Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 867
Filter
1.
Curr Osteoporos Rep ; 22(1): 11-27, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38198033

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to highlight the evidence of microvascular dysfunction in bone and marrow and its relation to poor skeletal outcomes in diabetes mellitus. RECENT FINDINGS: Diabetes mellitus is characterized by chronic hyperglycemia, which may lead to microangiopathy and macroangiopathy. Micro- and macroangiopathy have been diagnosed in Type 1 and Type 2 diabetes, coinciding with osteopenia, osteoporosis, enhanced fracture risk and delayed fracture healing. Microangiopathy has been reported in the skeleton, correlating with reduced blood flow and perfusion, vasomotor dysfunction, microvascular rarefaction, reduced angiogenic capabilities, and augmented vascular permeability. Microangiopathy within the skeleton may be detrimental to bone and manifest as, among other clinical abnormalities, reduced mass, enhanced fracture risk, and delayed fracture healing. More investigations are required to elucidate the various mechanisms by which diabetic microvascular dysfunction impacts the skeleton.


Subject(s)
Diabetes Mellitus, Type 2 , Fractures, Bone , Humans , Diabetes Mellitus, Type 2/complications , Bone Marrow/blood supply , Bone and Bones , Microvessels
2.
Adv Healthc Mater ; 12(8): e2201830, 2023 03.
Article in English | MEDLINE | ID: mdl-36521080

ABSTRACT

The mechanical stimuli generated by body exercise can be transmitted from cortical bone into the deep bone marrow (mechanopropagation). Excitingly, a mechanosensitive perivascular stem cell niche is recently identified within the bone marrow for osteogenesis and lymphopoiesis. Although it is long known that they are maintained by exercise-induced mechanical stimulation, the mechanopropagation from compact bone to deep bone marrow vasculature remains elusive of this fundamental mechanobiology field. No experimental system is available yet to directly understand such exercise-induced mechanopropagation at the bone-vessel interface. To this end, taking advantage of the revolutionary in vivo 3D deep bone imaging, an integrated computational biomechanics framework to quantitatively evaluate the mechanopropagation capabilities for bone marrow arterioles, arteries, and sinusoids is devised. As a highlight, the 3D geometries of blood vessels are smoothly reconstructed in the presence of vessel wall thickness and intravascular pulse pressure. By implementing the 5-parameter Mooney-Rivlin model that simulates the hyperelastic vessel properties, finite element analysis to thoroughly investigate the mechanical effects of exercise-induced intravascular vibratory stretching on bone marrow vasculature is performed. In addition, the blood pressure and cortical bone bending effects on vascular mechanoproperties are examined. For the first time, movement-induced mechanopropagation from the hard cortical bone to the soft vasculature in the bone marrow is numerically simulated. It is concluded that arterioles and arteries are much more efficient in propagating mechanical force than sinusoids due to their stiffness. In the future, this in-silico approach can be combined with other clinical imaging modalities for subject/patient-specific vascular reconstruction and biomechanical analysis, providing large-scale phenotypic data for personalized mechanobiology discovery.


Subject(s)
Bone Marrow , Tomography, X-Ray Computed , Humans , Bone Marrow/blood supply , Biomechanical Phenomena , Arterioles , Bone and Bones
3.
Nat Commun ; 13(1): 393, 2022 01 19.
Article in English | MEDLINE | ID: mdl-35046411

ABSTRACT

The fate of hematopoietic stem cells (HSCs) can be directed by microenvironmental factors including extracellular calcium ion concentration ([Ca2+]e), but the local [Ca2+]e around individual HSCs in vivo remains unknown. Here we develop intravital ratiometric analyses to quantify the absolute pH and [Ca2+]e in the mouse calvarial bone marrow, taking into account the pH sensitivity of the calcium probe and the wavelength-dependent optical loss through bone. Unexpectedly, the mean [Ca2+]e in the bone marrow (1.0 ± 0.54 mM) is not significantly different from the blood serum, but the HSCs are found in locations with elevated local [Ca2+]e (1.5 ± 0.57 mM). With aging, a significant increase in [Ca2+]e is found in M-type cavities that exclusively support clonal expansion of activated HSCs. This work thus establishes a tool to investigate [Ca2+]e and pH in the HSC niche with high spatial resolution and can be broadly applied to other tissue types.


Subject(s)
Bone Marrow/diagnostic imaging , Bone Marrow/metabolism , Calcium/metabolism , Intravital Microscopy , Aging/metabolism , Animals , Benzopyrans/chemistry , Bone Marrow/blood supply , Bone Remodeling , Cellular Microenvironment , Fluorescence , Hematopoietic Stem Cells/metabolism , Hydrogen-Ion Concentration , Mice, Inbred C57BL , Naphthols/chemistry , Rhodamines/chemistry
4.
Sci Rep ; 12(1): 1439, 2022 01 27.
Article in English | MEDLINE | ID: mdl-35087109

ABSTRACT

Multiple myeloma (MM) is an incurable B cell malignancy characterized by the accumulation of monoclonal abnormal plasma cells in the bone marrow (BM). It has been a significant challenge to study the spatiotemporal interactions of MM cancer cells with the embedded microenvironments of BM. Here we report a microfluidic device which was designed to mimic several physiological features of the BM niche: (1) sinusoidal circulation, (2) sinusoidal endothelium, and (3) stroma. The endothelial and stromal compartments were constructed and used to demonstrate the device's utility by spatiotemporally characterizing the CXCL12-mediated egression of MM cells from the BM stroma and its effects on the barrier function of endothelial cells (ECs). We found that the egression of MM cells resulted in less organized and loosely connected ECs, the widening of EC junction pores, and increased permeability through ECs, but without significantly affecting the number density of viable ECs. The results suggest that the device can be used to study the physical and secreted factors determining the trafficking of cancer cells through BM. The sinusoidal flow feature of the device provides an integral element for further creating systemic models of cancers that reside or metastasize to the BM niche.


Subject(s)
Bone Marrow/pathology , Lab-On-A-Chip Devices , Multiple Myeloma/pathology , Spatio-Temporal Analysis , Bone Marrow/blood supply , Capillaries/cytology , Capillaries/pathology , Cell Line , Endothelial Cells , Humans , Tumor Microenvironment
5.
J Mol Med (Berl) ; 100(1): 53-61, 2022 01.
Article in English | MEDLINE | ID: mdl-34709407

ABSTRACT

Endothelial cells play a critical role in supporting postnatal hematopoiesis in the bone marrow. Unique endothelial cells, together with various perivascular cells, form different types of vascular structures, constructing a vast microvascular delivery and trafficking network for blood cells, oxygen, and nutrition. These blood vessels build distinct hematopoietic stem and progenitor cell niches, which offer not only sites of residence for blood cells but also indispensable signals directing HSPC homing, self-renewal, and multilineage differentiation. Deep insight into the structure and function of the BM vasculature niche and its participation in hematopoiesis is necessary to develop advanced strategies for the reconstitution of hematopoiesis.


Subject(s)
Bone Marrow/blood supply , Hematopoiesis , Microvessels , Animals , Endothelial Cells , Hematopoietic Stem Cells , Humans
6.
Nat Commun ; 12(1): 6963, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34845225

ABSTRACT

Within the bone marrow microenvironment, endothelial cells (EC) exert important functions. Arterial EC support hematopoiesis while H-type capillaries induce bone formation. Here, we show that BM sinusoidal EC (BM-SEC) actively control erythropoiesis. Mice with stabilized ß-catenin in BM-SEC (Ctnnb1OE-SEC) generated by using a BM-SEC-restricted Cre mouse line (Stab2-iCreF3) develop fatal anemia. While activation of Wnt-signaling in BM-SEC causes an increase in erythroblast subsets (PII-PIV), mature erythroid cells (PV) are reduced indicating impairment of terminal erythroid differentiation/reticulocyte maturation. Transplantation of Ctnnb1OE-SEC hematopoietic stem cells into wildtype recipients confirms lethal anemia to be caused by cell-extrinsic, endothelial-mediated effects. Ctnnb1OE-SEC BM-SEC reveal aberrant sinusoidal differentiation with altered EC gene expression and perisinusoidal ECM deposition and angiocrine dysregulation with de novo endothelial expression of FGF23 and DKK2, elevated in anemia and involved in vascular stabilization, respectively. Our study demonstrates that BM-SEC play an important role in the bone marrow microenvironment in health and disease.


Subject(s)
Anemia/genetics , Bone Marrow/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Endothelium, Vascular/metabolism , Erythroblasts/metabolism , Erythropoiesis/genetics , beta Catenin/genetics , Anemia/metabolism , Anemia/mortality , Anemia/pathology , Animals , Bone Marrow/blood supply , Capillaries/cytology , Capillaries/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Differentiation , Endothelial Cells/classification , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Erythroblasts/classification , Erythroblasts/cytology , Female , Fibroblast Growth Factor-23/genetics , Fibroblast Growth Factor-23/metabolism , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Integrases/genetics , Integrases/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Transgenic , Osteogenesis , Reticulocytes/cytology , Reticulocytes/metabolism , Survival Analysis , Wnt Signaling Pathway , beta Catenin/metabolism
7.
J Hematol Oncol ; 14(1): 122, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34372909

ABSTRACT

BACKGROUND: During acute myeloid leukemia (AML) growth, the bone marrow (BM) niche acquires significant vascular changes that can be offset by therapeutic blast cytoreduction. The molecular mechanisms of this vascular plasticity remain to be fully elucidated. Herein, we report on the changes that occur in the vascular compartment of the FLT3-ITD+ AML BM niche pre and post treatment and their impact on leukemic stem cells (LSCs). METHODS: BM vasculature was evaluated in FLT3-ITD+ AML models (MllPTD/WT/Flt3ITD/ITD mouse and patient-derived xenograft) by 3D confocal imaging of long bones, calvarium vascular permeability assays, and flow cytometry analysis. Cytokine levels were measured by Luminex assay and miR-126 levels evaluated by Q-RT-PCR and miRNA staining. Wild-type (wt) and MllPTD/WT/Flt3ITD/ITD mice with endothelial cell (EC) miR-126 knockout or overexpression served as controls. The impact of treatment-induced BM vascular changes on LSC activity was evaluated by secondary transplantation of BM cells after administration of tyrosine kinase inhibitors (TKIs) to MllPTD/WT/Flt3ITD/ITD mice with/without either EC miR-126 KO or co-treatment with tumor necrosis factor alpha (TNFα) or anti-miR-126 miRisten. RESULTS: In the normal BM niche, CD31+Sca-1high ECs lining arterioles have miR-126 levels higher than CD31+Sca-1low ECs lining sinusoids. We noted that during FLT3-ITD+ AML growth, the BM niche lost arterioles and gained sinusoids. These changes were mediated by TNFα, a cytokine produced by AML blasts, which induced EC miR-126 downregulation and caused depletion of CD31+Sca-1high ECs and gain in CD31+Sca-1low ECs. Loss of miR-126high ECs led to a decreased EC miR-126 supply to LSCs, which then entered the cell cycle and promoted leukemia growth. Accordingly, antileukemic treatment with TKI decreased the BM blast-produced TNFα and increased miR-126high ECs and the EC miR-126 supply to LSCs. High miR-126 levels safeguarded LSCs, as shown by more severe disease in secondary transplanted mice. Conversely, EC miR-126 deprivation via genetic or pharmacological EC miR-126 knock-down prevented treatment-induced BM miR-126high EC expansion and in turn LSC protection. CONCLUSIONS: Treatment-induced CD31+Sca-1high EC re-vascularization of the leukemic BM niche may represent a LSC extrinsic mechanism of treatment resistance that can be overcome with therapeutic EC miR-126 deprivation.


Subject(s)
Bone Marrow/pathology , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Neoplastic Stem Cells/pathology , Animals , Bone Marrow/blood supply , Humans , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Mice , Mice, Inbred C57BL , Up-Regulation , fms-Like Tyrosine Kinase 3/genetics
8.
Neurosci Lett ; 763: 136179, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34416344

ABSTRACT

Neural differentiation of mesenchymal stromal cells has been widely studied. However, a comparative characterization of ultrastructural changes during neural differentiation has not been performed. In this study, we conducted scanning electron microscopy and transmission electron microscopy analysis to show the morphological changes in mesenchymal stromal cells upon induction of neural differentiation. In addition, transmission electron microscopy results demonstrated ultrastructural differences between human cranial bone marrow mesenchymal stromal cells and iliac crest bone marrow mesenchymal stromal cells. We propose that enriched microvesicles in cranial bone marrow mesenchymal stromal cells may be responsible for the increased efficiency of neural differentiation.


Subject(s)
Mesenchymal Stem Cells/ultrastructure , Neurogenesis , Skull/cytology , Bone Marrow/blood supply , Cells, Cultured , Humans , Ilium/cytology , Mesenchymal Stem Cells/physiology , Microscopy, Electron, Transmission , Microvessels/anatomy & histology , Primary Cell Culture
9.
Front Immunol ; 12: 658535, 2021.
Article in English | MEDLINE | ID: mdl-33936091

ABSTRACT

The bone marrow is a complex ecosystem in which hematopoietic and non-hematopoietic cells reside. In this review, we discuss the bone marrow niches in mice that facilitate the survival, maintenance, and differentiation of cells of hematopoietic origin based on the recent literature. Our review places a special focus on the hematopoietic multipotent progenitors and on plasma cells, corresponding to the last stage of the B-cell lineage, that play a key role in the humoral memory response. We highlight the similarities between the microenvironments necessary for the establishment and the maintenance of these two immune cell subsets, and how the chemokine CXCL12/CXCR4 signaling axis contributes to these processes. Finally, we bring elements to address the following question: are multipotent progenitors and plasma cells neighbors or roommates within the bone marrow?


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Lymphopoiesis , Plasma Cells/cytology , Plasma Cells/metabolism , Animals , Biomarkers , Bone Marrow/blood supply , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Communication , Cellular Microenvironment , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Stem Cell Niche
10.
Methods Mol Biol ; 2308: 139-150, 2021.
Article in English | MEDLINE | ID: mdl-34057721

ABSTRACT

The bone marrow is the major hematopoietic organ, consisting of distinct microenvironmental niches for the production of hematopoietic cells. Advanced visualizing methods are required to define and better understand the interactions between stromal and hematopoietic cells. In this chapter, we describe an ex vivo whole-mount imaging technique of the bone marrow, which allows for a fast, high-quality, and three-dimensional visualization of different bone marrow components. We provide a guide for conducting adoptive transfer experiments of fluorescently labeled leukocytes and visualizing their location in the bone marrow with respect to the bone marrow vasculature. This method presents a quick, easy, and inexpensive approach to image the bone marrow in three dimensions.


Subject(s)
Bone Marrow/blood supply , Cell Movement , Imaging, Three-Dimensional , Leukocytes/physiology , Microscopy, Confocal , Microscopy, Fluorescence , Tissue Embedding , Adoptive Transfer , Animals , Cellular Microenvironment , Fluorescent Dyes/metabolism , Image Processing, Computer-Assisted , Leukocytes/metabolism , Mice , Microtomy
11.
Leukemia ; 35(12): 3509-3525, 2021 12.
Article in English | MEDLINE | ID: mdl-34007044

ABSTRACT

Bone marrow (BM) angiogenesis significantly influences disease progression in multiple myeloma (MM) patients and correlates with adverse prognosis. The present study shows a statistically significant correlation of the AP-1 family member JunB with VEGF, VEGFB, and IGF1 expression levels in MM. In contrast to the angiogenic master regulator Hif-1α, JunB protein levels were independent of hypoxia. Results in tumor-cell models that allow the induction of JunB knockdown or JunB activation, respectively, corroborated the functional role of JunB in the production and secretion of these angiogenic factors (AFs). Consequently, conditioned media derived from MM cells after JunB knockdown or JunB activation either inhibited or stimulated in vitro angiogenesis. The impact of JunB on MM BM angiogenesis was finally confirmed in a dynamic 3D model of the BM microenvironment, a xenograft mouse model as well as in patient-derived BM sections. In summary, in continuation of our previous study (Fan et al., 2017), the present report reveals for the first time that JunB is not only a mediator of MM cell survival, proliferation, and drug resistance, but also a promoter of AF transcription and consequently of MM BM angiogenesis. Our results thereby underscore worldwide efforts to target AP-1 transcription factors such as JunB as a promising strategy in MM therapy.


Subject(s)
Bone Marrow/blood supply , Multiple Myeloma/blood supply , Transcription Factors/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line, Tumor , Female , Heterografts , Humans , Insulin-Like Growth Factor I/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Primary Cell Culture , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor B/metabolism
13.
Ann Hematol ; 100(5): 1169-1179, 2021 May.
Article in English | MEDLINE | ID: mdl-33704530

ABSTRACT

This translational study aimed at gaining insight into the effects of lenalidomide in acute myeloid leukemia (AML). Forty-one AML patients aged 66 or older of the Swiss cohort of the HOVON-103 AML/SAKK30/10 study were included. After randomization, they received standard induction chemotherapy with or without lenalidomide. Bone marrow biopsies at diagnosis and before the 2nd induction cycle were obtained to assess the therapeutic impact on leukemic blasts and microenvironment. Increased bone marrow angiogenesis, as assessed by microvessel density (MVD), was found at AML diagnosis and differed significantly between the WHO categories. Morphological analysis revealed a higher initial MVD in AML with myelodysplasia-related changes (AML-MRC) and a more substantial decrease of microvascularization after lenalidomide exposure. A slight increase of T-bet-positive TH1-equivalents was identifiable under lenalidomide. In the subgroup of patients with AML-MRC, the progression-free survival differed between the two treatment regimens, showing a potential but not significant benefit of lenalidomide. We found no correlation between the cereblon genotype (the target of lenalidomide) and treatment response or prognosis. In conclusion, addition of lenalidomide may be beneficial to elderly patients suffering from AML-MRC, where it leads to a reduction of microvascularization and, probably, to an intensified specific T cell-driven anti-leukemic response.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bone Marrow/drug effects , Lenalidomide/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Tumor Microenvironment/drug effects , Aged , Bone Marrow/blood supply , Bone Marrow/pathology , Cohort Studies , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
14.
Osteoarthritis Cartilage ; 29(5): 718-727, 2021 05.
Article in English | MEDLINE | ID: mdl-33577958

ABSTRACT

OBJECTIVE: The vascularization of subchondral bone plays a significant role in the progression of knee osteoarthritis (OA). Treatment with platelet-rich plasma (PRP) has positive effects on cartilage lesions. However, PRP's efficacy for subchondral bone marrow lesions and the relationship of these lesions to cartilage are still undiscovered. Therefore, our aims were first to longitudinally investigate the change in subchondral flow by dynamic contrast enhanced MRI and degeneration of cartilage by MRI T2∗ in an anterior cruciate transection rodent (ACLT) model, and second to examine changes in parameters after intra-articular PRP injection. DESIGN: A 32-week investigation in 18 rats allocated to sham-control, ACLT with normal saline injection (ACLT + NS), and ACLT with PRP injection groups ended with histological evaluation. Another rat was used as a donor of allogenic PRP. RESULTS: Compared to the sham-control group, the ACLT + NS group had higher subchondral blood volume A (0.051, 95% confidence interval: 0.009, 0.092) and lower venous washout kel (-0.030: -0.055, -0.005) from week 4; lower permeability kep from week 18 (-0.954: -1.339, -0.569); higher cartilage T2∗ values (1.803: 1.504, 2.102) reflecting collagen loss beginning at week 10. For the PRP treatment group, subchondral bone marrow A and cartilage T2∗ decreased from week 10. Histological results confirmed and were correlated with the MRI findings. CONCLUSION: Subchondral hyper-perfusion plays a vital role in the pathogenesis of OA and was associated with cartilage degeneration. The efficacy of PRP can be observed from reduced perfusion and MRI T2∗ values.


Subject(s)
Bone Marrow/blood supply , Bone Marrow/diagnostic imaging , Cartilage, Articular/diagnostic imaging , Magnetic Resonance Imaging , Platelet-Rich Plasma , Animals , Blood Volume , Disease Models, Animal , Injections, Intra-Articular , Osteoarthritis/diagnostic imaging , Osteoarthritis/therapy , Rats, Sprague-Dawley , Stifle/blood supply , Stifle/diagnostic imaging
16.
Pediatr Res ; 90(2): 341-346, 2021 08.
Article in English | MEDLINE | ID: mdl-33469189

ABSTRACT

BACKGROUND: Iron deficiency anemia (IDA)-induced reactive thrombocytosis can occur in children and adults. The underlying mechanism for this phenomenon is indeterminate. Traditional cytokines such as thrombopoietin (TPO), interleukin-6 (IL-6), and IL-11 involved in megakaryopoiesis have not been shown to be the cause. Recent studies suggest that growth factors and signaling molecules involved with angiogenesis influence the proliferation and differentiation of megakaryocytes. METHODS: We investigated the possible association between angiogenic cytokines with reactive thrombocytosis due to IDA in an iron-deficient (ID) rat model. Complete blood count, iron panels, and TPO levels were measured at baseline and 5 weeks later in both control (C) and ID rats. Angiogenic cytokines were evaluated in the bone marrow in all rats. RESULTS: We successfully induced IDA in our rats by phlebotomy and reduced iron diet. We did not find an increase of TPO in ID rats. A review of the bone marrow showed an increase in the number of megakaryocytes, vascular structures, as well as increased intensity of stain for vascular endothelial growth factor (VEGF), and CXC chemokine receptor 4 (CXCR4) in rats with IDA compared to controls. CONCLUSIONS: Our results of histological bone marrow data suggest an important role for angiogenesis in the development of IDA-induced thrombocytosis. IMPACT: Thrombocytosis is common with IDA in both children and adults, but the mechanism is unclear. We confirmed that TPO is not the major driver of iron deficiency-associated thrombocytosis. We confirmed the increase in the number of megakaryocytes in the bone marrow despite stable TPO levels. We provided evidence supporting an important role of angiogenesis in megakaryocytopoiesis/thrombopoiesis with increased vascular structures and angiogenic cytokines in the bone marrow of iron-deficient rats. The demonstration that angiogenesis may play an important role in secondary thrombocytosis could lead to a new approach in treating symptomatic reactive thrombocytosis by targeting angiogenesis.


Subject(s)
Anemia, Iron-Deficiency/complications , Bone Marrow/blood supply , Megakaryocytes/metabolism , Neovascularization, Pathologic , Receptors, CXCR4/metabolism , Thrombocytosis/etiology , Thrombopoiesis , Vascular Endothelial Growth Factor A/metabolism , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/pathology , Animals , Disease Models, Animal , Male , Megakaryocytes/pathology , Rats, Sprague-Dawley , Signal Transduction , Thrombocytosis/blood , Thrombocytosis/pathology , Thrombopoietin/metabolism
17.
Int J Radiat Oncol Biol Phys ; 109(1): 60-72, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32841681

ABSTRACT

PURPOSE: Although vascular alterations in solid tumor malignancies are known to decrease therapeutic delivery, the effects of leukemia-induced bone marrow vasculature (BMV) alterations on therapeutic delivery are not well known. Additionally, functional quantitative measurements of the leukemic BMV during chemotherapy and radiation therapy are limited, largely due to a lack of high-resolution imaging techniques available preclinically. This study develops a murine model using compartmental modeling for quantitative multiphoton microscopy (QMPM) to characterize the malignant BMV before and during treatment. METHODS AND MATERIALS: Using QMPM, live time-lapsed images of dextran leakage from the local BMV to the surrounding bone marrow of mice bearing acute lymphoblastic leukemia (ALL) were taken and fit to a 2-compartment model to measure the transfer rate (Ktrans), fractional extracellular extravascular space (νec), and vascular permeability parameters, as well as functional single-vessel characteristics. In response to leukemia-induced BMV alterations, the effects of 2 to 4 Gy low-dose radiation therapy (LDRT) on the BMV, drug delivery, and mouse survival were assessed post-treatment to determine whether neoadjuvant LDRT before chemotherapy improves treatment outcome. RESULTS: Mice bearing ALL had significantly altered Ktrans, increased νec, and increased permeability compared with healthy mice. Angiogenesis, decreased single-vessel perfusion, and decreased vessel diameter were observed. BMV alterations resulted in disease-dependent reductions in cellular uptake of Hoechst dye. LDRT to mice bearing ALL dilated BMV, increased single-vessel perfusion, and increased daunorubicin uptake by ALL cells. Consequently, LDRT administered to mice before receiving nilotinib significantly increased survival compared with mice receiving LDRT after nilotinib, demonstrating the importance of LDRT conditioning before therapeutic administration. CONCLUSION: The developed QMPM enables single-platform assessments of the pharmacokinetics of fluorescent agents and characterization of the BMV. Initial results suggest BMV alterations after neoadjuvant LDRT may contribute to enhanced drug delivery and increased treatment efficacy for ALL. The developed QMPM enables observations of the BMV for use in ALL treatment optimization.


Subject(s)
Bone Marrow/blood supply , Neoadjuvant Therapy , Neovascularization, Pathologic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Radiation Dosage , Animals , Cell Line, Tumor , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Radiotherapy Dosage , Tumor Microenvironment/radiation effects
19.
Sci Rep ; 10(1): 17319, 2020 10 14.
Article in English | MEDLINE | ID: mdl-33057033

ABSTRACT

Multiple Myeloma (MM) induces bone destruction, decreases bone formation, and increases marrow angiogenesis in patients. We reported that osteocytes (Ocys) directly interact with MM cells to increase tumor growth and expression of Ocy-derived factors that promote bone resorption and suppress bone formation. However, the contribution of Ocys to enhanced marrow vascularization in MM is unclear. Since the MM microenvironment is hypoxic, we assessed if hypoxia and/or interactions with MM cells increases pro-angiogenic signaling in Ocys. Hypoxia and/or co-culture with MM cells significantly increased Vegf-a expression in MLOA5-Ocys, and conditioned media (CM) from MLOA5s or MM-MLOA5 co-cultured in hypoxia, significantly increased endothelial tube length compared to normoxic CM. Further, Vegf-a knockdown in MLOA5s or primary Ocys co-cultured with MM cells or neutralizing Vegf-a in MM-Ocy co-culture CM completely blocked the increased endothelial activity. Importantly, Vegf-a-expressing Ocy numbers were significantly increased in MM-injected mouse bones, positively correlating with tumor vessel area. Finally, we demonstrate that direct contact with MM cells increases Ocy Fgf23, which enhanced Vegf-a expression in Ocys. Fgf23 deletion in Ocys blocked these changes. These results suggest hypoxia and MM cells induce a pro-angiogenic phenotype in Ocys via Fgf23 and Vegf-a signaling, which can promote MM-induced marrow vascularization.


Subject(s)
Bone Marrow/blood supply , Fibroblast Growth Factors/physiology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neovascularization, Pathologic/genetics , Osteocytes/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Bone Resorption/etiology , Cell Line , Female , Fibroblast Growth Factor-23 , Gene Expression/genetics , Humans , Mice , Mice, Inbred C57BL , Osteocytes/metabolism , Osteogenesis , Tumor Microenvironment
20.
Eur J Radiol ; 131: 109206, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32871293

ABSTRACT

PURPOSE: To describe bone perfusion and adiposity beyond the necrotic zone with quantitative MRI techniques in osteonecrosis of the femoral head (ONFH). METHOD: In this cross-sectional multicentre study, we recruited patients suffering from late-stage ONFH or hip osteoarthritis. Hip MRI included quantitative MRI sequences: chemical-shift imaging and dynamic contrast-enhanced MRI. We drew regions of interest inside the necrotic zone (inner necrosis and its border) and outside (femoral head, neck and greater trochanter) in ONFH. In the control group, regions of interest were drawn in the femoral head, femoral neck and the greater trochanter. For each region of interest, we measured fat fraction, and calculated semi-quantitative (area under the curve, initial slope) and pharmacokinetic perfusion parameters (Ktrans and Kep). RESULTS: Thirty-two male adults (mean age 58 ±â€¯9 years, range 38-74 years) were included. Sixteen patients formed the ONFH group and fifteen the control group; one was excluded. In the normal-appearing non-necrotic part of the femoral head, fat fraction was not significantly different in comparison with controls (p = 1), but Ktrans was significantly lower than in controls (0.012 ±â€¯0.018 vs. 0.027 ±â€¯0.045; p = 0.05). This perfusion parameter reflects exchanges between blood microvessels and bone marrow. CONCLUSIONS: Our results question the concept of adipose toxicity on the macroscopic scale, and bring up the concept of regional ischemic penumbra that goes beyond the visible necrotic zone. Further studies are required to test these hypotheses in larger populations and earlier disease states.


Subject(s)
Adiposity , Femur Head Necrosis/diagnostic imaging , Femur Head/blood supply , Magnetic Resonance Imaging , Adult , Aged , Area Under Curve , Body Mass Index , Bone Marrow/blood supply , Bone Marrow/diagnostic imaging , Bone Marrow/pathology , Cross-Sectional Studies , Femur/blood supply , Femur/diagnostic imaging , Femur Head/diagnostic imaging , Femur Head/pathology , Femur Head Necrosis/pathology , Femur Head Necrosis/physiopathology , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Osteoarthritis, Hip/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...