Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Nat Immunol ; 23(8): 1256-1272, 2022 08.
Article in English | MEDLINE | ID: mdl-35902638

ABSTRACT

The recombination-activating genes (RAG) 1 and 2 are indispensable for diversifying the primary B cell receptor repertoire and pruning self-reactive clones via receptor editing in the bone marrow; however, the impact of RAG1/RAG2 on peripheral tolerance is unknown. Partial RAG deficiency (pRD) manifesting with late-onset immune dysregulation represents an 'experiment of nature' to explore this conundrum. By studying B cell development and subset-specific repertoires in pRD, we demonstrate that reduced RAG activity impinges on peripheral tolerance through the generation of a restricted primary B cell repertoire, persistent antigenic stimulation and an inflammatory milieu with elevated B cell-activating factor. This unique environment gradually provokes profound B cell dysregulation with widespread activation, remarkable extrafollicular maturation and persistence, expansion and somatic diversification of self-reactive clones. Through the model of pRD, we reveal a RAG-dependent 'domino effect' that impacts stringency of tolerance and B cell fate in the periphery.


Subject(s)
B-Lymphocytes , DNA-Binding Proteins , Homeodomain Proteins , Nuclear Proteins , Cell Differentiation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Humans , Immune Tolerance , Lymphocyte Count , Nuclear Proteins/deficiency
2.
Am J Pathol ; 191(2): 396-414, 2021 02.
Article in English | MEDLINE | ID: mdl-33159887

ABSTRACT

Recruitment of naive T cells to lymph nodes is essential for the development of adaptive immunity. Upon pathogen infection, lymph nodes promptly increase the influx of naive T cells from the circulation in order to screen and prime the T cells. The precise contribution of the lymph node vasculature to the regulation of this process remains unclear. Here we show a role for the Ras GTPase, R-Ras, in the functional adaptation of high endothelial venules to increase naive T cell trafficking to the lymph nodes. R-Ras is transiently up-regulated in the endothelium of high endothelial venules by the inflammatory cytokine tumor necrosis factor (TNF) within 24 hours of pathogen inoculation. TNF induces R-Ras upregulation in endothelial cells via JNK and p38 mitogen-activated protein kinase but not NF-κB. Studies of T cell trafficking found that the loss of function of endothelial R-Ras impairs the rapid acceleration of naive T cell recruitment to the lymph nodes upon inflammation. This defect diminished the ability of naive OT-1 T cells to develop antitumor activity against ovalbumin-expressing melanoma. Proteomic analyses suggest that endothelial R-Ras facilitates TNF-dependent transendothelial migration (diapedesis) of naive T cells by modulating molecular assembly the at T cell-endothelial cell interface. These findings give new mechanistic insights into the functional adaptation of high endothelial venules to accelerate naive T cell recruitment to the lymph nodes.


Subject(s)
Chemotaxis, Leukocyte/physiology , T-Lymphocytes/immunology , Transendothelial and Transepithelial Migration/physiology , Tumor Necrosis Factor-alpha/metabolism , ras Proteins/metabolism , Animals , Endothelial Cells/metabolism , Humans , Lymph Nodes/blood supply , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , T-Lymphocytes/metabolism , Up-Regulation , Venules/immunology , Venules/metabolism
3.
J Vasc Res ; 58(4): 252-266, 2021.
Article in English | MEDLINE | ID: mdl-33873190

ABSTRACT

PURPOSE: The retinal vasculature is heavily invested by pericytes. Small GTPase R-Ras is highly expressed in endothelial cells and pericytes, suggesting importance of this Ras homolog for the regulation of the blood vessel wall. We investigated the specific contribution of pericyte-expressed R-Ras to the development of the retinal vasculature. METHODS: The effect of R-Ras deficiency in pericytes was analyzed in pericyte-targeted conditional Rras knockout mice at birth and during the capillary plexus formation in the neonatal retina. RESULTS: The offspring of these mice frequently exhibited unilateral microphthalmia. Analyses of the developing retinal vasculature in the eyes without microphthalmia revealed excessive endothelial cell proliferation, sprouting, and branching of the capillary plexus in these animals. These vessels were structurally defective with diminished pericyte coverage and basement membrane formation. Furthermore, these vessels showed reduced VE-cadherin staining and significantly elevated plasma leakage indicating the breakdown of the blood-retinal barrier. This defect was associated with considerable macrophage infiltration in the retina. CONCLUSIONS: The normal retinal vascular development is dependent on R-Ras expression in pericytes, and the absence of it leads to unattenuated angiogenesis and significantly weakens the blood-retinal barrier. Our findings underscore the importance of R-Ras for pericyte function during the normal eye development.


Subject(s)
Blood-Retinal Barrier/metabolism , Microphthalmos/metabolism , Neovascularization, Pathologic , Pericytes/metabolism , Retinal Vessels/metabolism , ras Proteins/deficiency , Animals , Animals, Newborn , Antigens, CD/metabolism , Blood-Retinal Barrier/pathology , Cadherins/metabolism , Cell Movement , Cell Proliferation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Genetic Predisposition to Disease , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Microphthalmos/genetics , Microphthalmos/pathology , Pericytes/pathology , Phenotype , Receptor, Platelet-Derived Growth Factor beta/deficiency , Receptor, Platelet-Derived Growth Factor beta/genetics , Retinal Vessels/pathology , ras Proteins/genetics
4.
Bioconjug Chem ; 30(9): 2312-2316, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31433175

ABSTRACT

Mitochondria are essential targets for treatment of diseases with mitochondrial disorders such as diabetes, cancer, and cardiovascular and neurodegenerative diseases. Mitochondria penetrating peptides (MPPs) are composed of cationic and hydrophobic amino acids that can target and permeate the mitochondrial membrane. Herein, a novel d-argine-phenylalanine-d-argine-phenylalanine-d-argine-phenylalanine-NH2 (rFrFrF) was tagged with a rhodamine-based fluorescent chromophore (TAMRA). This probe (TAMRA-rFrFrF) exhibited advantageous properties for long-term mitochondria tracking as demonstrated by fluorescence microscopy. Cell viability assays and oxygen consumption rates indicate low cytotoxicity and high biocompatibility of the new contrast agent. Colocalization studies suggest that TAMRA-rFrFrF is a promising candidate for continuous mitochondrial tracking for up to 3 days.


Subject(s)
Cell Tracking/methods , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/metabolism , Fluorescent Dyes/chemistry , Mitochondria/metabolism , Rhodamines/chemistry , Cell Survival , HeLa Cells , Humans
5.
FASEB J ; : fj201700818RRR, 2018 May 18.
Article in English | MEDLINE | ID: mdl-29775418

ABSTRACT

The increase in cAMP levels in endothelial cells triggers cellular signaling to alter vascular permeability. It is generally considered that cAMP signaling stabilizes the endothelial barrier function and reduces permeability. However, previous studies have only examined the permeability shortly after cAMP elevation and thus have only investigated acute responses. Because cAMP is a key regulator of gene expression, elevated cAMP may have a delayed but profound impact on the endothelial permeability by altering the expression of the genes that are vital for the vessel wall stability. The small guanosine triphosphate hydrolase Ras-related protein (R-Ras) stabilizes VE-cadherin clustering and enhances endothelial barrier function, thereby stabilizing the integrity of blood vessel wall. Here we show that cAMP controls endothelial permeability through RRAS gene regulation. The prolonged cAMP elevation transcriptionally repressed RRAS in endothelial cells via a cAMP response element-binding protein (CREB) 3-dependent mechanism and significantly disrupted the adherens junction. These effects resulted in a marked increase of endothelial permeability that was reversed by R-Ras transduction. Furthermore, cAMP elevation in the endothelium by prostaglandin E2 or phosphodiesterase type 4 inhibition caused plasma leakage from intact microvessels in mouse skin. Our study demonstrated that, contrary to the widely accepted notion, cAMP elevation in endothelial cells ultimately increases vascular permeability, and the cAMP-dependent RRAS repression critically contributes to this effect.-Perrot, C. Y., Sawada, J., Komatsu, M. Prolonged activation of cyclic AMP signaling leads to endothelial barrier disruption via transcriptional repression of RRAS.

6.
J Biol Chem ; 290(13): 8133-45, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25645912

ABSTRACT

Abnormal angiogenesis is associated with a broad range of medical conditions, including cancer. The formation of neovasculature with functionally defective blood vessels significantly impacts tumor progression, metastasis, and the efficacy of anticancer therapies. Vascular endothelial growth factor (VEGF) potently induces vascular permeability and vessel growth in the tumor microenvironment, and its inhibition normalizes tumor vasculature. In contrast, the signaling of the small GTPase R-Ras inhibits excessive angiogenic growth and promotes the maturation of regenerating blood vessels. R-Ras signaling counteracts VEGF-induced vessel sprouting, permeability, and invasive activities of endothelial cells. In this study, we investigated the effect of R-Ras on VEGF receptor 2 (VEGFR2) activation by VEGF, the key mechanism for angiogenic stimulation. We show that tyrosine phosphorylation of VEGFR2 is significantly elevated in the tumor vasculature and dermal microvessels of VEGF-injected skin in R-Ras knockout mice. In cultured endothelial cells, R-Ras suppressed the internalization of VEGFR2, which is required for full activation of the receptor by VEGF. Consequently, R-Ras strongly suppressed autophosphorylation of the receptor at all five major tyrosine phosphorylation sites. Conversely, silencing of R-Ras resulted in increased VEGFR2 phosphorylation. This effect of R-Ras on VEGFR2 was, at least in part, dependent on vascular endothelial cadherin. These findings identify a novel function of R-Ras to control the response of endothelial cells to VEGF and suggest an underlying mechanism by which R-Ras regulates angiogenesis.


Subject(s)
Carcinoma, Lewis Lung/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Melanoma, Experimental/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , ras Proteins/physiology , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Carcinoma, Lewis Lung/blood supply , Cell Line, Tumor , Dermis/blood supply , Melanoma, Experimental/blood supply , Mice, Knockout , Microvessels/metabolism , Neoplasm Transplantation , Phosphorylation , Protein Processing, Post-Translational , Protein Transport
7.
Am J Pathol ; 184(2): 369-75, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24401613

ABSTRACT

A major limitation in the pharmacological treatment of pulmonary arterial hypertension (PAH) is the lack of pulmonary vascular selectivity. Recent studies have identified a tissue-penetrating homing peptide, CARSKNKDC (CAR), which specifically homes to hypertensive pulmonary arteries but not to normal pulmonary vessels or other tissues. Some tissue-penetrating vascular homing peptides have a unique ability to facilitate transport of co-administered drugs into the targeted cells/tissues without requiring physical conjugation of the drug to the peptide (bystander effect). We tested the hypothesis that co-administered CAR would selectively enhance the pulmonary vascular effects of i.v. vasodilators in Sugen5416/hypoxia/normoxia-exposed PAH rats. Systemically administered CAR was predominantly detected in cells of remodeled pulmonary arteries. Intravenously co-administered CAR enhanced pulmonary, but not systemic, effects of the vasodilators, fasudil and imatinib, in PAH rats. CAR increased lung tissue imatinib concentration in isolated PAH lungs without increasing pulmonary vascular permeability. Sublingual CAR was also effective in selectively enhancing the pulmonary vasodilation by imatinib and sildenafil. Our results suggest a new paradigm in the treatment of PAH, using an i.v./sublingual tissue-penetrating homing peptide to selectively augment pulmonary vascular effects of nonselective drugs without the potentially problematic conjugation process. CAR may be particularly useful as an add-on therapy to selectively enhance the pulmonary vascular efficacy of any ongoing drug treatment in patients with PAH.


Subject(s)
Drug Delivery Systems/methods , Hypertension, Pulmonary/drug therapy , Peptides/chemistry , Vasodilator Agents/therapeutic use , Administration, Sublingual , Amino Acid Sequence , Animals , Arterial Occlusive Diseases/drug therapy , Arterial Occlusive Diseases/pathology , Benzamides/pharmacology , Benzamides/therapeutic use , Familial Primary Pulmonary Hypertension , Hemodynamics/drug effects , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Imatinib Mesylate , Infusions, Intravenous , Injections, Intravenous , Male , Molecular Sequence Data , Piperazines/pharmacology , Piperazines/therapeutic use , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Treatment Outcome , Vasodilator Agents/administration & dosage , Vasodilator Agents/pharmacology
8.
J Vasc Res ; 52(5): 347-59, 2015.
Article in English | MEDLINE | ID: mdl-27029009

ABSTRACT

R-Ras is a Ras family small GTPase that is highly expressed in mature functional blood vessels in normal tissues. It inhibits pathological angiogenesis and promotes vessel maturation and stabilization. Previous studies suggest that R-Ras affects cellular signaling in endothelial cells, pericytes and smooth-muscle cells to regulate vessel formation and remodeling in adult tissues. R-Ras suppresses VEGF-induced endothelial permeability and vessel sprouting while promoting normalization of pathologically developing vessels in mice. It attenuates VEGF receptor-2 (VEGFR2) activation by inhibiting internalization of the receptor upon VEGF ligand binding, leading to significant reduction of VEGFR2 autophosphorylation. Here, we show that R-Ras strongly suppresses the VEGF-dependent activation of stress-activated protein kinase-2/p38 mitogen-activated protein kinase (SAPK2/p38MAPK) and the phosphorylation of downstream heat-shock protein 27 (HSP27), a regulator of actin cytoskeleton organization, in endothelial cells. The suppression of p38MAPK activation and HSP27 phosphorylation by R-Ras concurred with altered actin cytoskeleton architecture, reduced membrane protrusion and inhibition of endothelial cell migration toward VEGF. Silencing of endogenous R-Ras by RNA interference increased membrane protrusion and cell migration stimulated by VEGF, and these effects were offset by p38MAPK inhibitor SB203580. These results suggest that R-Ras regulates angiogenic activities of endothelial cells in part via inhibition of the p38MAPK-HSP27 axis of VEGF signaling.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Mitogen-Activated Protein Kinase 11/metabolism , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/pharmacology , ras Proteins/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Cell Movement/drug effects , Cells, Cultured , Enzyme Activation , Heat-Shock Proteins , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Mitogen-Activated Protein Kinase 11/antagonists & inhibitors , Molecular Chaperones , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , Signal Transduction/drug effects , Time Factors , Transfection , ras Proteins/genetics
9.
Org Biomol Chem ; 13(43): 10716-25, 2015 Nov 21.
Article in English | MEDLINE | ID: mdl-26351137

ABSTRACT

Observation of the activation and inhibition of angiogenesis processes is important in the progression of cancer. Application of targeting peptides, such as a small peptide that contains adjacent L-arginine (R), glycine (G) and L-aspartic acid (D) residues can afford high selectivity and deep penetration in vessel imaging. To facilitate deep tissue vasculature imaging, probes that can be excited via two-photon absorption (2PA) in the near-infrared (NIR) and subsequently emit in the NIR are essential. In this study, the enhancement of tissue image quality with RGD conjugates was investigated with new NIR-emitting pyranyl fluorophore derivatives in two-photon fluorescence microscopy. Linear and nonlinear photophysical properties of the new probes were comprehensively characterized; significantly the probes exhibited good 2PA over a broad spectral range from 700-1100 nm. Cell and tissue images were then acquired and examined, revealing deep penetration and high contrast with the new pyranyl RGD-conjugates up to 350 µm in tumor tissue.


Subject(s)
Fluorescent Dyes/chemistry , Nanoparticles/chemistry , Neoplasms/blood supply , Oligopeptides/chemistry , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/diagnosis , Carcinoma, Lewis Lung/metabolism , Fluorescent Dyes/metabolism , Humans , Infrared Rays , Integrins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence, Multiphoton , Nanoparticles/metabolism , Neoplasms/diagnosis , Neoplasms/metabolism , Oligopeptides/metabolism , Optical Imaging , Photons
10.
Mol Pharm ; 11(12): 4374-84, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25333706

ABSTRACT

This study sought to develop a liposomal delivery system of fasudil--an investigational drug for the treatment of pulmonary arterial hypertension (PAH)--that will preferentially accumulate in the PAH lungs. Liposomal fasudil was prepared by film-hydration method, and the drug was encapsulated by active loading. The liposome surface was coated with a targeting moiety, CARSKNKDC, a cyclic peptide; the liposomes were characterized for size, polydispersity index, zeta potential, and storage and nebulization stability. The in vitro drug release profiles and uptake by TGF-ß activated pulmonary arterial smooth muscle cells (PASMC) and alveolar macrophages were evaluated. The pharmacokinetics were monitored in male Sprague-Dawley rats, and the pulmonary hemodynamics were studied in acute and chronic PAH rats. The size, polydispersity index (PDI), and zeta potential of the liposomes were 206-216 nm, 0.058-0.084, and -20-42.7 mV, respectively. The formulations showed minimal changes in structural integrity when nebulized with a commercial microsprayer. The optimized formulation was stable for >4 weeks when stored at 4 °C. Fasudil was released in a continuous fashion over 120 h with a cumulative release of 76%. Peptide-linked liposomes were taken up at a higher degree by TGF-ß activated PASMCs; but alveolar macrophages could not engulf peptide-coated liposomes. The formulations did not injure the lungs; the half-life of liposomal fasudil was 34-fold higher than that of plain fasudil after intravenous administration. Peptide-linked liposomal fasudil, as opposed to plain liposomes, reduced the mean pulmonary arterial pressure by 35-40%, without influencing the mean systemic arterial pressure. This study establishes that CAR-conjugated inhalable liposomal fasudil offers favorable pharmacokinetics and produces pulmonary vasculature specific dilatation.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Hypertension, Pulmonary/drug therapy , Liposomes/chemistry , Peptides/chemistry , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/chemistry , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Animals , Male , Rats , Rats, Sprague-Dawley , Vasodilator Agents/chemistry , Vasodilator Agents/therapeutic use
11.
Cell Rep ; 43(3): 113837, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38402584

ABSTRACT

Communication between adjacent endothelial cells is important for the homeostasis of blood vessels. We show that quiescent endothelial cells use Jagged1 to instruct neighboring endothelial cells to assume a quiescent phenotype and secure the endothelial barrier. This phenotype enforcement by neighboring cells is operated by R-Ras through activation of Akt3, which results in upregulation of a Notch ligand Jagged1 and consequential upregulation of Notch target genes, such as UNC5B, and VE-cadherin accumulation in the neighboring cells. These signaling events lead to the stable interaction between neighboring endothelial cells to continue to fortify juxtacrine signaling via Jagged1-Notch. This mode of intercellular signaling provides a positive feedback regulation of endothelial cell-cell interactions and cellular quiescence required for the stabilization of the endothelium.


Subject(s)
Endothelial Cells , Membrane Proteins , Serrate-Jagged Proteins , Endothelial Cells/metabolism , Membrane Proteins/metabolism , Calcium-Binding Proteins/genetics , Intercellular Signaling Peptides and Proteins , Receptors, Notch/metabolism , Jagged-1 Protein/genetics
12.
J Vasc Res ; 50(2): 134-44, 2013.
Article in English | MEDLINE | ID: mdl-23258211

ABSTRACT

Restenosis is a major complication of coronary angioplasty, at least partly due to the fact that the origin and identity of contributing cell types are not well understood. In this study, we have investigated whether pericyte-like cells or mesenchymal stem cells (MSCs) from the adventitia contribute to restenosis. We demonstrate that while cells expressing the pericyte markers NG2, platelet-derived growth factor receptor ß, and CD146 are rare in the adventitia of uninjured mouse femoral arteries, following injury their numbers strongly increase. Some of these adventitial pericyte-like cells acquire a more MSC-like phenotype (CD90+ and CD29+ are up-regulated) and also appear in the restenotic neointima. Via bone marrow transplantation and ex vivo artery culture approaches, we demonstrate that the pericyte-like MSCs of the injured femoral artery are not derived from the bone marrow, but originate in the adventitia itself mainly via the proliferation of resident pericyte-like cells. In summary, we have identified a population of resident adventitial pericyte-like cells or MSCs that contribute to restenosis following arterial injury. These cells are different from myofibroblasts, smooth muscle cells, and other progenitor populations that have been shown to participate in the restenotic process.


Subject(s)
Adventitia/pathology , Arterial Occlusive Diseases/physiopathology , Femoral Artery/injuries , Mesenchymal Stem Cells/physiology , Neointima/physiopathology , Pericytes/physiology , Animals , Antigens/analysis , Antigens/biosynthesis , Antigens/genetics , Antigens, CD/analysis , Aorta, Thoracic/cytology , Bone Marrow Transplantation , Cell Lineage , Constriction, Pathologic , Femoral Artery/pathology , Gene Expression Profiling , Genes, Reporter , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Proteoglycans/analysis , Proteoglycans/biosynthesis , Proteoglycans/genetics , Radiation Chimera , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , Recurrence
13.
Mol Pharm ; 10(5): 1655-67, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23485062

ABSTRACT

This study sought to investigate the efficacy of a noninvasive and long acting polymeric particle based formulation of prostaglandin E1 (PGE1), a potent pulmonary vasodilator, in alleviating the signs of pulmonary hypertension (PH) and reversing the biochemical changes that occur in the diseased lungs. PH rats, developed by a single subcutaneous injection of monocrotaline (MCT), were treated with two types of polymeric particles of PGE1, porous and nonporous, and intratracheal or intravenous plain PGE1. For chronic studies, rats received either intratracheal porous poly(lactic-co-glycolic acid) (PLGA) particles, once- or thrice-a-day, or plain PGE1 thrice-a-day for 10 days administered intratracheally or intravenously. The influence of formulations on disease progression was studied by measuring the mean pulmonary arterial pressure (MPAP), evaluating right ventricular hypertrophy and assessing various molecular and cellular makers including the degree of muscularization, platelet aggregation, matrix metalloproteinase-2 (MMP-2), and proliferating cell nuclear antigen (PCNA). Both plain PGE1 and large porous particles of PGE1 reduced MPAP and right ventricular hypertrophy (RVH) in rats that received the treatments for 10 days. Polymeric porous particles of PGE1 produced the same effects at a reduced dosing frequency compared to plain PGE1 and caused minimal off-target effects on systemic hemodynamics. Microscopic and immunohistochemical studies revealed that porous particles of PGE1 also reduced the degree of muscularization, von Willebrand factor (vWF), and PCNA expression in the lungs of PH rats. Overall, our study suggests that PGE1 loaded inhalable particulate formulations improve PH symptoms and arrest the progression of disease at a reduced dosing frequency compared to plain PGE1.


Subject(s)
Alprostadil/administration & dosage , Hypertension, Pulmonary/drug therapy , Administration, Inhalation , Animals , Delayed-Action Preparations , Disease Models, Animal , Disease Progression , Drug Carriers/chemistry , Hemodynamics/drug effects , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/pathology , Lactic Acid/chemistry , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Vasodilator Agents/administration & dosage , von Willebrand Factor/metabolism
14.
Am J Pathol ; 178(6): 2489-95, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21549345

ABSTRACT

Pulmonary arterial hypertension (PAH) is a disorder of the pulmonary vasculature associated with elevated pulmonary vascular resistance. Despite recent advances in the treatment of PAH, with eight approved clinical therapies and additional therapies undergoing clinical trials, PAH remains a serious life-threatening condition. The lack of pulmonary vascular selectivity and associated systemic adverse effects of these therapies remain the main obstacles to successful treatment. Peptide-mediated drug delivery that specifically targets the vasculature of PAH lungs may offer a solution to the lack of drug selectivity. Herein, we show highly selective targeting of rat PAH lesions by a novel cyclic peptide, CARSKNKDC (CAR). Intravenous administration of CAR peptide resulted in intense accumulation of the peptide in monocrotaline-induced and SU5416/hypoxia-induced hypertensive lungs but not in healthy lungs or other organs of PAH rats. CAR homed to all layers of remodeled pulmonary arteries, ie, endothelium, neointima, medial smooth muscle, and adventitia, in the hypertensive lungs. CAR also homed to capillary vessels and accumulated in the interstitial space of the PAH lungs, manifesting its extravasation activity. These results demonstrated the remarkable ability of CAR to selectively target PAH lung vasculature and effectively penetrate and spread throughout the diseased lung tissue. These results suggest the clinical utility of CAR in the targeted delivery of therapeutic compounds and imaging probes to PAH lungs.


Subject(s)
Drug Delivery Systems , Hypertension, Pulmonary/pathology , Peptides/pharmacology , Pulmonary Artery/pathology , Amino Acid Sequence , Animals , Humans , Hypertension, Pulmonary/complications , Hypoxia/complications , Indoles/pharmacology , Lung/blood supply , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Molecular Sequence Data , Monocrotaline , Peptides/administration & dosage , Peptides/chemistry , Pulmonary Artery/drug effects , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/metabolism , Time Factors
15.
Nat Med ; 11(12): 1346-50, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16286923

ABSTRACT

R-Ras is a small GTPase of the Ras family that regulates cell survival and integrin activity. Despite a number of in vitro studies, the in vivo function of R-Ras remains unclear. Here, we used R-Ras-null mice to explore the in vivo function of this small GTPase. Our results show a role for R-Ras as a regulator of vascular differentiation that primarily affects the remodeling of blood vessels. We show that R-Ras-null mice, although otherwise phenotypically normal, mount excessive vascular responses. We found that in vivo R-Ras expression is largely confined to fully differentiated smooth muscle cells, including those of blood vessels, and to endothelial cells. Challenging the R-Ras-null mice with arterial injury or tumor implantation showed exaggerated neointimal thickening in response to the injury and increased angiogenesis in the tumors. In wild-type mice, R-Ras expression was greatly reduced in hyperplastic neointimal smooth muscle cells and in angiogenic endothelial cells. Forced expression of activated R-Ras suppressed mitogenic and invasive activities of growth factor-stimulated vascular cells. These results establish an unexpected role for R-Ras in blood vessel homeostasis and suggest that R-Ras signaling may offer a target for therapeutic intervention in vascular diseases.


Subject(s)
GTP Phosphohydrolases/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Tunica Intima/pathology , ras Proteins/metabolism , Animals , DNA Primers , Endothelial Cells/metabolism , Fluorescent Antibody Technique , GTP Phosphohydrolases/genetics , Gene Deletion , Genetic Vectors , Hyperplasia/metabolism , Hyperplasia/pathology , Immunoblotting , Immunohistochemistry , Lentivirus , Mice , Myocytes, Smooth Muscle/metabolism , Neovascularization, Pathologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tunica Intima/metabolism , ras Proteins/genetics
16.
Cancer Immunol Res ; 10(4): 468-481, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35201289

ABSTRACT

High endothelial venules (HEV) are specialized post-capillary venules that recruit naïve lymphocytes to lymph nodes. HEVs are essential for the development of adaptive immunity. HEVs can also develop in tumors where they are thought to be important for recruiting naïve T cells and B cells into the tumors and locally enhancing antitumor immunity by supporting the formation of tertiary lymphoid structures. Herein, we used comparative transcriptome analysis of human breast cancer to investigate genes differentially expressed between tumor-associated HEVs and the rest of the tumor vasculature. Tumor vessels highly expressing HEV-upregulated genes, such as the homeobox gene MEOX2 and the tetraspanin gene TSPAN7, were associated with extensive infiltration of T and B cells and the occurrence of tertiary lymphoid structures, which is known to predict therapeutic responses to immune-checkpoint inhibitors. Moreover, high transcript counts of these genes in clinical tumor specimens were associated with a significant survival benefit in advanced breast cancer. The molecular signature of HEVs identified herein may be useful for guiding immunotherapies and provides a new direction for investigating tumor-associated HEVs and their clinical significance. See related Spotlight by Gallimore, p. 371.


Subject(s)
Breast Neoplasms , Tertiary Lymphoid Structures , Female , Humans , Lymph Nodes/pathology , Lymphocytes , Venules/pathology
17.
Bioconjug Chem ; 22(7): 1438-50, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21688841

ABSTRACT

A two-photon absorbing (2PA) and aggregation-enhanced near-infrared (NIR) emitting pyran derivative, encapsulated in and stabilized by silica nanoparticles (SiNPs), is reported as a nanoprobe for two-photon fluorescence microscopy (2PFM) bioimaging that overcomes the fluorescence quenching associated with high chromophore loading. The new SiNP probe exhibited aggregate-enhanced emission producing nearly twice as strong a signal as the unaggregated dye, a 3-fold increase in two-photon absorption relative to the DFP in solution, and approximately 4-fold increase in photostability. The surface of the nanoparticles was functionalized with a folic acid (FA) derivative for folate-mediated delivery of the nanoprobe for 2PFM bioimaging. Surface modification of SiNPs with the FA derivative was supported by zeta potential variation and (1)H NMR spectral characterization of the SiNPs as a function of surface modification. In vitro studies using HeLa cells expressing a folate receptor (FR) indicated specific cellular uptake of the functionalized nanoparticles. The nanoprobe was demonstrated for FR-targeted one-photon in vivo imaging of HeLa tumor xenograft in mice upon intravenous injection of the probe. The FR-targeting nanoprobe not only exhibited highly selective tumor targeting but also readily extravasated from tumor vessels, penetrated into the tumor parenchyma, and was internalized by the tumor cells. Two-photon fluorescence microscopy bioimaging provided three-dimensional (3D) cellular-level resolution imaging up to 350 µm deep in the HeLa tumor.


Subject(s)
Fluorescent Dyes , Folate Receptors, GPI-Anchored/metabolism , Folic Acid , Nanoparticles , Neoplasms/diagnosis , Pyrans , Silicon Dioxide , Animals , Female , Fluorescent Dyes/chemistry , Folic Acid/chemistry , HeLa Cells , Humans , Mice , Mice, Nude , Microscopy, Fluorescence, Multiphoton/methods , Nanoparticles/chemistry , Neoplasms/metabolism , Neoplasms/pathology , Pyrans/chemistry , Silicon Dioxide/chemistry
18.
Bioconjug Chem ; 22(10): 2060-71, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-21950485

ABSTRACT

We report the synthesis and characterization of two amine reactive fluorescent dyes with efficient two-photon absorption (2PA) properties and high fluorescence quantum yields. Bioconjugation of these dyes with the DC-101 antibody proved to be useful for selectively imaging the vascular endothelial growth factor receptor 2 (VEGFR-2) in cells expressing this receptor in vitro and in "whole" mounted excised tumors (ex vivo) by two-photon fluorescence microscopy (2PFM). The penetration depths reached within the tumors by 2PFM was over 800 µm. In addition, the concentration of dye required for incubation of these bioconjugates was in the picomolar domain, the probes possessed very good photostability, and the 2PFM setup did not require any additional means of increasing the collection efficiencies of fluorescent photons to achieve the relatively deep tissue imaging that was realized, due, in large part, to the favorable photophysical properties of the new probes.


Subject(s)
Antibodies, Monoclonal/chemistry , Carcinoma, Lewis Lung/diagnosis , Fluorescent Dyes/chemistry , Immunoconjugates/chemistry , Vascular Endothelial Growth Factor Receptor-2/analysis , Animals , Antibodies, Monoclonal/immunology , Cell Line , Humans , Immunoconjugates/immunology , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Models, Molecular , Molecular Imaging , Swine , Vascular Endothelial Growth Factor Receptor-2/immunology
19.
J Invest Dermatol ; 141(2): 385-394, 2021 02.
Article in English | MEDLINE | ID: mdl-32888955

ABSTRACT

MicroRNAs (miRs) are important posttranscriptional regulators of cell fate in both normal and disease states. miR-211 has previously been shown to be a direct regulator of metabolism in BRAFV600E-mutant melanoma cells in vitro. Here, we report that miR-211 expression promotes the aggressive growth of BRAFV600E-mutant melanoma xenografts in vivo. miR-211 promoted proliferation through the posttranscriptional activation of extracellular signal-regulated kinase (ERK) 5 signaling, which has recently been implicated in the resistance to BRAF and MAPK/ERK kinase inhibitors. We therefore examined whether miR-211 similarly modulated melanoma resistance to the BRAF inhibitor vemurafenib and the MAPK/ERK kinase inhibitor cobimetinib. Consistent with this model, miR-211 expression increased melanoma cell resistance to both the inhibitors, and this resistance was associated with an increased ERK5 phosphorylation. miR-211 mediates these effects by directly inhibiting the expression of DUSP6, an ERK5 pathway-specific phosphatase and now shown to be an miR-211 target gene. These results dissect the role of the miR-211-DUSP6-ERK5 axis in melanoma tumor growth and suggest a mechanism for the development of drug-resistant tumors and a target for overcoming resistance.


Subject(s)
Drug Resistance, Neoplasm/genetics , Dual Specificity Phosphatase 6/genetics , Melanoma/drug therapy , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Protein Kinase Inhibitors/pharmacology , Animals , Azetidines/pharmacology , Azetidines/therapeutic use , Cell Line, Tumor , Cell Proliferation/genetics , Dual Specificity Phosphatase 6/metabolism , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System/genetics , Melanoma/genetics , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinase 7/genetics , Mutation , Phosphorylation/genetics , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Vemurafenib/pharmacology , Vemurafenib/therapeutic use , Xenograft Model Antitumor Assays
20.
Sci Rep ; 10(1): 11186, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32636414

ABSTRACT

A close association between pericytes and endothelial cells (ECs) is crucial to the stability and function of capillary blood vessels and microvessels. The loss or dysfunction of pericytes results in significant disruption of these blood vessels as observed in pathological conditions, including cancer, diabetes, stroke, and Alzheimer's disease. Prostaglandin E2 (PGE2) is a lipid mediator of inflammation, and its tissue concentration is elevated in cancer and neurological disorders. Here, we show that the exposure to PGE2 switches pericytes to a fast-migrating, loosely adhered phenotype that fails to intimately interact with ECs. N-cadherin and connexin-43 in adherens junction and gap junction between pericytes and ECs are downregulated by EP-4 and EP-1-dependent mechanisms, leading to breakdown of the pericyte-EC interaction. Furthermore, R-Ras, a small GTPase important for vascular normalization and vessel stability, is transcriptionally repressed by PGE2 in an EP4-dependent manner. Mouse dermal capillary vessels lose pericyte coverage substantially upon PGE2 injection into the skin. Our results suggest that EP-mediated direct disruption of pericytes by PGE2 is a key process for vascular destabilization. Restoring pericyte-EC interaction using inhibitors of PGE2 signaling may offer a therapeutic strategy in cancer and neurological disorders, in which pericyte dysfunction contributes to the disease progression.


Subject(s)
Dinoprostone/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Pericytes/drug effects , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Cell Movement , Cells, Cultured , Connexin 43/metabolism , Down-Regulation , Gap Junctions/metabolism , HEK293 Cells , HT29 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Pericytes/metabolism , Pericytes/physiology , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL