Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
GE Port J Gastroenterol ; 31(5): 325-337, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39360169

RESUMEN

Introduction: Three years after the beginning of the SARS-CoV-2 pandemic, the safety and efficacy of COVID-19 vaccination in liver cirrhosis (LC) patients remain controversial. We aimed to study the safety, immunological, and clinical responses of LC patients to COVID-19 vaccination. Methods: Prospective multicentric study in adults with LC eligible for COVID-19 vaccination, without prior known infection. Patients were followed up until the timing of a booster dose, SARS-CoV-2 infection, or death. Spike-protein immunoglobulin G antibody titers for SARS-CoV-2 at 2 weeks, 3 months, and 6 months postvaccination were assessed. Antibody titers <33.8 binding antibody units (BAU)/mL were considered seronegative and <200 BAU/mL suboptimal. Postvaccination infection and its severity were registered. Results: We included 124 LC patients, 81% males, mean aged 61 ± 10 years, with a mean follow-up of 221 ± 26 days. Alcohol was the most common (61%) cause of cirrhosis, and 7% were under immunosuppressants for autoimmune hepatitis; 69% had portal hypertension, 42% had a previous decompensation, and 21% had a Child-Pugh-Turcotte score of B/C. The type of vaccine administrated was BNT162b2 (n = 59, 48%), ChAdOx1nCoV-19 (n = 45, 36%), mRNA-1273 (n = 14, 11%), and Ad26.COV2.S (n = 6, 5%). Eighteen percent of the patients reported adverse events after vaccination, none serious. Median [Q1; Q3] antibody titers were 1,185 [280; 2,080] BAU/mL at 2 weeks, 301 [72; 1,175] BAU/mL at 3 months, and 192 [49; 656] BAU/mL at 6 months. There were seronegative and suboptimal antibody responses in 8% and 23% of the patients at 2 weeks, 16% and 38% at 3 months, and 22% and 48% at 6 months. Older age and adenovirus vector vaccines were the only factors associated with seronegative and suboptimal responses at 2 weeks and 3 months (p < 0.05) in a multivariable logistic regression analysis. Eleven patients (9%) were infected with SARS-CoV-2 during follow-up (3.8-6.6 months postvaccination), all with mild disease. There were no differences regarding the type of vaccine, and 73% had antibody titers >200 BAU/mL at 3 months. Conclusion: COVID-19 vaccines in patients with LC were safe, without serious adverse events. The humoral and clinical responses were similar to the reported for the general population. Humoral response was adversely impacted by older age and adenovirus vector vaccines and unrelated to the liver disease severity.


Introdução: Três anos após o início da pandemia SARS-CoV-2, a segurança e eficácia da vacinação COVID-19 em doentes com cirrose hepática (CH) permanecem controversas. Pretendemos avaliar a segurança, respostas imunológica e clínica de doentes com CH às vacinas contra a COVID-19. Métodos: Estudo prospetivo multicêntrico em adultos com CH elegíveis para vacinação contra a COVID-19, sem infeção prévia conhecida. Os doentes foram acompanhados até ao momento da dose de reforço, infeção SARS-CoV-2 ou falecimento. Avaliámos os títulos de anticorpos IgG da proteína-Spike SARS-CoV-2 às 2 semanas, 3 meses e 6 meses. Títulos de anticorpos <33.8 BAU/mL foram considerados seronegativos e <200 BAU/mL subótimos. A ocorrência de infeção pós-vacinação e respetiva gravidade foram registadas. Resultados: Incluímos 124 doentes com CH, 81% homens, com idade média de 61 ± 10 anos e um seguimento médio de 221 ± 26 dias. A causa mais prevalente de cirrose foi o álcool (61%) e 7% dos doentes faziam terapêutica imunossupressora por hepatite autoimune. Existiam sinais de hipertensão portal em 69%, descompensação prévia em 42% e classificação de Child-Pugh-Turcotte B/C em 21%. O tipo de vacina administrada foi: BNT162b2 (n = 59, 48%), ChAdOx1nCoV-19 (n = 45, 36%), mRNA-1273 (n = 14, 11%) e Ad26.COV2.S (n = 6, 5%). Foram reportados efeitos adversos pós-vacinação em 18% dos participantes, nenhum deles grave. Os títulos medianos [Q1; Q3] de anticorpos foram 1.185 [280; 2.080] BAU/mL às 2 semanas, 301 [72; 1.175] BAU/mL aos 3 meses e 192 [49; 656] BAU/mL aos 6 meses. Observámos respostas humorais seronegativas e subótimas em 8% e 23% dos doentes às 2 semanas, 16% e 38% aos 3 meses e 22% e 48% aos 6 meses. A idade avançada e vacinas de vetor de adenovírus foram os únicos fatores associados a respostas seronegativas e subótimas às 2 semanas e 3 meses (p < 0.05) em análise de regressão logística multivariada. Onze doentes (9%) desenvolveram infeção SARS-CoV-2 durante o seguimento (3.8­6.6 meses pós vacinação), todos com doença ligeira. Não observámos diferenças relativamente ao tipo de vacina, apresentando 73% deles títulos de anticorpos >200 BAU/mL aos 3 meses. Conclusões: A vacinação contra a COVID-19 em doentes com CH foi segura, sem efeitos adversos graves. As respostas humoral e clínica foram semelhantes às reportadas na população geral. A resposta humoral foi afetada negativamente pela idade avançada e vacinas de vetor de adenovírus e não apresentou relação com a gravidade da doença hepática.

2.
Hepatol Int ; 18(4): 1178-1201, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38878111

RESUMEN

BACKGROUND: With the implementation of the 11th edition of the International Classification of Diseases (ICD-11) and the publication of the metabolic dysfunction-associated fatty liver disease (MAFLD) nomenclature in 2020, it is important to establish consensus for the coding of MAFLD in ICD-11. This will inform subsequent revisions of ICD-11. METHODS: Using the Qualtrics XM and WJX platforms, questionnaires were sent online to MAFLD-ICD-11 coding collaborators, authors of papers, and relevant association members. RESULTS: A total of 890 international experts in various fields from 61 countries responded to the survey. We also achieved full coverage of provincial-level administrative regions in China. 77.1% of respondents agreed that MAFLD should be represented in ICD-11 by updating NAFLD, with no significant regional differences (77.3% in Asia and 76.6% in non-Asia, p = 0.819). Over 80% of respondents agreed or somewhat agreed with the need to assign specific codes for progressive stages of MAFLD (i.e. steatohepatitis) (92.2%), MAFLD combined with comorbidities (84.1%), or MAFLD subtypes (i.e., lean, overweight/obese, and diabetic) (86.1%). CONCLUSIONS: This global survey by a collaborative panel of clinical, coding, health management and policy experts, indicates agreement that MAFLD should be coded in ICD-11. The data serves as a foundation for corresponding adjustments in the ICD-11 revision.


Asunto(s)
Clasificación Internacional de Enfermedades , Enfermedad del Hígado Graso no Alcohólico , Humanos , Enfermedad del Hígado Graso no Alcohólico/epidemiología , Enfermedad del Hígado Graso no Alcohólico/clasificación , Encuestas y Cuestionarios , Salud Global
4.
GE Port J Gastroenterol ; 31(1): 14-23, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38314031

RESUMEN

Liver cancer is globally the third leading cause of death from cancer. Hepatocellular carcinoma (HCC) develops in patients with underlying liver disease. The fraction of HCC attributed to nonalcoholic fatty liver disease (NAFLD) shows an accelerated increase in the last decades, being already responsible for 15% of all HCC cases. Similar to other causes of liver cirrhosis, patients with NAFLD-associated cirrhosis should be enrolled in HCC-screening programs, yet these patients are under-screened, and currently are less than half likely to be proposed for HCC screening as compared to patients with HCV-associated cirrhosis. NAFLD-associated HCC has the peculiarity of occurring in precirrhotic phases in 20-50% of the cases. Currently, HCC screening in precirrhotic NAFLD patients is not routinely recommended, since the risk of developing HCC is very low. However, because NAFLD affects one-third of the worldwide population, noncirrhotic NAFLD already accounts for 6% of HCC cases. As such, it is pressing to develop stratification tools, in order to personalize the individual risk of HCC development in a patient with NAFLD, allowing precision HCC-screening programs. This review summarizes the epidemiology of NAFLD-associated HCC with a critical analysis of current HCC-screening recommendations.


O cancro do fígado é, globalmente, a terceira causa de morte por cancro. O carcinoma hepatocelular (CHC) desenvolve-se em doentes com doença hepática crónica subjacente. A fracção de CHC atribuível ao fígado gordo não alcoólico (FGNA) tem vindo a aumentar com uma aceleração no seu crescimento nas últimas décadas, sendo atualmente responsável por 15% dos casos de CHC. À semelhança do que ocorre com outras causas de cirrose hepática, os doentes com cirrose associada a FGNA devem ser inseridos em programas de rastreio de CHC. Contudo, esses doentes são sub-rastreados, já que a probabilidade de serem incluídos em programas de rastreio de CHC é menos de metade comparando com doentes com cirrose associada a hepatite C crónica. O CHC associado ao FGNA tem a particularidade de ocorrer em fases pré-cirróticas em 20 a 50% dos casos. O rastreio de CHC em doentes com FGNA em fase pré-cirrótica não está recomendado por rotina, uma vez que, ainda assim, o risco destes doentes desenvolverem CHC é muito baixo. No entanto, uma vez que um terço da população mundial tem FGNA, o FGNA em não cirróticos corresponde a 6% de todos os casos de CHC. Assim sendo, é urgente o desenvolvimento de métodos de estratificação, por forma a personalizar o risco individual de desenvolvimento de CHC em doentes com FGNA, permitindo maior precisão nos programas de rastreio de CHC. Esta revisão sumariza a epidemiologia de CHC associado ao FGNA, com uma análise crítica das atuais recomendações de rastreio de CHC.

5.
Front Med (Lausanne) ; 10: 1316284, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38146424

RESUMEN

MASLD prevalence is growing towards the leading cause of end-stage liver disease. Up to today, the most effective treatment is weight loss. Weight loss interventions are moving from lifestyle changes to bariatric surgery or endoscopy, and, more recently, to a new wave of anti-obesity drugs that can compete with bariatric surgery. Liver-targeted therapy is a necessity for those patients who already present liver fibrosis. The field is moving fast, and in the near future, we will testify to a disruptive change in MASLD treatment, similar to the paradigm-shift that occurred for hepatitis C almost one decade ago with direct antiviral agents.

7.
Int J Mol Sci ; 24(2)2023 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-36674460

RESUMEN

Celiac disease (CD) is a common autoimmune disease affecting around 1% of the population. It consists of an immune-mediated enteropathy, triggered by gluten exposure in susceptible patients. All patients with CD, irrespective of the presence of symptoms, must endure a lifelong gluten-free diet (GFD). This is not an easy task due to a lack of awareness of the gluten content in foods and the extensive incorporation of gluten in processed foods. Furthermore, a GFD imposes a sense of limitation and might be associated with decreased quality of life in CD patients. This results in gluten contamination in the diet of four out of five celiac patients adhering to a GFD. Furthermore, one in three adult patients will report persistent symptoms and two in three will not achieve full histological recovery when on a GFD. In recent years, there has been extensive research conducted in the quest to find the holy grail of pharmacological treatment for CD. This review will present a concise description of the current rationale and main clinical trials related to CD drug therapy.


Asunto(s)
Enfermedad Celíaca , Adulto , Humanos , Enfermedad Celíaca/diagnóstico , Calidad de Vida , Glútenes , Dieta Sin Gluten , Alimentos
9.
Case Rep Gastroenterol ; 16(2): 452-455, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36157610

RESUMEN

A 42-year-old man with no relevant past medical history presented with intermittent mild icterus and no signs of chronic liver disease. Laboratory tests were notable for hyperbilirubinemia (total 7.97 mg/dL, direct 5.37 mg/dL), bilirubinuria, no signs of hemolysis, normal liver tests and lipids profile. Abdominal ultrasound was unremarkable. A panel of chronic liver diseases was negative except for increased serum (147.4 µg/dL) and urinary (179 µg/24 h) copper, with normal ceruloplasmin. No other Leipzig criteria for Wilson's disease were found, including a negative test for ATP7B gene mutations (by exome sequencing). Total urinary coproporphyrin was normal with predominance of isomer I (86% of total urinary coproporphyrin output). Clinical and laboratorial profile was compatible with Dubin-Johnson syndrome; however, exome sequencing and search for deletions in the ABBC2 gene (encoding MRP2) only found a heterozygous potentially pathogenic variant (c.1483A>G - p.Lys495Glu). Additional extended molecular analysis of genes implicated in bilirubin metabolism found a homozygous deletion of a region encompassing exons 4-16 of SLCO1B3 gene (encoding OATP1B3) and all SLCO1B1 exons (encoding OATP1B1), thereby establishing Rotor syndrome diagnosis. Rotor and Dubin-Johnson syndromes are rare autosomal recessive liver diseases characterized by chronic conjugated hyperbilirubinemia, caused by the absence of the hepatic function OATP1B1/B3 (leading to impaired hepatic bilirubin reuptake and storage) and MRP2 transporters (leading to impaired hepatic bilirubin excretion), respectively. We report a case of compound hereditary hyperbilirubinemia with a misleading presentation with special focus on its diagnosis, particularly the advantage of extensive unbiased genetic testing by dedicated laboratories. With this case, we aim to highlight the necessity of establishing a diagnosis, reassuring the patient, and avoiding unnecessary invasive and costly diagnostic procedures.

10.
United European Gastroenterol J ; 10(7): 745-753, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35860851

RESUMEN

Bilirubin, a breakdown product of heme, is normally glucuronidated and excreted by the liver into bile. Failure of this system can lead to a buildup of conjugated bilirubin in the blood, resulting in jaundice. Hyperbilirubinemia is an important clinical sign that needs to be investigated under a stepwise evaluation. Inherited non-hemolytic conjugated hyperbilirubinemic conditions include Dubin-Johnson syndrome (caused by mutations affecting ABCC2 gene) and Rotor syndrome (caused by the simultaneous presence of mutations in SLCO1B1 and SLCO1B3 genes). Although classically viewed as benign conditions requiring no treatment, they lately gained an increased interest since recent studies suggested that mutations in the responsible genes leading to hyperbilirubinemia, as well as minor genetic variants, may result in an increased susceptibility to drug toxicity. This article provides a comprehensive review on the pathophysiology of Dubin-Johnson and Rotor syndromes, presenting the current knowledge concerning the molecular details and basis of these conditions.


Asunto(s)
Hiperbilirrubinemia Hereditaria , Ictericia Idiopática Crónica , Bilirrubina , Hemo/metabolismo , Humanos , Hiperbilirrubinemia/etiología , Hiperbilirrubinemia/genética , Hiperbilirrubinemia Hereditaria/diagnóstico , Hiperbilirrubinemia Hereditaria/genética , Ictericia Idiopática Crónica/diagnóstico , Ictericia Idiopática Crónica/genética , Transportador 1 de Anión Orgánico Específico del Hígado/genética
11.
Diabetes Metab Syndr Obes ; 14: 3627-3645, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34408459

RESUMEN

Sedentarism is the pandemic of modern times. It is associated with several medical conditions including obesity, type 2 diabetes mellitus, cardiovascular diseases and also liver disease, particularly metabolic dysfunction associated fatty liver disease (MAFLD). In an era when MAFLD is the most prevalent chronic liver disease worldwide, whilst no pharmacological therapy has been approved for it, exercise has proved to be effective in improving liver steatosis. Interestingly, exercise decreases liver fat even in the absence of weight loss. The challenge for the clinician is to motivate the obese patient with MAFLD, and associated co-morbidities, who has crystallized a sedentary behavior, at times when every need is at the distance of a click on the Internet, and the entire world can be visited behind a screen. In this review, the aggregate evidence on the mechanisms and effects of exercise in the management of MAFLD is summarized, with simple recommendations for everyday clinical practice.

12.
PLoS One ; 16(5): e0252218, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34033642

RESUMEN

BACKGROUND: Most long-term heavy drinkers do not have clinically evident chronic liver disease (CLD). However, at any time-point, their risk of developing CLD remains unknown. We aimed to evaluate the long-term outcomes of a group of heavy drinkers, without evidence of CLD at baseline. METHODS: A cohort of 123 long-term heavy drinkers without CLD were prospectively recruited in 2002 and retrospectively followed until 2018. RESULTS: At baseline (2002), median alcohol consumption was 271±203g/day during 21.5±20 years, 65% being abstinent during the previous 1.75±5 months. Patients were followed for 14±3 years. During follow-up, 53% reported any alcohol intake. Alcohol consumption during follow-up associated weakly with either 1- or 6-months previous abstinence at baseline. Until 2018, progression to CLD occurred in 6%, associating with years of alcohol intake during follow-up (OR 1.15 [1.01-1.31]) and baseline alkaline-phosphatase (OR 1.05 [1.01-1.10]). During follow-up, being abstinent for at least 1 year positively associated with CLD-free survival. 27% died (55% of cancer-mostly oropharyngeal cancer, 27% of cardiovascular disease, and 9% of liver disease), with a mean age of 71 years [69-74] (10 years less than the expected in the Portuguese population). Achieving abstinence for at least 1 year positively associated with overall survival, while smoking, and hepatic steatosis at baseline associated negatively. CONCLUSION: Long-term heavy drinkers seemed to have a decreased life expectancy compared with the overall Portuguese population. Cancer was the main cause of death. Our results suggest that progression to CLD depends mostly on continued alcohol intake. Alcohol abstinence, even if temporary, seems to decrease the risks of CLD and mortality.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Hepatopatías/mortalidad , Neoplasias/mortalidad , Fosfatasa Alcalina/metabolismo , Enfermedades Cardiovasculares/metabolismo , Humanos , Hepatopatías/epidemiología , Neoplasias/epidemiología , Estudios Prospectivos , Factores de Riesgo
13.
Eur J Gastroenterol Hepatol ; 33(1S Suppl 1): e883-e897, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-35048655

RESUMEN

INTRODUCTION: Frailty and sarcopenia associate with increased mortality in patients with liver cirrhosis on the transplant waitlist. We conducted a systematic review on the impact of pretransplant frailty and sarcopenia on post-transplant outcomes in adult patients with liver cirrhosis. METHODS: We performed a search in Medline, Embase and Cochrane Central. Of the 12276 references initially recovered, 34 were included. RESULTS: Frailty and sarcopenia presented a negative impact on post-transplant outcomes and seemed to associate with an overall two-fold reduction in early and 50% reduction in late survival, for severe conditions, according to the largest cohorts. These patients required longer ICU and hospitalization time, had higher rates of sepsis and respiratory complications and lower graft-survival. The reversibility of frailty depended on the severity of functional impairment and on the co-morbidities contributing to frailty. Reversibility of sarcopenia occurred in only a minority of patients, in unbiased studies. CONCLUSION: Frailty and sarcopenia are double-edged swords: patients with frailty/sarcopenia should be prioritized for liver transplantation due to increased mortality on the waitlist; however, severe frailty/sarcopenia may justify delisting because it associates with dismal prognosis post-liver transplantation. Patients presenting mild to moderate frailty/sarcopenia, should be submitted to liver transplantation before those conditions worsen to a level that significantly impacts post-liver transplantation outcomes.


Asunto(s)
Fragilidad , Trasplante de Hígado , Sarcopenia , Adulto , Fragilidad/complicaciones , Fragilidad/diagnóstico , Humanos , Cirrosis Hepática/complicaciones , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/cirugía , Trasplante de Hígado/efectos adversos , Pronóstico , Sarcopenia/etiología
15.
Expert Rev Gastroenterol Hepatol ; 12(4): 351-367, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29224471

RESUMEN

INTRODUCTION: Nonalcoholic fatty liver disease (NAFLD) encompasses simple steatosis and steatohepatitis (NASH) with or without fibrosis/cirrhosis and hepatocellular carcinoma. NAFLD occurs epidemically in most areas of the world, contributes to cardiovascular events and liver-related mortality and therefore exacts a major economic toll. Areas covered: Here we summarize what clinicians should know about NAFLD histopathology in adults. We report on the individual histological features and scoring systems of NAFLD: the NAFLD activity score (NAS) introduced by the NASH-Clinical Research Network, the 'Fatty Liver Inhibition of Progression' algorithm and Steatosis, Activity, and Fibrosis (SAF) score. Pros and cons of histological classifications in NASH are discussed. Special emphasis is given to liver histopathology in some high-risk patient groups, such as those with severe obesity and type 2 diabetes. Moreover, we also examine the relationship between liver histopathology and clinical features, and the impact of liver histopathology on the long-term prognosis of NAFLD. Finally, we propose an integrated diagnostic approach which utilizes both non-invasive tools and liver biopsy in those individual patients with suspected NAFLD. Expert commentary: Based on expert opinions, we conclude with a research agenda on NAFLD which focuses on the most burning topics to be addressed over the next five years.


Asunto(s)
Carcinoma Hepatocelular/patología , Cirrosis Hepática/patología , Neoplasias Hepáticas/patología , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/patología , Biopsia , Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/epidemiología , Humanos , Cirrosis Hepática/diagnóstico por imagen , Cirrosis Hepática/epidemiología , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/epidemiología , Enfermedad del Hígado Graso no Alcohólico/clasificación , Enfermedad del Hígado Graso no Alcohólico/diagnóstico por imagen , Enfermedad del Hígado Graso no Alcohólico/epidemiología , Valor Predictivo de las Pruebas , Pronóstico , Factores de Riesgo , Índice de Severidad de la Enfermedad
16.
J Hepatol ; 68(3): 550-562, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29107151

RESUMEN

Liver disease remains a leading cause of mortality worldwide despite recent successes in the field of viral hepatitis, because increases in alcohol consumption and obesity are fuelling an epidemic of chronic fatty liver disease for which there are currently no effective medical therapies. About 20% of individuals with chronic liver injury ultimately develop end-stage liver disease due to cirrhosis. Hence, treatments to prevent and reverse cirrhosis in individuals with ongoing liver injury are desperately needed. The development of successful treatments requires an improved understanding of the mechanisms controlling liver disease progression. The liver responds to diverse insults with a conserved wound healing response, suggesting that it might be generally beneficial to optimise pathways that are crucial for effective liver repair. The Hedgehog pathway has emerged as a potential target based on compelling preclinical and clinical data, which demonstrate that it critically regulates the liver's response to injury. Herein, we will summarise evidence of the Hedgehog pathway's role in liver disease and discuss how modulating pathway activity might be applied to improve liver disease outcomes.


Asunto(s)
Proteínas Hedgehog/fisiología , Hepatopatías , Transducción de Señal , Manejo de la Enfermedad , Humanos , Hepatopatías/metabolismo , Hepatopatías/terapia , Pronóstico
17.
BMC Cell Biol ; 18(1): 20, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28427343

RESUMEN

BACKGROUND: Obesity is associated with multiple diseases, but it is unclear how obesity promotes progressive tissue damage. Recovery from injury requires repair, an energy-expensive process that is coupled to energy availability at the cellular level. The satiety factor, leptin, is a key component of the sensor that matches cellular energy utilization to available energy supplies. Leptin deficiency signals energy depletion, whereas activating the Hedgehog pathway drives energy-consuming activities. Tissue repair is impaired in mice that are obese due to genetic leptin deficiency. Tissue repair is also blocked and obesity enhanced by inhibiting Hedgehog activity. We evaluated the hypothesis that loss of leptin silences Hedgehog signaling in pericytes, multipotent leptin-target cells that regulate a variety of responses that are often defective in obesity, including tissue repair and adipocyte differentiation. RESULTS: We found that pericytes from liver and white adipose tissue require leptin to maintain expression of the Hedgehog co-receptor, Smoothened, which controls the activities of Hedgehog-regulated Gli transcription factors that orchestrate gene expression programs that dictate pericyte fate. Smoothened suppression prevents liver pericytes from being reprogrammed into myofibroblasts, but stimulates adipose-derived pericytes to become white adipocytes. Progressive Hedgehog pathway decay promotes senescence in leptin-deficient liver pericytes, which, in turn, generate paracrine signals that cause neighboring hepatocytes to become fatty and less proliferative, enhancing vulnerability to liver damage. CONCLUSIONS: Leptin-responsive pericytes evaluate energy availability to inform tissue construction by modulating Hedgehog pathway activity and thus, are at the root of progressive obesity-related tissue pathology. Leptin deficiency inhibits Hedgehog signaling in pericytes to trigger a pericytopathy that promotes both adiposity and obesity-related tissue damage.


Asunto(s)
Células Estrelladas Hepáticas/fisiología , Leptina/genética , Obesidad/fisiopatología , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/citología , Animales , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Proteínas Hedgehog/fisiología , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/metabolismo , Leptina/deficiencia , Leptina/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Obesos , Miofibroblastos/citología , Miofibroblastos/metabolismo , Obesidad/genética , Comunicación Paracrina/genética , Receptores de Leptina/metabolismo , Receptor Smoothened/agonistas
18.
Int J Mol Sci ; 17(4): 481, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27043550

RESUMEN

Recently, the importance of the gut-liver-adipose tissue axis has become evident. Nonalcoholic fatty liver disease (NAFLD) is the hepatic disease of a systemic metabolic disorder that radiates from energy-surplus induced adiposopathy. The gut microbiota has tremendous influences in our whole-body metabolism, and is crucial for our well-being and health. Microorganisms precede humans in more than 400 million years and our guest flora evolved with us in order to help us face aggressor microorganisms, to help us maximize the energy that can be extracted from nutrients, and to produce essential nutrients/vitamins that we are not equipped to produce. However, our gut microbiota can be disturbed, dysbiota, and become itself a source of stress and injury. Dysbiota may adversely impact metabolism and immune responses favoring obesity and obesity-related disorders such as insulin resistance/diabetes mellitus and NAFLD. In this review, we will summarize the latest evidence of the role of microbiota/dysbiota in diet-induced obesity and NAFLD, as well as the potential therapeutic role of targeting the microbiota in this set.


Asunto(s)
Dieta , Microbiota , Enfermedad del Hígado Graso no Alcohólico/microbiología , Obesidad/microbiología , Animales , Humanos , Intestinos/microbiología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/patología , Probióticos/administración & dosificación
19.
Gastroenterology ; 150(8): 1769-77, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26928243

RESUMEN

Nonalcoholic steatohepatitis (NASH) is a necro-inflammatory response that ensues when hepatocytes are injured by lipids (lipotoxicity). NASH is a potential outcome of nonalcoholic fatty liver (NAFL), a condition that occurs when lipids accumulate in hepatocytes. NASH may be reversible, but it can also result in cirrhosis and primary liver cancer. We are beginning to learn about the mechanisms of progression of NAFL and NASH. NAFL does not inevitably lead to NASH because NAFL is a heterogeneous condition. This heterogeneity exists because different types of lipids with different cytotoxic potential accumulate in the NAFL, and individuals with NAFL differ in their ability to defend against lipotoxicity. There are no tests that reliably predict which patients with NAFL will develop lipotoxicity. However, NASH encompasses the spectrum of wound-healing responses induced by lipotoxic hepatocytes. Differences in these wound-healing responses among individuals determine whether lipotoxic livers regenerate, leading to stabilization or resolution of NASH, or develop progressive scarring, cirrhosis, and possibly liver cancer. We review concepts that are central to the pathogenesis of NASH.


Asunto(s)
Progresión de la Enfermedad , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Lípidos/toxicidad , Enfermedad del Hígado Graso no Alcohólico/etiología , Humanos , Hígado/citología , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo
20.
GE Port J Gastroenterol ; 23(3): 126-129, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28868447
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA