Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Am J Hematol ; 99(3): 336-349, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38165047

RESUMO

Mechanisms through which mature megakaryocytes (Mks) and their progenitors sense the bone marrow extracellular matrix to promote lineage differentiation in health and disease are still partially understood. We found PIEZO1, a mechanosensitive cation channel, to be expressed in mouse and human Mks. Human mutations in PIEZO1 have been described to be associated with blood cell disorders. Yet, a role for PIEZO1 in megakaryopoiesis and proplatelet formation has never been investigated. Here, we show that activation of PIEZO1 increases the number of immature Mks in mice, while the number of mature Mks and Mk ploidy level are reduced. Piezo1/2 knockout mice show an increase in Mk size and platelet count, both at basal state and upon marrow regeneration. Similarly, in human samples, PIEZO1 is expressed during megakaryopoiesis. Its activation reduces Mk size, ploidy, maturation, and proplatelet extension. Resulting effects of PIEZO1 activation on Mks resemble the profile in Primary Myelofibrosis (PMF). Intriguingly, Mks derived from Jak2V617F PMF mice show significantly elevated PIEZO1 expression, compared to wild-type controls. Accordingly, Mks isolated from bone marrow aspirates of JAK2V617F PMF patients show increased PIEZO1 expression compared to Essential Thrombocythemia. Most importantly, PIEZO1 expression in bone marrow Mks is inversely correlated with patient platelet count. The ploidy, maturation, and proplatelet formation of Mks from JAK2V617F PMF patients are rescued upon PIEZO1 inhibition. Together, our data suggest that PIEZO1 places a brake on Mk maturation and platelet formation in physiology, and its upregulation in PMF Mks might contribute to aggravating some hallmarks of the disease.


Assuntos
Mielofibrose Primária , Trombocitemia Essencial , Humanos , Animais , Camundongos , Megacariócitos/metabolismo , Mielofibrose Primária/genética , Medula Óssea , Trombopoese/genética , Trombocitemia Essencial/metabolismo , Plaquetas/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo
2.
Blood Adv ; 7(15): 4003-4018, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37171626

RESUMO

Megakaryocytes (MKs) are the precursor cells of platelets, located in the bone marrow (BM). Once mature, they extend elongated projections named proplatelets through sinusoid vessels, emerging from the marrow stroma into the circulating blood. Not all signals from the microenvironment that regulate proplatelet formation are understood, particularly those from the BM biomechanics. We sought to investigate how MKs perceive and adapt to modifications of the stiffness of their environment. Although the BM is one of the softest tissue of the body, its rigidification results from excess fibronectin (FN), and other matrix protein deposition occur upon myelofibrosis. Here, we have shown that mouse MKs are able to detect the stiffness of a FN-coated substrate and adapt their morphology accordingly. Using a polydimethylsiloxane substrate with stiffness varying from physiological to pathological marrow, we found that a stiff matrix favors spreading, intracellular contractility, and FN fibrils assembly at the expense of proplatelet formation. Itgb3, but not Itgb1, is required for stiffness sensing, whereas both integrins are involved in fibrils assembly. In contrast, soft substrates promote proplatelet formation in an Itgb3-dependent manner, consistent with the ex vivo decrease in proplatelet formation and the in vivo decrease in platelet number in Itgb3-deficient mice. Our findings demonstrate the importance of environmental stiffness for MK functions with potential pathophysiological implications during pathologies that deregulate FN deposition and modulate stiffness in the marrow.


Assuntos
Fibronectinas , Megacariócitos , Animais , Camundongos , Plaquetas/metabolismo , Medula Óssea , Fibronectinas/metabolismo , Megacariócitos/metabolismo , Contagem de Plaquetas
3.
Psychooncology ; 31(8): 1412-1419, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35488808

RESUMO

BACKGROUND: Art therapy (AT) as supportive care may help patients cope with cancer treatments. This non-blinded randomized trial assessed the impact of creative AT on severe fatigue and quality of life (QoL) in localized breast cancer patients undergoing irradiation. MATERIAL AND METHODS: 320 patients were randomized to an AT group (ATG; 8 weekly sessions starting during irradiation) or to a standard group (SG). The primary endpoint was severe global fatigue (Functional Assessment of Chronic Therapy Fatigue subscale score <37) at 1 month post-irradiation. Quality of life (Fact-B), anxiety/depression (Hospital Anxiety and Depression Scale (HADS)) and different dimensions of fatigue 20-item Multidimensional Fatigue Inventory (MFI-20) were assessed at 1, 6 and 12 months post-irradiation. The secondary endpoints, fatigue among patients treated with chemotherapy, QoL (Fact-B), anxiety/depression (HADS) and different dimensions of fatigue (MFI-20) at 1, 6 and 12 months post-irradiation (with post hoc analysis in patients with treated with chemotherapy) were also assessed. RESULTS: 82% of patients completed ≥8 sessions. Severe initial global fatigue was observed in 43% of patients in each group, and among in 64% of patients whose treatment protocol contained chemotherapy. At 1 month post-irradiation, 45% in the ATG and 57% of patients in the SG reported severe global fatigue (p = 0.37); among patients with initial severe mental fatigue (MFF), 79% and 44% had improved MFF (p = 0.007) respectively; similarly 79% and 44% with initial poor motivation had better mental motivation (p = 0.03). At 6 and 12 months, social well-being scores in the ATG were higher (21.3 and 21.4 vs. 19.8 and 19.2, p = 0.05 and p < 0.01) with a significant improvement for patients who had chemotherapy (41% vs. 18%, p = 0.017). A positive association was observed between the number of AT sessions, fatigue and QoL (p < 0.01). CONCLUSION: AT did not significantly improve global severe fatigue among all cancer participants 1 month after radiation therapy, however it had a positive impact on social well-being and may improve MFF and motivation.


Assuntos
Arteterapia , Neoplasias da Mama , Ansiedade/terapia , Neoplasias da Mama/complicações , Neoplasias da Mama/radioterapia , Depressão/terapia , Feminino , Humanos , Qualidade de Vida
4.
J Vis Exp ; (174)2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34515681

RESUMO

The 3D environment leading to both confinement and mechanical constraints is increasingly recognized as an important determinant of cell behavior. 3D culture has thus been developed to better approach the in vivo situation. Megakaryocytes differentiate from hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM). The BM is one of the softest tissues of the body, confined inside the bone. The bone being poorly extensible at the cell scale, megakaryocytes are concomitantly subjected to a weak stiffness and high confinement. This protocol presents a method for the recovery of mouse lineage negative (Lin-) HSPCs by immuno-magnetic sorting and their differentiation into mature megakaryocytes in a 3D medium composed of methylcellulose. Methylcellulose is non-reactive towards megakaryocytes and its stiffness may be adjusted to that of normal bone marrow or increased to mimic a pathological fibrotic marrow. The process to recover the megakaryocytes for further cell analyses is also detailed in the protocol. Although proplatelet extension is prevented within the 3D milieu, it is described below how to resuspend the megakaryocytes in liquid medium and to quantify their capacity to extend proplatelets. Megakaryocytes grown in 3D hydrogel have a higher capacity to form proplatelets compared to those grown in a liquid milieu. This 3D culture allows i) to differentiate progenitors towards megakaryocytes reaching a higher maturation state, ii) to recapitulate phenotypes that may be observed in vivo but go unnoticed in classical liquid cultures, and iii) to study transduction pathways induced by the mechanical cues provided by a 3D environment.


Assuntos
Megacariócitos , Metilcelulose , Animais , Medula Óssea , Células da Medula Óssea , Diferenciação Celular , Células Cultivadas , Hidrogéis , Camundongos
5.
J Vis Exp ; (173)2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34398145

RESUMO

Platelets are produced by megakaryocytes, specialized cells located in the bone marrow. The possibility to image megakaryocytes in real time and their native environment was described more than 10 years ago and sheds new light on the process of platelet formation. Megakaryocytes extend elongated protrusions, called proplatelets, through the endothelial lining of sinusoid vessels. This paper presents a protocol to simultaneously image in real time fluorescently labeled megakaryocytes in the skull bone marrow and sinusoid vessels. This technique relies on a minor surgery that keeps the skull intact to limit inflammatory reactions. The mouse head is immobilized with a ring glued to the skull to prevent movements from breathing. Using two-photon microscopy, megakaryocytes can be visualized for up to a few hours, enabling the observation of cell protrusions and proplatelets in the process of elongation inside sinusoid vessels. This allows the quantification of several parameters related to the morphology of the protrusions (width, length, presence of constriction areas) and their elongation behavior (velocity, regularity, or presence of pauses or retraction phases). This technique also allows simultaneous recording of circulating platelets in sinusoid vessels to determine platelet velocity and blood flow direction. This method is particularly useful to study the role of genes of interest in platelet formation using genetically modified mice and is also amenable to pharmacological testing (study the mechanisms, evaluating drugs in the treatment of platelet production disorders). It has become an invaluable tool, especially to complement in vitro studies as it is now known that in vivo and in vitro proplatelet formation rely on different mechanisms. It has been shown, for example, that in vitro microtubules are required for proplatelet elongation per se. However, in vivo, they rather serve as a scaffold, elongation being mainly promoted by blood flow forces.


Assuntos
Medula Óssea , Megacariócitos , Animais , Plaquetas , Camundongos , Microtúbulos , Crânio/diagnóstico por imagem , Crânio/cirurgia
6.
Exp Clin Transplant ; 19(8): 877-879, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34085918

RESUMO

Portal vein thrombosis is a common complication in patients with cirrhosis and a challenge for the transplant team. Not so long ago, portal vein thrombosis was considered an absolute contraindication for liver transplant, but improvements in surgical techniques have overcome this problem in many transplant centers around the world. Here, we present the case of a 52-year-old female patient with cirrhosis from a primary biliary cholangitis and a complex portal vein thrombosis. She underwent a deceased donor liver transplant with a Model for End-Stage Liver Disease of 40. The portal thrombosis was handled using a portosystemic shunt from the splenic vein to the left ovarian vein, which was visualized on a computed tomography scan performed as part of the study protocol. The donor was a 52-year-old woman with brain death secondary to a vascular cerebral accident. A caval replacement technique was used with no complications during surgery. For the portal anastomosis, the dilated left ovarian vein was carefully dissected and brought through the lesser sac, behind the stomach, to obtain a suitable length. An end-to-end anastomosis of the graft portal vein to the left ovarian vein was performed with a 6-0 Prolene running suture. An abdominal computed tomography scan was performed 6 months after liver transplant showing patency of portal vein and no anastomotic defects, and after 24 months of follow-up the patient is in good clinical condition with normal laboratory values and Doppler ultrasonography with no vascular anomalies and adequate portal flow. To our knowledge, the use of a spleno-ovarian shunt has not been reported as an alternative for portal reconstruction in a case of thrombosis.


Assuntos
Doença Hepática Terminal , Hepatopatias , Transplante de Fígado , Trombose , Trombose Venosa , Doença Hepática Terminal/etiologia , Feminino , Humanos , Cirrose Hepática/complicações , Hepatopatias/complicações , Transplante de Fígado/efeitos adversos , Transplante de Fígado/métodos , Doadores Vivos , Pessoa de Meia-Idade , Veia Porta/diagnóstico por imagem , Veia Porta/cirurgia , Índice de Gravidade de Doença , Trombose/etiologia , Resultado do Tratamento , Trombose Venosa/diagnóstico por imagem , Trombose Venosa/etiologia , Trombose Venosa/cirurgia
7.
J Vis Exp ; (171)2021 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-34096921

RESUMO

The last stage of megakaryopoiesis leads to cytoplasmic extensions from mature megakaryocytes, the so-called proplatelets. Much has been learned about the proplatelet formation using in vitro-differentiated megakaryocytes; however, there is an increasing evidence that conventional culture systems do not faithfully recapitulate the differentiation/maturation process that takes places inside the bone marrow. In this manuscript, we present an explant method initially described in 1956 by Thiéry and Bessis to visualize megakaryocytes which have matured in their native environment, thus circumventing potential artifacts and misinterpretations. Fresh bone marrows are collected by flushing the femurs of mice, sliced into 0.5 mm cross sections, and placed in an incubation chamber at 37 °C containing a physiological buffer. Megakaryocytes become gradually visible at the explant periphery and are observed up to 6 hours under an inverted microscope coupled to a video camera. Over time, megakaryocytes change their shape, with some cells having a spherical form and others developing thick extensions or extending many thin proplatelets with extensive branching. Both qualitative and quantitative investigations are carried out. This method has the advantage of being simple, reproducible, and fast as numerous megakaryocytes are present, and classically half of them form proplatelets in 6 hours compared to 4 days for cultured mouse megakaryocytes. In addition to the study of mutant mice, an interesting application of this method is the straightforward evaluation of the pharmacological agents on the proplatelet extension process, without interfering with the differentiation process that may occur in cultures.


Assuntos
Plaquetas , Medula Óssea , Animais , Plaquetas/citologia , Diferenciação Celular , Células Cultivadas , Citoplasma , Megacariócitos/citologia , Camundongos
8.
J Cell Sci ; 133(20)2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-33127839

RESUMO

The main function of blood platelets is to ensure hemostasis and prevent hemorrhages. The 1011 platelets needed daily are produced in a well-orchestrated process. However, this process is not yet fully understood and in vitro platelet production is still inefficient. Platelets are produced in the bone marrow by megakaryocytes, highly specialized precursor cells that extend cytoplasmic projections called proplatelets (PPTs) through the endothelial barrier of sinusoid vessels. In this Cell Science at a Glance article and the accompanying poster we discuss the mechanisms and pathways involved in megakaryopoiesis and platelet formation processes. We especially address the - still underestimated - role of the microenvironment of the bone marrow, and present recent findings on how PPT extension in vivo differs from that in vitro and entails different mechanisms. Finally, we recapitulate old but recently revisited evidence that - although bone marrow does produce megakaryocytes and PPTs - remodeling and the release of bona fide platelets, mainly occur in the downstream microcirculation.


Assuntos
Plaquetas , Megacariócitos , Medula Óssea , Citoplasma , Trombopoese
9.
J Thromb Haemost ; 18(11): 2987-3001, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32702204

RESUMO

BACKGROUND: Blood platelets are anucleate cell fragments that prevent bleeding and minimize blood vessel injury. They are formed from the cytoplasm of megakaryocytes located in the bone marrow. For successful platelet production, megakaryocyte fragments must pass through the sinusoid endothelial barrier by a cell biology process unique to these giant cells as compared with erythrocytes and leukocytes. Currently, the mechanisms by which megakaryocytes interact and progress through the endothelial cells are not understood, resulting in a significant gap in our knowledge of platelet production. OBJECTIVE: The aim of this study was to investigate how megakaryocytes interact and progress through the endothelial cells of mouse bone marrow sinusoids. METHODS: We used a combination of fluorescence, electron, and three-dimensional microscopy to characterize the cellular events between megakaryocytes and endothelial cells. RESULTS: We identified protrusive, F-actin-based podosome-like structures, called in vivo-MK podosomes, which initiate the formation of pores through endothelial cells. These structures present a collective and spatial organization through their interconnection via a contractile network of actomyosin, essential to regulate the endothelial openings. This ensures proper passage of megakaryocyte-derived processes into the blood circulation to promote thrombopoiesis. CONCLUSION: This study provides novel insight into the in vivo function of podosomes of megakaryocytes with critical importance to platelet production.


Assuntos
Megacariócitos , Podossomos , Animais , Plaquetas , Medula Óssea , Capilares , Células Endoteliais , Camundongos , Trombopoese
10.
Transplant Proc ; 52(4): 1102-1105, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32204898

RESUMO

BACKGROUND: A short right renal vein (RRV) remains a challenge for renal transplant surgery, especially in the living donor. Different techniques exist to obtain an RRV with a suitable length in cadaveric donor; however, in living donors the options are limited. MATERIAL AND METHODS: We present 2 living kidney transplants in which we obtained a very short RRV, making the implantation very difficult. We describe our technique to overcome this problem by using cadaveric iliac vessels retrieved from previous cadaveric donations and preserved at 4°C in histidine-tryptophan-ketoglutarate (HTK) solution, without intraoperative or postoperative complications. We complied with the Helsinki Congress and the Istanbul Declaration regarding the donor source. RESULTS: In both cases, kidney grafts had optimal primary function, with good creatinine clearance after transplant and good patency of vascular anastomosis by Doppler ultrasounds. CONCLUSIONS: We believe the use of cadaveric vessel grafts in living donor kidney transplant is a valuable resource as a rescue tool in emergency situations like the ones being presented in this article in order to avoid discarding a kidney graft with damage or short vessels. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.


Assuntos
Artéria Ilíaca/transplante , Transplante de Rim/métodos , Doadores Vivos , Veias Renais , Aloenxertos , Cadáver , Humanos , Masculino , Pessoa de Meia-Idade , Transplante Homólogo
11.
Methods Mol Biol ; 1812: 139-153, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30171577

RESUMO

The differentiation and maturation of megakaryocytes (MKs) occurs in a 3D environment where the cells must constantly adapt to the external physical and mechanical constraints during their development and migration to sinusoid vessels. In this chapter, we present a method for culture of mouse MKs from bone marrow hematopoietic progenitor cells in a methylcellulose 3D medium with a stiffness mimicking that of bone marrow. In addition, we describe how the MKs can be recovered to allow for analysis of their differentiation and maturation state by transmission electron microscopy, immunofluorescence or flow cytometry techniques and to evaluate their ability to form proplatelets. This approach allows (1) generation of MKs with a morphology that more closely resembles the MKs that differentiate in vivo, (2) recovery of megakaryocyte phenotypes sometimes observed in vivo but not found in classical liquid cultures, and (3) study of mechanotransduction pathways induced by the stiffness of the medium.


Assuntos
Técnicas de Cultura de Células/métodos , Megacariócitos/citologia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Hidrogéis/química , Metilcelulose/química , Camundongos , Microscopia Eletrônica de Transmissão
12.
Am J Hematol ; 93(3): 430-441, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29247535

RESUMO

The extracellular matrix (ECM) is a key acellular structure in constant remodeling to provide tissue cohesion and rigidity. Deregulation of the balance between matrix deposition, degradation, and crosslinking results in fibrosis. Bone marrow fibrosis (BMF) is associated with several malignant and nonmalignant pathologies severely affecting blood cell production. BMF results from abnormal deposition of collagen fibers and enhanced lysyl oxidase-mediated ECM crosslinking within the marrow, thereby increasing marrow stiffness. Bone marrow stiffness has been recently recognized as an important regulator of blood cell development, notably by modifying the fate and differentiation process of hematopoietic or mesenchymal stem cells. This review surveys the different components of the ECM and their influence on stem cell development, with a focus on the impact of the ECM composition and stiffness on the megakaryocytic lineage in health and disease. Megakaryocyte maturation and the biogenesis of their progeny, the platelets, are thought to respond to environmental mechanical forces through a number of mechanosensors, including integrins and mechanosensitive ion channels, reviewed here.


Assuntos
Plaquetas/citologia , Medula Óssea/fisiologia , Matriz Extracelular/fisiologia , Hematopoese/fisiologia , Megacariócitos/citologia , Animais , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/fisiologia , Glicosaminoglicanos/fisiologia , Células-Tronco Hematopoéticas/citologia , Humanos , Integrinas/fisiologia , Canais Iônicos/fisiologia , Mecanotransdução Celular , Células-Tronco Mesenquimais/citologia , Camundongos , Proteínas de Neoplasias/fisiologia , Neoplasias/patologia , Mielofibrose Primária/patologia , Proteína-Lisina 6-Oxidase/fisiologia , Trombopoese/fisiologia
13.
Cell ; 171(6): 1368-1382.e23, 2017 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-29195076

RESUMO

Blood platelets are critical for hemostasis and thrombosis and play diverse roles during immune responses. Despite these versatile tasks in mammalian biology, their skills on a cellular level are deemed limited, mainly consisting in rolling, adhesion, and aggregate formation. Here, we identify an unappreciated asset of platelets and show that adherent platelets use adhesion receptors to mechanically probe the adhesive substrate in their local microenvironment. When actomyosin-dependent traction forces overcome substrate resistance, platelets migrate and pile up the adhesive substrate together with any bound particulate material. They use this ability to act as cellular scavengers, scanning the vascular surface for potential invaders and collecting deposited bacteria. Microbe collection by migrating platelets boosts the activity of professional phagocytes, exacerbating inflammatory tissue injury in sepsis. This assigns platelets a central role in innate immune responses and identifies them as potential targets to dampen inflammatory tissue damage in clinical scenarios of severe systemic infection.


Assuntos
Infecções Bacterianas/imunologia , Plaquetas/imunologia , Animais , Bactérias/classificação , Plaquetas/citologia , Vasos Sanguíneos/lesões , Vasos Sanguíneos/patologia , Cálcio/metabolismo , Movimento Celular , Polaridade Celular , Humanos , Inflamação/imunologia , Integrinas/metabolismo , Camundongos , Miosinas/metabolismo , Neutrófilos/citologia
14.
Haematologica ; 101(8): 896-908, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27478199

RESUMO

Constitutional thrombocytopenias result from platelet production abnormalities of hereditary origin. Long misdiagnosed and poorly studied, knowledge about these rare diseases has increased considerably over the last twenty years due to improved technology for the identification of mutations, as well as an improvement in obtaining megakaryocyte culture from patient hematopoietic stem cells. Simultaneously, the manipulation of mouse genes (transgenesis, total or conditional inactivation, introduction of point mutations, random chemical mutagenesis) have helped to generate disease models that have contributed greatly to deciphering patient clinical and laboratory features. Most of the thrombocytopenias for which the mutated genes have been identified now have a murine model counterpart. This review focuses on the contribution that these mouse models have brought to the understanding of hereditary thrombocytopenias with respect to what was known in humans. Animal models have either i) provided novel information on the molecular and cellular pathways that were missing from the patient studies; ii) improved our understanding of the mechanisms of thrombocytopoiesis; iii) been instrumental in structure-function studies of the mutated gene products; and iv) been an invaluable tool as preclinical models to test new drugs or develop gene therapies. At present, the genetic determinants of thrombocytopenia remain unknown in almost half of all cases. Currently available high-speed sequencing techniques will identify new candidate genes, which will in turn allow the generation of murine models to confirm and further study the abnormal phenotype. In a complementary manner, programs of random mutagenesis in mice should also identify new candidate genes involved in thrombocytopenia.


Assuntos
Trombocitopenia/etiologia , Trombocitopenia/metabolismo , Animais , Autoantígenos/metabolismo , Síndrome de Bernard-Soulier/etiologia , Síndrome de Bernard-Soulier/metabolismo , Plaquetas/metabolismo , Diferenciação Celular/genética , Citoesqueleto/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Iodeto Peroxidase/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Megacariócitos/citologia , Megacariócitos/metabolismo , Camundongos , Receptores de Trombopoetina/metabolismo , Transdução de Sinais , Trombocitopenia/diagnóstico , Trombopoese , Fatores de Transcrição/metabolismo , Síndrome de Wiskott-Aldrich/etiologia , Síndrome de Wiskott-Aldrich/metabolismo
15.
Blood ; 128(16): 2022-2032, 2016 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-27503502

RESUMO

Megakaryocyte (MK) differentiation occurs within the bone marrow (BM), a complex 3-dimensional (3D) environment of low stiffness exerting local external constraints. To evaluate the influence of the 3D mechanical constraints that MKs may encounter in vivo, we differentiated mouse BM progenitors in methylcellulose (MC) hydrogels tuned to mimic BM stiffness. We found that MKs grown in a medium of 30- to 60-Pa stiffness more closely resembled those in the BM in terms of demarcation membrane system (DMS) morphological aspect and exhibited higher ploidy levels, as compared with MKs in liquid culture. Following resuspension in a liquid medium, MC-grown MKs displayed twice as much proplatelet formation as cells grown in liquid culture. Thus, the MC gel, by mimicking external constraints, appeared to positively influence MK differentiation. To determine whether MKs adapt to extracellular stiffness through mechanotransduction involving actomyosin-based modulation of the intracellular tension, myosin-deficient (Myh9-/-) progenitors were grown in MC gels. Absence of myosin resulted in abnormal cell deformation and strongly decreased proplatelet formation, similarly to features observed for Myh9-/- MKs differentiated in situ but not in vitro. Moreover, megakaryoblastic leukemia 1 (MKL1), a well-known actor in mechanotransduction, was found to be preferentially relocated within the nucleus of MC-differentiated MKs, whereas its inhibition prevented MC-mediated increased proplatelet formation. Altogether, these data show that a 3D medium mimicking BM stiffness contributes, through the myosin IIA and MKL1 pathways, to a more favorable in vitro environment for MK differentiation, which ultimately translates into increased proplatelet production.


Assuntos
Plaquetas/metabolismo , Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Mecanotransdução Celular/fisiologia , Megacariócitos/metabolismo , Animais , Plaquetas/citologia , Células Cultivadas , Hidrogéis/química , Megacariócitos/citologia , Metilcelulose/química , Camundongos , Camundongos Knockout , Cadeias Pesadas de Miosina , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIA/metabolismo , Tensão Superficial , Transativadores/genética , Transativadores/metabolismo
16.
PLoS One ; 11(3): e0151848, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27010197

RESUMO

Microenvironment and activation signals likely imprint heterogeneity in the lymphatic endothelial cell (LEC) population. Particularly LECs of secondary lymphoid organs are exposed to different cell types and immune stimuli. However, our understanding of the nature of LEC activation signals and their cell source within the secondary lymphoid organ in the steady state remains incomplete. Here we show that integrin alpha 2b (ITGA2b), known to be carried by platelets, megakaryocytes and hematopoietic progenitors, is expressed by a lymph node subset of LECs, residing in medullary, cortical and subcapsular sinuses. In the subcapsular sinus, the floor but not the ceiling layer expresses the integrin, being excluded from ACKR4+ LECs but overlapping with MAdCAM-1 expression. ITGA2b expression increases in response to immunization, raising the possibility that heterogeneous ITGA2b levels reflect variation in exposure to activation signals. We show that alterations of the level of receptor activator of NF-κB ligand (RANKL), by overexpression, neutralization or deletion from stromal marginal reticular cells, affected the proportion of ITGA2b+ LECs. Lymph node LECs but not peripheral LECs express RANK. In addition, we found that lymphotoxin-ß receptor signaling likewise regulated the proportion of ITGA2b+ LECs. These findings demonstrate that stromal reticular cells activate LECs via RANKL and support the action of hematopoietic cell-derived lymphotoxin.


Assuntos
Células Endoteliais/imunologia , Linfonodos/citologia , Glicoproteína IIb da Membrana de Plaquetas/imunologia , Ligante RANK/imunologia , Animais , Células Cultivadas , Células Endoteliais/citologia , Fibronectinas/imunologia , Linfonodos/imunologia , Linfotoxina-beta/imunologia , Camundongos Endogâmicos C57BL , Transdução de Sinais
17.
Cell Stem Cell ; 14(1): 81-93, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24268694

RESUMO

Self-renewal and differentiation of stem cells depend on asymmetric division and polarized motility processes that in other cell types are modulated by nonmuscle myosin-II (MII) forces and matrix mechanics. Here, mass spectrometry-calibrated intracellular flow cytometry of human hematopoiesis reveals MIIB to be a major isoform that is strongly polarized in hematopoietic stem cells and progenitors (HSC/Ps) and thereby downregulated in differentiated cells via asymmetric division. MIIA is constitutive and activated by dephosphorylation during cytokine-triggered differentiation of cells grown on stiff, endosteum-like matrix, but not soft, marrow-like matrix. In vivo, MIIB is required for generation of blood, while MIIA is required for sustained HSC/P engraftment. Reversible inhibition of both isoforms in culture with blebbistatin enriches for long-term hematopoietic multilineage reconstituting cells by 5-fold or more as assessed in vivo. Megakaryocytes also become more polyploid, producing 4-fold more platelets. MII is thus a multifunctional node in polarized division and niche sensing.


Assuntos
Diferenciação Celular , Movimento Celular , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Contração Muscular/fisiologia , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Apoptose , Western Blotting , Técnicas de Cultura de Células , Linhagem da Célula , Proliferação de Células , Citometria de Fluxo , Células-Tronco Hematopoéticas/fisiologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Fosforilação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Nicho de Células-Tronco/fisiologia
18.
Blood ; 123(6): 921-30, 2014 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-24152908

RESUMO

The demarcation membrane system (DMS) in megakaryocytes forms the plasma membrane (PM) of future platelets. Using confocal microscopy, electron tomography, and large volume focused ion beam/scanning electron microscopy (FIB/SEM), we determined the sequential steps of DMS formation. We identified a pre-DMS that initiated at the cell periphery and was precisely located between the nuclear lobes. At all developmental stages, the DMS remained continuous with the cell surface. The number of these connections correlated well with the nuclear lobulation, suggesting a relationship with cleavage furrow formation and abortive cytokinesis. On DMS expansion, Golgi complexes assembled around the pre-DMS, and fusion profiles between trans-golgi network-derived vesicles and the DMS were observed. Brefeldin-A reduced DMS expansion, indicating that the exocytic pathway is essential for DMS biogenesis. Close contacts between the endoplasmic reticulum (ER) and the DMS were detected, suggesting physical interaction between the 2 membrane systems. FIB/SEM revealed that the DMS forms an intertwined tubular membrane network resembling the platelet open canalicular system. We thus propose the following steps in DMS biogenesis: (1) focal membrane assembly at the cell periphery; (2) PM invagination and formation of a perinuclear pre-DMS; (3) expansion through membrane delivery from Golgi complexes; and (4) ER-mediated lipid transfer.


Assuntos
Medula Óssea/metabolismo , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Megacariócitos/citologia , Células-Tronco/metabolismo , Rede trans-Golgi/metabolismo , Animais , Células Cultivadas , Megacariócitos/metabolismo , Camundongos , Microscopia de Fluorescência , Células-Tronco/citologia
19.
Nat Commun ; 3: 717, 2012 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-22395608

RESUMO

Megakaryocytes are unique mammalian cells that undergo polyploidization (endomitosis) during differentiation, leading to an increase in cell size and protein production that precedes platelet production. Recent evidence demonstrates that endomitosis is a consequence of a late failure in cytokinesis associated with a contractile ring defect. Here we show that the non-muscle myosin IIB heavy chain (MYH10) is expressed in immature megakaryocytes and specifically localizes in the contractile ring. MYH10 downmodulation by short hairpin RNA increases polyploidization by inhibiting the return of 4N cells to 2N, but other regulators, such as of the G1/S transition, might regulate further polyploidization of the 4N cells. Conversely, re-expression of MYH10 in the megakaryocytes prevents polyploidization and the transition of 2N to 4N cells. During polyploidization, MYH10 expression is repressed by the major megakaryocyte transcription factor RUNX1. Thus, RUNX1-mediated silencing of MYH10 is required for the switch from mitosis to endomitosis, linking polyploidization with megakaryocyte differentiation.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Megacariócitos/citologia , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIB/genética , Poliploidia , Animais , Antígenos CD34/biossíntese , Linhagem Celular , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Citocinese , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Mitose , Cadeias Pesadas de Miosina/biossíntese , Cadeias Pesadas de Miosina/metabolismo , Miosina não Muscular Tipo IIB/biossíntese , Miosina não Muscular Tipo IIB/metabolismo , Interferência de RNA , RNA Interferente Pequeno
20.
Blood ; 119(14): 3333-41, 2012 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-22234693

RESUMO

Macrothrombocytopenia in MYH9-related disease (MYH9-RD) results from defects in nonmuscular myosin-IIA function. Thrombopoietin receptor agonists (eltrombopag; romiplostim) seem to improve hemostasis, but little is known about their biologic effects in MYH9-RD. We administered romiplostim to Myh9(-/-) mice (100 µg/kg, every 3 days, during 1 month). MKs increased to similar numbers in Myh9(-/-) and wild-type (WT) mice (with an increase in immature MKs), but Myh9(-/-) platelet count response was much less (2.5-fold vs 8-fold increase). A strong increase in MK nuclei emboli in the lung, in WT and Myh9(-/-) mice, indicates increased transmigration of MKs from the BM. Prolonged (but not acute) treatment with romiplostim decreased expression of GPIb-IX-V complex and GPVI, but not of GPIIbIIIa, and bleeding time increased in WT mice. Microcirculation was not altered by the increased number of large platelets in any of the assessed organs, but in Myh9(-/-) mice a much stronger increase in BM reticulin fibers was present after 4 weeks of romiplostim treatment vs WT mice. These data further encourage short-term use of thrombopoietic agents in patients with MYH9-RDs; however, myelofibrosis has to be considered as a potential severe adverse effect during longer treatment. Reduction of GPIbIX/GPVI expression by romiplostim requires further studies.


Assuntos
Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Miosina não Muscular Tipo IIA/genética , Mielofibrose Primária/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombopoetina/farmacologia , Animais , Tempo de Sangramento , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Medula Óssea/metabolismo , Medula Óssea/patologia , Embolia/induzido quimicamente , Fator IX/metabolismo , Fator V/metabolismo , Feminino , Contagem de Leucócitos , Pulmão/patologia , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cadeias Pesadas de Miosina , Miosina não Muscular Tipo IIA/metabolismo , Selectina-P/metabolismo , Contagem de Plaquetas , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Fc/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Reticulina/metabolismo , Baço/citologia , Trombopoetina/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA