Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Curr Atheroscler Rep ; 26(7): 317-329, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38753254

RESUMEN

PURPOSE OF REVIEW: Low-density lipoprotein (LDL) poses a risk for atherosclerotic cardiovascular disease (ASCVD). As LDL comprises various subtypes differing in charge, density, and size, understanding their specific impact on ASCVD is crucial. Two highly atherogenic LDL subtypes-electronegative LDL (L5) and Lp(a)-induce vascular cell apoptosis and atherosclerotic changes independent of plasma cholesterol levels, and their mechanisms warrant further investigation. Here, we have compared the roles of L5 and Lp(a) in the development of ASCVD. RECENT FINDINGS: Lp(a) tends to accumulate in artery walls, promoting plaque formation and potentially triggering atherosclerosis progression through prothrombotic or antifibrinolytic effects. High Lp(a) levels correlate with calcific aortic stenosis and atherothrombosis risk. L5 can induce endothelial cell apoptosis and increase vascular permeability, inflammation, and atherogenesis, playing a key role in initiating atherosclerosis. Elevated L5 levels in certain high-risk populations may serve as a distinctive predictor of ASCVD. L5 and Lp(a) are both atherogenic lipoproteins contributing to ASCVD through distinct mechanisms. Lp(a) has garnered attention, but equal consideration should be given to L5.


Asunto(s)
Aterosclerosis , Lipoproteína(a) , Humanos , Lipoproteína(a)/sangre , Lipoproteína(a)/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/sangre , Lipoproteínas LDL/sangre , Lipoproteínas LDL/metabolismo , Apoptosis , Animales
2.
J Biol Chem ; 288(27): 19414-28, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23703610

RESUMEN

Activation of the integrin family of cell adhesion receptors on progenitor cells may be a viable approach to enhance the effects of stem cell-based therapies by improving cell retention and engraftment. Here, we describe the synthesis and characterization of the first small molecule agonist identified for the integrin α4ß1 (also known as very late antigen-4 or VLA-4). The agonist, THI0019, was generated via two structural modifications to a previously identified α4ß1 antagonist. THI0019 greatly enhanced the adhesion of cultured cell lines and primary progenitor cells to α4ß1 ligands VCAM-1 and CS1 under both static and flow conditions. Furthermore, THI0019 facilitated the rolling and spreading of cells on VCAM-1 and the migration of cells toward SDF-1α. Molecular modeling predicted that the compound binds at the α/ß subunit interface overlapping the ligand-binding site thus indicating that the compound must be displaced upon ligand binding. In support of this model, an analog of THI0019 modified to contain a photoreactive group was used to demonstrate that when cross-linked to the integrin, the compound behaves as an antagonist instead of an agonist. In addition, THI0019 showed cross-reactivity with the related integrin α4ß7 as well as α5ß1 and αLß2. When cross-linked to αLß2, the photoreactive analog of THI0019 remained an agonist, consistent with it binding at the α/ß subunit interface and not at the ligand-binding site in the inserted ("I") domain of the αL subunit. Co-administering progenitor cells with a compound such as THI0019 may provide a mechanism for enhancing stem cell therapy.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Integrina alfa4beta1/agonistas , Modelos Moleculares , Células Madre/metabolismo , Antígeno CD11a/genética , Antígeno CD11a/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfa4beta1/genética , Integrina alfa4beta1/metabolismo , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Células Jurkat , Células Madre/citología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
3.
J Biol Chem ; 287(39): 32981-92, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22833667

RESUMEN

The molecular mechanisms that regulate the endothelial response during transendothelial migration (TEM) of invasive cancer cells remain elusive. Tyrosine phosphorylation of vascular endothelial cadherin (VE-cad) has been implicated in the disruption of endothelial cell adherens junctions and in the diapedesis of metastatic cancer cells. We sought to determine the signaling mechanisms underlying the disruption of endothelial adherens junctions after the attachment of invasive breast cancer cells. Attachment of invasive breast cancer cells (MDA-MB-231) to human umbilical vein endothelial cells induced tyrosine phosphorylation of VE-cad, dissociation of ß-catenin from VE-cad, and retraction of endothelial cells. Breast cancer cell-induced tyrosine phosphorylation of VE-cad was mediated by activation of the H-Ras/Raf/MEK/ERK signaling cascade and depended on the phosphorylation of endothelial myosin light chain (MLC). The inhibition of H-Ras or MLC in endothelial cells inhibited TEM of MDA-MB-231 cells. VE-cad tyrosine phosphorylation in endothelial cells induced by the attachment of MDA-MB-231 cells was mediated by MDA-MB-231 α(2)ß(1) integrin. Compared with highly invasive MDA-MB-231 breast cancer cells, weakly invasive MCF-7 breast cancer cells expressed lower levels of α(2)ß(1) integrin. TEM of MCF-7 as well as induction of VE-cad tyrosine phosphorylation and dissociation of ß-catenin from the VE-cad complex by MCF-7 cells were lower than in MDA-MB-231 cells. These processes were restored when MCF-7 cells were treated with ß(1)-activating antibody. Moreover, the response of endothelial cells to the attachment of prostatic (PC-3) and ovarian (SKOV3) invasive cancer cells resembled the response to MDA-MB-231 cells. Our study showed that the MDA-MB-231 cell-induced disruption of endothelial adherens junction integrity is triggered by MDA-MB-231 cell α(2)ß(1) integrin and is mediated by H-Ras/MLC-induced tyrosine phosphorylation of VE-cad.


Asunto(s)
Uniones Adherentes/metabolismo , Antígenos CD/metabolismo , Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Integrina alfa2beta1/metabolismo , Uniones Adherentes/genética , Uniones Adherentes/patología , Antígenos CD/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Cadherinas/genética , Línea Celular Tumoral , Endotelio Vascular/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Integrina alfa2beta1/genética , Sistema de Señalización de MAP Quinasas/genética , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Invasividad Neoplásica , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Fosforilación/genética
4.
Biomater Sci ; 10(21): 6217-6229, 2022 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-36102692

RESUMEN

Adjuvants play a critical role in enhancing vaccine efficacy; however, there is a need to develop new immunomodulatory compounds to address emerging pathogens and to expand the use of immunotherapies. Multidomain peptides (MDPs) are materials composed of canonical amino acids that form injectable supramolecular hydrogels under physiological salt and pH conditions. MDP hydrogels are rapidly infiltrated by immune cells in vivo and have previously been shown to influence cytokine production. Therefore, we hypothesized that these immunostimulatory characteristics would allow MDPs to function as vaccine adjuvants. Herein, we demonstrate that loading antigen into MDP hydrogels does not interfere with their rheological properties and that positively charged MDPs can act as antigen depots, as demonstrated by their ability to release ovalbumin (OVA) over a period of 7-9 days in vivo. Mice vaccinated with MDP-adjuvanted antigen generated significantly higher IgG titers than mice treated with the unadjuvanted control, suggesting that these hydrogels potentiate humoral immunity. Interestingly, MDP hydrogels did not elicit a robust cellular immune response, as indicated by the lower production of IgG2c and smaller populations of tetramer-positive CD8+ T splenocytes compared to mice vaccinated alum-adjuvanted OVA. Together, the data suggest that MDP hydrogel adjuvants strongly bias the immune response towards humoral immunity while evoking a very limited cellular immune response. As a result, MDPs may have the potential to serve as adjuvants for applications that benefit exclusively from humoral immunity.


Asunto(s)
Hidrogeles , Inmunidad Humoral , Ratones , Animales , Ovalbúmina , Adyuvantes Inmunológicos/química , Antígenos , Péptidos , Adyuvantes Farmacéuticos , Inmunoglobulina G , Aminoácidos , Citocinas
5.
J Clin Invest ; 132(13)2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35552271

RESUMEN

The inability of CD8+ effector T cells (Teffs) to reach tumor cells is an important aspect of tumor resistance to cancer immunotherapy. The recruitment of these cells to the tumor microenvironment (TME) is regulated by integrins, a family of adhesion molecules that are expressed on T cells. Here, we show that 7HP349, a small-molecule activator of lymphocyte function-associated antigen-1 (LFA-1) and very late activation antigen-4 (VLA-4) integrin cell-adhesion receptors, facilitated the preferential localization of tumor-specific T cells to the tumor and improved antitumor response. 7HP349 monotherapy had modest effects on anti-programmed death 1-resistant (anti-PD-1-resistant) tumors, whereas combinatorial treatment with anti-cytotoxic T lymphocyte-associated protein 4 (anti-CTLA-4) increased CD8+ Teff intratumoral sequestration and synergized in cooperation with neutrophils in inducing cancer regression. 7HP349 intratumoral CD8+ Teff enrichment activity depended on CXCL12. We analyzed gene expression profiles using RNA from baseline and on treatment tumor samples of 14 melanoma patients. We identified baseline CXCL12 gene expression as possibly improving the likelihood or response to anti-CTLA-4 therapies. Our results provide a proof-of-principle demonstration that LFA-1 activation could convert a T cell-exclusionary TME to a T cell-enriched TME through mechanisms involving cooperation with innate immune cells.


Asunto(s)
Antígeno-1 Asociado a Función de Linfocito , Melanoma , Linfocitos T CD8-positivos , Antígeno CTLA-4 , Humanos , Inmunoterapia/métodos , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Linfocitos Infiltrantes de Tumor , Melanoma/tratamiento farmacológico , Melanoma/genética , Receptor de Muerte Celular Programada 1 , Linfocitos T/metabolismo , Microambiente Tumoral
6.
Bioconjug Chem ; 22(8): 1706-14, 2011 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-21749133

RESUMEN

A key issue regarding the use of stem cells in cardiovascular regenerative medicine is their retention in target tissues. Here, we have generated and assessed a bispecific antibody heterodimer designed to improve the retention of bone-marrow-derived multipotent stromal cells (BMMSC) in cardiac tissue damaged by myocardial infarction. The heterodimer comprises an anti-human CD90 monoclonal antibody (mAb) (clone 5E10) and an anti-myosin light chain 1 (MLC1) mAb (clone MLM508) covalently cross-linked by a bis-arylhydrazone. We modified the anti-CD90 antibody with a pegylated-4-formylbenzamide moiety to a molar substitution ratio (MSR) of 2.6 and the anti-MLC1 antibody with a 6-hydrazinonicotinamide moiety to a MSR of 0.9. The covalent modifications had no significant deleterious effect on mAb epitope binding. Furthermore, the binding of anti-CD90 antibody to BMMSCs did not prevent their differentiation into adipo-, chondro-, or osteogenic lineages. Modified antibodies were combined under mild conditions (room temperature, pH 6, 1 h) in the presence of a catalyst (aniline) to allow for rapid generation of the covalent bis-arylhydrazone, which was monitored at A(354). We evaluated epitope immunoreactivity for each mAb in the construct. Flow cytometry demonstrated binding of the bispecific construct to BMMSCs that was competed by free anti-CD90 mAb, verifying that modification and cross-linking were not detrimental to the anti-CD90 complementarity-determining region. Similarly, ELISA-based assays demonstrated bispecific antibody binding to plastic-immobilized recombinant MLC1. Excess anti-MLC1 mAb competed for bispecific antibody binding. Finally, the anti-CD90 × anti-MLC1 bispecific antibody construct induced BMMSC adhesion to plastic-immobilized MLC1 that was resistant to shear stress, as measured in parallel-plate flow chamber assays. We used mAbs that bind both human antigens and the respective pig homologues. Thus, the anti-CD90 × anti-MLC1 bispecific antibody may be used in large animal studies of acute myocardial infarction and may provide a starting point for clinical studies.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Terapia Molecular Dirigida/métodos , Células Madre Multipotentes/inmunología , Infarto del Miocardio/tratamiento farmacológico , Cadenas Ligeras de Miosina/inmunología , Células del Estroma/inmunología , Antígenos Thy-1/inmunología , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/inmunología , Células de la Médula Ósea , Humanos , Infarto del Miocardio/patología , Miocardio , Porcinos
7.
NPJ Vaccines ; 6(1): 114, 2021 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-34497271

RESUMEN

The development of suitable safe adjuvants to enhance appropriate antigen-driven immune responses remains a challenge. Here we describe the adjuvant properties of a small molecule activator of the integrins αLß2 and α4ß1, named 7HP349, which can be safely delivered systemically independent of antigen. 7HP349 directly activates integrin cell adhesion receptors crucial for the generation of an immune response. When delivered systemically in a model of Chagas disease following immunization with a DNA subunit vaccine encoding candidate T. cruzi antigens, TcG2 and TcG4, 7HP349 enhanced the vaccine efficacy in both prophylactic and therapeutic settings. In a prophylactic setting, mice immunized with 7HP349 adjuvanted vaccine exhibited significantly improved control of acute parasite burden in cardiac and skeletal muscle as compared to vaccination alone. When administered with vaccine therapeutically, parasite burden was again decreased, with the greatest adjuvant effect of 7HP349 being noted in skeletal muscle. In both settings, adjuvantation with 7HP349 was effective in decreasing pathological inflammatory infiltrate, improving the integrity of tissue, and controlling tissue fibrosis in the heart and skeletal muscle of acutely and chronically infected Chagas mice. The positive effects correlated with increased splenic frequencies of CD8+T effector cells and an increase in the production of IFN-γ and cytolytic molecules (perforin and granzyme) by the CD4+ and CD8+ effector and central memory subsets in response to challenge infection. This demonstrates that 7HP349 can serve as a systemically administered adjuvant to enhance T cell-mediated immune responses to vaccines. This approach could be applied to numerous vaccines with no reformulation of existing stockpiles.

8.
Biochem Biophys Res Commun ; 400(4): 619-24, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20807504

RESUMEN

The development of antagonists to the α4 integrin family of cell adhesion molecules has been an active area of pharmaceutical research to treat inflammatory and autoimmune diseases. Presently being tested in human clinical trials are compounds selective for α4ß1 (VLA-4) as well as several dual antagonists that inhibit both α4ß1 and α4ß7. The value of a dual versus a selective small molecule antagonist as well as the consequences of inhibiting different affinity states of the α4 integrins have been debated in the literature. Here, we characterize TBC3486, a N,N-disubstituted amide, which represents a unique structural class of non-peptidic, small molecule VLA-4 antagonists. Using a variety of adhesion assay formats as well as flow cytometry experiments using mAbs specific for certain activation-dependent integrin epitopes we demonstrate that TBC3486 preferentially targets the high affinity conformation of α4ß1 and behaves as a ligand mimetic. The antagonist is capable of blocking integrin-dependent T-cell co-activation in vitro as well as proves to be efficacious in vivo at low doses in two animal models of allergic inflammation. These data suggest that a small molecule α4 integrin antagonist selective for α4ß1 over α4ß7 and, specifically, selective for the high affinity conformation of α4ß1 may prove to be an effective therapy for multiple inflammatory diseases in humans.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Inflamación/tratamiento farmacológico , Integrina alfa4beta1/antagonistas & inhibidores , Tiofenos/farmacología , Urea/análogos & derivados , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Humanos , Hipersensibilidad/tratamiento farmacológico , Integrina alfa4beta1/química , Células Jurkat , Activación de Linfocitos/efectos de los fármacos , Ratones , Conformación Proteica/efectos de los fármacos , Eosinofilia Pulmonar/tratamiento farmacológico , Linfocitos T/efectos de los fármacos , Tiofenos/uso terapéutico , Urea/farmacología , Urea/uso terapéutico
9.
Biomaterials ; 231: 119667, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31855625

RESUMEN

Multidomain Peptide (MDP) hydrogels are nanofibrous materials with many potential biomedical applications. The peptide sequence design of these materials offers high versatility and allows for the incorporation of various chemical functionalities into the nanofibrous scaffold. It is known that host response to biomaterials is strongly affected by factors such as size, shape, stiffness, and chemistry. However, there is a lack of fundamental understanding of the host response to different MDP hydrogels. In particular, it is unknown what effect the chemical functionality displayed on the nanofiber has on biological activity. Here we evaluated the early inflammatory host response to four MDP hydrogels displaying amines, guanidinium ions, and carboxylates in a subcutaneous injection model. While all the studied peptide materials possess similar nanostructure and physical properties, they trigger markedly different inflammatory responses. These were characterized by immunophenotyping of the cellular infiltrate using multi-color flow cytometry. The negatively-charged peptides elicit minimal inflammation characterized by tissue-resident macrophage infiltration, fast remodeling, and no collagen deposition or blood vessel formation within the implants. In contrast, the positively-charged peptides are highly infiltrated by immune cells, are remodeled at a slower rate, promote angiogenesis, and result in a high degree of collagen deposition. The presence of dynamic cell phenotypes characterizes the inflammation caused by the lysine-based peptide, including inflammatory monocytes, macrophages, and lymphoid cells, which is seen to be resolving over time. The arginine-based hydrogel shows higher inflammatory response with a persistent and significant infiltration of polymorphonuclear myeloid-derived cells, even ten days after implantation. This understanding of the immune response to peptide biomaterials improves our ability to design effective materials and to tailor their use for specific biomedical applications.


Asunto(s)
Hidrogeles , Nanofibras , Materiales Biocompatibles , Inmunidad , Péptidos
10.
Front Immunol ; 11: 575085, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33488575

RESUMEN

Leukocyte inflammatory responses require integrin cell-adhesion molecule signaling through spleen tyrosine kinase (Syk), a non-receptor kinase that binds directly to integrin ß-chain cytoplasmic domains. Here, we developed a high-throughput screen to identify small molecule inhibitors of the Syk-integrin cytoplasmic domain interactions. Screening small molecule compound libraries identified the ß-lactam antibiotics cefsulodin and ceftazidime, which inhibited integrin ß-subunit cytoplasmic domain binding to the tandem SH2 domains of Syk (IC50 range, 1.02-4.9 µM). Modeling suggested antagonist binding to Syk outside the pITAM binding site. Ceftazidime inhibited integrin signaling via Syk, including inhibition of adhesion-dependent upregulation of interleukin-1ß and monocyte chemoattractant protein-1, but did not inhibit ITAM-dependent phosphorylation of Syk mediated by FcγRI signaling. Our results demonstrate a novel means to target Syk independent of its kinase and pITAM binding sites such that integrin signaling via this kinase is abrogated but ITAM-dependent signaling remains intact. As integrin signaling through Syk is essential for leukocyte activation, this may represent a novel approach to target inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Cefsulodina/farmacología , Ceftazidima/farmacología , Cadenas beta de Integrinas/efectos de los fármacos , Leucocitos/efectos de los fármacos , Quinasa Syk/antagonistas & inhibidores , Antiinflamatorios/química , Cefsulodina/química , Ceftazidima/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Cadenas beta de Integrinas/química , Cadenas beta de Integrinas/metabolismo , Leucocitos/enzimología , Masculino , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Bibliotecas de Moléculas Pequeñas , Quinasa Syk/química , Quinasa Syk/metabolismo , Células THP-1
11.
BioDrugs ; 22(2): 85-100, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18345706

RESUMEN

Chronic obstructive pulmonary disease (COPD) and asthma are inflammatory diseases of the lung where a hallmark feature is excessive leukocyte infiltration that leads to tissue injury. Cell adhesion molecules (e.g. selectins and integrins) play a key role in cell trafficking, and in the lung they regulate leukocyte extravasation, migration within the interstitium, cellular activation, and tissue retention. All selectin family members (including L-selectin, P-selectin, and E-selectin) and many of the beta1 and beta2 integrins appear to be important therapeutic targets, as numerous animal studies have demonstrated essential roles for these cell adhesion molecules in lung inflammation. Not surprisingly, these families of adhesion molecules have been under intense investigation by the pharmaceutical industry for the development of novel therapeutics. Integrins are validated drug targets, as drugs that antagonize integrin alphaIIbbeta3 (e.g. abciximab), integrin alphaLbeta2 (efalizumab), and integrin alpha4beta1 (natalizumab) are currently US FDA-approved for acute coronary syndromes, psoriasis, and multiple sclerosis, respectively. However, none has been approved for indications related to asthma or COPD. Here, we provide an overview of roles played by selectins and integrins in lung inflammation. We also describe recent clinical results (both failures and successes) in developing adhesion molecule antagonists, with specific emphasis on those targets that may have potential benefit in asthma and COPD. Early clinical trials using selectin and integrin antagonists have met with limited success. However, recent positive phase II clinical trials with a small-molecule selectin antagonist (bimosiamose) and a small-molecule integrin alpha4beta1 antagonist (valategrast [R411]), have generated enthusiastic anticipation that novel strategies to treat asthma and COPD may be forthcoming.


Asunto(s)
Antiinflamatorios/uso terapéutico , Asma/tratamiento farmacológico , Quimiotaxis de Leucocito/efectos de los fármacos , Integrinas/antagonistas & inhibidores , Leucocitos/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Selectinas/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Asma/metabolismo , Diseño de Fármacos , Humanos , Integrinas/química , Integrinas/metabolismo , Leucocitos/metabolismo , Modelos Moleculares , Conformación Proteica , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Selectinas/química , Selectinas/metabolismo , Resultado del Tratamiento
12.
Sci Rep ; 8(1): 3733, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29487319

RESUMEN

Inflammation drives the degradation of atherosclerotic plaque, yet there are no non-invasive techniques available for imaging overall inflammation in atherosclerotic plaques, especially in the coronary arteries. To address this, we have developed a clinically relevant system to image overall inflammatory cell burden in plaque. Here, we describe a targeted contrast agent (THI0567-targeted liposomal-Gd) that is suitable for magnetic resonance (MR) imaging and binds with high affinity and selectivity to the integrin α4ß1(very late antigen-4, VLA-4), a key integrin involved in recruiting inflammatory cells to atherosclerotic plaques. This liposomal contrast agent has a high T1 relaxivity (~2 × 105 mM-1s-1 on a particle basis) resulting in the ability to image liposomes at a clinically relevant MR field strength. We were able to visualize atherosclerotic plaques in various regions of the aorta in atherosclerosis-prone ApoE-/- mice on a 1 Tesla small animal MRI scanner. These enhanced signals corresponded to the accumulation of monocyte/macrophages in the subendothelial layer of atherosclerotic plaques in vivo, whereas non-targeted liposomal nanoparticles did not demonstrate comparable signal enhancement. An inflammatory cell-targeted method that has the specificity and sensitivity to measure the inflammatory burden of a plaque could be used to noninvasively identify patients at risk of an acute ischemic event.


Asunto(s)
Integrina alfa4beta1/química , Integrina alfa4beta1/metabolismo , Imagen por Resonancia Magnética , Placa Aterosclerótica/diagnóstico por imagen , Placa Aterosclerótica/metabolismo , Animales , Modelos Animales de Enfermedad , Integrina alfa4beta1/antagonistas & inhibidores , Ligandos , Liposomas , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Noqueados , Modelos Moleculares , Conformación Molecular , Placa Aterosclerótica/patología , Unión Proteica , Relación Estructura-Actividad
13.
Nat Commun ; 7: 11276, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27080438

RESUMEN

Promoting the paracrine effects of human mesenchymal stem cell (hMSC) therapy may contribute to improvements in patient outcomes. Here we develop an innovative strategy to enhance the paracrine effects of hMSCs. In a mouse hindlimb ischaemia model, we examine the effects of hMSCs in which a novel triple-catalytic enzyme is introduced to stably produce prostacyclin (PGI2-hMSCs). We show that PGI2-hMSCs facilitate perfusion recovery and enhance running capability as compared with control hMSCs or iloprost (a stable PGI2 analogue). Transplanted PGI2-hMSCs do not incorporate long term into host tissue, but rather they mediate host regeneration and muscle mass gain in a paracrine manner. Mechanistically, this involves long noncoding RNA H19 in promoting PGI2-hMSC-associated survival and proliferation of host progenitor cells under hypoxic conditions. Together, our data reveal the novel ability of PGI2-hMSCs to stimulate host regenerative processes and improve physical function by regulating long noncoding RNA in resident progenitor cells.


Asunto(s)
Epoprostenol/metabolismo , Miembro Posterior/irrigación sanguínea , Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , ARN Largo no Codificante/genética , Animales , Línea Celular , Células Cultivadas , Terapia Combinada , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/metabolismo , Células HEK293 , Humanos , Iloprost/farmacología , Ratones Endogámicos NOD , Ratones SCID , Actividad Motora , Carrera , Trasplante Heterólogo , Resultado del Tratamiento , Vasodilatadores/farmacología
14.
Biomaterials ; 98: 113-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27182813

RESUMEN

For a proangiogenic therapy to be successful, it must promote the development of mature vasculature for rapid reperfusion of ischemic tissue. Whole growth factor, stem cell, and gene therapies have yet to achieve the clinical success needed to become FDA-approved revascularization therapies. Herein, we characterize a biodegradable peptide-based scaffold engineered to mimic VEGF and self-assemble into a nanofibrous, thixotropic hydrogel, SLanc. We found that this injectable hydrogel was rapidly infiltrated by host cells and could be degraded while promoting the generation of neovessels. In mice with induced hind limb ischemia, this synthetic peptide scaffold promoted angiogenesis and ischemic tissue recovery, as shown by Doppler-quantified limb perfusion and a treadmill endurance test. Thirteen-month-old mice showed significant recovery within 7 days of treatment. Biodistribution studies in healthy mice showed that the hydrogel is safe when administered intramuscularly, subcutaneously, or intravenously. These preclinical studies help establish the efficacy of this treatment for peripheral artery disease due to diminished microvascular perfusion, a necessary step before clinical translation. This peptide-based approach eliminates the need for cell transplantation or viral gene transfection (therapies currently being assessed in clinical trials) and could be a more effective regenerative medicine approach to microvascular tissue engineering.


Asunto(s)
Miembro Posterior/irrigación sanguínea , Isquemia/tratamiento farmacológico , Nanofibras/uso terapéutico , Neovascularización Fisiológica , Péptidos/uso terapéutico , Animales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Isquemia/patología , Ratones Endogámicos C57BL , Músculos/patología , Neovascularización Fisiológica/efectos de los fármacos , Péptidos/farmacología , Reperfusión , Distribución Tisular/efectos de los fármacos
15.
Sci STKE ; 2002(124): pe14, 2002 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-11904458

RESUMEN

Transmembrane proteins, such as G protein-coupled receptors (GPCRs) and integrins, activate intracellular signaling pathways through interactions with downstream binding partners. Woodside discusses two examples in which GPCRs and integrins interact in a noncompeting manner with more than one partner. The specific GPCR described is the thrombin receptor, in experiments where G protein peptides selectively block signaling through a particular G protein that does not appear to inhibit coupling of the receptor to other G proteins. The second system described is the alphaIIbbeta3 integrin and its activation of the nonreceptor tyrosine kinase Syk. Syk appeared capable of interacting with both the integrin and intracellular domains of immune response receptors, because binding of Syk to the integrin was not inhibited by peptides based on the Syk binding site in immune response receptors. Thus, multiple, noncompeting binding partners add to the complexity of signal transduction outputs from a single receptor complex.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Integrinas/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas de Unión al GTP/fisiología , Guanosina Trifosfato/metabolismo , Guanosina Trifosfato/fisiología , Humanos , Integrinas/fisiología , Receptores de Superficie Celular/fisiología , Transducción de Señal/fisiología
16.
BMC Cell Biol ; 4: 2, 2003 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-12600279

RESUMEN

BACKGROUND: Rho family GTPase regulation of the actin cytoskeleton governs a variety of cell responses. In this report, we have analyzed the role of the GTPase Rho in maintenance of the T lymphocyte actin cytoskeleton. RESULTS: Inactivation of the GTPase Rho in the human T lymphocytic cell line HPB-ALL does not inhibit constitutively high adhesion to the integrin beta1 substrate fibronectin. It did however result in the aberrant extension of finger-like dendritic processes on the substrates VCAM-1, Fn, and mAb specific to beta1 integrins. Time-lapse video microscopy demonstrated that C3 induced extensions were primarily the result of an altered pseudopod elongation rather than retraction. Once the stellate pseudopodia extended, none retracted, and cells became completely immobile. Filipodial structures were absent and the dendritic-like processes in C3 treated cells were rich in filamentous actin. Immunolocalization of RhoA in untreated HPB-ALL cells spreading on fibronectin demonstrated a diffuse staining pattern within the pseudopodia. In C3 treated cells, clusters of RhoA were pronounced and localized within the altered extensions. CONCLUSIONS: GTPase Rho is actively involved in the regulation of T lymphocyte morphology and motility.


Asunto(s)
Linfocitos T/enzimología , Proteínas de Unión al GTP rho/metabolismo , ADP Ribosa Transferasas/metabolismo , ADP Ribosa Transferasas/farmacología , Actinas/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Toxinas Botulínicas/metabolismo , Toxinas Botulínicas/farmacología , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Extensiones de la Superficie Celular/efectos de los fármacos , Extensiones de la Superficie Celular/metabolismo , Fibronectinas/farmacología , Humanos , Microscopía por Video/métodos , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Células Tumorales Cultivadas , Proteína de Unión al GTP rhoA/metabolismo
17.
FEBS Lett ; 588(13): 2133-40, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24846140

RESUMEN

Lectin-like oxidized low-density lipoprotein receptor (LOX-1) is a scavenger receptor that binds oxidized low-density lipoprotein (OxLDL) and has a role in atherosclerosis development. The N-terminus intracellular region (cytoplasmic domain) of LOX-1 mediates receptor internalization and trafficking, potentially through intracellular protein interactions. Using affinity isolation, we identified 6 of the 8 components of the chaperonin-containing TCP-1 (CCT) complex bound to LOX-1 cytoplasmic domain, which we verified by coimmunoprecipitation and immunostaining in human umbilical vein endothelial cells. We found that the interaction between CCT and LOX-1 is direct and ATP-dependent and that OxLDL suppressed this interaction. Understanding the association between LOX-1 and the CCT complex may facilitate the design of novel therapies for cardiovascular disease.


Asunto(s)
Chaperonina con TCP-1/química , Chaperonina con TCP-1/metabolismo , Receptores Depuradores de Clase E/química , Receptores Depuradores de Clase E/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Chaperonina con TCP-1/genética , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ligandos , Lipoproteínas LDL/metabolismo , Datos de Secuencia Molecular , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína , ARN Interferente Pequeño/genética , Receptores Depuradores de Clase E/genética
18.
Immunol Cell Biol ; 87(4): 324-36, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19139760

RESUMEN

Lipid rafts are small laterally mobile microdomains that are highly enriched in lymphocyte signaling molecules. GM1 gangliosides are a common lipid raft component and have been shown to be important in many T-cell functions. The aggregation of specific GM1 lipid rafts can control many T-cell activation events, including their novel association with T-cell integrins. We found that clustering GM1 lipid rafts can regulate beta1 integrin function. This was apparent through increased resistance to shear flow-dependent detachment of T cells adherent to the alpha4beta1 and alpha5beta1 integrin ligand fibronectin (FN). Adhesion strengthening as a result of clustering GM1 enriched lipid rafts correlated with increased cellular rigidity and morphology through the localization of cortical F-actin, the resistance to shear-induced cell stretching, and an increase in the surface area and symmetry of the contact area between the cell surface and adhesive substrate. Furthermore, clustering GM1 lipid rafts could initiate integrin 'inside-out' signaling mechanisms. This was seen through increased integrin-cytoskeleton associations and enhanced soluble binding of FN and VCAM-1, suggesting the induction of high-affinity integrin conformations. The activation of these adhesion-strengthening characteristics appears to be specific for the aggregation of GM1 lipid rafts as the aggregation of the heterogeneous raft-associated molecule CD59 failed to activate these functions. These findings indicate a novel mechanism to signal to beta1 integrins and to activate adhesion-strengthening processes.


Asunto(s)
Fibronectinas/inmunología , Gangliósido G(M1)/inmunología , Integrinas/inmunología , Microdominios de Membrana/inmunología , Linfocitos T/inmunología , Adhesión Celular/inmunología , Línea Celular Tumoral , Quimiotaxis/inmunología , Citoesqueleto/inmunología , Humanos , Células Jurkat , Transducción de Señal , Molécula 1 de Adhesión Celular Vascular/inmunología
19.
Expert Opin Investig Drugs ; 15(10): 1235-55, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16989599

RESUMEN

Integrins are a family of heterodimeric cell surface receptors that mediate adhesion events crucial to cellular migration, proliferation and activation. Although critical to a normal immune response, integrins can also facilitate the progression of many inflammatory and autoimmune disorders. As such, they have attracted the attention of the pharmaceutical industry. Several humanised monoclonal antibodies directed against integrin targets have proven to be successful in clinical trials and have been approved for use in humans. This has not only resulted in effective therapies for patients, but also has provided important proof-of-concept studies for the development of small-molecule antagonists. This review focuses on those integrin subclasses that are being evaluated for their potential role in pulmonary, dermatological, gastrointestinal or rheumatic diseases. These include the alpha4 and beta2 integrins, as well as an emerging group of targets from the collagen-binding family of integrins. Interfering with integrin signalling pathways represents a future area of interest. The rationale for pursuing these targets, as well as the drugs presently under development, are discussed.


Asunto(s)
Mediadores de Inflamación/farmacología , Mediadores de Inflamación/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Integrinas/antagonistas & inhibidores , Integrinas/clasificación , Animales , Humanos , Inflamación/inmunología , Integrinas/inmunología , Integrinas/fisiología
20.
J Immunol ; 176(8): 5041-9, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16585601

RESUMEN

Cell adhesion mediated by the interaction between integrin alpha4beta1 and VCAM-1 is important in normal physiologic processes and in inflammatory and autoimmune disease. Numerous studies have mapped the alpha4beta1 binding sites in VCAM-1 that mediate cell adhesion; however, little is known about the regions in VCAM-1 important for regulating soluble binding. In the present study, we demonstrate that 6D VCAM-1 (an alternatively spliced isoform of VCAM-1 lacking Ig-like domain 4) binds alpha4beta1 with a higher relative affinity than does the full-length form of VCAM-1 containing 7 Ig-like extracellular domains (7D VCAM-1). In indirect binding assays, the EC50 of soluble 6D VCAM-1 binding to alpha4beta1 on Jurkat cells (in 1 mM MnCl2) was 2 x 10(-9) M, compared with 7D VCAM-1 at 11 x 10(-9) M. When used in solution to inhibit alpha4beta1 mediated cell adhesion, the IC50 of 6D VCAM-1 was 13 x 10(-9) M, compared with 7D VCAM-1 measured at 150 x 10(-9) M. Removal of Ig-like domains 4, 5, or 6, or simply substituting Asp328 in domain 4 of 7D VCAM-1 with alanine, caused increased binding of soluble 7D VCAM-1 to alpha4beta1. In contrast, cells adhered more avidly to 7D VCAM-1 under shear force, as it induced cell spreading at lower concentrations than did 6D VCAM-1. Finally, soluble 6D VCAM-1 acts as an agonist through alpha4beta1 by augmenting cell migration and inducing cell aggregation. These results indicate that the domain 4 of VCAM-1 plays a contrasting role when VCAM-1 is presented in solution or as a cell surface-expressed adhesive substrate.


Asunto(s)
Adhesión Celular/fisiología , Integrina alfa4beta1/metabolismo , Molécula 1 de Adhesión Celular Vascular/química , Molécula 1 de Adhesión Celular Vascular/fisiología , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/genética , Agregación Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , ADN/genética , Humanos , Técnicas In Vitro , Molécula 1 de Adhesión Intercelular/fisiología , Células Jurkat , Cinética , Mutagénesis , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Solubilidad , Molécula 1 de Adhesión Celular Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA