Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 665: 141-151, 2023 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-37163934

RESUMEN

Traumatic brain injury (TBI) can negatively impact systemic organs, which can lead to more death and disability. However, the mechanism underlying the effect of TBI on systemic organs remains unclear. In previous work, we found that brain-derived extracellular vesicles (BDEVs) released from the injured brain can induce systemic coagulation with a widespread fibrin deposition in the microvasculature of the lungs, kidney, and heart in a mouse model of TBI. In this study, we investigated whether BDEVs can induce heart, lung, liver, and kidney injury in TBI mice. The results of pathological staining and related biomarkers indicated that BDEVs can induce histological damage and systematic dysfunction. In vivo imaging system demonstrated that BDEVs can gather in systemic organs. We also found that BDEVs could induce cell apoptosis in the lung, liver, heart, and kidney. Furthermore, we discovered that BDEVs could cause multi-organ endothelial cell damage. Finally, this secondary multi-organ damage could be relieved by removing circulating BDEVs. Our research provides a novel perspective and potential mechanism of TBI-associated multi-organ damage.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Vesículas Extracelulares , Ratones , Animales , Encéfalo/patología , Lesiones Encefálicas/patología , Apoptosis , Vesículas Extracelulares/patología
2.
Mol Cell Biochem ; 478(1): 103-119, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35731343

RESUMEN

The ongoing pandemic (also known as coronavirus disease-19; COVID-19) by a constantly emerging viral agent commonly referred as the severe acute respiratory syndrome corona virus 2 or SARS-CoV-2 has revealed unique pathological findings from infected human beings, and the postmortem observations. The list of disease symptoms, and postmortem observations is too long to mention; however, SARS-CoV-2 has brought with it a whole new clinical syndrome in "long haulers" including dyspnea, chest pain, tachycardia, brain fog, exercise intolerance, and extreme fatigue. We opine that further improvement in delivering effective treatment, and preventive strategies would be benefited from validated animal disease models. In this context, we designed a study, and show that a genetically engineered mouse expressing the human angiotensin converting enzyme 2; ACE-2 (the receptor used by SARS-CoV-2 agent to enter host cells) represents an excellent investigative resource in simulating important clinical features of the COVID-19. The ACE-2 mouse model (which is susceptible to SARS-CoV-2) when administered with a recombinant SARS-CoV-2 spike protein (SP) intranasally exhibited a profound cytokine storm capable of altering the physiological parameters including significant changes in cardiac function along with multi-organ damage that was further confirmed via histological findings. More importantly, visceral organs from SP treated mice revealed thrombotic blood clots as seen during postmortem examination. Thus, the ACE-2 engineered mouse appears to be a suitable model for studying intimate viral pathogenesis thus paving the way for identification, and characterization of appropriate prophylactics as well as therapeutics for COVID-19 management.


Asunto(s)
COVID-19 , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/genética
3.
Int J Mol Sci ; 23(8)2022 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-35456997

RESUMEN

INTRODUCTION AND PURPOSE OF THE STUDY: SARS-CoV-2 virus does not only affect the respiratory system. It may cause damage to many organ systems with long-term effects. The latest scientific reports inform that this virus leaves a long-term trace in the nervous, circulatory, respiratory, urinary and reproductive systems. It manifests itself in disturbances in the functioning of the organs of these systems, causing serious health problems. The aim of the study was to review the latest research into the long-term effects of COVID-19 and determine how common these symptoms are and who is most at risk. Based on a literature review using the electronic scientific databases of PubMed and Web of Science on the long-term effects of SARS-CoV-2 infection, 88 studies were included in the analysis. The information contained in the analyzed literature shows that the SARS-CoV-2 virus can cause multi-organ damage, causing a number of long-term negative health complications. CONCLUSIONS: There is evidence that the virus can cause long-term complications lasting more than six months. They mainly concern disturbances in the functioning of the nervous, circulatory and respiratory systems. However, these studies are small or short-lasting, and many are speculative.


Asunto(s)
COVID-19 , SARS-CoV-2 , Cuerpo Humano , Humanos , Inflamación
4.
Clin Exp Med ; 24(1): 96, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38717644

RESUMEN

Primary Sjögren's Syndrome (pSS) falls within the category of connective tissue diseases, characterized by the presence of autoantibodies such as antinuclear antibodies (ANA). However, according to the classification criteria for pSS, some patients may exhibit a negative result for autoantibodies. Patients with a negative result for autoantibodies may lack typical features of connective tissue diseases, and the immunological state as well as the extent of organ involvement and damage may differ from those with positive autoantibodies. This study aims to compare the clinical phenotypes of patients with positive and negative autoantibodies, providing insights for disease classification and treatment selection for clinicians. Patients with pSS were grouped based on the presence and titers of their autoantibodies. Subsequently, differences in organ damage and laboratory indicators were compared between these groups, aiming to analyze the value of autoantibody titers in assessing the condition of pSS. (1) Patients with positive ANA exhibited elevated levels of inflammatory indicators, including ESR, IgG levels, lip gland biopsy pathology grade, and overall organ involvement, in comparison with patients with negative ANA (P < 0.05). Furthermore, ANA-positivity correlated with a higher occurrence of multi-organ damage, particularly affecting the skin, mucous membranes, and the hematological system (P < 0.05). (2) As ANA titers increased, patients demonstrated elevated levels of IgG and an escalation in organ involvement (P < 0.05). (3) Patients in the positive autoantibody group (positive for antinuclear antibodies, anti-SSA, or anti-SSB antibodies) had higher IgG levels compared to the negative group (P < 0.05). (4) Patients with positive anti-SSA and anti-SSB antibodies exhibited higher levels of inflammatory indicators and IgG compared to other patients (P < 0.05); however, no significant differences were observed in terms of organ involvement and organ damage. Patients with positive ANA in pSS typically exhibit higher levels of inflammation and an increased likelihood of experiencing multi-organ damage. Furthermore, as the ANA titers increase, both inflammation levels and the risk of multi-organ damage also escalate. Additionally, the presence of anti-SSA and anti-SSB antibodies may contribute to an elevated risk of increased inflammation levels, but does not increase the risk of organ damage.


Asunto(s)
Anticuerpos Antinucleares , Síndrome de Sjögren , Humanos , Anticuerpos Antinucleares/sangre , Anticuerpos Antinucleares/inmunología , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/patología , Síndrome de Sjögren/sangre , Femenino , Persona de Mediana Edad , Masculino , Adulto , Anciano , Inflamación/inmunología , Inflamación/patología , Inmunoglobulina G/sangre
5.
Inflammation ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38884700

RESUMEN

Acute pancreatitis (AP) is one of the most common gastrointestinal emergencies, often resulting in self-digestion, edema, hemorrhage, and even necrosis of pancreatic tissue. When AP progresses to severe acute pancreatitis (SAP), it often causes multi-organ damage, leading to a high mortality rate. However, the molecular mechanisms underlying SAP-mediated organ damage remain unclear. This study aims to systematically mine SAP data from public databases and combine experimental validation to identify key molecules involved in multi-organ damage caused by SAP. Retrieve transcriptomic data of mice pancreatic tissue for AP, lung and liver tissue for SAP, and corresponding normal tissue from the Gene Expression Omnibus (GEO) database. Conduct gene differential analysis using Limma and DEseq2 methods. Perform enrichment analysis using the clusterProfiler package in R software. Score immune cells and immune status in various organs using single-sample gene set enrichment analysis (ssGSEA). Evaluate mRNA expression levels of core genes using reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry. Validate serum amylase, TNF-α, IL-1ß, and IL-6 levels in peripheral blood using enzyme-linked immunosorbent assay (ELISA), and detect the formation of neutrophil extracellular traps (NETs) in mice pancreatic, liver, and lung tissues using immunofluorescence. Differential analysis reveals that 46 genes exhibit expression dysregulation in mice pancreatic tissue for AP, liver and lung tissue for SAP, as well as peripheral blood in humans. Functional enrichment analysis indicates that these genes are primarily associated with neutrophil-related biological processes. ROC curve analysis indicates that 12 neutrophil-related genes have diagnostic potential for SAP. Immune infiltration analysis reveals high neutrophil infiltration in various organs affected by SAP. Single-cell sequencing analysis shows that these genes are predominantly expressed in neutrophils and macrophages. FPR1, ITGAM, and C5AR1 are identified as key genes involved in the formation of NETs and activation of neutrophils. qPCR and IHC results demonstrate upregulation of FPR1, ITGAM, and C5AR1 expression in pancreatic, liver, and lung tissues of mice with SAP. Immunofluorescence staining shows increased levels of neutrophils and NETs in SAP mice. Inhibition of NETs formation can alleviate the severity of SAP as well as the levels of inflammation in the liver and lung tissues. This study identified key genes involved in the formation of NETs, namely FPR1, ITGAM, and C5AR1, which are upregulated during multi-organ damage in SAP. Inhibition of NETs release effectively reduces the systemic inflammatory response and liver-lung damage in SAP. This research provides new therapeutic targets for the multi-organ damage associated with SAP.

6.
Front Pediatr ; 11: 1223266, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37650048

RESUMEN

Background: Multisystem inflammatory syndrome in children (MIS-c) emerged during the coronavirus disease 2019 pandemic and is associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Despite the extensively studied clinical manifestation of acute condition, the short- and long-term effects of MIS-c on children's health are unknown. Methods: This was a prospective longitudinal cohort study. Children aged <18 years who met the Centers for Disease Prevention and Control (CDC) diagnostic criteria and who were admitted to the Children's Clinical University Hospital of Latvia (CCUH) between July 1, 2020, and April 15, 2022, were enrolled in the study. An outpatient follow-up program was initiated in July 2020. All children were evaluated at 2 weeks, 2 months (1-3 months), and 6 months (5-7 months) after discharge. The face-to-face interviews comprised four domains as follows: symptom assessment, physical examination, laboratory testing, and cardiological investigation [including electrocardiogram (ECG) and echocardiography (echo)]. Results: Overall, 21 patients with MIS-c were enrolled. The median age of the study group was 6 years. At the 2-week follow-up, almost half of the patients (N = 10, 47.6%) reported exercise intolerance with provoked tiredness. Laboratory tests showed a considerable increase in blood cell count, with a near doubling of leukocyte and neutrophil counts and a tripling of thrombocyte levels. However, a decline in the levels of inflammatory and organ-specific markers was observed. Cardiological investigation showed significant improvement with gradual resolution of the acute-phase pathological findings. Within 2 months, improvement in exercise capacity was observed with 5-fold and 2-fold reductions in physical intolerance (N = 2, 9.5%) and physical activity-induced fatigue (N = 5, 23.8%), respectively. Normalization of all blood cell lines was observed, and cardiological investigation showed no persistent changes. At the 6-month visit, further improvement in the children's exercise capacity was observed, and both laboratory and cardiological investigation showed no pathological changes. Conclusions: Most persistent symptoms were reported within the first 2 weeks after the acute phase, with decreased physical activity tolerance and activity-induced fatigue as the main features. A positive trend was observed at each follow-up visit as the spectrum of the children's complaints decreased. Furthermore, rapid normalization of laboratory markers and cardiac abnormalities was observed.

7.
Inflammation ; 45(1): 279-296, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34564825

RESUMEN

Sepsis leads to systemic hypotension, disturbed perfusion, inflammation, and tissue toxicity in vital organs. Neuropeptide W (NPW) has modulatory effects in the control of blood pressure and inflammatory processes, implicating a potential beneficial effect against sepsis-induced oxidative damage. Under anesthesia, male Sprague Dawley rats underwent cecal ligation and puncture. Immediately after surgery, either saline or TNF-alpha inhibitor (etanercept; 1 mg/kg) antibiotic (ceftriaxon; 10 mg/kg) combination or NPW (0.1, 1, or 3 µg/kg) was given subcutaneously, and injections were repeated on the 12th and 24th h. The sham-operated control group was treated with saline at the same time points. All rats were euthanized on the 25th h of surgery. Sepsis resulted in oxidative damage of the brain, heart, lung, liver, and kidney. Elevations in blood urea nitrogen and alkaline phosphatase, showing renal and hepatic dysfunction, were not evident when septic rats were treated with NPW. NPW reduced serum levels of C-reactive protein, corticosterone, and interleukin-6, while histopathologically verified tissue damage in all the studied tissues was ameliorated. NPW treatment suppressed lipid peroxidation in the heart, lung, and brain, and the depleted antioxidant GSH levels of the brain and heart were replenished by NPW. Moreover, sepsis-related neutrophil recruitment to the liver and lung was also suppressed by NPW. Although the survival rate of the rats was not significantly prolonged by NPW, most of these improvements in systemic and local inflammatory events were comparable with those reached by the etanercept and antibiotic combination, suggesting the therapeutic impact of NPW during the acute period of sepsis.


Asunto(s)
Antiinflamatorios/farmacología , Insuficiencia Multiorgánica/prevención & control , Neuropéptidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Sepsis/tratamiento farmacológico , Animales , Antiinflamatorios/uso terapéutico , Biomarcadores/metabolismo , Masculino , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/metabolismo , Neuropéptidos/uso terapéutico , Ratas , Ratas Sprague-Dawley , Sepsis/metabolismo , Sepsis/fisiopatología , Resultado del Tratamiento
8.
Front Cardiovasc Med ; 9: 854750, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35463767

RESUMEN

In COVID-19 the development of severe viral pneumonia that is coupled with systemic inflammatory response triggers multi-organ failure and is of major concern. Cardiac involvement occurs in nearly 60% of patients with pre-existing cardiovascular conditions and heralds worse clinical outcome. Diagnoses carried out in the acute phase of COVID-19 rely upon increased levels of circulating cardiac injury biomarkers and transthoracic echocardiography. These diagnostics, however, were unable to pinpoint the mechanisms of cardiac injury in COVID-19 patients. Identifying the main features of cardiac injury remains an urgent yet unmet need in cardiology, given the potential clinical consequences. Cardiovascular magnetic resonance (CMR) provides an unparalleled opportunity to gain a deeper insight into myocardial injury given its unique ability to interrogate the properties of myocardial tissue. This endeavor is particularly important in convalescent COVID-19 patients as many continue to experience chest pain, palpitations, dyspnea and exertional fatigue, six or more months after the acute illness. This review will provide a critical appraisal of research on cardiovascular damage in convalescent adult COVID-19 patients with an emphasis on the use of CMR and its value to our understanding of organ damage.

9.
Infect Genet Evol ; 87: 104647, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33264669

RESUMEN

The devastating pandemic of coronavirus disease 2019 (COVID-19) has caused thousands of deaths and left millions of restless patients suffering from its complications. Increasing data indicate that the disease presents in a severe form in patients with pre-existing chronic conditions like cardiovascular diseases, diabetes, respiratory system diseases, and renal diseases. This denotes that these patients are more susceptible to COVID-19 and have higher mortality rates compared to patients with no comorbid conditions. Several factors can explain the heightened susceptibility and fatal presentation of COVID-19 in these patients, for example, the enhanced expression of the angiotensin-converting enzyme-2 (ACE2) in specific organs, cytokine storm, and drug interactions contribute to the increased morbidity and mortality. Adding to the findings that individuals with pre-existing conditions may be more susceptible to COVID-19, it has also been shown that COVID-19 can induce chronic diseases in previously healthy patients. Therefore, understanding the interlinked relationship between COVID-19 and chronic diseases helps in optimizing the management of susceptible patients. This review comprehensively described the molecular mechanisms that contribute to worse COVID-19 prognosis in patients with pre-existing comorbidities such as diabetes, cardiovascular diseases, respiratory diseases, gastrointestinal and renal diseases, blood disorders, autoimmune diseases, and finally, obesity. It also focused on how COVID-19 could, in some cases, lead to chronic conditions as a result of long-term multi-organ damage. Lastly, this work carefully discussed the tailored management plans for each specific patient population, aiming to achieve the best therapeutic outcome with minimum complications.


Asunto(s)
COVID-19/complicaciones , COVID-19/virología , Enfermedad Crónica , Comorbilidad , Humanos , Pronóstico , Factores de Riesgo , SARS-CoV-2/aislamiento & purificación
10.
Mol Immunol ; 137: 105-113, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34242919

RESUMEN

Underlying mechanisms of multi-organ manifestations and exacerbated inflammation in COVID-19 are yet to be delineated. The hypothesis of SARS-CoV-2 triggering autoimmunity is gaining attention and, in the present study, we have identified 28 human proteins harbouring regions homologous to SARS-CoV-2 peptides that could possibly be acting as autoantigens in COVID-19 patients displaying autoimmune conditions. Interestingly, these conserved regions are amongst the experimentally validated B cell epitopes of SARS-CoV-2 proteins. The reported human proteins have demonstrated presence of autoantibodies against them in typical autoimmune conditions which may explain the frequent occurrence of autoimmune conditions following SARS-CoV-2 infection. Moreover, the proposed autoantigens' widespread tissue distribution is suggestive of their involvement in multi-organ manifestations via molecular mimicry. We opine that our report may aid in directing subsequent necessary antigen-specific studies, results of which would be of long-term relevance in management of extrapulmonary symptoms of COVID-19.


Asunto(s)
Autoantígenos/inmunología , Enfermedades Autoinmunes/complicaciones , COVID-19/etiología , Epítopos de Linfocito B/inmunología , SARS-CoV-2/inmunología , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/virología , Autoinmunidad/inmunología , COVID-19/inmunología , COVID-19/patología , Humanos , Imitación Molecular/inmunología
11.
Phenomics ; 1(3): 113-128, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35233559

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has created an immense menace to public health worldwide, exerting huge effects on global economic and political conditions. Understanding the biology and pathogenesis mechanisms of this novel virus, in large parts, relies on optimal physiological models that allow replication and propagation of SARS-CoV-2. Human organoids, derived from stem cells, are three-dimensional cell cultures that recapitulate the cellular organization, transcriptional and epigenetic signatures of their counterpart organs. Recent studies have indicated their great values as experimental virology platforms, making human organoid an ideal tool for investigating host-pathogen interactions. Here, we summarize research developments for SARS-CoV-2 infection of various human organoids involved in multiple systems, including lung, liver, brain, intestine, kidney and blood vessel organoids. These studies help us reveal the pathogenesis mechanism of COVID-19, and facilitate the development of effective vaccines and drugs as well as other therapeutic regimes.

12.
Expert Opin Biol Ther ; 21(5): 615-626, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33161757

RESUMEN

Introduction: The coronavirus disease 2019 (COVID-19) pandemic, caused by a newly discovered coronavirus (severe acute respiratory syndrome coronavirus 2, or SARS-CoV-2), continues to spread all around the world. Despite the emergency of COVID-19 worldwide, remdesivir is the only treatment that has been recently approved to treat the diseases, and other effective therapies are still lacking. SARS-CoV-2 may cause severe illness in 20% of patients. Based on available data, there is an association between interleukin-6 (IL-6) and severe COVID-19. Sarilumab is a fully human immunoglobulin G1 monoclonal antibody binding to both membrane-bound and soluble IL-6 receptors with high affinity and has been considered for off-label use in the treatment of COVID-19.Areas covered: The present article reviews recently published literature focusing on the pathophysiology of COVID-19 induced cytokine storm, the potential therapeutic role, and important clinical issues of sarilumab in the treatment of COVID-19 patients.Expert opinion: The off-label treatment administration is unavoidable in the critical situation of the COVID-19 pandemic. Further efforts should be directed to determine mechanisms of SARS-CoV-2 induced immune dysregulation as well as indications of sarilumab in the patients with COVID-19 to minimize concerns regarding its off-label administration.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/uso terapéutico , Alanina/análogos & derivados , Alanina/metabolismo , Alanina/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados/metabolismo , Anticuerpos Monoclonales Humanizados/farmacología , Antivirales/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/epidemiología , COVID-19/metabolismo , Síndrome de Liberación de Citoquinas/epidemiología , Síndrome de Liberación de Citoquinas/metabolismo , Humanos , Pandemias , SARS-CoV-2/metabolismo
13.
Front Mol Biosci ; 7: 573327, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33330617

RESUMEN

BACKGROUND: To this day, the molecular mechanism of endotoxin-induced multi-organ failure has not been completely clarified. This study aimed to construct an miRNA-mRNA regulatory network and identify main pathways and key genes in multi-organ of LPS-mediated endotoxemic mice. METHODS: Public datasets from six mRNA and three miRNA microarray datasets were downloaded from the GEO website to screen final differentially expressed genes (FDEGs) and hub genes in the heart, lung, liver, and kidney of LPS-mediated endotoxemic mice. Functional and pathway enrichment analysis of FDEGs was used to identify the main pathways in multi-organ damage of LPS-treated mice. Finally, hub genes of each organ were intersected to obtain the key genes of multi-organ. RESULTS: Firstly, 158, 358, 299, and 91 FDEGs were identified in the heart, lung, liver, and kidney, respectively. The pathway enrichment analysis of the FDEGs then showed that the TNF signaling pathway, Toll-like receptor signaling pathway, and some viral-infection-related pathways (influenza A, measles, and herpes simplex) were the main pathways in multi-organ damage of LPS-mediated endotoxemic mice. Moreover, miRNA-mRNA or PPI regulatory networks were constructed based on FDEGs. According to these networks, 31, 34, 34, and 31 hub genes were identified in the heart, lung, liver, and kidney, respectively. Among them, nine key genes (Cd274, Cxcl1, Cxcl9, Icam1, Ifit2, Isg15, Stat1, Tlr2, and Usp18) were enriched in Toll-like receptor signaling pathway and chemokine signaling pathway. Finally, seven potential drugs were predicted based on these key genes. CONCLUSION: The shared underlying molecular pathways in endotoxin-induced multi-organ damage that have been identified include Toll-like receptor signaling pathway and TNF signaling pathway. Besides, nine key genes (Cd274, Cxcl1, Cxcl9, Icam1, Ifit2, Isg15, Stat1, Tlr2, and Usp18) and seven potential drugs were identified. Our data provide a new sight and potential target for future therapy in endotoxemia-induced multi-organ failure.

14.
Artículo en Inglés | MEDLINE | ID: mdl-31149326

RESUMEN

BACKGROUND: Lead is a nephrotoxicant probably implicated in the rising incidence of chronic kidney injury in sub-Sahara Africa. With the prohibitive cost and unavailability of metal chelators, chronic kidney disease CKD prevention is very difficult hence the search for affordable alternative. Costus afer have been shown to be organo-protective. The present research investigated the nephroprotective effect of aqueous leaf extract of Costus afer on lead induced nephrotoxicity in male rats. METHODS: Adult male rats were weight matched into five groups of five rats each. Groups 1 & 2 serve as normal and toxic control receiving deionized and leaded (CH3COO)2Pb. 3H2O water respectively. Groups 3, 4 and 5 were administered peroral 750, 1500 and 2250 mg/kg of aqueous leaf extract of Costus afer respectively while receiving Pb2+ water ad libitum. Hematological, antioxidant and histological parameters obtained from the result serve as scientific evidence in the study. RESULTS: Costus afer treatment significantly reversed (P < 0.05) the decrease in the levels of gluthatione peroxidase (GSH-PX), superoxide dismutase (SOD), catalase (CAT), Glutathione-S-trasferase activity (GST) seen in the lead acetate only treated group. Similarly, the increased malondialdehyde (MDA) level in the lead acetate only treated group was significantly (P < 0.05) reduced in the Costus afer treated groups. There were significant (P < 0.05) decreases in serum serum level of sodium (146 ± 2.1 to 133 ± 6.0) and potassium (5.1 ± 0.4 to 4.4 ± 0.3) in lead acetate alone and treated group respectively. Also recorded was a significant (P < 0.05) decrease in serum levels of total protein and albumin (67 ± 7.9 to 47 ± 5.0 g/dl) and (45 ± 4.4 to 33 ± 5.5 g/dl) in lead acetate alone and Costus afer treated groups respectively. CONCLUSIONS: Aqueous leaf extract of Costus afer may be nephroprotective in albino rats.

15.
Pathology ; 49(1): 1-9, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27914684

RESUMEN

Sickle cell disease (SCD) is an inherited haemoglobin disorder, associated with recurrent painful episodes, ongoing haemolytic anaemia and progressive multi-organ damage. Until the early 1990s, survival beyond the fourth decade for a patient with SCD was considered unusual and prompted case reports. Nowadays, in countries with developed health care systems, more than 90 percent of newborns with SCD survive into adulthood. Nevertheless, their life expectancy is still shortened by more than two decades compared to the general population. With an increasing life expectancy, SCD has now evolved into a debilitating disorder with substantial morbidity resulting from ongoing sickle cell vasculopathy and multi-organ damage. Limited data on health care issues of older adults with SCD poses multiple challenges to patients, their families and health care providers. In this review, we will address and discuss acute and chronic complications of SCD with a special focus on the older adult.


Asunto(s)
Envejecimiento/fisiología , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/patología , Órganos en Riesgo/patología , Dolor/fisiopatología , Anemia de Células Falciformes/diagnóstico , Atención a la Salud , Humanos , Dimensión del Dolor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA