10.3390/v15020290] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/09/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND In 2019, the world witnessed the onset of an unprecedented pandemic. By February 2022, the infection by SARS-CoV-2 has already been responsible for the death of more than 5 million people worldwide. Recently, we and other groups discovered that SARS-CoV-2 infection induces ER stress and activation of the unfolded protein response (UPR) pathway. Degradation of misfolded/unfolded proteins is an essential element of proteostasis and occurs mainly in lysosomes or proteasomes. The N-terminal arginylation of proteins is characterized as an inducer of ubiquitination and proteasomal degradation by the N-degron pathway. RESULTS The role of protein arginylation during SARS-CoV-2 infection was elucidated. Protein arginylation was studied in Vero CCL-81, macrophage-like THP1, and Calu-3 cells infected at different times. A reanalysis of in vivo and in vitro public omics data combined with immunoblotting was performed to measure levels of arginyl-tRNA-protein transferase (ATE1) and its substrates. Dysregulation of the N-degron pathway was specifically identified during coronavirus infections compared to other respiratory viruses. We demonstrated that during SARS-CoV-2 infection, there is an increase in ATE1 expression in Calu-3 and Vero CCL-81 cells. On the other hand, infected macrophages showed no enzyme regulation. ATE1 and protein arginylation was variant-dependent, as shown using P1 and P2 viral variants and HEK 293T cells transfection with the spike protein and receptor-binding domains (RBD). In addition, we report that ATE1 inhibitors, tannic acid and merbromine (MER) reduce viral load. This finding was confirmed in ATE1-silenced cells. CONCLUSIONS We demonstrate that ATE1 is increased during SARS-CoV-2 infection and its inhibition has potential therapeutic value.
Collapse
Affiliation(s)
- Janaina Macedo-da-Silva
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, São Paulo 05508-000, Brazil
| | - Livia Rosa-Fernandes
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, São Paulo 05508-000, Brazil
- Laboratory of Experimental Immunoparasitology, Department of Parasitology, ICB, University of São Paulo, São Paulo 05508-000, Brazil
| | - Vinicius de Morais Gomes
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, São Paulo 05508-000, Brazil
| | - Veronica Feijoli Santiago
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, São Paulo 05508-000, Brazil
| | - Deivid Martins Santos
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Bruno Rafael Barboza
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, São Paulo 05508-000, Brazil
| | - Edmarcia Elisa de Souza
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo 05508-000, Brazil
| | - Rodolfo Ferreira Marques
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo 05508-000, Brazil
| | - Silvia Beatriz Boscardin
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo 05508-000, Brazil
| | - Edison Luiz Durigon
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, São Paulo 05508-000, Brazil
| | - Claudio Romero Farias Marinho
- Laboratory of Experimental Immunoparasitology, Department of Parasitology, ICB, University of São Paulo, São Paulo 05508-000, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo 05508-000, Brazil
| | - Suely Kazue Nagahashi Marie
- Laboratory of Molecular and Cellular Biology (LIM 15), Department of Neurology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Giuseppe Palmisano
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, São Paulo 05508-000, Brazil
- School of Natural Sciences, Macquarie University, Sydney 2109, Australia
- Correspondence: or ; Tel.: +55-11-99920-8662
| |
Collapse
|
15
|
Novel Molecular Insights into Leukemic Evolution of Myeloproliferative Neoplasms: A Single Cell Perspective. Int J Mol Sci 2022; 23:ijms232315256. [PMID: 36499582 PMCID: PMC9740017 DOI: 10.3390/ijms232315256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are clonal disorders originated by the serial acquisition of somatic mutations in hematopoietic stem/progenitor cells. The major clinical entities are represented by polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), that are caused by driver mutations affecting JAK2, MPL or CALR. Disease progression is related to molecular and clonal evolution. PV and ET can progress to secondary myelofibrosis (sMF) but can also evolve to secondary acute myeloid leukemia (sAML). PMF is associated with the highest frequency of leukemic transformation, which represents the main cause of death. sAML is associated with a dismal prognosis and clinical features that differ from those of de novo AML. The molecular landscape distinguishes sAML from de novo AML, since the most frequent hits involve TP53, epigenetic regulators, spliceosome modulators or signal transduction genes. Single cell genomic studies provide novel and accurate information about clonal architecture and mutation acquisition order, allowing the reconstruction of clonal dynamics and molecular events that accompany leukemic transformation. In this review, we examine our current understanding of the genomic heterogeneity in MPNs and how it affects disease progression and leukemic transformation. We focus on molecular events elicited by somatic mutations acquisition and discuss the emerging findings coming from single cell studies.
Collapse
|
16
|
Calreticulin mutations affect its chaperone function and perturb the glycoproteome. Cell Rep 2022; 41:111689. [DOI: 10.1016/j.celrep.2022.111689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/17/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022] Open
|
17
|
Ibarra J, Elbanna YA, Kurylowicz K, Ciboddo M, Greenbaum HS, Arellano NS, Rodriguez D, Evers M, Bock-Hughes A, Liu C, Smith Q, Lutze J, Baumeister J, Kalmer M, Olschok K, Nicholson B, Silva D, Maxwell L, Dowgielewicz J, Rumi E, Pietra D, Casetti IC, Catricala S, Koschmieder S, Gurbuxani S, Schneider RK, Oakes SA, Elf SE. Type I but Not Type II Calreticulin Mutations Activate the IRE1α/XBP1 Pathway of the Unfolded Protein Response to Drive Myeloproliferative Neoplasms. Blood Cancer Discov 2022; 3:298-315. [PMID: 35405004 PMCID: PMC9338758 DOI: 10.1158/2643-3230.bcd-21-0144] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/21/2022] [Accepted: 04/08/2022] [Indexed: 01/09/2023] Open
Abstract
Approximately 20% of patients with myeloproliferative neoplasms (MPN) harbor mutations in the gene calreticulin (CALR), with 80% of those mutations classified as either type I or type II. While type II CALR-mutant proteins retain many of the Ca2+ binding sites present in the wild-type protein, type I CALR-mutant proteins lose these residues. The functional consequences of this differential loss of Ca2+ binding sites remain unexplored. Here, we show that the loss of Ca2+ binding residues in the type I mutant CALR protein directly impairs its Ca2+ binding ability, which in turn leads to depleted endoplasmic reticulum (ER) Ca2+ and subsequent activation of the IRE1α/XBP1 pathway of the unfolded protein response. Genetic or pharmacologic inhibition of IRE1α/XBP1 signaling induces cell death in type I mutant but not type II mutant or wild-type CALR-expressing cells, and abrogates type I mutant CALR-driven MPN disease progression in vivo. SIGNIFICANCE Current targeted therapies for CALR-mutated MPNs are not curative and fail to differentiate between type I- versus type II-driven disease. To improve treatment strategies, it is critical to identify CALR mutation type-specific vulnerabilities. Here we show that IRE1α/XBP1 represents a unique, targetable dependency specific to type I CALR-mutated MPNs. This article is highlighted in the In This Issue feature, p. 265.
Collapse
Affiliation(s)
- Juan Ibarra
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Yassmin A. Elbanna
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Katarzyna Kurylowicz
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Michele Ciboddo
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Harrison S. Greenbaum
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Nicole S. Arellano
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Deborah Rodriguez
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Maria Evers
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Althea Bock-Hughes
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Chenyu Liu
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Quinn Smith
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Julian Lutze
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois
| | - Julian Baumeister
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Milena Kalmer
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Kathrin Olschok
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Benjamin Nicholson
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Diane Silva
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Luke Maxwell
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Jonathan Dowgielewicz
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Elisa Rumi
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Daniela Pietra
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | | | - Silvia Catricala
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | | | - Rebekka K. Schneider
- Department of Cell Biology, Institute for Biomedical Technologies, RWTH Aachen University, Aachen, Germany
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Scott A. Oakes
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Shannon E. Elf
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| |
Collapse
|
18
|
Hippo pathway-related genes expression is deregulated in myeloproliferative neoplasms. Med Oncol 2022; 39:97. [DOI: 10.1007/s12032-022-01696-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/22/2022] [Indexed: 10/18/2022]
|
19
|
Genovese E, Mirabile M, Rontauroli S, Sartini S, Fantini S, Tavernari L, Maccaferri M, Guglielmelli P, Bianchi E, Parenti S, Carretta C, Mallia S, Castellano S, Colasante C, Balliu M, Bartalucci N, Palmieri R, Ottone T, Mora B, Potenza L, Passamonti F, Voso MT, Luppi M, Vannucchi AM, Tagliafico E, Manfredini R. The Response to Oxidative Damage Correlates with Driver Mutations and Clinical Outcome in Patients with Myelofibrosis. Antioxidants (Basel) 2022; 11:antiox11010113. [PMID: 35052617 PMCID: PMC8772737 DOI: 10.3390/antiox11010113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/29/2021] [Accepted: 01/01/2022] [Indexed: 12/12/2022] Open
Abstract
Myelofibrosis (MF) is the Philadelphia-negative myeloproliferative neoplasm characterized by the worst prognosis and no response to conventional therapy. Driver mutations in JAK2 and CALR impact on JAK-STAT pathway activation but also on the production of reactive oxygen species (ROS). ROS play a pivotal role in inflammation-induced oxidative damage to cellular components including DNA, therefore leading to greater genomic instability and promoting cell transformation. In order to unveil the role of driver mutations in oxidative stress, we assessed ROS levels in CD34+ hematopoietic stem/progenitor cells of MF patients. Our results demonstrated that ROS production in CD34+ cells from CALR-mutated MF patients is far greater compared with patients harboring JAK2 mutation, and this leads to increased oxidative DNA damage. Moreover, CALR-mutant cells show less superoxide dismutase (SOD) antioxidant activity than JAK2-mutated ones. Here, we show that high plasma levels of total antioxidant capacity (TAC) correlate with detrimental clinical features, such as high levels of lactate dehydrogenase (LDH) and circulating CD34+ cells. Moreover, in JAK2-mutated patients, high plasma level of TAC is also associated with a poor overall survival (OS), and multivariate analysis demonstrated that high TAC classification is an independent prognostic factor allowing the identification of patients with inferior OS in both DIPSS lowest and highest categories. Altogether, our data suggest that a different capability to respond to oxidative stress can be one of the mechanisms underlying disease progression of myelofibrosis.
Collapse
Affiliation(s)
- Elena Genovese
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Margherita Mirabile
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Stefano Sartini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sebastian Fantini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Lara Tavernari
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Monica Maccaferri
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy;
| | - Paola Guglielmelli
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Elisa Bianchi
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sandra Parenti
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Chiara Carretta
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Selene Mallia
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sara Castellano
- Center for Genome Research, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.C.); (E.T.)
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Corrado Colasante
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Manjola Balliu
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Niccolò Bartalucci
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Raffaele Palmieri
- Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy; (R.P.); (T.O.); (M.T.V.)
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy; (R.P.); (T.O.); (M.T.V.)
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, 00179 Rome, Italy
| | - Barbara Mora
- Division of Hematology, Ospedale ASST Sette Laghi, University of Insubria, 21110 Varese, Italy; (B.M.); (F.P.)
| | - Leonardo Potenza
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Francesco Passamonti
- Division of Hematology, Ospedale ASST Sette Laghi, University of Insubria, 21110 Varese, Italy; (B.M.); (F.P.)
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy; (R.P.); (T.O.); (M.T.V.)
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, 00179 Rome, Italy
| | - Mario Luppi
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Alessandro Maria Vannucchi
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.C.); (E.T.)
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Rossella Manfredini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
- Correspondence:
| | | |
Collapse
|
20
|
Bartalucci N, Galluzzi L. Philadelphia-negative myeloproliferative neoplasms: From origins to new perspectives. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 366:ix-xx. [PMID: 35153008 DOI: 10.1016/s1937-6448(22)00019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Affiliation(s)
- Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; DENOThe Excellence Center, University of Florence, Florence, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States; Department of Dermatology, Yale School of Medicine, New Haven, CT, United States; Université de Paris, Paris, France.
| |
Collapse
|
21
|
Calabresi L, Balliu M, Bartalucci N. Immunoblotting-assisted assessment of JAK/STAT and PI3K/Akt/mTOR signaling in myeloproliferative neoplasms CD34+ stem cells. Methods Cell Biol 2022; 171:81-109. [DOI: 10.1016/bs.mcb.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
22
|
Claudiani S, Mason CC, Milojkovic D, Bianchi A, Pellegrini C, Di Marco A, Fiol CR, Robinson M, Ponnusamy K, Mokretar K, Chowdhury A, Albert M, Reid AG, Deininger MW, Naresh K, Apperley JF, Khorashad JS. Carfilzomib Enhances the Suppressive Effect of Ruxolitinib in Myelofibrosis. Cancers (Basel) 2021; 13:cancers13194863. [PMID: 34638347 PMCID: PMC8507927 DOI: 10.3390/cancers13194863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022] Open
Abstract
As the first FDA-approved tyrosine kinase inhibitor for treatment of patients with myelofibrosis (MF), ruxolitinib improves clinical symptoms but does not lead to eradication of the disease or significant reduction of the mutated allele burden. The resistance of MF clones against the suppressive action of ruxolitinib may be due to intrinsic or extrinsic mechanisms leading to activity of additional pro-survival genes or signalling pathways that function independently of JAK2/STAT5. To identify alternative therapeutic targets, we applied a pooled-shRNA library targeting ~5000 genes to a JAK2V617F-positive cell line under a variety of conditions, including absence or presence of ruxolitinib and in the presence of a bone marrow microenvironment-like culture medium. We identified several proteasomal gene family members as essential to HEL cell survival. The importance of these genes was validated in MF cells using the proteasomal inhibitor carfilzomib, which also enhanced lethality in combination with ruxolitinib. We also showed that proteasome gene expression is reduced by ruxolitinib in MF CD34+ cells and that additional targeting of proteasomal activity by carfilzomib enhances the inhibitory action of ruxolitinib in vitro. Hence, this study suggests a potential role for proteasome inhibitors in combination with ruxolitinib for management of MF patients.
Collapse
Affiliation(s)
- Simone Claudiani
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Clinton C. Mason
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT 84108, USA;
| | - Dragana Milojkovic
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Andrea Bianchi
- Department of Information Engineering, University of L’Aquila, 67100 L’Aquila, Italy; (A.B.); (A.D.M.)
| | - Cristina Pellegrini
- Department of Biotechnological and Applied Clinical Science, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Antinisca Di Marco
- Department of Information Engineering, University of L’Aquila, 67100 L’Aquila, Italy; (A.B.); (A.D.M.)
| | - Carme R. Fiol
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Mark Robinson
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Kanagaraju Ponnusamy
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Katya Mokretar
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Avirup Chowdhury
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Michael Albert
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Alistair G. Reid
- Molecular Pathology Unit, Liverpool University, Liverpool L7 8XP, UK;
| | - Michael W. Deininger
- Versiti Blood Research Institute, Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Kikkeri Naresh
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Jane F. Apperley
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Jamshid S. Khorashad
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
- Correspondence:
| |
Collapse
|
23
|
Camargo AC, Remoli B, Portela LM, Fioretto MN, Chuffa LG, Moreno CS, Justulin LA. Transcriptomic landscape of male and female reproductive cancers: Similar pathways and molecular signatures predicting response to endocrine therapy. Mol Cell Endocrinol 2021; 535:111393. [PMID: 34245846 DOI: 10.1016/j.mce.2021.111393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/21/2021] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
Reproductive cancers in both genders represent serious health problems, whose incidence has significantly risen over the past decades. Although considerable differences among reproductive cancers exist, we aimed to identify similar signaling pathways and key molecular oncomarkers shared among six human reproductive cancers that can advance the current knowledge of cancer biology to propose new strategies for more effective therapies. Using a computational analysis approach, here we uncover aberrant miRNAs-mRNAs networks shared in six reproductive tumor types, and identify common molecular mechanisms strictly associated with cancer promotion and aggressiveness. Based on the fact that estrogenic and androgenic signaling pathways were most active in prostate and breast cancers, we further demonstrated that both androgen and estrogen deprivation therapy are capable of regulating the expression of the same key molecular sensors associated with endoplasmic reticulum dysfunction and cell cycle in these cancers. Overall, our data reveal a potential mechanistic framework of cellular processes that are shared among reproductive cancers, and particularly, highlight the importance of hormonal deprivation in breast and prostate cancers and potentially new biomarkers of response to these therapeutic approaches.
Collapse
Affiliation(s)
- Ana Cl Camargo
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, São Paulo, Brazil
| | - Beatriz Remoli
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, São Paulo, Brazil
| | - Luiz Mf Portela
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, São Paulo, Brazil
| | - Mateus N Fioretto
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, São Paulo, Brazil
| | - Luiz Ga Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, São Paulo, Brazil
| | - Carlos S Moreno
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Luis A Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, São Paulo, Brazil.
| |
Collapse
|
24
|
Impact of Calreticulin and Its Mutants on Endoplasmic Reticulum Function in Health and Disease. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021. [PMID: 34050866 DOI: 10.1007/978-3-030-67696-4_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
The endoplasmic reticulum (ER) performs key cellular functions including protein synthesis, lipid metabolism and signaling. While these functions are spatially isolated in structurally distinct regions of the ER, there is cross-talk between the pathways. One vital player that is involved in ER function is the ER-resident protein calreticulin (CALR). It is a calcium ion-dependent lectin chaperone that primarily assists in glycoprotein synthesis in the ER as part of the protein quality control machinery. CALR also buffers calcium ion release and mediates other glycan-independent protein interactions. Mutations in CALR have been reported in a subset of chronic blood tumors called myeloproliferative neoplasms. The mutations consist of insertions or deletions in the CALR gene that all cause a + 1 bp shift in the reading frame and lead to a dramatic alteration of the amino acid sequence of the C-terminal domain of CALR. This alters CALR function and affects cell homeostasis. This chapter will discuss how CALR and mutant CALR affect ER health and disease.
Collapse
|
25
|
Efficient Neuroprotective Rescue of Sacsin-Related Disease Phenotypes in Zebrafish. Int J Mol Sci 2021; 22:ijms22168401. [PMID: 34445111 PMCID: PMC8395086 DOI: 10.3390/ijms22168401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/24/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is a multisystem hereditary ataxia associated with mutations in SACS, which encodes sacsin, a protein of still only partially understood function. Although mouse models of ARSACS mimic largely the disease progression seen in humans, their use in the validation of effective therapies has not yet been proposed. Recently, the teleost Danio rerio has attracted increasing attention as a vertebrate model that allows rapid and economical screening, of candidate molecules, and thus combines the advantages of whole-organism phenotypic assays and in vitro high-throughput screening assays. Through CRISPR/Cas9-based mutagenesis, we generated and characterized a zebrafish sacs-null mutant line that replicates the main features of ARSACS. The sacs-null fish showed motor impairment, hindbrain atrophy, mitochondrial dysfunction, and reactive oxygen species accumulation. As proof of principle for using these mutant fish in high-throughput screening studies, we showed that both acetyl-DL-leucine and tauroursodeoxycholic acid improved locomotor and biochemical phenotypes in sacs−/− larvae treated with these neuroprotective agents, by mediating significant rescue of the molecular functions altered by sacsin loss. Taken together, the evidence here reported shows the zebrafish to be a valuable model organism for the identification of novel molecular mechanisms and for efficient and rapid in vivo optimization and screening of potential therapeutic compounds. These findings may pave the way for new interventions targeting the earliest phases of Purkinje cell degeneration in ARSACS.
Collapse
|
26
|
Shide K. Calreticulin mutations in myeloproliferative neoplasms. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 365:179-226. [PMID: 34756244 DOI: 10.1016/bs.ircmb.2021.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Calreticulin (CALR) is a chaperone present in the endoplasmic reticulum, which is involved in the quality control of N-glycosylated proteins and storage of calcium ions. In 2013, the C-terminal mutation in CALR was identified in half of the patients with essential thrombocythemia and primary myelofibrosis who did not have a JAK2 or MPL mutation. The results of 8 years of intensive research are changing the clinical practice associated with treating myeloproliferative neoplasms (MPNs). The presence or absence of CALR mutations and their mutation types already provide important information for diagnosis and treatment decision making. In addition, the interaction with the thrombopoietin receptor MPL, which is the main mechanism of transformation by CALR mutation, and the expression of the mutant protein on the cell surface have a great potential as targets for molecular-targeted drugs and immunotherapy. This chapter presents recent findings on the clinical significance of the CALR mutation and the molecular basis by which this mutation drives MPNs.
Collapse
Affiliation(s)
- Kotaro Shide
- Division of Haematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
| |
Collapse
|
27
|
Immunomodulatory effects of different intravenous immunoglobulin preparations in chronic lymphocytic leukemia. Sci Rep 2021; 11:12926. [PMID: 34155276 PMCID: PMC8217488 DOI: 10.1038/s41598-021-92412-8] [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: 11/13/2020] [Accepted: 06/10/2021] [Indexed: 11/08/2022] Open
Abstract
Hypogammaglobulinemia is the most frequently observed immune defect in chronic lymphocytic leukemia (CLL). Although CLL patients usually have low serum levels of all isotypes (IgG, IgM and IgA), standard immunoglobulin (Ig) preparations for replacement therapy administrated to these patients contain more than 95% of IgG. Pentaglobin is an Ig preparation of intravenous application (IVIg) enriched with IgM and IgA (IVIgGMA), with the potential benefit to restore the Ig levels of all isotypes. Because IVIg preparations at high doses have well-documented anti-inflammatory and immunomodulatory effects, we aimed to evaluate the capacity of Pentaglobin and a standard IVIg preparation to affect leukemic and T cells from CLL patients. In contrast to standard IVIg, we found that IVIgGMA did not modify T cell activation and had a lower inhibitory effect on T cell proliferation. Regarding the activation of leukemic B cells through BCR, it was similarly reduced by both IVIgGMA and IVIgG. None of these IVIg preparations modified spontaneous apoptosis of T or leukemic B cells. However, the addition of IVIgGMA on in vitro cultures decreased the apoptosis of T cells induced by the BCL-2 inhibitor, venetoclax. Importantly, IVIgGMA did not impair venetoclax-induced apoptosis of leukemic B cells. Overall, our results add new data on the effects of different preparations of IVIg in CLL, and show that the IgM/IgA enriched preparation not only affects relevant mechanisms involved in CLL pathogenesis but also has a particular profile of immunomodulatory effects on T cells that deserves further investigation.
Collapse
|
28
|
Guijarro-Hernández A, Vizmanos JL. A Broad Overview of Signaling in Ph-Negative Classic Myeloproliferative Neoplasms. Cancers (Basel) 2021; 13:cancers13050984. [PMID: 33652860 PMCID: PMC7956519 DOI: 10.3390/cancers13050984] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary There is growing evidence that Ph-negative myeloproliferative neoplasms are disorders in which multiple signaling pathways are significantly disturbed. The heterogeneous phenotypes observed among patients have highlighted the importance of having a comprehensive knowledge of the molecular mechanisms behind these diseases. This review aims to show a broad overview of the signaling involved in myeloproliferative neoplasms (MPNs) and other processes that can modify them, which could be helpful to better understand these diseases and develop more effective targeted treatments. Abstract Ph-negative myeloproliferative neoplasms (polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF)) are infrequent blood cancers characterized by signaling aberrations. Shortly after the discovery of the somatic mutations in JAK2, MPL, and CALR that cause these diseases, researchers extensively studied the aberrant functions of their mutant products. In all three cases, the main pathogenic mechanism appears to be the constitutive activation of JAK2/STAT signaling and JAK2-related pathways (MAPK/ERK, PI3K/AKT). However, some other non-canonical aberrant mechanisms derived from mutant JAK2 and CALR have also been described. Moreover, additional somatic mutations have been identified in other genes that affect epigenetic regulation, tumor suppression, transcription regulation, splicing and other signaling pathways, leading to the modification of some disease features and adding a layer of complexity to their molecular pathogenesis. All of these factors have highlighted the wide variety of cellular processes and pathways involved in the pathogenesis of MPNs. This review presents an overview of the complex signaling behind these diseases which could explain, at least in part, their phenotypic heterogeneity.
Collapse
Affiliation(s)
- Ana Guijarro-Hernández
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
| | - José Luis Vizmanos
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Correspondence:
| |
Collapse
|
29
|
Carretta C, Mallia S, Genovese E, Parenti S, Rontauroli S, Bianchi E, Fantini S, Sartini S, Tavernari L, Tagliafico E, Manfredini R. Genomic Analysis of Hematopoietic Stem Cell at the Single-Cell Level: Optimization of Cell Fixation and Whole Genome Amplification (WGA) Protocol. Int J Mol Sci 2020; 21:E7366. [PMID: 33036143 PMCID: PMC7582552 DOI: 10.3390/ijms21197366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 12/15/2022] Open
Abstract
Single-cell genomics has become the method of choice for the study of heterogeneous cell populations and represents an elective application in defining the architecture and clonal evolution in hematological neoplasms. Reconstructing the clonal evolution of a neoplastic population therefore represents the main way to understand more deeply the pathogenesis of the neoplasm, but it is also a potential tool to understand the evolution of the tumor population with respect to its response to therapy. Pre-analytical phase for single-cell genomics analysis is crucial to obtain a cell population suitable for single-cell sorting, and whole genome amplification is required to obtain the necessary amount of DNA from a single cell in order to proceed with sequencing. Here, we evaluated the impact of different methods of cellular immunostaining, fixation and whole genome amplification on the efficiency and yield of single-cell sequencing.
Collapse
Affiliation(s)
- Chiara Carretta
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Selene Mallia
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Elena Genovese
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Sandra Parenti
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Elisa Bianchi
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Sebastian Fantini
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Stefano Sartini
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Lara Tavernari
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, 41125 Modena, Italy;
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| |
Collapse
|
30
|
Abstract
Calreticulin (CALR) is an endoplasmic reticulum (ER)-resident protein involved in a spectrum of cellular processes. In healthy cells, CALR operates as a chaperone and Ca2+ buffer to assist correct protein folding within the ER. Besides favoring the maintenance of cellular proteostasis, these cell-intrinsic CALR functions support Ca2+-dependent processes, such as adhesion and integrin signaling, and ensure normal antigen presentation on MHC Class I molecules. Moreover, cancer cells succumbing to immunogenic cell death (ICD) expose CALR on their surface, which promotes the uptake of cell corpses by professional phagocytes and ultimately supports the initiation of anticancer immunity. Thus, loss-of-function CALR mutations promote oncogenesis not only as they impair cellular homeostasis in healthy cells, but also as they compromise natural and therapy-driven immunosurveillance. However, the prognostic impact of total or membrane-exposed CALR levels appears to vary considerably with cancer type. For instance, while genetic CALR defects promote pre-neoplastic myeloproliferation, patients with myeloproliferative neoplasms bearing CALR mutations often experience improved overall survival as compared to patients bearing wild-type CALR. Here, we discuss the context-dependent impact of CALR on malignant transformation, tumor progression and response to cancer therapy.
Collapse
|
31
|
Edahiro Y, Araki M, Komatsu N. Mechanism underlying the development of myeloproliferative neoplasms through mutant calreticulin. Cancer Sci 2020; 111:2682-2688. [PMID: 32462673 PMCID: PMC7419020 DOI: 10.1111/cas.14503] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/12/2020] [Accepted: 05/22/2020] [Indexed: 01/14/2023] Open
Abstract
Deregulation of cytokine signaling is frequently associated with various pathological conditions, including malignancies. In patients with myeloproliferative neoplasms (MPNs), recurrent somatic mutations in the calreticulin (CALR) gene, which encodes a molecular chaperone that resides in the endoplasmic reticulum, have been reported. Studies have defined mutant CALR as an oncogene promoting the development of MPN, and deciphered a novel molecular mechanism by which mutant CALR constitutively activates thrombopoietin receptor MPL and its downstream molecules to induce cellular transformation. The mechanism of interaction and activation of MPL by mutant CALR is unique, not only due to the latter forming a homomultimeric complex through a novel mutant‐specific sequence generated by frameshift mutation, but also for its ability to interact with immature asparagine‐linked glycan for eventual engagement with immature MPL in the endoplasmic reticulum. The complex formed between mutant CALR and MPL is then transported to the cell surface, where it induces constitutive activation of downstream kinase JAK2 bound to MPL. Refined structural and cell biological studies can provide an in‐depth understanding of this unusual mechanism of receptor activation by a mutant molecular chaperone. Mutant CALR is also involved in modulation of the immune response, transcription, and intracellular homeostasis, which could contribute to the development of MPN. In the present article, we comprehensively review the current understanding of the underlying molecular mechanisms for mutant molecular chaperone‐induced cellular transformation.
Collapse
Affiliation(s)
- Yoko Edahiro
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Marito Araki
- Department of Transfusion Medicine and Stem Cell Regulation, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
32
|
The Expression of Myeloproliferative Neoplasm-Associated Calreticulin Variants Depends on the Functionality of ER-Associated Degradation. Cancers (Basel) 2019; 11:cancers11121921. [PMID: 31810292 PMCID: PMC6966542 DOI: 10.3390/cancers11121921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Mutations in CALR observed in myeloproliferative neoplasms (MPN) were recently shown to be pathogenic via their interaction with MPL and the subsequent activation of the Janus Kinase – Signal Transducer and Activator of Transcription (JAK-STAT) pathway. However, little is known on the impact of those variant CALR proteins on endoplasmic reticulum (ER) homeostasis. Methods: The impact of the expression of Wild Type (WT) or mutant CALR on ER homeostasis was assessed by quantifying the expression level of Unfolded Protein Response (UPR) target genes, splicing of X-box Binding Protein 1 (XBP1), and the expression level of endogenous lectins. Pharmacological and molecular (siRNA) screens were used to identify mechanisms involved in CALR mutant proteins degradation. Coimmunoprecipitations were performed to define more precisely actors involved in CALR proteins disposal. Results: We showed that the expression of CALR mutants alters neither ER homeostasis nor the sensitivity of hematopoietic cells towards ER stress-induced apoptosis. In contrast, the expression of CALR variants is generally low because of a combination of secretion and protein degradation mechanisms mostly mediated through the ER-Associated Degradation (ERAD)-proteasome pathway. Moreover, we identified a specific ERAD network involved in the degradation of CALR variants. Conclusions: We propose that this ERAD network could be considered as a potential therapeutic target for selectively inhibiting CALR mutant-dependent proliferation associated with MPN, and therefore attenuate the associated pathogenic outcomes.
Collapse
|