47 results on '"Irina Pleines"'
Search Results
2. Critical redundant functions of the adapters Grb2 and Gads in platelet (hem)ITAM signaling in mice
- Author
-
Timo Vögtle, Ayesha A. Baig, Julia Volz, Timothy B. Duchow, Irina Pleines, Sebastian Dütting, Lars Nitschke, Stephen P. Watson, and Bernhard Nieswandt
- Subjects
adapter proteins ,hemostasis ,itam ,mice ,signaling ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Platelets are essential for normal hemostasis; however, pathological conditions can also trigger unwanted platelet activation precipitating thrombosis and ischemic damage of vital organs such as the heart or brain. Glycoprotein (GP)VI- and C-type lectin-like receptor 2 (CLEC-2)-mediated (hem)immunoreceptor tyrosine-based activation motif (ITAM) signaling represents a major pathway for platelet activation. The two members of the Growth-factor receptor-bound protein 2 (Grb2) family of adapter proteins expressed in platelets – Grb2 and Grb2-related adapter protein downstream of Shc (Gads) – are part of the hem(ITAM) signaling cascade by forming an adapter protein complex with linker for activation of T cells (LAT). To date, a possible functional redundancy between these two adapters in platelet activation has not been investigated. We here generated megakaryocyte- and platelet-specific Grb2/Gads double knockout (DKO) mice and analyzed their platelet function in vitro and in vivo. The DKO platelets exhibited virtually abolished (hem)ITAM signaling whereas only partial defects were seen in Grb2 or Gads single-deficient platelets. This was based on impaired phosphorylation of key molecules in the (hem)ITAM signaling cascade and translated into impaired hemostasis and partially defective arterial thrombosis, thereby exceeding the defects in either Grb2 KO or Gads KO mice. Despite this severe (hem)ITAM signaling defect, CLEC-2 dependent regulation of blood-lymphatic vessel separation was not affected in the DKO animals. These results provide direct evidence for critically redundant roles of Grb2 and Gads for platelet function in hemostasis and thrombosis, but not development.
- Published
- 2020
- Full Text
- View/download PDF
3. Blood collection, platelet isolation and measurement of platelet count and size in mice—a practical guide
- Author
-
Katja Aurbach, Markus Spindler, Elizabeth J. Haining, Markus Bender, and Irina Pleines
- Subjects
hematology ,mice ,platelet count ,platelet isolation ,platelet size ,protocol ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Inherited or acquired disorders of platelet production and function can result in thrombocytopenia and bleeding. Mouse models have proven useful for investigating the mechanisms that underlie these defects in humans. Precise methods for blood withdrawal, platelet isolation and measurement of platelet parameters are key for the generation of reproducible and conclusive data. Here, we provide three different protocols for mouse platelet isolation to encourage research knowledge transfer between experienced laboratories, while at the same time enabling less experienced researchers to implement a protocol that best suits their local expertise and equipment. We also address the issue that reported mouse platelet count and size vary considerably in the literature by investigating different factors that influence these important platelet parameters, namely: 1) genetic background and gender, 2) choice of analysis method (hematological analyzer or flow cytometry), 3) dilution of the blood sample and 4) choice of anticoagulant. The herein presented results and considerations may serve as a practical guide for both experienced and new researchers in the platelet field.
- Published
- 2019
- Full Text
- View/download PDF
4. Rho GTPases and their downstream effectors in megakaryocyte biology
- Author
-
Irina Pleines, Deya Cherpokova, and Markus Bender
- Subjects
cdc42 ,cytoskeleton ,megakaryocyte ,platelet ,rac1 ,rhoa ,rho gtpases ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Megakaryocytes differentiate from hematopoietic stem cells in the bone marrow. The transition of megakaryocytes to platelets is a complex process. Thereby, megakaryocytes extend proplatelets into sinusoidal blood vessels, where the proplatelets undergo fission to release platelets. Defects in platelet production can lead to a low platelet count (thrombocytopenia) with increased bleeding risk. Rho GTPases comprise a family of small signaling G proteins that have been shown to be master regulators of the cytoskeleton controlling many aspects of intracellular processes. The generation of Pf4-Cre transgenic mice was a major breakthrough that enabled studies in megakaryocyte-/platelet-specific knockout mouse lines and provided new insights into the central regulatory role of Rho GTPases in megakaryocyte maturation and platelet production. In this review, we will summarize major findings on the role of Rho GTPases in megakaryocyte biology with a focus on mouse lines in which knockout strategies have been applied to study the function of the best-characterized members Rac1, Cdc42 and RhoA and their downstream effector proteins.
- Published
- 2019
- Full Text
- View/download PDF
5. Coactosin-like 1 integrates signaling critical for shear-dependent thrombus formation in mouse platelets
- Author
-
Inga Scheller, Simon Stritt, Sarah Beck, Bing Peng, Irina Pleines, Katrin G. Heinze, Attila Braun, Oliver Otto, Robert Ahrends, Albert Sickmann, Markus Bender, and Bernhard Nieswandt
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Platelet aggregate formation is a multistep process involving receptor-mediated, as well as biomechanical, signaling cascades, which are highly dependent on actin dynamics. We have previously shown that actin depolymerizing factor (ADF)/n-cofilin and Twinfilin 2a, members of the ADF homology (ADF-H) protein family, have distinct roles in platelet formation and function. Coactosin-like 1 (Cotl1) is another ADF-H protein that binds actin and was also shown to enhance biosynthesis of pro-inflammatory leukotrienes (LT) in granulocytes. Here, we generated mice lacking Cotl1 in the megakaryocyte lineage (Cotl1−/−) to investigate its role in platelet production and function. Absence of Cotl1 had no impact on platelet counts, platelet activation or cytoskeletal reorganization under static conditions in vitro. In contrast, Cotl1 deficiency markedly affected platelet aggregate formation on collagen and adhesion to immobilized von Willebrand factor at high shear rates in vitro, pointing to an impaired function of the platelet mechanoreceptor glycoprotein (GP) Ib. Furthermore, Cotl1−/−platelets exhibited increased deformability at high shear rates, indicating that the GPIb defect may be linked to altered biomechanical properties of the deficient cells. In addition, we found that Cotl1 deficiency markedly affected platelet LT biosynthesis. Strikingly, exogenous LT addition restored defective aggregate formation of Cotl1−/− platelets at high shear in vitro, indicating a critical role of platelet-derived LT in thrombus formation. In vivo, Cotl1 deficiency translated into prolonged tail bleeding times and protection from occlusive arterial thrombus formation. Together, our results show that Cotl1 in platelets is an integrator of biomechanical and LT signaling in hemostasis and thrombosis.
- Published
- 2020
- Full Text
- View/download PDF
6. A Cdc42/RhoA regulatory circuit downstream of glycoprotein Ib guides transendothelial platelet biogenesis
- Author
-
Sebastian Dütting, Frederique Gaits-Iacovoni, David Stegner, Michael Popp, Adrien Antkowiak, Judith M.M. van Eeuwijk, Paquita Nurden, Simon Stritt, Tobias Heib, Katja Aurbach, Oguzhan Angay, Deya Cherpokova, Niels Heinz, Ayesha A. Baig, Maximilian G. Gorelashvili, Frank Gerner, Katrin G. Heinze, Jerry Ware, Georg Krohne, Zaverio M. Ruggeri, Alan T. Nurden, Harald Schulze, Ute Modlich, Irina Pleines, Cord Brakebusch, and Bernhard Nieswandt
- Subjects
Science - Abstract
Platelets derive from large precursor cells (megakaryocytes) in the bone marrow. Düttinget al. show that megakaryocyte polarization and platelet biogenesis in the bone-marrow sinusoids are directed by adhesion receptor GPIb signalling and resulting balanced antagonism between RhoA (stop-signal) and Cdc42 (go-signal).
- Published
- 2017
- Full Text
- View/download PDF
7. Comparative Analysis of Microfluidics Thrombus Formation in Multiple Genetically Modified Mice: Link to Thrombosis and Hemostasis
- Author
-
Magdolna Nagy, Johanna P. van Geffen, David Stegner, David J. Adams, Attila Braun, Susanne M. de Witt, Margitta Elvers, Mitchell J. Geer, Marijke J. E. Kuijpers, Karl Kunzelmann, Jun Mori, Cécile Oury, Joachim Pircher, Irina Pleines, Alastair W. Poole, Yotis A. Senis, Remco Verdoold, Christian Weber, Bernhard Nieswandt, Johan W. M. Heemskerk, and Constance C. F. M. J. Baaten
- Subjects
arterial thrombus formation ,bleeding ,collagen ,glycoprotein VI ,platelets ,microfluidics ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Genetically modified mice are indispensable for establishing the roles of platelets in arterial thrombosis and hemostasis. Microfluidics assays using anticoagulated whole blood are commonly used as integrative proxy tests for platelet function in mice. In the present study, we quantified the changes in collagen-dependent thrombus formation for 38 different strains of (genetically) modified mice, all measured with the same microfluidics chamber. The mice included were deficient in platelet receptors, protein kinases or phosphatases, small GTPases or other signaling or scaffold proteins. By standardized re-analysis of high-resolution microscopic images, detailed information was obtained on altered platelet adhesion, aggregation and/or activation. For a subset of 11 mouse strains, these platelet functions were further evaluated in rhodocytin- and laminin-dependent thrombus formation, thus allowing a comparison of glycoprotein VI (GPVI), C-type lectin-like receptor 2 (CLEC2) and integrin α6β1 pathways. High homogeneity was found between wild-type mice datasets concerning adhesion and aggregation parameters. Quantitative comparison for the 38 modified mouse strains resulted in a matrix visualizing the impact of the respective (genetic) deficiency on thrombus formation with detailed insight into the type and extent of altered thrombus signatures. Network analysis revealed strong clusters of genes involved in GPVI signaling and Ca2+ homeostasis. The majority of mice demonstrating an antithrombotic phenotype in vivo displayed with a larger or smaller reduction in multi-parameter analysis of collagen-dependent thrombus formation in vitro. Remarkably, in only approximately half of the mouse strains that displayed reduced arterial thrombosis in vivo, this was accompanied by impaired hemostasis. This was also reflected by comparing in vitro thrombus formation (by microfluidics) with alterations in in vivo bleeding time. In conclusion, the presently developed multi-parameter analysis of thrombus formation using microfluidics can be used to: (i) determine the severity of platelet abnormalities; (ii) distinguish between altered platelet adhesion, aggregation and activation; and (iii) elucidate both collagen and non-collagen dependent alterations of thrombus formation. This approach may thereby aid in the better understanding and better assessment of genetic variation that affect in vivo arterial thrombosis and hemostasis.
- Published
- 2019
- Full Text
- View/download PDF
8. G6b-B regulates an essential step in megakaryocyte maturation
- Author
-
Isabelle C. Becker, Zoltan Nagy, Georgi Manukjan, Melanie Haffner-Luntzer, Maximilian Englert, Tobias Heib, Timo Vögtle, Carina Gross, Richa Bharti, Sascha Dietrich, Kristina Mott, Johannes Heck, Sebastian Stegmaier, Anke Baranowsky, Thorsten Schinke, Nicolas Schlegel, Tobias Heckel, David Stegner, Irina Pleines, Anita Ignatius, Harald Schulze, and Bernhard Nieswandt
- Subjects
Blood Platelets ,Mice, Inbred C57BL ,Mice ,Nucleotides ,Primary Myelofibrosis ,Animals ,Hematology ,Megakaryocytes ,Thrombocytopenia - Abstract
G6b-B is a megakaryocyte lineage-specific immunoreceptor tyrosine-based inhibition motif–containing receptor, essential for platelet homeostasis. Mice with a genomic deletion of the entire Mpig6b locus develop severe macrothrombocytopenia and myelofibrosis, which is reflected in humans with null mutations in MPIG6B. The current model proposes that megakaryocytes lacking G6b-B develop normally, whereas proplatelet release is hampered, but the underlying molecular mechanism remains unclear. We report on a spontaneous recessive single nucleotide mutation in C57BL/6 mice, localized within the intronic region of the Mpig6b locus that abolishes G6b-B expression and reproduces macrothrombocytopenia, myelofibrosis, and osteosclerosis. As the mutation is based on a single-nucleotide exchange, Mpig6bmut mice represent an ideal model to study the role of G6b-B. Megakaryocytes from these mice were smaller, displayed a less-developed demarcation membrane system, and had a reduced expression of receptors. RNA sequencing revealed a striking global reduction in the level of megakaryocyte-specific transcripts, in conjunction with decreased protein levels of the transcription factor GATA-1 and impaired thrombopoietin signaling. The reduced number of mature MKs in the bone marrow was corroborated on a newly developed Mpig6b-null mouse strain. Our findings highlight an unexpected essential role of G6b-B in the early differentiation within the megakaryocytic lineage.
- Published
- 2022
9. G6b-B regulates an essential step in megakaryocyte maturation
- Author
-
Harald Schulze, J. Heck, T. Heib, R. Bharti, M. Haffner-Luntzer, Bernhard Nieswandt, A. Ignatius, Georgi Manukjan, M. Englert, S. Dietrich, K. Mott, I. C. Becker, Irina Pleines, N. Schlegel, T. Schinke, T. Heckel, A. Jeschke, Zoltan Nagy, C. Gross, T. Voegtle, and David Stegner
- Subjects
Mutation ,RNA ,Biology ,medicine.disease_cause ,medicine.disease ,Cell biology ,medicine.anatomical_structure ,Megakaryocyte ,medicine ,Tyrosine ,Receptor ,Myelofibrosis ,Transcription factor ,Thrombopoietin - Abstract
G6b-B is a megakaryocyte lineage-specific immunoreceptor tyrosine-based inhibition motif (ITIM)-containing receptor, essential for platelet homeostasis. Mice with a genomic deletion of the entire Mpig6b locus develop severe macrothrombocytopenia and myelofibrosis, which is reflected in humans with null-mutations in MPIG6B. The current model proposes that megakaryocytes lacking G6b-B develop normally, while proplatelet release is hampered, but the underlying molecular mechanism remains unclear. Here, we report on a spontaneous recessive single nucleotide mutation in C57BL/6 mice, localized within the intronic region of the Mpig6b locus that abolishes G6b-B expression and reproduces macrothrombocytopenia, myelofibrosis and osteosclerosis. As the mutation is based on a single nucleotide exchange, Mpig6bmut mice represent an ideal model to study the role of G6b-B. Megakaryocytes from these mice were smaller in size, displayed a less developed demarcation membrane system and reduced expression of receptors. RNA sequencing revealed a striking global reduction in the level of megakaryocyte-specific transcripts, in conjunction with decreased protein levels of the transcription factor GATA-1, and impaired thrombopoietin signaling. The reduced number of mature MKs in the bone marrow was corroborated on a newly developed Mpig6b null mouse strain. Our findings highlight an unexpected essential role of G6b-B in the early differentiation within the megakaryocytic lineage.
- Published
- 2021
10. Impaired microtubule dynamics contribute to microthrombocytopenia in RhoB-deficient mice
- Author
-
Maximilian Englert, Katja Aurbach, Isabelle C. Becker, Annika Gerber, Tobias Heib, Lou M. Wackerbarth, Charly Kusch, Kristina Mott, Gabriel H. M. Araujo, Ayesha A. Baig, Sebastian Dütting, Ulla G. Knaus, Christian Stigloher, Harald Schulze, Bernhard Nieswandt, Irina Pleines, and Zoltan Nagy
- Subjects
Blood Platelets ,Mice ,Tubulin ,Animals ,Hematology ,rhoB GTP-Binding Protein ,Megakaryocytes ,Microtubules ,Thrombocytopenia - Abstract
Megakaryocytes are large cells in the bone marrow that give rise to blood platelets. Platelet biogenesis involves megakaryocyte maturation, the localization of the mature cells in close proximity to bone marrow sinusoids, and the formation of protrusions, which are elongated and shed within the circulation. Rho GTPases play important roles in platelet biogenesis and function. RhoA-deficient mice display macrothrombocytopenia and a striking mislocalization of megakaryocytes into bone marrow sinusoids and a specific defect in G-protein signaling in platelets. However, the role of the closely related protein RhoB in megakaryocytes or platelets remains unknown. In this study, we show that, in contrast to RhoA deficiency, genetic ablation of RhoB in mice results in microthrombocytopenia (decreased platelet count and size). RhoB-deficient platelets displayed mild functional defects predominantly upon induction of the collagen/glycoprotein VI pathway. Megakaryocyte maturation and localization within the bone marrow, as well as actin dynamics, were not affected in the absence of RhoB. However, in vitro–generated proplatelets revealed pronouncedly impaired microtubule organization. Furthermore, RhoB-deficient platelets and megakaryocytes displayed selective defects in microtubule dynamics/stability, correlating with reduced levels of acetylated α-tubulin. Our findings imply that the reduction of this tubulin posttranslational modification results in impaired microtubule dynamics, which might contribute to microthrombocytopenia in RhoB-deficient mice. Importantly, we demonstrate that RhoA and RhoB are localized differently and have selective, nonredundant functions in the megakaryocyte lineage.
- Published
- 2021
11. Microthrombocytopenia caused by impaired microtubule stability in RhoB-deficient mice
- Author
-
Maximilian Englert, Katja Aurbach, Annika Gerber, Tobias Heib, Isabelle C. Becker, Lou M. Wackerbarth, Charly Kusch, Ayesha A. Baig, Sebastian Dütting, Ulla G. Knaus, Christian Stigloher, Bernhard Nieswandt, Irina Pleines, and Zoltan Nagy
- Abstract
Megakaryocytes are large cells in the bone marrow, which give rise to blood platelets. Platelet biogenesis involves megakaryocyte maturation, the localization of mature cells in close proximity to bone marrow sinusoids and the formation of protrusions, which are shed into the circulation. Rho GTPases play important roles in platelet biogenesis and function. RhoA-deficient mice display macrothrombocytopenia and a striking mislocalization of megakaryocytes into bone marrow sinusoids and a specific defect in G-protein signaling in platelets. However, the role of the closely related protein RhoB in megakaryocytes or platelets remains unknown. In this study, we show that, in contrast to RhoA deficiency, genetic ablation of RhoB in mice results in microthrombocytopenia (decreased platelet count and size). RhoB-deficient platelets displayed mild functional defects predominantly upon induction of the collagen/glycoprotein VI pathway. Megakaryocyte maturation and localization within the bone marrow, as well as actin dynamics were not affected in the absence of RhoB. However, in vitro generated proplatelets revealed pronouncedly impaired microtubule organization. Furthermore, RhoB-deficient platelets and megakaryocytes displayed selective defects in microtubule dynamics/stability, correlating with pronouncedly reduced levels of acetylated α-tubulin. Our findings imply that absence of this tubulin posttranslational modification results in decreased microtubule stability leading to microthrombocytopenia in RhoB-deficient mice. Our data thus points to specifically impaired microtubule - but not actin - dynamics as a general mechanism underlying the manifestation of microthrombocytopenia in vivo. We furthermore demonstrate that RhoA and RhoB have specific, non-redundant functions in the megakaryocyte lineage.KEY POINTSRhoB-deficient mice display microthrombocytopeniaRhoB has different functions in the megakaryocyte lineage than RhoA and regulates microtubule dynamics
- Published
- 2021
12. Coactosin-like 1 integrates signaling critical for shear-dependent thrombus formation in mouse platelets
- Author
-
Irina Pleines, Albert Sickmann, Katrin G. Heinze, Oliver Otto, Bernhard Nieswandt, Sarah Beck, Simon Stritt, Markus Bender, Attila Braun, Inga Scheller, Robert Ahrends, and Bing Peng
- Subjects
Blood Platelets ,Platelet Biology & its Disorders ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Megakaryocyte ,Von Willebrand factor ,von Willebrand Factor ,medicine ,Animals ,Platelet ,Platelet activation ,Thrombus ,Actin ,Mice, Knockout ,biology ,Chemistry ,Microfilament Proteins ,Thrombosis ,Articles ,Hematology ,Platelet Activation ,medicine.disease ,Cell biology ,medicine.anatomical_structure ,Platelet Glycoprotein GPIb-IX Complex ,Actin depolymerizing factor ,Hemostasis ,biology.protein ,030215 immunology - Abstract
Platelet aggregate formation is a multistep process involving receptor-mediated, as well as biomechanical, signaling cascades, which are highly dependent on actin dynamics. We have previously shown that actin depolymerizing factor (ADF)/n-cofilin and Twinfilin 2a, members of the ADF homology (ADF-H) protein family, have distinct roles in platelet formation and function. Coactosin-like 1 (Cotl1) is another ADF-H protein that binds actin and was also shown to enhance biosynthesis of pro-inflammatory leukotrienes (LT) in granulocytes. Here, we generated mice lacking Cotl1 in the megakaryocyte lineage (Cotl1−/−) to investigate its role in platelet production and function. Absence of Cotl1 had no impact on platelet counts, platelet activation or cytoskeletal reorganization under static conditions in vitro. In contrast, Cotl1 deficiency markedly affected platelet aggregate formation on collagen and adhesion to immobilized von Willebrand factor at high shear rates in vitro, pointing to an impaired function of the platelet mechanoreceptor glycoprotein (GP) Ib. Furthermore, Cotl1−/−platelets exhibited increased deformability at high shear rates, indicating that the GPIb defect may be linked to altered biomechanical properties of the deficient cells. In addition, we found that Cotl1 deficiency markedly affected platelet LT biosynthesis. Strikingly, exogenous LT addition restored defective aggregate formation of Cotl1−/− platelets at high shear in vitro, indicating a critical role of platelet-derived LT in thrombus formation. In vivo, Cotl1 deficiency translated into prolonged tail bleeding times and protection from occlusive arterial thrombus formation. Together, our results show that Cotl1 in platelets is an integrator of biomechanical and LT signaling in hemostasis and thrombosis.
- Published
- 2019
13. RhoA/Cdc42 signaling drives cytoplasmic maturation but not endomitosis in megakaryocytes
- Author
-
Annika Gerber, Isabelle C. Becker, Heike M. Hermanns, Philipp Burkard, Mara Meub, Bernhard Nieswandt, Charly Kusch, Deya Cherpokova, Daniel Jahn, Harald Schulze, Cord Brakebusch, Tobias Heib, Georgi Manukjan, Zoltan Nagy, Maximilian Englert, Thomas Dandekar, Irina Pleines, Christian Stigloher, Johannes Balkenhol, Sarah Beck, Lou Martha Wackerbarth, Sebastian Dütting, and Markus Sauer
- Subjects
0301 basic medicine ,Cytoplasm ,Myosin light-chain kinase ,RHOA ,proplatelet formation ,CDC42 ,macromolecular substances ,General Biochemistry, Genetics and Molecular Biology ,Lim kinase ,megakaryocyte ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Megakaryocyte ,Downregulation and upregulation ,medicine ,Animals ,Humans ,Cdc42 ,Cytoskeleton ,cdc42 GTP-Binding Protein ,platelet ,endomitosis ,biology ,Chemistry ,Rho GTPase ,cytoskeleton ,RhoA ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Bone marrow ,rhoA GTP-Binding Protein ,transcription ,Megakaryocytes ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Summary Megakaryocytes (MKs), the precursors of blood platelets, are large, polyploid cells residing mainly in the bone marrow. We have previously shown that balanced signaling of the Rho GTPases RhoA and Cdc42 is critical for correct MK localization at bone marrow sinusoids in vivo. Using conditional RhoA/Cdc42 double-knockout (DKO) mice, we reveal here that RhoA/Cdc42 signaling is dispensable for the process of polyploidization in MKs but essential for cytoplasmic MK maturation. Proplatelet formation is virtually abrogated in the absence of RhoA/Cdc42 and leads to severe macrothrombocytopenia in DKO animals. The MK maturation defect is associated with downregulation of myosin light chain 2 (MLC2) and β1-tubulin, as well as an upregulation of LIM kinase 1 and cofilin-1 at both the mRNA and protein level and can be linked to impaired MKL1/SRF signaling. Our findings demonstrate that MK endomitosis and cytoplasmic maturation are separately regulated processes, and the latter is critically controlled by RhoA/Cdc42.
- Published
- 2021
14. Isolation of murine bone marrow by centrifugation or flushing for the analysis of hematopoietic cells - a comparative study
- Author
-
David Stegner, Tobias Heib, Martha-Lena Müller, Irina Pleines, and Carina Gross
- Subjects
0301 basic medicine ,Male ,Centrifugation ,Cell Separation ,030204 cardiovascular system & hematology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Bone Marrow ,medicine ,Animals ,Humans ,Platelet ,Chemistry ,Compartment (ship) ,Hematology ,General Medicine ,Hematopoietic Stem Cells ,In vitro ,Cell biology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Bone marrow ,Stem cell - Abstract
Investigation of the bone marrow as the main compartment of hematopoiesis is critical in many research fields. Here, we adapted a centrifugation-based method for the isolation of murine bone marrow and compared it to the traditional flushing method. Analysis of primary hematopoietic stem cells, immune cells, and megakaryocytes revealed a comparable distribution of cellular (sub)populations. Furthermore, in vitro differentiated megakaryocytes displayed unaltered proplatelet formation. Strikingly, bone marrow isolation by centrifugation was considerably faster than the flushing method and significantly increased the cell yield. Thus, the centrifugation-based isolation method is highly suitable for the study of murine bone marrow cells.
- Published
- 2020
15. Actin/microtubule crosstalk during platelet biogenesis in mice is critically regulated by Twinfilin1 and Cofilin1
- Author
-
Markus Spindler, Markus Bender, Georgi Manukjan, Pekka Lappalainen, Katja Aurbach, Sarah Beck, Zoltan Nagy, Harald Schulze, Carina Gross, Walter Witke, Tobias Heib, Irina Pleines, Bernhard Nieswandt, Lou Martha Wackerbarth, Inga Scheller, Isabelle C. Becker, Institute of Biotechnology, and University Management
- Subjects
Blood Platelets ,Cofilin 1 ,ADF/COFILIN FAMILY ,Platelet disorder ,TUBULIN ,PROTEIN ,macromolecular substances ,Microtubules ,Thrombopoiesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Microtubule ,Animals ,Platelet ,Cytoskeleton ,ACTIN POLYMERIZATION ,PROPLATELET FORMATION ,030304 developmental biology ,0303 health sciences ,MICROTUBULE STABILITY ,Chemistry ,MUTATIONS ,Microfilament Proteins ,Hematology ,Actin cytoskeleton ,Platelets and Thrombopoiesis ,Actins ,Cell biology ,Crosstalk (biology) ,MEGAKARYOCYTE ,ENDS ,030220 oncology & carcinogenesis ,1182 Biochemistry, cell and molecular biology ,TURNOVER ,MDia1 ,Megakaryocytes - Abstract
Rearrangements of the microtubule (MT) and actin cytoskeleton are pivotal for platelet biogenesis. Hence, defects in actin- or MT-regulatory proteins are associated with platelet disorders in humans and mice. Previous studies in mice revealed that loss of the actin-depolymerizing factor homology (ADF-H) protein Cofilin1 (Cof1) in megakaryocytes (MKs) results in a moderate macrothrombocytopenia but normal MK numbers, whereas deficiency in another ADF-H protein, Twinfilin1 (Twf1), does not affect platelet production or function. However, recent studies in yeast have indicated a critical synergism between Twf1 and Cof1 in the regulation of actin dynamics. We therefore investigated platelet biogenesis and function in mice lacking both Twf1 and Cof1 in the MK lineage. In contrast to single deficiency in either protein, Twf1/Cof1 double deficiency (DKO) resulted in a severe macrothrombocytopenia and dramatically increased MK numbers in bone marrow and spleen. DKO MKs exhibited defective proplatelet formation in vitro and in vivo as well as impaired spreading and altered assembly of podosome-like structures on collagen and fibrinogen in vitro. These defects were associated with aberrant F-actin accumulation and, remarkably, the formation of hyperstable MT, which appears to be caused by dysregulation of the actin- and MT-binding proteins mDia1 and adenomatous polyposis coli. Surprisingly, the mild functional defects described for Cof1-deficient platelets were only slightly aggravated in DKO platelets suggesting that both proteins are largely dispensable for platelet function in the peripheral blood. In summary, these findings reveal critical redundant functions of Cof1 and Twf1 in ensuring balanced actin/microtubule crosstalk during thrombopoiesis in mice and possibly humans.
- Published
- 2020
16. Loss of Hem1 disrupts macrophage function and impacts on migration, phagocytosis and integrin-mediated adhesion
- Author
-
Stephanie Stahnke, Frieda Kage, Michael Sixt, Aleks Guledani, Klemens Rottner, Theresia E.B. Stradal, Anika Steffen, Charly Kusch, Bernhard Nieswandt, Hermann Döring, Sebastian Dütting, Manfred Rohde, Michael Schnoor, Irina Pleines, David J. J. de Gorter, Mathias Müsken, Jan Faix, and Robert Geffers
- Subjects
Focal adhesion ,biology ,Podosome ,Chemistry ,Integrin ,biology.protein ,Null cell ,Adhesion ,Lamellipodium ,Cell adhesion ,Paxillin ,Cell biology - Abstract
The hematopoietic-specific protein 1 (Hem1) comprises an essential subunit of the WAVE Regulatory Complex (WRC) in immune cells. WRC has a fundamental role in Arp2/3 complex activation and the protrusion of branched actin networks in motile cells.Hem1 deficiency leads to suppression of the entire WRC in immune cells. Defective WRC function in macrophages results in loss of lamellipodia and migration defects. Moreover, phagocytosis, commonly accompanied by lamellipodium protrusion during cup formation, is altered in Hem1 null cells concerning frequency and efficacy. When analyzing cell spreading, adhesion and podosome formation, we found that Hem1 null cells are capable, in principle, of podosome formation and consequently, do not show any quantitative differences in extracellular matrix degradation. Their adhesive behavior, however, was significantly altered. Specifically, adhesion as well as de-adhesion of Hem1 null cells was strongly compromised, likely contributing to the observed reduced efficiency of phagocytosis. In line with this, phosphorylation of the prominent adhesion component paxillin was diminished. Non-hematopoietic somatic cells disrupted in expression for both Hem1 and its ubiquitous orthologue Nck-associated protein 1 (Nap1) or the essential WRC components Sra-1/PIR121 did not only confirm defective paxillin phosphorylation, but also revealed that paxillin turnover in focal adhesions is accelerated in the absence of WRC. Finally, adhesion assays using platelets lacking functional WRC as model system unmasked radically decreased αIIbβ3 integrin activation.Our results thus demonstrate that WRC-driven actin networks impact on integrin-dependent processes controlling formation and dismantling of different types of cell-substratum adhesion.One sentence summaryInterference of Hem1 function in mice and cells uncovers a hitherto unrecognized role in integrin-mediated cell adhesion that is crucial for macrophage function and connects to recently discovered immunodeficiencies in patients carrying Hem1 mutations.
- Published
- 2020
17. Blood collection, platelet isolation and measurement of platelet count and size in mice—a practical guide
- Author
-
Markus Spindler, Katja Aurbach, Elizabeth J. Haining, Irina Pleines, and Markus Bender
- Subjects
Blood Platelets ,Male ,0301 basic medicine ,medicine.medical_specialty ,Isolation (health care) ,Blood withdrawal ,030204 cardiovascular system & hematology ,Bioinformatics ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Platelet production ,medicine ,Animals ,Platelet ,Protocol (science) ,Hematology ,Platelet Count ,business.industry ,General Medicine ,Blood collection ,030104 developmental biology ,Research knowledge ,business - Abstract
Inherited or acquired disorders of platelet production and function can result in thrombocytopenia and bleeding. Mouse models have proven useful for investigating the mechanisms that underlie these defects in humans. Precise methods for blood withdrawal, platelet isolation and measurement of platelet parameters are key for the generation of reproducible and conclusive data. Here, we provide three different protocols for mouse platelet isolation to encourage research knowledge transfer between experienced laboratories, while at the same time enabling less experienced researchers to implement a protocol that best suits their local expertise and equipment. We also address the issue that reported mouse platelet count and size vary considerably in the literature by investigating different factors that influence these important platelet parameters, namely: 1) genetic background and gender, 2) choice of analysis method (hematological analyzer or flow cytometry), 3) dilution of the blood sample and 4) choice of anticoagulant. The herein presented results and considerations may serve as a practical guide for both experienced and new researchers in the platelet field.
- Published
- 2018
18. CK2β regulates thrombopoiesis and Ca2+-triggered platelet activation in arterial thrombosis
- Author
-
Katja Aurbach, Sascha Geue, Christoph Kleinschnitz, Harald Schulze, Britta Walker-Allgaier, Oliver Borst, Daniela Semeniak, Madhumita Chatterjee, Irina Pleines, Melanie Märklin, Thierry Buchou, Eva Geuss, Meinrad Gawaz, David Stegner, Patrick Münzer, Helmut R. Salih, David W. Litchfield, Friederike Langhauser, Irene Gonzalez Menendez, Florian Lang, Bernhard Nieswandt, and Leticia Quintanilla-Martinez
- Subjects
0301 basic medicine ,medicine.medical_specialty ,medicine.diagnostic_test ,Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Vascular occlusion ,03 medical and health sciences ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Megakaryocyte ,Bleeding time ,Hemostasis ,Internal medicine ,medicine ,Platelet ,Platelet activation ,Thrombopoiesis ,medicine.symptom ,Thrombus - Abstract
Platelets, anucleated megakaryocyte (MK)-derived cells, play a major role in hemostasis and arterial thrombosis. Although protein kinase casein kinase 2 (CK2) is readily detected in MKs and platelets, the impact of CK2-dependent signaling on MK/platelet (patho-)physiology has remained elusive. The present study explored the impact of the CK2 regulatory β-subunit on platelet biogenesis and activation. MK/platelet-specific genetic deletion of CK2β (ck2β-/- ) in mice resulted in a significant macrothrombocytopenia and an increased extramedullar megakaryopoiesis with an enhanced proportion of premature platelets. Although platelet life span was only mildly affected, ck2β-/- MK displayed an abnormal microtubule structure with a drastically increased fragmentation within bone marrow and a significantly reduced proplatelet formation in vivo. In ck2β-/- platelets, tubulin polymerization was disrupted, resulting in an impaired thrombopoiesis and an abrogated inositol 1,4,5-triphosphate receptor-dependent intracellular calcium (Ca2+) release. Presumably due to a blunted increase in the concentration of cytosolic Ca2+, activation-dependent increases of α and dense-granule secretion and integrin αIIbβ3 activation, and aggregation were abrogated in ck2β-/- platelets. Accordingly, thrombus formation and stabilization under high arterial shear rates were significantly diminished, and thrombotic vascular occlusion in vivo was significantly blunted in ck2β-/- mice, accompanied by a slight prolongation of bleeding time. Following transient middle cerebral artery occlusion, ck2β-/- mice displayed significantly reduced cerebral infarct volumes, developed significantly less neurological deficits, and showed significantly better outcomes after ischemic stroke than ck2βfl/fl mice. The present observations reveal CK2β as a novel powerful regulator of thrombopoiesis, Ca2+-dependent platelet activation, and arterial thrombosis in vivo.
- Published
- 2017
19. Correction: An essential role for α4A-tubulin in platelet biogenesis
- Author
-
Catherine Strassel, Maria M Magiera, Arnaud Dupuis, Morgane Batzenschlager, Agnès Hovasse, Irina Pleines, Paul Guéguen, Anita Eckly, Sylvie Moog, Léa Mallo, Quentin Kimmerlin, Stéphane Chappaz, Jean-Marc Strub, Natarajan Kathiresan, Henri de la Salle, Alain Van Dorsselaer, Claude Ferec, Jean-Yves Py, Christian Gachet, Christine Schaeffer-Reiss, Benjamin T Kile, Carsten Janke, and François Lanza
- Subjects
Ecology ,biology ,Health, Toxicology and Mutagenesis ,Published Erratum ,macromolecular substances ,Plant Science ,Computational biology ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Blot ,Tubulin ,biology.protein ,Platelet ,Research Articles ,Biogenesis ,Research Article - Abstract
Alpha4A-tubulin is the predominant α-tubulin isotype in platelets. Mutations in α4A-tubulin cause abnormal platelet biogenesis and marginal band formation in mice and in a patient, establishing an essential role of this tubulin isotype., During platelet biogenesis, microtubules (MTs) are arranged into submembranous structures (the marginal band) that encircle the cell in a single plane. This unique MT array has no equivalent in any other mammalian cell, and the mechanisms responsible for this particular mode of assembly are not fully understood. One possibility is that platelet MTs are composed of a particular set of tubulin isotypes that carry specific posttranslational modifications. Although β1-tubulin is known to be essential, no equivalent roles of α-tubulin isotypes in platelet formation or function have so far been reported. Here, we identify α4A-tubulin as a predominant α-tubulin isotype in platelets. Similar to β1-tubulin, α4A-tubulin expression is up-regulated during the late stages of megakaryocyte differentiation. Missense mutations in the α4A-tubulin gene cause macrothrombocytopenia in mice and humans. Defects in α4A-tubulin lead to changes in tubulin tyrosination status of the platelet tubulin pool. Ultrastructural defects include reduced numbers and misarranged MT coils in the platelet marginal band. We further observed defects in megakaryocyte maturation and proplatelet formation in Tuba4a-mutant mice. We have, thus, discovered an α-tubulin isotype with specific and essential roles in platelet biogenesis.
- Published
- 2021
20. Pivotal role of PDK1 in megakaryocyte cytoskeletal dynamics and polarization during platelet biogenesis
- Author
-
Patrick Münzer, Peter Seizer, Dominik Rath, Harald Schulze, Leticia Quintanilla-Fend, Caroline Brähler, Carla E. Borst, Helmut R. Salih, Tobias Geisler, Meinrad Gawaz, Florian Lang, Sascha Geue, Katja Aurbach, Georgi Manukjan, Oliver Borst, Britta Walker-Allgaier, Bernhard Nieswandt, Irina Pleines, Mailin-Christin Manke, David Stegner, and Melanie Märklin
- Subjects
0301 basic medicine ,Blood Platelets ,animal structures ,Podosome ,Immunology ,macromolecular substances ,030204 cardiovascular system & hematology ,Biochemistry ,Filamentous actin ,Thrombopoiesis ,3-Phosphoinositide-Dependent Protein Kinases ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Megakaryocyte ,medicine ,Animals ,Humans ,Platelet activation ,Cytoskeleton ,Mice, Knockout ,Chemistry ,Cell Biology ,Hematology ,Cofilin ,Actin cytoskeleton ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Megakaryocytes - Abstract
During thrombopoiesis, megakaryocytes (MKs) form proplatelets within the bone marrow (BM) and release platelets into BM sinusoids. Phosphoinositide-dependent protein kinase-1 (PDK1) is required for Ca2+-dependent platelet activation, but its role in MK development and regulation of platelet production remained elusive. The present study explored the role of PDK1 in the regulation of MK maturation and polarization during thrombopoiesis using a MK/platelet-specific knockout approach. Pdk1-deficient mice (Pdk1-/-) developed a significant macrothrombocytopenia as compared with wild-type mice (Pdk1fl/fl). Pdk1 deficiency further dramatically increased the number of MKs without sinusoidal contact within the BM hematopoietic compartment, resulting in a pronounced MK hyperplasia and a significantly increased extramedullary thrombopoiesis. Cultured Pdk1-/- BM-MKs showed impaired spreading on collagen, associated with an altered actin cytoskeleton structure with less filamentous actin (F-actin) and diminished podosome formation, whereas the tubulin cytoskeleton remained unaffected. This phenotype was associated with abrogated phosphorylation of p21-activated kinase (PAK) as well as its substrates LIM domain kinase and cofilin, supporting the hypothesis that the defective F-actin assembly results from increased cofilin activity in Pdk1-deficient MKs. Pdk1-/- BM-MKs developed increased ploidy and exhibited an abnormal ultrastructure with disrupted demarcation membrane system (DMS). Strikingly, Pdk1-/- BM-MKs displayed a pronounced defect in DMS polarization and produced significantly less proplatelets, indicating that PDK1 is critically required for proplatelet formation. In human MKs, genetic PDK1 knockdown resulted in increased maturity but reduced platelet-like particles formation. The present observations reveal a pivotal role of PDK1 in the regulation of MK cytoskeletal dynamics and polarization, proplatelet formation, and thrombopoiesis.
- Published
- 2019
21. An essential role for α4A-tubulin in platelet biogenesis
- Author
-
Alain Van Dorsselaer, Claude Férec, Arnaud Dupuis, Benjamin T. Kile, Christine Schaeffer-Reiss, Carsten Janke, Morgane Batzenschlager, Irina Pleines, François Lanza, Paul Gueguen, Léa Mallo, Anita Eckly, Maria M. Magiera, Jean Marc Strub, Catherine Strassel, Christian Gachet, Sylvie Moog, Jean-Yves Py, Agnès Hovasse, Quentin Kimmerlin, Stephane Chappaz, Natarajan Kathiresan, Biologie et pharmacologie des plaquettes sanguines: hémostase, thrombose, transfusion, Université de Strasbourg (UNISTRA)-EFS-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Curie [Paris], Stress génotoxiques et cancer, Université Paris-Sud - Paris 11 (UP11)-Institut Curie [Paris]-Centre National de la Recherche Scientifique (CNRS), Laboratoire de chimie de coordination (LCC), Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Institut de Chimie de Toulouse (ICT-FR 2599), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), Laboratoire d'Analyses de Biologie Médicale (LABM), Institut National de la Santé et de la Recherche Médicale (INSERM)-EFS Alsace, Département Sciences Analytiques et Interactions Ioniques et Biomoléculaires (DSA-IPHC), Institut Pluridisciplinaire Hubert Curien (IPHC), Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS), Génétique moléculaire et génétique épidémiologique, Université de Brest (UBO)-Institut National de la Santé et de la Recherche Médicale (INSERM), Division of Developmental Immunology (DKBW), University of Basel (Unibas), Etablissement Français du Sang Bretagne, EFS, Institut de Chimie de Toulouse (ICT), Institut de Recherche pour le Développement (IRD)-Université Toulouse III - Paul Sabatier (UT3), Université de Toulouse (UT)-Université de Toulouse (UT)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Institut National Polytechnique (Toulouse) (Toulouse INP), Université de Toulouse (UT)-Institut de Recherche pour le Développement (IRD)-Université Toulouse III - Paul Sabatier (UT3), Université de Toulouse (UT)-Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Centre National de la Recherche Scientifique (CNRS), Institut de Chimie de Toulouse (ICT-FR 2599), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Institut de Recherche pour le Développement (IRD)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS), and Université de Strasbourg (UNISTRA)-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Blood Platelets ,Male ,0301 basic medicine ,Alkylating Agents ,Health, Toxicology and Mutagenesis ,Megakaryocyte differentiation ,Mutation, Missense ,Antigens, CD34 ,Plant Science ,macromolecular substances ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,Microtubules ,Corrections ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Thrombopoiesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Megakaryocyte ,Tubulin ,Microtubule ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,medicine ,Animals ,Humans ,Platelet ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,Cells, Cultured ,Mice, Inbred BALB C ,Ecology ,biology ,Platelet Count ,Chemistry ,Correction ,Thrombocytopenia ,Isotype ,Tissue Donors ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Ethylnitrosourea ,030220 oncology & carcinogenesis ,biology.protein ,Megakaryocytes ,Biogenesis - Abstract
During platelet biogenesis, microtubules (MTs) are arranged into submembranous structures (the marginal band) that encircle the cell in a single plane. This unique MT array has no equivalent in any other mammalian cell, and the mechanisms responsible for this particular mode of assembly are not fully understood. One possibility is that platelet MTs are composed of a particular set of tubulin isotypes that carry specific posttranslational modifications. Although β1-tubulin is known to be essential, no equivalent roles of α-tubulin isotypes in platelet formation or function have so far been reported. Here, we identify α4A-tubulin as a predominant α-tubulin isotype in platelets. Similar to β1-tubulin, α4A-tubulin expression is up-regulated during the late stages of megakaryocyte differentiation. Missense mutations in the α4A-tubulin gene cause macrothrombocytopenia in mice and humans. Defects in α4A-tubulin lead to changes in tubulin tyrosination status of the platelet tubulin pool. Ultrastructural defects include reduced numbers and misarranged MT coils in the platelet marginal band. We further observed defects in megakaryocyte maturation and proplatelet formation inTuba4a-mutant mice. We have, thus, discovered an α-tubulin isotype with specific and essential roles in platelet biogenesis.
- Published
- 2019
22. Rho GTPases and their downstream effectors in megakaryocyte biology
- Author
-
Deya Cherpokova, Irina Pleines, and Markus Bender
- Subjects
0301 basic medicine ,Blood Platelets ,rho GTP-Binding Proteins ,RHOA ,G protein ,RAC1 ,CDC42 ,030204 cardiovascular system & hematology ,Biology ,Thrombopoiesis ,03 medical and health sciences ,0302 clinical medicine ,Megakaryocyte ,medicine ,Animals ,Humans ,Cytoskeleton ,Hematology ,General Medicine ,Cell biology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Knockout mouse ,biology.protein ,Megakaryocytes ,Biomarkers ,Signal Transduction - Abstract
Megakaryocytes differentiate from hematopoietic stem cells in the bone marrow. The transition of megakaryocytes to platelets is a complex process. Thereby, megakaryocytes extend proplatelets into sinusoidal blood vessels, where the proplatelets undergo fission to release platelets. Defects in platelet production can lead to a low platelet count (thrombocytopenia) with increased bleeding risk. Rho GTPases comprise a family of small signaling G proteins that have been shown to be master regulators of the cytoskeleton controlling many aspects of intracellular processes. The generation of Pf4-Cre transgenic mice was a major breakthrough that enabled studies in megakaryocyte-/platelet-specific knockout mouse lines and provided new insights into the central regulatory role of Rho GTPases in megakaryocyte maturation and platelet production. In this review, we will summarize major findings on the role of Rho GTPases in megakaryocyte biology with a focus on mouse lines in which knockout strategies have been applied to study the function of the best-characterized members Rac1, Cdc42 and RhoA and their downstream effector proteins.
- Published
- 2018
23. Regulation of cell proliferation by ERK and signal-dependent nuclear translocation of ERK is dependent on Tm5NM1-containing actin filaments
- Author
-
Anthony J. Kee, Vanessa B. Sequeira, Peter W. Gunning, Elizabeth A. Musgrove, Benjamin T. Kile, Bin Wang, Thomas Fath, Christine A. Lucas, Galina Schevzov, Edna C. Hardeman, Jason D. Coombes, Rony Seger, Alexandra Cretu, Justine R. Stehn, Richard K. Assoian, Jeff Hook, Tamar Hanoch, and Irina Pleines
- Subjects
Male ,MAPK/ERK pathway ,endocrine system ,MAP Kinase Signaling System ,Active Transport, Cell Nucleus ,Mice, Transgenic ,macromolecular substances ,Tropomyosin ,Mitogen-activated protein kinase kinase ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Animals ,Humans ,Phosphorylation ,Casein Kinase II ,Molecular Biology ,Cytoskeleton ,Actin ,Cell Proliferation ,030304 developmental biology ,Mice, Knockout ,Mitogen-Activated Protein Kinase Kinases ,0303 health sciences ,Cell growth ,Articles ,Cell Biology ,Actin cytoskeleton ,Molecular biology ,Cell biology ,Mice, Inbred C57BL ,Actin Cytoskeleton ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer cell ,Casein kinase 2 - Abstract
Tropomyosin Tm5NM1 regulates cell proliferation and organ size. It mediates this effect by regulating the interaction of pERK and Imp7, leading to the regulation of pERK nuclear translocation. This demonstrates a role for a specific population of actin filaments in regulating a critical step in the MAPK/ERK signaling pathway., ERK-regulated cell proliferation requires multiple phosphorylation events catalyzed first by MEK and then by casein kinase 2 (CK2), followed by interaction with importin7 and subsequent nuclear translocation of pERK. We report that genetic manipulation of a core component of the actin filaments of cancer cells, the tropomyosin Tm5NM1, regulates the proliferation of normal cells both in vitro and in vivo. Mouse embryo fibroblasts (MEFs) lacking Tm5NM1, which have reduced proliferative capacity, are insensitive to inhibition of ERK by peptide and small-molecule inhibitors, indicating that ERK is unable to regulate proliferation of these knockout (KO) cells. Treatment of wild-type MEFs with a CK2 inhibitor to block phosphorylation of the nuclear translocation signal in pERK resulted in greatly decreased cell proliferation and a significant reduction in the nuclear translocation of pERK. In contrast, Tm5NM1 KO MEFs, which show reduced nuclear translocation of pERK, were unaffected by inhibition of CK2. This suggested that it is nuclear translocation of CK2-phosphorylated pERK that regulates cell proliferation and this capacity is absent in Tm5NM1 KO cells. Proximity ligation assays confirmed a growth factor–stimulated interaction of pERK with Tm5NM1 and that the interaction of pERK with importin7 is greatly reduced in the Tm5NM1 KO cells.
- Published
- 2015
24. Intrinsic apoptosis circumvents the functional decline of circulating platelets but does not cause the storage lesion
- Author
-
Simone M. Schoenwaelder, Shaun P. Jackson, Jonathan P. Bernardini, Rachael M. Lane, Benjamin T. Kile, Amanda E. Au, Maria Kauppi, Marion Lebois, Emma C. Josefsson, Jason Corbin, Katya J. Henley, Donald Metcalf, Pradnya Gangatirkar, Sarah Ellis, Ping Cannon, Imala Alwis, Kate E. Jarman, and Irina Pleines
- Subjects
0301 basic medicine ,Blood Platelets ,Male ,Bleeding Time ,Genotype ,Cell Survival ,Immunology ,Population ,bcl-X Protein ,Apoptosis ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Bcl-2-associated X protein ,Animals ,Platelet ,Hemostatic function ,education ,Blood Coagulation ,bcl-2-Associated X Protein ,Mice, Knockout ,education.field_of_study ,biology ,Intrinsic apoptosis ,Cell Biology ,Hematology ,Phosphatidylserine ,Cell biology ,Blood Cell Count ,Mitochondria ,030104 developmental biology ,chemistry ,Hemostasis ,Caspases ,biology.protein ,Female ,Disease Susceptibility ,Biomarkers ,Signal Transduction - Abstract
The circulating life span of blood platelets is regulated by the prosurvival protein BCL-XL It restrains the activity of BAK and BAX, the essential prodeath mediators of intrinsic apoptosis. Disabling the platelet intrinsic apoptotic pathway in mice by deleting BAK and BAX results in a doubling of platelet life span and concomitant thrombocytosis. Apoptotic platelets expose phosphatidylserine (PS) via a mechanism that is distinct from that driven by classical agonists. Whether there is any role for apoptotic PS in platelet function in vivo, however, is unclear. Apoptosis has also been associated with the platelet storage lesion (PSL), the constellation of biochemical deteriorations that occur during blood bank storage. In this study, we investigated the role of BAK/BAX-mediated apoptosis in hemostasis and thrombosis and in the development of the PSL. We show that although intrinsic apoptosis is rapidly induced during storage at 37°C, it is not detected when platelets are kept at the standard storage temperature of 22°C. Remarkably, loss of BAK and BAX did not prevent the development of the PSL at either temperature. BAK/BAX-deficient mice exhibited increased bleeding times and unstable thrombus formation. This phenotype was not caused by impaired PS exposure, but was associated with a defect in granule release from aged platelets. Strikingly, rejuvenation of BAK/BAX-deficient platelets in vivo completely rescued the observed hemostatic defects. Thus, apoptotic culling of old platelets from the bloodstream is essential to maintain a functional, hemostatically reactive platelet population. Inhibiting intrinsic apoptosis in blood banked platelets is unlikely to yield significant benefit.
- Published
- 2017
25. CK2β regulates thrombopoiesis and Ca
- Author
-
Patrick, Münzer, Britta, Walker-Allgaier, Sascha, Geue, Friederike, Langhauser, Eva, Geuss, David, Stegner, Katja, Aurbach, Daniela, Semeniak, Madhumita, Chatterjee, Irene, Gonzalez Menendez, Melanie, Märklin, Leticia, Quintanilla-Martinez, Helmut R, Salih, David W, Litchfield, Thierry, Buchou, Christoph, Kleinschnitz, Florian, Lang, Bernhard, Nieswandt, Irina, Pleines, Harald, Schulze, Meinrad, Gawaz, and Oliver, Borst
- Subjects
Blood Platelets ,Mice, Knockout ,Mice ,Animals ,Thrombosis ,Calcium Signaling ,Casein Kinase II ,Platelet Activation ,Megakaryocytes ,Peptide Fragments ,Thrombopoiesis - Abstract
Platelets, anucleated megakaryocyte (MK)-derived cells, play a major role in hemostasis and arterial thrombosis. Although protein kinase casein kinase 2 (CK2) is readily detected in MKs and platelets, the impact of CK2-dependent signaling on MK/platelet (patho-)physiology has remained elusive. The present study explored the impact of the CK2 regulatory β-subunit on platelet biogenesis and activation. MK/platelet-specific genetic deletion of CK2β (
- Published
- 2017
26. A Cdc42/RhoA regulatory circuit downstream of glycoprotein Ib guides transendothelial platelet biogenesis
- Author
-
Judith M.M. van Eeuwijk, Ute Modlich, Alan T. Nurden, Katja Aurbach, Zaverio M. Ruggeri, Frank Gerner, Tobias Heib, Cord Brakebusch, Frédérique Gaits-Iacovoni, Simon Stritt, Paquita Nurden, Adrien Antkowiak, Harald Schulze, Jerry Ware, Niels Heinz, Ayesha A Baig, Michael Popp, Irina Pleines, Katrin G. Heinze, Bernhard Nieswandt, Deya Cherpokova, Georg Krohne, Oguzhan Angay, Sebastian Dütting, Maximilian G. Gorelashvili, and David Stegner
- Subjects
Blood Platelets ,0301 basic medicine ,RHOA ,Science ,General Physics and Astronomy ,CDC42 ,GTPase ,030204 cardiovascular system & hematology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell polarity ,Animals ,Humans ,Platelet ,ddc:610 ,cdc42 GTP-Binding Protein ,Mice, Knockout ,Multidisciplinary ,biology ,Cell Polarity ,Endothelial Cells ,Platelet Glycoprotein GPIb-IX Complex ,General Chemistry ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Glycoprotein Ib ,Cdc42 GTP-Binding Protein ,biology.protein ,Female ,rhoA GTP-Binding Protein ,Megakaryocytes - Abstract
Blood platelets are produced by large bone marrow (BM) precursor cells, megakaryocytes (MKs), which extend cytoplasmic protrusions (proplatelets) into BM sinusoids. The molecular cues that control MK polarization towards sinusoids and limit transendothelial crossing to proplatelets remain unknown. Here, we show that the small GTPases Cdc42 and RhoA act as a regulatory circuit downstream of the MK-specific mechanoreceptor GPIb to coordinate polarized transendothelial platelet biogenesis. Functional deficiency of either GPIb or Cdc42 impairs transendothelial proplatelet formation. In the absence of RhoA, increased Cdc42 activity and MK hyperpolarization triggers GPIb-dependent transmigration of entire MKs into BM sinusoids. These findings position Cdc42 (go-signal) and RhoA (stop-signal) at the centre of a molecular checkpoint downstream of GPIb that controls transendothelial platelet biogenesis. Our results may open new avenues for the treatment of platelet production disorders and help to explain the thrombocytopenia in patients with Bernard–Soulier syndrome, a bleeding disorder caused by defects in GPIb-IX-V., Platelets derive from large precursor cells (megakaryocytes) in the bone marrow. Dütting et al. show that megakaryocyte polarization and platelet biogenesis in the bone-marrow sinusoids are directed by adhesion receptor GPIb signalling and resulting balanced antagonism between RhoA (stop-signal) and Cdc42 (go-signal).
- Published
- 2017
27. RhoA/ROCK guides NMII on the way to MK polyploidy
- Author
-
Irina Pleines and Bernhard Nieswandt
- Subjects
0301 basic medicine ,Gene isoform ,RHOA ,Cell division ,Immunology ,Biochemistry ,Polyploidy ,03 medical and health sciences ,Megakaryocyte ,Nonmuscle myosin ,medicine ,Humans ,Cleavage furrow ,Mitosis ,rho-Associated Kinases ,biology ,Chemistry ,DNA replication ,Cell Biology ,Hematology ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,rhoA GTP-Binding Protein - Abstract
A unique feature of megakaryocyte maturation is the switch from mitosis to replication of DNA without cell division, a process termed endomitosis. In this issue of Blood, Roy et al elegantly demonstrate that RhoA/ROCK signaling is critical for the differential activity and localization of nonmuscle myosin (NM) IIA and IIB isoforms at the megakaryocyte cleavage furrow, a key step in the induction of endomitosis.1
- Published
- 2016
28. Defective tubulin organization and proplatelet formation in murine megakaryocytes lacking Rac1 and Cdc42
- Author
-
Imke Meyer, Najet Debili, Harald Schulze, Sebastian Dütting, Anita Eckly, Cord Brakebusch, Christian Gachet, Bernhard Nieswandt, Martina Morowski, Deya Cherpokova, Georg Krohne, and Irina Pleines
- Subjects
rac1 GTP-Binding Protein ,Mice, 129 Strain ,RHOA ,Blotting, Western ,Immunology ,macromolecular substances ,Microtubules ,Biochemistry ,Abnormal platelet morphology ,Mice ,Microscopy, Electron, Transmission ,Tubulin ,Microtubule ,Animals ,Platelet ,Pseudopodia ,cdc42 GTP-Binding Protein ,Cytoskeleton ,Megakaryocyte Progenitor Cells ,Mice, Knockout ,Hemostasis ,Microscopy, Confocal ,biology ,Thrombosis ,Cell Biology ,Hematology ,Thrombocytopenia ,Cell biology ,Mice, Inbred C57BL ,Cdc42 GTP-Binding Protein ,Microscopy, Electron, Scanning ,biology.protein ,Megakaryocytes - Abstract
Blood platelets are anuclear cell fragments that are essential for blood clotting. Platelets are produced by bone marrow megakaryocytes (MKs), which extend protrusions, or so-called proplatelets, into bone marrow sinusoids. Proplatelet formation requires a profound reorganization of the MK actin and tubulin cytoskeleton. Rho GTPases, such as RhoA, Rac1, and Cdc42, are important regulators of cytoskeletal rearrangements in platelets; however, the specific roles of these proteins during platelet production have not been established. Using conditional knockout mice, we show here that Rac1 and Cdc42 possess redundant functions in platelet production and function. In contrast to a single-deficiency of either protein, a double-deficiency of Rac1 and Cdc42 in MKs resulted in macrothrombocytopenia, abnormal platelet morphology, and impaired platelet function. Double-deficient bone marrow MKs matured normally in vivo but displayed highly abnormal morphology and uncontrolled fragmentation. Consistently, a lack of Rac1/Cdc42 virtually abrogated proplatelet formation in vitro. Strikingly, this phenotype was associated with severely defective tubulin organization, whereas actin assembly and structure were barely affected. Together, these results suggest that the combined action of Rac1 and Cdc42 is crucial for platelet production, particularly by regulating microtubule dynamics.
- Published
- 2013
29. Regulation of Megakaryocyte and Platelet Survival
- Author
-
Amanda E. Au, Marion Lebois, Irina Pleines, and Emma C. Josefsson
- Subjects
Programmed cell death ,medicine.anatomical_structure ,Megakaryocyte ,Apoptosis ,Chemistry ,Hemostasis ,Intrinsic apoptosis ,medicine ,Platelet ,Bone marrow ,Wound healing ,Cell biology - Abstract
Platelets play vital roles in hemostasis, wound healing, and a range of other processes [1]. Their number is tightly controlled within narrow physiological ranges. This occurs through a dynamic balance between platelet production and consumption/clearance rates, so as to ensure that the total platelet mass remains constant. Megakaryocytes in the bone marrow produce around 100 billion platelets per day. In a healthy individual, the majority of platelets are not consumed by hemostatic processes. It is therefore imperative that platelet lifespan is strictly regulated. Recent work has demonstrated that the survival of megakaryocytes and platelets is controlled by programmed cell death, apoptosis [2]. Both cell types possess a classical Bak- and Bax-mediated intrinsic, mitochondrial, apoptosis pathway that must be restrained in order for them to develop and survive. In addition, recent work has revealed that the glycosylation state of platelet surface proteins is an indicator of platelet age and that aged, desialylated platelets stimulate platelet production [3].
- Published
- 2016
30. Defective diacylglycerol‐induced Ca2+entry but normal agonist‐induced activation responses in TRPC6‐deficient mouse platelets
- Author
-
Irina Pleines, Alexander Dietrich, Bernhard Nieswandt, Ina Thielmann, Attila Braun, G. Ramanathan, Shuchi Gupta, Frauke May, Christine Mannhalter, and David Varga-Szabo
- Subjects
Blood Platelets ,medicine.medical_specialty ,Time Factors ,ORAI1 Protein ,Platelet Glycoprotein GPIIb-IIIa Complex ,Ferric Compounds ,TRPC6 ,Diglycerides ,Mice ,Transient receptor potential channel ,Chlorides ,Internal medicine ,TRPC6 Cation Channel ,medicine ,Animals ,Platelet ,Protease-activated receptor ,Calcium Signaling ,RNA, Messenger ,Stromal Interaction Molecule 1 ,Platelet activation ,TRPC ,TRPC Cation Channels ,Diacylglycerol kinase ,Mice, Knockout ,Membrane Glycoproteins ,ORAI1 ,Chemistry ,Secretory Vesicles ,Thrombin ,Thrombosis ,Hematology ,Platelet Activation ,Cell biology ,Adenosine Diphosphate ,Mice, Inbred C57BL ,Disease Models, Animal ,C-Reactive Protein ,Endocrinology ,Gene Expression Regulation ,Calcium Channels - Abstract
Summary. Background: Platelet adhesion, activation and aggregation at sites of vascular injury are essential processes for primary hemostasis. Elevation of the intracellular Ca2+ concentration is a central event in platelet activation but the underlying mechanisms are not fully understood. Store-operated calcium entry (SOCE) through Orai1 was shown to be the main Ca2+ influx pathway in murine platelets, but there are additional non-store-operated Ca2+ (non-SOC) and receptor operated Ca2+ (ROC) channels expressed in the platelet plasma membrane.Objective: Canonical transient receptor potential (TRPC) channel 6 is found both in human and murine platelets and has been proposed to mediate diacylglycerol (DAG) activated ROCE but also a role in the regulation of SOCE has been suggested.Methods: To investigate the function of TRPC6 in platelet Ca2+ signaling and activation, we analyzed platelets from mice deficient in TRPC6 using a wide range of in vitro and in vivo assays.Results: In the mutant platelets, DAG activated Ca2+ influx was found to be abolished. However, this did not significantly affect SOCE or agonist induced Ca2+ responses. Platelet function in vitro and in vivo was also unaltered in the absence of TRPC6.Conclusion: Our results indicate that DAG activated ROCE is mediated exclusively by TRPC6 in murine platelets, but this Ca2+ influx has no major functional relevance for hemostasis and thrombosis. Further, in contrast to previous suggestions, based on studies with human platelets, TRPC6 appears to play an insignificant role in the regulation of SOCE in murine platelets.
- Published
- 2012
31. ADF/n-cofilin–dependent actin turnover determines platelet formation and sizing
- Author
-
Irina Pleines, Walter Witke, John H. Hartwig, Christian Gachet, Antje Gohla, Christine B. Gurniak, Anita Eckly, Georg Krohne, Bernhard Nieswandt, Margitta Elvers, Elisabeth Jeanclos, Shuchi Gupta, and Markus Bender
- Subjects
Blood Platelets ,Cofilin 1 ,Time Factors ,Cell Survival ,Blotting, Western ,Immunology ,Arp2/3 complex ,macromolecular substances ,Biology ,Biochemistry ,Mice ,Microscopy, Electron, Transmission ,Animals ,Platelet ,Cell Shape ,Cytoskeleton ,Actin ,Cell Size ,Mice, Knockout ,Platelet Count ,Thrombin ,Fibrinogen ,Actin remodeling ,Cell Biology ,Hematology ,Cofilin ,Microtubule sliding ,Actin cytoskeleton ,Actins ,Cell biology ,Actin Cytoskeleton ,Destrin ,Splenomegaly ,Microscopy, Electron, Scanning ,biology.protein ,MDia1 ,Megakaryocytes - Abstract
The cellular and molecular mechanisms orchestrating the complex process by which bone marrow megakaryocytes form and release platelets remain poorly understood. Mature megakaryocytes generate long cytoplasmic extensions, proplatelets, which have the capacity to generate platelets. Although microtubules are the main structural component of proplatelets and microtubule sliding is known to drive proplatelet elongation, the role of actin dynamics in the process of platelet formation has remained elusive. Here, we tailored a mouse model lacking all ADF/n-cofilin–mediated actin dynamics in megakaryocytes to specifically elucidate the role of actin filament turnover in platelet formation. We demonstrate, for the first time, that in vivo actin filament turnover plays a critical role in the late stages of platelet formation from megakaryocytes and the proper sizing of platelets in the periphery. Our results provide the genetic proof that platelet production from megakaryocytes strictly requires dynamic changes in the actin cytoskeleton.
- Published
- 2010
32. Platelet hyperreactivity and a prothrombotic phenotype in mice with a gain-of-function mutation in phospholipase C gamma 2
- Author
-
P. Yu, R. Pozgaj, Margitta Elvers, J. M. W. Heemskerk, Bernhard Nieswandt, Irina Pleines, Marijke J.E. Kuijpers, Frauke May, Biochemie, and RS: CARIM School for Cardiovascular Diseases
- Subjects
Blood Platelets ,Heterozygote ,Platelet Aggregation ,integrin ,Integrin ,Clot retraction ,Biology ,Collagen receptor ,Mice ,PLC gamma 2 ,Animals ,Platelet ,Platelet activation ,thrombosis ,Diacylglycerol kinase ,platelet ,Phospholipase C gamma ,Hematology ,Flow Cytometry ,Cell biology ,Phenotype ,Biochemistry ,Mutation ,biology.protein ,Signal transduction ,GPVI ,signaling - Abstract
Summary. Background: Agonist-induced platelet activation involves different signaling pathways leading to the activation of phospholipase C (PLC) β or PLCγ2. Activated PLC produces inositol 1,4,5-trisphosphate and diacylglycerol, which trigger Ca2+ mobilization and the activation of protein kinase C, respectively. PLCβ is activated downstream of Gq-coupled receptors for soluble agonists with only short interaction times in flowing blood. In contrast, PLCγ2 becomes activated downstream of receptors that interact with immobilized ligands such as the collagen receptor glycoprotein (GP) VI or activated integrins. Objective and methods: We speculated that PLCγ2 activity might be optimized for sustained but submaximal signaling to control relatively slow platelet responses. To test this hypothesis, we analyzed platelets from mice heterozygous for a gain-of-function mutation in the Plcg2 gene (Plcg2Ali5/+). Results: Plcg2Ali5/+ platelets showed enhanced Ca2+ mobilization, integrin activation, granule secretion and phosphatidylserine exposure upon GPVI or C-type lectin-like receptor-2 stimulation. Furthermore, integrin αIIbβ3 outside-in signaling was markedly enhanced in the mutant platelets, as shown by accelerated spreading on different matrices and faster clot retraction. These defects translated into virtually unlimited thrombus formation on collagen under flow in vitro and a prothrombotic phenotype in vivo. Conclusions: These results demonstrate that the enzymatic activity of PLCγ2 is tightly regulated to ensure efficient but limited platelet activation at sites of vascular injury.
- Published
- 2010
33. Factor XIIa Inhibitor Recombinant Human Albumin Infestin-4 Abolishes Occlusive Arterial Thrombus Formation Without Affecting Bleeding
- Author
-
Christoph Kleinschnitz, Guido Stoll, Ulrich Kronthaler, Ina Hagedorn, Gerhard Dickneite, Irina Pleines, Stefan Schmidbauer, and Bernhard Nieswandt
- Subjects
DNA, Complementary ,Factor XIIa ,Arterial Occlusive Diseases ,Coagulation Factor XII ,Pharmacology ,Fibrin ,Brain Ischemia ,Mice ,Albumins ,Physiology (medical) ,medicine ,Animals ,Humans ,Thrombus ,Hemostasis ,Factor XII ,medicine.diagnostic_test ,biology ,business.industry ,Infarction, Middle Cerebral Artery ,Rats, Inbred Strains ,Thrombosis ,medicine.disease ,Mice, Mutant Strains ,Recombinant Proteins ,Rats ,Mice, Inbred C57BL ,Coagulation ,Immunology ,biology.protein ,Insect Proteins ,Partial Thromboplastin Time ,Cardiology and Cardiovascular Medicine ,business ,Partial thromboplastin time - Abstract
Background— Blood coagulation is a tightly regulated process of sequentially activated serine proteases culminating in fibrin formation, which is critical for limiting posttraumatic blood loss but also may contribute to acute thrombotic diseases, most notably myocardial infarction and stroke. Recent studies with factor XII–deficient mice revealed that the factor XII–induced intrinsic coagulation pathway is essential for pathological thrombus formation but dispensable for hemostasis. Consequently, these findings led to the hypothesis that factor XII could be a promising pharmacological target for safe antithrombotic therapy. Methods and Results— The complementary DNA of the previously described factor XIIa inhibitor Infestin-4, cloned from the midgut of Triatoma infestans , was fused to recombinant human albumin (rHA) and analyzed in vitro. The resulting protein rHA-Infestin-4 specifically inhibits factor XIIa and causes prolonged activated partial thromboplastin time in human, mouse, and rat plasma. To assess its inhibitory potency in vivo, mice and rats were injected with rHA-Infestin-4 and challenged in pathological thrombus formation models. In addition, bleeding assays were performed. rHA-Infestin-4 completely abolished occlusive arterial thrombus formation in mice and rats while leaving hemostasis fully intact. Furthermore, rHA-Infestin-4 was highly protective in a murine model of ischemic stroke. Conclusion— These results identify rHA-Infestin-4 as a promising agent to achieve powerful protection from ischemic cardiovascular and cerebrovascular events without affecting hemostasis.
- Published
- 2010
34. CLEC-2 is an essential platelet-activating receptor in hemostasis and thrombosis
- Author
-
Frauke May, Ina Hagedorn, Irina Pleines, Markus Bender, Timo Vögtle, Johannes Eble, Margitta Elvers, and Bernhard Nieswandt
- Subjects
Blood Platelets ,medicine.medical_specialty ,Platelet Aggregation ,Immunology ,Excipient ,Pharmacology ,Biochemistry ,Antibodies ,Mice ,Platelet Adhesiveness ,Internal medicine ,Animals ,Humans ,Medicine ,Lectins, C-Type ,Platelet ,Thrombus ,Receptor ,Hematology ,business.industry ,Activator (genetics) ,Thrombosis ,Cell Biology ,medicine.disease ,Extracellular Matrix ,Hemostasis ,Endothelium, Vascular ,business ,medicine.drug - Abstract
The present invention relates to a pharmaceutical composition comprising an inhibitor of C-type lectin-like receptor 2 (CLEC-2) or an inhibitor of an activator of CLEC-2, each of which induces an irreversible inactivation or degradation of CLEC-2 on platelets, and optionally a pharmaceutically acceptable carrier, excipient and/or diluent. Furthermore, the present invention relates to an inhibitor of C-type lectin-like receptor 2 (CLEC-2) or an inhibitor of an activator of CLEC-2, each of which induces an irreversible inactivation or degradation of CLEC-2 on platelets, for use in treating or preventing a disorder related to venous or arterial thrombus formation and to the use of such an inhibitor as a lead compound for developing a drug for treating or preventing a disorder related to venous or arterial thrombus formation. The present invention also relates to a method of treating and/or preventing a disorder related to venous or arterial thrombus formation comprising administering a pharmaceutically effective amount of an inhibitor of C-type lectin-like receptor 2 (CLEC-2) or an inhibitor of an activator of CLEC-2, each of which induces an irreversible inactivation or degradation of CLEC-2 on platelets, to a subject in need thereof. The present invention further relates to methods of identifying a compound suitable as a lead compound and/or as a medicament for the treatment and/or prevention of a disorder related to venous or arterial thrombus formation.
- Published
- 2009
35. Orai1 (CRACM1) is the platelet SOC channel and essential for pathological thrombus formation
- Author
-
Attila Braun, David Varga-Szabo, Christoph Kleinschnitz, Irina Pleines, Markus Bender, Madeleine Austinat, Michael Bösl, Guido Stoll, and Bernhard Nieswandt
- Subjects
Blood Platelets ,Pathology ,medicine.medical_specialty ,ORAI1 Protein ,Immunology ,Myocardial Ischemia ,Ischemia ,Excipient ,Pharmacology ,Biochemistry ,Brain Ischemia ,Pathogenesis ,Mice ,medicine ,Animals ,Humans ,Platelet ,Stromal Interaction Molecule 1 ,Thrombus ,Mice, Knockout ,Membrane Glycoproteins ,business.industry ,ORAI1 ,Activator (genetics) ,Body Weight ,Thrombosis ,Cell Biology ,Hematology ,medicine.disease ,Mice, Inbred C57BL ,Calcium ,Female ,Calcium Channels ,business ,medicine.drug - Abstract
Platelet activation and aggregation at sites of vascular injury are essential for primary hemostasis, but are also major pathomechanisms underlying myocardial infarction and stroke. Changes in [Ca2+]i are a central step in platelet activation. In nonexcitable cells, receptor-mediated depletion of intracellular Ca2+ stores triggers Ca2+ entry through store-operated calcium (SOC) channels. STIM1 has been identified as an endoplasmic reticulum (ER)–resident Ca2+ sensor that regulates store-operated calcium entry (SOCE) in immune cells and platelets, but the identity of the platelet SOC channel has remained elusive. Orai1 (CRACM1) is the recently discovered SOC (CRAC) channel in T cells and mast cells but its role in mammalian physiology is unknown. Here we report that Orai1 is strongly expressed in human and mouse platelets. To test its role in blood clotting, we generated Orai1-deficient mice and found that their platelets display severely defective SOCE, agonist-induced Ca2+ responses, and impaired activation and thrombus formation under flow in vitro. As a direct consequence, Orai1 deficiency in mice results in resistance to pulmonary thromboembolism, arterial thrombosis, and ischemic brain infarction, but only mild bleeding time prolongation. These results establish Orai1 as the long-sought platelet SOC channel and a crucial mediator of ischemic cardiovascular and cerebrovascular events.
- Published
- 2009
36. Rac1 is essential for phospholipase C-γ2 activation in platelets
- Author
-
Cord Brakebusch, David Varga-Szabo, Frauke May, Margitta Elvers, Irina Pleines, Miroslava Pozgajova, Bernhard Nieswandt, Anna Chrostek-Grashoff, and Amrei Strehl
- Subjects
Blood Platelets ,rac1 GTP-Binding Protein ,Platelet Aggregation ,Physiology ,Clinical Biochemistry ,Platelet Membrane Glycoproteins ,Biology ,Mice ,chemistry.chemical_compound ,Physiology (medical) ,Immunoreceptor tyrosine-based activation motif ,Animals ,Lectins, C-Type ,Protease-activated receptor ,Platelet ,Platelet activation ,Mice, Knockout ,Phospholipase C ,Phospholipase C gamma ,Thrombosis ,Tyrosine phosphorylation ,Platelet Activation ,Cell biology ,Adenosine Diphosphate ,Poly I-C ,Biochemistry ,chemistry ,15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid ,Calcium ,GPVI ,Signal transduction - Abstract
Platelet activation at sites of vascular injury is triggered through different signaling pathways leading to activation of phospholipase (PL) Cbeta or PLCgamma2. Active PLCs trigger Ca(2+) mobilization and entry, which is a prerequisite for adhesion, secretion, and thrombus formation. PLCbeta isoenzymes are activated downstream of G protein-coupled receptors (GPCRs), whereas PLCgamma2 is activated downstream of immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptors, such as the major platelet collagen receptor glycoprotein (GP) VI or CLEC-2. The mechanisms underlying PLC regulation are not fully understood. An involvement of small GTPases of the Rho family (Rho, Rac, Cdc42) in PLC activation has been proposed but this has not been investigated in platelets. We here show that murine platelets lacking Rac1 display severely impaired GPVI- or CLEC-2-dependent activation and aggregation. This defect was associated with impaired production of inositol 1,4,5-trisphosphate (IP(3)) and intracellular calcium mobilization suggesting inappropriate activation of PLCgamma2 despite normal tyrosine phosphorylation of the enzyme. Rac1 ( -/- ) platelets displayed defective thrombus formation on collagen under flow conditions which could be fully restored by co-infusion of ADP and the TxA(2) analog U46619, indicating that impaired GPVI-, but not G-protein signaling, was responsible for the observed defect. In line with this, Rac1 ( -/- ) mice were protected in two collagen-dependent arterial thrombosis models. Together, these results demonstrate that Rac1 is essential for ITAM-dependent PLCgamma2 activation in platelets and that this is critical for thrombus formation in vivo.
- Published
- 2008
37. Loss of talin1 in platelets abrogates integrin activation, platelet aggregation, and thrombus formation in vitro and in vivo
- Author
-
Markus Moser, David Varga-Szabo, Reinhard Fässler, David R. Critchley, Susan J. Monkley, Irina Pleines, and Bernhard Nieswandt
- Subjects
Blood Platelets ,Talin ,Cytoplasm ,Integrins ,Platelet Aggregation ,Immunology ,Integrin ,Mice, Transgenic ,030204 cardiovascular system & hematology ,Ferric Compounds ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Platelet Adhesiveness ,Chlorides ,In vivo ,Platelet adhesiveness ,medicine ,Immunology and Allergy ,Animals ,Platelet ,Thrombus ,Receptor ,030304 developmental biology ,0303 health sciences ,Microscopy ,biology ,Models, Genetic ,Chemistry ,Brief Definitive Report ,Thrombosis ,Cell Biology ,medicine.disease ,Flow Cytometry ,Talin binding ,3. Good health ,Cell biology ,Arterioles ,Biochemistry ,Hemostasis ,biology.protein ,Brief Definitive Reports ,030215 immunology - Abstract
Platelet adhesion and aggregation at sites of vascular injury are essential for normal hemostasis but may also lead to pathological thrombus formation, causing diseases such as myocardial infarction or stroke. Heterodimeric receptors of the integrin family play a central role in the adhesion and aggregation of platelets. In resting platelets, integrins exhibit a low affinity state for their ligands, and they shift to a high affinity state at sites of vascular injury. It has been proposed that direct binding of the cytoskeletal protein talin1 to the cytoplasmic domain of the integrin β subunits is necessary and sufficient to trigger the activation of integrins to this high affinity state, but direct in vivo evidence in support of this hypothesis is still lacking. Here, we show that platelets from mice lacking talin1 are unable to activate integrins in response to all known major platelet agonists while other cellular functions are still preserved. As a consequence, mice with talin-deficient platelets display a severe hemostatic defect and are completely resistant to arterial thrombosis. Collectively, these experiments demonstrate that talin is required for inside-out activation of platelet integrins in hemostasis and thrombosis.
- Published
- 2007
38. Expansion of the neonatal platelet mass is achieved via an extension of platelet lifespan
- Author
-
Chaitanya Chavda, Marlyse A. Debrincat, Benjamin T. Kile, Haley E. Ramsey, Peter Veng-Pedersen, Karin M. Hoffmeister, Sihem Ait-Oudhia, Renata Grozovsky, Zhi-Jian Liu, Irina Pleines, Donald E. Mager, Zhongbo Hu, Joseph E. Italiano, Martha Sola-Visner, and Emma C. Josefsson
- Subjects
Blood Platelets ,medicine.medical_specialty ,Cell Survival ,Immunology ,Plenary Paper ,Blood volume ,Apoptosis ,Biology ,Biochemistry ,Piperazines ,Thrombopoiesis ,Nitrophenols ,Mice ,Internal medicine ,medicine ,Animals ,Humans ,Platelet ,Mean platelet volume ,Fetus ,Sulfonamides ,Platelet Count ,Biphenyl Compounds ,Infant, Newborn ,Cell Biology ,Hematology ,Biphenyl compound ,Haematopoiesis ,medicine.anatomical_structure ,Endocrinology ,Animals, Newborn ,Liver ,Proto-Oncogene Proteins c-bcl-2 ,Bone marrow ,Mean Platelet Volume ,Megakaryocytes ,Spleen - Abstract
The fetal/neonatal hematopoietic system must generate enough blood cells to meet the demands of rapid growth. This unique challenge might underlie the high incidence of thrombocytopenia among preterm neonates. In this study, neonatal platelet production and turnover were investigated in newborn mice. Based on a combination of blood volume expansion and increasing platelet counts, the platelet mass increased sevenfold during the first 2 weeks of murine life, a time during which thrombopoiesis shifted from liver to bone marrow. Studies applying in vivo biotinylation and mathematical modeling showed that newborn and adult mice had similar platelet production rates, but neonatal platelets survived 1 day longer in circulation. This prolonged lifespan fully accounted for the rise in platelet counts observed during the second week of murine postnatal life. A study of pro-apoptotic and anti-apoptotic Bcl-2 family proteins showed that neonatal platelets had higher levels of the anti-apoptotic protein Bcl-2 and were more resistant to apoptosis induced by the Bcl-2/Bcl-xL inhibitor ABT-737 than adult platelets. However, genetic ablation or pharmacologic inhibition of Bcl-2 alone did not shorten neonatal platelet survival or reduce platelet counts in newborn mice, indicating the existence of redundant or alternative mechanisms mediating the prolonged lifespan of neonatal platelets.
- Published
- 2014
39. Interleukin-11 is the dominant IL-6 family cytokine during gastrointestinal tumorigenesis and can be targeted therapeutically
- Author
-
Yeliz Boglev, Adele Preaudet, Matthias Ernst, Rita A. Busuttil, Tracy L Putoczki, Andrea Loving, Brent S. McKenzie, David Horst, Irina Pleines, Oliver M. Sieber, Paul Ziegler, Andrew G. Jarnicki, Nicholas J. Wilson, Stefan Thiem, Ryan Farid, Paul M Nguyen, Kirsten Edwards, Rodney B. Luwor, Benjamin T. Kile, Florian R. Greten, Andrew D. Nash, Joan K. Heath, and Alex Boussioutas
- Subjects
0303 health sciences ,Cancer Research ,medicine.medical_treatment ,Cancer ,Interleukin ,Cell Biology ,Biology ,Glycoprotein 130 ,medicine.disease ,medicine.disease_cause ,Stat3 Signaling Pathway ,3. Good health ,Interleukin 11 ,03 medical and health sciences ,0302 clinical medicine ,Cytokine ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,medicine ,biology.protein ,Interleukin 6 ,Carcinogenesis ,030304 developmental biology - Abstract
SummaryAmong the cytokines linked to inflammation-associated cancer, interleukin (IL)-6 drives many of the cancer “hallmarks” through downstream activation of the gp130/STAT3 signaling pathway. However, we show that the related cytokine IL-11 has a stronger correlation with elevated STAT3 activation in human gastrointestinal cancers. Using genetic mouse models, we reveal that IL-11 has a more prominent role compared to IL-6 during the progression of sporadic and inflammation-associated colon and gastric cancers. Accordingly, in these models and in human tumor cell line xenograft models, pharmacologic inhibition of IL-11 signaling alleviated STAT3 activation, suppressed tumor cell proliferation, and reduced the invasive capacity and growth of tumors. Our results identify IL-11 signaling as a potential therapeutic target for the treatment of gastrointestinal cancers.
- Published
- 2013
- Full Text
- View/download PDF
40. Extended Platelet In Vivo Survival Results in Exhausted Platelets
- Author
-
Irina Pleines, Benjamin T. Kile, Kate E. Jarman, Simone M. Schoenwaelder, Sarah Ellis, Emma C. Josefsson, Jason Corbin, Shaun P. Jackson, Ping Cannon, Amanda E.-L. Au, Marion Lebois, Rachael M. Lane, and Katya J. Henley
- Subjects
medicine.medical_specialty ,Thromboxane ,Immunology ,Degranulation ,Convulxin ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Endocrinology ,Apoptosis ,Internal medicine ,medicine ,Platelet ,Platelet activation ,biological phenomena, cell phenomena, and immunity ,Dense granule ,GPVI - Abstract
Platelet lifespan is limited to 10 days in humans and 5 days in mice. The intrinsic apoptosis pathway regulates the survival of platelets, where the pro-survival protein Bcl-xLrestrains the essential death mediators Bak and Bax (Mason et al., Cell 2007). Hence, platelet lifespan and platelet counts in mice are increased in the absence of Bak and Bak/Bax. While platelet production in mice is normal in the absence of intrinsic apoptosis (Josefsson et al., J Exp Med 2011), the function of these long-lived platelets has not been investigated. In the current study we examined the functional outcome of extended platelet survival. We found that washed platelets from mice with a constitutive deletion of Bak and a platelet-specific deletion of Bax (Bak-/-BaxPf4Δ/Pf4Δ) were fully resistant to apoptosis induced by the BH3-mimetic ABT-737, as demonstrated by lack of phosphatidylserine exposure (binding of AnnexinV) and unaltered mitochondrial membrane potential. Tail bleeding times into 37°C saline, were extended in the absence of either Bak alone or both Bak and Bax. Furthermore, the electrolytic thrombosis model showed that despite normal time to arterial occlusion, the thrombi formed in Bak-/- BaxPf4Δ/Pf4Δ mice were unstable, a trend also observed in Bak-/-Baxfl/fl mice. The formation of stable thrombi is dependent on the release of secondary agonists, such as ADP and Thromboxane, from activated platelets. To investigate potential defects in platelet signaling pathways in the absence of Bax and/or Bak, we performed in vitro platelet activation assays. Flow cytometric measurements revealed that activation of the PAR4 receptor (by PAR4-AP) or GPVI (by convulxin) led to reduced integrin activation (JON/A) and degranulation (P-selectin exposure) in the absence of Bak and Bak/Bax, while loss of Bax alone had no effect. In contrast, the response to activation with ADP, which does not induce granule release, was similar in platelets from all genotypes. Similarly, platelet aggregation in response to intermediate concentrations of PAR4-AP was severely reduced in the absence of Bak and Bak/Bax, but normal in response to ADP. We next investigated if abnormal degranulation in response to agonists could explain the aggregation defect. Platelet aggregation was performed with PAR4-AP and the platelet supernatants were collected after centrifugation. Dense granule release (ATP and serotonin) and alpha granule release (PF4) were significantly reduced from platelets deficient in Bak, Bak/Bax, but not Bax alone. Untreated resting platelets of all genotypes contained similar amount of granular proteins (ATP, serotonin and PF4). Hence, altered granule content was not the reason behind the abnormality. We next explored if platelet age was a factor behind the observed functional differences. To be able to directly compare platelet function in Bak/Bax deficient mice and wild-type controls, we synchronized platelet age to ~3 days in all genotypes. Platelets were depleted in vivo by injection of anti-platelet serum (APS). Newly generated platelets were collected at 72 h post injection, a time-point were platelet counts had returned to normal. Remarkably, synchronized platelet age normalized PAR4-AP and convulxin dependent integrin activation (JON/A) and degranulation (P-selectin exposure) in the absence of Bak and Bak/Bax to control levels. Similarly, the platelet aggregation and release defects were rescued. Lastly we investigated if synchronizing platelet age would revert the hemostatic defect of Bak/Bax mice in vivo. We determined tail bleeding times using mice, which were either untreated or depleted of platelets 72 h prior to the experiment. Strikingly, synchronization of platelet age to 3 days rescued the hemostatic defect in Bak-/-BaxPf4Δ/Pf4Δ mice. We conclude that extended platelet survival leads to platelet exhaustion, with reduced ability to mobilize granular release. Our studies suggest that, in the context of blood bank storage, extending platelet survival times by pharmacologically inhibiting apoptosis may result in a hemostatically compromised product. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
41. Megakaryocyte-specific RhoA deficiency causes macrothrombocytopenia and defective platelet activation in hemostasis and thrombosis
- Author
-
Christoph Kleinschnitz, Jolanda van Hengel, Shuchi Gupta, Stefan Offermanns, Irina Pleines, Lidija Chakarova, Georg Krohne, Cord Brakebusch, Ina Hagedorn, Frauke May, and Bernhard Nieswandt
- Subjects
Blood Platelets ,Brain Infarction ,rho GTP-Binding Proteins ,medicine.medical_specialty ,RHOA ,Bleeding Time ,Immunology ,Clot Retraction ,Clot retraction ,Biology ,Fibrinogen ,Biochemistry ,GTP-Binding Protein alpha Subunits, G12-G13 ,Mice ,Megakaryocyte ,Internal medicine ,medicine ,Animals ,Platelet ,Platelet activation ,Calcium Signaling ,Cell Shape ,Cell Size ,Mice, Knockout ,Hemostasis ,Platelet Count ,Thrombosis ,Cell Biology ,Hematology ,Platelet Activation ,Thrombocytopenia ,Kinetics ,Endocrinology ,medicine.anatomical_structure ,biology.protein ,GTP-Binding Protein alpha Subunits, Gq-G11 ,Signal transduction ,rhoA GTP-Binding Protein ,Megakaryocytes ,medicine.drug - Abstract
Vascular injury initiates rapid platelet activation that is critical for hemostasis, but it also may cause thrombotic diseases, such as myocardial infarction or ischemic stroke. Reorganizations of the platelet cytoskeleton are crucial for platelet shape change and secretion and are thought to involve activation of the small GTPase RhoA. In this study, we analyzed the in vitro and in vivo consequences of megakaryocyte- and platelet-specific RhoA gene deletion in mice. We found a pronounced macrothrombocytopenia in RhoA-deficient mice, with platelet counts of approximately half that of wild-type controls. The mutant cells displayed an altered shape but only a moderately reduced life span. Shape change of RhoA-deficient platelets in response to G13-coupled agonists was abolished, and it was impaired in response to Gq stimulation. Similarly, RhoA was required for efficient secretion of α and dense granules downstream of G13 and Gq. Furthermore, RhoA was essential for integrin-mediated clot retraction but not for actomyosin rearrangements and spreading of activated platelets on fibrinogen. In vivo, RhoA deficiency resulted in markedly prolonged tail bleeding times but also significant protection in different models of arterial thrombosis and in a model of ischemic stroke. Together, these results establish RhoA as an important regulator of platelet function in thrombosis and hemostasis.
- Published
- 2011
42. Multiple alterations of platelet functions dominated by increased secretion in mice lacking Cdc42 in platelets
- Author
-
Anita Eckly, Christian Gachet, Bernhard Nieswandt, Ina Hagedorn, Cord Brakebusch, François Lanza, Sandra Eliautou, Xunwei Wu, Irina Pleines, Margitta Elvers, and Markus Bender
- Subjects
Blood Platelets ,Immunology ,Blotting, Western ,macromolecular substances ,CDC42 ,Cell Separation ,Biology ,Biochemistry ,chemistry.chemical_compound ,Mice ,GTP-Binding Protein Regulators ,Animals ,Secretion ,Platelet ,Platelet activation ,Mice, Knockout ,Hemostasis ,Degranulation ,Cell Biology ,Hematology ,Flow Cytometry ,Platelet Activation ,Cell biology ,Adenosine diphosphate ,Microscopy, Electron ,chemistry ,Filopodia ,Adenosine triphosphate - Abstract
Platelet activation at sites of vascular injury is crucial for hemostasis, but it may also cause myocardial infarction or stroke. Cytoskeletal reorganization is essential for platelet activation and secretion. The small GTPase Cdc42 has been implicated as an important mediator of filopodia formation and exocytosis in various cell types, but its exact function in platelets is not established. Here, we show that the megakaryocyte/platelet-specific loss of Cdc42 leads to mild thrombocytopenia and a small increase in platelet size in mice. Unexpectedly, Cdc42-deficient platelets were able to form normally shaped filopodia and spread fully on fibrinogen upon activation, whereas filopodia formation upon selective induction of GPIb signaling was reduced compared with wild-type platelets. Furthermore, Cdc42-deficient platelets showed enhanced secretion of α granules, a higher adenosine diphosphate (ADP)/adenosine triphosphate (ATP) content, increased aggregation at low agonist concentrations, and enhanced aggregate formation on collagen under flow. In vivo, lack of Cdc42 resulted in faster occlusion of ferric chloride–injured arterioles. The life span of Cdc42-deficient platelets was markedly reduced, suggesting increased clearing of the cells under physiologic conditions. These data point to novel multiple functions of Cdc42 in the regulation of platelet activation, granule organization, degranulation, and a specific role in GPIb signaling.
- Published
- 2010
43. Deficiency of the tetraspanin CD63 associated with kidney pathology but normal lysosomal function
- Author
-
Nina Himmerkus, Paul Saftig, Irina Pleines, Friedrich Koch-Nolte, Markus Bleich, Renate Lüllmann-Rauch, Zane Orinska, Bernhard Nieswandt, Bernd Schröder, and Jenny Schröder
- Subjects
Blood Platelets ,Platelet Function Tests ,Endosome ,Molecular Sequence Data ,Platelet Membrane Glycoproteins ,Biology ,Urinalysis ,Mice ,Immune system ,Tetraspanin ,Antigen ,Antigens, CD ,Animals ,Amino Acid Sequence ,Kidney Tubules, Collecting ,Molecular Biology ,Inclusion Bodies ,Genome ,Membrane Glycoproteins ,CD63 ,Tetraspanin 30 ,Gene targeting ,Cell Biology ,Articles ,Fibroblasts ,Water-Electrolyte Balance ,Chromosomes, Mammalian ,Immunohistochemistry ,Cell biology ,Diuresis ,Immune System ,Knockout mouse ,Immunology ,Gene Targeting ,Female ,Kidney Diseases ,Lysosomes ,Intracellular ,Pseudogenes - Abstract
CD63 is a member of the tetraspanin superfamily that constitutes a main component of the lysosomal membrane. In mice, two CD63 gene loci are present, with only one of these two being functional. We generated and analyzed mice deficient for active CD63. Disruption of CD63 results in a complete loss of CD63 protein expression. Despite its abundance in late endosomes/lysosomes, the lack of CD63 does not cause obvious endosomal/lysosomal abnormalities. CD63 knockout mice are viable and fertile without gross morphological abnormalities in the majority of tissues. No alterations in the populations of immune cells and only minor differences in platelet function were observed. This suggests that the lack of CD63 could be successfully compensated for, most likely by other tetraspanins. However, CD63 deficiency leads to an altered water balance. CD63 knockout mice show an increased urinary flow, water intake, reduced urine osmolality, and a higher fecal water content. In principle cells of the collecting duct of CD63-deficient mice, abnormal intracellular lamellar inclusions were observed. This indicates that the sorting of apical transport proteins might be impaired in these cells. CD63 knockout mice provide an important tool for analyzing the various postulated functions of CD63 in vivo.
- Published
- 2008
44. The calcium sensor STIM1 is an essential mediator of arterial thrombosis and ischemic brain infarction
- Author
-
Christoph Kleinschnitz, Guido Stoll, David Varga-Szabo, Markus Bender, Irina Pleines, Bernhard Nieswandt, Mirko Pham, Attila Braun, and Thomas Renné
- Subjects
inorganic chemicals ,Brain Infarction ,medicine.medical_specialty ,Bleeding Time ,Platelet Aggregation ,T-Lymphocytes ,Immunology ,Infarction ,Hemorrhage ,Mice ,Internal medicine ,medicine ,Immunology and Allergy ,Animals ,Platelet ,Platelet activation ,Stromal Interaction Molecule 1 ,Thrombus ,Growth Disorders ,Mice, Knockout ,Membrane Glycoproteins ,Voltage-dependent calcium channel ,business.industry ,Endoplasmic reticulum ,Brief Definitive Report ,STIM1 ,Thrombosis ,medicine.disease ,Sarcoplasmic Reticulum ,Endocrinology ,Brief Definitive Reports ,Calcium ,Calcium Channels ,business - Abstract
Platelet activation and aggregation are essential to limit posttraumatic blood loss at sites of vascular injury but also contributes to arterial thrombosis, leading to myocardial infarction and stroke. Agonist-induced elevation of [Ca2+]i is a central step in platelet activation, but the underlying mechanisms are not fully understood. A major pathway for Ca2+ entry in nonexcitable cells involves receptor-mediated release of intracellular Ca2+ stores, followed by activation of store-operated calcium (SOC) channels in the plasma membrane. Stromal interaction molecule 1 (STIM1) has been identified as the Ca2+ sensor in the endoplasmic reticulum (ER) that activates Ca2+ release–activated channels in T cells, but its role in mammalian physiology is unknown. Platelets express high levels of STIM1, but its exact function has been elusive, because these cells lack a normal ER and Ca2+ is stored in a tubular system referred to as the sarcoplasmatic reticulum. We report that mice lacking STIM1 display early postnatal lethality and growth retardation. STIM1-deficient platelets have a marked defect in agonist-induced Ca2+ responses, and impaired activation and thrombus formation under flow in vitro. Importantly, mice with STIM1-deficient platelets are significantly protected from arterial thrombosis and ischemic brain infarction but have only a mild bleeding time prolongation. These results establish STIM1 as an important mediator in the pathogenesis of ischemic cardio- and cerebrovascular events.
- Published
- 2008
45. Cell adhesion mechanisms in platelets
- Author
-
Irina Pleines, David Varga-Szabo, and Bernhard Nieswandt
- Subjects
Blood Platelets ,Platelet Aggregation ,Integrin ,Platelet Membrane Glycoproteins ,Sensitivity and Specificity ,Collagen receptor ,Extracellular matrix ,Mice ,Platelet Adhesiveness ,Von Willebrand factor ,Cell Adhesion ,Animals ,Humans ,Platelet ,Cell adhesion ,Molecular Biology ,biology ,Chemistry ,Adhesion ,Cell biology ,Extracellular Matrix ,Immunology ,biology.protein ,GPVI ,Cardiology and Cardiovascular Medicine ,Signal Transduction - Abstract
At sites of vascular injury, platelets come into contact with the subendothelial extracellular matrix which triggers their activation and the formation of a hemostatic plug. This process is crucial for normal hemostasis, but may also lead to pathological thrombus formation causing diseases such as myocardial infarction or stroke. The initial capture of flowing platelets is mediated by the interaction of the glycoprotein (GP) Ib-V-IX complex with von Willebrand factor (vWF) immobilized on exposed collagens. This interaction allows the binding of the collagen receptor GPVI to its ligand and to initiate cellular activation, a process that is reinforced by locally produced thrombin and soluble mediators released from platelets. These events lead to the shift of β1 and β3 integrins on the platelet surface from a low to a high affinity state, thereby enabling them to bind their ligands and to mediate firm adhesion, spreading, coagulant activity, and aggregation. This review summarizes the most important structural and functional properties of these adhesion receptors and briefly discusses their potential as targets for antithrombotic therapy.
- Published
- 2008
46. BCL-2 is dispensable for thrombopoiesis and platelet survival
- Author
-
Marlyse A. Debrincat, Warren S. Alexander, Andreas Strasser, Emma C. Josefsson, Ashley P. Ng, Jason Corbin, Marion Lebois, Rachael M. Lane, Martha Sola-Visner, M L Holmes, Cassandra J. Vandenberg, Benjamin T. Kile, Irina Pleines, and Philippe Bouillet
- Subjects
Genetically modified mouse ,Blood Platelets ,Cancer Research ,Programmed cell death ,Cell Survival ,Transgene ,Immunology ,bcl-X Protein ,Mice, Transgenic ,Biology ,Thrombopoiesis ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Mice ,Megakaryocyte ,medicine ,Animals ,Platelet ,Navitoclax ,Cell Biology ,Thrombocytopenia ,medicine.anatomical_structure ,chemistry ,Proto-Oncogene Proteins c-bcl-2 ,Apoptosis ,Cancer research ,Original Article - Abstract
Navitoclax (ABT-263), an inhibitor of the pro-survival BCL-2 family proteins BCL-2, BCL-XL and BCL-W, has shown clinical efficacy in certain BCL-2-dependent haematological cancers, but causes dose-limiting thrombocytopaenia. The latter effect is caused by Navitoclax directly inducing the apoptotic death of platelets, which are dependent on BCL-XL for survival. Recently, ABT-199, a selective BCL-2 antagonist, was developed. It has shown promising anti-leukaemia activity in patients whilst sparing platelets, suggesting that the megakaryocyte lineage does not require BCL-2. In order to elucidate the role of BCL-2 in megakaryocyte and platelet survival, we generated mice with a lineage-specific deletion of Bcl2, alone or in combination with loss of Mcl1 or Bclx. Platelet production and platelet survival were analysed. Additionally, we made use of BH3 mimetics that selectively inhibit BCL-2 or BCL-XL. We show that the deletion of BCL-2, on its own or in concert with MCL-1, does not affect platelet production or platelet lifespan. Thrombocytopaenia in Bclx-deficient mice was not affected by additional genetic loss or pharmacological inhibition of BCL-2. Thus, BCL-2 is dispensable for thrombopoiesis and platelet survival in mice.
- Published
- 2015
47. Mutations in Tropomyosin 4 Cause Macrothrombocytopenia in Mice and Humans
- Author
-
Joanne Woods, Willem H. Ouwehand, Rachael M. Lane, Nicola Foad, Peter W. Gunning, Galina Schevzov, Ernest Turro, Sarah K Westbury, Harriet Manning, Andrew D Mumford, Rémi Favier, Stephane Chappaz, Irina Pleines, Benjamin T. Kile, and Marloes R. Tijssen
- Subjects
education.field_of_study ,Platelet disorder ,Immunology ,Population ,Platelet Glycoprotein GPIb-IX Complex ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,medicine.anatomical_structure ,Megakaryocyte ,medicine ,Missense mutation ,Platelet ,Bone marrow ,Mean platelet volume ,education - Abstract
A genome-wide scan in nearly 70,000 individuals showed that the common SNP rs8109288 in the first intron of the human TPM4 gene exerts an effect on the volume and count of platelets (Gieger et al. Nature 2011). We isolated a mouse line with an ENU-induced missense mutation in Tpm4. Mice carrying this mutation exhibited dose-dependent macrothrombocytopenia, while other blood cell counts were normal. Bone marrow transplant experiments demonstrated that the phenotype is intrinsic to hematopoietic cells. Notably, Tpm4 insufficiency did not affect the life span or in vitro function of mutant platelets, and there was no evidence of an increased propensity to bleeding. Megakaryocyte numbers in the bone marrow were increased, although maturation as measured by ploidy appeared normal. Mutant megakaryocytes displayed altered morphology indicating fragmentation, and markedly decreased proplatelet formation in vitro. Based on Gieger et al., we examined the functional requirement for TPM4 in human megakaryocytes. We found that the localisation of TPM4 in proplatelet-forming megakaryocytes was extremely similar to the localisation in their mouse counterparts, suggesting an identical role. Furthermore, knock down of TPM4by shRNA in human megakaryocytes did not affect maturation as measured by CD41 and CD42 expression, but significantly reduced the number of proplatelet-forming cells. The occasional megakaryocyte that did form proplatelets did not exhibit the typical “beads-on-a-string” phenotype. Typically, one large bulb at the end of a protrusion or a string with no clearly distinguishable beads was observed. We therefore performed a look-up in the BRIDGE consortium database, which enrolled 542 cases with inherited bleeding and platelet disorders of unknown aetiology in the NIHR BioResource for exome sequencing. Calling of variants revealed single nucleotide variants with consequences in TPM4which were absent from ~30,000 control haplotypes, in three BRIDGE cases. Two cases with a stop codon at residue 105 and R91H variant presented with macrothrombocytopenia and mild bleeding symptoms with platelet counts of 103 and 128 x10e9/L and volumes of 15.10 and 14.00 fl, respectively. The remaining case with variant D20N (not conserved, genomic evolutionary rate profiling score of 2.68 compared to 4.62 for the other variants) had a count in the normal range (232 x10e9/L) and a reduced platelet volume of 7.20 fl. Together, these results provide compelling evidence that Tropomyosin 4 is a crucial regulator of platelet production in mice and humans, being specifically required for the terminal stages of platelet formation. Our studies demonstrate that the common intronic variant exerts a subtle effect, whilst two extremely rare variants have a more robust effect on platelet formation leading to counts and volumes at the tails of the population distribution. The lack of concordance between mice and humans with regard to bleeding may be explained by strong modifiers at loci other than TPM4. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.