Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Bone Miner Res ; 37(2): 215-225, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34633122

RESUMEN

The possible mechanisms underlying the quantitative and qualitative effects of cinacalcet on bone were explored in a chronic kidney disease-mineral and bone disorder (CKD-MBD) mouse model in relation to the influence of the interactions among the osteoclast (OC) endoplasmic reticulum (ER) stress, autophagy and apoptosis pathways on OC differentiation. Body weight and biochemical parameters improved significantly in the CKD + cinacalcet groups compared to the CKD group. Micro-computed tomography (µCT) revealed both cortical and trabecular parameters deteriorated significantly in the CKD group and were reversed by cinacalcet in a dose-dependent manner. Nanoindentation analysis of bone quality proved that both cortical hardness and elastic modulus improved significantly with high dose cinacalcet treatment. In vitro studies revealed that cinacalcet inhibited receptor activator of NF-κB ligand (RANKL)/receptor activator of NF-κB (RANK)-induced OC differentiation in a concentration-dependent manner through a close interaction between activation of caspase-related apoptosis, reversal of OC autophagy through the protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and adenosine monophosphate-activated protein kinase (AMPK) pathways, and attenuation of the OC ER stress/CREBH/NFATc1 signaling pathway. Cinacalcet improves both bone quantity and bone quality in CKD mouse model and inhibits OC differentiation through regulation of the interactions among the apoptosis, ER stress, and autophagy pathways within OCs. © 2021 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica , Osteoclastos , Animales , Autofagia , Diferenciación Celular , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/tratamiento farmacológico , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/metabolismo , Cinacalcet/farmacología , Cinacalcet/uso terapéutico , Estrés del Retículo Endoplásmico , Ratones , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Microtomografía por Rayos X
2.
Int J Mol Sci ; 22(20)2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34681927

RESUMEN

Uremic toxins, such as indoxyl sulfate (IS) and kynurenine, accumulate in the blood in the event of kidney failure and contribute to further bone damage. To maintain the homeostasis of the skeletal system, bone remodeling is a persistent process of bone formation and bone resorption that depends on a dynamic balance of osteoblasts and osteoclasts. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates the toxic effects of uremic toxins. IS is an endogenous AhR ligand and is metabolized from tryptophan. In osteoclastogenesis, IS affects the expression of the osteoclast precursor nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) through AhR signaling. It is possible to increase osteoclast differentiation with short-term and low-dose IS exposure and to decrease differentiation with long-term and/or high-dose IS exposure. Coincidentally, during osteoblastogenesis, through the AhR signaling pathway, IS inhibits the phosphorylation of ERK, and p38 reduces the expression of the transcription factor 2 (Runx2), disturbing osteoblastogenesis. The AhR antagonist resveratrol has a protective effect on the IS/AhR pathway. Therefore, it is necessary to understand the multifaceted role of AhR in CKD, as knowledge of these transcription signals could provide a safe and effective method to prevent and treat CKD mineral bone disease.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Indicán/toxicidad , Osteoblastos/citología , Osteoclastos/citología , Receptores de Hidrocarburo de Aril/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Indicán/orina , Factores de Transcripción NFATC/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/orina , Transducción de Señal/efectos de los fármacos
3.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34445700

RESUMEN

Severe acute respiratory syndrome coronavirus 2 is a new, highly pathogenic virus that has recently elicited a global pandemic called the 2019 coronavirus disease (COVID-19). COVID-19 is characterized by significant immune dysfunction, which is caused by strong but unregulated innate immunity with depressed adaptive immunity. Reduced and delayed responses to interferons (IFN-I/IFN-III) can increase the synthesis of proinflammatory cytokines and extensive immune cell infiltration into the airways, leading to pulmonary disease. The development of effective treatments for severe COVID-19 patients relies on our knowledge of the pathophysiological components of this imbalanced innate immune response. Strategies to address innate response factors will be essential. Significant efforts are currently underway to develop vaccines against SARS-CoV-2. COVID-19 vaccines, such as inactivated DNA, mRNA, and protein subunit vaccines, have already been applied in clinical use. Various vaccines display different levels of effectiveness, and it is important to continue to optimize and update their composition in order to increase their effectiveness. However, due to the continuous emergence of variant viruses, improving the immunity of the general public may also increase the effectiveness of the vaccines. Many observational studies have demonstrated that serum levels of vitamin D are inversely correlated with the incidence or severity of COVID-19. Extensive evidence has shown that vitamin D supplementation could be vital in mitigating the progression of COVID-19 to reduce its severity. Vitamin D defends against SARS-CoV-2 through a complex mechanism through interactions between the modulation of innate and adaptive immune reactions, ACE2 expression, and inhibition of the renin-angiotensin system (RAS). However, it remains unclear whether Vit-D also plays an important role in the effectiveness of different COVID-19 vaccines. Based on analysis of the molecular mechanism involved, we speculated that vit-D, via various immune signaling pathways, plays a complementary role in the development of vaccine efficacy.


Asunto(s)
Vacunas contra la COVID-19/administración & dosificación , COVID-19/prevención & control , Vitamina D/administración & dosificación , Vitamina D/sangre , Animales , COVID-19/sangre , COVID-19/inmunología , Vacunas contra la COVID-19/inmunología , Ensayos Clínicos como Asunto , Humanos , Inmunogenicidad Vacunal , Pandemias/prevención & control , Ensayos Clínicos Controlados Aleatorios como Asunto , SARS-CoV-2/aislamiento & purificación , Vitamina D/inmunología
4.
Int J Mol Sci ; 22(10)2021 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-34065735

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is still an ongoing global health crisis. Immediately after the inhalation of SARS-CoV-2 viral particles, alveolar type II epithelial cells harbor and initiate local innate immunity. These particles can infect circulating macrophages, which then present the coronavirus antigens to T cells. Subsequently, the activation and differentiation of various types of T cells, as well as uncontrollable cytokine release (also known as cytokine storms), result in tissue destruction and amplification of the immune response. Vitamin D enhances the innate immunity required for combating COVID-19 by activating toll-like receptor 2. It also enhances antimicrobial peptide synthesis, such as through the promotion of the expression and secretion of cathelicidin and ß-defensin; promotes autophagy through autophagosome formation; and increases the synthesis of lysosomal degradation enzymes within macrophages. Regarding adaptive immunity, vitamin D enhances CD4+ T cells, suppresses T helper 17 cells, and promotes the production of virus-specific antibodies by activating T cell-dependent B cells. Moreover, vitamin D attenuates the release of pro-inflammatory cytokines by CD4+ T cells through nuclear factor κB signaling, thereby inhibiting the development of a cytokine storm. SARS-CoV-2 enters cells after its spike proteins are bound to angiotensin-converting enzyme 2 (ACE2) receptors. Vitamin D increases the bioavailability and expression of ACE2, which may be responsible for trapping and inactivating the virus. Activation of the renin-angiotensin-aldosterone system (RAS) is responsible for tissue destruction, inflammation, and organ failure related to SARS-CoV-2. Vitamin D inhibits renin expression and serves as a negative RAS regulator. In conclusion, vitamin D defends the body against SARS-CoV-2 through a novel complex mechanism that operates through interactions between the activation of both innate and adaptive immunity, ACE2 expression, and inhibition of the RAS system. Multiple observation studies have shown that serum concentrations of 25 hydroxyvitamin D are inversely correlated with the incidence or severity of COVID-19. The evidence gathered thus far, generally meets Hill's causality criteria in a biological system, although experimental verification is not sufficient. We speculated that adequate vitamin D supplementation may be essential for mitigating the progression and severity of COVID-19. Future studies are warranted to determine the dosage and effectiveness of vitamin D supplementation among different populations of individuals with COVID-19.


Asunto(s)
Inmunidad Adaptativa , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/inmunología , Inmunidad Innata , SARS-CoV-2/inmunología , Vitamina D/metabolismo , Vitamina D/farmacología , COVID-19/mortalidad , COVID-19/fisiopatología , COVID-19/virología , Síndrome de Liberación de Citoquinas/complicaciones , Citocinas/metabolismo , Humanos , Receptores Virales/metabolismo , Sistema Renina-Angiotensina/fisiología
5.
Sci Rep ; 11(1): 1171, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441921

RESUMEN

Identifying modifiable risk factors of peritoneal dialysis (PD)-related peritonitis is of clinical importance in patient care. Mineral bone disease (MBD) has been associated with mortality and morbidity in end-stage kidney disease (ESKD) patients. However, its influence on PD related peritonitis due to altered host immunity remains elusive. This study investigated whether abnormal biomarkers of MBD are associated with the development of peritonitis in patients undergoing maintenance PD. We conducted a retrospective observational cohort study, analysing data derived from a nationwide dialysis registry database in Taiwan, from 2005 to 2012. A total of 5750 ESKD patients commencing PD therapy during this period were enrolled and followed up to 60 months or by the end of the study period. The patients were stratified based on their baseline serum parathyroid hormone (PTH) levels, calcium (Ca) levels or phosphorus (P) levels, respectively or in combinations. The primary outcome was the occurrence of first episode of peritonitis, and patient outcomes such as deaths, transfer to haemodialysis or receiving renal transplantation were censored. Peritonitis-free survival and the influence of PTH, Ca, P (individual or in combination) on the peritonitis occurrence were analysed. A total of 5750 PD patients was enrolled. Of them, 1611 patients experienced their first episode of peritonitis during the study period. Patients with low PTH, high Ca or low P levels, respectively or in combination, had the lowest peritonitis-free survival. After adjusting for age, sex and serum albumin levels, we found that the combinations of low PTH levels with either high Ca levels or low/normal P levels were significant risk factors of developing peritonitis. Abnormal mineral bone metabolism in maintenance PD patients with low serum PTH levels, in combination with either high Ca levels or low/normal P levels, could be novel risk factors of PD-related peritonitis.


Asunto(s)
Calcio/sangre , Hormona Paratiroidea/sangre , Diálisis Peritoneal/efectos adversos , Peritonitis/sangre , Peritonitis/etiología , Fósforo/sangre , Biomarcadores/sangre , Enfermedades Óseas/sangre , Calcio de la Dieta/sangre , Humanos , Fallo Renal Crónico/sangre , Estudios Retrospectivos , Factores de Riesgo , Taiwán
6.
Int J Med Sci ; 18(2): 314-324, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390800

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic is the largest health crisis ever faced worldwide. It has resulted in great health and economic costs because no effective treatment is currently available. Since infected persons vary in presentation from healthy asymptomatic mild symptoms to those who need intensive care support and eventually succumb to the disease, this illness is considered to depend primarily on individual immunity. Demographic distribution and disease severity in several regions of the world vary; therefore, it is believed that natural inherent immunity provided through dietary sources and traditional medicines could play an important role in infection prevention and disease progression. People can boost their immunity to prevent them from infection after COVID-19 exposure and can reduce their inflammatory reactions to protect their organ deterioration in case suffering from the disease. Some drugs with in-situ immunomodulatory and anti-inflammatory activity are also identified as adjunctive therapy in the COVID-19 era. This review discusses the importance of COVID-19 interactions with immune cells and inflammatory cells; and further emphasizes the possible pathways related with traditional herbs, medications and nutritional products. We believe that such pathophysiological pathway approach treatment is rational and important for future development of new therapeutic agents for prevention or cure of COVID-19 infection.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Interacciones Huésped-Patógeno , Medicina Tradicional , COVID-19/prevención & control , COVID-19/virología , Quimioterapia Combinada , Humanos , Inmunomodulación , Terapia Molecular Dirigida , Fitoterapia , Extractos Vegetales/uso terapéutico , SARS-CoV-2/fisiología , Vitaminas/uso terapéutico , Zinc/uso terapéutico
7.
Int J Mol Sci ; 21(7)2020 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-32252330

RESUMEN

Vascular calcification, which involves the deposition of calcifying particles within the arterial wall, is mediated by atherosclerosis, vascular smooth muscle cell osteoblastic changes, adventitial mesenchymal stem cell osteoblastic differentiation, and insufficiency of the calcification inhibitors. Recent observations implied a role for mesenchymal stem cells and endothelial progenitor cells in vascular calcification. Mesenchymal stem cells reside in the bone marrow and the adventitial layer of arteries. Endothelial progenitor cells that originate from the bone marrow are an important mechanism for repairing injured endothelial cells. Mesenchymal stem cells may differentiate osteogenically by inflammation or by specific stimuli, which can activate calcification. However, the bioactive substances secreted from mesenchymal stem cells have been shown to mitigate vascular calcification by suppressing inflammation, bone morphogenetic protein 2, and the Wingless-INT signal. Vitamin D deficiency may contribute to vascular calcification. Vitamin D supplement has been used to modulate the osteoblastic differentiation of mesenchymal stem cells and to lessen vascular injury by stimulating adhesion and migration of endothelial progenitor cells. This narrative review clarifies the role of mesenchymal stem cells and the possible role of vitamin D in the mechanisms of vascular calcification.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Calcificación Vascular/etiología , Calcificación Vascular/metabolismo , Vitamina D/metabolismo , Animales , Biomarcadores , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Células Progenitoras Endoteliales/efectos de los fármacos , Humanos , Inmunofenotipificación , Células Madre Mesenquimatosas/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Calcificación Vascular/tratamiento farmacológico , Calcificación Vascular/patología , Vitamina D/farmacología , Vitamina D/uso terapéutico
8.
Int J Mol Sci ; 21(5)2020 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-32111067

RESUMEN

Vascular calcification (VC) is highly associated with cardiovascular disease and all-cause mortality in patients with chronic kidney disease. Dysregulation of endothelial cells and vascular smooth muscle cells (VSMCs) is related to VC. Sirtuin-1 (Sirt1) deacetylase encompasses a broad range of transcription factors that are linked to an extended lifespan. Sirt1 enhances endothelial NO synthase and upregulates FoxOs to activate its antioxidant properties and delay cell senescence. Sirt1 reverses osteogenic phenotypic transdifferentiation by influencing RUNX2 expression in VSMCs. Low Sirt1 hardly prevents acetylation by p300 and phosphorylation of ß-catenin that, following the facilitation of ß-catenin translocation, drives osteogenic phenotypic transdifferentiation. Hyperphosphatemia induces VC by osteogenic conversion, apoptosis, and senescence of VSMCs through the Pit-1 cotransporter, which can be retarded by the sirt1 activator resveratrol. Proinflammatory adipocytokines released from dysfunctional perivascular adipose tissue (PVAT) mediate medial calcification and arterial stiffness. Sirt1 ameliorates release of PVAT adipokines and increases adiponectin secretion, which interact with FoxO 1 against oxidative stress and inflammatory arterial insult. Conclusively, Sirt1 decelerates VC by means of influencing endothelial NO bioavailability, senescence of ECs and VSMCs, osteogenic phenotypic transdifferentiation, apoptosis of VSMCs, ECM deposition, and the inflammatory response of PVAT. Factors that aggravate VC include vitamin D deficiency-related macrophage recruitment and further inflammation responses. Supplementation with vitamin D to adequate levels is beneficial in improving PVAT macrophage infiltration and local inflammation, which further prevents VC.


Asunto(s)
Sirtuina 1/metabolismo , Calcificación Vascular/metabolismo , Adipoquinas , Tejido Adiposo/metabolismo , Animales , Apoptosis , Enfermedades Cardiovasculares/metabolismo , Transdiferenciación Celular , Células Endoteliales/metabolismo , Proteína Forkhead Box O1/metabolismo , Humanos , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Osteogénesis/fisiología , Factores de Transcripción , Calcificación Vascular/prevención & control , Rigidez Vascular , beta Catenina/metabolismo
9.
Nutrients ; 11(1)2019 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-30642029

RESUMEN

Vascular calcification is a critical complication in patients with chronic kidney disease (CKD) because it is predictive of cardiovascular events and mortality. In addition to the traditional mechanisms associated with endothelial dysfunction and the osteoblastic transformation of vascular smooth muscle cells (VSMCs), the regulation of calcification inhibitors, such as calciprotein particles (CPPs) and matrix vesicles plays a vital role in uremic vascular calcification in CKD patients because of the high prevalence of vitamin K deficiency. Vitamin K governs the gamma-carboxylation of matrix Gla protein (MGP) for inhibiting vascular calcification, and the vitamin D binding protein receptor is related to vitamin K gene expression. For patients with chronic kidney disease, adequate use of vitamin D supplements may play a role in vascular calcification through modulation of the calciprotein particles and matrix vesicles (MVs).


Asunto(s)
Insuficiencia Renal Crónica/tratamiento farmacológico , Calcificación Vascular/tratamiento farmacológico , Vitamina D/farmacología , Vitamina K/farmacología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Suplementos Dietéticos , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Hiperfosfatemia/sangre , Hiperfosfatemia/tratamiento farmacológico , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/sangre , Calcificación Vascular/etiología , Deficiencia de Vitamina D/sangre , Deficiencia de Vitamina D/complicaciones , Deficiencia de Vitamina D/tratamiento farmacológico , Deficiencia de Vitamina K/sangre , Deficiencia de Vitamina K/tratamiento farmacológico , Proteína Gla de la Matriz
10.
Nutrients ; 10(12)2018 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-30513912

RESUMEN

In chronic kidney disease (CKD), hyperphosphatemia induces fibroblast growth factor-23 (FGF-23) expression that disturbs renal 1,25-dihydroxy vitamin D (1,25D) synthesis; thereby increasing parathyroid hormone (PTH) production. FGF-23 acts on the parathyroid gland (PTG) to increase 1α-hydroxylase activity and results in increase intra-gland 1,25D production that attenuates PTH secretion efficiently if sufficient 25D are available. Interesting, calcimimetics can further increase PTG 1α-hydroxylase activity that emphasizes the demand for nutritional vitamin D (NVD) under high PTH status. In addition, the changes in hydroxylase enzyme activity highlight the greater parathyroid 25-hydroxyvitmain D (25D) requirement in secondary hyperparathyroidism (SHPT); the higher proportion of oxyphil cells as hyperplastic parathyroid progression; lower cytosolic vitamin D binding protein (DBP) content in the oxyphil cell; and calcitriol promote vitamin D degradation are all possible reasons supports nutritional vitamin D (NVD; e.g., Cholecalciferol) supplement is crucial in SHPT. Clinically, NVD can effectively restore serum 25D concentration and prevent the further increase in PTH level. Therefore, NVD might have the benefit of alleviating the development of SHPT in early CKD and further lowering PTH in moderate to severe SHPT in dialysis patients.


Asunto(s)
Hiperparatiroidismo Secundario/complicaciones , Insuficiencia Renal Crónica/complicaciones , Vitamina D/administración & dosificación , Suplementos Dietéticos , Factor-23 de Crecimiento de Fibroblastos , Humanos , Glándulas Paratiroides/metabolismo
11.
Nutrients ; 10(2)2018 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-29439405

RESUMEN

We evaluated the improvement of intact parathyroid hormone (iPTH) levels and bone parameters by supplementing nutritional vitamin D (cholecalciferol) to combined calcimimetic (cinacalcet) and active vitamin D analog (calcitriol) among severe secondary hyperparathyroidism (SHPT) hemodialysis (HD) patients. A randomized, controlled open-label study was undertaken in 60 HD patients with serum iPTH > 1000 pg/mL or persistently high iPTH ≥ 600 pg/mL even after >3 months of calcitriol (3 µg/week). The study group received oral cholecalciferol (5000 IU/ day) and the control group received a placebo. All patients received fixed dose cinacalcet (30 mg/day, orally) and calcitriol. Calcitriol was reduced if iPTH ≤ 300 pg/mL and cinacalcet was withdrawn if serum iPTH was persistently low (iPTH ≤ 300 pg/mL) for 4 weeks after the reduction of calcitriol. A significantly lower iPTH level was noted from the 20th week in the study group compared to the placebo group, and the target iPTH ≤ 300 pg/mL was achieved at the 24th week in the study group. Most patients achieved serum 25-(OH)D3 ≥ 30 ng/mL in the study group. Nearly 40% of study patients gained >10% improvement in femoral neck (FN) bone mineral density (BMD). We conclude that cholecalciferol additively reduced serum iPTH levels, improved 25-(OH)D3 levels and improved FN BMD when used together with cinacalcet/calcitriol in severe SHPT HD patients.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Calcitriol/uso terapéutico , Colecalciferol/uso terapéutico , Cinacalcet/uso terapéutico , Hiperparatiroidismo Secundario/tratamiento farmacológico , Hormona Paratiroidea/sangre , Diálisis Renal/efectos adversos , Anciano , Calcifediol/sangre , Calcimiméticos/sangre , Calcimiméticos/farmacología , Calcimiméticos/uso terapéutico , Calcitriol/sangre , Calcitriol/farmacología , Colecalciferol/sangre , Colecalciferol/farmacología , Cinacalcet/sangre , Cinacalcet/farmacología , Suplementos Dietéticos , Femenino , Cuello Femoral/efectos de los fármacos , Cuello Femoral/metabolismo , Humanos , Hiperparatiroidismo Secundario/sangre , Hiperparatiroidismo Secundario/etiología , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Vitaminas/sangre , Vitaminas/farmacología , Vitaminas/uso terapéutico
12.
Biomed Res Int ; 2017: 2803579, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28286758

RESUMEN

The risk of cardiovascular death is 10 times higher in patients with CKD (chronic kidney disease) than in those without CKD. Vascular calcification, common in patients with CKD, is a predictor of cardiovascular mortality. Vitamin D deficiency, another complication of CKD, is associated with vascular calcification in patients with CKD. GFR decline, proteinuria, tubulointerstitial injury, and the therapeutic dose of active form vitamin D aggravate vitamin D deficiency and reduce its pleiotropic effect on the cardiovascular system. Vitamin D supplement for CKD patients provides a protective role in vascular calcification on the endothelium by (1) renin-angiotensin-aldosterone system inactivation, (2) alleviating insulin resistance, (3) reduction of cholesterol and inhibition of foam cell and cholesterol efflux in macrophages, and (4) modulating vascular regeneration. For the arterial calcification, vitamin D supplement provides adjunctive role in regressing proteinuria, reverse renal osteodystrophy, and restoring calcification inhibitors. Recently, adventitial progenitor cell has been linked to be involved in the vascular calcification. Vitamin D may provide a role in modulating adventitial progenitor cells. In summary, vitamin D supplement may provide an ancillary role for ameliorating uremic vascular calcification.


Asunto(s)
Endotelio Vascular , Insuficiencia Renal Crónica , Uremia , Calcificación Vascular , Vitamina D/uso terapéutico , Adventicia/metabolismo , Adventicia/patología , Animales , Colesterol/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Células Espumosas/metabolismo , Células Espumosas/patología , Humanos , Resistencia a la Insulina , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Sistema Renina-Angiotensina/efectos de los fármacos , Uremia/complicaciones , Uremia/tratamiento farmacológico , Uremia/metabolismo , Uremia/patología , Calcificación Vascular/tratamiento farmacológico , Calcificación Vascular/etiología , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
13.
Nutrients ; 8(11)2016 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-27827962

RESUMEN

BACKGROUND: Active Vitamin D analogues are used clinically for prevention and treatment of secondary hyperparathyroidism (SHPT) in hemodialysis (HD) patients. Nutritional vitamin D supplementation is used for additional local parathyroid (PTH) suppression, with lower incidence of hypercalcemia and hyperphosphatemia. This study evaluates the possible beneficial effects of combined vitamin D treatment (paricalcitol and cholecalciferol). METHODS: Sixty HD patients with serum parathyroid hormone (iPTH) >300 pg/mL were enrolled. All patients administered 2 mcg/day of paricalcitol and were randomly allocated into control group (placebo) or study group (cholecalciferol) for 16 weeks. Serum 25(OH)D3, iPTH and human cathelicidin (hCAP-18) were measured at baseline and during follow-up. RESULTS: iPTH levels decreased in the study group appropriately and were more significantly decreased at 16 weeks. Study group had significantly increased 25(OH)D3 levels. In addition, the study group had significantly increased serum hCAP-18 levels compared with control group. Correlation analysis showed a significant correlation between the percentage increase in serum hCAP-18 and 25(OH)D3 levels. CONCLUSIONS: Cholecalciferol, in combination with paricalcitol, additively lowers the iPTH levels in a significant number of patients after 16 weeks of supplementation. A dose of 5000 IU/week of cholecalciferol could maintain serum 25(OH)D3 levels above 30 ng/dL as early as 8 weeks after beginning supplementation. Doubling of serum cathelicidin levels were noted after 16 weeks of cholecalciferol supplementation in 40% of study patients.


Asunto(s)
Colecalciferol/administración & dosificación , Ergocalciferoles/uso terapéutico , Hiperparatiroidismo Secundario/tratamiento farmacológico , Hormona Paratiroidea/sangre , Diálisis Renal , Vitamina D/sangre , Anciano , Péptidos Catiónicos Antimicrobianos/sangre , Calcifediol/sangre , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Humanos , Hiperparatiroidismo Secundario/etiología , Hiperparatiroidismo Secundario/prevención & control , Fallo Renal Crónico/complicaciones , Masculino , Persona de Mediana Edad , Placebos , Resultado del Tratamiento , Catelicidinas
14.
Clin Chim Acta ; 450: 135-44, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26291576

RESUMEN

The common causes of death in chronic kidney disease (CKD) patients are cardiovascular events and infectious disease. These patients are also predisposed to the development of vitamin D deficiency, which leads to an increased risk of immune dysfunction. Many extra-renal cells possess the capability to produce local active 1,25(OH)2D in an intracrine or paracrine fashion, even without kidney function. Vitamin D affects both the innate and adaptive immune systems. In innate immunity, vitamin D promotes production of cathelicidin and ß-defensin 2 and enhances the capacity for autophagy via toll-like receptor activation as well as affects complement concentrations. In adaptive immunity, vitamin D suppresses the maturation of dendritic cells and weakens antigen presentation. Vitamin D also increases T helper (Th) 2 cytokine production and the efficiency of Treg lymphocytes but suppresses the secretion of Th1 and Th17 cytokines. In addition, vitamin D can decrease autoimmune disease activity. Vitamin D has been shown to play an important role in maintaining normal immune function and crosstalk between the innate and adaptive immune systems. Vitamin D deficiency may also contribute to deterioration of immune function and infectious disorders in CKD patients. However, it needs more evidence to support the requirements for vitamin D supplementation.


Asunto(s)
Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/metabolismo , Vitamina D/metabolismo , Inmunidad Adaptativa , Animales , Humanos , Inmunidad Innata
15.
J Chin Med Assoc ; 77(12): 642-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25443802

RESUMEN

BACKGROUND: No evidence exists from randomized trials to support using cloud-based manometers integrated with available physician order entry systems for tracking patient blood pressure (BP) to assist in the control of renal function deterioration. We investigated how integrating cloud-based manometers with physician order entry systems benefits our outpatient chronic kidney disease patients compared with typical BP tracking systems. METHODS: We randomly assigned 36 chronic kidney disease patients to use cloud-based manometers integrated with physician order entry systems or typical BP recording sheets, and followed the patients for 6 months. The composite outcome was that the patients saw improvement both in BP and renal function. RESULTS: We compared the systolic and diastolic BP (SBP and DBP), and renal function of our patients at 0 months, 3 months, and 6 months after using the integrated manometers and typical BP monitoring sheets. Nighttime SBP and DBP were significantly lower in the study group compared with the control group. Serum creatinine level in the study group improved significantly compared with the control group after the end of Month 6 (2.83 ± 2.0 vs. 4.38 ± 3.0, p = 0.018). Proteinuria improved nonsignificantly in Month 6 in the study group compared with the control group (1.05 ± 0.9 vs. 1.90 ± 1.3, p = 0.09). Both SBP and DBP during the nighttime hours improved significantly in the study group compared with the baseline. CONCLUSION: In pre-end-stage renal disease patients, regularly monitoring BP by integrating cloud-based manometers appears to result in a significant decrease in creatinine and improvement in nighttime BP control. Estimated glomerular filtration rate and proteinuria were found to be improved nonsignificantly, and thus, larger population and longer follow-up studies may be needed.


Asunto(s)
Prestación Integrada de Atención de Salud , Riñón/fisiopatología , Sistemas de Entrada de Órdenes Médicas , Insuficiencia Renal Crónica/fisiopatología , Anciano , Presión Sanguínea , Femenino , Humanos , Masculino , Manometría , Persona de Mediana Edad
16.
Clin Biochem ; 45(16-17): 1286-94, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22683753

RESUMEN

Patients on dialysis have a substantially higher mortality rate compared with the general population. Dialysis is usually associated with an increased plasma level of homocysteine (Hcy). Hcy is viewed as a nontraditional marker of the prognosis of cardiovascular disease (CVD) in the general population and in patients with chronic kidney disease. The effects of Hcy-lowering therapy in patients with end-stage renal disease (ESRD) remain controversial. We searched multiple databases including PubMed, MEDLINE, and OVID, and conducted a systematic review of the literature. Possible therapeutic measures were also surveyed. Our review shows that effective normalization of plasma Hcy level may decrease CVD-related morbidity and mortality in nondiabetic ESRD patients. Hyperglycemia in association with diabetes mellitus makes ESRD patients resistant to Hcy-lowering therapy. Folic acid fortification may attenuate the beneficial effects of Hcy-lowering therapy. Supraphysiological doses of folic acid and vitamin B supplementation might be needed in ESRD patients with diabetes or high Hcy levels. The response to Hcy-lowering therapy may be influenced by differences within and between populations in sex, genotype, nutrition, and mandatory fortification. Treatment resistance found mainly in diabetic ESRD patients but not in nondiabetic ESRD patients that may need other therapeutic approaches.


Asunto(s)
Homocisteína/sangre , Fallo Renal Crónico/sangre , Animales , Diálisis , Homocisteína/metabolismo , Homocisteína/fisiología , Humanos , Hiperglucemia/sangre , Hiperglucemia/terapia , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/terapia , Riñón/fisiopatología , Fallo Renal Crónico/mortalidad , Fallo Renal Crónico/terapia , Metaanálisis como Asunto , Diálisis Renal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA