Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 131(1): 59-73, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35574842

RESUMEN

BACKGROUND: Chronic renal inflammation has been widely recognized as a major promoter of several forms of high blood pressure including salt-sensitive hypertension. In diabetes, IL (interleukin)-6 induces salt sensitivity through a dysregulation of the epithelial sodium channel. However, the origin of this inflammatory process and the molecular events that culminates with an abnormal regulation of epithelial sodium channel and salt sensitivity in diabetes are largely unknown. METHODS: Both in vitro and in vivo approaches were used to investigate the molecular and cellular contributors to the renal inflammation associated with diabetic kidney disease and how these inflammatory components interact to develop salt sensitivity in db/db mice. RESULTS: Thirty-four-week-old db/db mice display significantly higher levels of IL-1ß in renal tubules compared with nondiabetic db/+ mice. Specific suppression of IL-1ß in renal tubules prevented salt sensitivity in db/db mice. A primary culture of renal tubular epithelial cells from wild-type mice releases significant levels of IL-1ß when exposed to a high glucose environment. Coculture of tubular epithelial cells and bone marrow-derived macrophages revealed that tubular epithelial cell-derived IL-1ß promotes the polarization of macrophages towards a proinflammatory phenotype resulting in IL-6 secretion. To evaluate whether macrophages are the cellular target of IL-1ß in vivo, diabetic db/db mice were transplanted with the bone marrow of IL-1R1 (IL-1 receptor type 1) knockout mice. db/db mice harboring an IL-1 receptor type 1 knockout bone marrow remained salt resistant, display lower renal inflammation and lower expression and activity of epithelial sodium channel compared with db/db transplanted with a wild-type bone marrow. CONCLUSIONS: Renal tubular epithelial cell-derived IL-1ß polarizes renal macrophages towards a proinflammatory phenotype that promotes salt sensitivity through the accumulation of renal IL-6. When tubular IL-1ß synthesis is suppressed or in db/db mice in which immune cells lack the IL-1R1, macrophage polarization is blunted resulting in no salt-sensitive hypertension.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Hipertensión , Nefritis , Animales , Diabetes Mellitus/metabolismo , Nefropatías Diabéticas/genética , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Inflamación/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Nefritis/metabolismo , Receptores de Interleucina-1/metabolismo , Cloruro de Sodio Dietético/toxicidad
2.
Nat Immunol ; 12(11): 1078-85, 2011 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-21964607

RESUMEN

The surface presentation of peptides by major histocompatibility complex (MHC) class I molecules is critical to CD8(+) T cell-mediated adaptive immune responses. Aminopeptidases have been linked to the editing of peptides for MHC class I loading, but carboxy-terminal editing is thought to be due to proteasome cleavage. By analysis of wild-type mice and mice genetically deficient in or overexpressing the dipeptidase angiotensin-converting enzyme (ACE), we have now identified ACE as having a physiological role in the processing of peptides for MHC class I. ACE edited the carboxyl terminus of proteasome-produced MHC class I peptides. The lack of ACE exposed new antigens but also abrogated some self antigens. ACE had substantial effects on the surface expression of MHC class I in a haplotype-dependent manner. We propose a revised model of peptide processing for MHC class I by introducing carboxypeptidase activity into the process.


Asunto(s)
Selección Clonal Mediada por Antígenos , Antígenos de Histocompatibilidad Clase I/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Linfocitos T/metabolismo , Inmunidad Adaptativa , Animales , Presentación de Antígeno/genética , Autoantígenos/inmunología , Autoantígenos/metabolismo , Células Cultivadas , Selección Clonal Mediada por Antígenos/genética , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/inmunología , Unión Proteica/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/genética , Linfocitos T/citología , Linfocitos T/inmunología , Transgenes/genética
3.
Transfusion ; 63(6): 1241-1245, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37096845

RESUMEN

BACKGROUND: There is a small but growing number of thrombotic thrombocytopenic purpura (TTP) cases attributed to immune checkpoint inhibitor therapy, with nivolumab and ipilimumab therapy being the most frequently described in the literature. STUDY DESIGN AND METHODS: This report evaluates the course of a patient with a history of metastatic adenocarcinoma of the lung who developed TTP following treatment with the PD-1 inhibitor Pembrolizumab. The patient was treated with six sessions of therapeutic plasma exchange and appeared to be in remission. Exacerbation occurred 4 days later, and seven more sessions of plasma exchange were performed along with four total doses of Rituximab, and a steroid taper with monitoring of platelet counts and ADAMTS13 activity. RESULTS: His platelet count recovered to a peak of 318,000 UL with an ADAMTS13 activity of 77% at the time of discharge. The patient has been following up regularly for outpatient testing with no TTP relapse as of the completion of this report. DISCUSSION: This is one of a few cases of Pembrolizumab-associated TTP reported in the literature with successful complete remission following treatment. Plasma exchange in this setting may be an especially beneficial therapeutic intervention because of the removal of both the anti-ADAMTS13 antibody as well as the immune system upregulating anti-PDL1 monoclonal antibody with replacement of ADAMTS13 from donor plasma. Longer duration of plasma exchange and monitoring for normalization of ADAMTS13 levels in addition to platelet count before cessation of treatment may improve durable remission rates in this entity.


Asunto(s)
Púrpura Trombocitopénica Trombótica , Humanos , Púrpura Trombocitopénica Trombótica/inducido químicamente , Púrpura Trombocitopénica Trombótica/tratamiento farmacológico , Proteínas ADAM/uso terapéutico , Recurrencia Local de Neoplasia/terapia , Rituximab/uso terapéutico , Intercambio Plasmático/efectos adversos , Proteína ADAMTS13
4.
J Am Soc Nephrol ; 32(5): 1131-1149, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33731332

RESUMEN

BACKGROUND: Hypertension is considered a major risk factor for the progression of diabetic kidney disease. Type 2 diabetes is associated with increased renal sodium reabsorption and salt-sensitive hypertension. Clinical studies show that men have higher risk than premenopausal women for the development of diabetic kidney disease. However, the renal mechanisms that predispose to salt sensitivity during diabetes and whether sexual dimorphism is associated with these mechanisms remains unknown. METHODS: Female and male db/db mice exposed to a high-salt diet were used to analyze the progression of diabetic kidney disease and the development of hypertension. RESULTS: Male, 34-week-old, db/db mice display hypertension when exposed to a 4-week high-salt treatment, whereas equivalently treated female db/db mice remain normotensive. Salt-sensitive hypertension in male mice was associated with no suppression of the epithelial sodium channel (ENaC) in response to a high-salt diet, despite downregulation of several components of the intrarenal renin-angiotensin system. Male db/db mice show higher levels of proinflammatory cytokines and more immune-cell infiltration in the kidney than do female db/db mice. Blocking inflammation, with either mycophenolate mofetil or by reducing IL-6 levels with a neutralizing anti-IL-6 antibody, prevented the development of salt sensitivity in male db/db mice. CONCLUSIONS: The inflammatory response observed in male, but not in female, db/db mice induces salt-sensitive hypertension by impairing ENaC downregulation in response to high salt. These data provide a mechanistic explanation for the sexual dimorphism associated with the development of diabetic kidney disease and salt sensitivity.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Canales Epiteliales de Sodio/fisiología , Hipertensión/etiología , Cloruro de Sodio Dietético/administración & dosificación , Animales , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Femenino , Hipertensión/metabolismo , Hipertensión/patología , Inflamación , Masculino , Ratones , Factores Sexuales , Cloruro de Sodio Dietético/efectos adversos
5.
J Biol Chem ; 295(5): 1369-1384, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31871049

RESUMEN

Angiotensin-converting enzyme (ACE) affects blood pressure. In addition, ACE overexpression in myeloid cells increases their immune function. Using MS and chemical analysis, we identified marked changes of intermediate metabolites in ACE-overexpressing macrophages and neutrophils, with increased cellular ATP (1.7-3.0-fold) and Krebs cycle intermediates, including citrate, isocitrate, succinate, and malate (1.4-3.9-fold). Increased ATP is due to ACE C-domain catalytic activity; it is reversed by an ACE inhibitor but not by an angiotensin II AT1 receptor antagonist. In contrast, macrophages from ACE knockout (null) mice averaged only 28% of the ATP levels found in WT mice. ACE overexpression does not change cell or mitochondrial size or number. However, expression levels of the electron transport chain proteins NDUFB8 (complex I), ATP5A, and ATP5ß (complex V) are significantly increased in macrophages and neutrophils, and COX1 and COX2 (complex IV) are increased in macrophages overexpressing ACE. Macrophages overexpressing ACE have increased mitochondrial membrane potential (24% higher), ATP production rates (29% higher), and maximal respiratory rates (37% higher) compared with WT cells. Increased cellular ATP underpins increased myeloid cell superoxide production and phagocytosis associated with increased ACE expression. Myeloid cells overexpressing ACE indicate the existence of a novel pathway in which myeloid cell function can be enhanced, with a key feature being increased cellular ATP.


Asunto(s)
Adenosina Trifosfato/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Células Mieloides/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Animales , Ciclo del Ácido Cítrico , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Neutrófilos/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Peptidil-Dipeptidasa A/genética , Regulación hacia Arriba
6.
Curr Hypertens Rep ; 22(1): 4, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31916032

RESUMEN

PURPOSE OF REVIEW: To review recent studies exploring how myeloid cell overexpression of angiotensin-converting enzyme (ACE) affects the immune response and to formulate an approach for considering the effectiveness of inflammation in cardiovascular disease RECENT FINDINGS: While it is widely appreciated that the renin-angiotensin system affects aspects of inflammation through the action of angiotensin II, new studies reveal a previously unknown role of ACE in myeloid cell biology. This was apparent from analysis of two mouse lines genetically modified to overexpress ACE in monocytes/macrophages or neutrophils. Cells overexpressing ACE demonstrated an increased immune response. For example, mice with increased macrophage ACE expression have increased resistance to melanoma, methicillin-resistant Staphylococcus aureus, a mouse model of Alzheimer's disease, and ApoE-knockout-induced atherosclerosis. These data indicate the profound effect of increasing myeloid cell function. Further, they suggest that an appropriate way to evaluate inflammation in both acute and chronic diseases is to ask whether the inflammatory infiltrate is sufficient to eliminate the immune challenge. The expression of ACE by myeloid cells induces a heightened immune response by these cells. The overexpression of ACE is associated with immune function beyond that possible by wild type (WT) myeloid cells. A heightened immune response effectively resolves disease in a variety of acute and chronic models of disease including models of Alzheimer's disease and atherosclerosis.


Asunto(s)
Hipertensión , Inflamación , Staphylococcus aureus Resistente a Meticilina , Peptidil-Dipeptidasa A , Animales , Enfermedad Crónica , Humanos , Ratones , Células Mieloides , Peptidil-Dipeptidasa A/metabolismo
7.
Cell Mol Biol Lett ; 25: 31, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32508938

RESUMEN

Angiotensin-converting enzyme (ACE), a dicarboxypeptidase, plays a major role in the regulation of blood pressure by cleaving angiotensin I into angiotensin II (Ang II), a potent vasoconstrictor. Because of its wide substrate specificity and tissue distribution, ACE affects many diverse biological processes. In inflammatory diseases, including granuloma, atherosclerosis, chronic kidney disease and bacterial infection, ACE expression gets upregulated in immune cells, especially in myeloid cells. With increasing evidences connecting ACE functions to the pathogenesis of these acquired diseases, it is suggested that ACE plays a vital role in immune functions. Recent studies with mouse models of bacterial infection and tumor suggest that ACE plays an important role in the immune responses of myeloid cells. Inhibition of ACE suppresses neutrophil immune response to bacterial infection. In contrast, ACE overexpression in myeloid cells strongly induced bacterial and tumor resistance in mice. A detailed biochemical understanding of how ACE activates myeloid cells and which ACE peptide(s) (substrate or product) mediate these effects could lead to the development of novel therapies for boosting immunity against a variety of stimuli, including bacterial infection and tumor.


Asunto(s)
Hematopoyesis/inmunología , Inflamación/inmunología , Células Mieloides/inmunología , Peptidil-Dipeptidasa A/fisiología , Inmunidad Adaptativa , Animales , Infecciones Bacterianas/inmunología , Humanos , Ratones , Neoplasias/inmunología , Peptidil-Dipeptidasa A/inmunología
8.
Biochem Biophys Res Commun ; 520(3): 573-579, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31615657

RESUMEN

BACKGROUND: Macrophages are ubiquitous in all stages of atherosclerosis, exerting tremendous impact on lesion progression and plaque stability. Because macrophages in atherosclerotic plaques express angiotensin-converting enzyme (ACE), current dogma posits that local myeloid-mediated effects worsen the disease. In contrast, we previously reported that myeloid ACE overexpression augments macrophage resistance to various immune challenges, including tumors, bacterial infection and Alzheimer's plaque deposition. Here, we sought to assess the impact of myeloid ACE on atherosclerosis. METHODS: A mouse model in which ACE is overexpressed in myelomonocytic lineage cells, called ACE10, was generated and sequentially crossed with ApoE-deficient mice to create ACE10/10ApoE-/- (ACE10/ApoE). Control mice were ACEWT/WTApoE-/- (WT/ApoE). Atherosclerosis was induced using an atherogenic diet alone, or in combination with unilateral nephrectomy plus deoxycorticosterone acetate (DOCA) salt for eight weeks. RESULTS: With an atherogenic diet alone or in combination with DOCA, the ACE10/ApoE mice showed significantly less atherosclerotic plaques compared to their WT/ApoE counterparts (p < 0.01). When recipient ApoE-/- mice were reconstituted with ACE10/10 bone marrow, these mice showed significantly reduced lesion areas compared to recipients reconstituted with wild type bone marrow. Furthermore, transfer of ACE-deficient bone marrow had no impact on lesion area. CONCLUSION: Our data indicate that while myeloid ACE may not be required for atherosclerosis, enhanced ACE expression paradoxically reduced disease progression.


Asunto(s)
Aterosclerosis/enzimología , Aterosclerosis/prevención & control , Células Mieloides/enzimología , Peptidil-Dipeptidasa A/metabolismo , Animales , Aterosclerosis/genética , Presión Sanguínea , Trasplante de Médula Ósea , Linaje de la Célula/genética , Colesterol/sangre , Dieta Aterogénica , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Macrófagos/enzimología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Células Mieloides/patología , Peptidil-Dipeptidasa A/genética , Regulación hacia Arriba
9.
Clin Sci (Lond) ; 133(2): 321-334, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30622219

RESUMEN

Acute respiratory distress syndrome (ARDS) in a deadly disease that can be brought on by endotoxins such as lipopolysaccharide (LPS). ARDS is characterized by vascular permeability, a severe inflammatory response, lung leukocyte infiltration, and resultant lung edema. Polymerase δ-interacting protein 2 (Poldip2) is a novel regulator of blood-brain barrier permeability; however, its role in regulating lung permeability and vascular inflammation is unknown. Here, the role of Poldip2 in regulating vascular permeability and inflammation in a mouse model of ARDS was assessed. Heterozygous deletion of Poldip2 was found to reduce LPS-induced mortality within 20 h, lung inflammatory signaling, and leukocyte infiltration. Moreover, reduced Poldip2-suppressed LP-induced vascular cell adhesion molecule (VCAM)-1 induction, leukocyte recruitment, and mitochondrial reactive oxygen species (ROS) production in vitro These data indicate that Poldip2 is an important regulator of the debilitating consequences of ARDS, potentially through the regulation of mitochondrial ROS-induced inflammatory signaling. Consequently, inhibition of Poldip2 may be a viable option for therapeutic discovery moving forward.


Asunto(s)
Permeabilidad Capilar , Células Endoteliales/metabolismo , Pulmón/irrigación sanguínea , Proteínas Mitocondriales/deficiencia , Proteínas Nucleares/deficiencia , Edema Pulmonar/prevención & control , Síndrome de Dificultad Respiratoria/metabolismo , Vasculitis/prevención & control , Animales , Adhesión Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Humanos , Leucocitos/metabolismo , Leucocitos/patología , Masculino , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Edema Pulmonar/genética , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Especies Reactivas de Oxígeno/metabolismo , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/patología , Transducción de Señal , Células THP-1 , Molécula 1 de Adhesión Celular Vascular/metabolismo , Vasculitis/genética , Vasculitis/metabolismo , Vasculitis/patología
10.
J Interv Cardiol ; 2019: 8947204, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31772549

RESUMEN

OBJECTIVE: To assess 1-year mortality after transcatheter aortic valve replacement (TAVR) in patients with bicuspid aortic stenosis (AS). BACKGROUND: Clinical trials have proven the beneficial effect of TAVR on mortality in patients with tricuspid AS. Individuals with bicuspid AS were excluded from these trials. METHODS: A meta-analysis using literature search from the Cochrane, PubMed, ClinicalTrials, SCOPUS, and EMBASE databases was conducted to determine the effect of TAVR on 1-year mortality in patients with bicuspid AS. Short-term outcomes that could potentially impact one-year mortality were analyzed. RESULTS: After evaluating 380 potential articles, 5 observational studies were selected. A total of 3890 patients treated with TAVR were included: 721 had bicuspid and 3,169 had tricuspid AS. No statistically significant difference between the baseline characteristics of the two groups of patients was seen outside of mean aortic gradient. Our primary endpoint of one-year all-cause mortality revealed 85 deaths in 719 patients (11.82%) with bicuspid AS compared to 467 deaths in 3100 patients (15.06%) with tricuspid AS, with no difference between both groups [relative risk (RR) 1.03; 95% CI 0.70-1.51]. Patients with bicuspid AS were associated with a decrease in device success (RR 0.62; 95% CI 0.45-0.84) and an increase in moderate-to-severe prosthetic valve regurgitation (RR 1.55; 95% CI 1.07-2.22) after TAVR compared to patients with tricuspid AS. The effect of meta-regression coefficients on one-year all-cause mortality was not statistically significant for any patient baseline characteristics. CONCLUSION: When comparing TAVR procedure in tricuspid AS versus bicuspid AS, there was no difference noted in one-year all-cause mortality.


Asunto(s)
Estenosis de la Válvula Aórtica/cirugía , Válvula Aórtica , Complicaciones Posoperatorias/mortalidad , Reemplazo de la Válvula Aórtica Transcatéter , Válvula Aórtica/diagnóstico por imagen , Válvula Aórtica/patología , Válvula Aórtica/cirugía , Estenosis de la Válvula Aórtica/diagnóstico , Humanos , Evaluación de Resultado en la Atención de Salud , Reemplazo de la Válvula Aórtica Transcatéter/efectos adversos , Reemplazo de la Válvula Aórtica Transcatéter/métodos
11.
Arterioscler Thromb Vasc Biol ; 38(5): 1125-1133, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29545241

RESUMEN

OBJECTIVE: The adaptive response to vascular injury is the formation of functional collateral vessels to maintain organ integrity. Many of the clinical complications associated with sickle cell disease can be attributed to repeated bouts of vascular insufficiency, yet the detailed mechanisms of collateral vessel formation after injury are largely unknown in sickle cell disease. Here, we characterize postischemic neovascularization in sickle cell disease and the role of neutrophils in the production of reactive oxygen species. APPROACH AND RESULTS: We induced hindlimb ischemia by ligation of the femoral artery in Townes SS (sickle cell) mice compared with AA (wild type) mice. Perfusion recovery, ascertained using LASER (light amplification by stimulated emission of radiation) Doppler perfusion imaging, showed significant diminution in collateral vessel formation in SS mice after hindlimb ischemia (76±13% AA versus 34±10% in SS by day 28; P<0.001; n=10 per group). The incidence of amputation (25% versus 5%) and foot necrosis (80% versus 15%) after hindlimb ischemia was significantly increased in the SS mice. Motor function recovery evaluation by the running wheel assay was also impaired in SS mice (36% versus 97% at 28 days post-hindlimb ischemia; P<0.001). This phenotype was associated with persistent and excessive production of reactive oxygen species by neutrophils. Importantly, neutrophil depletion or treatment with the antioxidant N-acetylcysteine reduced oxidative stress and improved functional collateral formation in the SS mice. CONCLUSIONS: Our data suggest dysfunctional collateral vessel formation in SS mice after vascular injury and provide a mechanistic basis for the multiple vascular complications of sickle cell disease.


Asunto(s)
Anemia de Células Falciformes/fisiopatología , Circulación Colateral , Isquemia/fisiopatología , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Acetilcisteína/farmacología , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/metabolismo , Animales , Antioxidantes/farmacología , Velocidad del Flujo Sanguíneo , Circulación Colateral/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Miembro Posterior , Peróxido de Hidrógeno/metabolismo , Isquemia/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Fisiológica/efectos de los fármacos , Neutrófilos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fenotipo , Flujo Sanguíneo Regional , Factores de Tiempo
13.
J Neurooncol ; 136(2): 289-298, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29124649

RESUMEN

Preclinical evidence suggests angiotensin blockade therapy (ABT) decreases late radiation toxicities. This study aims to investigate the association between ABT and symptomatic radiation necrosis (SRN) following stereotactic radiosurgery (SRS). Resected brain metastases (rBM) and arteriovenous malformation (AVM) patients treated with SRS from 2002 to 2015 were identified. Patients in the ABT cohort were on therapy during SRS and at 1-month follow up. Kaplan Meier method and cumulative incidence model were used to analyze overall survival (OS) and intracranial outcomes. 228 consecutive patients were treated with SRS: 111 with rBM and 117 with AVM. Overall, 51 (22.4%) patients were in the ABT group: 32 (28.8%) in the rBM and 19 (16.2%) in AVM cohorts. Baseline characteristics were similar, except for higher Graded Prognostic Analysis (3-4) in the rBM (ABT: 25.0% vs. non-ABT: 49.0%, p = 0.033) and median age in the AVM (ABT: 51.4 vs. non-ABT: 35.4, p < 0.001) cohorts. In both populations, OS and intracranial efficacy (rBM-local control; AVM-obliteration rates) were statistically similar between the cohorts. ABT was associated with lower 1-year SRN rates in both populations: rBM, 3.1 versus 25.3% (p = 0.003); AVM, 6.7 vs. 14.6% (p = 0.063). On multivariate analysis, ABT was a significant predictive factor for rBM (HR: 0.17; 95% CI 0.03-0.88, p = 0.035), but did not reach statistical significance for AVM (HR: 0.36; 95% CI 0.09-1.52, p = 0.165). ABT use appears to be associated with a reduced risk of SRN following SRS, without detriment to OS or intracranial efficacy. A prospective trial to validate these findings is warranted.


Asunto(s)
Antagonistas de Receptores de Angiotensina/uso terapéutico , Neoplasias Encefálicas/radioterapia , Malformaciones Arteriovenosas Intracraneales/radioterapia , Traumatismos por Radiación/prevención & control , Radiocirugia/efectos adversos , Adulto , Anciano , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/cirugía , Estudios de Cohortes , Femenino , Humanos , Malformaciones Arteriovenosas Intracraneales/patología , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Necrosis , Resultado del Tratamiento
14.
Lab Invest ; 94(5): 536-44, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24614194

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells which accumulate in cancer, infection and chronic inflammation. These cells suppress T-cell function and the immune response. Angiotensin-converting enzyme (ACE) is a peptidase that is now known to regulate aspects of myelopoiesis. Here, we show that ACE expression correlates with myeloid maturation in vitro. Forced ACE overexpression in monocytic cells reduces the generation of MDSCs. In vivo, mice with a genetic change resulting in myeloid cell ACE overexpression have reduced numbers of blood and splenic MDSCs in a tumor model and in a model of chronic inflammation induced by complete Freund's adjuvant. In contrast, ACE-null mice produce large numbers of MDSCs during chronic inflammation. Macrophages from mice with myeloid ACE overexpressing are more pro-inflammatory and have more tumor-killing activity than cells from wild-type mice. Thus, manipulating myeloid ACE activity can interfere with MDSC development and the maturation of myeloid cells.


Asunto(s)
Células Progenitoras Mieloides/fisiología , Mielopoyesis , Peptidil-Dipeptidasa A/metabolismo , Animales , Melanoma Experimental/enzimología , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/enzimología , Fenotipo
15.
Ann Surg Oncol ; 21(1): 93-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24081796

RESUMEN

BACKGROUND: The purpose of this study was to describe the incidence and location of ipsilateral breast tumor recurrence (IBTR) among breast cancer patients treated with oncoplastic reduction mammoplasty (ORM) and radiotherapy (RT). METHODS: The medical records of 86 consecutive women with ductal carcinoma in situ (DCIS) (n = 11) or invasive carcinoma of the breast (n = 75) treated with ORM at Emory University between January 1994 and December 2010 were reviewed. RESULTS: Following ORM, prolonged wound healing or surgical complications led to delay of adjuvant chemotherapy or RT in 11 patients. Surgical clips were found outside the primary tumor breast quadrant in 43 % of the patients with available RT planning CT images. When the clips were found outside the primary tumor quadrant, the RT boost was more frequently delivered outside versus inside the primary tumor quadrant (67 vs. 33 %, p < 0.001). After a median follow-up period of 4.5 years (range 0.1-17.9), 6 patients developed an IBTR and only 1 IBTR occurred outside the primary tumor quadrant. The 5-year ipsilateral breast tumor control rates were 91 % (95 % CI 0.82-0.99) and 93 % (95 % CI 0.90-0.97) for patients with DCIS and invasive carcinoma, respectively. CONCLUSIONS: The use of ORM yields acceptable rates of IBTR. ORM may displace breast tissue and surgical clips to breast quadrants outside of the original tumor location, but the majority of IBTRs still occur in the original tumor quadrant. This area remains at highest risk of in-breast recurrence in women treated with ORM irrespective of surgical clip location.


Asunto(s)
Neoplasias de la Mama/terapia , Carcinoma Ductal de Mama/terapia , Carcinoma Intraductal no Infiltrante/terapia , Carcinoma Lobular/terapia , Mamoplastia , Mastectomía Segmentaria , Recurrencia Local de Neoplasia/diagnóstico , Radioterapia , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/mortalidad , Carcinoma Ductal de Mama/secundario , Carcinoma Intraductal no Infiltrante/mortalidad , Carcinoma Intraductal no Infiltrante/secundario , Carcinoma Lobular/mortalidad , Carcinoma Lobular/secundario , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Metástasis Linfática , Persona de Mediana Edad , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Tasa de Supervivencia
16.
Cancer Discov ; 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38552005

RESUMEN

Tumor-associated macrophages are transcriptionally heterogeneous, but the spatial distribution and cell interactions that shape macrophage tissue roles remain poorly characterized. Here, we spatially resolve five distinct human macrophage populations in normal and malignant human breast and colon tissue and reveal their cellular associations. This spatial map reveals that distinct macrophage populations reside in spatially segregated micro-environmental niches with conserved cellular compositions that are repeated across healthy and diseased tissue. We show that IL4I1+ macrophages phagocytose dying cells in areas with high cell turnover and predict good outcome in colon cancer. In contrast, SPP1+ macrophages are enriched in hypoxic and necrotic tumor regions and portend worse outcome in colon cancer. A subset of FOLR2+ macrophages is embedded in plasma cell niches. NLRP3+ macrophages co-localize with neutrophils and activate an inflammasome in tumors. Our findings indicate that a limited number of unique human macrophage niches function as fundamental building blocks in tissue.

17.
Am J Physiol Regul Integr Comp Physiol ; 305(3): R205-15, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23739345

RESUMEN

The renin angiotensin system (RAS) is a network of enzymes and peptides that coalesce primarily on the angiotensin II type 1 receptor (AT1R) to induce cell proliferation, angiogenesis, fibrosis, and blood pressure control. Angiotensin-converting enzyme (ACE), the key peptidase of the RAS, is promiscuous in that it cleaves other substrates such as substance P and bradykinin. Accumulating evidence implicates ACE in the pathophysiology of carcinogenesis. While the role of ACE and its peptide network in modulating angiogenesis via the AT1R is well documented, its involvement in shaping other aspects of the tumor microenvironment remains largely unknown. Here, we review the role of ACE in modulating the immune compartment of the tumor microenvironment, which encompasses the immunosuppressive, cancer-promoting myeloid-derived suppressor cells, alternatively activated tumor-associated macrophages, and T regulatory cells. We also discuss the potential roles of peptides that accumulate in the setting of chronic ACE inhibitor use, such as bradykinin, substance P, and N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP), and how they may undercut the gains of anti-angiogenesis from ACE inhibition. These emerging mechanisms may harmonize the often-conflicting results on the role of ACE inhibitors and ACE polymorphisms in various cancers and call for further investigations into the potential benefit of ACE inhibitors in some neoplasms.


Asunto(s)
Neoplasias/enzimología , Neoplasias/fisiopatología , Neovascularización Patológica/fisiopatología , Peptidil-Dipeptidasa A/metabolismo , Microambiente Tumoral , Animales , Humanos , Células Mieloides/fisiología , Neovascularización Fisiológica/fisiología , Sistema Renina-Angiotensina/fisiología , Linfocitos T/fisiología
18.
Cardiovasc Res ; 119(9): 1825-1841, 2023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37225143

RESUMEN

AIMS: The metabolic failure of macrophages to adequately process lipid is central to the aetiology of atherosclerosis. Here, we examine the role of macrophage angiotensin-converting enzyme (ACE) in a mouse model of PCSK9-induced atherosclerosis. METHODS AND RESULTS: Atherosclerosis in mice was induced with AAV-PCSK9 and a high-fat diet. Animals with increased macrophage ACE (ACE 10/10 mice) have a marked reduction in atherosclerosis vs. WT mice. Macrophages from both the aorta and peritoneum of ACE 10/10 express increased PPARα and have a profoundly altered phenotype to process lipids characterized by higher levels of the surface scavenger receptor CD36, increased uptake of lipid, increased capacity to transport long chain fatty acids into mitochondria, higher oxidative metabolism and lipid ß-oxidation as determined using 13C isotope tracing, increased cell ATP, increased capacity for efferocytosis, increased concentrations of the lipid transporters ABCA1 and ABCG1, and increased cholesterol efflux. These effects are mostly independent of angiotensin II. Human THP-1 cells, when modified to express more ACE, increase expression of PPARα, increase cell ATP and acetyl-CoA, and increase cell efferocytosis. CONCLUSION: Increased macrophage ACE expression enhances macrophage lipid metabolism, cholesterol efflux, efferocytosis, and it reduces atherosclerosis. This has implications for the treatment of cardiovascular disease with angiotensin II receptor antagonists vs. ACE inhibitors.


Asunto(s)
Aterosclerosis , Proproteína Convertasa 9 , Humanos , Animales , Ratones , Proproteína Convertasa 9/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Metabolismo de los Lípidos , Colesterol/metabolismo , Macrófagos/metabolismo , Aterosclerosis/genética , Aterosclerosis/prevención & control , Angiotensinas/metabolismo , Adenosina Trifosfato/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo
19.
Front Immunol ; 14: 1278383, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37928535

RESUMEN

The pathogenesis of atherosclerosis is defined by impaired lipid handling by macrophages which increases intracellular lipid accumulation. This dysregulation of macrophages triggers the accumulation of apoptotic cells and chronic inflammation which contributes to disease progression. We previously reported that mice with increased macrophage-specific angiotensin-converting enzyme, termed ACE10/10 mice, resist atherosclerosis in an adeno-associated virus-proprotein convertase subtilisin/kexin type 9 (AAV-PCSK9)-induced model. This is due to increased lipid metabolism by macrophages which contributes to plaque resolution. However, the importance of ACE in peripheral blood monocytes, which are the primary precursors of lesional-infiltrating macrophages, is still unknown in atherosclerosis. Here, we show that the ACE-mediated metabolic phenotype is already triggered in peripheral blood circulating monocytes and that this functional modification is directly transferred to differentiated macrophages in ACE10/10 mice. We found that Ly-6Clo monocytes were increased in atherosclerotic ACE10/10 mice. The monocytes isolated from atherosclerotic ACE10/10 mice showed enhanced lipid metabolism, elevated mitochondrial activity, and increased adenosine triphosphate (ATP) levels which implies that ACE overexpression is already altered in atherosclerosis. Furthermore, we observed increased oxygen consumption (VO2), respiratory exchange ratio (RER), and spontaneous physical activity in ACE10/10 mice compared to WT mice in atherosclerotic conditions, indicating enhanced systemic energy consumption. Thus, ACE overexpression in myeloid lineage cells modifies the metabolic function of peripheral blood circulating monocytes which differentiate to macrophages and protect against atherosclerotic lesion progression due to better lipid metabolism.


Asunto(s)
Aterosclerosis , Proproteína Convertasa 9 , Animales , Ratones , Aterosclerosis/patología , Lípidos , Células Mieloides/patología
20.
FASEB J ; 25(4): 1145-55, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21148418

RESUMEN

Inhibition of angiotensin-converting enzyme (ACE) induces anemia in humans and mice, but it is unclear whether ACE is involved in other aspects of hematopoiesis. Here, we systemically evaluated ACE-knockout (KO) mice and found myelopoietic abnormalities characterized by increased bone marrow myeloblasts and myelocytes, as well as extramedullary myelopoiesis. Peritoneal macrophages from ACE-KO mice were deficient in the production of effector molecules, such as tumor necrosis factor-α, interleukin-12p40, and CD86 when stimulated with lipopolysaccharide and interferon-γ. ACE-KO mice were more susceptible to Staphylococcus aureus infection. Further studies using total or fractionated bone marrows revealed that ACE regulates myeloid proliferation, differentiation, and functional maturation via angiotensin II and substance P and through the angiotensin II receptor type 1 and substance P neurokinin 1 receptors. Angiotensin II was correlated with CCAAT-enhancer-binding protein-α up-regulation during myelopoiesis. Angiotensin II supplementation of ACE-KO mice rescued macrophage functional maturation. These results demonstrate a previous unrecognized significant role for ACE in myelopoiesis and imply new perspectives for manipulating myeloid cell expansion and maturation.


Asunto(s)
Mielopoyesis/fisiología , Peptidil-Dipeptidasa A/fisiología , Angiotensina II/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Lisinopril/farmacología , Macrófagos/fisiología , Ratones , Ratones Noqueados , Mielopoyesis/genética , Sustancia P/fisiología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA