Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 143(25): 2666-2670, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38635757

ABSTRACT

ABSTRACT: Lysyl oxidase (LOX) is a facilitator of extracellular matrix cross-linking. Using newly developed megakaryocyte-specific LOX knockout mice, we show that LOX expressed in these scarce bone marrow cells affects bone volume and collagen architecture in a sex-dependent manner.


Subject(s)
Megakaryocytes , Mice, Knockout , Protein-Lysine 6-Oxidase , Animals , Protein-Lysine 6-Oxidase/metabolism , Protein-Lysine 6-Oxidase/genetics , Megakaryocytes/metabolism , Megakaryocytes/cytology , Mice , Male , Female , Bone and Bones/metabolism , Sex Characteristics , Collagen/metabolism , Gene Deletion , Sex Factors , Extracellular Matrix Proteins
2.
Blood ; 139(21): 3127-3137, 2022 05 26.
Article in English | MEDLINE | ID: mdl-34428274

ABSTRACT

Myeloproliferative neoplasms (MPNs) are a heterogeneous group of chronic hematological diseases that arise from the clonal expansion of abnormal hematopoietic stem cells, of which polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) have been extensively reviewed in the context of control of clonal expansion, fibrosis, and other phenotypes. Herein, we review current knowledge on the influence of different forms of MPN on bone health. In studies, murine models and human data have implicated various degrees of effect of different forms of MPN on bone density and on osteoblast proliferation and differentiation. Most results have shown that bone volume is generally increased in patients with PMF, whereas it is slightly decreased or not altered in patients with ET or PV, although possible differences between male and female phenotypes were not fully explored in most MPN forms. Osteosclerosis in patients with PMF is a serious complication that can lead to bone marrow failure, and the loss of bone reported in some patients with ET or PV can lead to osteoporotic fractures. Some MPN forms are associated with an increased number of megakaryocytes (MKs), and several of the MK-associated factors in MPN are known to affect bone development. We review known mechanisms involved in these processes, with a focus on the role of MKs and secreted factors. Understanding MPN-associated changes in bone health could improve early intervention and treatment of this side effect of the pathology.


Subject(s)
Myeloproliferative Disorders , Polycythemia Vera , Thrombocythemia, Essential , Animals , Female , Homeostasis , Humans , Male , Megakaryocytes/pathology , Mice , Myeloproliferative Disorders/genetics , Polycythemia Vera/genetics , Thrombocythemia, Essential/genetics
3.
Am J Hematol ; 99(7): 1230-1239, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38654461

ABSTRACT

Venous thromboembolism (VTE) poses a significant risk to cancer patients receiving systemic therapy. The generalizability of pan-cancer models to lymphomas is limited. Currently, there are no reliable risk prediction models for thrombosis in patients with lymphoma. Our objective was to create a risk assessment model (RAM) specifically for lymphomas. We performed a retrospective cohort study to develop Fine and Gray sub-distribution hazard model for VTE and pulmonary embolism (PE)/ lower extremity deep vein thrombosis (LE-DVT) respectively in adult lymphoma patients from the Veterans Affairs national healthcare system (VA). External validations were performed at the Harris Health System (HHS) and the MD Anderson Cancer Center (MDACC). Time-dependent c-statistic and calibration curves were used to assess discrimination and fit. There were 10,313 (VA), 854 (HHS), and 1858 (MDACC) patients in the derivation and validation cohorts with diverse baseline. At 6 months, the VTE incidence was 5.8% (VA), 8.2% (HHS), and 8.8% (MDACC), respectively. The corresponding estimates for PE/LE-DVT were 3.9% (VA), 4.5% (HHS), and 3.7% (MDACC), respectively. The variables in the final RAM included lymphoma histology, body mass index, therapy type, recent hospitalization, history of VTE, history of paralysis/immobilization, and time to treatment initiation. The RAM had c-statistics of 0.68 in the derivation and 0.69 and 0.72 in the two external validation cohorts. The two models achieved a clear differentiation in risk stratification in each cohort. Our findings suggest that easy-to-implement, clinical-based model could be used to predict personalized VTE risk for lymphoma patients.


Subject(s)
Lymphoma , Venous Thromboembolism , Humans , Retrospective Studies , Lymphoma/complications , Lymphoma/epidemiology , Middle Aged , Female , Male , Aged , Risk Assessment , Venous Thromboembolism/etiology , Venous Thromboembolism/epidemiology , Adult , Pulmonary Embolism/etiology , Pulmonary Embolism/epidemiology , Venous Thrombosis/etiology , Venous Thrombosis/epidemiology , Risk Factors , Incidence , Aged, 80 and over
4.
Am J Hematol ; 99(3): 336-349, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38165047

ABSTRACT

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.


Subject(s)
Primary Myelofibrosis , Thrombocythemia, Essential , Humans , Animals , Mice , Megakaryocytes/metabolism , Primary Myelofibrosis/genetics , Bone Marrow , Thrombopoiesis/genetics , Thrombocythemia, Essential/metabolism , Blood Platelets/metabolism , Ion Channels/genetics , Ion Channels/metabolism
5.
Mol Cell Proteomics ; 21(4): 100213, 2022 04.
Article in English | MEDLINE | ID: mdl-35182768

ABSTRACT

Primary myelofibrosis (PMF) is a neoplasm prone to leukemic transformation, for which limited treatment is available. Among individuals diagnosed with PMF, the most prevalent mutation is the JAK2V617F somatic point mutation that activates the Janus kinase 2 (JAK2) enzyme. Our earlier reports on hyperactivity of ß1 integrin and enhanced adhesion activity of the α2ß1 complex in JAK2V617F megakaryocytes (MKs) led us to examine the new hypothesis that this mutation leads to posttranslational modification via changes in glycosylation. Samples were derived from immunoprecipitation of MKs obtained from Vav1-hJAK2V617F and WT mice. Immunoprecipitated fractions were separated by SDS-PAGE and analyzed using LC-MS/MS techniques in a bottom-up glycoproteomics workflow. In the immunoprecipitate, glycopeptiforms corresponding to 11 out of the 12 potential N-glycosylation sites of integrin ß1 and to all nine potential glycosylation sites of integrin α2 were observed. Glycopeptiforms were compared across WT and JAK2V617F phenotypes for both integrins. The overall trend observed is that JAK2V617F mutation in PMF MKs leads to changes in ß1 glycosylation; in most cases, it results in an increase in the integrated area of glycopeptiforms. We also observed that in mutated MKs, changes in integrin α2 glycosylation were more substantial than those observed for integrin ß1 glycosylation, a finding that suggests that altered integrin α2 glycosylation may also affect activation. Additionally, the identification of proteins associated to the cytoskeleton that were co-immunoprecipitated with integrins α2 and ß1 demonstrated the potential of the methodology employed in this study to provide some insight, at the peptide level, into the consequences of integrin activation in MKs. The extensive and detailed glycosylation patterns we uncovered provide a basis for future functional studies of each site in control cells as compared to JAK2V617F-mutated cells. Data are available via ProteomeXchange with identifier PXD030550.


Subject(s)
Janus Kinase 2/genetics , Megakaryocytes , Primary Myelofibrosis , Animals , Chromatography, Liquid , Integrin alpha2/genetics , Integrin alpha2/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Megakaryocytes/metabolism , Mice , Mutation , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/genetics , Tandem Mass Spectrometry
6.
Am J Hematol ; 98(8): 1214-1222, 2023 08.
Article in English | MEDLINE | ID: mdl-37161855

ABSTRACT

It remains unclear if immune checkpoint inhibitor (ICI) therapy is associated with higher rate of venous thromboembolism (VTE) compared with cytotoxic chemotherapy (chemo) in patients with comparable cancer type, staging, and comorbidities. Using the national Veterans Affairs healthcare system database from 2016 to 2021, we performed a propensity score (PS)-weighted retrospective cohort study to compare the incidence of VTE in patients with selected stage III/IV cancer receiving first-line ICI versus chemo. The PS model utilized overlap weights to balance age, sex, race, treatment year, VTE history, paralysis/immobilization, prolonged hospitalization, cancer type, staging, time between diagnosis and treatment, and National Cancer Institute comorbidity index. Weighted Cox regressions with robust standard error were used to assess the hazard ratio (HR) and 95% confidence interval (CI). We found that among comparable advanced cancers, first-line ICI (n = 1823) and first-line chemo (n = 6345) had similar rates of VTE (8.49% for ICI and 8.36% for chemo at 6 months). The weighted HR was 1.06 (95% CI 0.88-1.26) for ICI versus chemo. In a subgroup analysis restricted to lung cancers, first-line ICI/chemo (n = 828), ICI monotherapy (n = 428), and chemo monotherapy (n = 4371) had similar rates of VTE (9.60% for ICI/chemo, 10.04% for ICI, and 8.91% for chemo at 6 months). The weighted HR was 1.05 (95% CI 0.77-1.42) for ICI versus chemo, and 1.08 (95% CI 0.83-1.42) for ICI/chemo versus chemo. In conclusion, ICI as a systemic therapy has a similarly elevated risk as cytotoxic chemo for VTE occurrence in cancer patients. This finding can inform future prospective studies exploring thromboprophylaxis strategies.


Subject(s)
Antineoplastic Agents , Immune Checkpoint Inhibitors , Venous Thromboembolism , Humans , Venous Thromboembolism/epidemiology , Venous Thromboembolism/etiology , Neoplasms/therapy , Antineoplastic Agents/therapeutic use , Retrospective Studies , Incidence , Male , Female , Middle Aged , Aged , Aged, 80 and over
7.
Blood ; 135(25): 2286-2291, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32294178

ABSTRACT

Excessive accumulation of extracellular matrix (ECM) is a hallmark of bone marrow (BM) milieu in primary myelofibrosis (PMF). Because cells have the ability to adhere to the surrounding ECM through integrin receptors, we examined the hypothesis that an abnormal ECM-integrin receptor axis contributes to BM megakaryocytosis in JAK2V617F+ PMF. Secretion of ECM protein fibronectin (FN) by BM stromal cells from PMF patients correlates with fibrosis and disease severity. Here, we show that Vav1-hJAK2V617F transgenic mice (JAK2V617F+) have high BM FN content associated with megakaryocytosis and fibrosis. Further, megakaryocytes from JAK2V617F+ mice have increased cell surface expression of the α5 subunit of the α5ß1 integrin, the major FN receptor in megakaryocytes, and augmented adhesion to FN compared with wild-type controls. Reducing adhesion to FN by an inhibitory antibody to the α5 subunit effectively reduces the percentage of CD41+ JAK2V617F+ megakaryocytes in vitro and in vivo. Corroborating our findings in mice, JAK2V617F+ megakaryocytes from patients showed elevated expression of α5 subunit, and a neutralizing antibody to α5 subunit reduced adhesion to FN and megakaryocyte number derived from CD34+ cells. Our findings reveal a previously unappreciated contribution of FN-α5ß1 integrin to megakaryocytosis in JAK2V617F+ PMF.


Subject(s)
Integrin alpha5beta1/physiology , Megakaryocytes/pathology , Primary Myelofibrosis/pathology , Animals , Bone Marrow/metabolism , Cell Adhesion , Cells, Cultured , Extracellular Matrix/metabolism , Female , Humans , Integrin alpha5/biosynthesis , Integrin alpha5/genetics , Integrin alpha5/immunology , Integrin alpha5beta1/antagonists & inhibitors , Janus Kinase 2/genetics , Male , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Primary Myelofibrosis/genetics
8.
J Am Soc Nephrol ; 32(11): 2834-2850, 2021 11.
Article in English | MEDLINE | ID: mdl-34716244

ABSTRACT

BACKGROUND: CKD, characterized by retained uremic solutes, is a strong and independent risk factor for thrombosis after vascular procedures . Urem ic solutes such as indoxyl sulfate (IS) and kynurenine (Kyn) mediate prothrombotic effect through tissue factor (TF). IS and Kyn biogenesis depends on multiple enzymes, with therapeutic implications unexplored. We examined the role of indoleamine 2,3-dioxygenase-1 (IDO-1), a rate-limiting enzyme of kynurenine biogenesis, in CKD-associated thrombosis after vascular injury. METHODS: IDO-1 expression in mice and human vessels was examined. IDO-1-/- mice, IDO-1 inhibitors, an adenine-induced CKD, and carotid artery injury models were used. RESULTS: Both global IDO-1-/- CKD mice and IDO-1 inhibitor in wild-type CKD mice showed reduced blood Kyn levels, TF expression in their arteries, and thrombogenicity compared with respective controls. Several advanced IDO-1 inhibitors downregulated TF expression in primary human aortic vascular smooth muscle cells specifically in response to uremic serum. Further mechanistic probing of arteries from an IS-specific mouse model, and CKD mice, showed upregulation of IDO-1 protein, which was due to inhibition of its polyubiquitination and degradation by IS in vascular smooth muscle cells. In two cohorts of patients with advanced CKD, blood IDO-1 activity was significantly higher in sera of study participants who subsequently developed thrombosis after endovascular interventions or vascular surgery. CONCLUSION: Leveraging genetic and pharmacologic manipulation in experimental models and data from human studies implicate IS as an inducer of IDO-1 and a perpetuator of the thrombotic milieu and supports IDO-1 as an antithrombotic target in CKD.


Subject(s)
Indican/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/blood , Kynurenine/physiology , Molecular Targeted Therapy , Postoperative Complications/enzymology , Renal Insufficiency, Chronic/enzymology , Thrombosis/enzymology , Vascular Surgical Procedures/adverse effects , Animals , Aorta , Carotid Artery Injuries/complications , Carotid Artery Thrombosis/etiology , Carotid Artery Thrombosis/prevention & control , Culture Media/pharmacology , Enzyme Induction/drug effects , Feedback, Physiological , Female , HEK293 Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Kynurenine/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Postoperative Complications/blood , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Renal Insufficiency, Chronic/drug therapy , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/etiology , Thrombosis/prevention & control , Tryptophan/metabolism , Uremia/blood
9.
Blood ; 134(26): 2399-2413, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31877217

ABSTRACT

Patients with malignancy are at 4- to 7-fold higher risk of venous thromboembolism (VTE), a potentially fatal, yet preventable complication. Although general mechanisms of thrombosis are enhanced in these patients, malignancy-specific triggers and their therapeutic implication remain poorly understood. Here we examined a colon cancer-specific VTE model and probed a set of metabolites with prothrombotic propensity in the inferior vena cava (IVC) ligation model. Athymic mice injected with human colon adenocarcinoma cells exhibited significantly higher IVC clot weights, a biological readout of venous thrombogenicity, compared with the control mice. Targeted metabolomics analysis of plasma of mice revealed an increase in the blood levels of kynurenine and indoxyl sulfate (tryptophan metabolites) in xenograft-bearing mice, which correlated positively with the increase in the IVC clot size. These metabolites are ligands of aryl hydrocarbon receptor (AHR) signaling. Accordingly, plasma from the xenograft-bearing mice activated the AHR pathway and augmented tissue factor (TF) and plasminogen activator inhibitor 1 (PAI-1) levels in venous endothelial cells in an AHR-dependent manner. Consistent with these findings, the endothelium from the IVC of xenograft-bearing animals revealed nuclear AHR and upregulated TF and PAI-1 expression, telltale signs of an activated AHR-TF/PAI-1 axis. Importantly, pharmacological inhibition of AHR activity suppressed TF and PAI-1 expression in endothelial cells of the IVC and reduced clot weights in both kynurenine-injected and xenograft-bearing mice. Together, these data show dysregulated tryptophan metabolites in a mouse cancer model, and they reveal a novel link between these metabolites and the control of the AHR-TF/PAI-1 axis and VTE in cancer.


Subject(s)
Colonic Neoplasms/complications , Disease Models, Animal , Metabolome , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Thromboplastin/metabolism , Venous Thromboembolism/etiology , Animals , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , Male , Mice , Mice, Nude , Signal Transduction , Tryptophan/metabolism , Venous Thromboembolism/metabolism , Venous Thromboembolism/pathology , Xenograft Model Antitumor Assays
10.
Arterioscler Thromb Vasc Biol ; 40(10): e262-e272, 2020 10.
Article in English | MEDLINE | ID: mdl-32814440

ABSTRACT

OBJECTIVE: The risk of thrombosis in myeloproliferative neoplasms, such as primary myelofibrosis varies depending on the type of key driving mutation (JAK2 [janus kinase 2], CALR [calreticulin], and MPL [myeloproliferative leukemia protein or thrombopoietin receptor]) and the accompanying mutations in other genes. In the current study, we sought to examine the propensity for thrombosis, as well as platelet activation properties in a mouse model of primary myelofibrosis induced by JAK2V617F (janus kinase 2 with valine to phenylalanine substitution on codon 617) mutation. Approach and Results: Vav1-hJAK2V617F transgenic mice show hallmarks of primary myelofibrosis, including significant megakaryocytosis and bone marrow fibrosis, with a moderate increase in red blood cells and platelet number. This mouse model was used to study responses to 2 models of vascular injury and to investigate platelet properties. Platelets derived from the mutated mice have reduced aggregation in response to collagen, reduced thrombus formation and thrombus size, as demonstrated using laser-induced or FeCl3-induced vascular injury models, and increased bleeding time. Strikingly, the mutated platelets had a significantly reduced number of dense granules, which could explain impaired ADP secretion upon platelet activation, and a diminished second wave of activation. CONCLUSIONS: Together, our study highlights for the first time the influence of a hyperactive JAK2 on platelet activation-induced ADP secretion and dense granule homeostasis, with consequent effects on platelet activation properties.


Subject(s)
Blood Coagulation , Blood Platelets/enzymology , Carotid Artery Injuries/enzymology , Janus Kinase 2/blood , Megakaryocytes/enzymology , Platelet Activation , Primary Myelofibrosis/enzymology , Thrombosis/enzymology , Animals , Carotid Artery Injuries/blood , Carotid Artery Injuries/genetics , Disease Models, Animal , Janus Kinase 2/genetics , Mice, Transgenic , Mutation , Platelet Aggregation , Primary Myelofibrosis/blood , Primary Myelofibrosis/genetics , Thrombopoiesis , Thrombosis/blood , Thrombosis/genetics
11.
J Immunol ; 202(2): 527-538, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30530591

ABSTRACT

G2A is a GPCR abundantly expressed in immune cells. G2A-/- mice showed higher lethality, higher plasma cytokines, and an impaired bacterial clearance in response to a murine model of sepsis (cecal ligation and puncture), which were blocked by GdCl3, an inhibitor of Kupffer cells. Anti-IL-10 Ab reversed the impaired bacterial clearance in G2A-/- mice. Indomethacin effectively blocked both the increased i.p. IL-10 levels and the impaired bacterial clearance, indicating that disturbed PG system is the proximal cause of these phenomena. Stimulation with LPS/C5a induced an increase in Escherichia coli phagocytosis and intracellular cAMP levels in G2A+/+ peritoneal macrophages but not G2A-/- cells, which showed more PGE2/nitrite release and intracellular reactive oxygen species levels. Heterologous coexpression of G2A and adenosine receptor type 2b (A2bAR) induced a synergistic increase in cAMP signaling in a ligand-independent manner, with the evidence of physical interaction of G2A with A2bAR. BAY 60-6583, a specific agonist for A2bAR, increased intracellular cAMP levels in Kupffer cells from G2A+/+ but not from G2A-/- mice. Both G2A and A2bAR were required for antiseptic action of lysophosphatidylcholine. These results show inappropriate activation of G2A-/- Kupffer cells to septic insults due to an impaired cAMP signaling possibly by lack of interaction with A2bAR.


Subject(s)
Cell Cycle Proteins/metabolism , Escherichia coli Infections/immunology , Escherichia coli/physiology , Kupffer Cells/immunology , Macrophages, Peritoneal/physiology , Receptor, Adenosine A2B/metabolism , Receptors, G-Protein-Coupled/metabolism , Sepsis/metabolism , Animals , Antibodies, Blocking , Cell Cycle Proteins/genetics , Cells, Cultured , Cyclic AMP/metabolism , Disease Models, Animal , Humans , Interleukin-10/immunology , Interleukin-10/metabolism , Macrophages, Peritoneal/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Protein Binding , Reactive Oxygen Species/metabolism , Receptor Cross-Talk , Receptor, Adenosine A2B/genetics , Receptors, G-Protein-Coupled/genetics , Sepsis/genetics , Signal Transduction
12.
Kidney Int ; 97(3): 538-550, 2020 03.
Article in English | MEDLINE | ID: mdl-31932072

ABSTRACT

Emerging evidence in animal models of chronic kidney disease (CKD) implicates Aryl Hydrocarbon Receptor (AHR) signaling as a mediator of uremic toxicity. However, details about its tissue-specific and time-dependent activation in response to various renal pathologies remain poorly defined. Here, a comprehensive analysis of AHR induction was conducted in response to discrete models of kidney diseases using a transgenic mouse line expressing the AHR responsive-promoter tethered to a ß-galactosidase reporter gene. Following validation using a canonical AHR ligand (a dioxin derivative), the transgenic mice were subjected to adenine-induced and ischemia/reperfusion-induced injury models representing CKD and acute kidney injury (AKI), respectively, in humans. Indoxyl sulfate was artificially increased in mice through the drinking water and by inhibiting its excretion into the urine. Adenine-fed mice showed a distinct and significant increase in ß-galactosidase in the proximal and distal renal tubules, cardiac myocytes, hepatocytes, and microvasculature in the cerebral cortex. The pattern of ß-galactosidase increase coincided with the changes in serum indoxyl sulfate levels. Machine-learning-based image quantification revealed positive correlations between indoxyl sulfate levels and ß-galactosidase expression in various tissues. This pattern of ß-galactosidase expression was recapitulated in the indoxyl sulfate-specific model. The ischemia/reperfusion injury model showed increase in ß-galactosidase in renal tubules that persisted despite reduction in serum indoxyl sulfate and blood urea nitrogen levels. Thus, our results demonstrate a relationship between AHR activation in various tissues of mice with CKD or AKI and the levels of indoxyl sulfate. This study demonstrates the use of a reporter gene mouse to probe tissue-specific manifestations of uremia in translationally relevant animal models and provide hypothesis-generating insights into the mechanism of uremic toxicity that warrant further investigation.


Subject(s)
Renal Insufficiency, Chronic , Uremia , Animals , Indican , Mice , Mice, Transgenic , Receptors, Aryl Hydrocarbon/genetics , Renal Insufficiency, Chronic/genetics
13.
Int J Mol Sci ; 21(20)2020 Oct 10.
Article in English | MEDLINE | ID: mdl-33050467

ABSTRACT

Adenosine is an extracellular signaling molecule that is particularly relevant in times of cellular stress, inflammation and metabolic disturbances when the levels of the purine increase. Adenosine acts on two G-protein-coupled stimulatory and on two G-protein-coupled inhibitory receptors, which have varying expression profiles in different tissues and conditions, and have different affinities for the endogenous ligand. Studies point to significant roles of adenosine and its receptors in metabolic disease and bone health, implicating the receptors as potential therapeutic targets. This review will highlight our current understanding of the dichotomous effects of adenosine and its receptors on adipogenesis versus osteogenesis within the bone marrow to maintain bone health, as well as its relationship to obesity. Therapeutic implications will also be reviewed.


Subject(s)
Adipogenesis/genetics , Adipose Tissue/metabolism , Bone Marrow/metabolism , Osteogenesis/genetics , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Signal Transduction
14.
J Am Soc Nephrol ; 29(3): 1063-1072, 2018 03.
Article in English | MEDLINE | ID: mdl-29343519

ABSTRACT

Individuals with CKD are particularly predisposed to thrombosis after vascular injury. Using mouse models, we recently described indoxyl sulfate, a tryptophan metabolite retained in CKD and an activator of tissue factor (TF) through aryl hydrocarbon receptor (AHR) signaling, as an inducer of thrombosis across the CKD spectrum. However, the translation of findings from animal models to humans is often challenging. Here, we investigated the uremic solute-AHR-TF thrombosis axis in two human cohorts, using a targeted metabolomics approach to probe a set of tryptophan products and high-throughput assays to measure AHR and TF activity. Analysis of baseline serum samples was performed from 473 participants with advanced CKD from the Dialysis Access Consortium Clopidogrel Prevention of Early AV Fistula Thrombosis trial. Participants with subsequent arteriovenous thrombosis had significantly higher levels of indoxyl sulfate and kynurenine, another uremic solute, and greater activity of AHR and TF, than those without thrombosis. Pattern recognition analysis using the components of the thrombosis axis facilitated clustering of the thrombotic and nonthrombotic groups. We further validated these findings using 377 baseline samples from participants in the Thrombolysis in Myocardial Infarction II trial, many of whom had CKD stage 2-3. Mechanistic probing revealed that kynurenine enhances thrombosis after vascular injury in an animal model and regulates thrombosis in an AHR-dependent manner. This human validation of the solute-AHR-TF axis supports further studies probing its utility in risk stratification of patients with CKD and exploring its role in other diseases with heightened risk of thrombosis.


Subject(s)
Indican/blood , Kynurenine/blood , Receptors, Aryl Hydrocarbon/blood , Renal Insufficiency, Chronic/blood , Thromboplastin/metabolism , Thrombosis/blood , Vascular System Injuries/blood , Vascular System Injuries/complications , Adult , Aged , Clinical Trials as Topic , Female , Humans , Male , Metabolomics , Middle Aged , Pattern Recognition, Automated , Renal Insufficiency, Chronic/complications , Signal Transduction , Thrombosis/etiology , Uremia/blood , Uremia/complications
15.
Blood ; 127(11): 1493-501, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26755713

ABSTRACT

Lysyl oxidase (LOX) is overexpressed in various pathologies associated with thrombosis, such as arterial stenosis and myeloproliferative neoplasms (MPNs). LOX is elevated in the megakaryocytic lineage of mouse models of MPNs and in patients with MPNs. To gain insight into the role of LOX in thrombosis and platelet function without compounding the influences of other pathologies, transgenic mice expressing LOX in wild-type megakaryocytes and platelets (Pf4-Lox(tg/tg)) were generated. Pf4-Lox(tg/tg) mice had a normal number of platelets; however, time to vessel occlusion after endothelial injury was significantly shorter in Pf4-Lox(tg/tg) mice, indicating a higher propensity for thrombus formation in vivo. Exploring underlying mechanisms, we found that Pf4-Lox(tg/tg) platelets adhere better to collagen and have greater aggregation response to lower doses of collagen compared with controls. Platelet activation in response to the ligand for collagen receptor glycoprotein VI (cross-linked collagen-related peptide) was unaffected. However, the higher affinity of Pf4-Lox(tg/tg) platelets to the collagen sequence GFOGER implies that the collagen receptor integrin α2ß1 is affected by LOX. Taken together, our findings demonstrate that LOX enhances platelet activation and thrombosis.


Subject(s)
Blood Platelets/drug effects , Collagen/pharmacology , Platelet Activation/physiology , Protein-Lysine 6-Oxidase/physiology , Thrombophilia/enzymology , Animals , Blood Platelets/cytology , Carotid Artery Injuries/complications , Carotid Artery Thrombosis/etiology , Integrin alpha2beta1/physiology , Megakaryocytes/enzymology , Mice , Mice, Transgenic , Peptide Fragments/pharmacology , Platelet Adhesiveness/genetics , Platelet Adhesiveness/physiology , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Platelet Factor 4/genetics , Promoter Regions, Genetic , Protein-Lysine 6-Oxidase/genetics , Rats , Thrombophilia/genetics
16.
Am J Hematol ; 93(3): 430-441, 2018 03.
Article in English | MEDLINE | ID: mdl-29247535

ABSTRACT

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.


Subject(s)
Blood Platelets/cytology , Bone Marrow/physiology , Extracellular Matrix/physiology , Hematopoiesis/physiology , Megakaryocytes/cytology , Animals , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/physiology , Glycosaminoglycans/physiology , Hematopoietic Stem Cells/cytology , Humans , Integrins/physiology , Ion Channels/physiology , Mechanotransduction, Cellular , Mesenchymal Stem Cells/cytology , Mice , Neoplasm Proteins/physiology , Neoplasms/pathology , Primary Myelofibrosis/pathology , Protein-Lysine 6-Oxidase/physiology , Thrombopoiesis/physiology
17.
Am J Kidney Dis ; 70(5): 715-721, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28720207

ABSTRACT

Thrombotic microangiopathy (TMA) is characterized by the presence of microangiopathic hemolytic anemia and thrombocytopenia along with organ dysfunction, and pathologically, by the presence of microthrombi in multiple microvascular beds. Delays in diagnosis and initiation of therapy are common due to the low incidence, variable presentation, and poor awareness of these diseases, underscoring the need for interdisciplinary approaches to clinical care for TMA. We describe a new approach to improve clinical management via a TMA team that originally stemmed from an Affinity Research Collaborative team focused on thrombosis and hemostasis. The TMA team consists of clinical faculty from different disciplines who together are charged with the responsibility to quickly analyze clinical presentations, guide laboratory testing, and streamline prompt institution of treatment. The TMA team also includes faculty members from a broad range of disciplines collaborating to elucidate the pathogenesis of TMA. To this end, a clinical database and biorepository have been constructed. TMA leaders educate front-line providers from other departments through presentations in various forums across multiple specialties. Facilitated by an Affinity Research Collaborative mechanism, we describe an interdisciplinary team dedicated to improving both clinical care and translational research in TMA.


Subject(s)
Patient Care Team/organization & administration , Plasma Exchange , Renal Dialysis , Thrombotic Microangiopathies/therapy , Databases, Factual , Hematology , Humans , Nephrologists , Pharmacists , Thrombotic Microangiopathies/diagnosis , Translational Research, Biomedical
18.
Haematologica ; 102(7): 1150-1160, 2017 07.
Article in English | MEDLINE | ID: mdl-28411253

ABSTRACT

Megakaryocytes (MK) in the bone marrow (BM) are immersed in a network of extracellular matrix components that regulates platelet release into the circulation. Combining biological and bioengineering approaches, we found that the activation of transient receptor potential cation channel subfamily V member 4 (TRPV4), a mechano-sensitive ion channel, is induced upon MK adhesion on softer matrices. This response promoted platelet production by triggering a cascade of events that lead to calcium influx, ß1 integrin activation and internalization, and Akt phosphorylation, responses not found on stiffer matrices. Lysyl oxidase (LOX) is a physiological modulator of BM matrix stiffness via collagen crosslinking. In vivo inhibition of LOX and consequent matrix softening lead to TRPV4 activation cascade and increased platelet levels. At the same time, in vitro proplatelet formation was reduced on a recombinant enzyme-mediated stiffer collagen. These results suggest a novel mechanism by which MKs, through TRPV4, sense extracellular matrix environmental rigidity and release platelets accordingly.


Subject(s)
Blood Platelets/cytology , Blood Platelets/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Thrombopoiesis , Animals , Calcium/metabolism , Cell Adhesion , Cell Differentiation , Collagen Type I/metabolism , Collagen Type IV/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Integrin beta1/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport , Protein-Lysine 6-Oxidase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TRPV Cation Channels/metabolism
19.
J Immunol ; 195(8): 3828-37, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26355158

ABSTRACT

The priming of macrophages with IFN-γ prior to TLR stimulation results in enhanced and prolonged inflammatory cytokine production. In this study, we demonstrate that, following TLR stimulation, macrophages upregulate the adenosine 2b receptor (A2bR) to enhance their sensitivity to immunosuppressive extracellular adenosine. This upregulation of A2bR leads to the induction of macrophages with an immunoregulatory phenotype and the downregulation of inflammation. IFN-γ priming of macrophages selectively prevents the induction of the A2bR in macrophages to mitigate sensitivity to adenosine and to prevent this regulatory transition. IFN-γ-mediated A2bR blockade leads to a prolonged production of TNF-α and IL-12 in response to TLR ligation. The pharmacologic inhibition or the genetic deletion of the A2bR results in a hyperinflammatory response to TLR ligation, similar to IFN-γ treatment of macrophages. Conversely, the overexpression of A2bR on macrophages blunts the IFN-γ effects and promotes the development of immunoregulatory macrophages. Thus, we propose a novel mechanism whereby IFN-γ contributes to host defense by desensitizing macrophages to the immunoregulatory effects of adenosine. This mechanism overcomes the transient nature of TLR activation, and prolongs the antimicrobial state of the classically activated macrophage. This study may offer promising new targets to improve the clinical outcome of inflammatory diseases in which macrophage activation is dysregulated.


Subject(s)
Interferon-gamma/immunology , Macrophage Activation/immunology , Macrophages/immunology , Receptor, Adenosine A2B/immunology , Up-Regulation/immunology , Animals , Female , Interferon-gamma/genetics , Interleukin-12/genetics , Interleukin-12/immunology , Macrophage Activation/genetics , Mice , Mice, Knockout , Receptor, Adenosine A2B/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/genetics
20.
J Immunol ; 195(4): 1732-43, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26136425

ABSTRACT

The adenosine A2b receptor (Adora2b) has been implicated in cardioprotection from myocardial ischemia. As such, Adora2b was found to be critical in ischemic preconditioning (IP) or ischemia/reperfusion (IR) injury of the heart. Whereas Adora2b is present on various cells types, the tissue-specific role of Adora2b in cardioprotection is still unknown. To study the tissue-specific role of Adora2b signaling on inflammatory cells, endothelia, or myocytes during myocardial ischemia in vivo, we intercrossed floxed Adora2b mice with Lyz2-Cre(+), VE-cadherin-Cre(+), or myosin-Cre(+) transgenic mice, respectively. Mice were exposed to 60 min of myocardial ischemia with or without IP (four times for 5 min) followed by 120 min of reperfusion. Cardioprotection by IP was abolished in Adora2b(f/f)-VE-cadherin-Cre(+) or Adora2b(f/f)-myosin-Cre(+), indicating that Adora2b signaling on endothelia or myocytes mediates IP. In contrast, primarily Adora2b signaling on inflammatory cells was necessary to provide cardioprotection in IR injury, indicated by significantly larger infarcts and higher troponin levels in Adora2b(f/f)-Lyz2-Cre(+) mice only. Cytokine profiling of IR injury in Adora2b(f/f)-Lyz2-Cre(+) mice pointed toward polymorphonuclear neutrophils (PMNs). Analysis of PMNs from Adora2b(f/f)-Lyz2-Cre(+) confirmed PMNs as one source of identified tissue cytokines. Finally, adoptive transfer of Adora2b(-/-) PMNs revealed a critical role of Adora2b on PMNs in cardioprotection from IR injury. Adora2b signaling mediates different types of cardioprotection in a tissue-specific manner. These findings have implications for the use of Adora2b agonists in the treatment or prevention of myocardial injury by ischemia.


Subject(s)
Gene Expression Regulation , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/metabolism , Adoptive Transfer , Animals , Bone Marrow Cells/metabolism , Disease Models, Animal , Endothelium/metabolism , Gene Deletion , Ischemic Preconditioning, Myocardial , Male , Mice , Mice, Knockout , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Neutrophils/immunology , Organ Specificity/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL