Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Mol Pharm ; 20(12): 6492-6503, 2023 Dec 04.
Article in English | MEDLINE | ID: mdl-37975733

ABSTRACT

mRNA LNPs can experience a decline in activity over short periods (ranging from weeks to months). As a result, they require frozen storage and transportation conditions to maintain their full functionality when utilized. Currently approved commercially available mRNA LNP vaccines also necessitate frozen storage and supply chain management. Overcoming this significant inconvenience in the future is crucial to reducing unnecessary costs and challenges associated with storage and transport. In this study, our objective was to illuminate the potential time frame for nonfrozen storage and transportation conditions of mRNA LNPs without compromising their activity. To achieve this goal, we conducted a stability assessment and an in vitro cell culture delivery study involving five mRNA LNPs. These LNPs were constructed by using a standard formulation similar to that employed in the three commercially available LNP formulations. Among these formulations, we selected five structurally diverse ionizable lipids─C12-200, CKK-E12, MC3, SM-102, and lipid 23─from the existing literature. We incorporated these lipids into a standard LNP formulation, keeping all other components identical. The LNPs, carrying mRNA payloads, were synthesized by using microfluidic mixing technology. We evaluated the shelf life stability of these LNPs over a span of 9 weeks at temperatures of 2-8, 25, and 40 °C, utilizing an array of analytical techniques. Our findings indicated minimal impact on the hydrodynamic diameter, zeta potential, encapsulation efficiency, and polydispersity of all LNPs across the various temperatures over the studied period. The RiboGreen assay analysis of LNPs showed consistent mRNA contents over several weeks at various nonfrozen storage temperatures, leading to the incorrect assumption of intact and functional LNPs. This misunderstanding was rectified by the significant differences observed in EGFP protein expression in an in vitro cell culture (using HEK293 cells) across the five LNPs. Specifically, only LNP 1 (C12-200) and LNP 4 (SM-102) exhibited high levels of EGFP expression at the start (T0), with over 90% of HEK293 cells transfected and mean fluorescence intensity (MFI) levels exceeding 1. Interestingly, LNP 1 (C12-200) maintained largely unchanged levels of in vitro activity over 11 weeks when stored at both 2-8 and 25 °C. In contrast, LNP 4 (SM-102) retained its functionality when stored at 2-8 °C over 11 weeks but experienced a gradual decline of in vitro activity when stored at room temperature over the same period. Importantly, we observed distinct LNP architectures for the five formulations through cryo-EM imaging. This highlights the necessity for a deeper comprehension of structure-activity relationships within these complex nanoparticle structures. Enhancing our understanding in this regard is vital for overcoming storage and stability limitations, ultimately facilitating the broader application of this technology beyond vaccines.


Subject(s)
Nanoparticles , Vaccines , Humans , HEK293 Cells , Lipids/chemistry , Nanoparticles/chemistry , RNA, Messenger/genetics , RNA, Small Interfering/chemistry
2.
Nat Cancer ; 4(8): 1193-1209, 2023 08.
Article in English | MEDLINE | ID: mdl-37550517

ABSTRACT

Aging facilitates the expansion of hematopoietic stem cells (HSCs) carrying clonal hematopoiesis-related somatic mutations and the development of myeloid malignancies, such as myeloproliferative neoplasms (MPNs). While cooperating mutations can cause transformation, it is unclear whether distinct bone marrow (BM) HSC-niches can influence the growth and therapy response of HSCs carrying the same oncogenic driver. Here we found different BM niches for HSCs in MPN subtypes. JAK-STAT signaling differentially regulates CDC42-dependent HSC polarity, niche interaction and mutant cell expansion. Asymmetric HSC distribution causes differential BM niche remodeling: sinusoidal dilation in polycythemia vera and endosteal niche expansion in essential thrombocythemia. MPN development accelerates in a prematurely aged BM microenvironment, suggesting that the specialized niche can modulate mutant cell expansion. Finally, dissimilar HSC-niche interactions underpin variable clinical response to JAK inhibitor. Therefore, HSC-niche interactions influence the expansion rate and therapy response of cells carrying the same clonal hematopoiesis oncogenic driver.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Humans , Aged , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/therapy , Myeloproliferative Disorders/pathology , Bone Marrow/pathology , Bone Marrow/physiology , Hematopoietic Stem Cells/pathology , Bone and Bones/pathology , Tumor Microenvironment/genetics
3.
Chem Sci ; 13(19): 5539-5545, 2022 May 18.
Article in English | MEDLINE | ID: mdl-35694350

ABSTRACT

Despite its essential role in the (patho)physiology of several diseases, CB2R tissue expression profiles and signaling mechanisms are not yet fully understood. We report the development of a highly potent, fluorescent CB2R agonist probe employing structure-based reverse design. It commences with a highly potent, preclinically validated ligand, which is conjugated to a silicon-rhodamine fluorophore, enabling cell permeability. The probe is the first to preserve interspecies affinity and selectivity for both mouse and human CB2R. Extensive cross-validation (FACS, TR-FRET and confocal microscopy) set the stage for CB2R detection in endogenously expressing living cells along with zebrafish larvae. Together, these findings will benefit clinical translatability of CB2R based drugs.

4.
Front Pharmacol ; 13: 841766, 2022.
Article in English | MEDLINE | ID: mdl-35645832

ABSTRACT

The distribution and roles of the cannabinoid CB2 receptor in the CNS are still a matter of debate. Recent data suggest that, in addition to its presence in microglial cells, the CB2 receptor may be also expressed at low levels, yet biologically relevant, in other cell types such as neurons. It is accepted that the expression of CB2 receptors in the CNS is low under physiological conditions and is significantly elevated in chronic neuroinflammatory states associated with neurodegenerative diseases such as Alzheimer's disease. By using a novel mouse model (CB2 EGFP/f/f), we studied the distribution of cannabinoid CB2 receptors in the 5xFAD mouse model of Alzheimer's disease (by generating 5xFAD/CB2 EGFP/f/f mice) and explored the roles of CB2 receptors in microglial function. We used a novel selective and brain penetrant CB2 receptor agonist (RO6866945) as well as mice lacking the CB2 receptor (5xFAD/CB2 -/-) for these studies. We found that CB2 receptors are expressed in dystrophic neurite-associated microglia and that their modulation modifies the number and activity of microglial cells as well as the metabolism of the insoluble form of the amyloid peptide. These results support microglial CB2 receptors as potential targets for the development of amyloid-modulating therapies.

5.
Cell Stem Cell ; 29(4): 528-544.e9, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35276096

ABSTRACT

The autonomic nervous system is a master regulator of homeostatic processes and stress responses. Sympathetic noradrenergic nerve fibers decrease bone mass, but the role of cholinergic signaling in bone has remained largely unknown. Here, we describe that early postnatally, a subset of sympathetic nerve fibers undergoes an interleukin-6 (IL-6)-induced cholinergic switch upon contacting the bone. A neurotrophic dependency mediated through GDNF-family receptor-α2 (GFRα2) and its ligand, neurturin (NRTN), is established between sympathetic cholinergic fibers and bone-embedded osteocytes, which require cholinergic innervation for their survival and connectivity. Bone-lining osteoprogenitors amplify and propagate cholinergic signals in the bone marrow (BM). Moderate exercise augments trabecular bone partly through an IL-6-dependent expansion of sympathetic cholinergic nerve fibers. Consequently, loss of cholinergic skeletal innervation reduces osteocyte survival and function, causing osteopenia and impaired skeletal adaptation to moderate exercise. These results uncover a cholinergic neuro-osteocyte interface that regulates skeletogenesis and skeletal turnover through bone-anabolic effects.


Subject(s)
Interleukin-6 , Osteogenesis , Cholinergic Agents , Cholinergic Fibers , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology
6.
Nat Commun ; 13(1): 543, 2022 01 27.
Article in English | MEDLINE | ID: mdl-35087060

ABSTRACT

The sympathetic nervous system has been evolutionary selected to respond to stress and activates haematopoietic stem cells via noradrenergic signals. However, the pathways preserving haematopoietic stem cell quiescence and maintenance under proliferative stress remain largely unknown. Here we found that cholinergic signals preserve haematopoietic stem cell quiescence in bone-associated (endosteal) bone marrow niches. Bone marrow cholinergic neural signals increase during stress haematopoiesis and are amplified through cholinergic osteoprogenitors. Lack of cholinergic innervation impairs balanced responses to chemotherapy or irradiation and reduces haematopoietic stem cell quiescence and self-renewal. Cholinergic signals activate α7 nicotinic receptor in bone marrow mesenchymal stromal cells leading to increased CXCL12 expression and haematopoietic stem cell quiescence. Consequently, nicotine exposure increases endosteal haematopoietic stem cell quiescence in vivo and impairs hematopoietic regeneration after haematopoietic stem cell transplantation in mice. In humans, smoking history is associated with delayed normalisation of platelet counts after allogeneic haematopoietic stem cell transplantation. These results suggest that cholinergic signals preserve stem cell quiescence under proliferative stress.


Subject(s)
Cholinergic Agents/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Animals , Bone Marrow/metabolism , Chemokine CXCL12/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Hematopoietic Stem Cell Transplantation , Humans , Mesenchymal Stem Cells/metabolism , Mice , Receptors, Adrenergic, beta-3/metabolism , Risk Factors
7.
Eye (Lond) ; 35(5): 1305-1316, 2021 May.
Article in English | MEDLINE | ID: mdl-33564135

ABSTRACT

The angopoietin/tyrosine kinase with immunoglobulin and epidermal growth factor homology domains (Ang/Tie) pathway is an emerging key regulator in vascular development and maintenance. Its relevance to clinicians and basic scientists as a potential therapeutic target in retinal and choroidal vascular diseases is highlighted by recent preclinical and clinical evidence. The Ang/Tie pathway plays an important role in the regulation of vascular stability, in angiogenesis under physiological and pathological conditions, as well as in inflammation. Under physiological conditions, angiopoietin-1 (Ang-1) binds to and phosphorylates the Tie2 receptor, leading to downstream signalling that promotes cell survival and vascular stability. Angiopoietin-2 (Ang-2) is upregulated under pathological conditions and acts as a context-dependent agonist/antagonist of the Ang-1/Tie2 axis, causing vascular destabilisation and sensitising blood vessels to the effects of vascular endothelial growth factor-A (VEGF-A). Ang-2 and VEGF-A synergistically drive vascular leakage, neovascularisation and inflammation, key components of retinal vascular diseases. Preclinical evidence suggests that modulating the Ang/Tie pathway restores vascular stabilisation and reduces inflammation. This review discusses how targeting the Ang/Tie pathway or applying Ang-2/VEGF-A combination therapy may be a valuable therapeutic strategy for restoring vascular stability and reducing inflammation in the treatment of retinal and choroidal vascular diseases.


Subject(s)
Angiopoietins , Vascular Diseases , Humans , Receptor, TIE-2/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A
8.
Nat Commun ; 12(1): 608, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504783

ABSTRACT

Haematopoietic stem cells (HSCs) are characterized by their self-renewal potential associated to dormancy. Here we identify the cell surface receptor neogenin-1 as specifically expressed in dormant HSCs. Loss of neogenin-1 initially leads to increased HSC expansion but subsequently to loss of self-renewal and premature exhaustion in vivo. Its ligand netrin-1 induces Egr1 expression and maintains quiescence and function of cultured HSCs in a Neo1 dependent manner. Produced by arteriolar endothelial and periarteriolar stromal cells, conditional netrin-1 deletion in the bone marrow niche reduces HSC numbers, quiescence and self-renewal, while overexpression increases quiescence in vivo. Ageing associated bone marrow remodelling leads to the decline of netrin-1 expression in niches and a compensatory but reversible upregulation of neogenin-1 on HSCs. Our study suggests that niche produced netrin-1 preserves HSC quiescence and self-renewal via neogenin-1 function. Decline of netrin-1 production during ageing leads to the gradual decrease of Neo1 mediated HSC self-renewal.


Subject(s)
Hematopoietic Stem Cells/metabolism , Membrane Proteins/metabolism , Netrin-1/metabolism , Stem Cell Niche , Animals , Arterioles/metabolism , Cell Differentiation , Cell Proliferation , Cellular Senescence , Gene Deletion , Hematopoietic Stem Cell Transplantation , Mice, Mutant Strains , Mice, Transgenic , Signal Transduction
9.
J Am Chem Soc ; 142(40): 16953-16964, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32902974

ABSTRACT

Pharmacological modulation of cannabinoid type 2 receptor (CB2R) holds promise for the treatment of numerous conditions, including inflammatory diseases, autoimmune disorders, pain, and cancer. Despite the significance of this receptor, researchers lack reliable tools to address questions concerning the expression and complex mechanism of CB2R signaling, especially in cell-type and tissue-dependent contexts. Herein, we report for the first time a versatile ligand platform for the modular design of a collection of highly specific CB2R fluorescent probes, used successfully across applications, species, and cell types. These include flow cytometry of endogenously expressing cells, real-time confocal microscopy of mouse splenocytes and human macrophages, as well as FRET-based kinetic and equilibrium binding assays. High CB2R specificity was demonstrated by competition experiments in living cells expressing CB2R at native levels. The probes were effectively applied to FACS analysis of microglial cells derived from a mouse model relevant to Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Fluorescent Dyes/chemistry , Microglia/metabolism , Receptor, Cannabinoid, CB2/analysis , Animals , CHO Cells , Cricetulus , Disease Models, Animal , Flow Cytometry , Fluorescence Resonance Energy Transfer , Humans , Ligands , Mice , Molecular Docking Simulation , Molecular Probes/chemistry , Optical Imaging , Sensitivity and Specificity , Signal Transduction
10.
Cell Stem Cell ; 25(3): 407-418.e6, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31303548

ABSTRACT

Hematopoietic stem cells (HSCs) residing in the bone marrow (BM) accumulate during aging but are functionally impaired. However, the role of HSC-intrinsic and -extrinsic aging mechanisms remains debated. Megakaryocytes promote quiescence of neighboring HSCs. Nonetheless, whether megakaryocyte-HSC interactions change during pathological/natural aging is unclear. Premature aging in Hutchinson-Gilford progeria syndrome recapitulates physiological aging features, but whether these arise from altered stem or niche cells is unknown. Here, we show that the BM microenvironment promotes myelopoiesis in premature/physiological aging. During physiological aging, HSC-supporting niches decrease near bone but expand further from bone. Increased BM noradrenergic innervation promotes ß2-adrenergic-receptor(AR)-interleukin-6-dependent megakaryopoiesis. Reduced ß3-AR-Nos1 activity correlates with decreased endosteal niches and megakaryocyte apposition to sinusoids. However, chronic treatment of progeroid mice with ß3-AR agonist decreases premature myeloid and HSC expansion and restores the proximal association of HSCs to megakaryocytes. Therefore, normal/premature aging of BM niches promotes myeloid expansion and can be improved by targeting the microenvironment.


Subject(s)
Aging, Premature/pathology , Aging/physiology , Bone Marrow/physiology , Hematopoietic Stem Cells/physiology , Megakaryocytes/physiology , Myeloid Cells/physiology , Progeria/pathology , Adrenergic Agonists/administration & dosage , Aging/metabolism , Aging, Premature/metabolism , Animals , Cell Differentiation , Cell Encapsulation , Cell Proliferation , Disease Models, Animal , Humans , Interleukin-6/metabolism , Mice , Nitric Oxide Synthase Type I/metabolism , Progeria/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Stem Cell Niche
11.
PLoS Genet ; 15(6): e1008216, 2019 06.
Article in English | MEDLINE | ID: mdl-31246957

ABSTRACT

ASAP1 is a multi-domain adaptor protein that regulates cytoskeletal dynamics, receptor recycling and intracellular vesicle trafficking. Its expression is associated with poor prognosis for a variety of cancers, and promotes cell migration, invasion and metastasis. Little is known about its physiological role. In this study, we used mice with a gene-trap inactivated ASAP1 locus to study the functional role of ASAP1 in vivo, and found defects in tissues derived from mesenchymal progenitor cells. Loss of ASAP1 led to growth retardation and delayed ossification typified by enlarged hypertrophic zones in growth plates and disorganized chondro-osseous junctions. Furthermore, loss of ASAP1 led to delayed adipocyte development and reduced fat depot formation. Consistently, deletion of ASAP1 resulted in accelerated chondrogenic differentiation of mesenchymal cells in vitro, but suppressed osteo- and adipogenic differentiation. Mechanistically, we found that FAK/Src and PI3K/AKT signaling is compromised in Asap1GT/GT MEFs, leading to impaired adipogenic differentiation. Dysregulated FAK/Src and PI3K/AKT signaling is also associated with attenuated osteogenic differentiation. Together these observations suggest that ASAP1 plays a decisive role during the differentiation of mesenchymal progenitor cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adipogenesis/genetics , Chondrogenesis/genetics , Osteogenesis/genetics , Animals , Cell Differentiation/genetics , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Developmental/genetics , Growth Plate/growth & development , Growth Plate/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics , src-Family Kinases/genetics
12.
Blood ; 133(3): 224-236, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30361261

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) and leukocytes circulate between the bone marrow (BM) and peripheral blood following circadian oscillations. Autonomic sympathetic noradrenergic signals have been shown to regulate HSPC and leukocyte trafficking, but the role of the cholinergic branch has remained unexplored. We have investigated the role of the cholinergic nervous system in the regulation of day/night traffic of HSPCs and leukocytes in mice. We show here that the autonomic cholinergic nervous system (including parasympathetic and sympathetic) dually regulates daily migration of HSPCs and leukocytes. At night, central parasympathetic cholinergic signals dampen sympathetic noradrenergic tone and decrease BM egress of HSPCs and leukocytes. However, during the daytime, derepressed sympathetic noradrenergic activity causes predominant BM egress of HSPCs and leukocytes via ß3-adrenergic receptor. This egress is locally supported by light-triggered sympathetic cholinergic activity, which inhibits BM vascular cell adhesion and homing. In summary, central (parasympathetic) and local (sympathetic) cholinergic signals regulate day/night oscillations of circulating HSPCs and leukocytes. This study shows how both branches of the autonomic nervous system cooperate to orchestrate daily traffic of HSPCs and leukocytes.


Subject(s)
Cell Movement , Cholinergic Agents/pharmacology , Circadian Rhythm , Hematopoietic Stem Cells/physiology , Leukocytes/physiology , Parasympathetic Nervous System/physiology , Sympathetic Nervous System/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Adhesion , Cells, Cultured , Chemotaxis , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Leukocytes/cytology , Leukocytes/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Adrenergic, beta-2 , Receptors, Adrenergic, beta-3/physiology , Receptors, G-Protein-Coupled/physiology
13.
Arterioscler Thromb Vasc Biol ; 37(3): 495-505, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28126825

ABSTRACT

OBJECTIVE: Vascular smooth muscle cells (VSMC) play a key role in the pathogenesis of atherosclerosis, the globally leading cause of death. The transmembrane orphan receptor endosialin (CD248) has been characterized as an activation marker of cells of the mesenchymal lineage including tumor-associated pericytes, stromal myofibroblasts, and activated VSMC. We, therefore, hypothesized that VSMC-expressed endosialin may display functional involvement in the pathogenesis of atherosclerosis. APPROACH AND RESULTS: Expression of endosialin was upregulated during atherosclerosis in apolipoprotein E (ApoE)-null mice and human atherosclerotic samples analyzed by quantitative real-time polymerase chain reaction and immunohistochemistry. Atherosclerosis, assessed by Oil Red O staining of the descending aorta, was significantly reduced in ApoE/endosialin-deficient mice on Western-type diet. Marker analysis of VSMC in lesions induced by shear stress-modifying cast implantation around the right carotid artery identified a more pronounced contractile VSMC phenotype in the absence of endosialin. Moreover, in addition to contributing to neointima formation, endosialin also potentially regulated the proinflammatory phenotype of VSMC as evidenced in surrogate cornea pocket assay experiments in vivo and corresponding flow cytometry and ELISA analyses in vitro. CONCLUSIONS: The experiments identify endosialin as a potential regulator of phenotypic remodeling of VSMC contributing to atherosclerosis. The association of endosialin with atherosclerosis and its absent expression in nonatherosclerotic samples warrant further consideration of endosialin as a therapeutic target and biomarker.


Subject(s)
Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neoplasm Proteins/metabolism , Vascular Remodeling , Aged , Aged, 80 and over , Animals , Antigens, CD/genetics , Antigens, Neoplasm/genetics , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Atherosclerosis/prevention & control , Case-Control Studies , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/pathology , Neointima , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Phenotype , Signal Transduction , Vasoconstriction
14.
Blood ; 129(7): 811-822, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28064238

ABSTRACT

Research in the last few years has revealed a sophisticated interaction network between multiple bone marrow cells that regulate different hematopoietic stem cell (HSC) properties such as proliferation, differentiation, localization, and self-renewal during homeostasis. These mechanisms are essential to keep the physiological HSC numbers in check and interfere with malignant progression. In addition to the identification of multiple mutations and chromosomal aberrations driving the progression of myeloid malignancies, alterations in the niche compartment recently gained attention for contributing to disease progression. Leukemic cells can remodel the niche into a permissive environment favoring leukemic stem cell expansion over normal HSC maintenance, and evidence is accumulating that certain niche alterations can even induce leukemic transformation. Relapse after chemotherapy is still a major challenge during treatment of myeloid malignancies, and cure is only rarely achieved. Recent progress in understanding the niche-imposed chemoresistance mechanisms will likely contribute to the improvement of current therapeutic strategies. This article discusses the role of different niche cells and their stage- and disease-specific roles during progression of myeloid malignancies and in response to chemotherapy.


Subject(s)
Hematopoietic Stem Cells/pathology , Leukemia, Myeloid/pathology , Myeloid Cells/pathology , Neoplastic Stem Cells/pathology , Antineoplastic Agents/therapeutic use , Bone Marrow/blood supply , Bone Marrow/drug effects , Bone Marrow/pathology , Hematopoietic Stem Cells/drug effects , Humans , Leukemia, Myeloid/drug therapy , Myeloid Cells/drug effects , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/pathology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Stem Cell Niche/drug effects , Tumor Microenvironment/drug effects
15.
Dev Cell ; 36(1): 79-93, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26766444

ABSTRACT

The WNT signaling enhancer R-spondin3 (RSPO3) is prominently expressed in the vasculature. Correspondingly, embryonic lethality of Rspo3-deficient mice is caused by vessel remodeling defects. Yet the mechanisms underlying vascular RSPO3 function remain elusive. Inducible endothelial Rspo3 deletion (Rspo3-iECKO) resulted in perturbed developmental and tumor vascular remodeling. Endothelial cell apoptosis and vascular pruning led to reduced microvessel density in Rspo3-iECKO mice. Rspo3-iECKO mice strikingly phenocopied the non-canonical WNT signaling-induced vascular defects of mice deleted for the WNT secretion factor Evi/Wls. An endothelial screen for RSPO3 and EVI/WLS co-regulated genes identified Rnf213, Usp18, and Trim30α. RNF213 targets filamin A and NFAT1 for proteasomal degradation attenuating non-canonical WNT/Ca(2+) signaling. Likewise, USP18 and TRIM5α inhibited NFAT1 activation. Consequently, NFAT protein levels were decreased in endothelial cells of Rspo3-iECKO mice and pharmacological NFAT inhibition phenocopied Rspo3-iECKO mice. The data identify endothelial RSPO3-driven non-canonical WNT/Ca(2+)/NFAT signaling as a critical maintenance pathway of the remodeling vasculature.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Signal Transduction/genetics , Thrombospondins/metabolism , Transcription Factors/metabolism , Animals , Calcium/metabolism , Mice, Transgenic , NFATC Transcription Factors/metabolism , Thrombospondins/genetics , Wnt Proteins/metabolism
16.
Cell Rep ; 12(11): 1761-73, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26344773

ABSTRACT

Tie1 is a mechanistically poorly characterized endothelial cell (EC)-specific orphan receptor. Yet, Tie1 deletion is embryonic lethal and Tie1 has been implicated in critical vascular pathologies, including atherosclerosis and tumor angiogenesis. Here, we show that Tie1 does not function independently but exerts context-dependent effects on the related receptor Tie2. Tie1 was identified as an EC activation marker that is expressed during angiogenesis by a subset of angiogenic tip and remodeling stalk cells and downregulated in the adult quiescent vasculature. Functionally, Tie1 expression by angiogenic EC contributes to shaping the tip cell phenotype by negatively regulating Tie2 surface presentation. In contrast, Tie1 acts in remodeling stalk cells cooperatively to sustain Tie2 signaling. Collectively, our data support an interactive model of Tie1 and Tie2 function, in which dynamically regulated Tie1 versus Tie2 expression determines the net positive or negative effect of Tie1 on Tie2 signaling.


Subject(s)
Receptor, TIE-1/physiology , Receptor, TIE-2/physiology , Vascular Remodeling/physiology , Animals , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Endothelial Cells/enzymology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Retinal Vessels/physiology , Signal Transduction
17.
Dev Cell ; 34(1): 5-17, 2015 Jul 06.
Article in English | MEDLINE | ID: mdl-26151903

ABSTRACT

The field of angiogenesis research has primarily focused on the mechanisms of sprouting angiogenesis. Yet vascular networks formed by vessel sprouting subsequently undergo extensive vascular remodeling to form a functional and mature vasculature. This "trimming" includes distinct processes of vascular pruning, the regression of selected vascular branches. In some situations complete vascular networks may undergo physiological regression. Vessel regression is an understudied yet emerging field of research. This review summarizes the state-of-the-art of vessel pruning and regression with a focus on the cellular processes and the molecular regulators of vessel maintenance and regression.


Subject(s)
Blood Vessels/pathology , Endothelial Cells/physiology , Morphogenesis/physiology , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/physiology , Animals , Humans , Models, Biological
18.
EMBO Mol Med ; 7(3): 332-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25680861

ABSTRACT

Liver fibrosis is a reversible wound-healing response to injury reflecting the critical balance between liver repair and scar formation. Chronic damage leads to progressive substitution of liver parenchyma by scar tissue and ultimately results in liver cirrhosis. Stromal cells (hepatic stellate cells [HSC] and endothelial cells) have been proposed to control the balance between liver fibrosis and regeneration. Here, we show that endosialin, a C-type lectin, expressed in the liver exclusively by HSC and portal fibroblasts, is upregulated in liver fibrosis in mouse and man. Chronic chemically induced liver damage resulted in reduced fibrosis and enhanced hepatocyte proliferation in endosialin-deficient (EN(KO)) mice. Correspondingly, acute-liver-damage-induced hepatocyte proliferation (partial hepatectomy) was increased in EN(KO) mice. A candidate-based screen of known regulators of hepatocyte proliferation identified insulin-like growth factor 2 (IGF2) as selectively endosialin-dependent hepatocyte mitogen. Collectively, the study establishes a critical role of HSC in the reciprocal regulation of fibrogenesis vs. hepatocyte proliferation and identifies endosialin as a therapeutic target in non-neoplastic settings.


Subject(s)
Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Cell Proliferation , Hepatic Stellate Cells/metabolism , Hepatocytes/cytology , Liver Cirrhosis/pathology , Animals , Humans , Liver Cirrhosis/chemically induced , Liver Regeneration , Mice , Mice, Knockout , Neoplasm Proteins/deficiency , Neoplasm Proteins/metabolism
19.
Cancer Cell ; 26(6): 880-895, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25490450

ABSTRACT

Antiangiogenic tumor therapy has failed in the adjuvant setting. Here we show that inhibition of the Tie2 ligand angiopoietin-2 (Ang2) effectively blocks metastatic growth in preclinical mouse models of postsurgical adjuvant therapy. Ang2 antibody treatment combines well with low-dose metronomic chemotherapy (LDMC) in settings in which maximum-dose chemotherapy does not prove effective. Mechanistically, Ang2 blockade could be linked to quenching the inflammatory and angiogenic response of endothelial cells (ECs) in the metastatic niche. Reduced EC adhesion molecule and chemokine expression inhibits the recruitment of tumor-promoting CCR2(+)Tie2(-) metastasis-associated macrophages. Moreover, LDMC contributes to therapeutic efficacy by inhibiting the recruitment of protumorigenic bone marrow-derived myeloid cells. Collectively, these data provide a rationale for mechanism-guided adjuvant tumor therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/drug therapy , Neoplasm Metastasis/drug therapy , Neovascularization, Pathologic/drug therapy , Adjuvants, Pharmaceutic/administration & dosage , Adjuvants, Pharmaceutic/adverse effects , Administration, Metronomic , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Animals , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Cell Line, Tumor , Endothelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mammary Neoplasms, Experimental , Maximum Tolerated Dose , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Xenograft Model Antitumor Assays
20.
Development ; 141(8): 1757-66, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24715464

ABSTRACT

Multiple cell types involved in the regulation of angiogenesis express Wnt ligands. Although ß-catenin dependent and independent Wnt signaling pathways have been shown to control angiogenesis, the contribution of individual cell types to activate these downstream pathways in endothelial cells (ECs) during blood vessel formation is still elusive. To investigate the role of ECs in contributing Wnt ligands for regulation of blood vessel formation, we conditionally deleted the Wnt secretion factor Evi in mouse ECs (Evi-ECKO). Evi-ECKO mice showed decreased microvessel density during physiological and pathological angiogenesis in the postnatal retina and in tumors, respectively. The reduced microvessel density resulted from increased vessel regression accompanied by decreased EC survival and proliferation. Concomitantly, survival-related genes were downregulated and cell cycle arrest- and apoptosis-inducing genes were upregulated. EVI silencing in cultured HUVECs showed similar target gene regulation, supporting a mechanism of EC-derived Wnt ligands in controlling EC function. ECs preferentially expressed non-canonical Wnt ligands and canonical target gene expression was unaffected in Evi-ECKO mice. Furthermore, the reduced vascularization of Matrigel plugs in Evi-ECKO mice could be rescued by introduction of non-canonical Wnt5a. Treatment of mouse pups with the non-canonical Wnt inhibitor TNP470 resulted in increased vessel regression accompanied by decreased EC proliferation, thus mimicking the proliferation-dependent Evi-ECKO remodeling phenotype. Taken together, this study identified EC-derived non-canonical Wnt ligands as regulators of EC survival, proliferation and subsequent vascular pruning during developmental and pathological angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Wnt Proteins/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/genetics , Autocrine Communication/drug effects , Autocrine Communication/genetics , Cell Count , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclohexanes/pharmacology , DNA-Binding Proteins/metabolism , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ligands , MDS1 and EVI1 Complex Locus Protein , Mice , Mice, Transgenic , Models, Biological , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , O-(Chloroacetylcarbamoyl)fumagillol , Phenotype , Proto-Oncogenes , Retina/growth & development , Retina/metabolism , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...