Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Clin Oral Investig ; 28(2): 147, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38351377

RESUMO

BACKGROUND: Phosphorous necrosis of the jaw (PNJ) exhibits similar clinical and pathological features as medical-related osteonecrosis of the jaw (MRONJ). This study aims at comparing the similarities and differences between PNJ and MRONJ regarding pathological features and to provide a theoretical basis for the clinical diagnosis and management of PNJ. MATERIAL AND METHODS: A retrospective analysis was conducted to assess clinical differences among 38 PNJ patients and 31 MRONJ patients, who were diagnosed and treated between January 2009 and October 2022. Pathological alterations in bone tissue were evaluated using EDS, H&E, Masson, and TRAP staining on five specimens from both MRONJ and PNJ cases; furthermore, immunohistochemistry was used to determine the expression levels of OPG, RANKL, and Runx2. The mandibular coronoid process was removed from individuals with temporomandibular joint ankylosis to serve as a control. RESULTS: CBCT imaging demonstrated necrotic bone formation in block, strip, or plaque shapes. EDS analysis showed that the calcium/phosphorus ratio in the bone tissue of PNJ and MRONJ was significantly lower than that of the control group (P < 0.05). Additionally, staining indicated reduced osteoblast counts, disrupted bone trabecular structure, and decreased collagen fiber content in the bone tissues of PNJ and MRONJ. Immunohistochemistry demonstrated that RANKL expression was significantly lower in MRONJ compared to PNJ and control groups (P < 0.05). Conversely, Runx2 expression was significantly higher in PNJ than in MRONJ and control groups (P < 0.05), and there was no significant difference in OPG expression. CONCLUSION: PNJ and MRONJ demonstrate comparable clinical manifestations and pathological traits, although disparities may exist in their underlying exhibit comparable clinical manifestations, pathological traits, and molecular mechanisms.


Assuntos
Osteonecrose da Arcada Osseodentária Associada a Difosfonatos , Conservadores da Densidade Óssea , Transtornos da Articulação Temporomandibular , Humanos , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/patologia , Subunidade alfa 1 de Fator de Ligação ao Core , Estudos Retrospectivos , Arcada Osseodentária , Difosfonatos/uso terapêutico
2.
Oral Dis ; 2023 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-37498913

RESUMO

OBJECTIVE: To investigate the effects of key pathogenic genes involved in the development of jaw ameloblastoma (AB) and its associated extracellular matrix (ECM) on osteogenic differentiation in order to provide a theoretical foundation for future research into bone aggressiveness of AB. METHODS: The essential genes were identified by five AB patients for whole-exome sequencing and the microarray datasets GES38494 and GES132472. Moreover, the expression of key genes and their encoded proteins in AB tissues was explored. In addition, AB-derived the decellularized ECM (ABdECM) tissues were generated by the decellularization technique. Furthermore, the osteogenic development of periodontal ligament stem cells (PDLSCs) was mimicked by simulating the effects of the AB tumor microenvironment (TME). RESULTS: The AB essential genes including COL1A2, COL4A2, FBN1, and HPSE were discovered. Among them, the expression of HPSE was down-regulated, while that of COL1A2, COL4A2, and FBN1 was noticeably upregulated in AB compared with normal gingival tissues of the jaws. In vitro osteogenic differentiation of PDLSCs was suppressed by the ABdECM. CONCLUSIONS: Abnormal ECM proteins encoded by COL4A2, COL1A2, FBN1, and HPSE genes can cause disturbance in the ECM environment of AB and promote bone resorption.

3.
World J Emerg Med ; 14(3): 217-223, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37152534

RESUMO

BACKGROUND: Targeted temperature management (TTM), as a therapeutic temperature control strategy for cardiac arrest (CA), is recommended by guidelines. However, the relationship between post-rewarming fever (PRF) and the prognosis of CA patients is unclear. Therefore, we aim to summarize the studies regarding the influence of PRF on patients with CA. METHODS: EMBASE, PubMed, and Cochrane Central databases were searched from inception to March 13, 2022. Randomized clinical trials (RCTs) and cohort studies on PRF in CA patients were included. According to the heterogeneity, the meta-analysis was performed using a random effects model or fixed effects model to calculate the pooled odds ratios (ORs) and corresponding 95% confidence intervals (CI s). The outcome data were unfavorable neurological outcome and mortality. RESULTS: The meta-analysis included 11 observational studies involving 3,246 patients. The results of the meta-analysis show that PRF (body temperature >38.0 °C) has no effect on the neurological outcome of CA patients (OR 0.71, 95% CI 0.43-1.17, I 2 82%) and has a significant relationship with lower mortality (OR 0.63; 95% CI 0.49-0.80, I 2 39%). However, PRF with a stricter definition (body temperature >38.5 °C ) was associated with worse neurological outcome (OR 1.44, 95% CI 1.08-1.92, I 2 45%) and higher mortality (OR 1.71, 95% CI 1.25-2.35, I 2 47%). CONCLUSION: This study suggests that PRF >38.0 °C may not affect the neurological outcome and have a lower mortality in CA patients who completed TTM. However, PRF >38.5 °C is a potential prognostic factor for worse outcomes in CA patients.

4.
Acta Neurol Belg ; 123(4): 1331-1337, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36922484

RESUMO

Acute lung injury or acute respiratory distress syndrome (ALI/ARDS) is a common complication after aneurysmal subarachnoid hemorrhage (aSAH), and is associated with worse neurologic outcomes and longer hospitalization. However, the effect of ALI/ARDS in SAH has not been well elucidated. The purpose of this study was to determine the incidence of ALI/ARDS in a cohort of patients with SAH and to determine the risk factors for ALI/ARDS and their impact on patient prognosis. We performed a retrospective analysis of 167 consecutive patients with aSAH enrolled. ALI/ARDS patients were rigorously adjudicated using North American-European Consensus Conference definition. Regression analyses were used to test the risk factors for ALI/ARDS in patients with SAH. A total of 167 patients fulfilled the inclusion criteria, and 27% patients (45 of 167) developed ALI. Among all 45 ALI patients, 33 (20%, 33 of 167) patients met criteria for ARDS. On multivariate analysis, elderly patients, lower glasgow coma scale (GCS), higher Hunt-Hess grade, higher simplified acute physiology score (SAPS) II score, pre-existing pneumonia, gastric aspiration, hypoxemia, and tachypnea were the strongest risk factor for ALI/ARDS. Patients with ALI/ARDS showed worse clinical outcomes measured at 30 days. Development of ALI/ARDS was associated with a statistically significant increasing the odds of tracheostomy and hospital complications, and increasing duration of mechanical ventilation, intensive care unit (ICU) length and hospitalization stay. Development of ALI/ARDS is a severe complication of SAH and is associated with a poor clinical outcome, and further studies should focus on both prevention and management strategies specific to SAH-associated ALI/ARDS.


Assuntos
Lesão Pulmonar Aguda , Síndrome do Desconforto Respiratório , Hemorragia Subaracnóidea , Humanos , Idoso , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/epidemiologia , Estudos Retrospectivos , Síndrome do Desconforto Respiratório/epidemiologia , Síndrome do Desconforto Respiratório/etiologia , Prognóstico , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/complicações
5.
Biophys J ; 122(7): 1198-1210, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-36772795

RESUMO

Cancer stem cells (CSCs) are known to have a high capacity for tumor initiation and the formation of metastases. We have previously shown that in collagen constructs mimetic of aligned extracellular matrix architectures observed in carcinomas, breast CSCs demonstrate enhanced directional and total motility compared with more differentiated carcinoma populations. Here, we show that CSCs maintain increased motility in diverse environments including on 2D elastic polyacrylamide gels of various stiffness, 3D randomly oriented collagen matrices, and ectopic cerebral slices representative of a common metastatic site. A consistent twofold increase of CSC motility across platforms suggests a general shift in cell migration mechanics between well-differentiated carcinoma cells and their stem-like counterparts. To further elucidate the source of differences in migration, we demonstrate that CSCs are less contractile than the whole population (WP) and develop fewer and smaller focal adhesions and show that enhanced CSC migration can be tuned via contractile forces. The WP can be shifted to a CSC-like migratory phenotype using partial myosin II inhibition. Inversely, CSCs can be shifted to a less migratory WP-like phenotype using microtubule-destabilizing drugs that increase contractility or by directly enhancing contractile forces. This work begins to reveal the mechanistic differences driving CSC migration and raises important implications regarding the potentially disparate effects of microtubule-targeting agents on the motility of different cell populations.


Assuntos
Carcinoma , Colágeno , Humanos , Linhagem Celular Tumoral , Colágeno/metabolismo , Movimento Celular , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Carcinoma/metabolismo , Carcinoma/patologia
6.
Mol Med Rep ; 24(6)2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34664679

RESUMO

Cerebral vasospasm (CVS) is a common complication of subarachnoid hemorrhage (SAH) with high deformity rates and cerebral vascular smooth muscle cells (VSMCs) phenotypic switch is considered to be involved in the regulation of CVS. However, to the best of the authors' knowledge, its underlying molecular mechanism remains to be elucidated. Peroxisome proliferator­activated receptor ß/δ (PPARß/δ) has been demonstrated to be involved in the modulation of vascular cells proliferation and maintains the autoregulation function of blood vessels. The present study investigated the potential effect of PPARß/δ on CVS following SAH. A model of SAH was established by endovascular perforation on male adult Sprague­Dawley rats, and the adenovirus PPARß/δ (Ad­PPARß/δ) was injected via intracerebroventricular administration prior to SAH. The expression levels of phenotypic markers α­smooth muscle actin and embryonic smooth muscle myosin heavy chain were measured via western blotting or immunofluorescence staining. The basilar artery diameter and vessel wall thickness were evaluated under fluorescence microscopy. SAH grade, neurological scores, brain water content and brain swelling were measured to study the mechanisms of PPARß/δ on vascular smooth muscle phenotypic transformation. It was revealed that the expression levels of synthetic proteins were upregulated in rats with SAH and this was accompanied by CVS. Activation of PPARß/δ using Ad­PPARß/δ markedly upregulated the contractile proteins elevation, restrained the synthetic proteins expression and attenuated SAH­induced CVS by regulating the phenotypic switch in VSMCs at 72 h following SAH. Furthermore, the preliminary study demonstrated that PPARß/δ downregulated ERK activity and decreased the expression of phosphorylated (p­)ETS domain­containing protein Elk­1 and p­p90 ribosomal S6 kinase, which have been demonstrated to serve an important role in VSMC phenotypic change. Additionally, it was revealed that Ad­PPARß/δ could positively improve CVS by ameliorating the diameter of the basilar artery and mitigating the thickness of the vascular wall. Furthermore, subsequent experiments demonstrated that Ad­PPARß/δ markedly reduced the brain water content and brain swelling and improved the neurological outcome. Taken together, the present study identified PPARß/δ as a useful regulator for the VSMCs phenotypic switch and attenuating CVS following SAH, thereby providing novel insights into the therapeutic strategies of delayed cerebral ischemia.


Assuntos
Músculo Liso Vascular/metabolismo , PPAR delta/metabolismo , PPAR beta/metabolismo , Hemorragia Subaracnóidea/metabolismo , Vasoespasmo Intracraniano/metabolismo , Actinas/metabolismo , Animais , Edema Encefálico/genética , Edema Encefálico/metabolismo , Modelos Animais de Doenças , Sistema de Sinalização das MAP Quinases/genética , Masculino , Miócitos de Músculo Liso/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Doenças do Sistema Nervoso/complicações , Doenças do Sistema Nervoso/genética , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/mortalidade , Vasoespasmo Intracraniano/complicações , Vasoespasmo Intracraniano/genética , Proteínas Elk-1 do Domínio ets/metabolismo
7.
IUBMB Life ; 73(10): 1244-1256, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34318585

RESUMO

The 1-year mortality and health consequences of COVID-19 in cancer patients are relatively underexplored. In this multicenter cohort study, 166 COVID-19 patients with cancer were compared with 498 non-cancer COVID-19 patients and 498 non-COVID cancer patients. The 1-year all-cause mortality and hospital mortality rates in Cancer COVID-19 Cohort (30% and 20%) were significantly higher than those in COVID-19 Cohort (9% and 8%, both P < .001) and Cancer Cohort (16% and 2%, both P < 0.001). The 12-month all-cause post-discharge mortality rate in survival discharged Cancer COVID-19 Cohort (8%) was higher than that in COVID-19 Cohort (0.4%, P < .001) but similar to that in Cancer Cohort (15%, P = .084). The incidence of sequelae in Cancer COVID-19 Cohort (23%, 26/114) is similar to that in COVID-19 Cohort (30%, 130/432, P = .13). The 1-year all-cause mortality was high among patients with hematologic malignancies (59%), followed by those who have nasopharyngeal, brain, and skin tumors (45%), digestive system neoplasm (43%), and lung cancers (32%). The rate was moderate among patients with genitourinary (14%), female genital (13%), breast (11%), and thyroid tumors (0). COVID-19 patients with cancer showed a high rate of in-hospital mortality and 1-year all-cause mortality, but the 12-month all-cause post-discharge mortality rate in survival discharged cancer COVID-19 patients was similar to that in Cancer Cohort. Comparing to COVID-19 Cohort, risk stratification showed that hematologic, nasopharyngeal, brain, digestive system, and lung tumors were high risk (44% vs 9%, P < 0.001), while genitourinary, female genital, breast, and thyroid tumors had moderate risk (10% vs 9%, P = .85) in COVID-19 Cancer Cohort. Different tumor subtypes had different effects on COVID-19. But if cancer patients with COVID-19 manage to survive their COVID-19 infections, then long-term mortality appears to be similar to the cancer patients without COVID-19, and their long-term clinical sequelae were similar to the COVID-19 patients without cancer.


Assuntos
COVID-19/mortalidade , Neoplasias/complicações , Idoso , COVID-19/complicações , COVID-19/virologia , Estudos de Coortes , Feminino , Mortalidade Hospitalar , Humanos , Masculino , SARS-CoV-2/isolamento & purificação
9.
Cell Death Dis ; 12(1): 9, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33414448

RESUMO

The level of microRNA-9-5p (miRNA-9-5p) in brain tissues is significantly changed in the chronic phase after traumatic brain injury (TBI). However, the effect of miRNA-9-5p on brain function after TBI has not been elucidated. In this study, we used a controlled cortical impact (CCI) model to induce TBI in Sprague-Dawley rats. Brain microvascular endothelial cells (BMECs), astrocytes, and neurons were extracted from immature Sprague-Dawley rats and cocultured to reconstruct the neurovascular unit (NVU) in vitro. The results showed that downregulation of miRNA-9-5p in the chronic phase contributed to neurological function recovery by promoting astrocyte proliferation and increasing the release of astrocyte-derived neurotrophic factors around injured brain tissues after TBI. A dual-luciferase reporter assay validated that miRNA-9-5p was a post-transcriptional modulator of thrombospondin 2 (Thbs-2), and downregulation of miRNA-9-5p promoted Thbs-2 expression in astrocytes. Furthermore, we verified that Thbs-2 can promote Notch pathway activation by directly binding to Jagged and Notch. Through in vitro experiments, we found that the expression of synaptic proteins and the number of synaptic bodies were increased in neurons in the NVU, which was constructed using astrocytes pretreated with miRNA-9-5p inhibitor. Moreover, we also found that downregulation of miRNA-9-5p promoted Thbs-2 expression in astrocytes, which activated the Notch/cylindromatosis/transforming growth factor-ß-activated kinase 1 pathway in neurons and promoted the expression of synaptic proteins, including post-synaptic density protein 95 and synaptotagmin. Based on these results, miRNA-9-5p may be a new promising prognostic marker and treatment target for TBI.


Assuntos
Lesões Encefálicas Traumáticas/genética , MicroRNAs/metabolismo , Sinapses/metabolismo , Animais , Regulação para Baixo , Ratos , Ratos Sprague-Dawley
10.
FASEB J ; 35(1): e21157, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33155736

RESUMO

The angiotensin II type 1 receptor (AT1R) signaling pathway is reported to modulate glucose metabolism. Targeting AT1R, our group invented ATRQß-001 vaccine, a novel immunotherapeutic strategy to block the activation of AT1R. Here, we evaluated the therapeutic efficacy of ATRQß-001 vaccine in insulin resistance, and investigated the mechanism. Our results showed that ATRQß-001 vaccine and specific monoclonal antibody against epitope ATR-001 (McAb-ATR) decreased fasting serum insulin concentration and improved glucose and insulin tolerance in ob/ob mice. These beneficial effects were verified in high-fat diet-induced obese mice. McAb-ATR activated insulin signaling in skeletal muscle and insulin-resistant C2C12 myotubes without affecting liver or white adipose tissue of ob/ob mice. Mechanistically, the favorable impact of McAb-ATR on insulin resistance was abolished in db/db mice and in C2C12 myotubes with leptin receptor knockdown. AT1R knockdown also eradicated the effects of McAb-ATR in C2C12 myotubes. Furthermore, McAb-ATR treatment was able to activate the leptin receptor-mediated JAK2/STAT3 signaling in skeletal muscle of ob/ob mice and C2C12 myotubes. Additionally, angiotensin II downregulated the leptin signaling in skeletal muscle of ob/ob and diet-induced obese mice. We demonstrated that ATRQß-001 vaccine and McAb-ATR improved whole-body insulin resistance and regulated glucose metabolism in skeletal muscle in a leptin receptor-dependent manner. Our data suggest that immunotherapy targeting AT1R is a novel strategy for treating insulin resistance.


Assuntos
Imunoterapia , Resistência à Insulina/imunologia , Receptores de Angiotensina/imunologia , Receptores para Leptina/imunologia , Vacinas de Partículas Semelhantes a Vírus/farmacologia , Animais , Masculino , Camundongos , Vacinas de Partículas Semelhantes a Vírus/imunologia
11.
Front Neurosci ; 14: 685, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32733194

RESUMO

Subarachnoid hemorrhage (SAH) is a complicated clinical syndrome, which is caused by several kinds of cerebrovascular disorders, with high morbidity, disability and mortality rate. In recent years, several studies have shown that early brain injury (EBI) is an important factor leading to the poor prognosis of SAH. A major cause of EBI has been attributed that hematoma components invade into the brain parenchyma, resulting in neuronal cell death. Therefore, the clearance of hematoma components is essential in the clinical outcome of patients after SAH. Here, in the review, we provide a summary of the current known hematoma components clearance mechanisms and simultaneously propose a new hypothesis for hematoma components clearance.

12.
J Neurochem ; 153(6): 710-726, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31951014

RESUMO

The level of microRNA-9-5p (miRNA-9-5p) in brain tissues is significantly changed after traumatic brain injury (TBI). However, the effect of miRNA-9-5p for brain function in TBI has not been elucidated. In this study, a controlled cortical impact model was used to induce TBI in Sprague-Dawley rats, and an oxygen glucose deprivation model was used to mimic the pathological state in vitro. Brain microvascular endothelial cells (BMECs) and astrocytes were extracted from immature Sprague-Dawley rats and cocultured to reconstruct blood-brain barrier (BBB) in vitro. The results show that the level of miRNA-9-5p was significantly increased in brain tissues after TBI, and up-regulation of miRNA9-5p contributed to the recovery of neurological function. Up-regulation of miRNA-9-5p with miRNA agomir may significantly alleviate apoptosis, neuroinflammation, and BBB damage in rats after TBI. Moreover, a dual luciferase reporter assay confirmed that miRNA-9-5p is a post-transcriptional modulator of Ptch-1. In in vitro experiments, the results confirmed that up-regulation of miRNA-9-5p with miRNA mimic alleviates cellular apoptosis, inflammatory response, and BBB damage mainly by inhibiting Ptch-1. In addition, we found that the activation of Hedgehog pathway was accompanied by inhibition of NF-κB/MMP-9 pathway in the BMECs treated with miRNA-9-5p mimic. Taken together, these results indicate that up-regulation of miRNA-9-5p alleviates BBB damage and neuroinflammatory responses by activating the Hedgehog pathway and inhibiting NF-κB/MMP-9 pathway, which promotes the recovery of neurological function after TBI.


Assuntos
Barreira Hematoencefálica/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Mediadores da Inflamação/metabolismo , MicroRNAs/biossíntese , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/patologia , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Proteínas Hedgehog/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Masculino , MicroRNAs/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
13.
Br J Pharmacol ; 177(2): 402-419, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31625597

RESUMO

BACKGROUND AND PURPOSE: Hypertension has been the leading preventable cause of premature death worldwide. The aim of this study was to design a more efficient vaccine against novel targets for the treatment of hypertension. EXPERIMENTAL APPROACH: The epitope CE12, derived from the human L-type calcium channel (CaV 1.2), was designed and conjugated with Qß bacteriophage virus-like particles to test the efficacy in hypertensive animals. Further, the hepatitis B core antigen (HBcAg)-CE12-CQ10 vaccine, a bivalent vaccine based on HBcAg virus-like particles and targeting both human angiotensin AT1 receptors and CaV 1.2 channels, was developed and evaluated in hypertensive rodents. KEY RESULTS: The Qß-CE12 vaccine effectively decreased the BP in hypertensive rodents. A monoclonal antibody against CE12 specifically bound to L-type calcium channels and inhibited channel activity. Injection with monoclonal antibody against CE12 effectively reduced the BP in angiotensin II-induced hypertensive mice. The HBcAg-CE12-CQ10 vaccine showed antihypertensive effects in hypertensive mice and relatively superior antihypertensive effects in spontaneously hypertensive rats and ameliorated L-NAME-induced renal injury. In addition, no obvious immune-mediated damage or electrophysiological adverse effects were detected. CONCLUSION AND IMPLICATIONS: Immunotherapy against both AT1 receptors and CaV 1.2 channels decreased the BP in hypertensive rodents effectively and provided protection against hypertensive target organ damage without obvious feedback activation of renin-angiotensin system or induction of dominant antibodies against the carrier protein. Thus, the HBcAg-CE12-CQ10 vaccine may provide a novel and promising therapeutic approach for hypertension.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Canais de Cálcio Tipo L/imunologia , Hipertensão/prevenção & controle , Receptor Tipo 1 de Angiotensina/imunologia , Vacinas Combinadas/farmacologia , Vacinas de Partículas Semelhantes a Vírus/farmacologia , Angiotensina II , Animais , Canais de Cálcio Tipo L/metabolismo , Modelos Animais de Doenças , Epitopos , Hipertensão/imunologia , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , Camundongos Endogâmicos BALB C , Ratos Endogâmicos SHR , Receptor Tipo 1 de Angiotensina/metabolismo , Vacinação
14.
J Am Heart Assoc ; 8(18): e012341, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31512549

RESUMO

Background We have developed a peptide vaccine named ATRQß-001, which was proved to retard signal transduction initiated by angiotensin II (Ang II). Ang II was implicated in abdominal aortic aneurysm (AAA) progression, but whether the ATRQß-001 vaccine would prevent AAA is unknown. Methods and Results Ang II-infused ApoE-/- mice and calcium phosphate-induced AAA in C57BL/6 mice were used to verify the efficiency of ATRQß-001 vaccine in AAA. Results demonstrated that the vaccine effectively restrained the aneurysmal dilation and vascular wall destruction of aorta in both animal models, beyond anti-hypertensive effects. In Ang II-induced AAA vascular sections, Immunohistochemical staining showed that the vaccine notably constrained vascular inflammation and vascular smooth muscle cell (VSMC) phenotypic transition, concurrently reduced macrophages infiltration. In cultured VSMC, the anti-ATR-001 antibody inhibited osteopontin secretion induced by Ang II, thereby impeded macrophage migration while co-culture. Furthermore, metalloproteinases and other matrix proteolytic enzymes were also found to be limited by the vaccine in vivo and in vitro. Conclusions ATRQß-001 vaccine prevented AAA initiation and progression in both Ang II and calcium phosphate-induced AAA models. And the beneficial effects were played beyond decrease of blood pressure, which provided a novel and promising method to take precautions against AAA.


Assuntos
Aorta/efeitos dos fármacos , Aneurisma da Aorta Abdominal/patologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Vacinas de Partículas Semelhantes a Vírus/farmacologia , Angiotensina II/toxicidade , Animais , Aorta/patologia , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/metabolismo , Fosfatos de Cálcio/toxicidade , Modelos Animais de Doenças , Inflamação , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Camundongos Knockout para ApoE , Músculo Liso Vascular/citologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Osteopontina/efeitos dos fármacos , Osteopontina/metabolismo , Distribuição Aleatória , Vasoconstritores/toxicidade
15.
J Am Coll Cardiol ; 73(20): 2567-2580, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31118151

RESUMO

BACKGROUND: Pulmonary arterial hypertension (PAH) is a chronic fatal disease. The treatment of PAH is less than ideal and the control is far from satisfactory worldwide. Vaccination provides a promising approach for treatment of PAH. OBJECTIVES: This study sought to find a vaccine against endothelin-1 (ET-1) receptor type A (ETAR) for treating PAH. METHODS: The ETRQß-002 vaccine was screened and the specific antibodies against epitope ETR-002 belonging to the second extracellular loop of ETAR (including the polyclonal and monoclonal antibody) were produced. The effect of the antibodies on Ca2+-dependent signal transduction events was investigated. In vivo, ETRQß-002 vaccine was used to vaccinate monocrotaline (MCT)- and Sugen/hypoxia-induced pulmonary hypertension animals. The monoclonal antibody (mAb) against ETR-002 was also injected into the PAH animals. The effect of ETRQß-002 vaccine on pulmonary hypertension and remodeling of pulmonary arterioles and right ventricle (RV) was carefully evaluated. Further, the possible immune-mediated damage was detected in normal vaccinated animals. RESULTS: ETR-002 peptide has perfect immunogenicity and ETRQß-002 vaccine could induce strong antibody production. In vitro, the anti-ETR-002 antibody bound to ETAR and inhibited Ca2+-dependent signal transduction events, including extracellular signal-regulated kinase phosphorylation and elevation of intracellular Ca2+ concentration induced by ET-1. In vivo, both ETRQß-002 vaccine and the mAb significantly decreased the RV systolic pressure up to 20 mm Hg and 10 mm Hg in MCT-exposed rats and Sugen/hypoxia-exposed mice, respectively. Also, ETRQß-002 vaccine/mAb obviously ameliorated pathological remodeling of pulmonary arterioles and hypertrophy of the RV in PAH animals. Additionally, no significant immune-mediated damage was detected in vaccinated animals. CONCLUSIONS: ETRQß-002 vaccine/mAb attenuated remodeling of pulmonary arterioles and RV in MCT- and Sugen/hypoxia-induced PAH animals and decreased RV systolic pressure effectively through diminishing the pressure response and inhibiting signal transduction initiated by ET-1. ETRQß-002 vaccine/mAb may provide a novel and promising method for PAH treatment.


Assuntos
Hipertensão Arterial Pulmonar/terapia , Artéria Pulmonar/ultraestrutura , Receptor de Endotelina A/imunologia , Vacinação/métodos , Vacinas de Subunidades/uso terapêutico , Animais , Anticorpos Monoclonais/imunologia , Células Cultivadas , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Imunoterapia/métodos , Masculino , Microscopia Eletrônica de Transmissão , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/fisiopatologia , Pressão Propulsora Pulmonar , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina A/metabolismo
16.
Hypertens Res ; 42(3): 329-340, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30587854

RESUMO

We invented the ATRQß-001 hypertension vaccine, which targeted angiotensin II type 1 receptor (AT1R) and showed a desirable blocking effect for AT1R. The purpose of this study was to investigate whether the ATRQß-001 vaccine could improve cardiac function and prevent cardiac remodeling after acute myocardial infarction (AMI). C57BL/6 male mice were randomly assigned into four groups: sham + VLP, MI + VLP, MI + ATRQß-001, and MI + valsartan. Mice were administered Qß virus-like particle (Qß-VLP, 100 µg/time), ATRQß-001 vaccine (100 µg/time), and valsartan (6 mg/kg/day) before AMI, which was induced by permanently ligating the left anterior descending coronary artery. The effect of the ATRQß-001 vaccine on cardiac function and cardiac remodeling was observed by following up for 1 week, 4 weeks, and 12 weeks post MI. The ATRQß-001 vaccine significantly reduced sudden cardiac death and increased survival rates (compared with MI + VLP, 80% versus 55% and mean estimate (days) 68.4 ± 7.0 versus 47.8 ± 8.9, respectively; p = 0.046) post MI. Echocardiography showed that the ATRQß-001 vaccine remarkably improved cardiac function (left ventricular ejection fraction, 24.8 ± 7.0% versus 13.2 ± 3.8%, p = 0.005) post MI. Histological analysis revealed that the ATRQß-001 vaccine obviously mitigated myocardial inflammation, apoptosis, and fibrosis after AMI. Further, the ATRQß-001 vaccine significantly inhibited the TGF-ß1/Smad2/3 signaling pathway. Assessment of the renin-angiotensin system (RAS) demonstrated that the ATRQß-001 vaccine did not cause obvious feedback of circulating RAS, but prominently attenuated the expression of AT1R, compared with the other groups at 4 and 12 weeks after AMI. In conclusion, the ATRQß-001 vaccine decreased mortality and improved cardiac function and remodeling after AMI.


Assuntos
Coração/fisiopatologia , Hipertensão/terapia , Infarto do Miocárdio/fisiopatologia , Receptor Tipo 1 de Angiotensina/imunologia , Vacinas de Partículas Semelhantes a Vírus/uso terapêutico , Vacinas/uso terapêutico , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Animais , Anti-Hipertensivos/uso terapêutico , Ecocardiografia , Coração/diagnóstico por imagem , Hipertensão/diagnóstico por imagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/diagnóstico por imagem , Sistema Renina-Angiotensina/efeitos dos fármacos , Análise de Sobrevida , Valsartana/uso terapêutico , Remodelação Ventricular
17.
Int J Med Sci ; 15(10): 1072-1082, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30013449

RESUMO

Background: As a major antioxidant in serum, uric acid (UA) was once considered only as the leading cause of gout; however, recent studies have validated its neuroprotective role in ischemic stroke. Because the potential protective effects of UA in traumatic brain injury (TBI) remain largely unknown, this study investigated the role of UA in TBI in both clinical patients and experimental animals. Methods: In TBI patients, serum UA concentrations were measured within 3 days after injury. Clinical outcomes at discharge were classified according to the Glasgow Outcome Scale: good outcome (4-5) and poor outcome (1-3). Risk factors for good outcome were identified via backward logistic regression analysis. For the animal study, a controlled cortical impact (CCI) injury model was established in mice. These mice were given UA at different doses intraperitoneally, and subsequent UA concentrations in mouse serum and brain tissue were determined. Neurological function, oxidative stress, inflammatory response, neuronal maintenance, cerebral blood flow, and lesion size were also assessed. Results: The serum UA level was significantly lower in TBI patients who had a good outcome (P<0.01), and low serum UA was an independent predictor of good outcome after TBI (P<0.01; odds ratio, 0.023; 95% confidence interval, 0.006-0.082). Consistently, decreased levels of serum UA were observed in both TBI patients and CCI animals (P<0.05), whereas the UA concentration was increased in CCI brain tissue (P<0.05). Administration of UA further increased the UA level in brain tissue as compared to that in control animals (P<0.05). Among the different doses administered, 16 mg/kg UA improved sensorimotor functional recovery, spatial learning, and memory in CCI mice (P<0.05). Moreover, oxidative stress and the inflammatory response were inhibited by UA treatment (P<0.05). UA treatment also improved neuronal maintenance and cortical blood flow (P<0.05) but not lesion size (P>0.05). Conclusions: UA acted to attenuate neuronal loss, cerebral perfusion impairment and neurological deficits in TBI mice through suppression of neuronal and vascular oxidative stress. Following TBI, active antioxidant defense in the brain may result in consumption of UA in the serum, and thus, a decreased serum UA level could be predictive of good clinical recovery.


Assuntos
Lesões Encefálicas Traumáticas , Ácido Úrico , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Ácido Úrico/sangue , Ácido Úrico/urina
18.
Neurobiol Dis ; 117: 114-124, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29886067

RESUMO

Traumatic brain injury (TBI) causes a high rate of mortality and disability worldwide, and there exists almost none effective drugs to protect against TBI. Neurotoxicity occurring after TBI can be derived from microglia and astrocytes, and causes neuronal death and synapse loss. Bexarotene has been demonstrated to protect neurons in CNS diseases. In the present study, we aimed to investigate the potential role of bexarotene in protecting against neurotoxicity after TBI, as well as the underlying mechanism. The controlled cortical impact (CCI) model was established on adult C57BL/6 mice, followed by intraperitoneal administration of bexarotene for 14 consecutive days. We found that bexarotene improved sensorimotor function and cognitive recovery in CCI mice. In addition, bexarotene decreased neuronal death and synapse loss, as well as inhibited apoptotic cascade. Moreover, bexarotene treatment reduced M1 microglia polarization, microglia-derived pro-inflammatory cytokines, and the number of A1 astrocytes after CCI. These effects of bexarotene were partially abolished by T0070907, an antagonist of peroxisome proliferator-activated receptor gamma (PPARγ). Additionally, bexarotene enhanced nuclear translocation and transcriptional activity of PPARγ. These findings show that bexarotene inhibits neurotoxicity in mice after TBI, at least in part through a PPARγ-dependent mechanism.


Assuntos
Bexaroteno/uso terapêutico , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , PPAR gama/metabolismo , Animais , Benzamidas/toxicidade , Bexaroteno/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Piridinas/toxicidade
19.
Sci Rep ; 7(1): 12534, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28970592

RESUMO

Vaccination provides a promising approach for treatment of hypercholesterolemia and improvement in compliance. In this study, the appropriate virus-like particle (VLP)-peptide vaccines targeting proprotein convertase subtilisin/kexin type 9 (PCSK9) were screened. The screening criteria of target peptides were as follows: (1) located in catalytic domain of PCSK9, or regulating the binding of PCSK9 and LDL receptors (LDLR); (2) having low/no-similarity when matched with the host proteome; (3) possessing ideal antigenicity and hydrophilicity; (4) including the functional mutation site of PCSK9. It was found that mice vaccinated with VLP -PCSK9 peptide vaccines, especially PCSK9Qß-003 vaccine, developed high titer IgG antibodies against PCSK9. PCSK9Qß-003 vaccine obviously decreased plasma total cholesterol in both Balb/c mice and LDLR+/- mice. Also, PCSK9Qß-003 vaccine decreased plasma PCSK9 level and up-regulated LDLR expression in liver. Additionally, PCSK9Qß-003 vaccine injection was associated with significant up-regulation of sterol-regulatory element-binding protein-2 (SREBP-2), hepatocyte nuclear factor 1α (HNF-1α), and 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase in LDLR+/- mice. No obvious immune injury was detected in vaccinated animals. The PCSK9Qß-003 vaccine, therefore, may be an attractive treatment approach for hypercholesterolemia through decreasing cholesterol and regulating lipid homeostasis.


Assuntos
Hipercolesterolemia/tratamento farmacológico , Pró-Proteína Convertase 9/genética , Vacinas de Subunidades/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Animais , Domínio Catalítico/efeitos dos fármacos , Colesterol/sangue , Modelos Animais de Doenças , Regulação da Expressão Gênica , Fator 1-alfa Nuclear de Hepatócito/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Hipercolesterolemia/genética , Hipercolesterolemia/imunologia , Hipercolesterolemia/prevenção & controle , Imunogenicidade da Vacina/genética , Imunogenicidade da Vacina/imunologia , Imunoglobulina G/sangue , Camundongos , Camundongos Knockout , Mutação , Inibidores de PCSK9 , Fosfoproteínas Fosfatases/genética , Receptores de LDL/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/genética
20.
Sci Rep ; 7(1): 12580, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28974760

RESUMO

We developed a virus-like particle (VLP)-based therapeutic vaccine against angiotensin II receptor type 1, ATR-AP205-001, which could significantly reduce the blood pressure and protect target organs of hypertensive animals. In this study, we focused on the immunological effect and safety of the VLP-based vaccine. By comparing to the depolymerized dimeric vaccine ATR-Dimer-001, we found that ATR-AP205-001 reached subcapsular sinus of lymph node shortly after administration, followed by accumulation on follicle dendritic cells via follicle B cell transportation, while ATR-Dimer-001 vaccine showed no association with FDCs. ATR-AP205-001 vaccine strongly activated dendritic cells, which promoted T cells differentiation to follicular helper T cells. ATR-AP205-001 vaccine induced powerful germinal center reaction, which was translated to a boost of specific antibody production and long-lasting B cell memory, far superior to ATR-Dimer-001 vaccine. Moreover, neither cytotoxic T cells, nor Th1/Th17 cell-mediated inflammation was observed in ATR-AP205-001 vaccine, similar to ATR-Dimer-001 vaccine. We concluded that ATR-AP205-001 vaccine quickly induced potent humoral immunity through collaboration of B cells, follicular dendritic cells and follicular helper T cells, providing an effective and safe intervention for hypertension in the future clinical application.


Assuntos
Hipertensão/tratamento farmacológico , Imunidade Inata/efeitos dos fármacos , Imunoconjugados/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Animais , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Centro Germinativo/efeitos dos fármacos , Centro Germinativo/imunologia , Humanos , Hipertensão/imunologia , Imunidade Inata/imunologia , Imunoconjugados/genética , Imunoconjugados/imunologia , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/patologia , Camundongos , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia , Vacinas Conjugadas/administração & dosagem , Vacinas Conjugadas/efeitos adversos , Vacinas de Partículas Semelhantes a Vírus/efeitos adversos , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...