Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Biochem Biophys Res Commun ; 525(3): 693-698, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32139120

RESUMO

The levels of the anti-aging protein α-Klotho, in its soluble form (s-Klotho), are depressed in the circulation of patients with type 1 diabetes (T1D) or type 2 diabetes (T2D). Gene transfer experiments have suggested a protective role for ß-cell specific expression of α-Klotho in murine models of T1D and T1D, but these approaches are not easily translatable to clinical therapy. It is unknown whether systemic s-Klotho protein treatment ameliorates disease in T1D, which is characterized by autoimmune destruction of ß cells. We previously reported from in vitro experiments with ß cells that s-Klotho increases insulin secretion, reduces cells death and promotes ß-cell replication. Here, we investigated s-Klotho protein therapy in NOD mice, which have autoimmune T1D. We observed that diabetic NOD mice have significantly lower plasma levels of s-Klotho, compared to their non-diabetic counterparts. To examine in vivo effects of Klotho, we treated NOD mice with s-Klotho protein, or with a Klotho blocking antibody. Systemic treatment with s-Klotho ameliorated diabetes; notably increasing ß-cell replication and total ß-cell mass. Klotho expression was increased locally in the islets. s-Klotho also markedly reduced immune-cell infiltration of islets (insulitis). In contrast, administration of the Klotho antibody was detrimental, and aggravated the loss of ß-cell mass. Thus, s-Klotho has protective effects in this model of T1D, and this appears to depend on a combination of increased ß-cell replication and reduced insulitis. These findings suggest that s-Klotho might be effective as a new therapeutic agent for T1D.


Assuntos
Glucuronidase/uso terapêutico , Células Secretoras de Insulina/patologia , Animais , Proliferação de Células , Feminino , Glucuronidase/sangue , Humanos , Proteínas Klotho , Camundongos Endogâmicos NOD , Tamanho do Órgão
2.
J Cell Physiol ; 234(9): 14432-14444, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30693506

RESUMO

Diabetes occurs when pancreatic ß-cell death exceeds ß-cell growth, which leads to loss of ß-cell mass. An effective therapy must have two actions: promotion of ß-cell replication and suppression of ß-cell death. Previous studies have established an important role for γ-aminobutyric acid (GABA) in islet-cell hormone homeostasis, as well as the maintenance of the ß-cell mass. GABA exerts paracrine actions on α cells in suppressing glucagon secretion, and it has autocrine actions on ß cells that increase insulin secretion. Multiple studies have shown that GABA increases the mitotic rate of ß cells. In mice, following ß-cell depletion with streptozotocin, GABA therapy can restore the ß-cell mass. Enhanced ß-cell replication appears to depend on growth and survival pathways involving Akt activation. Some studies have also suggested that it induces transdifferentiation of α cells into ß cells, but this has been disputed and requires further investigation. In addition to proliferative effects, GABA protects ß cells against injury and markedly reduces their apoptosis under a variety of conditions. The antiapoptotic effects depend at least in part on the enhancement of sirtuin-1 and Klotho activity, which both inhibit activation of the NF-κB inflammatory pathway. Importantly, in xenotransplanted human islets, GABA therapy stimulates ß-cell replication and insulin secretion. Thus, the intraislet GABAergic system is a target for the amelioration of diabetes therapy, including ß-cell survival and regeneration. GABA (or GABAergic drugs) can be combined with other antidiabetic drugs for greater effect.

3.
Biochem Biophys Res Commun ; 493(4): 1542-1547, 2017 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-28993191

RESUMO

Systemic gamma-aminobutyric acid (GABA) therapy prevents or ameliorates type 1 diabetes (T1D), by suppressing autoimmune responses and stimulating pancreatic beta cells. In beta cells, it increases insulin secretion, prevents apoptosis, and induces regeneration. It is unclear how GABA mediates these effects. We hypothesized that Klotho is involved. It is a multi-functional protein expressed in the kidneys, brain, pancreatic beta cells, other tissues, and is cell-bound or soluble. Klotho knockout mice display accelerated aging, and in humans Klotho circulating levels decline with age, renal disease and diabetes. Here, we report that GABA markedly increased circulating levels of Klotho in streptozotocin (STZ)-induced diabetes. GABA also increased Klotho in the islet of Langerhans of normal mice, as well as the islets and kidneys of STZ-treated mice. In vitro, GABA stimulated production and secretion of Klotho by human islet cells. Knockdown (KD) of Klotho with siRNA in INS-1E insulinoma cells abrogated the protective effects of GABA against STZ toxicity. Following KD, soluble Klotho reversed the effects of Klotho deficiency. In human islet cells soluble Klotho protected against cell death, and stimulated proliferation and insulin secretion. NF-κB activation triggers beta-cell apoptosis, and both GABA and Klotho suppress this pathway. We found Klotho KD augmented NF-κB p65 expression, and abrogated the ability of GABA to block NF-κB activation. This is the first report that GABAergic stimulation increases Klotho expression. Klotho protected and stimulated beta cells and lack of Klotho (KD) was reversed by soluble Klotho. These findings have important implications for the treatment of T1D.


Assuntos
Glucuronidase/biossíntese , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Ácido gama-Aminobutírico/farmacologia , Envelhecimento/efeitos dos fármacos , Envelhecimento/metabolismo , Animais , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Técnicas de Silenciamento de Genes , Glucuronidase/antagonistas & inibidores , Glucuronidase/genética , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Proteínas Klotho , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores
4.
Blood ; 126(2): 247-56, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-26036802

RESUMO

Immune thrombocytopenia (ITP) is a common autoimmune bleeding disorder characterized by autoantibodies targeting platelet surface proteins, most commonly GPIIbIIIa (αIIbß3 integrin), leading to platelet destruction. Recently, CD8(+) cytotoxic T-lymphocytes (CTLs) targeting platelets and megakaryocytes have also been implicated in thrombocytopenia. Because steroids are the most commonly administered therapy for ITP worldwide, we established both active (immunized splenocyte engraftment) and passive (antibody injection) murine models of steroid treatment. Surprisingly, we found that, in both models, CD8(+) T cells limited the severity of the thrombocytopenia and were required for an efficacious response to steroid therapy. Conversely, CD8(+) T-cell depletion led to more severe thrombocytopenia, whereas CD8(+) T-cell transfusion ameliorated thrombocytopenia. CD8(+) T-regulatory cell (Treg) subsets were detected, and interestingly, dexamethasone (DEX) treatment selectively expanded CD8(+) Tregs while decreasing CTLs. In vitro coculture studies revealed CD8(+) Tregs suppressed CD4(+) and CD19(+) proliferation, platelet-associated immunoglobulin G generation, CTL cytotoxicity, platelet apoptosis, and clearance. Furthermore, we found increased production of anti-inflammatory interleukin-10 in coculture studies and in vivo after steroid treatment. Thus, we uncovered subsets of CD8(+) Tregs and demonstrated their potent immunosuppressive and protective roles in experimentally induced thrombocytopenia. The data further elucidate mechanisms of steroid treatment and suggest therapeutic potential for CD8(+) Tregs in immune thrombocytopenia.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Dexametasona/uso terapêutico , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Púrpura Trombocitopênica Idiopática/imunologia , Animais , Plaquetas/imunologia , Linfócitos T CD8-Positivos/transplante , Terapia Combinada , Modelos Animais de Doenças , Imunoterapia Adotiva , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/imunologia , Púrpura Trombocitopênica Idiopática/terapia , Linfócitos T Citotóxicos , Resultado do Tratamento
5.
Biochem Biophys Res Commun ; 452(3): 649-54, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25193706

RESUMO

We have previously shown that GABA protects pancreatic islet cells against apoptosis and exerts anti-inflammatory effects. Notably, GABA inhibited the activation of NF-κB in both islet cells and lymphocytes. NF-κB activation is detrimental to beta cells by promoting apoptosis. However, the mechanisms by which GABA mediates these effects are unknown. Because the above-mentioned effects mimic the activity of sirtuin 1 (SIRT1) in beta cells, we investigated whether it is involved. SIRT1 is an NAD(+)-dependent deacetylase that enhances insulin secretion, and counteracts inflammatory signals in beta cells. We found that the incubation of a clonal beta-cell line (rat INS-1) with GABA increased the expression of SIRT1, as did GABA receptor agonists acting on either type A or B receptors. NAD(+) (an essential cofactor of SIRT1) was also increased. GABA augmented SIRT1 enzymatic activity, which resulted in deacetylation of the p65 component of NF-κB, and this is known to interfere with the activation this pathway. GABA increased insulin production and reduced drug-induced apoptosis, and these actions were reversed by SIRT1 inhibitors. We examined whether SIRT1 is similarly induced in newly isolated human islet cells. Indeed, GABA increased both NAD(+) and SIRT1 (but not sirtuins 2, 3 and 6). It protected human islet cells against spontaneous apoptosis in culture, and this was negated by a SIRT1 inhibitor. Thus, our findings suggest that major beneficial effects of GABA on beta cells are due to increased SIRT1 and NAD(+), and point to a new pathway for diabetes therapy.


Assuntos
Apoptose/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Sirtuína 1/genética , Fator de Transcrição RelA/metabolismo , Ácido gama-Aminobutírico/farmacologia , Animais , Baclofeno/análogos & derivados , Baclofeno/farmacologia , Linhagem Celular Tumoral , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Regulação da Expressão Gênica , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Muscimol/farmacologia , NAD/metabolismo , Picrotoxina/farmacologia , Cultura Primária de Células , Ratos , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Transdução de Sinais , Sirtuína 1/metabolismo , Fator de Transcrição RelA/genética , Ácido gama-Aminobutírico/metabolismo
6.
Proc Natl Acad Sci U S A ; 108(28): 11692-7, 2011 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-21709230

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease characterized by insulitis and islet ß-cell loss. Thus, an effective therapy may require ß-cell restoration and immune suppression. Currently, there is no treatment that can achieve both goals efficiently. We report here that GABA exerts antidiabetic effects by acting on both the islet ß-cells and immune system. Unlike in adult brain or islet α-cells in which GABA exerts hyperpolarizing effects, in islet ß-cells, GABA produces membrane depolarization and Ca(2+) influx, leading to the activation of PI3-K/Akt-dependent growth and survival pathways. This provides a potential mechanism underlying our in vivo findings that GABA therapy preserves ß-cell mass and prevents the development of T1D. Remarkably, in severely diabetic mice, GABA restores ß-cell mass and reverses the disease. Furthermore, GABA suppresses insulitis and systemic inflammatory cytokine production. The ß-cell regenerative and immunoinhibitory effects of GABA provide insights into the role of GABA in regulating islet cell function and glucose homeostasis, which may find clinical application.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Células Secretoras de Insulina/efeitos dos fármacos , Ácido gama-Aminobutírico/farmacologia , Animais , Apoptose/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/biossíntese , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Feminino , Hiperglicemia/prevenção & controle , Imunossupressores/farmacologia , Mediadores da Inflamação/metabolismo , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/fisiologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regeneração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Ácido gama-Aminobutírico/fisiologia
7.
Cells ; 13(17)2024 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-39272986

RESUMO

The α-Klotho protein (hereafter Klotho) is an obligate coreceptor for fibroblast growth factor 23 (FGF23). It is produced in the kidneys, brain and other sites. Klotho insufficiency causes hyperphosphatemia and other anomalies. Importantly, it is associated with chronic pathologies (often age-related) that have an inflammatory component. This includes atherosclerosis, diabetes and Alzheimer's disease. Its mode of action in these diseases is not well understood, but it inhibits or regulates multiple major pathways. Klotho has a membrane form and a soluble form (s-Klotho). Cytosolic Klotho is postulated but not well characterized. s-Klotho has endocrine properties that are incompletely elucidated. It binds to the FGF receptor 1c (FGFR1c) that is widely expressed (including endothelial cells). It also attaches to soluble FGF23, and FGF23/Klotho binds to FGFRs. Thus, s-Klotho might be a roaming FGF23 coreceptor, but it has other functions. Notably, Klotho (cell-bound or soluble) counteracts inflammation and appears to mitigate related aging (inflammaging). It inhibits NF-κB and the NLRP3 inflammasome. This inflammasome requires priming by NF-κB and produces active IL-1ß, membrane pores and cell death (pyroptosis). In accord, Klotho countered inflammation and cell injury induced by toxins, damage-associated molecular patterns (DAMPs), cytokines, and reactive oxygen species (ROS). s-Klotho also blocks the TGF-ß receptor and Wnt ligands, which lessens fibrotic disease. Low Klotho is associated with loss of muscle mass (sarcopenia), as occurs in aging and chronic diseases. s-Klotho counters the inhibitory effects of myostatin and TGF-ß on muscle, reduces inflammation, and improves muscle repair following injury. The inhibition of TGF-ß and other factors may also be protective in diabetic retinopathy and age-related macular degeneration (AMD). This review examines Klotho functions especially as related to inflammation and potential applications.


Assuntos
Envelhecimento , Fator de Crescimento de Fibroblastos 23 , Glucuronidase , Proteínas Klotho , Humanos , Envelhecimento/metabolismo , Fator de Crescimento de Fibroblastos 23/metabolismo , Animais , Glucuronidase/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/uso terapêutico
8.
Front Endocrinol (Lausanne) ; 14: 1283739, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38149094

RESUMO

Background: Osteoporosis (OP) is a systemic skeletal disease characterized by compromised bone strength leading to an increased risk of fracture. There is an ongoing debate on whether non-alcoholic fatty liver disease (NAFLD) is an active contributor or an innocent bystander in the pathogenesis of OP. The aim of this study was to assess the causal association between NAFLD and OP. Methods: We performed two-sample Mendelian randomization (MR) analyses to investigate the causal association between genetically predicted NAFLD [i.e., imaging-based liver fat content (LFC), chronically elevated serum alanine aminotransferase (cALT) and biopsy-confirmed NAFLD] and risk of OP. The inverse variant weighted method was performed as main analysis to obtain the causal estimates. Results: Imaging-based LFC and biopsy-confirmed NAFLD demonstrated a suggestive causal association with OP ([odds ratio (OR): 1.003, 95% CI: 1.001-1.004, P < 0.001; OR: 1.001, 95% CI: 1.000-1.002, P = 0.031]). The association between cALT and OP showed a similar direction, but was not statistically significant (OR: 1.001, 95% CI: 1.000-1.002, P = 0.079). Repeated analyses after exclusion of genes associated with confounding factors showed consistent results. Sensitivity analysis indicated low heterogeneity, high reliability and low pleiotropy of the causal estimates. Conclusion: The two-sample MR analyses suggest a causal association between genetically predicted NAFLD and OP.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Osteoporose , Humanos , Hepatopatia Gordurosa não Alcoólica/epidemiologia , Hepatopatia Gordurosa não Alcoólica/genética , Análise da Randomização Mendeliana , Reprodutibilidade dos Testes , Osteoporose/etiologia , Osteoporose/genética
9.
Mol Cell Endocrinol ; 559: 111755, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36049597

RESUMO

We recently demonstrated that combined therapy of GABA and sitagliptin promoted beta-cell proliferation, and decreased beta-cell apoptosis in a multiple low-dose streptozotocin (STZ)-induced beta-cell injury mouse model. In this study, we examined whether this combined therapy is effective in ameliorating the impairment of beta-cell function caused by high-fat diet (HFD) feeding in mice. Male C57BL/6J mice were fed normal chow diet, HFD, or HFD combined with GABA, sitagliptin, or both drugs. Oral drug daily administration was initiated one week before HFD and maintained for two weeks. After two weeks of intervention, we found that GABA or sitagliptin administration ameliorated the impairment of glucose tolerance induced by HFD. This was associated with improved insulin secretion in vivo. Notably, combined administration of GABA and sitagliptin significantly enhanced these effects as compared to each of the monotherapies. Combined GABA and sitagliptin was superior at increasing beta-cell mass, and associated Ki67+ and PDX-1+ beta-cell counts. In addition, we found that HFD-induced compensatory beta-cell proliferation was associated with increased activation of unfolded protein response (UPR), as indicated by BiP expression. This could be an important mechanism of compensatory beta-cell proliferation, and beta cells treated with GABA and sitagliptin showed greater UPR activation. Our results suggest that the combined use of these agents produces superior therapeutic outcomes.


Assuntos
Traumatismo Múltiplo , Fosfato de Sitagliptina , Masculino , Camundongos , Animais , Camundongos Endogâmicos C57BL , Fosfato de Sitagliptina/farmacologia , Fosfato de Sitagliptina/uso terapêutico , Dieta Hiperlipídica/efeitos adversos , Estreptozocina , Ácido gama-Aminobutírico/farmacologia
10.
Biochem Biophys Res Commun ; 425(4): 775-80, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22885184

RESUMO

Cancer stem cells (CSCs) initiate tumors and have a high resistance to conventional cancer therapy. Tranilast is an orally active drug of low toxicity that exerts inhibitory effects on breast CSCs. This appears to depend on its aryl hydrocarbon receptor (AHR) agonistic activity, but this receptor has diverse functions and it is unclear how CSCs are inhibited. CSCs generate tumor spheres in low-adherence cultures, and we employed the mammosphere-forming assay as a functional test for breast CSCs. Because NF-κB has a key role in mammosphere formation and CSC-mediated tumor initiation, we examined that pathway. We also examined the role of neuropilin-1 (Nrp1), which is a growth factor coreceptor linked to the tumorigenicity of some CSCs. We found that tranilast concurrently suppressed mammosphere formation, Nrp1 expression and constitutive NF-κB activation. Flow cytometric analysis revealed that a subpopulation of breast cancer cells bearing breast CSC markers also expressed Nrp1. A blocking anti-Nrp1 antibody suppressed mammosphere formation. We examined whether there was a link between Nrp1 and NF-κB activation. The siRNA knockdown of Nrp1 severely suppressed NF-κB activation and mammosphere formation. The phosphorylation of Akt and ERK1/2 was also reduced, but to a lesser extent. We conclude that Nrp1 plays a key role in mammosphere formation and this activity is linked to NF-κB activation. Thus, Nrp1 might be a target for therapy against breast CSCs, and the anticancer drug tranilast suppresses its expression.


Assuntos
Neoplasias da Mama/metabolismo , NF-kappa B/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neuropilina-1/biossíntese , Esferoides Celulares/metabolismo , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Proteína Quinase 3 Ativada por Mitógeno , NF-kappa B/agonistas , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Neuropilina-1/genética , Proteínas Proto-Oncogênicas c-akt , RNA Interferente Pequeno/genética , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia , ortoaminobenzoatos/farmacologia
11.
Front Aging ; 3: 931331, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35903083

RESUMO

The α-Klotho protein (henceforth denoted Klotho) has antiaging properties, as first observed in mice homozygous for a hypomorphic Klotho gene (kl/kl). These mice have a shortened lifespan, stunted growth, renal disease, hyperphosphatemia, hypercalcemia, vascular calcification, cardiac hypertrophy, hypertension, pulmonary disease, cognitive impairment, multi-organ atrophy and fibrosis. Overexpression of Klotho has opposite effects, extending lifespan. In humans, Klotho levels decline with age, chronic kidney disease, diabetes, Alzheimer's disease and other conditions. Low Klotho levels correlate with an increase in the death rate from all causes. Klotho acts either as an obligate coreceptor for fibroblast growth factor 23 (FGF23), or as a soluble pleiotropic endocrine hormone (s-Klotho). It is mainly produced in the kidneys, but also in the brain, pancreas and other tissues. On renal tubular-cell membranes, it associates with FGF receptors to bind FGF23. Produced in bones, FGF23 regulates renal excretion of phosphate (phosphaturic effect) and vitamin D metabolism. Lack of Klotho or FGF23 results in hyperphosphatemia and hypervitaminosis D. With age, human renal function often deteriorates, lowering Klotho levels. This appears to promote age-related pathology. Remarkably, Klotho inhibits four pathways that have been linked to aging in various ways: Transforming growth factor ß (TGF-ß), insulin-like growth factor 1 (IGF-1), Wnt and NF-κB. These can induce cellular senescence, apoptosis, inflammation, immune dysfunction, fibrosis and neoplasia. Furthermore, Klotho increases cell-protective antioxidant enzymes through Nrf2 and FoxO. In accord, preclinical Klotho therapy ameliorated renal, cardiovascular, diabetes-related and neurodegenerative diseases, as well as cancer. s-Klotho protein injection was effective, but requires further investigation. Several drugs enhance circulating Klotho levels, and some cross the blood-brain barrier to potentially act in the brain. In clinical trials, increased Klotho was noted with renin-angiotensin system inhibitors (losartan, valsartan), a statin (fluvastatin), mTOR inhibitors (rapamycin, everolimus), vitamin D and pentoxifylline. In preclinical work, antidiabetic drugs (metformin, GLP-1-based, GABA, PPAR-γ agonists) also enhanced Klotho. Several traditional medicines and/or nutraceuticals increased Klotho in rodents, including astaxanthin, curcumin, ginseng, ligustilide and resveratrol. Notably, exercise and sport activity increased Klotho. This review addresses molecular, physiological and therapeutic aspects of Klotho.

12.
Eur J Pharm Sci ; 175: 106218, 2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35618199

RESUMO

We demonstrated recently that supaglutide, a novel GLP-1 mimetic generated by recombinant fusion protein techniques, exerted hypoglycemic effects in type 2 diabetes db/db mice and spontaneous diabetic monkeys. In this study, we investigated the pharmacokinetics and pharmacodynamics of supaglutide by single subcutaneous and intravenous injection(s) in rats and rhesus monkeys, as well as fourconsecutive subcutaneous injections in monkeys.We found the half-life (t1/2) of supaglutide was 39.7 h and 35.8 h at dosing 0.1 mg/kg upon subcutaneous or intravenous administration respectively, in rhesus monkeys. The plasma supaglutide peaked at 8-10 h, while the plasma drug exposure levels increased with the increase of dose, showing approximately a linear pharmacokinetic characteristic. The elimination kinetics (Ke) were found to be similar between subcutaneous (∼0.025 in rats and ∼0.018 in monkeys) and intravenous administration (0.021 in rats and 0.020 in monkeys), whereas the bioavailability was found to be 31.1% in rats and 63.9% in monkeys. In monkeys, a single dose injection of supaglutide markedly decreased the random blood glucose levels that reaching the maxima effects in 14-16 h, gradually recovered and returned to the baseline level approximately after 72 h. 125I-supaglutide was found mainly distributed in the serum and organs rich in blood supply. Urine was found to be the primary excretion route of supaglutide, following by feces, but mostly not in bile.Our results show that supaglutide possess linear pharmacokinetic characteristics associated with prolonged hypoglycemic effects inanimals,suggestinga potential weekly dosing therapeutic reagent for the treatment of type 2 diabetes and metabolic diseases.


Assuntos
Diabetes Mellitus Tipo 2 , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Injeções Intravenosas , Macaca fascicularis , Macaca mulatta , Camundongos , Ratos , Ratos Sprague-Dawley
13.
Carcinogenesis ; 32(4): 613-21, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21186301

RESUMO

Neuropilin (Nrp)-1 and Nrp-2 are multifunctional proteins frequently expressed by cancer cells and contribute to tumor progression by mechanisms that are not well understood. They are co-receptors for vascular endothelial growth factor and class 3 semaphorins, but recently we found that Nrp1 also binds latent and active transforming growth factor (TGF)-ß1, and activates the latent form latency-associated peptide (LAP)-TGF-ß1. Here, we report that Nrp1 has affinity for TGF-ß receptors TßRI and TßRII, the signaling TGF-ß receptors, as well as TßRIII (betaglycan), as determined in binding assays, pull down assays and confocal microscopy. Nrp1 had a higher affinity for TßRI than TßRII and could form a complex with these receptors. In breast cancer cells, Nrp1 and TßRI cointernalized in the presence of TGF-ß1. Nrp1 acted as a TGF-ß co-receptor by augmenting canonical Smad2/3 signaling. Importantly, Nrp-positive cancer cells, unlike negative cells, were able to activate latent TGF-ß1 and respond. We examined two other membrane proteins that bind LAP-TGF-ß, i.e. an RGD-binding integrin (αvß3) and Glycoprotein A repetitions predominant (CLRRC32). RGD-binding integrins are frequently expressed by cancer cells, and glycoprotein A repetitions predominant is expressed by activated regulatory T cells that appear linked to poor tumor immunity. In vitro, these receptors did not activate LAP-TGF-ß1, but subsequent addition of Nrp1 activated the cytokine. Thus, Nrp1 might collaborate with other latent TGF-ß receptors in TGF-ß capture and activation. We also show that Nrp2 has activities similar to Nrp1. We conclude that Nrp1 is a co-receptor for TGF-ß1 and augments responses to latent and active TGF-ß. Since TGF-ß promotes metastasis this is highly relevant to cancer biology.


Assuntos
Neoplasias/metabolismo , Neuropilina-1/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta1/fisiologia , Linhagem Celular Tumoral , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Proteoglicanas/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Transdução de Sinais , Proteínas Smad/fisiologia
14.
J Gene Med ; 13(9): 470-7, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21786368

RESUMO

BACKGROUND: Therapeutic xenoproteins are immunogenic and can induce neutralizing antibodies. When delivered by intramuscular injection of a plasmid vector, this mimics classical DNA vaccination. To demonstrate this, we chose Exendin-4 (Ex4), which is a glucagon-like peptide-1 mimetic xenoprotein in clinical use for treating type 2 diabetes. We constructed an Ex4 and mouse immunoglobulin (Ig)G1-Fc fusion fragment (Ex4/Fc), and hypothesized that it would have minimal immunogenicity as a result of its capacity to bind the inhibitory Fc receptor FcγRIIb expressed by B lymphocytes. METHODS: Plasmid vectors encoding Ex4/Fc constructs, with wild-type or mutant Fc, were injected intramuscularly into mice, and local electroporation was applied to enhance gene transfer. Gene transfer was performed in both wild-type and FcγRIIb knockout mice. Antibody production was detected in serum by an enzyme-linked immunosorbent assay. RESULTS: Recombinant Ex4/Fc bound only to B cells expressing FcγRIIb. This binding was dependent on a motif in the Fc region, which we mutated to abolish binding (Ex4/Fcmut). Ex4 antibody was detected in mice treated with Ex4, as well as Ex4/Fcmut, but not in those treated with Ex4/Fc. Thus, wild-type Fc was associated with reduced immunogenicity. To confirm this was related to the presence of inhibitory Fc receptors, we also performed experiments in FcγRIIb-null mice. Mice lacking this receptor produced antibodies against all Ex4 constructs, including the wild-type Fc (Ex4/Fc). CONCLUSIONS: The present study shows that inhibitory FcγRIIb receptors interacting with the wild-type IgG1-Fc reduce immunity against Ex4/Fc, suggesting an approach for reducing the immunogenicity of therapeutic proteins in the context of gene therapy.


Assuntos
Terapia Genética , Hipoglicemiantes/imunologia , Fragmentos Fc das Imunoglobulinas/genética , Peptídeos/genética , Peptídeos/imunologia , Receptores de IgG/metabolismo , Peçonhas/genética , Peçonhas/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Diabetes Mellitus Tipo 2/terapia , Exenatida , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Fragmentos Fc das Imunoglobulinas/metabolismo , Injeções Intramusculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmídeos/administração & dosagem , Plasmídeos/genética , Ligação Proteica , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Transdução Genética
15.
Blood ; 113(3): 622-5, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19023118

RESUMO

Acute lymphopenia-induced homeostatic proliferation (HP) of T cells promotes antitumor immunity, but the mechanism is unclear. We hypothesized that this is due to a lack of inhibitory signals that allows activation of T cells with low affinity for self-antigens. Tumors resist immunity in part by expressing inhibitory molecules such as PD-1 ligand 1 (PD-L1), B7-H4, and TGF-beta. In irradiated mice undergoing HP, we found that T cells displayed a severe deficit in the activation-induced expression of inhibitory molecules PD-1 and CTLA-4, and TGF-beta1-induced expression of Foxp3. HP T cells were also less suppressed by B7-H4/Ig and, unlike control T cells, failed to produce IL-10 in response to this molecule. This deficiency in regulation was reversed as normal T-cell numbers were restored. We conclude that T cells are weakly regulated by inhibitory molecules during the acute phase of HP, which could explain their increased effectiveness in cancer immunotherapy.


Assuntos
Homeostase/imunologia , Linfopenia/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD/biossíntese , Antígenos CD/imunologia , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/imunologia , Autoantígenos/imunologia , Antígeno B7-1/biossíntese , Antígeno B7-1/imunologia , Antígeno CTLA-4 , Proliferação de Células , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/imunologia , Camundongos , Receptor de Morte Celular Programada 1 , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/imunologia , Inibidor 1 da Ativação de Células T com Domínio V-Set
16.
Exp Mol Pathol ; 90(1): 116-22, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21040720

RESUMO

In the treatment of breast cancer, although a wide of choice of drugs and treatment modalities are available, drug resistance or drug toxicity poses a considerable challenge. Tranilast is a well tolerated drug used in the treatment of allergic disorders. Previous works in various models have shown that tranilast has the potential to be used as an anti-cancer drug. Hence, in this study using human breast cancer cell lines BT-474 and MDA-MB-231, we studied the effect of tranilast on cell growth, migration and ability to prevent colony formation in vitro, properties that are relevant to a possible therapeutic effect in breast cancer. We found that tranilast inhibits the growth of both breast cancer cell lines. In the cell migration experiments, the tumor cells exhibit significantly slower wound closure after tranilast treatment, as well as reduced migration using an insert system. Downregulation of MRTF-A, a global cytoskeleton regulator was observed after tranilast treatment. Additionally, tranilast treatment increased levels of cleaved PARP in both cell lines tested indicating a stimulation of apoptosis. A significant reduction in colony size and number was observed in soft agar clonogenic assays in both cell lines after tranilast treatment. BT-474 cells were more responsive to tranilast treatment compared to MDA-MB-231 cells, suggesting a difference in modes of action, or sensitivity, possibly related to their different receptor status. Based on these changes in cancer cell lines, we conclude that tranilast exerts effects that set a rationale for future preclinical studies in animal models of breast cancer.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , ortoaminobenzoatos/uso terapêutico , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ensaio de Unidades Formadoras de Colônias , Relação Dose-Resposta a Droga , Feminino , Humanos , Poli(ADP-Ribose) Polimerases/metabolismo , ortoaminobenzoatos/farmacologia
17.
J Endocrinol ; 248(2): 133-143, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33258801

RESUMO

γ-Aminobutyric acid (GABA) and glucagon-like peptide-1 receptor agonist (GLP-1RA) improve rodent ß-cell survival and function. In human ß-cells, GABA exerts stimulatory effects on proliferation and anti-apoptotic effects, whereas GLP-1RA drugs have only limited effects on proliferation. We previously demonstrated that GABA and sitagliptin (Sita), a dipeptidyl peptidase-4 inhibitor which increases endogenous GLP-1 levels, mediated a synergistic ß-cell protective effect in mice islets. However, it remains unclear whether this combination has similar effects on human ß-cell. To address this question, we transplanted a suboptimal mass of human islets into immunodeficient NOD-scid-gamma mice with streptozotocin-induced diabetes, and then treated them with GABA, Sita, or both. The oral administration of either GABA or Sita ameliorated blood glucose levels, increased transplanted human ß-cell counts and plasma human insulin levels. Importantly, the combined administration of the drugs generated significantly superior results in all these responses, as compared to the monotherapy with either one of them. The proliferation and/or regeneration, improved by the combination, were demonstrated by increased Ki67+, PDX-1+, or Nkx6.1+ ß-cell numbers. Protection against apoptosis was also significantly improved by the drug combination. The expression level of α-Klotho, a protein with protective and stimulatory effects on ß cells, was also augmented. Our study indicates that combined use of GABA and Sita produced greater therapeutic benefits, which are likely due to an enhancement of ß-cell proliferation and a decrease in apoptosis.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , GABAérgicos/uso terapêutico , Fosfato de Sitagliptina/uso terapêutico , Ácido gama-Aminobutírico/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidores da Dipeptidil Peptidase IV/farmacologia , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , GABAérgicos/farmacologia , Humanos , Ilhotas Pancreáticas/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos Endogâmicos NOD , Pessoa de Meia-Idade , Fosfato de Sitagliptina/farmacologia , Ácido gama-Aminobutírico/farmacologia
18.
Biochem Biophys Res Commun ; 403(2): 172-7, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21070745

RESUMO

The incretin hormone glucagon-like peptide-1 (GLP-1) exerts important functions in controlling glucose and energy homeostasis. Endogenous GLP-1 has a very short half-life due to DPP-IV-mediated degradation and renal clearance, which limits the therapeutic use of native GLP-1. We have shown previously that immunoglobulin fragment-fused GLP-1 (GLP-1/Fc) is a structurally stable GLP-1 analog. Here, we report a non-viral GLP-1/Fc gene therapy strategy utilizing a REP78-in-trans and REB-in-cis element system to achieve a site-specific genomic integration. For this purpose, the GLP-1/Fc expression cassette, which is fused with the RBE element, was co-injected with the Rep78 plasmid into the muscles of transgenic mice carrying the AAVS1 locus of human chromosome 19. The Rep protein-mediated site-specific integration was demonstrated by nested PCR, dot-blot, and Southern blotting. We found that this approach reduced weight gain and improved lipid profiles in the AAVS1-mice on high-fat diet challenge. Our observations reveal a new GLP-1 therapeutic strategy with an apparent absence of side effects, which may find applications in diabetes treatment and obesity prevention.


Assuntos
Terapia Genética/métodos , Peptídeo 1 Semelhante ao Glucagon/genética , Músculo Esquelético/metabolismo , Aumento de Peso/genética , Animais , Cromossomos Humanos Par 19/genética , Proteínas de Ligação a DNA , Metabolismo Energético/genética , Engenharia Genética , Loci Gênicos , Genoma , Homeostase/genética , Humanos , Camundongos , Camundongos Transgênicos , Plasmídeos/genética , Recombinação Genética , Proteínas Virais
19.
Anticancer Drugs ; 21(4): 351-61, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20145538

RESUMO

The malignant transformation of breast epithelium involves a number of cellular pathways, including those dependent on signaling from TGF beta. Tranilast [N-(3, 4-dimethoxycinnamonyl)-anthranilic acid] is a drug that is used in Japan to control allergic disorders in patients, and its mechanism of action involves TGF beta. In view of the multiple roles of TGF beta in tumor progression, we hypothesized in this study that tranilast impacts cell proliferation, apoptosis, and migration. Using the mouse breast cancer cell line 4T1, our studies showed that tranilast increases AKT1 phosphorylation and decreases ERK1/2 phosphorylation. Alterations in the cell cycle mediators' cyclin D1, p27, cyclin A, pRB, cyclin B, and Cdc2 were observed after exposure to tranilast, favoring cell arrest beyond the G1/S phase. Tranilast reduced tumor cell proliferation even when it was amplified by exogenous TGF beta. TGF beta-neutralizing antibody did not cause a significant decrease in cell proliferation. Tranilast treatment upregulates p53, induces PARP cleavage in vitro, consistent with a promotion of tumor cell apoptosis. TGF beta-neutralizing antibody downregulates endoglin and matrix metalloproteinases (MMP)-9 levels in vitro indicating that the tranilast effect is mediated through TGF beta modulation. Tranilast treatment results in the inhibition of cell migration and invasion. Western blot analysis of tumor lysates from tranilast-treated mice shows decreased levels of TGF beta1, endoglin, and significantly higher levels of p53 and cleaved PARP. Cleaved caspase 3 expression is significantly elevated in tranilast-treated mouse breast tumors. To conclude, tranilast induces cellular and molecular changes in murine breast cancer that can be exploited in preclinical therapeutic trials.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Mamárias Experimentais/tratamento farmacológico , ortoaminobenzoatos/uso terapêutico , Animais , Antineoplásicos/farmacologia , Biomarcadores/metabolismo , Caspase 3/metabolismo , Ciclo Celular/efeitos dos fármacos , Endoglina , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica/prevenção & controle , Fosforilação/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , ortoaminobenzoatos/farmacologia
20.
J Endocrinol ; 246(3): 207-222, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32544878

RESUMO

Gamma-aminobutyric acid (GABA) administration attenuates streptozotocin (STZ)-induced diabetes in rodent models with unclear underlying mechanisms. We found that GABA and Sitagliptin possess additive effect on pancreatic ß-cells, which prompted us to ask the existence of common or unique targets of GLP-1 and GABA in pancreatic ß-cells. Effect of GABA on expression of thioredoxin-interacting protein (TxNIP) was assessed in the INS-1 832/13 (INS-1) cell line, WT and GLP-1R-/- mouse islets. GABA was also orally administrated in STZ-challenged WT or GLP-1R-/- mice, followed by immunohistochemistry assessment of pancreatic islets. Effect of GABA on Wnt pathway effector ß-catenin (ß-cat) was examined in INS-1 cells, WT and GLP-1R-/- islets. We found that GABA shares a common feature with GLP-1 on inhibiting TxNIP, while this function was attenuated in GLP-1R-/- islets. In WT mice with STZ challenge, GABA alleviated several 'diabetic syndromes', associated with increased ß-cell mass. These features were virtually absent in GLP-1R-/- mice. Knockdown TxNIP in INS-1 cells increased GLP-1R, Pdx1, Nkx6.1 and Mafa levels, associated with increased responses to GABA or GLP-1 on stimulating insulin secretion. Cleaved caspase-3 level can be induced by high-glucose, dexamethasone, or STZ in INS-1 cell, while GABA treatment blocked the induction. Finally, GABA treatment increased cellular cAMP level and ß-cat S675 phosphorylation in WT but not GLP-1R-/- islets. We, hence, identified TxNIP as a common target of GABA and GLP-1 and suggest that, upon STZ or other stress challenge, the GLP-1R-cAMP-ß-cat signaling cascade also mediates beneficial effects of GABA in pancreatic ß-cell, involving TxNIP reduction.


Assuntos
Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Células Secretoras de Insulina/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Caspase 3/metabolismo , Linhagem Celular , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Fatores de Transcrição Maf Maior/genética , Fatores de Transcrição Maf Maior/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fosforilação , Transativadores/genética , Transativadores/metabolismo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa