Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
Pharmacol Res ; 170: 105724, 2021 08.
Article En | MEDLINE | ID: mdl-34116209

Telmisartan prevents diet-induced obesity (DIO) in rodents. Given that the precise underlying mechanism is not known, we examined whether a gut-related mechanism might be involved. Sprague-Dawley rats received cafeteria diet (CD) for 3 months to develop DIO and were administered either telmisartan (8 mg/kgbw) or vehicle. In addition, pair-fed (PF) rats received CD adjusted to TEL and control rats (CON) only received chow. Stool samples were analysed by 16 S rRNA gene amplicon sequencing. CD-fed rats became obese while TEL, PF and CON rats remained lean. Alpha diversity analyses indicated that bacterial diversity was similar before the study but changed over time. Beta diversity revealed a time-, CD- and telmisartan-dependent effect. The Firmicutes/Bacteroidetes ratio and the abundance of Blautia, Allobaculum and Parasutterella were higher in DIO and PF than in CON, but not in TEL. Enterotype (ET)-like clustering analyses, Kleinberg's hub network scoring and random forest analyses also indicated that telmisartan induced a specific signature of gut microbiota. In response to stool transfer from telmisartan-pre-treated donor to high-fat fed acceptor mice, body weight gain was slightly attenuated. We attribute the anti-obesity action of telmisartan treatment to diet-independent alterations in gut microbiota as the microbiota from telmisartan-treated, CD-fed rats clearly differed from those of DIO and PF rats. ET-like clustering network, random forest classification and the higher stability in bacterial co-occurrence network analyses indicate that there is more than one indicator species for telmisartan's specific signature, which is further strengthened by the fact that we cannot identify a single indicator species.


Angiotensin II Type 1 Receptor Blockers/pharmacology , Anti-Obesity Agents/pharmacology , Bacteria/drug effects , Gastrointestinal Microbiome/drug effects , Obesity/drug therapy , Telmisartan/pharmacology , Weight Gain/drug effects , Animals , Bacteria/growth & development , Diet/adverse effects , Disease Models, Animal , Dysbiosis , Fecal Microbiota Transplantation , Feces/microbiology , Mice , Obesity/etiology , Obesity/microbiology , Obesity/physiopathology , Rats , Rats, Sprague-Dawley
2.
J Cereb Blood Flow Metab ; 41(9): 2356-2369, 2021 09.
Article En | MEDLINE | ID: mdl-33730932

Angiotensin II receptor blockers (telmisartan) prevent rodents from diet-induced obesity and improve their metabolic status. Hyperglycemia and obesity are associated with reduced cerebral blood flow and neurovascular uncoupling which may lead to behavioral deficits. We wanted to know whether a treatment with telmisartan prevents these changes in obesity.We put young mice on high-fat diet and simultaneously treated them with telmisartan. At the end of treatment, we performed laser speckle imaging and magnetic resonance imaging to assess the effect on neurovascular coupling and cerebral blood flow. Different behavioral tests were used to investigate cognitive function.Mice developed diet-induced obesity and after 16, not 8 weeks of high-fat diet, however, the response to whisker pad stimulation was about 30% lower in obese compared to lean mice. Simultaneous telmisartan treatment increased the response again by 10% compared to obese mice. Moreover, telmisartan treatment normalized high-fat diet-induced reduction of cerebral blood flow and prevented a diet-induced anxiety-like behavior. In addition to that, telmisartan affects cellular senescence and string vessel formation in obesity.We conclude, that telmisartan protects against neurovascular unit impairments in a diet-induced obesity setting and may play a role in preventing obesity related cognitive deficits in Alzheimer's disease.


Angiotensin II Type 1 Receptor Blockers/therapeutic use , Anxiety/drug therapy , Diet, High-Fat/adverse effects , Obesity/physiopathology , Telmisartan/therapeutic use , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Disease Models, Animal , Male , Mice , Telmisartan/pharmacology
3.
Proc Natl Acad Sci U S A ; 117(3): 1753-1761, 2020 01 21.
Article En | MEDLINE | ID: mdl-31896584

Carbon dioxide (CO2), the major product of metabolism, has a strong impact on cerebral blood vessels, a phenomenon known as cerebrovascular reactivity. Several vascular risk factors such as hypertension or diabetes dampen this response, making cerebrovascular reactivity a useful diagnostic marker for incipient vascular pathology, but its functional relevance, if any, is still unclear. Here, we found that GPR4, an endothelial H+ receptor, and endothelial Gαq/11 proteins mediate the CO2/H+ effect on cerebrovascular reactivity in mice. CO2/H+ leads to constriction of vessels in the brainstem area that controls respiration. The consequential washout of CO2, if cerebrovascular reactivity is impaired, reduces respiration. In contrast, CO2 dilates vessels in other brain areas such as the amygdala. Hence, an impaired cerebrovascular reactivity amplifies the CO2 effect on anxiety. Even at atmospheric CO2 concentrations, impaired cerebrovascular reactivity caused longer apneic episodes and more anxiety, indicating that cerebrovascular reactivity is essential for normal brain function. The site-specific reactivity of vessels to CO2 is reflected by regional differences in their gene expression and the release of vasoactive factors from endothelial cells. Our data suggest the central nervous system (CNS) endothelium as a target to treat respiratory and affective disorders associated with vascular diseases.


Anxiety/metabolism , Cardiovascular System/metabolism , Endothelium/metabolism , Respiration Disorders/metabolism , Amygdala , Animals , Arterioles/pathology , Brain/physiology , Brain Stem/metabolism , Carbon Dioxide/metabolism , Central Nervous System/metabolism , Disease Models, Animal , Endothelium/pathology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression , Humans , Hypercapnia/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Respiration , Risk Factors , Signal Transduction
4.
Pflugers Arch ; 470(11): 1673-1689, 2018 11.
Article En | MEDLINE | ID: mdl-29978352

Obesity is a global health problem and treatment options are still insufficient. When chronically treated with the angiotensin II receptor blocker telmisartan (TEL), rodents do not develop diet-induced obesity (DIO). However, the underlying mechanism for this is still unclear. Here we investigated whether TEL prevents leptin resistance by enhancing leptin uptake across the blood-brain barrier (BBB). To address this question, we fed C57BL/6 mice a high-fat diet (HFD) and treated them daily with TEL by oral gavage. In addition to broadly characterizing the metabolism of leptin, we determined leptin uptake into the brain by measuring BBB transport of radioactively labeled leptin after long-term and short-term TEL treatment. Additionally, we assessed BBB integrity in response to angiotensin II in vitro and in vivo. We found that HFD markedly increased body weight, energy intake, and leptin concentration but that this effect was abolished under TEL treatment. Furthermore, glucose control and, most importantly, leptin uptake across the BBB were impaired in mice on HFD, but, again, both were preserved under TEL treatment. BBB integrity was not impaired due to angiotensin II or blocking of angiotensin II receptors. However, TEL did not exhibit an acute effect on leptin uptake across the BBB. Our results confirm that TEL prevents DIO and show that TEL preserves leptin transport and thereby prevents leptin resistance. We conclude that the preservation of leptin sensitivity is, however, more a consequence than the cause of TEL preventing body weight gain.


Angiotensin II Type 1 Receptor Blockers/therapeutic use , Anti-Obesity Agents/therapeutic use , Blood-Brain Barrier/metabolism , Leptin/metabolism , Obesity/drug therapy , Telmisartan/therapeutic use , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Anti-Obesity Agents/pharmacology , Blood-Brain Barrier/drug effects , Body Weight , Cell Line , Diet, High-Fat/adverse effects , Energy Metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Telmisartan/pharmacology
5.
Pharmacol Res ; 125(Pt A): 72-90, 2017 Nov.
Article En | MEDLINE | ID: mdl-28687340

Cardiovascular diseases (CVD) are among the main causes of death globally and in this context hypertension represents one of the key risk factors for developing a CVD. It is well established that the peripheral renin-angiotensin system (RAS) plays an important role in regulating blood pressure (BP). All components of the classic RAS can also be found in the brain but, in contrast to the peripheral RAS, how the endogenous RAS is involved in modulating cardiovascular effects in the brain is not fully understood yet. It is a complex system that may work differently in diverse areas of the brain and is linked to the peripheral system by the circumventricular organs (CVO), which do not have a blood brain barrier (BBB). In this review, we focus on the brain angiotensin peptides, their interactions with each other, and the consequences in the central nervous system (CNS) concerning cardiovascular control. Additionally, we present potential drug targets in the brain RAS for the treatment of hypertension.


Angiotensins/metabolism , Brain/physiopathology , Cardiovascular Diseases/pathology , Renin-Angiotensin System/physiology , Animals , Blood Pressure/physiology , Brain/metabolism , Cardiovascular Diseases/metabolism , Humans
...