Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell ; 186(2): 382-397.e24, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36669473

RESUMEN

Blood and lymphatic vessels form a versatile transport network and provide inductive signals to regulate tissue-specific functions. Blood vessels in bone regulate osteogenesis and hematopoiesis, but current dogma suggests that bone lacks lymphatic vessels. Here, by combining high-resolution light-sheet imaging and cell-specific mouse genetics, we demonstrate presence of lymphatic vessels in mouse and human bones. We find that lymphatic vessels in bone expand during genotoxic stress. VEGF-C/VEGFR-3 signaling and genotoxic stress-induced IL6 drive lymphangiogenesis in bones. During lymphangiogenesis, secretion of CXCL12 from proliferating lymphatic endothelial cells is critical for hematopoietic and bone regeneration. Moreover, lymphangiocrine CXCL12 triggers expansion of mature Myh11+ CXCR4+ pericytes, which differentiate into bone cells and contribute to bone and hematopoietic regeneration. In aged animals, such expansion of lymphatic vessels and Myh11-positive cells in response to genotoxic stress is impaired. These data suggest lymphangiogenesis as a therapeutic avenue to stimulate hematopoietic and bone regeneration.


Asunto(s)
Regeneración Ósea , Vasos Linfáticos , Anciano , Animales , Humanos , Ratones , Células Endoteliales , Linfangiogénesis
2.
Annu Rev Cell Dev Biol ; 32: 649-675, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27576121

RESUMEN

In addition to their conventional role as a versatile transport system, blood vessels provide signals controlling organ development, regeneration, and stem cell behavior. In the skeletal system, certain capillaries support perivascular osteoprogenitor cells and thereby control bone formation. Blood vessels are also a critical component of niche microenvironments for hematopoietic stem cells. Here we discuss key pathways and factors controlling endothelial cell behavior in bone, the role of vessels in osteogenesis, and the nature of vascular stem cell niches in bone marrow.


Asunto(s)
Vasos Sanguíneos/metabolismo , Hematopoyesis , Osteogénesis , Transducción de Señal , Animales , Médula Ósea/irrigación sanguínea , Células Endoteliales/metabolismo , Humanos
3.
EMBO J ; 40(1): e105242, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33215738

RESUMEN

Age-associated alterations of the hormone-secreting endocrine system cause organ dysfunction and disease states. However, the cell biology of endocrine tissue ageing remains poorly understood. Here, we perform comparative 3D imaging to understand age-related perturbations of the endothelial cell (EC) compartment in endocrine glands. Datasets of a wide range of markers highlight a decline in capillary and artery numbers, but not of perivascular cells in pancreas, testis and thyroid gland, with age in mice and humans. Further, angiogenesis and ß-cell expansion in the pancreas are coupled by a distinct age-dependent subset of ECs. While this EC subpopulation supports pancreatic ß cells, it declines during ageing concomitant with increased expression of the gap junction protein Gja1. EC-specific ablation of Gja1 restores ß-cell expansion in the aged pancreas. These results provide a proof of concept for understanding age-related vascular changes and imply that therapeutic targeting of blood vessels may restore aged endocrine tissue function. This comprehensive data atlas offers over > 1,000 multicolour volumes for exploration and research in endocrinology, ageing, matrix and vascular biology.


Asunto(s)
Envejecimiento/fisiología , Sistema Endocrino/fisiología , Células Endoteliales/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Vasos Sanguíneos , Glándulas Endocrinas/fisiología , Femenino , Humanos , Imagenología Tridimensional/métodos , Células Secretoras de Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica/patología , Páncreas/fisiología , Testículo/fisiología , Glándula Tiroides/fisiología , Adulto Joven
4.
Nature ; 532(7599): 380-4, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27074508

RESUMEN

Blood vessels define local microenvironments in the skeletal system, play crucial roles in osteogenesis and provide niches for haematopoietic stem cells. The properties of niche-forming vessels and their changes in the ageing organism remain incompletely understood. Here we show that Notch signalling in endothelial cells leads to the expansion of haematopoietic stem cell niches in bone, which involves increases in CD31-positive capillaries and platelet-derived growth factor receptor-ß (PDGFRß)-positive perivascular cells, arteriole formation and elevated levels of cellular stem cell factor. Although endothelial hypoxia-inducible factor signalling promotes some of these changes, it fails to enhance vascular niche function because of a lack of arterialization and expansion of PDGFRß-positive cells. In ageing mice, niche-forming vessels in the skeletal system are strongly reduced but can be restored by activation of endothelial Notch signalling. These findings indicate that vascular niches for haematopoietic stem cells are part of complex, age-dependent microenvironments involving multiple cell populations and vessel subtypes.


Asunto(s)
Envejecimiento/fisiología , Arteriolas/fisiología , Huesos/irrigación sanguínea , Capilares/fisiología , Células Madre Hematopoyéticas/citología , Nicho de Células Madre , Animales , Arteriolas/citología , Huesos/citología , Huesos/metabolismo , Capilares/citología , Recuento de Células , Células Endoteliales/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Osteogénesis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Factor de Células Madre/metabolismo
5.
Nature ; 532(7599): 323-8, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27074509

RESUMEN

Bone marrow endothelial cells (BMECs) form a network of blood vessels that regulate both leukocyte trafficking and haematopoietic stem and progenitor cell (HSPC) maintenance. However, it is not clear how BMECs balance these dual roles, and whether these events occur at the same vascular site. We found that mammalian bone marrow stem cell maintenance and leukocyte trafficking are regulated by distinct blood vessel types with different permeability properties. Less permeable arterial blood vessels maintain haematopoietic stem cells in a low reactive oxygen species (ROS) state, whereas the more permeable sinusoids promote HSPC activation and are the exclusive site for immature and mature leukocyte trafficking to and from the bone marrow. A functional consequence of high permeability of blood vessels is that exposure to blood plasma increases bone marrow HSPC ROS levels, augmenting their migration and differentiation, while compromising their long-term repopulation and survival. These findings may have relevance for clinical haematopoietic stem cell transplantation and mobilization protocols.


Asunto(s)
Vasos Sanguíneos/citología , Vasos Sanguíneos/fisiología , Médula Ósea/irrigación sanguínea , Hematopoyesis , Animales , Antígenos Ly/metabolismo , Arterias/citología , Arterias/fisiología , Células de la Médula Ósea/citología , Diferenciación Celular , Movimiento Celular , Autorrenovación de las Células , Supervivencia Celular , Quimiocina CXCL12/metabolismo , Células Endoteliales/fisiología , Femenino , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Leucocitos/citología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Nestina/metabolismo , Pericitos/fisiología , Permeabilidad , Plasma/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores CXCR4/metabolismo
6.
Nature ; 507(7492): 323-328, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24646994

RESUMEN

The mammalian skeletal system harbours a hierarchical system of mesenchymal stem cells, osteoprogenitors and osteoblasts sustaining lifelong bone formation. Osteogenesis is indispensable for the homeostatic renewal of bone as well as regenerative fracture healing, but these processes frequently decline in ageing organisms, leading to loss of bone mass and increased fracture incidence. Evidence indicates that the growth of blood vessels in bone and osteogenesis are coupled, but relatively little is known about the underlying cellular and molecular mechanisms. Here we identify a new capillary subtype in the murine skeletal system with distinct morphological, molecular and functional properties. These vessels are found in specific locations, mediate growth of the bone vasculature, generate distinct metabolic and molecular microenvironments, maintain perivascular osteoprogenitors and couple angiogenesis to osteogenesis. The abundance of these vessels and associated osteoprogenitors was strongly reduced in bone from aged animals, and pharmacological reversal of this decline allowed the restoration of bone mass.


Asunto(s)
Vasos Sanguíneos/fisiología , Huesos/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Osteogénesis/fisiología , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Vasos Sanguíneos/anatomía & histología , Vasos Sanguíneos/citología , Vasos Sanguíneos/crecimiento & desarrollo , Huesos/citología , Células Endoteliales/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/metabolismo , Oxígeno/metabolismo , Células Madre/citología , Células Madre/metabolismo
7.
Nature ; 507(7492): 376-380, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24647000

RESUMEN

Blood vessel growth in the skeletal system and osteogenesis seem to be coupled, suggesting the existence of molecular crosstalk between endothelial and osteoblastic cells. Understanding the nature of the mechanisms linking angiogenesis and bone formation should be of great relevance for improved fracture healing or prevention of bone mass loss. Here we show that vascular growth in bone involves a specialized, tissue-specific form of angiogenesis. Notch signalling promotes endothelial cell proliferation and vessel growth in postnatal long bone, which is the opposite of the well-established function of Notch and its ligand Dll4 in the endothelium of other organs and tumours. Endothelial-cell-specific and inducible genetic disruption of Notch signalling in mice not only impaired bone vessel morphology and growth, but also led to reduced osteogenesis, shortening of long bones, chondrocyte defects, loss of trabeculae and decreased bone mass. On the basis of a series of genetic experiments, we conclude that skeletal defects in these mutants involved defective angiocrine release of Noggin from endothelial cells, which is positively regulated by Notch. Administration of recombinant Noggin, a secreted antagonist of bone morphogenetic proteins, restored bone growth and mineralization, chondrocyte maturation, the formation of trabeculae and osteoprogenitor numbers in endothelial-cell-specific Notch pathway mutants. These findings establish a molecular framework coupling angiogenesis, angiocrine signals and osteogenesis, which may prove significant for the development of future therapeutic applications.


Asunto(s)
Huesos/irrigación sanguínea , Huesos/metabolismo , Endotelio Vascular/metabolismo , Neovascularización Fisiológica , Osteogénesis , Receptores Notch/metabolismo , Animales , Animales Recién Nacidos , Vasos Sanguíneos/crecimiento & desarrollo , Desarrollo Óseo/efectos de los fármacos , Huesos/citología , Huesos/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Proteínas Portadoras/administración & dosificación , Proteínas Portadoras/metabolismo , Proteínas Portadoras/farmacología , Proliferación Celular , Condrocitos/citología , Condrocitos/efectos de los fármacos , Endotelio Vascular/citología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Osteogénesis/efectos de los fármacos , Transducción de Señal/genética
10.
Nat Cardiovasc Res ; 1: 918-932, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36531334

RESUMEN

The mammalian skeletal system shows sex differences in structure, functions, ageing and disease incidences. The role of blood vessels in physiological, regenerative and pathological bone functions indicates the requisite to understanding their sex specificity. Here, we find oestrogen regulates blood vessel physiology during pregnancy and menopause through oestrogen receptor alpha (ERα) and G-protein coupled oestrogen receptor-1 (Gper1) but not ERß-dependent signalling in mice. Oestrogen regulates BECs' lipid use and promotes lipolysis of adipocytes and FA uptake from the microenvironment. Low oestrogen conditions skew endothelial FA metabolism to accumulate lipid peroxides (LPO), leading to vascular ageing. High ferrous ion levels in female BECs intensify LPO accumulation and accelerate the ageing process. Importantly, inhibiting LPO generation using liproxstatin-1 in aged mice significantly improved bone heath. Thus, our findings illustrate oestrogen's effects on BECs and suggest LPO targeting could be an efficient strategy to manage blood and bone health in females.

11.
Sci Adv ; 7(6)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33536212

RESUMEN

Blood vessels provide supportive microenvironments for maintaining tissue functions. Age-associated vascular changes and their relation to tissue aging and pathology are poorly understood. Here, we perform 3D imaging of young and aging vascular beds. Multiple organs in mice and humans demonstrate an age-dependent decline in vessel density and pericyte numbers, while highly remodeling tissues such as skin preserve the vasculature. Vascular attrition precedes the appearance of cellular hallmarks of aging such as senescence. Endothelial VEGFR2 loss-of-function mice demonstrate that vascular perturbations are sufficient to stimulate cellular changes coupled with aging. Age-associated tissue-specific molecular changes in the endothelium drive vascular loss and dictate pericyte to fibroblast differentiation. Lineage tracing of perivascular cells with inducible PDGFRß and NG2 Cre mouse lines demonstrated that increased pericyte to fibroblast differentiation distinguishes injury-induced organ fibrosis and zymosan-induced arthritis. To spur further discoveries, we provide a freely available resource with 3D vascular and tissue maps.

12.
Front Cell Dev Biol ; 8: 602278, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33330496

RESUMEN

Recent advances in our understanding of blood vessels and vascular niches in bone convey their critical importance in regulating bone development and physiology. The contribution of blood vessels in bone functions and remodeling has recently gained enormous interest because of their therapeutic potential. The mammalian skeletal system performs multiple functions in the body to regulate growth, homeostasis and metabolism. Blood vessels provide support to various cell types in bone and maintain functional niches in the bone marrow microenvironment. Heterogeneity within blood vessels and niches indicate the importance of specialized vascular niches in regulating skeletal functions. In this review, we discuss physiology of bone vasculature and their specialized niches for hematopoietic stem cells and mesenchymal progenitor cells. We provide clinical and experimental information available on blood vessels during physiological bone remodeling.

13.
J Bone Miner Res ; 35(11): 2103-2120, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32845550

RESUMEN

Bone vasculature and bone marrow vascular niches supply oxygen, nutrients, and secrete angiocrine factors required for the survival, maintenance, and self-renewal of stem and progenitor cells. In the skeletal system, vasculature creates nurturing niches for bone and blood-forming stem cells. Blood vessels regulate hematopoiesis and drive bone formation during development, repair, and regeneration. Dysfunctional vascular niches induce skeletal aging, bone diseases, and hematological disorders. Recent cellular and molecular characterization of the bone marrow microenvironment has provided unprecedented insights into the complexity, heterogeneity, and functions of the bone vasculature and vascular niches. The bone vasculature is composed of distinct vessel subtypes that differentially regulate osteogenesis, hematopoiesis, and disease conditions in bones. Further, bone marrow vascular niches supporting stem cells are often complex microenvironments involving multiple different cell populations and vessel subtypes. This review provides an overview of the emerging vascular cell heterogeneity in bone and the new roles of the bone vasculature and associated vascular niches in health and disease. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Enfermedades Óseas , Médula Ósea , Huesos , Hematopoyesis , Humanos , Osteogénesis , Nicho de Células Madre
14.
Nat Cell Biol ; 21(4): 430-441, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30936475

RESUMEN

Growth plate cartilage contributes to the generation of a large variety of shapes and sizes of skeletal elements in the mammalian system. The removal of cartilage and how this process regulates bone shape are not well understood. Here we identify a non-bone-resorbing osteoclast subtype termed vessel-associated osteoclast (VAO). Endothelial cells at the bone/cartilage interface support VAOs through a RANKL-RANK signalling mechanism. In contrast to classical bone-associated osteoclasts, VAOs are dispensable for cartilage resorption and regulate anastomoses of type H vessels. Remarkably, proteinases including matrix metalloproteinase-9 (Mmp9) released from endothelial cells, not osteoclasts, are essential for resorbing cartilage to lead directional bone growth. Importantly, disrupting the orientation of angiogenic blood vessels by misdirecting them results in contorted bone shape. This study identifies proteolytic functions of endothelial cells in cartilage and provides a framework to explore tissue-lytic features of blood vessels in fracture healing, arthritis and cancer.


Asunto(s)
Cartílago/enzimología , Endotelio/enzimología , Osteoclastos/fisiología , Osteogénesis , Péptido Hidrolasas/metabolismo , Animales , Resorción Ósea , Huesos/irrigación sanguínea , Huesos/citología , Cartílago/metabolismo , Endotelio/metabolismo , Placa de Crecimiento/anatomía & histología , Ratones Endogámicos C57BL , Osteoclastos/clasificación , Osteoclastos/metabolismo
15.
JCI Insight ; 4(13)2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31292293

RESUMEN

Bone provides supportive microenvironments for hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) and is a frequent site of metastasis. While incidences of bone metastases increase with age, the properties of the bone marrow microenvironment that regulate dormancy and reactivation of disseminated tumor cells (DTCs) remain poorly understood. Here, we elucidate the age-associated changes in the bone secretome that trigger proliferation of HSCs, MSCs, and DTCs in the aging bone marrow microenvironment. Remarkably, a bone-specific mechanism involving expansion of pericytes and induction of quiescence-promoting secretome rendered this proliferative microenvironment resistant to radiation and chemotherapy. This bone-specific expansion of pericytes was triggered by an increase in PDGF signaling via remodeling of specialized type H blood vessels in response to therapy. The decline in bone marrow pericytes upon aging provides an explanation for loss of quiescence and expansion of cancer cells in the aged bone marrow microenvironment. Manipulation of blood flow - specifically, reduced blood flow - inhibited pericyte expansion, regulated endothelial PDGF-B expression, and rendered bone metastatic cancer cells susceptible to radiation and chemotherapy. Thus, our study provides a framework to recognize bone marrow vascular niches in age-associated increases in metastasis and to target angiocrine signals in therapeutic strategies to manage bone metastasis.


Asunto(s)
Envejecimiento/patología , Médula Ósea/patología , Neoplasias Óseas/terapia , Microambiente Tumoral/fisiología , Antagonistas de Receptores Adrenérgicos alfa 1/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Médula Ósea/irrigación sanguínea , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/secundario , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Resistencia a Antineoplásicos/fisiología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/patología , Células Madre Mesenquimatosas/efectos de la radiación , Ratones , Pericitos/efectos de los fármacos , Pericitos/patología , Pericitos/efectos de la radiación , Prazosina/administración & dosificación , Tolerancia a Radiación/fisiología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de la radiación , Irradiación Corporal Total , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cell Stem Cell ; 22(1): 64-77.e6, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29276143

RESUMEN

Bone marrow vascular niches sustain hematopoietic stem cells (HSCs) and are drastically remodeled in leukemia to support pathological functions. Acute myeloid leukemia (AML) cells produce angiogenic factors, which likely contribute to this remodeling, but anti-angiogenic therapies do not improve AML patient outcomes. Using intravital microscopy, we found that AML progression leads to differential remodeling of vasculature in central and endosteal bone marrow regions. Endosteal AML cells produce pro-inflammatory and anti-angiogenic cytokines and gradually degrade endosteal endothelium, stromal cells, and osteoblastic cells, whereas central marrow remains vascularized and splenic vascular niches expand. Remodeled endosteal regions have reduced capacity to support non-leukemic HSCs, correlating with loss of normal hematopoiesis. Preserving endosteal endothelium with the small molecule deferoxamine or a genetic approach rescues HSCs loss, promotes chemotherapeutic efficacy, and enhances survival. These findings suggest that preventing degradation of the endosteal vasculature may improve current paradigms for treating AML.


Asunto(s)
Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/patología , Nicho de Células Madre , Animales , Médula Ósea/irrigación sanguínea , Médula Ósea/patología , Recuento de Células , Hematopoyesis , Humanos , Microscopía Intravital , Ratones Endogámicos C57BL , Bazo/patología , Células del Estroma/patología , Factores de Tiempo , Microambiente Tumoral
17.
Stem Cells Int ; 2017: 5046953, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29104595

RESUMEN

Bone provides nurturing microenvironments for an array of cell types that coordinate important physiological functions of the skeleton, such as energy metabolism, mineral homeostasis, osteogenesis, and haematopoiesis. Endothelial cells form an intricate network of blood vessels that organises and sustains various microenvironments in bone. The recent identification of heterogeneity in the bone vasculature supports the existence of multiple vascular niches within the bone marrow compartment. A unique combination of cells and factors defining a particular microenvironment, supply regulatory signals to mediate a specific function. This review discusses recent developments in our understanding of vascular niches in bone that play a critical role in regulating the behaviour of multipotent haematopoietic and mesenchymal stem cells during development and homeostasis.

18.
Cell Rep ; 18(7): 1804-1816, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28199850

RESUMEN

Measurements of flow velocities at the level of individual arterial vessels and sinusoidal capillaries are crucial for understanding the dynamics of hematopoietic stem and progenitor cell homing in the bone marrow vasculature. We have developed two complementary intravital two-photon imaging approaches to determine blood flow dynamics and velocities in multiple vessel segments by capturing the motion of red blood cells. High-resolution spatiotemporal measurements through a cranial window to determine short-time dynamics of flowing blood cells and repetitive centerline scans were used to obtain a detailed flow-profile map with hemodynamic parameters. In addition, we observed the homing of individual hematopoietic stem and progenitor cells and obtained detailed information on their homing behavior. With our imaging setup, we determined flow patterns at cellular resolution, blood flow velocities and wall shear stress in small arterial vessels and highly branched sinusoidal capillaries, and the cellular dynamics of hematopoietic stem and progenitor cell homing.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Células de la Médula Ósea/fisiología , Médula Ósea/fisiología , Células Madre Hematopoyéticas/fisiología , Microvasos/fisiología , Animales , Movimiento Celular/fisiología , Hemodinámica/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Resistencia al Corte/fisiología , Estrés Fisiológico/fisiología
19.
Bonekey Rep ; 5: 851, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28018584

RESUMEN

Confocal and two-photon microscopy has been widely used in bone research to not only produce high quality, three-dimensional images but also to provide valuable structural and quantitative information. In this article, we describe step-by-step protocols for confocal and two-photon microscopy to investigate earlier cellular events during colonisation of cancer cells in bone using xenograft mouse models. This includes confocal/two-photon microscopy imaging of paraformaldehyde fixed thick bone sections and frozen bone samples.

20.
Nat Commun ; 7: 13601, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27922003

RESUMEN

While blood vessels play important roles in bone homeostasis and repair, fundamental aspects of vascular function in the skeletal system remain poorly understood. Here we show that the long bone vasculature generates a peculiar flow pattern, which is important for proper angiogenesis. Intravital imaging reveals that vessel growth in murine long bone involves the extension and anastomotic fusion of endothelial buds. Impaired blood flow leads to defective angiogenesis and osteogenesis, and downregulation of Notch signalling in endothelial cells. In aged mice, skeletal blood flow and endothelial Notch activity are also reduced leading to decreased angiogenesis and osteogenesis, which is reverted by genetic reactivation of Notch. Blood flow and angiogenesis in aged mice are also enhanced on administration of bisphosphonate, a class of drugs frequently used for the treatment of osteoporosis. We propose that blood flow and endothelial Notch signalling are key factors controlling ageing processes in the skeletal system.


Asunto(s)
Huesos/irrigación sanguínea , Osteogénesis , Flujo Sanguíneo Regional/fisiología , Alendronato/farmacología , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Huesos/efectos de los fármacos , Huesos/fisiología , Difosfonatos/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Neovascularización Fisiológica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Receptores Notch/metabolismo , Flujo Sanguíneo Regional/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA