Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Genes Dev ; 35(1-2): 133-146, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33334822

RESUMO

The cJun NH2-terminal kinase (JNK) signaling pathway is activated by metabolic stress and promotes the development of metabolic syndrome, including hyperglycemia, hyperlipidemia, and insulin resistance. This integrated physiological response involves cross-talk between different organs. Here we demonstrate that JNK signaling in adipocytes causes an increased circulating concentration of the hepatokine fibroblast growth factor 21 (FGF21) that regulates systemic metabolism. The mechanism of organ crosstalk is mediated by a feed-forward regulatory loop caused by JNK-regulated FGF21 autocrine signaling in adipocytes that promotes increased expression of the adipokine adiponectin and subsequent hepatic expression of the hormone FGF21. The mechanism of organ cross-talk places circulating adiponectin downstream of autocrine FGF21 expressed by adipocytes and upstream of endocrine FGF21 expressed by hepatocytes. This regulatory loop represents a novel signaling paradigm that connects autocrine and endocrine signaling modes of the same hormone in different tissues.


Assuntos
Tecido Adiposo/fisiologia , Comunicação Autócrina/genética , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica/genética , Transdução de Sinais/genética , Adipócitos/metabolismo , Adiponectina/metabolismo , Tecido Adiposo/fisiopatologia , Animais , Sistema Endócrino/metabolismo , Metabolismo Energético/genética , Retroalimentação Fisiológica/fisiologia , Fatores de Crescimento de Fibroblastos/sangue , Hepatócitos/metabolismo , Resistência à Insulina/genética , Fígado/metabolismo , MAP Quinase Quinase 4/deficiência , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos
2.
Development ; 150(20)2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-37260408

RESUMO

Nutritional and metabolic cues are integral to animal development. Organisms use them both as sustenance and environmental indicators, fueling, informing and influencing developmental decisions. Classical examples, such as the Warburg effect, clearly illustrate how genetic programs control metabolic changes. However, the way that nutrition and metabolism can also modulate or drive genetic programs to instruct developmental trajectories is much more elusive, owing to several difficulties including uncoupling permissive and instructive functions. Here, we discuss recent advancements in the field that highlight the developmental role of nutritional and metabolic cues across multiple levels of organismal complexity.


Assuntos
Fenômenos Fisiológicos da Nutrição , Animais
3.
Neurourol Urodyn ; 43(5): 1066-1074, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38289317

RESUMO

INTRODUCTION: Lower urinary tract symptoms (LUTSs) are a diverse array of urinary and pelvic dysfunctions that can emerge from childhood, extend through adulthood, and persist into older age. This narrative review aims to provide a comprehensive perspective on the continuum of LUTS and shed light on the underlying mechanisms and clinical implications that span across the lower urinary tract. METHODS: A panel of five experts from Belgium, the Netherlands, India, Denmark, and the United States participated in an intensive research to explore and pinpoint existing insights into the lifelong concept of LUTS, particularly at the pelvic level. The experts reviewed the existing literature and held a webinar to discuss their findings. RESULTS: Childhood LUTS can persist, resolve, or progress into bladder underactivity, dysfunctional voiding, or pain syndromes. The Lifelong character can be explained by pelvic organ cross-talk facilitated through complex neurological and nonneurological interactions. At the molecular level, the role of vasopressin receptors in the bladder's modulation and their potential relevance to therapeutic strategies for LUTS are explored. Frailty emerges as a parallel concept to lifelong LUTS, with a complex and synergistic relationship. Frailty, not solely an age-related condition, accentuates LUTS severity with insufficient evidence regarding the effectiveness and safety profile of the available therapeutic modalities. CONCLUSION: Understanding lifelong LUTSs offers insights into genetic, anatomical, neurological, and molecular mechanisms. Further research could identify predictive biomarkers, elucidate the role of clinically translatable elements in pelvic cross-talk, and uncover molecular signatures for personalized management.


Assuntos
Sintomas do Trato Urinário Inferior , Bexiga Urinária , Humanos , Sintomas do Trato Urinário Inferior/fisiopatologia , Bexiga Urinária/fisiopatologia
4.
BMC Med Inform Decis Mak ; 24(1): 35, 2024 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-38310256

RESUMO

OBJECTIVE: Acute kidney injury (AKI) is a clinical syndrome that occurs as a result of a dramatic decline in kidney function caused by a variety of etiological factors. Its main biomarkers, serum creatinine and urine output, are not effective in diagnosing early AKI. For this reason, this study provides insight into this syndrome by exploring the comorbidities of AKI, which may facilitate the early diagnosis of AKI. In addition, organ crosstalk in AKI was systematically explored based on comorbidities to obtain clinically reliable results. METHODS: We collected data from the Medical Information Mart for Intensive Care-IV database on patients aged [Formula: see text] 18 years in intensive care units (ICU) who were diagnosed with AKI using the criteria proposed by Kidney Disease: Improving Global Outcomes. The Apriori algorithm was used to mine association rules on the diagnoses of 55,486 AKI and non-AKI patients in the ICU. The comorbidities of AKI mined were validated through the Electronic Intensive Care Unit database, the Colombian Open Health Database, and medical literature, after which comorbidity results were visualized using a disease network. Finally, organ diseases were identified and classified from comorbidities to investigate renal crosstalk with other distant organs in AKI. RESULTS: We found 579 AKI comorbidities, and the main ones were disorders of lipoprotein metabolism, essential hypertension, and disorders of fluid, electrolyte, and acid-base balance. Of the 579 comorbidities, 554 were verifiable and 25 were new and not previously reported. In addition, crosstalk between the kidneys and distant non-renal organs including the liver, heart, brain, lungs, and gut was observed in AKI with the strongest heart-kidney crosstalk, followed by lung-kidney crosstalk. CONCLUSION: The comorbidities mined in this study using association rules are scientific and may be used for the early diagnosis of AKI and the construction of AKI predictive models. Furthermore, the organ crosstalk results obtained through comorbidities may provide supporting information for the management of short- and long-term treatment practices for organ dysfunction.


Assuntos
Injúria Renal Aguda , Classificação Internacional de Doenças , Humanos , Idoso , Estudos Prospectivos , Injúria Renal Aguda/diagnóstico , Injúria Renal Aguda/epidemiologia , Injúria Renal Aguda/etiologia , Comorbidade , Biomarcadores , Unidades de Terapia Intensiva
5.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-39000150

RESUMO

Neuroinflammation, crucial in neurological disorders like Alzheimer's disease, multiple sclerosis, and hepatic encephalopathy, involves complex immune responses. Extracellular vesicles (EVs) play a pivotal role in intercellular and inter-organ communication, influencing disease progression. EVs serve as key mediators in the immune system, containing molecules capable of activating molecular pathways that exacerbate neuroinflammatory processes in neurological disorders. However, EVs from mesenchymal stem cells show promise in reducing neuroinflammation and cognitive deficits. EVs can cross CNS barriers, and peripheral immune signals can influence brain function via EV-mediated communication, impacting barrier function and neuroinflammatory responses. Understanding EV interactions within the brain and other organs could unveil novel therapeutic targets for neurological disorders.


Assuntos
Vesículas Extracelulares , Doenças Neuroinflamatórias , Vesículas Extracelulares/metabolismo , Humanos , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/patologia , Animais , Comunicação Celular , Encéfalo/metabolismo , Encéfalo/patologia , Células-Tronco Mesenquimais/metabolismo , Barreira Hematoencefálica/metabolismo , Inflamação/metabolismo , Inflamação/patologia
6.
Proteomics ; : e2300078, 2023 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-37525338

RESUMO

Lifestyle modification represents the first-line strategy for the prevention and treatment of type 2 diabetes mellitus (T2DM), which is frequently associated with obesity and characterized by defective pancreatic insulin secretion and/or insulin resistance. Exercise training is an essential component of lifestyle modification and has been shown to ameliorate insulin resistance by reducing body fat mass and by enhancing skeletal muscle mitochondrial biogenesis and insulin-independent glucose uptake. Additionally, exercising stimulates the release of exerkines such as metabolites or cytokines, but also long non-coding RNA, microRNAs, cell-free DNA (cf-DNA), and extracellular vesicles (EVs), which contribute to inter-tissue communication. There is emerging evidence that EV number and content are altered in obesity and T2DM and may be involved in several metabolic processes, specifically either worsening or improving insulin resistance. This review summarizes the current knowledge on the metabolic effects of exercise training and on the potential role of humoral factors and EV as new biomarkers for early diagnosis and tailored treatment of T2DM.

7.
Rev Cardiovasc Med ; 24(2): 40, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39077395

RESUMO

Acute cardiorenal syndrome (CRS) is often observed in patients with acute kidney injury (AKI) in the cardiac intensive care unit and is reported to be associated with poor prognosis. Volume disorder or re-distribution, renin-angiotensin-aldosterone system activation, and neurohormonal and sympathetic nervous system activation have been suggested to be related to the occurrence of acute CRS. There is a lack of biomarkers that can identify changes in renal function in patients with acute CRS. Evidence-based medications are limited in the management of acute CRS in AKI. Therefore, we reviewed the epidemiology, pathophysiology, clinical assessment, and treatment of acute CRS in AKI.

8.
Int Immunol ; 34(2): 81-95, 2022 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-34173833

RESUMO

The heart is highly innervated by autonomic neurons, and dynamic autonomic regulation of the heart and blood vessels is essential for animals to carry out the normal activities of life. Cardiovascular diseases, including heart failure and myocardial infarction, are characterized in part by an imbalance in autonomic nervous system activation, with excess sympathetic and diminished parasympathetic activation. Notably, however, this is often accompanied by chronic inflammation within the cardiovascular tissues, which suggests there are interactions between autonomic dysregulation and inflammation. Recent studies have been unraveling the mechanistic links between autonomic nerves and immune cells within the cardiovascular system. The autonomic nervous system and immune system also act in concert to coordinate the actions of multiple organs that not only maintain homeostasis but also likely play key roles in disease-disease interactions, such as cardiorenal syndrome and multimorbidity. In this review, we summarize the physiological and pathological interactions between autonomic nerves and macrophages in the context of cardiovascular disease.


Assuntos
Doenças Cardiovasculares , Animais , Sistema Nervoso Autônomo/fisiologia , Coração/inervação , Inflamação , Macrófagos
9.
Calcif Tissue Int ; 113(1): 21-38, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37193929

RESUMO

Maintenance of skeletal health is tightly regulated by osteocytes, osteoblasts, and osteoclasts via coordinated secretion of bone-derived factors, termed osteokines. Disruption of this coordinated process due to aging and metabolic disease promotes loss of bone mass and increased risk of fracture. Indeed, growing evidence demonstrates that metabolic diseases, including type 2 diabetes, liver disease and cancer are accompanied by bone loss and altered osteokine levels. With the persistent prevalence of cancer and the growing epidemic of metabolic disorders, investigations into the role of inter-tissue communication during disease progression are on the rise. While osteokines are imperative for bone homeostasis, work from us and others have identified that osteokines possess endocrine functions, exerting effects on distant tissues including skeletal muscle and liver. In this review we first discuss the prevalence of bone loss and osteokine alterations in patients with type 2 diabetes, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, cirrhosis, and cancer. We then discuss the effects of osteokines in mediating skeletal muscle and liver homeostasis, including RANKL, sclerostin, osteocalcin, FGF23, PGE2, TGF-ß, BMPs, IGF-1 and PTHrP. To better understand how inter-tissue communication contributes to disease progression, it is essential that we include the bone secretome and the systemic roles of osteokines.


Assuntos
Doenças Ósseas Metabólicas , Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Osso e Ossos , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Densidade Óssea , Doenças Ósseas Metabólicas/metabolismo
10.
J Biol Chem ; 296: 100575, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33757768

RESUMO

How organs sense circulating metabolites is a key question. Here, we show that the multispecific organic anion transporters of drugs, OAT1 (SLC22A6 or NKT) and OAT3 (SLC22A8), play a role in organ sensing. Metabolomics analyses of the serum of Oat1 and Oat3 knockout mice revealed changes in tryptophan derivatives involved in metabolism and signaling. Several of these metabolites are derived from the gut microbiome and are implicated as uremic toxins in chronic kidney disease. Direct interaction with the transporters was supported with cell-based transport assays. To assess the impact of the loss of OAT1 or OAT3 function on the kidney, an organ where these uptake transporters are highly expressed, knockout transcriptomic data were mapped onto a "metabolic task"-based computational model that evaluates over 150 cellular functions. Despite the changes of tryptophan metabolites in both knockouts, only in the Oat1 knockout were multiple tryptophan-related cellular functions increased. Thus, deprived of the ability to take up kynurenine, kynurenate, anthranilate, and N-formylanthranilate through OAT1, the kidney responds by activating its own tryptophan-related biosynthetic pathways. The results support the Remote Sensing and Signaling Theory, which describes how "drug" transporters help optimize levels of metabolites and signaling molecules by facilitating organ cross talk. Since OAT1 and OAT3 are inhibited by many drugs, the data implies potential for drug-metabolite interactions. Indeed, treatment of humans with probenecid, an OAT-inhibitor used to treat gout, elevated circulating tryptophan metabolites. Furthermore, given that regulatory agencies have recommended drugs be tested for OAT1 and OAT3 binding or transport, it follows that these metabolites can be used as endogenous biomarkers to determine if drug candidates interact with OAT1 and/or OAT3.


Assuntos
Rim/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Triptofano/metabolismo , Animais , Rim/citologia , Camundongos , Estresse Oxidativo , Transporte Proteico , Transdução de Sinais
11.
EMBO J ; 37(22)2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30389664

RESUMO

The endoplasmic reticulum-associated protein degradation (ERAD) is responsible for recognizing and retro-translocating protein substrates, misfolded or not, from the ER for cytosolic proteasomal degradation. HMG-CoA Reductase (HMGCR) Degradation protein-HRD1-was initially identified as an E3 ligase critical for ERAD. However, its physiological functions remain largely undefined. Herein, we discovered that hepatic HRD1 expression is induced in the postprandial condition upon mouse refeeding. Mice with liver-specific HRD1 deletion failed to repress FGF21 production in serum and liver even in the refeeding condition and phenocopy the FGF21 gain-of-function mice showing growth retardation, female infertility, and diurnal circadian behavior disruption. HRD1-ERAD facilitates the degradation of the liver-specific ER-tethered transcription factor CREBH to downregulate FGF21 expression. HRD1-ERAD catalyzes polyubiquitin conjugation onto CREBH at lysine 294 for its proteasomal degradation, bridging a multi-organ crosstalk in regulating growth, circadian behavior, and female fertility through regulating the CREBH-FGF21 regulatory axis.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Degradação Associada com o Retículo Endoplasmático , Fatores de Crescimento de Fibroblastos/biossíntese , Fígado/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Feminino , Fertilidade/genética , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Células HEK293 , Humanos , Fígado/patologia , Masculino , Camundongos , Camundongos Transgênicos , Poliubiquitina/genética , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/genética
12.
Nephrol Dial Transplant ; 37(7): 1218-1228, 2022 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-33527986

RESUMO

Several theories regarding acute kidney injury (AKI)-related mortality have been entertained, although mounting evidence supports the paradigm that impaired kidney function directly and adversely affects the function of several remote organs. The kidneys and liver are fundamental to human metabolism and detoxification, and it is therefore hardly surprising that critical illness complicated by hepatorenal dysfunction portends a poor prognosis. Several diseases can simultaneously impact the proper functioning of the liver and kidneys, although this review will address the impact of AKI on liver function. While evidence for this relationship in humans remains sparse, we present supportive studies and then discuss the most likely mechanisms by which AKI can cause liver dysfunction. These include 'traditional' complications of AKI (uremia, volume overload and acute metabolic acidosis, among others) as well as systemic inflammation, hepatic leukocyte infiltration, cytokine-mediated liver injury and hepatic oxidative stress. We conclude by addressing the therapeutic implications of these findings to clinical medicine.


Assuntos
Injúria Renal Aguda , Hepatopatias , Injúria Renal Aguda/terapia , Estado Terminal , Humanos , Rim , Hepatopatias/etiologia
13.
BMC Nephrol ; 23(1): 202, 2022 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-35659594

RESUMO

BACKGROUND: In critically ill patients, acute pain occurs frequently, causes sympathetic activation, release of inflammatory mediators, and potential organ dysfunction, with the kidneys potentially sensitive to inflammation-mediated injury. This study aimed to explore the association between acute pain in critically ill patients and the occurrence of acute kidney injury (AKI). METHODS: Data from a retrospective cohort of adult patients admitted between June 2013 and June 2016 to the Intensive Care Unit (ICU) of a tertiary hospital in São Paulo, Brazil, were analyzed. The main exclusion criteria were ICU length of stay < 48 h, coma, and prior kidney dysfunction. The outcome (AKI) was defined as an elevation in the baseline serum creatinine level of ≥ 0.3 mg/dl and/or > 50% at any time after the first 48 h in the ICU. Multivariable logistic regression and hierarchical cluster analysis were performed. RESULTS: The isolated incidence of pain was 23.6%, and the incidence of pain duration > 5 days was 10.6%. AKI occurred in 31.7% of the cohort. In multivariable logistic analysis, duration of pain > 5 days (OR 5.25 CI 2.19-12.57 p < 0.01) and mechanical ventilation (MV) ≥ 3 days (OR 5.5 CI 2.3-13.5 p < 0.01) were the variables with positive association with AKI. The hierarchical cluster analysis reinforced the relation between AKI, MV and duration of pain. CONCLUSIONS: Pain is an especially important issue in critically ill patients and in this exploratory study it appears to be associated with AKI development. The search for more rigorous pain control in ICU is crucial and can influence organ dysfunction.


Assuntos
Injúria Renal Aguda , Dor Aguda , Injúria Renal Aguda/etiologia , Dor Aguda/complicações , Adulto , Brasil/epidemiologia , Estudos de Coortes , Estado Terminal/epidemiologia , Humanos , Unidades de Terapia Intensiva , Insuficiência de Múltiplos Órgãos , Estudos Retrospectivos , Fatores de Risco
14.
Int J Mol Sci ; 23(15)2022 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-35897797

RESUMO

The prevalence of non-alcoholic steatohepatitis (NASH) is rapidly increasing and associated with cardiovascular disease (CVD), the major cause of mortality in NASH patients. Although sharing common risk factors, the mechanisms by which NASH may directly contribute to the development to CVD remain poorly understood. The aim of this study is to gain insight into key molecular processes of NASH that drive atherosclerosis development. Thereto, a time-course study was performed in Ldlr-/-.Leiden mice fed a high-fat diet to induce NASH and atherosclerosis. The effects on NASH and atherosclerosis were assessed and transcriptome analysis was performed. Ldlr-/-.Leiden mice developed obesity, hyperlipidemia and insulin resistance, with steatosis and hepatic inflammation preceding atherosclerosis development. Transcriptome analysis revealed a time-dependent increase in pathways related to NASH and fibrosis followed by an increase in pro-atherogenic processes in the aorta. Gene regulatory network analysis identified specific liver regulators related to lipid metabolism (SC5D, LCAT and HMGCR), inflammation (IL1A) and fibrosis (PDGF, COL3A1), linked to a set of aorta target genes related to vascular inflammation (TNFA) and atherosclerosis signaling (CCL2 and FDFT1). The present study reveals pathogenic liver processes that precede atherosclerosis development and identifies hepatic key regulators driving the atherogenic pathways and regulators in the aorta.


Assuntos
Aterosclerose , Hepatopatia Gordurosa não Alcoólica , Animais , Aterosclerose/genética , Aterosclerose/patologia , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Fibrose , Inflamação/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/genética
15.
Int J Mol Sci ; 23(9)2022 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-35563026

RESUMO

The skeletal muscle is the largest organ in the body and secretes circulating factors, including myokines, which are involved in various cellular signaling processes. Skeletal muscle is vital for metabolism and physiology and plays a crucial role in insulin-mediated glucose disposal. Myokines have autocrine, paracrine, and endocrine functions, serving as critical regulators of myogenic differentiation, fiber-type switching, and maintaining muscle mass. Myokines have profound effects on energy metabolism and inflammation, contributing to the pathophysiology of type 2 diabetes (T2D) and other metabolic diseases. Myokines have been shown to increase insulin sensitivity, thereby improving glucose disposal and regulating glucose and lipid metabolism. Many myokines have now been identified, and research on myokine signaling mechanisms and functions is rapidly emerging. This review summarizes the current state of the field regarding the role of myokines in tissue cross-talk, including their molecular mechanisms, and their potential as therapeutic targets for T2D.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Humanos , Resistência à Insulina/fisiologia , Músculo Esquelético/metabolismo
16.
Pflugers Arch ; 473(12): 1813-1839, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34553265

RESUMO

Duchenne muscular dystrophy is a highly progressive muscle wasting disorder due to primary abnormalities in one of the largest genes in the human genome, the DMD gene, which encodes various tissue-specific isoforms of the protein dystrophin. Although dystrophinopathies are classified as primary neuromuscular disorders, the body-wide abnormalities that are associated with this disorder and the occurrence of organ crosstalk suggest that a multi-systems pathophysiological view should be taken for a better overall understanding of the complex aetiology of X-linked muscular dystrophy. This article reviews the molecular and cellular effects of deficiency in dystrophin isoforms in relation to voluntary striated muscles, the cardio-respiratory system, the kidney, the liver, the gastrointestinal tract, the nervous system and the immune system. Based on the establishment of comprehensive biomarker signatures of X-linked muscular dystrophy using large-scale screening of both patient specimens and genetic animal models, this article also discusses the potential usefulness of novel disease markers for more inclusive approaches to differential diagnosis, prognosis and therapy monitoring that also take into account multi-systems aspects of dystrophinopathy. Current therapeutic approaches to combat muscular dystrophy are summarised.


Assuntos
Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Animais , Biomarcadores/metabolismo , Distrofina/metabolismo , Humanos , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Distrofia Muscular de Duchenne/metabolismo
17.
Tohoku J Exp Med ; 254(4): 245-252, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34373421

RESUMO

In insulin-resistant states such as obesity, pancreatic ß-cells proliferate to prevent blood glucose elevations. Failure of this ß-cells proliferative response leads to the development of diabetes. On the other hand, when organs are damaged, cells proliferate to repair the organs. Therefore, these proliferations are compensatory mechanisms aimed at maintaining whole-body homeostasis. We previously discovered vagal signal-mediated systems regulating adaptive proliferation of ß-cells and hepatocytes. Neuron-mediated liver-ß-cell inter-organ crosstalk is involved in promotion of ß-cell proliferation during obesity, and in this system, vagal signals directly stimulate ß-cell proliferation. Meanwhile, in the liver, the multi-step mechanisms whereby vagal nerve signals activate hepatic resident macrophages are involved in hepatocyte proliferation after severe injury. Diabetes mellitus develops on the pathological basis of insufficient insulin action. Insulin action insufficiency is attributable to insulin resistance, i.e., the failure of insulin to exert sufficient effects, and/or to impairment of insulin secretion. Impairment of insulin secretion is attributable not only to the ß-cell dysfunction but also to functional ß-cell mass reduction. In this regard, there are already therapeutic options to increase insulin secretion from residual ß-cells, such as sulfonyl urea and incretin-related drugs. In contrast, there are as yet no applicable therapeutic strategies to increase functional ß-cell mass in vivo. Therefore, we have conducted the basic investigations to tackle this issue based on the discovery of neuron-mediated liver-ß-cell inter-organ crosstalk. This review introduces vagal signal-mediated regulatory systems of adaptive cell proliferation in vivo and efforts to develop cell-increasing therapies based on vagal nerve-mediated cell proliferation.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Proliferação de Células , Homeostase , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo
18.
Int J Mol Sci ; 23(1)2021 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-35008586

RESUMO

Ischemic stroke is characterized by an occlusion of a cerebral blood vessel resulting in neuronal cell death due to nutritional and oxygen deficiency. Additionally, post-ischemic cell death is augmented after reperfusion. These events are paralleled by dysregulated miRNA expression profiles in the peri-infarct area. Understanding the underlying molecular mechanism in the peri-infarct region is crucial for developing promising therapeutics. Utilizing a tMCAo (transient Middle Cerebral Artery occlusion) model in rats, we studied the expression levels of the miRNAs (miR) 223-3p, 155-5p, 3473, and 448-5p in the cortex, amygdala, thalamus, and hippocampus of both the ipsi- and contralateral hemispheres. Additionally, the levels in the blood serum, spleen, and liver and the expression of their target genes, namely, Nlrp3, Socs1, Socs3, and Vegfa, were assessed. We observed an increase in all miRNAs on the ipsilateral side of the cerebral cortex in a time-dependent manner and increased miRNAs levels (miR-223-3p, miR-3473, and miR-448-5p) in the contralateral hemisphere after 72 h. Besides the cerebral cortex, the amygdala presented increased expression levels, whereas the thalamus and hippocampus showed no alterations. Different levels of the investigated miRNAs were detected in blood serum, liver, and spleen. The gene targets were altered not only in the peri-infarct area of the cortex but selectively increased in the investigated non-affected brain regions along with the spleen and liver during the reperfusion time up to 72 h. Our results suggest a supra-regional influence of miRNAs following ischemic stroke, which should be studied to further identify whether miRNAs are transported or locally upregulated.


Assuntos
Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Ataque Isquêmico Transitório/metabolismo , Fígado/metabolismo , MicroRNAs/metabolismo , Soro/metabolismo , Baço/metabolismo , Animais , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Ratos , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Acidente Vascular Cerebral/metabolismo
19.
Kidney Int ; 97(4): 741-752, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32061437

RESUMO

Acute kidney injury is a common complication of advanced liver disease and increased mortality of these patients. Here, we analyzed the role of Y-box protein-1 (YB-1), a nucleic acid binding protein, in the bile duct ligation model of liver fibrosis and monitored liver and subsequent kidney damage. Following bile duct ligation, both serum levels of liver enzymes and expression of hepatic extracellular matrix components such as type I collagen were significantly reduced in mice with half-maximal YB-1 expression (Yb1+/-) as compared to their wild-type littermates. By contrast, expression of the chemokine CXCL1 was significantly augmented in these Yb1+/- mice. YB-1 was identified as a potent transcriptional repressor of the Cxcl1 gene. Precision-cut kidney slices from Yb1+/- mice revealed higher expression of the CXCL1 receptor CXCR2 as well as enhanced responsivity to CXCL1 compared to those from wild-type mice. Increased CXCL1 content in Yb1+/- mice led to pronounced bile duct ligation-induced damage of the kidneys monitored as parameters of tubular epithelial injury and immune cell infiltration. Pharmacological blockade of CXCR2 as well as application of an inhibitory anti-CXCL1 antibody significantly mitigated early systemic effects on the kidneys following bile duct ligation whereas it had only a modest impact on hepatic inflammation and function. Thus, our analyses provide direct evidence that YB-1 crucially contributes to hepatic fibrosis and modulates liver-kidney crosstalk by maintaining tight control over chemokine CXCL1 expression.


Assuntos
Cirrose Hepática , Ácidos Nucleicos , Fatores de Transcrição , Animais , Proteínas de Transporte , Rim , Ligadura , Fígado/patologia , Cirrose Hepática/genética , Camundongos , Camundongos Endogâmicos C57BL
20.
Cell Tissue Res ; 380(2): 341-378, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31932949

RESUMO

The worldwide prevalence of diabetes mellitus and obesity is rapidly increasing not only in adults but also in children and adolescents. Diabetes is associated with macrovascular complications increasing the risk for cardiovascular disease and stroke, as well as microvascular complications leading to diabetic nephropathy, retinopathy and neuropathy. Animal models are essential for studying disease mechanisms and for developing and testing diagnostic procedures and therapeutic strategies. Rodent models are most widely used but have limitations in translational research. Porcine models have the potential to bridge the gap between basic studies and clinical trials in human patients. This article provides an overview of concepts for the development of porcine models for diabetes and obesity research, with a focus on genetically engineered models. Diabetes-associated ocular, cardiovascular and renal alterations observed in diabetic pig models are summarized and their similarities with complications in diabetic patients are discussed. Systematic multi-organ biobanking of porcine models of diabetes and obesity and molecular profiling of representative tissue samples on different levels, e.g., on the transcriptome, proteome, or metabolome level, is proposed as a strategy for discovering tissue-specific pathomechanisms and their molecular key drivers using systems biology tools. This is exemplified by a recent study providing multi-omics insights into functional changes of the liver in a transgenic pig model for insulin-deficient diabetes mellitus. Collectively, these approaches will provide a better understanding of organ crosstalk in diabetes mellitus and eventually reveal new molecular targets for the prevention, early diagnosis and treatment of diabetes mellitus and its associated complications.


Assuntos
Complicações do Diabetes/diagnóstico , Diabetes Mellitus Tipo 2/complicações , Animais , Modelos Animais de Doenças , Humanos , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA