Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hypertension ; 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690653

RESUMEN

BACKGROUND: Ssmall-interfering RNA (siRNA) targeting hepatic AGT (angiotensinogen) mRNA depletes AGT, lowering blood pressure for up to 6 months. However, certain situations may require a rapid angiotensin increase. The reverse siRNA silencing, RVR technology is a potential approach to counteract siRNA effects. METHODS: Spontaneously hypertensive rats received 10 mg/kg AGT siRNA, and 3 weeks later were given AGT-RVR (1, 10, or 20 mg/kg). One week after AGT-RVR dosing, a redose of AGT siRNA assessed its post-AGT-RVR effectiveness for 2 weeks. Additionally, the impact of AGT-RVR after an equihypotensive dose of valsartan (4 mg/kg per day) was examined. RESULTS: Baseline mean arterial pressure (MAP) was 144±1 mm Hg. AGT siRNA reduced MAP by ≈16 mm Hg and AGT by >95%, while renin increased 25-fold. All AGT-RVR doses restored MAP to baseline within 4 to 7 days. Notably, 10 and 20 mg/kg restored AGT and renin to baseline, while 1 mg/kg allowed ≈50% AGT restoration, with renin remaining above baseline. A second AGT siRNA treatment, following 1-mg/kg AGT-RVR, reduced MAP to the same degree as the initial dose, while following 10 mg/kg AGT-RVR, it resulted in ≈50% of the first dose's MAP effect at 2 weeks. The valsartan-induced MAP reduction was unaffected by AGT-RVR. CONCLUSIONS: In spontaneously hypertensive rats, angiotensinogen-RVR dose-dependently reversed AGT siRNA-induced AGT reduction, normalizing MAP. MAP normalization persisted even with 50% recovered AGT levels, likely due to upregulated renin maintaining adequate angiotensin generation. Post-AGT-RVR dosing, a second AGT siRNA dose lowered MAP again.

3.
Hypertension ; 81(4): 861-875, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38361240

RESUMEN

BACKGROUND: Chemerin, an inflammatory adipokine, is upregulated in preeclampsia, and its placental overexpression results in preeclampsia-like symptoms in mice. Statins may lower chemerin. METHODS: Chemerin was determined in a prospective cohort study in women suspected of preeclampsia and evaluated as a predictor versus the sFlt-1 (soluble fms-like tyrosine kinase-1)/PlGF (placental growth factor) ratio. Chemerin release was studied in perfused placentas and placental explants with or without the statins pravastatin and fluvastatin. We also addressed statin placental passage and the effects of chemerin in chorionic plate arteries. RESULTS: Serum chemerin was elevated in women with preeclampsia, and its addition to a predictive model yielded significant effects on top of the sFlt-1/PlGF ratio to predict preeclampsia and its fetal complications. Perfused placentas and explants of preeclamptic women released more chemerin and sFlt-1 and less PlGF than those of healthy pregnant women. Statins reversed this. Both statins entered the fetal compartment, and the fetal/maternal concentration ratio of pravastatin was twice that of fluvastatin. Chemerin constricted plate arteries, and this was blocked by a chemerin receptor antagonist and pravastatin. Chemerin did not potentiate endothelin-1 in chorionic plate arteries. In explants, statins upregulated low-density lipoprotein receptor expression, which relies on the same transcription factor as chemerin, and NO release. CONCLUSIONS: Chemerin is a biomarker for preeclampsia, and statins both prevent its placental upregulation and effects, in an NO and low-density lipoprotein receptor-dependent manner. Combined with their capacity to improve the sFlt-1/PlGF ratio, this offers an attractive mechanism by which statins may prevent or treat preeclampsia.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Preeclampsia , Humanos , Embarazo , Femenino , Animales , Ratones , Placenta/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Factor de Crecimiento Placentario , Pravastatina/farmacología , Regulación hacia Arriba , Estudios Prospectivos , Preeclampsia/tratamiento farmacológico , Preeclampsia/prevención & control , Fluvastatina/metabolismo , Fluvastatina/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacología , Biomarcadores , Quimiocinas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo
4.
J Hypertens ; 42(5): 883-892, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38088400

RESUMEN

BACKGROUND: Sodium-glucose cotransporter-2 (SGLT2) inhibition exerts cardioprotective and renoprotective effects, often on top of renin-angiotensin system (RAS) blockade. We investigated this in diabetic hypertensive (mREN2)27 rats. METHODS: Rats were made diabetic with streptozotocin and treated with vehicle, the angiotensin receptor blocker valsartan, the SGLT2 inhibitor empagliflozin, or their combination. Blood pressure (BP) was measured by telemetry. RESULTS: Diabetes resulted in albuminuria, accompanied by glomerulosclerosis, without a change in glomerular filtration rate. Empagliflozin did not lower BP, while valsartan did, and when combined the BP drop was largest. Only dual blockade reduced cardiac hypertrophy and prevented left ventricular dilatation. Valsartan, but not empagliflozin, increased renin, and the largest renin rise occurred during dual blockade, resulting in plasma angiotensin II [but not angiotensin-(1-7)] upregulation. In contrast, in the kidney, valsartan lowered angiotensin II and angiotensin-(1-7), and empagliflozin did not alter this. Although both valsartan and empagliflozin alone tended to diminish albuminuria, the reduction was significant only when both drugs were combined. This was accompanied by reduced glomerulosclerosis, no change in glomerular filtration rate, and a favorable expression pattern of fibrosis and inflammatory markers (including SGLT2) in the kidney. CONCLUSION: RAS blockade and SGLT2 inhibition display synergistic beneficial effects on BP, kidney injury and cardiac hypertrophy in a rat with hypertension and diabetes. The synergy does not involve upregulation of angiotensin-(1-7), but may relate to direct RAS-independent effects of empagliflozin in the heart and kidney.


Asunto(s)
Compuestos de Bencidrilo , Diabetes Mellitus , Glucósidos , Hipertensión , Ratas , Animales , Sistema Renina-Angiotensina , Renina , Angiotensina II/farmacología , Albuminuria , Transportador 2 de Sodio-Glucosa/metabolismo , Valsartán/farmacología , Valsartán/uso terapéutico , Presión Sanguínea/fisiología , Cardiomegalia , Glucosa/farmacología , Glucosa/uso terapéutico , Sodio/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 43(12): 2256-2264, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37855126

RESUMEN

Blood pressure management involves antihypertensive therapies blocking the renin-angiotensin system (RAS). Yet, it might be inadequate due to poor patient adherence or the so-called RAS escape phenomenon, elicited by the compensatory renin elevation upon RAS blockade. Recently, evidence points toward targeting hepatic AGT (angiotensinogen) as a novel approach to block the RAS pathway that could circumvent the RAS escape phenomenon. Removing AGT, from which all angiotensins originate, should prevent further angiotensin generation, even when renin rises. Furthermore, by making use of a trivalent N-acetylgalactosamine ligand-conjugated small interfering RNA that specifically targets the degradation of hepatocyte-produced mRNAs in a highly potent and specific manner, it may be possible in the future to manage hypertension with therapy that is administered 1 to 2× per year, thereby supporting medication adherence. This review summarizes all current findings on AGT small interfering RNA in preclinical models, making a comparison versus classical RAS blockade with either ACE (angiotensin-converting enzyme) inhibitors or AT1 (angiotensin II type 1) receptor antagonists and AGT suppression with antisense oligonucleotides. It ends with discussing the first-in-human study with AGT small interfering RNA.


Asunto(s)
Angiotensinógeno , Hipertensión , Humanos , Acetilgalactosamina , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Presión Sanguínea , Hipertensión/terapia , Hipertensión/tratamiento farmacológico , Renina/metabolismo , Sistema Renina-Angiotensina/fisiología , ARN Interferente Pequeño/farmacología
6.
Br J Pharmacol ; 180(1): 80-93, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36106615

RESUMEN

BACKGROUND AND PURPOSE: Small interfering RNA (siRNA) targeting liver angiotensinogen lowers blood pressure, but its effects in hypertensive diabetes are unknown. EXPERIMENTAL APPROACH: To address this, TGR (mRen2)27 rats (angiotensin II-dependent hypertension model) were made diabetic with streptozotocin over 18 weeks and treated with either vehicle, angiotensinogen siRNA, the AT1 antagonist valsartan, the ACE inhibitor captopril, valsartan + siRNA or valsartan + captopril for the final 3 weeks. Mean arterial pressure (MAP) was measured via radiotelemetry. KEY RESULTS: MAP before treatment was 153 ± 2 mmHg. Diabetes resulted in albuminuria, accompanied by glomerulosclerosis and podocyte effacement, without a change in glomerular filtration rate. All treatments lowered MAP and cardiac hypertrophy, and the largest drop in MAP was observed with siRNA + valsartan. Treatment with siRNA lowered circulating angiotensinogen by >99%, and the lowest circulating angiotensin II and aldosterone levels occurred in the dual treatment groups. Angiotensinogen siRNA did not affect renal angiotensinogen mRNA expression, confirming its liver-specificity. Furthermore, only siRNA with or without valsartan lowered renal angiotensin I. All treatments lowered renal angiotensin II and the reduction was largest (>95%) in the siRNA + valsartan group. All treatments identically lowered albuminuria, whereas only siRNA with or without valsartan restored podocyte foot processes and reduced glomerulosclerosis. CONCLUSION AND IMPLICATIONS: Angiotensinogen siRNA exerts renoprotection in diabetic TGR (mRen2)27 rats and this relies, at least in part, on the suppression of renal angiotensin II formation from liver-derived angiotensinogen. Clinical trials should now address whether this is also beneficial in human diabetic kidney disease.


Asunto(s)
Angiotensina II , Diabetes Mellitus Experimental , Hipertensión , Enfermedades Renales , ARN Interferente Pequeño , Animales , Humanos , Ratas , Albuminuria , Angiotensina II/efectos de los fármacos , Angiotensina II/genética , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Hipertensión/tratamiento farmacológico , Hígado/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina , Valsartán/farmacología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Enfermedades Renales/prevención & control , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico
7.
Hypertension ; 79(10): 2115-2126, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35904033

RESUMEN

Multiple types of renin-angiotensin system (RAS) blockers exist, allowing interference with the system at the level of renin, angiotensin-converting enzyme, or the angiotensin II receptor. Yet, in particular, for the treatment of hypertension, the number of patients with uncontrolled hypertension continues to rise, either due to patient noncompliance or because of the significant renin rises that may, at least partially, overcome the effect of RAS blockade (RAS escape). New approaches to target the RAS are either direct antisense oligonucleotides that inhibit angiotensinogen RNA translation, or small interfering RNA (siRNA) that function via the RNA interference pathway. Since all angiotensins stem from angiotensinogen, lowering angiotensinogen has the potential to circumvent the RAS escape phenomenon. Moreover, antisense oligonucleotides and small interfering RNA require injections only every few weeks to months, which might reduce noncompliance. Of course, angiotensinogen suppression also poses a threat in situations where the RAS is acutely needed, for instance in women becoming pregnant during treatment, or in cases of emergency, when severe hypotension occurs. This review discusses all preclinical data on angiotensinogen suppression, as well as the limited clinical data that are currently available. It concludes that it is an exciting new tool to target the RAS with high specificity and a low side effect profile. Its long-term action might revolutionize pharmacotherapy, as it could overcome compliance problems. Preclinical and clinical programs are now carefully investigating its efficacy and safety profile, allowing an optimal introduction as a novel drug to treat cardiovascular and renal diseases in due time.


Asunto(s)
Hipertensión , Enfermedades Renales , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Femenino , Humanos , Oligonucleótidos Antisentido/metabolismo , Embarazo , ARN Interferente Pequeño/farmacología , Renina/metabolismo , Sistema Renina-Angiotensina
8.
J Cardiovasc Pharmacol ; 78(Suppl 6): S53-S62, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34840262

RESUMEN

ABSTRACT: In recent years, perivascular adipose tissue (PVAT) research has gained special attention in an effort to understand its involvement in vascular function. PVAT is recognized as an important endocrine organ that secretes procontractile and anticontractile factors, including components of the renin-angiotensin-aldosterone system, particularly angiotensinogen (AGT). This review critically addresses the occurrence of AGT in PVAT, its release into the blood stream, and its contribution to the generation and effects of angiotensins (notably angiotensin-(1-7) and angiotensin II) in the vascular wall. It describes that the introduction of transgenic animals, expressing AGT at 0, 1, or more specific location(s), combined with the careful measurement of angiotensins, has revealed that the assumption that PVAT independently generates angiotensins from locally synthesized AGT is incorrect. Indeed, selective deletion of AGT from adipocytes did not lower circulating AGT, neither under a control diet nor under a high-fat diet, and only liver-specific AGT deletion resulted in the disappearance of AGT from blood plasma and adipose tissue. An entirely novel scenario therefore develops, supporting local angiotensin generation in PVAT that depends on the uptake of both AGT and renin from blood, in addition to the possibility that circulating angiotensins exert vascular effects. The review ends with a summary of where we stand now and recommendations for future research.


Asunto(s)
Tejido Adiposo/metabolismo , Angiotensinógeno/metabolismo , Vasos Sanguíneos/metabolismo , Sistema Renina-Angiotensina , Vasoconstricción , Vasodilatación , Tejido Adiposo/fisiopatología , Vasos Sanguíneos/fisiopatología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Humanos , Transducción de Señal
9.
Clin Sci (Lond) ; 135(14): 1727-1731, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34291792

RESUMEN

Although the existence of a brain renin-angiotensin system (RAS) had been proposed five decades ago, we still struggle to understand how it functions. The main reason for this is the virtual lack of renin at brain tissue sites. Moreover, although renin's substrate, angiotensinogen, appears to be synthesized locally in the brain, brain angiotensin (Ang) II disappeared after selective silencing of hepatic angiotensinogen. This implies that brain Ang generation depends on hepatic angiotensinogen after all. Rodrigues et al. (Clin Sci (Lond) (2021) 135:1353-1367) generated a transgenic mouse model overexpressing full-length rat angiotensinogen in astrocytes, and observed massively elevated brain Ang II levels, increased sympathetic nervous activity and vasopressin, and up-regulated erythropoiesis. Yet, blood pressure and kidney function remained unaltered, and surprisingly no other Ang metabolites occurred in the brain. Circulating renin was suppressed. This commentary critically discusses these findings, concluding that apparently in the brain, overexpressed angiotensinogen can be cleaved by an unidentified non-renin enzyme, yielding Ang II directly, which then binds to Ang receptors, allowing no metabolism by angiotensinases like ACE2 and aminopeptidase A. Future studies should now unravel the identity of this non-renin enzyme, and determine whether it also contributes to Ang II generation at brain tissue sites in wildtype animals. Such studies should also re-evaluate the concept that Ang-(1-7) and Ang III, generated by ACE2 and aminopeptidase A, respectively, have important functions in the brain.


Asunto(s)
Encéfalo/metabolismo , Hipertensión/fisiopatología , Sistema Renina-Angiotensina/fisiología , Renina/metabolismo , Angiotensina II/metabolismo , Humanos , Hipertensión/metabolismo , Receptores de Angiotensina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...