1
|
de Castro Sampaio SS, Ramalho MCC, de Souza CS, de Almeida Rodrigues B, de Mendonça GRS, Lazarini M. RHO subfamily of small GTPases in the development and function of hematopoietic cells. J Cell Physiol 2025; 240:e31469. [PMID: 39434451 DOI: 10.1002/jcp.31469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/16/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024]
Abstract
RHOA, RHOB, and RHOC comprise a subfamily of RHO GTPase proteins famed for controlling cytoskeletal dynamics. RHO proteins operate downstream of multiple signals emerging from the microenvironment, leading to diverse cell responses, such as proliferation, adhesion, and migration. Therefore, RHO signaling has been centrally placed in the regulation of blood cells. Despite their high homology, unique roles of RHOA, RHOB, and RHOC have been described in hematopoietic cells. In this article, we overview the contribution of RHO proteins in the development and function of each blood cell lineage. Additionally, we highlight the aberrations of the RHO signaling pathways found in hematological malignancies, providing clues for the identification of new therapeutic targets.
Collapse
Affiliation(s)
| | | | - Caroline Santos de Souza
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Mariana Lazarini
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Pozzi G, Presta V, Masselli E, Condello G, Cortellazzi S, Arcari ML, Micheloni C, Vitale M, Gobbi G, Mirandola P, Carubbi C. Interplay between Protein Kinase C Epsilon and Reactive Oxygen Species during Myogenic Differentiation. Cells 2023; 12:1792. [PMID: 37443826 PMCID: PMC10340168 DOI: 10.3390/cells12131792] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Reactive oxygen species (ROS) are currently recognized as a key driver of several physiological processes. Increasing evidence indicates that ROS levels can affect myogenic differentiation, but the molecular mechanisms still need to be elucidated. Protein kinase C (PKC) epsilon (PKCe) promotes muscle stem cell differentiation and regeneration of skeletal muscle after injury. PKCs play a tissue-specific role in redox biology, with specific isoforms being both a target of ROS and an up-stream regulator of ROS production. Therefore, we hypothesized that PKCe represents a molecular link between redox homeostasis and myogenic differentiation. We used an in vitro model of a mouse myoblast cell line (C2C12) to study the PKC-redox axis. We demonstrated that the transition from a myoblast to myotube is typified by increased PKCe protein content and decreased ROS. Intriguingly, the expression of the antioxidant enzyme superoxide dismutase 2 (SOD2) is significantly higher in the late phases of myogenic differentiation, mimicking PKCe protein content. Furthermore, we demonstrated that PKCe inhibition increases ROS and reduces SOD2 protein content while SOD2 silencing did not affect PKCe protein content, suggesting that the kinase could be an up-stream regulator of SOD2. To support this hypothesis, we found that in C2C12 cells, PKCe interacts with Nrf2, whose activation induces SOD2 transcription. Overall, our results indicate that PKCe is capable of activating the antioxidant signaling preventing ROS accumulation in a myotube, eventually promoting myogenic differentiation.
Collapse
Affiliation(s)
- Giulia Pozzi
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
| | - Valentina Presta
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
| | - Elena Masselli
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
| | - Giancarlo Condello
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
| | - Samuele Cortellazzi
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
| | - Maria Luisa Arcari
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
| | - Cristina Micheloni
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
| | - Marco Vitale
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
- Italian Foundation for Research in Balneotherapy (FoRST), 00198 Rome, Italy
| | - Giuliana Gobbi
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
| | - Prisco Mirandola
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
| | - Cecilia Carubbi
- Department of Medicine and Surgery (DiMeC), University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (G.P.); (V.P.); (E.M.); (G.C.); (M.L.A.); (C.M.); (M.V.); (C.C.)
| |
Collapse
|
3
|
Wang Q, Wei J, Jia X, Feng X, Ji Z, Ji X, Shao X. Downregulation of ADAM17 in pediatric immune thrombocytopenia impairs proplatelet formation. BMC Pediatr 2022; 22:164. [PMID: 35354403 PMCID: PMC8966352 DOI: 10.1186/s12887-022-03237-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Immune thrombocytopenia (ITP) is the most common etiology of acquired thrombocytopenia diseases in children. ITP is characterized by the immune-mediated decreased formation and excessive destruction of platelets. The pathogenesis and management of pediatric ITP are distinct from adult ITP. A disintegrin and metalloproteinase 17 (ADAM17) mediates the shedding of platelet receptor glycoprotein Ib α (GPIb α) in extracellular domain, functioning in the platelet activation and clearance. Our study aims to probe the roles and mechanisms of ADAM17 in pediatric ITP. METHODS The differently expressed ADAM17 in megakaryocytes was obtained from children with ITP through the next-generation RNA-Sequence. Hematoxylin-eosin and Giemsa staining were performed for cell morphology identification. Flow cytometry was applied to assess autoantibodies against platelets, subtypes of lymphocytes, the surface expression level of ADAM17 and polyploidization of megakaryocytes, as well as the full-length GP Ib α. RESULTS ADAM17 was significantly downregulated in megakaryocytes and platelets in children with ITP. Higher values of PDW and positive autoantibodies presence were observed in children with ITP. Loss of ADAM17 in mice led to defects in proplatelet formation and significantly elevated expression of phosphorylated myosin light chain (p-MLC) in megakaryocytes. CONCLUSIONS Our study indicated that the downregulation of ADAM17 might be an innate cause of inefficient platelet production in pediatric ITP.
Collapse
Affiliation(s)
- Qi Wang
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu Province, China.
| | - Jia Wei
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu Province, China
| | - Xi Jia
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu Province, China
| | - Xiao Feng
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu Province, China
| | - Zhenghua Ji
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu Province, China
| | - Xueqiang Ji
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu Province, China
| | - Xuejun Shao
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu Province, China.
| |
Collapse
|
4
|
Vainchenker W, Arkoun B, Basso-Valentina F, Lordier L, Debili N, Raslova H. Role of Rho-GTPases in megakaryopoiesis. Small GTPases 2021; 12:399-415. [PMID: 33570449 PMCID: PMC8583283 DOI: 10.1080/21541248.2021.1885134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Megakaryocytes (MKs) are the bone marrow (BM) cells that generate blood platelets by a process that requires: i) polyploidization responsible for the increased MK size and ii) cytoplasmic organization leading to extension of long pseudopods, called proplatelets, through the endothelial barrier to allow platelet release into blood. Low level of localized RHOA activation prevents actomyosin accumulation at the cleavage furrow and participates in MK polyploidization. In the platelet production, RHOA and CDC42 play opposite, but complementary roles. RHOA inhibits both proplatelet formation and MK exit from BM, whereas CDC42 drives the development of the demarcation membranes and MK migration in BM. Moreover, the RhoA or Cdc42 MK specific knock-out in mice and the genetic alterations in their down-stream effectors in human induce a thrombocytopenia demonstrating their key roles in platelet production. A better knowledge of Rho-GTPase signalling is thus necessary to develop therapies for diseases associated with platelet production defects. Abbreviations: AKT: Protein Kinase BARHGEF2: Rho/Rac Guanine Nucleotide Exchange Factor 2ARP2/3: Actin related protein 2/3BM: Bone marrowCDC42: Cell division control protein 42 homologCFU-MK: Colony-forming-unit megakaryocyteCIP4: Cdc42-interacting protein 4mDIA: DiaphanousDIAPH1; Protein diaphanous homolog 1ECT2: Epithelial Cell Transforming Sequence 2FLNA: Filamin AGAP: GTPase-activating proteins or GTPase-accelerating proteinsGDI: GDP Dissociation InhibitorGEF: Guanine nucleotide exchange factorHDAC: Histone deacetylaseLIMK: LIM KinaseMAL: Megakaryoblastic leukaemiaMARCKS: Myristoylated alanine-rich C-kinase substrateMKL: Megakaryoblastic leukaemiaMLC: Myosin light chainMRTF: Myocardin Related Transcription FactorOTT: One-Twenty Two ProteinPACSIN2: Protein Kinase C And Casein Kinase Substrate In Neurons 2PAK: P21-Activated KinasePDK: Pyruvate Dehydrogenase kinasePI3K: Phosphoinositide 3-kinasePKC: Protein kinase CPTPRJ: Protein tyrosine phosphatase receptor type JRAC: Ras-related C3 botulinum toxin substrate 1RBM15: RNA Binding Motif Protein 15RHO: Ras homologousROCK: Rho-associated protein kinaseSCAR: Suppressor of cAMP receptorSRF: Serum response factorSRC: SarcTAZ: Transcriptional coactivator with PDZ motifTUBB1: Tubulin β1VEGF: Vascular endothelial growth factorWAS: Wiskott Aldrich syndromeWASP: Wiskott Aldrich syndrome proteinWAVE: WASP-family verprolin-homologous proteinWIP: WASP-interacting proteinYAP: Yes-associated protein
Collapse
Affiliation(s)
- William Vainchenker
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France.,GrEX, Sorbonne Paris Cité, Paris, France
| | - Brahim Arkoun
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France.,GrEX, Sorbonne Paris Cité, Paris, France
| | - Francesca Basso-Valentina
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France.,Université Sorbonne Paris Cité/Université Paris Dideront, Paris, France
| | - Larissa Lordier
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France
| | - Najet Debili
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France
| | - Hana Raslova
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France
| |
Collapse
|
5
|
Egot M, Lasne D, Poirault-Chassac S, Mirault T, Pidard D, Dreano E, Elie C, Gandrille S, Marchelli A, Baruch D, Rendu J, Fauré J, Flaujac C, Gratacap MP, Sié P, Gaussem P, Salomon R, Baujat G, Bachelot-Loza C. Role of oculocerebrorenal syndrome of Lowe (OCRL) protein in megakaryocyte maturation, platelet production and functions: a study in patients with Lowe syndrome. Br J Haematol 2021; 192:909-921. [PMID: 33528045 DOI: 10.1111/bjh.17346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/16/2020] [Accepted: 01/03/2021] [Indexed: 11/29/2022]
Abstract
Lowe syndrome (LS) is an oculocerebrorenal syndrome of Lowe (OCRL1) genetic disorder resulting in a defect of the OCRL protein, a phosphatidylinositol-4,5-bisphosphate 5-phosphatase containing various domains including a Rho GTPase-activating protein (RhoGAP) homology domain catalytically inactive. We previously reported surgery-associated bleeding in patients with LS, suggestive of platelet dysfunction, accompanied with a mild thrombocytopenia in several patients. To decipher the role of OCRL in platelet functions and in megakaryocyte (MK) maturation, we conducted a case-control study on 15 patients with LS (NCT01314560). While all had a drastically reduced expression of OCRL, this deficiency did not affect platelet aggregability, but resulted in delayed thrombus formation on collagen under flow conditions, defective platelet spreading on fibrinogen and impaired clot retraction. We evidenced alterations of the myosin light chain phosphorylation (P-MLC), with defective Rac1 activity and, inversely, elevated active RhoA. Altered cytoskeleton dynamics was also observed in cultured patient MKs showing deficient proplatelet extension with increased P-MLC that was confirmed using control MKs transfected with OCRL-specific small interfering(si)RNA (siOCRL). Patients with LS also had an increased proportion of circulating barbell-shaped proplatelets. Our present study establishes that a deficiency of the OCRL protein results in a defective actomyosin cytoskeleton reorganisation in both MKs and platelets, altering both thrombopoiesis and some platelet responses to activation necessary to ensure haemostasis.
Collapse
Affiliation(s)
- Marion Egot
- Université de Paris, Innovations Thérapeutiques en Hémostase, Paris, INSERM U1140, France
| | - Dominique Lasne
- Université de Paris, Innovations Thérapeutiques en Hémostase, Paris, INSERM U1140, France.,AP-HP, Laboratoire d'Hématologie, Hôpital Necker-Enfants Malades, Paris, France
| | - Sonia Poirault-Chassac
- Université de Paris, Innovations Thérapeutiques en Hémostase, Paris, INSERM U1140, France
| | - Tristan Mirault
- Université de Paris, Innovations Thérapeutiques en Hémostase, Paris, INSERM U1140, France.,AP-HP, Service de Médecine Vasculaire, Hôpital Européen Georges-Pompidou, Paris, France
| | - Dominique Pidard
- Université de Paris, Innovations Thérapeutiques en Hémostase, Paris, INSERM U1140, France
| | - Elise Dreano
- Université de Paris, Innovations Thérapeutiques en Hémostase, Paris, INSERM U1140, France
| | - Caroline Elie
- AP-HP, Unité de Recherche Clinique, Hôpital Necker-Enfants Malades, Paris, France
| | - Sophie Gandrille
- Université de Paris, Innovations Thérapeutiques en Hémostase, Paris, INSERM U1140, France
| | - Aurore Marchelli
- Université de Paris, Innovations Thérapeutiques en Hémostase, Paris, INSERM U1140, France
| | - Dominique Baruch
- Université de Paris, Innovations Thérapeutiques en Hémostase, Paris, INSERM U1140, France
| | - John Rendu
- University Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble, France
| | - Julien Fauré
- University Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble, France
| | - Claire Flaujac
- Centre hospitalier de Versailles, André Mignot, Service de Biologie Médicale, Secteur Hémostase, Le Chesnay, France
| | - Marie-Pierre Gratacap
- INSERM U1048 and Université Toulouse 3, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), CHU-Rangueil, Toulouse, France
| | - Pierre Sié
- INSERM U1048 and Université Toulouse 3, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), CHU-Rangueil, Toulouse, France.,CHU de Toulouse, Laboratoire d'Hématologie, Toulouse, France
| | - Pascale Gaussem
- Université de Paris, Innovations Thérapeutiques en Hémostase, Paris, INSERM U1140, France.,AP-HP, Service d'Hématologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| | - Rémi Salomon
- AP-HP, Service de Néphrologie Pédiatrique, Hôpital Necker-Enfants Malades, INSERM U983, Paris, France
| | - Geneviève Baujat
- AP-HP, Service de Génétique, Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
| | | |
Collapse
|
6
|
Nakamura S, Sugimoto N, Eto K. Development of platelet replacement therapy using human induced pluripotent stem cells. Dev Growth Differ 2021; 63:178-186. [PMID: 33507533 PMCID: PMC8048793 DOI: 10.1111/dgd.12711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 12/13/2022]
Abstract
In the body, platelets mainly work as a hemostatic agent, and the lack of platelets can cause serious bleeding. Induced pluripotent stem (iPS) cells potentially allow for a stable supply of platelets that are independent of donors and eliminate the risk of infection. However, a major challenge in iPS cell-based systems is producing the number of platelets required for a single transfusion (more than 200 billion in Japan). Thus, development in large-scale culturing technology is required. In previous studies, we generated a self-renewable, immortalized megakaryocyte cell line by transfecting iPS cell-derived hematopoietic progenitor cells with c-MYC, BMI1, and BCL-XL genes. Optimization of the culture conditions, including the discovery of a novel fluid-physical factor, turbulence, in the production of platelets in vivo, and the development of bioreactors that apply turbulence have enabled us to generate platelets of clinical quality and quantity. We have further generated platelets deleted of HLA class I expression by using genetic modification technology for patients suffering from alloimmune transfusion refractoriness, since these patients are underserved by current blood donation systems. In this review, we highlight current research and our recent work on iPS cell-derived platelet induction.
Collapse
Affiliation(s)
- Sou Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Naoshi Sugimoto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
7
|
Generation and manipulation of human iPSC-derived platelets. Cell Mol Life Sci 2021; 78:3385-3401. [PMID: 33439272 PMCID: PMC7804213 DOI: 10.1007/s00018-020-03749-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/01/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022]
Abstract
The discovery of iPSCs has led to the ex vivo production of differentiated cells for regenerative medicine. In the case of transfusion products, the derivation of platelets from iPSCs is expected to complement our current blood-donor supplied transfusion system through donor-independent production with complete pathogen-free assurance. This derivation can also overcome alloimmune platelet transfusion refractoriness by resulting in autologous, HLA-homologous or HLA-deficient products. Several developments were necessary to produce a massive number of platelets required for a single transfusion. First, expandable megakaryocytes were established from iPSCs through transgene expression. Second, a turbulent-type bioreactor with improved platelet yield and quality was developed. Third, novel drugs that enabled efficient feeder cell-free conditions were developed. Fourth, the platelet-containing suspension was purified and resuspended in an appropriate buffer. Finally, the platelet product needed to be assured for competency and safety including non-tumorigenicity through in vitro and in vivo preclinical tests. Based on these advancements, a clinical trial has started. The generation of human iPSC-derived platelets could evolve transfusion medicine to the next stage and assure a ubiquitous, safe supply of platelet products. Further, considering the feasibility of gene manipulations in iPSCs, other platelet products may bring forth novel therapeutic measures.
Collapse
|
8
|
The PKC universe keeps expanding: From cancer initiation to metastasis. Adv Biol Regul 2020; 78:100755. [PMID: 33017725 DOI: 10.1016/j.jbior.2020.100755] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 02/08/2023]
Abstract
Classical and novel protein kinase C (PKC) isozymes (c/nPKCs), members of the PKC family that become activated by the lipid second messenger diacylglycerol (DAG) and phorbol esters, exert a myriad of cellular effects that impact proliferative and motile cellular responses. While c/nPKCs have been indisputably associated with tumor promotion, their roles exceed by far their sole involvement as promoter kinases. Indeed, this original dogma has been subsequently redefined by the introduction of several new concepts: the identification of tumor suppressing roles for c/nPKCs, and their participation in early and late stages of carcinogenesis. This review dives deep into the intricate roles of c/nPKCs in cancer initiation as well as in the different stages of the metastatic cascade, with great emphasis in their involvement in cancer cell motility via regulation of small Rho GTPases, the production of extracellular matrix (ECM)-degrading proteases, and the epithelial-to-mesenchymal transition (EMT) program required for the acquisition of highly invasive traits. Here, we highlight functional interplays between either PKCα or PKCε and mesenchymal features that may ultimately contribute to anticancer drug resistance in cellular and animal models. We also introduce the novel hypothesis that c/nPKCs may be implicated in the control of immune evasion through the regulation of immune checkpoint protein expression. In summary, dissecting the colossal complexity of c/nPKC signaling in the wide spectrum of cancer progression may bring new opportunities for the development of meaningful tools aiding for cancer prognosis and therapy.
Collapse
|
9
|
Kong B, Lv ZD, Xia J, Jin LY, Yang ZC. DLC-3 suppresses cellular proliferation, migration, and invasion in triple-negative breast cancer by the Wnt/β-catenin pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1224-1232. [PMID: 31933937 PMCID: PMC6947060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/22/2019] [Indexed: 06/10/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype. Our study investigated the functional role of DLC-3 in TNBC. The expression of DLC-3 was assessed by immunohistochemistry in TNBC to evaluate the clinicopathologic significance of DLC-3. Recombinant lentiviral vectors encoding the DLC-3 gene were constructed for transfection into MDA-MB-231. Real-time qPCR and western blot analysis were employed to evaluate the expression of DLC-3, β-catenin, GSK-3β and c-myc in DLC-3-transfected cells. Moreover, cell proliferation assays, cell colony formation assays, and cell migration and invasion assays were performed to elucidate the role of DLC-3 in TNBC development and progression. Our data revealed that DLC-3 was downregulated in TNBC, and its expression level was associated with lymph node status and differentiation grade in breast cancer. Both real-time qPCR and western blot analyses showed that the DLC-3 gene and protein were overexpressed in the DLC-3-transfected MDA-MB-231 cells. In addition, the expression of GSK-3β was upregulated and the expression of β-catenin and c-myc gene was downregulated in the DLC-3-transfected cells. Furthermore, DLC-3 overexpression inhibited cell proliferation, colony formation, migration, and invasion in vitro. DLC-3, functioning as a tumor-suppressor gene, inhibits cell growth and invasion in TNBC, possibly through regulation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Bin Kong
- Breast Center, The Affiliated Hospital of Qingdao UniversityQingdao 266003, P. R. China
| | - Zhi-Dong Lv
- Breast Center, The Affiliated Hospital of Qingdao UniversityQingdao 266003, P. R. China
| | - Jing Xia
- Breast Center, The Affiliated Hospital of Qingdao UniversityQingdao 266003, P. R. China
| | - Li-Ying Jin
- Cerebrovascular Disease Research Institute, The Affiliated Hospital of Qingdao UniversityQingdao 266003, P. R. China
| | - Zhao-Chuan Yang
- Department of Child Health Care, The Affiliated Hospital of Qingdao UniversityQingdao 266003, P. R. China
| |
Collapse
|
10
|
Distinctive requirement of PKCε in the control of Rho GTPases in epithelial and mesenchymally transformed lung cancer cells. Oncogene 2019; 38:5396-5412. [PMID: 30923343 PMCID: PMC6609469 DOI: 10.1038/s41388-019-0796-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/17/2022]
Abstract
Diacylglycerol (DAG)/phorbol ester-regulated protein kinase C (PKC) isozymes have been widely linked to tumor promotion and the development of a metastatic phenotype. PKCε, an oncogenic member of the PKC family, is abnormally overexpressed in lung cancer and other cancer types. This kinase plays significant roles in proliferation, survival and migration; however its role in epithelial-to-mesenchymal transition (EMT) has been scarcely studied. Silencing experiments in non-small lung cancer (NSCLC) cells revealed that PKCε or other DAG-regulated PKCs (PKCα and PKCδ) were dispensable for the acquisition of a mesenchymal phenotype induced by transforming growth factor beta (TGF-β). Unexpectedly, we found a nearly complete down-regulation of PKCε expression in TGF-β-mesenchymally transformed NSCLC cells. PMA and AJH-836 (a DAG-mimetic that preferentially activates PKCε) promote ruffle formation in NSCLC cells via Rac1, however they fail to induce these morphological changes in TGF-β-mesenchymally transformed cells despite their elevated Rac1 activity. Several Rac Guanine nucleotide Exchange-Factors (Rac-GEFs) were also up-regulated in TGF-β-treated NSCLC cells, including Trio and Tiam2, which were required for cell motility. Lastly, we found that silencing or inhibiting PKCε enhances RhoA activity and stress fiber formation, a phenotype also observed in TGF-β-transformed cells. Our studies established a distinctive involvement of PKCε in epithelial and mesenchymal NSCLC cells, and identified a complex interplay between PKCε and small GTPases that contributes to regulation of NSCLC cell morphology and motile activity.
Collapse
|
11
|
Dhenge A, Kuhikar R, Kale V, Limaye L. Regulation of differentiation of MEG01 to megakaryocytes and platelet-like particles by Valproic acid through Notch3 mediated actin polymerization. Platelets 2018; 30:780-795. [PMID: 30332548 DOI: 10.1080/09537104.2018.1528344] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Valproic acid (VPA) is one of the HDAC inhibitors used for the treatment of neurological disorders and hematological malignancies. Its role in self-renewal and proliferation of hematopoietic stem cells (HSCs) is well studied, but little is known about its involvement in regulating megakaryopoiesis and thrombopoiesis. In this study, we evaluated the role of VPA in megakaryopoiesis by using MEG-01, a megakaryoblast cell line. Our results show that VPA treatment differentiates MEG-01 cells to megakaryocytes (MK) and platelet-like particles. It was confirmed by augmented expression of MK and PLT-specific markers, higher ploidy, and PLT functionality. We assessed the molecular events underlying megakaryopoiesis. In the present study, we found an upregulation of Notch3 and its downstream target PDGFR-β upon VPA treatment. The direct role of Notch3 in megakaryopoiesis has not yet been studied. PDGFR-β is known to control actin organization during vascular smooth muscle cell differentiation. The actin cytoskeleton plays important role during proplatelet and PLT formation. We found an upregulation of Rac/Cdc42 GTPase and its downstream effectors that are the key players during actin polymerization events. We speculate that VPA induces PLT formation through Notch-3 signaling that in turn modulates actin polymerization that is one of the crucial steps necessary for thrombopoiesis. These studies were also confirmed with knockdown of Notch3 in MEG01 by using ShRNA approach as well as with apheresis-derived CD34+ cells. Altogether, these findings provide an evidence for a novel role of Notch3 in regulating platelet formation.
Collapse
Affiliation(s)
- Ankita Dhenge
- a National Centre for Cell Science, NCCS Complex , Savitribai Phule Pune University Campus , Pune , India
| | - Rutuja Kuhikar
- a National Centre for Cell Science, NCCS Complex , Savitribai Phule Pune University Campus , Pune , India
| | - Vaijayanti Kale
- a National Centre for Cell Science, NCCS Complex , Savitribai Phule Pune University Campus , Pune , India
| | - Lalita Limaye
- a National Centre for Cell Science, NCCS Complex , Savitribai Phule Pune University Campus , Pune , India
| |
Collapse
|
12
|
Ito Y, Nakamura S, Sugimoto N, Shigemori T, Kato Y, Ohno M, Sakuma S, Ito K, Kumon H, Hirose H, Okamoto H, Nogawa M, Iwasaki M, Kihara S, Fujio K, Matsumoto T, Higashi N, Hashimoto K, Sawaguchi A, Harimoto KI, Nakagawa M, Yamamoto T, Handa M, Watanabe N, Nishi E, Arai F, Nishimura S, Eto K. Turbulence Activates Platelet Biogenesis to Enable Clinical Scale Ex Vivo Production. Cell 2018; 174:636-648.e18. [PMID: 30017246 DOI: 10.1016/j.cell.2018.06.011] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 03/30/2018] [Accepted: 05/23/2018] [Indexed: 12/14/2022]
Abstract
The ex vivo generation of platelets from human-induced pluripotent cells (hiPSCs) is expected to compensate donor-dependent transfusion systems. However, manufacturing the clinically required number of platelets remains unachieved due to the low platelet release from hiPSC-derived megakaryocytes (hiPSC-MKs). Here, we report turbulence as a physical regulator in thrombopoiesis in vivo and its application to turbulence-controllable bioreactors. The identification of turbulent energy as a determinant parameter allowed scale-up to 8 L for the generation of 100 billion-order platelets from hiPSC-MKs, which satisfies clinical requirements. Turbulent flow promoted the release from megakaryocytes of IGFBP2, MIF, and Nardilysin to facilitate platelet shedding. hiPSC-platelets showed properties of bona fide human platelets, including circulation and hemostasis capacities upon transfusion in two animal models. This study provides a concept in which a coordinated physico-chemical mechanism promotes platelet biogenesis and an innovative strategy for ex vivo platelet manufacturing.
Collapse
Affiliation(s)
- Yukitaka Ito
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Kyoto Development Center, Megakaryon Corporation, Kyoto, Japan
| | - Sou Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Naoshi Sugimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | | | - Yoshikazu Kato
- Mixing Technology Laboratory, SATAKE Chemical Equipment Manufacturing Ltd., Saitama, Japan
| | - Mikiko Ohno
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Japan
| | - Shinya Sakuma
- Department of Micro-Nano Systems Engineering, Nagoya University, Nagoya, Japan
| | - Keitaro Ito
- Department of Micro-Nano Systems Engineering, Nagoya University, Nagoya, Japan
| | - Hiroki Kumon
- Department of Micro-Nano Systems Engineering, Nagoya University, Nagoya, Japan
| | - Hidenori Hirose
- Kyoto Development Center, Megakaryon Corporation, Kyoto, Japan
| | - Haruki Okamoto
- Kyoto Development Center, Megakaryon Corporation, Kyoto, Japan
| | - Masayuki Nogawa
- Center for Transfusion Medicine and Cell Therapy, Keio University School of Medicine, Tokyo, Japan
| | - Mio Iwasaki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shunsuke Kihara
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kosuke Fujio
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Takuya Matsumoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Natsumi Higashi
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kazuya Hashimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Akira Sawaguchi
- Department of Anatomy, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Ken-Ichi Harimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masato Nakagawa
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; AMED-CREST, AMED, Tokyo, Japan
| | - Makoto Handa
- Center for Transfusion Medicine and Cell Therapy, Keio University School of Medicine, Tokyo, Japan
| | - Naohide Watanabe
- Center for Transfusion Medicine and Cell Therapy, Keio University School of Medicine, Tokyo, Japan
| | - Eiichiro Nishi
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Japan
| | - Fumihito Arai
- Department of Micro-Nano Systems Engineering, Nagoya University, Nagoya, Japan
| | - Satoshi Nishimura
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan.
| |
Collapse
|
13
|
Martini S, Pozzi G, Carubbi C, Masselli E, Galli D, Di Nuzzo S, Banchini A, Gobbi G, Vitale M, Mirandola P. PKCε promotes human Th17 differentiation: Implications in the pathophysiology of psoriasis. Eur J Immunol 2018; 48:644-654. [DOI: 10.1002/eji.201747102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 12/01/2017] [Accepted: 12/15/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Silvia Martini
- Department of Medicine & Surgery (DiMeC); University of Parma; Parma IT
- CoreLab; Azienda Ospedaliero-Universitaria di Parma; Parma IT
| | - Giulia Pozzi
- Department of Medicine & Surgery (DiMeC); University of Parma; Parma IT
| | - Cecilia Carubbi
- Department of Medicine & Surgery (DiMeC); University of Parma; Parma IT
- CoreLab; Azienda Ospedaliero-Universitaria di Parma; Parma IT
| | - Elena Masselli
- Department of Medicine & Surgery (DiMeC); University of Parma; Parma IT
| | - Daniela Galli
- Department of Medicine & Surgery (DiMeC); University of Parma; Parma IT
| | - Sergio Di Nuzzo
- Department of Medicine & Surgery (DiMeC); University of Parma; Parma IT
| | - Antonio Banchini
- Department of Medicine & Surgery (DiMeC); University of Parma; Parma IT
| | - Giuliana Gobbi
- Department of Medicine & Surgery (DiMeC); University of Parma; Parma IT
- CoreLab; Azienda Ospedaliero-Universitaria di Parma; Parma IT
| | - Marco Vitale
- Department of Medicine & Surgery (DiMeC); University of Parma; Parma IT
- CoreLab; Azienda Ospedaliero-Universitaria di Parma; Parma IT
| | - Prisco Mirandola
- Department of Medicine & Surgery (DiMeC); University of Parma; Parma IT
- CoreLab; Azienda Ospedaliero-Universitaria di Parma; Parma IT
| |
Collapse
|
14
|
Kostyak JC, Liverani E, Kunapuli SP. PKC-epsilon deficiency alters progenitor cell populations in favor of megakaryopoiesis. PLoS One 2017; 12:e0182867. [PMID: 28783756 PMCID: PMC5544228 DOI: 10.1371/journal.pone.0182867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/25/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND It has long been postulated that Protein Kinase C (PKC) is an important regulator of megakaryopoiesis. Recent contributions to the literature have outlined the functions of several individual PKC isoforms with regard to megakaryocyte differentiation and platelet production. However, the exact role of PKCε remains elusive. OBJECTIVE To delineate the role of PKCε in megakaryopoiesis. APPROACH AND RESULTS We used a PKCε knockout mouse model to examine the effect of PKCε deficiency on platelet mass, megakaryocyte mass, and bone marrow progenitor cell distribution. We also investigated platelet recovery in PKCε null mice and TPO-mediated signaling in PKCε null megakaryocytes. PKCε null mice have higher platelet counts due to increased platelet production compared to WT littermate controls (p<0.05, n = 8). Furthermore, PKCε null mice have more bone marrow megakaryocyte progenitor cells than WT littermate control mice. Additionally, thrombopoietin-mediated signaling is perturbed in PKCε null mice as Akt and ERK1/2 phosphorylation are enhanced in PKCε null megakaryocytes stimulated with thrombopoietin. Finally, in response to immune-induced thrombocytopenia, PKCε null mice recovered faster and had higher rebound thrombocytosis than WT littermate control mice. CONCLUSIONS Enhanced platelet recovery could be due to an increase in megakaryocyte progenitor cells found in PKCε null mice as well as enhanced thrombopoietin-mediated signaling observed in PKCε deficient megakaryocytes. These data suggest that PKCε is a negative regulator of megakaryopoiesis.
Collapse
Affiliation(s)
- John C. Kostyak
- Sol Sherry Thrombosis Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Elisabetta Liverani
- Center for Inflammation, Translational and Clinical Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Satya P. Kunapuli
- Sol Sherry Thrombosis Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Pharmacology, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Physiology, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
15
|
Wang JY, Ye S, Zhong H. The role of bone marrow microenvironment in platelet production and their implications for the treatment of thrombocytopenic diseases. ACTA ACUST UNITED AC 2017; 22:630-639. [PMID: 28569613 DOI: 10.1080/10245332.2017.1333274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Impaired platelet production has been found to be an important pathological mechanism of thrombocytopenia in many diseases. Platelet generation is a complex process that mainly occurs in the bone marrow, and thus is closely regulated by the bone marrow microenvironment. This review attempts to summarize the most current knowledge referring the role of bone marrow microenvironment in the regulation of platelet production. METHODS The effects of multiple microenvironment ingredients in regulating megakaryopoiesis and thrombocytopoiesis have been discussed. Abnormalities of these components in thrombocytopenic diseases are also described. DISCUSSIONS Thrombocytopenia is a common clinical manifestation of a variety of diseases. The functional importance of platelets has driven the developments of a broad range of studies. Platelet generation mainly occurs within the bone marrow, where the cells, soluble factors, and extracellular matrix proteins collaboratively form a complex regulatory network, directing megakaryocytic proliferation and differentiation. Alteration in any part of the regulating network may result in defective platelet formation, and eventually lead to thrombocytopenia. A variety of thrombocytopenic diseases have been found to be related with the disregulated bone marrow microenvironment. Identification of the variations of these niche ingredients in certain diseases has facilitated the developments of multiple therapeutic regimes. Further studies that can combine these niche factors with their downstream regulatory factors will be beneficial for developing more effective therapies. CONCLUSIONS Further definition of the role of bone marrow microenvironment in platelet generation may deepen our understanding of the underlying mechanisms as well as provide new therapeutic targets for thrombocytopenic diseases.
Collapse
Affiliation(s)
- Jun-Ying Wang
- a Department of Hematology, South Campus Ren Ji Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai , PR China
| | - Shuang Ye
- b Department of Rheumatology, South Campus Ren Ji Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai , PR China
| | - Hua Zhong
- a Department of Hematology, South Campus Ren Ji Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai , PR China
| |
Collapse
|
16
|
Carubbi C, Masselli E, Martini S, Galli D, Aversa F, Mirandola P, Italiano JE, Gobbi G, Vitale M. Human thrombopoiesis depends on Protein kinase Cδ/protein kinase Cε functional couple. Haematologica 2016; 101:812-20. [PMID: 27081176 DOI: 10.3324/haematol.2015.137984] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 04/12/2016] [Indexed: 01/12/2023] Open
Abstract
A deeper understanding of the molecular events driving megakaryocytopoiesis and thrombopoiesis is essential to regulate in vitro and in vivo platelet production for clinical applications. We previously documented the crucial role of PKCε in the regulation of human and mouse megakaryocyte maturation and platelet release. However, since several data show that different PKC isoforms fulfill complementary functions, we targeted PKCε and PKCδ, which show functional and phenotypical reciprocity, at the same time as boosting platelet production in vitro. Results show that PKCδ, contrary to PKCε, is persistently expressed during megakaryocytic differentiation, and a forced PKCδ down-modulation impairs megakaryocyte maturation and platelet production. PKCδ and PKCε work as a functional couple with opposite roles on thrombopoiesis, and the modulation of their balance strongly impacts platelet production. Indeed, we show an imbalance of PKCδ/PKCε ratio both in primary myelofibrosis and essential thrombocythemia, featured by impaired megakaryocyte differentiation and increased platelet production, respectively. Finally, we demonstrate that concurrent molecular targeting of both PKCδ and PKCε represents a strategy for in vitro platelet factories.
Collapse
Affiliation(s)
- Cecilia Carubbi
- Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Italy
| | - Elena Masselli
- Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Italy
| | - Silvia Martini
- Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Italy
| | - Daniela Galli
- Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Italy
| | - Franco Aversa
- Department of Clinical and Experimental Medicine, University of Parma, Italy
| | - Prisco Mirandola
- Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Italy
| | - Joseph E Italiano
- Hematology Division, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Giuliana Gobbi
- Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Italy
| | - Marco Vitale
- Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Italy
| |
Collapse
|
17
|
Sympathetic stimulation facilitates thrombopoiesis by promoting megakaryocyte adhesion, migration, and proplatelet formation. Blood 2016; 127:1024-35. [DOI: 10.1182/blood-2015-07-660746] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/28/2015] [Indexed: 12/22/2022] Open
Abstract
Key Points
NE and EPI promote megakaryocyte adhesion, migration, and proplatelet formation via α2-adrenoceptor-ERK1/2 signaling. Sympathetic stimulation enhances platelet production, which may facilitate recovery of thrombocytopenia or aggravate atherosclerosis.
Collapse
|
18
|
Di Marcantonio D, Galli D, Carubbi C, Gobbi G, Queirolo V, Martini S, Merighi S, Vaccarezza M, Maffulli N, Sykes SM, Vitale M, Mirandola P. PKCε as a novel promoter of skeletal muscle differentiation and regeneration. Exp Cell Res 2015; 339:10-9. [PMID: 26431586 DOI: 10.1016/j.yexcr.2015.09.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 09/23/2015] [Accepted: 09/26/2015] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Satellite cells are muscle resident stem cells and are responsible for muscle regeneration. In this study we investigate the involvement of PKCε during muscle stem cell differentiation in vitro and in vivo. Here, we describe the identification of a previously unrecognized role for the PKCε-HMGA1 signaling axis in myoblast differentiation and regeneration processes. METHODS PKCε expression was modulated in the C2C12 cell line and primary murine satellite cells in vitro, as well as in an in vivo model of muscle regeneration. Immunohistochemistry and immunofluorescence, RT-PCR and shRNA silencing techniques were used to determine the role of PKCε and HMGA1 in myogenic differentiation. RESULTS PKCε expression increases and subsequently re-localizes to the nucleus during skeletal muscle cell differentiation. In the nucleus, PKCε blocks Hmga1 expression to promote Myogenin and Mrf4 accumulation and myoblast formation. Following in vivo muscle injury, PKCε accumulates in regenerating, centrally-nucleated myofibers. Pharmacological inhibition of PKCε impairs the expression of two crucial markers of muscle differentiation, namely MyoD and Myogenin, during injury induced muscle regeneration. CONCLUSION This work identifies the PKCε-HMGA1 signaling axis as a positive regulator of skeletal muscle differentiation.
Collapse
Affiliation(s)
- D Di Marcantonio
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Via Gramsci, 14, 43100 Parma, Italy; Immune Cell Development and Host Defense, Research Institute of Fox Chase Cancer Center, Philadelphia, PA, USA
| | - D Galli
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Via Gramsci, 14, 43100 Parma, Italy; Centre for Molecular and Translational Oncology (COMT), University of Parma, Italy; Sport and Exercise Medicine Center (SEM), University of Parma, Italy
| | - C Carubbi
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Via Gramsci, 14, 43100 Parma, Italy
| | - G Gobbi
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Via Gramsci, 14, 43100 Parma, Italy; Centre for Molecular and Translational Oncology (COMT), University of Parma, Italy; Sport and Exercise Medicine Center (SEM), University of Parma, Italy
| | - V Queirolo
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Via Gramsci, 14, 43100 Parma, Italy
| | - S Martini
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Via Gramsci, 14, 43100 Parma, Italy
| | - S Merighi
- Department of Medical Science, University of Ferrara, Italy
| | - M Vaccarezza
- Department of Human Sciences, Society and Health (HSSH), University of Cassino, FR, Italy; School of Biomedical Sciences, University of Queensland, Brisbane, QLD 4072, Australia
| | - N Maffulli
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK; Department of Musculoskeletal Disorders, University of Salerno School of Medicine and Surgery, Salerno, Italy
| | - S M Sykes
- Immune Cell Development and Host Defense, Research Institute of Fox Chase Cancer Center, Philadelphia, PA, USA
| | - M Vitale
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Via Gramsci, 14, 43100 Parma, Italy; Centre for Molecular and Translational Oncology (COMT), University of Parma, Italy; Sport and Exercise Medicine Center (SEM), University of Parma, Italy.
| | - P Mirandola
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Via Gramsci, 14, 43100 Parma, Italy; Centre for Molecular and Translational Oncology (COMT), University of Parma, Italy; Sport and Exercise Medicine Center (SEM), University of Parma, Italy
| |
Collapse
|
19
|
Poulter NS, Thomas SG. Cytoskeletal regulation of platelet formation: Coordination of F-actin and microtubules. Int J Biochem Cell Biol 2015. [PMID: 26210823 DOI: 10.1016/j.biocel.2015.07.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Platelets are small, anucleate blood cells which play an important role in haemostasis. Thrombocytopenia is a condition where the platelet count falls below 150×10(9)/l and patients suffering from severe forms of this condition can experience life-threatening bleeds requiring platelet transfusions. Platelets are produced from large progenitor cells called megakaryocytes which are found in the bone marrow. The process of megakaryocyte maturation and the formation of proplatelets are essential steps in the production of mature platelets and both depend heavily on the actin and microtubule cytoskeletons. Understanding these processes is important for the development of in vitro platelet production which will help to treat thrombocytopenia as well as produce model systems for studying platelet-associated disorders. This review will highlight some of the recent advances in our understanding of the role of the cytoskeleton in platelet production, especially the key molecules and signalling pathways that regulate actin and microtubule crosstalk.
Collapse
Affiliation(s)
- Natalie S Poulter
- Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Steven G Thomas
- Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
20
|
Masselli E, Carubbi C, Gobbi G, Mirandola P, Galli D, Martini S, Bonomini S, Crugnola M, Craviotto L, Aversa F, Vitale M. Protein kinase Cɛ inhibition restores megakaryocytic differentiation of hematopoietic progenitors from primary myelofibrosis patients. Leukemia 2015; 29:2192-201. [PMID: 26183534 DOI: 10.1038/leu.2015.150] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/07/2015] [Accepted: 05/29/2015] [Indexed: 01/02/2023]
Abstract
Among the three classic Philadelphia chromosome-negative myeloproliferative neoplasms, primary myelofibrosis (PMF) is the most severe in terms of disease biology, survival and quality of life. Abnormalities in the process of differentiation of PMF megakaryocytes (MKs) are a hallmark of the disease. Nevertheless, the molecular events that lead to aberrant megakaryocytopoiesis have yet to be clarified. Protein kinase Cɛ (PKCɛ) is a novel serine/threonine kinase that is overexpressed in a variety of cancers, promoting aggressive phenotype, invasiveness and drug resistance. Our previous findings on the role of PKCɛ in normal (erythroid and megakaryocytic commitment) and malignant (acute myeloid leukemia) hematopoiesis prompted us to investigate whether it could be involved in the pathogenesis of PMF MK-impaired differentiation. We demonstrate that PMF megakaryocytic cultures express higher levels of PKCɛ than healthy donors, which correlate with higher disease burden but not with JAK2V617F mutation. Inhibition of PKCɛ function (by a negative regulator of PKCɛ translocation) or translation (by target small hairpin RNA) leads to reduction in PMF cell growth, restoration of PMF MK differentiation and inhibition of PKCɛ-related anti-apoptotic signaling (Bcl-xL). Our data suggest that targeting PKCɛ directly affects the PMF neoplastic clone and represent a proof-of-concept for PKCɛ inhibition as a novel therapeutic strategy in PMF.
Collapse
Affiliation(s)
- E Masselli
- Hematology and BMT Unit, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy.,Unit of Human Anatomy and Histology, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| | - C Carubbi
- Unit of Human Anatomy and Histology, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| | - G Gobbi
- Unit of Human Anatomy and Histology, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| | - P Mirandola
- Unit of Human Anatomy and Histology, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| | - D Galli
- Unit of Human Anatomy and Histology, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| | - S Martini
- Unit of Human Anatomy and Histology, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| | - S Bonomini
- Hematology and BMT Unit, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - M Crugnola
- Hematology and BMT Unit, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - L Craviotto
- Department of Clinical and Experimental Medicine, Hematology and BMT Unit, University of Parma, Parma, Italy
| | - F Aversa
- Hematology and BMT Unit, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy.,Department of Clinical and Experimental Medicine, Hematology and BMT Unit, University of Parma, Parma, Italy
| | - M Vitale
- Unit of Human Anatomy and Histology, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| |
Collapse
|
21
|
Shi DS, Smith MCP, Campbell RA, Zimmerman PW, Franks ZB, Kraemer BF, Machlus KR, Ling J, Kamba P, Schwertz H, Rowley JW, Miles RR, Liu ZJ, Sola-Visner M, Italiano JE, Christensen H, Kahr WHA, Li DY, Weyrich AS. Proteasome function is required for platelet production. J Clin Invest 2014; 124:3757-66. [PMID: 25061876 DOI: 10.1172/jci75247] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 06/05/2014] [Indexed: 01/03/2023] Open
Abstract
The proteasome inhibiter bortezomib has been successfully used to treat patients with relapsed multiple myeloma; however, many of these patients become thrombocytopenic, and it is not clear how the proteasome influences platelet production. Here we determined that pharmacologic inhibition of proteasome activity blocks proplatelet formation in human and mouse megakaryocytes. We also found that megakaryocytes isolated from mice deficient for PSMC1, an essential subunit of the 26S proteasome, fail to produce proplatelets. Consistent with decreased proplatelet formation, mice lacking PSMC1 in platelets (Psmc1(fl/fl) Pf4-Cre mice) exhibited severe thrombocytopenia and died shortly after birth. The failure to produce proplatelets in proteasome-inhibited megakaryocytes was due to upregulation and hyperactivation of the small GTPase, RhoA, rather than NF-κB, as has been previously suggested. Inhibition of RhoA or its downstream target, Rho-associated protein kinase (ROCK), restored megakaryocyte proplatelet formation in the setting of proteasome inhibition in vitro. Similarly, fasudil, a ROCK inhibitor used clinically to treat cerebral vasospasm, restored platelet counts in adult mice that were made thrombocytopenic by tamoxifen-induced suppression of proteasome activity in megakaryocytes and platelets (Psmc1(fl/fl) Pdgf-Cre-ER mice). These results indicate that proteasome function is critical for thrombopoiesis, and suggest inhibition of RhoA signaling as a potential strategy to treat thrombocytopenia in bortezomib-treated multiple myeloma patients.
Collapse
|
22
|
Machlus KR, Thon JN, Italiano JE. Interpreting the developmental dance of the megakaryocyte: a review of the cellular and molecular processes mediating platelet formation. Br J Haematol 2014; 165:227-36. [PMID: 24499183 DOI: 10.1111/bjh.12758] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Platelets are essential for haemostasis, and thrombocytopenia (platelet counts <150 × 10(9) /l) is a major clinical problem encountered across a number of conditions, including immune thrombocytopenic purpura, myelodysplastic syndromes, chemotherapy, aplastic anaemia, human immunodeficiency virus infection, complications during pregnancy and delivery, and surgery. Circulating blood platelets are specialized cells that function to prevent bleeding and minimize blood vessel injury. Platelets circulate in their quiescent form, and upon stimulation, activate to release their granule contents and spread on the affected tissue to create a physical barrier that prevents blood loss. The current model of platelet formation states that large progenitor cells in the bone marrow, called megakaryocytes, release platelets by extending long, branching processes, designated proplatelets, into sinusoidal blood vessels. This review will focus on different factors that impact megakaryocyte development, proplatelet formation and platelet release. It will highlight recent studies on thrombopoeitin-dependent megakaryocyte maturation, endomitosis and granule formation, cytoskeletal contributions to proplatelet formation, the role of apoptosis, and terminal platelet formation and release.
Collapse
Affiliation(s)
- Kellie R Machlus
- Hematology Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
23
|
Avanzi MP, Goldberg F, Davila J, Langhi D, Chiattone C, Mitchell WB. Rho kinase inhibition drives megakaryocyte polyploidization and proplatelet formation through MYC and NFE2 downregulation. Br J Haematol 2014; 164:867-76. [PMID: 24383889 DOI: 10.1111/bjh.12709] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/08/2013] [Indexed: 01/08/2023]
Abstract
The processes of megakaryocyte polyploidization and demarcation membrane system (DMS) formation are crucial for platelet production, but the mechanisms controlling these processes are not fully determined. Inhibition of Rho kinase (ROCK) signalling leads to increased polyploidization in umbilical cord blood-derived megakaryocytes. To extend these findings we determined the effect of ROCK inhibition on development of the DMS and on proplatelet formation. The underlying mechanisms were explored by analysing the effect of ROCK inhibition on the expression of MYC and NFE2, which encode two transcription factors critical for megakaryocyte development. ROCK inhibition promoted DMS formation, and increased proplatelet formation and platelet release. Rho kinase inhibition also downregulated MYC and NFE2 expression in mature megakaryocytes, and this down-regulation correlated with increased proplatelet formation. Our findings suggest a model whereby ROCK inhibition drives polyploidization, DMS growth and proplatelet formation late in megakaryocyte maturation through downregulation of MYC and NFE2 expression.
Collapse
Affiliation(s)
- Mauro P Avanzi
- Platelet Biology Laboratory, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA; Cellular Therapy Laboratory, Hematology Division, Santa Casa Medical School, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|