Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Transplant ; 24(2): 177-189, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37813189

RESUMEN

Present-day islet culture methods provide short-term maintenance of cell viability and function, limiting access to islet transplantation. Attempts to lengthen culture intervals remain unsuccessful. A new method was developed to permit the long-term culture of islets. Human islets were embedded in polysaccharide 3D-hydrogel in cell culture inserts or gas-permeable chambers with serum-free CMRL 1066 supplemented media for up to 8 weeks. The long-term cultured islets maintained better morphology, cell mass, and viability at 4 weeks than islets in conventional suspension culture. In fact, islets cultured in the 3D-hydrogel retained ß cell mass and function on par with freshly isolated islets in vitro and, when transplanted into diabetic mice, restored glucose balance similar to fresh islets. Using gas-permeable chambers, the 3D-hydrogel culture method was scaled up over 10-fold and maintained islet viability and function, although the cell mass recovery rate was 50%. Additional optimization of scale-up methods continues. If successful, this technology could afford flexibility and expand access to islet transplantation, especially single-donor islet-after-kidney transplantation.


Asunto(s)
Diabetes Mellitus Experimental , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Humanos , Ratones , Animales , Técnicas de Cultivo de Célula , Hidrogeles , Insulina , Supervivencia Celular
2.
Am J Physiol Endocrinol Metab ; 324(4): E347-E357, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36791324

RESUMEN

Thrombospondin-1 (TSP1) is a secreted protein minimally expressed in health but increased in disease and age. TSP1 binds to the cell membrane receptor CD47, which itself engages signal regulatory protein α (SIRPα), and the latter creates a checkpoint for immune activation. Individuals with cancer administered checkpoint-blocking molecules developed insulin-dependent diabetes. Relevant to this, CD47 blocking antibodies and SIRPα fusion proteins are in clinical trials. We characterized the molecular signature of TSP1, CD47, and SIRPα in human islets and pancreata. Fresh islets and pancreatic tissue from nondiabetic individuals were obtained. The expression of THBS1, CD47, and SIRPA was determined using single-cell mRNA sequencing, immunofluorescence microscopy, Western blot, and flow cytometry. Islets were exposed to diabetes-affiliated inflammatory cytokines and changes in protein expression were determined. CD47 mRNA was expressed in all islet cell types. THBS1 mRNA was restricted primarily to endothelial and mesenchymal cells, whereas SIRPA mRNA was found mostly in macrophages. Immunofluorescence staining showed CD47 protein expressed by ß cells and present in the exocrine pancreas. TSP1 and SIRPα proteins were not seen in islets or the exocrine pancreas. Western blot and flow cytometry confirmed immunofluorescent expression patterns. Importantly, human islets produced substantial quantities of secreted TSP1. Human pancreatic exocrine and endocrine tissue expressed CD47, whereas fresh islets displayed cell surface CD47 and secreted TSP1 at baseline and in inflammation. These findings suggest unexpected effects on islets from agents that intersect TSP1-CD47-SIRPα.NEW & NOTEWORTHY CD47 is a cell surface receptor with two primary ligands, soluble thrombospondin-1 (TSP1) and cell surface signal regulatory protein alpha (SIRPα). Both interactions provide checkpoints for immune cell activity. We determined that fresh human islets display CD47 and secrete TSP1. However, human islet endocrine cells lack SIRPα. These gene signatures are likely important given the increasing use of CD47 and SIRPα blocking molecules in individuals with cancer.


Asunto(s)
Antígeno CD47 , Neoplasias , Humanos , Antígeno CD47/genética , Antígeno CD47/metabolismo , Macrófagos/metabolismo , Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Trombospondinas/metabolismo , Trombospondinas/uso terapéutico , Trombospondina 1/genética , Trombospondina 1/metabolismo
3.
Cancer Immunol Immunother ; 72(9): 2879-2888, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37217603

RESUMEN

The use of treatments, such as programmed death protein 1 (PD1) or cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) antibodies, that loosen the natural checks upon immune cell activity to enhance cancer killing have shifted clinical practice and outcomes for the better. Accordingly, the number of antibodies and engineered proteins that interact with the ligand-receptor components of immune checkpoints continue to increase along with their use. It is tempting to view these molecular pathways simply from an immune inhibitory perspective. But this should be resisted. Checkpoint molecules can have other cardinal functions relevant to the development and use of blocking moieties. Cell receptor CD47 is an example of this. CD47 is found on the surface of all human cells. Within the checkpoint paradigm, non-immune cell CD47 signals through immune cell surface signal regulatory protein alpha (SIRPα) to limit the activity of the latter, the so-called trans signal. Even so, CD47 interacts with other cell surface and soluble molecules to regulate biogas and redox signaling, mitochondria and metabolism, self-renewal factors and multipotency, and blood flow. Further, the pedigree of checkpoint CD47 is more intricate than supposed. High-affinity interaction with soluble thrombospondin-1 (TSP1) and low-affinity interaction with same-cell SIRPα, the so-called cis signal, and non-SIRPα ectodomains on the cell membrane suggests that multiple immune checkpoints converge at and through CD47. Appreciation of this may provide latitude for pathway-specific targeting and intelligent therapeutic effect.


Asunto(s)
Antígeno CD47 , Neoplasias , Humanos , Antígenos de Diferenciación/farmacología , Receptores Inmunológicos/metabolismo , Neoplasias/terapia , Anticuerpos/farmacología , Proteínas Portadoras , Fagocitosis
4.
Am J Physiol Cell Physiol ; 321(2): C201-C213, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34106789

RESUMEN

Thrombospondin-1 (TSP1) is the prototypical member of a family of secreted proteins that modulate cell behavior by engaging with molecules in the extracellular matrix and with receptors on the cell surface. CD47 is widely displayed on many, if not all, cell types and is a high-affinity TSP1 receptor. CD47 is a marker of self that limits innate immune cell activities, a feature recently exploited to enhance cancer immunotherapy. Another major role for CD47 in health and disease is to mediate TSP1 signaling. TSP1 acting through CD47 contributes to mitochondrial, metabolic, and endocrine dysfunction. Studies in animal models found that elevated TSP1 expression, acting in part through CD47, causes mitochondrial and metabolic dysfunction. Clinical studies established that abnormal TSP1 expression positively correlates with obesity, fatty liver disease, and diabetes. The unabated increase in these conditions worldwide and the availability of CD47 targeting drugs justify a closer look into how TSP1 and CD47 disrupt metabolic balance and the potential for therapeutic intervention.


Asunto(s)
Antígeno CD47/metabolismo , Diabetes Mellitus/metabolismo , Células Endoteliales/metabolismo , Mitocondrias/metabolismo , Trombospondina 1/metabolismo , Animales , Membrana Celular/metabolismo , Humanos
5.
Am J Physiol Cell Physiol ; 319(1): C45-C63, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32374675

RESUMEN

Numerous age-dependent alterations at the molecular, cellular, tissue and organ systems levels underlie the pathophysiology of aging. Herein, the focus is upon the secreted protein thrombospondin-1 (TSP1) as a promoter of aging and age-related diseases. TSP1 has several physiological functions in youth, including promoting neural synapse formation, mediating responses to ischemic and genotoxic stress, minimizing hemorrhage, limiting angiogenesis, and supporting wound healing. These acute functions of TSP1 generally require only transient expression of the protein. However, accumulating basic and clinical data reinforce the view that chronic diseases of aging are associated with accumulation of TSP1 in the extracellular matrix, which is a significant maladaptive contributor to the aging process. Identification of the relevant cell types that chronically produce and respond to TSP1 and the molecular mechanisms that mediate the resulting maladaptive responses could direct the development of therapeutic agents to delay or revert age-associated maladies.


Asunto(s)
Envejecimiento/genética , Envejecimiento/metabolismo , Trombospondina 1/biosíntesis , Trombospondina 1/genética , Animales , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/terapia , Daño del ADN/fisiología , Humanos , Enfermedades Musculoesqueléticas/genética , Enfermedades Musculoesqueléticas/metabolismo , Enfermedades Musculoesqueléticas/terapia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Transducción de Señal/fisiología , Trombospondina 1/antagonistas & inhibidores , Cicatrización de Heridas/fisiología
6.
Am J Physiol Lung Cell Mol Physiol ; 316(6): L1150-L1164, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30892078

RESUMEN

Pulmonary hypertension (PH) is a leading cause of death in sickle cell disease (SCD) patients. Hemolysis and oxidative stress contribute to SCD-associated PH. We have reported that the protein thrombospondin-1 (TSP1) is elevated in the plasma of patients with SCD and, by interacting with its receptor CD47, limits vasodilation of distal pulmonary arteries ex vivo. We hypothesized that the TSP1-CD47 interaction may promote PH in SCD. We found that TSP1 and CD47 are upregulated in the lungs of Berkeley (BERK) sickling (Sickle) mice and patients with SCD-associated PH. We then generated chimeric animals by transplanting BERK bone marrow into C57BL/6J (n = 24) and CD47 knockout (CD47KO, n = 27) mice. Right ventricular (RV) pressure was lower in fully engrafted Sickle-to-CD47KO than Sickle-to-C57BL/6J chimeras, as shown by the reduced maximum RV pressure (P = 0.013) and mean pulmonary artery pressure (P = 0.020). The afterload of the sickle-to-CD47KO chimeras was also lower, as shown by the diminished pulmonary vascular resistance (P = 0.024) and RV effective arterial elastance (P = 0.052). On myography, aortic segments from Sickle-to-CD47KO chimeras showed improved relaxation to acetylcholine. We hypothesized that, in SCD, TSP1-CD47 signaling promotes PH, in part, by increasing reactive oxygen species (ROS) generation. In human pulmonary artery endothelial cells, treatment with TSP1 stimulated ROS generation, which was abrogated by CD47 blockade. Explanted lungs of CD47KO chimeras had less vascular congestion and a smaller oxidative footprint. Our results show that genetic absence of CD47 ameliorates SCD-associated PH, which may be due to decreased ROS levels. Modulation of TSP1-CD47 may provide a new molecular approach to the treatment of SCD-associated PH.


Asunto(s)
Anemia de Células Falciformes/patología , Antígeno CD47/metabolismo , Hipertensión Pulmonar/patología , Arteria Pulmonar/patología , Trombospondina 1/metabolismo , Anemia de Células Falciformes/genética , Animales , Antígeno CD47/antagonistas & inhibidores , Antígeno CD47/genética , Células Cultivadas , Células Endoteliales/patología , Humanos , Hipertensión Pulmonar/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Arteria Pulmonar/citología , Especies Reactivas de Oxígeno/metabolismo , Función Ventricular Derecha/fisiología
7.
Pediatr Nephrol ; 34(12): 2479-2494, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30392076

RESUMEN

Ischemia reperfusion (IR) injury is a process defined by the temporary loss of blood flow and tissue perfusion followed later by restoration of the same. Brief periods of IR can be tolerated with little permanent deficit, but sensitivity varies for different target cells and tissues. Ischemia reperfusion injuries have multiple causes including peripheral vascular disease and surgical interventions that disrupt soft tissue and organ perfusion as occurs in general and reconstructive surgery. Ischemia reperfusion injury is especially prominent in organ transplantation where substantial effort has been focused on protecting the transplanted organ from the consequences of IR. A number of factors mediate IR injury including the production of reactive oxygen species and inflammatory cell infiltration and activation. In the kidney, IR injury is a major cause of acute injury and secondary loss of renal function. Transplant-initiated renal IR is also a stimulus for innate and adaptive immune-mediated transplant dysfunction. The cell surface molecule CD47 negatively modulates cell and tissue responses to stress through limitation of specific homeostatic pathways and initiation of cell death pathways. Herein, a summary of the maladaptive activities of renal CD47 will be considered as well as the possible therapeutic benefit of interfering with CD47 to limit renal IR.


Asunto(s)
Antígeno CD47/metabolismo , Daño por Reperfusión/fisiopatología , Factores de Edad , Animales , Antígenos de Diferenciación/metabolismo , Humanos , Riñón/irrigación sanguínea , Enfermedades Renales/etiología , Receptores Inmunológicos/metabolismo , Daño por Reperfusión/terapia , Transducción de Señal
8.
J Cell Mol Med ; 22(4): 2086-2097, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29441713

RESUMEN

Thrombospondin-1 (TSP-1), a matricellular protein and one of the first endogenous anti-angiogenic molecules identified, has long been considered a potent modulator of human diseases. While the therapeutic effect of TSP-1 to suppress cancer was investigated in both research and clinical settings, the mechanisms of how TSP-1 is regulated in cancer remain elusive, and the scientific answers to the question of whether TSP-1 expressions can be utilized as diagnostic or prognostic marker for patients with cancer are largely inconsistent. Moreover, TSP-1 plays crucial functions in angiogenesis, inflammation and tissue remodelling, which are essential biological processes in the progression of many cardiovascular diseases, and therefore, its dysregulated expressions in such conditions may have therapeutic significance. Herein, we critically analysed the literature pertaining to TSP-1 expression in circulating blood and pathological tissues in various types of cancer as well as cardiovascular and inflammation-related diseases in humans. We compare the secretion rates of TSP-1 by different cancer and non-cancer cells and discuss the potential connection between the expression changes of TSP-1 and vascular endothelial growth factor (VEGF) observed in patients with cancer. Moreover, the pattern and emerging significance of TSP-1 profiles in cardiovascular disease, such as peripheral arterial disease, diabetes and other related non-cancer disorders, are highlighted. The analysis of published TSP-1 data presented in this review may have implications for the future exploration of novel TSP-1-based treatment strategies for cancer and cardiovascular-related diseases.


Asunto(s)
Enfermedad , Regulación de la Expresión Génica , Trombospondina 1/metabolismo , Humanos , Modelos Biológicos , Células del Estroma/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo
9.
PLoS Comput Biol ; 13(1): e1005272, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28045898

RESUMEN

Hypoxia is an important physiological stress signal that drives angiogenesis, the formation of new blood vessels. Besides an increase in the production of pro-angiogenic signals such as vascular endothelial growth factor (VEGF), hypoxia also stimulates the production of anti-angiogenic signals. Thrombospondin-1 (TSP-1) is one of the anti-angiogenic factors whose synthesis is driven by hypoxia. Cellular synthesis of TSP-1 is tightly regulated by different intermediate biomolecules including proteins that interact with hypoxia-inducible factors (HIFs), transcription factors that are activated by receptor and intracellular signaling, and microRNAs which are small non-coding RNA molecules that function in post-transcriptional modification of gene expression. Here we present a computational model that describes the mechanistic interactions between intracellular biomolecules and cooperation between signaling pathways that together make up the complex network of TSP-1 regulation both at the transcriptional and post-transcriptional level. Assisted by the model, we conduct in silico experiments to compare the efficacy of different therapeutic strategies designed to modulate TSP-1 synthesis in conditions that simulate tumor and peripheral arterial disease microenvironment. We conclude that TSP-1 production in endothelial cells depends on not only the availability of certain growth factors but also the fine-tuned signaling cascades that are initiated by hypoxia.


Asunto(s)
Regulación de la Expresión Génica/genética , Modelos Biológicos , Trombospondina 1/genética , Trombospondina 1/metabolismo , Hipoxia de la Célula/fisiología , Biología Computacional , Simulación por Computador , Humanos , Oxígeno/metabolismo , Proteínas Smad/metabolismo , Trombospondina 1/análisis , Factores de Transcripción
10.
Am J Physiol Cell Physiol ; 312(3): C254-C262, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27974299

RESUMEN

Nitric oxide (NO) is one of the critical components of the vasculature, regulating key signaling pathways in health. In macrovessels, NO functions to suppress cell inflammation as well as adhesion. In this way, it inhibits thrombosis and promotes blood flow. It also functions to limit vessel constriction and vessel wall remodeling. In microvessels and particularly capillaries, NO, along with growth factors, is important in promoting new vessel formation, a process termed angiogenesis. With age and cardiovascular disease, animal and human studies confirm that NO is dysregulated at multiple levels including decreased production, decreased tissue half-life, and decreased potency. NO has also been implicated in diseases that are related to neurotransmission and cancer although it is likely that these processes involve NO at higher concentrations and from nonvascular cell sources. Conversely, NO and drugs that directly or indirectly increase NO signaling have found clinical applications in both age-related diseases and in younger individuals. This focused review considers recently reported advances being made in the field of NO signaling regulation at several levels including enzymatic production, receptor function, interacting partners, localization of signaling, matrix-cellular and cell-to-cell cross talk, as well as the possible impact these newly described mechanisms have on health and disease.


Asunto(s)
Vasos Sanguíneos/fisiopatología , Enfermedades Cardiovasculares/fisiopatología , Modelos Cardiovasculares , Neovascularización Fisiológica/fisiología , Óxido Nítrico/metabolismo , Animales , Velocidad del Flujo Sanguíneo , Humanos
12.
J Biol Chem ; 290(52): 31113-25, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26534964

RESUMEN

Signal regulatory protein α (SIRPα) is a membrane glycoprotein immunoreceptor abundant in cells of monocyte lineage. SIRPα ligation by a broadly expressed transmembrane protein, CD47, results in phosphorylation of the cytoplasmic immunoreceptor tyrosine-based inhibitory motifs, resulting in the inhibition of NF-κB signaling in macrophages. Here we observed that proteolysis of SIRPα during inflammation is regulated by a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10), resulting in the generation of a membrane-associated cleavage fragment in both THP-1 monocytes and human lung epithelia. We mapped a charge-dependent putative cleavage site near the membrane-proximal domain necessary for ADAM10-mediated cleavage. In addition, a secondary proteolytic cleavage within the membrane-associated SIRPα fragment by γ-secretase was identified. Ectopic expression of a SIRPα mutant plasmid encoding a proteolytically resistant form in HeLa cells inhibited activation of the NF-κB pathway and suppressed STAT1 phosphorylation in response to TNFα to a greater extent than expression of wild-type SIRPα. Conversely, overexpression of plasmids encoding the proteolytically cleaved SIRPα fragments in cells resulted in enhanced STAT-1 and NF-κB pathway activation. Thus, the data suggest that combinatorial actions of ADAM10 and γ-secretase on SIRPα cleavage promote inflammatory signaling.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Antígenos de Diferenciación/metabolismo , Proteínas de la Membrana/metabolismo , Proteolisis , Receptores Inmunológicos/metabolismo , Transducción de Señal , Proteínas ADAM/genética , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/genética , Antígenos de Diferenciación/genética , Células HeLa , Humanos , Inflamación/genética , Inflamación/metabolismo , Proteínas de la Membrana/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Receptores Inmunológicos/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo
13.
Kidney Int ; 90(2): 334-347, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27259369

RESUMEN

Defects in renal tubular epithelial cell repair contribute to renal ischemia reperfusion injury, cause acute kidney damage, and promote chronic renal disease. The matricellular protein thrombospondin-1 and its receptor CD47 are involved in experimental renal ischemia reperfusion injury, although the role of this interaction in renal recovery is unknown. We found upregulation of self-renewal genes (transcription factors Oct4, Sox2, Klf4 and cMyc) in the kidney of CD47(-/-) mice after ischemia reperfusion injury. Wild-type animals had minimal self-renewal gene expression, both before and after injury. Suggestive of cell autonomy, CD47(-/-) renal tubular epithelial cells were found to increase expression of the self-renewal genes. This correlated with enhanced proliferative capacity compared with cells from wild-type mice. Exogenous thrombospondin-1 inhibited self-renewal gene expression in renal tubular epithelial cells from wild-type but not CD47(-/-) mice, and this was associated with decreased proliferation. Treatment of renal tubular epithelial cells with a CD47 blocking antibody or CD47-targeting small interfering RNA increased expression of some self-renewal transcription factors and promoted cell proliferation. In a syngeneic kidney transplant model, treatment with a CD47 blocking antibody increased self-renewal transcription factor expression, decreased tissue damage, and improved renal function compared with that in control mice. Thus, thrombospondin-1 via CD47 inhibits renal tubular epithelial cell recovery after ischemia reperfusion injury through inhibition of proliferation/self-renewal.


Asunto(s)
Antígeno CD47/metabolismo , Células Epiteliales/fisiología , Túbulos Renales/fisiología , Regeneración , Daño por Reperfusión/complicaciones , Trombospondina 1/metabolismo , Animales , Antígeno CD47/genética , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Trasplante de Riñón , Túbulos Renales/citología , Túbulos Renales/patología , Factor 4 Similar a Kruppel , Masculino , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal , Factores de Transcripción/metabolismo , Regulación hacia Arriba
14.
Herz ; 41(1): 57-62, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26135468

RESUMEN

OBJECTIVE: Atrial fibrillation (AF) is a condition where platelet hyperaggregability is commonly present. We examined potential physiological bases for platelet hyperaggregability in a cohort of patients with acute and chronic AF. In particular, we sought to identify the impact of inflammation [myeloperoxidase (MPO) and C-reactive protein (CRP)] and impaired nitric oxide (NO) signaling. METHODS: Clinical and biochemical determinants of adenosine diphosphate (ADP)-induced platelet aggregation were sought in patients (n = 106) hospitalized with AF via univariate and multivariate analysis. RESULTS: Hyper-responsiveness of platelets to ADP was directly (r = 0.254, p < 0.01) correlated with plasma concentrations of thrombospondin-1 (TSP-1), a matricellular protein that impairs NO responses and contributes to development of oxidative stress. In turn, plasma TSP-1 concentrations were directly correlated with MPO concentrations (r = 0.221, p < 0.05), while MPO concentrations correlated with those of asymmetric dimethylarginine (ADMA, r = 0.220, p < 0.05), and its structural isomer symmetric dimethylarginine (SDMA, r = 0.192, p = 0.05). Multivariate analysis identified TSP-1 (ß = 0.276, p < 0.05) concentrations, as well as female sex (ß = 0.199, p < 0.05), as direct correlates of platelet aggregability, and SDMA concentrations (ß = - 0.292, p < 0.05) as an inverse correlate. CONCLUSION: We conclude that platelet hyperaggregability, where present in the context of AF, may be engendered by impaired availability of NO, as well as via MPO-related inflammatory activation.


Asunto(s)
Fibrilación Atrial/inmunología , Miocarditis/inmunología , Óxido Nítrico/inmunología , Agregación Plaquetaria/inmunología , Especies Reactivas de Oxígeno/inmunología , Anciano , Anciano de 80 o más Años , Fibrilación Atrial/patología , Femenino , Humanos , Masculino , Miocarditis/patología , Óxido Nítrico/sangre , Peroxidasa/sangre , Peroxidasa/inmunología , Especies Reactivas de Oxígeno/sangre
16.
J Biol Chem ; 289(9): 5904-13, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24403062

RESUMEN

Protein-tyrosine phosphatase 4A3 (PTP4A3) is highly expressed in multiple human cancers and is hypothesized to have a critical, albeit poorly defined, role in the formation of experimental tumors in mice. PTP4A3 is broadly expressed in many tissues so the cellular basis of its etiological contributions to carcinogenesis may involve both tumor and stromal cells. In particular, PTP4A3 is expressed in the tumor vasculature and has been proposed to be a direct target of vascular endothelial growth factor (VEGF) signaling in endothelial cells. We now provide the first in vivo experimental evidence that PTP4A3 participates in VEGF signaling and contributes to the process of pathological angiogenesis. Colon tumor tissue isolated from Ptp4a3-null mice revealed reduced tumor microvessel density compared with wild type controls. Additionally, vascular cells derived from Ptp4a3-null tissues exhibited decreased invasiveness in an ex vivo wound healing assay. When primary endothelial cells were isolated and cultured in vitro, Ptp4a3-null cells displayed greatly reduced migration compared with wild type cells. Exposure to VEGF led to an increase in Src phosphorylation in wild type endothelial cells, a response that was completely ablated in Ptp4a3-null cells. In loss-of-function studies, reduced VEGF-mediated migration was also observed when human endothelial cells were treated with a small molecule inhibitor of PTP4A3. VEGF-mediated in vivo vascular permeability was significantly attenuated in PTP4A3-deficient mice. These findings strongly support a role for PTP4A3 as an important contributor to endothelial cell function and as a multimodal target for cancer therapy and mitigating VEGF-regulated angiogenesis.


Asunto(s)
Movimiento Celular , Neoplasias del Colon/metabolismo , Células Endoteliales/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Células Endoteliales/patología , Humanos , Proteínas Inmediatas-Precoces/genética , Ratones , Ratones Mutantes , Proteínas de Neoplasias/genética , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Proteínas Tirosina Fosfatasas/genética , Factor A de Crecimiento Endotelial Vascular/genética
17.
J Am Soc Nephrol ; 25(6): 1171-86, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24511121

RESUMEN

Ischemia reperfusion injury (IRI) causes tissue and organ injury, in part, through alterations in tissue blood flow and the production of reactive oxygen species. The cell surface receptor signal-regulatory protein-α (SIRP-α) is expressed on inflammatory cells and suppresses phagocytosis, but the function of SIRP-α in IRI has not been determined. We reported previously that the matricellular protein thrombospondin-1 is upregulated in IRI. Here, we report a novel interaction between thrombospondin-1 and SIRP-α on nonphagocytic cells. In cell-free experiments, thrombospondin-1 bound SIRP-α. In vascular smooth muscle cells and renal tubular epithelial cells, treatment with thrombospondin-1 led to phosphorylation of SIRP-α and downstream activation of Src homology domain 2-containing phosphatase-1. Thrombospondin-1 also stimulated phosphorylation of p47(phox) (an organizer subunit for nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1/2) and increased production of superoxide, both of which were abrogated by knockdown or antibody blockade of SIRP-α. In rodent aortic rings, treatment with thrombospondin-1 increased the production of superoxide and inhibited nitric oxide-mediated vasodilation in a SIRP-α-dependent manner. Renal IRI upregulated the thrombospondin-1-SIRP-α signaling axis and was associated with increased superoxide production and cell death. A SIRP-α antibody that blocks thrombospondin-1 activation of SIRP-α mitigated the effects of renal IRI, increasing blood flow, suppressing production of reactive oxygen species, and preserving cellular architecture. A role for CD47 in SIRP-α activation in these pathways is also described. Overall, these results suggest that thrombospondin-1 binding to SIRP-α on nonphagocytic cells activates NADPH oxidase, limits vasodilation, and promotes renal IRI.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Enfermedades Renales/metabolismo , Receptores Inmunológicos/metabolismo , Daño por Reperfusión/metabolismo , Trombospondina 1/metabolismo , Animales , Antígeno CD47/metabolismo , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología
19.
Clin Transl Med ; 14(2): e1584, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38362603

RESUMEN

Cluster of differentiation 47 (CD47) occupies the outer membrane of human cells, where it binds to soluble and cell surface receptors on the same and other cells, sculpting their topography and resulting in a pleiotropic receptor-multiligand interaction network. It is a focus of drug development to temper and accentuate CD47-driven immune cell liaisons, although consideration of on-target CD47 effects remain neglected. And yet, a late clinical trial of a CD47-blocking antibody was discontinued, existent trials were restrained, and development of CD47-targeting agents halted by some pharmaceutical companies. At this point, if CD47 can be exploited for clinical advantage remains to be determined. Herein an airing is made of the seemingly conflicting actions of CD47 that reflect its position as a junction connecting receptors and signalling pathways that impact numerous human cell types. Prospects of CD47 boosting and blocking are considered along with potential therapeutic implications for autoimmune diseases and cancer.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Fagocitosis , Antígeno CD47/metabolismo , Inmunoterapia/métodos , Neoplasias/metabolismo , Antineoplásicos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA