Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Physiol Biochem ; 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38587595

RESUMO

Fasting and fasting-mimicking conditions modulate tumor metabolism and remodel the tumor microenvironment (TME), which could be exploited for the treatment of tumors. A body of evidence demonstrates that fasting and fasting-mimicking conditions can kill cancer cells, or sensitize them to the antitumor activity of standard-of-care drugs while protecting normal cells against their toxic side effects. Pre- and clinical data also suggest that immune responses are involved in these therapeutic effects. Therefore, there is increasing interest in evaluating the impact of fasting-like conditions in the efficacy of antitumor therapies based on the restoration or activation of antitumor immune responses. Here, we review the recent progress in the intersection of fasting-like conditions and current cancer treatments, with an emphasis on cancer immunotherapy.

2.
Clin Cancer Res ; 30(1): 176-186, 2024 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-37527007

RESUMO

PURPOSE: The aim of our study was to elucidate the impact of bevacizumab added to neoadjuvant chemotherapy (NACT) on the tumor immune microenvironment and correlate the changes with the clinical outcome of the patients. EXPERIMENTAL DESIGN: IHC and multiplex immunofluorescence for lymphoid and myeloid lineage markers were performed in matched tumor samples from 23 patients with ovarian cancer enrolled in GEICO 1205/NOVA clinical study before NACT and at the time of interval cytoreductive surgery. RESULTS: Our results showed that the addition of bevacizumab to NACT plays a role mainly on lymphoid populations at the stromal compartment, detecting a significant decrease of CD4+ T cells, an increase of CD8+ T cells, and an upregulation in effector/regulatory cell ratio (CD8+/CD4+FOXP3+). None of the changes observed were detected in the intra-epithelial site in any arm (NACT or NACT-bevacizumab). No differences were found in myeloid lineage (macrophage-like). The percentage of Treg populations and effector/regulatory cell ratio in the stroma were the only two variables significantly associated with progression-free survival (PFS). CONCLUSIONS: The addition of bevacizumab to NACT did not have an impact on PFS in the GEICO 1205 study. However, at the cellular level, changes in CD4+, CD8+ lymphocyte populations, and CD8+/CD4+FOXP3 ratio have been detected only at the stromal site. On the basis of our results, we hypothesize about the existence of mechanisms of resistance that could prevent the trafficking of T-effector cells into the epithelial component of the tumor as a potential explanation for the lack of efficacy of ICI in the first-line treatment of advanced epithelial ovarian cancer. See related commentary by Soberanis Pina and Oza, p. 12.


Assuntos
Terapia Neoadjuvante , Neoplasias Ovarianas , Humanos , Feminino , Carcinoma Epitelial do Ovário/tratamento farmacológico , Bevacizumab/uso terapêutico , Terapia Neoadjuvante/métodos , Microambiente Tumoral , Neoplasias Ovarianas/patologia , Fatores de Transcrição Forkhead , Quimioterapia Adjuvante
4.
EMBO Rep ; 24(8): e55884, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37366231

RESUMO

Recent studies highlight the importance of baseline functional immunity for immune checkpoint blockade therapies. High-dimensional systemic immune profiling is performed in a cohort of non-small-cell lung cancer patients undergoing PD-L1/PD-1 blockade immunotherapy. Responders show high baseline myeloid phenotypic diversity in peripheral blood. To quantify it, we define a diversity index as a potential biomarker of response. This parameter correlates with elevated activated monocytic cells and decreased granulocytic phenotypes. High-throughput profiling of soluble factors in plasma identifies fractalkine (FKN), a chemokine involved in immune chemotaxis and adhesion, as a biomarker of response to immunotherapy that also correlates with myeloid cell diversity in human patients and murine models. Secreted FKN inhibits lung adenocarcinoma growth in vivo through a prominent contribution of systemic effector NK cells and increased tumor immune infiltration. FKN sensitizes murine lung cancer models refractory to anti-PD-1 treatment to immune checkpoint blockade immunotherapy. Importantly, recombinant FKN and tumor-expressed FKN are efficacious in delaying tumor growth in vivo locally and systemically, indicating a potential therapeutic use of FKN in combination with immunotherapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Humanos , Camundongos , Antígeno B7-H1/genética , Biomarcadores , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/uso terapêutico , Neoplasias Pulmonares/genética
5.
Cardiovasc Diabetol ; 22(1): 84, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-37046242

RESUMO

AIM: Obesity is linked to cardiometabolic diseases, however non-obese individuals are also at risk for type 2 diabetes (T2D) and cardiovascular disease (CVD). White adipose tissue (WAT) is known to play a role in both T2D and CVD, but the contribution of WAT inflammatory status especially in non-obese patients with cardiometabolic diseases is less understood. Therefore, we aimed to find associations between WAT inflammatory status and cardiometabolic diseases in non-obese individuals. METHODS: In a population-based cohort containing non-obese healthy (n = 17), T2D (n = 16), CVD (n = 18), T2D + CVD (n = 19) individuals, seventeen different cytokines were measured in WAT and in circulation. In addition, 13-color flow cytometry profiling was employed to phenotype the immune cells. Human T cell line (Jurkat T cells) was stimulated by rCCL18, and conditioned media (CM) was added to the in vitro cultures of human adipocytes. Lipolysis was measured by glycerol release. Blocking antibodies against IFN-γ and TGF-ß were used in vitro to prove a role for these cytokines in CCL18-T-cell-adipocyte lipolysis regulation axis. RESULTS: In CVD, T2D and CVD + T2D groups, CCL18 and CD4+ T cells were upregulated significantly compared to healthy controls. WAT CCL18 secretion correlated with the amounts of WAT CD4+ T cells, which also highly expressed CCL18 receptors suggesting that WAT CD4+ T cells are responders to this chemokine. While direct addition of rCCL18 to mature adipocytes did not alter the adipocyte lipolysis, CM from CCL18-treated T cells increased glycerol release in in vitro cultures of adipocytes. IFN-γ and TGF-ß secretion was significantly induced in CM obtained from T cells treated with CCL18. Blocking these cytokines in CM, prevented CM-induced upregulation of adipocyte lipolysis. CONCLUSION: We suggest that in T2D and CVD, increased production of CCL18 recruits and activates CD4+ T cells to secrete IFN-γ and TGF-ß. This, in turn, promotes adipocyte lipolysis - a possible risk factor for cardiometabolic diseases.


Assuntos
Doenças Cardiovasculares , Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/metabolismo , Glicerol/metabolismo , Linfócitos T/metabolismo , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Citocinas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/metabolismo , Quimiocinas CC/metabolismo
6.
Front Endocrinol (Lausanne) ; 13: 996954, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36313784

RESUMO

Background: Obesity-associated metabolic complications display sexual dimorphism and can be impacted by cytokines. We previously showed that interleukin-10 (IL-10) was upregulated in white adipose tissue (WAT) of obese women with type 2 diabetes (T2D). Whether this pertains to men is unknown. The aim of this study was to compare the impact of obesity and T2D on WAT IL-10 levels in men versus women. Methods: Plasma and subcutaneous WAT biopsies were obtained from 108 metabolically well-characterized individuals. WAT IL10 expression/secretion and WAT-resident IL-10-secreting macrophage number were measured. Circulating sex hormone levels were correlated to WAT IL10 expression in 22 individuals and sex hormone effects on macrophage IL10 expression were investigated in vitro. Results: Obese women with T2D showed increased IL10 expression/secretion and IL-10-secreting WAT macrophage number compared to other female groups. This difference was absent in men. Non-obese women and men with T2D showed similar IL-10 levels compared to healthy controls, indicating that T2D alone does not regulate IL-10. Although WAT IL10 expression correlated with serum estrone (E1) concentrations, recombinant E1 did not affect macrophage IL10 expression in vitro. Conclusion: WAT IL-10 levels are higher in women with obesity and T2D, but not in men and this effect is primarily attributed to obesity per se. This is less likely to be driven by circulating sex hormones. We propose that the WAT IL-10 might exert protective effects in obesity-associated chronic inflammation in women which could be one of the contributing factors for the decreased morbidity observed in women during obesity than men.


Assuntos
Diabetes Mellitus Tipo 2 , Interleucina-10 , Masculino , Humanos , Feminino , Interleucina-10/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo
7.
Cancer Lett ; 549: 215900, 2022 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-36087681

RESUMO

Tumor progression relies on the interaction between tumor cells and their surrounding tumor microenvironment (TME), which also influences therapeutic responses. The complement system, an essential part of innate immunity, has been traditionally considered an effector arm against tumors. However, established tumors co-opt complement-mediated immune responses in the TME to support chronic inflammation, activate cancer-related signaling pathways and hamper antitumor immune responses. In this context, myeloid-derived suppressor cells (MDSCs), a heterogeneous population of myeloid progenitors with immunosuppressive functions, are recognized as major mediators of tumor-associated complement activities. This review focuses on the impact of complement activation within the TME, with a special emphasis on MDSC functions and the involvement of the C5a/C5aR1 axis. We also discuss the translation of these findings into therapeutic advances based on complement inhibition.


Assuntos
Células Supressoras Mieloides , Neoplasias , Humanos , Imunidade , Neoplasias/patologia , Transdução de Sinais , Microambiente Tumoral
8.
Front Immunol ; 13: 946522, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091057

RESUMO

Numerous publications have underlined the link between complement C5a and the clinical course of COVID-19. We previously reported that levels of C5a remain high in the group of severely ill patients up to 90 days after hospital discharge. We have now evaluated which complement pathway fuels the elevated levels of C5a during hospitalization and follow-up. The alternative pathway (AP) activation marker C3bBbP and the soluble fraction of C4d, a footprint of the classical/lectin (CP/LP) pathway, were assessed by immunoenzymatic assay in a total of 188 serial samples from 49 patients infected with SARS-CoV-2. Unlike C5a, neither C3bBbP nor C4d readouts rose proportionally to the severity of the disease. Detailed correlation analyses in hospitalization and follow-up samples collected from patients of different disease severity showed significant positive correlations of AP and CP/LP markers with C5a in certain groups, except for the follow-up samples of the patients who suffered from highly severe COVID-19 and presented the highest C5a readouts. In conclusion, there is not a clear link between persistently high levels of C5a after hospital discharge and markers of upstream complement activation, suggesting the existence of a non-canonical source of C5a in patients with a severe course of COVID-19.


Assuntos
COVID-19 , Ativação do Complemento , Complemento C3b , Complemento C4b , Complemento C5a , Fator B do Complemento , Fragmentos de Peptídeos , Biomarcadores/sangue , COVID-19/sangue , COVID-19/imunologia , Ativação do Complemento/imunologia , Complemento C3b/imunologia , Complemento C4b/imunologia , Complemento C5a/análise , Complemento C5a/imunologia , Fator B do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Humanos , Fragmentos de Peptídeos/imunologia , SARS-CoV-2
9.
N Engl J Med ; 386(26): 2471-2481, 2022 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-35767439

RESUMO

BACKGROUND: Pediatric patients with diffuse intrinsic pontine glioma (DIPG) have a poor prognosis, with a median survival of less than 1 year. Oncolytic viral therapy has been evaluated in patients with pediatric gliomas elsewhere in the brain, but data regarding oncolytic viral therapy in patients with DIPG are lacking. METHODS: We conducted a single-center, dose-escalation study of DNX-2401, an oncolytic adenovirus that selectively replicates in tumor cells, in patients with newly diagnosed DIPG. The patients received a single virus infusion through a catheter placed in the cerebellar peduncle, followed by radiotherapy. The primary objective was to assess the safety and adverse-event profile of DNX-2401. The secondary objectives were to evaluate the effect of DNX-2401 on overall survival and quality of life, to determine the percentage of patients who have an objective response, and to collect tumor-biopsy and peripheral-blood samples for correlative studies of the molecular features of DIPG and antitumor immune responses. RESULTS: A total of 12 patients, 3 to 18 years of age, with newly diagnosed DIPG received 1×1010 (the first 4 patients) or 5×1010 (the subsequent 8 patients) viral particles of DNX-2401, and 11 received subsequent radiotherapy. Adverse events among the patients included headache, nausea, vomiting, and fatigue. Hemiparesis and tetraparesis developed in 1 patient each. Over a median follow-up of 17.8 months (range, 5.9 to 33.5), a reduction in tumor size, as assessed on magnetic resonance imaging, was reported in 9 patients, a partial response in 3 patients, and stable disease in 8 patients. The median survival was 17.8 months. Two patients were alive at the time of preparation of the current report, 1 of whom was free of tumor progression at 38 months. Examination of a tumor sample obtained during autopsy from 1 patient and peripheral-blood studies revealed alteration of the tumor microenvironment and T-cell repertoire. CONCLUSIONS: Intratumoral infusion of oncolytic virus DNX-2401 followed by radiotherapy in pediatric patients with DIPG resulted in changes in T-cell activity and a reduction in or stabilization of tumor size in some patients but was associated with adverse events. (Funded by the European Research Council under the European Union's Horizon 2020 Research and Innovation Program and others; EudraCT number, 2016-001577-33; ClinicalTrials.gov number, NCT03178032.).


Assuntos
Neoplasias do Tronco Encefálico , Glioma Pontino Intrínseco Difuso , Terapia Viral Oncolítica , Vírus Oncolíticos , Adenoviridae , Adolescente , Astrocitoma/radioterapia , Astrocitoma/terapia , Neoplasias do Tronco Encefálico/mortalidade , Neoplasias do Tronco Encefálico/patologia , Neoplasias do Tronco Encefálico/radioterapia , Neoplasias do Tronco Encefálico/terapia , Criança , Pré-Escolar , Glioma Pontino Intrínseco Difuso/mortalidade , Glioma Pontino Intrínseco Difuso/radioterapia , Glioma Pontino Intrínseco Difuso/terapia , Glioma/radioterapia , Glioma/terapia , Humanos , Infusões Intralesionais , Terapia Viral Oncolítica/efeitos adversos , Terapia Viral Oncolítica/métodos , Qualidade de Vida , Microambiente Tumoral
10.
Acta Diabetol ; 59(5): 687-696, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35098372

RESUMO

AIM: To evaluate the long-term effect of intra-lymphatic administration of GAD-alum and a booster dose 2.5 years after the first intervention (DIAGNODE Extension study) in patients with recent-onset type 1 diabetes. METHODS: DIAGNODE-1: Samples were collected from 12 patients after 30 months who had received 3 injections of 4 µg GAD-alum into a lymph node with one-month interval. DIAGNODE Extension study: First in human, a fourth booster dose of autoantigen (GAD-alum) was given to 3 patients at 31.5 months, who were followed for another 12 months. C-peptide was measured during mixed meal tolerance tests (MMTTs). GADA, IA-2A, GADA subclasses, GAD65-induced cytokines, PBMCs proliferation and T cells markers were analyzed. RESULTS: After 30-month treatment, efficacy was still seen in 8/12 patients (good responders, GR). Partial remission (IDAA1c < 9) had decreased compared to 15 months, but did not differ from baseline, and HbA1c remained stable. GAD65-specific immune responses induced by the treatment started to wane after 30 months, and most changes observed at 15 months were undetectable. GADA subclasses IgG2, IgG3 and IgG4 were predominant in the GR along with IgG1. A fourth intra-lymphatic GAD-alum dose to three patients after 31.5 months gave no adverse events. In all three patients, C-peptide seemed to increase the first 6 months, and thereafter, C-peptide, HbA1c, insulin requirement and IDAA1c remained stable. CONCLUSION: The effect of intra-lymphatic injections of GAD-alum had decreased after 30 months. Good responders showed a specific immune response. Administration of a fourth booster dose after 31.5 months was safe, and there was no decline in C-peptide observed during the 12-month follow-up.


Assuntos
Diabetes Mellitus Tipo 1 , Compostos de Alúmen , Autoanticorpos , Peptídeo C , Diabetes Mellitus Tipo 1/terapia , Seguimentos , Glutamato Descarboxilase , Hemoglobinas Glicadas , Humanos , Imunoglobulina G
11.
Diabetes Metab Res Rev ; 38(3): e3500, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34611978

RESUMO

AIMS: Immunomodulation with autoantigens potentially constitutes a specific and safe treatment for type 1 diabetes (T1D). Studies with GAD-alum administrated subcutaneously have shown to be safe, but its efficacy has been inconclusive. Administration of GAD-alum into the lymph nodes, aimed to optimise antigen presentation, has shown promising results in an open-label clinical trial. Herein, we compared the immune response of the individuals included in the trial with a group who received GAD-alum subcutaneously in a previous study. MATERIALS AND METHODS: Samples from T1D individuals collected 15 months after administration of either three doses 1 month apart of 4 µg GAD-alum into lymph nodes (LN, n = 12) or two doses 1 month apart of 20 µg subcutaneously (SC, n = 12) were studied. GADA, GADA subclasses, GAD65 -induced cytokines, peripheral blood mononuclear cell proliferation, and T cells markers were analysed. RESULTS: Low doses of GAD-alum into the lymph nodes induced higher GADA levels than higher doses administrated subcutaneously. Immune response in the LN group was characterised by changes in GADA subclasses, with a relative reduction of IgG1 and enhanced IgG2, IgG3, and IgG4 proportion, higher GAD65 -induced secretion of IL-5, IL-10, and TNF-α, and reduction of cell proliferation and CD8+ T cells. These changes were not observed after subcutaneous (SC) injections of GAD-alum. CONCLUSIONS: GAD-specific immune responses 15 months after lymph node injections of GAD-alum differed from the ones induced by SC administration of the same autoantigen.


Assuntos
Diabetes Mellitus Tipo 1 , Compostos de Alúmen , Linfócitos T CD8-Positivos , Glutamato Descarboxilase , Humanos , Imunidade , Leucócitos Mononucleares
12.
Cancer Lett ; 529: 70-84, 2022 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-34971753

RESUMO

Myeloid-derived suppressor cells (MDSCs) play a major role in cancer progression. In this study, we investigated the mechanisms by which complement C5a increases the capacity of polymorphonuclear MDSCs (PMN-MDSCs) to promote tumor growth and metastatic spread. Stimulation of PMN-MDSCs with C5a favored the invasion of cancer cells via a process dependent on the formation of neutrophil extracellular traps (NETs). NETosis was dependent on the production of high mobility group box 1 (HMGB1) by cancer cells. Moreover, C5a induced the surface expression of the HMGB1 receptors TLR4 and RAGE in PMN-MDSCs. In a mouse lung metastasis model, inhibition of C5a, C5a receptor-1 (C5aR1) or NETosis reduced the number of circulating-tumor cells (CTCs) and the metastatic burden. In support of the translational relevance of these findings, C5a was able to stimulate migration and NETosis in PMN-MDSCs obtained from lung cancer patients. Furthermore, myeloperoxidase (MPO)-DNA complexes, as markers of NETosis, were elevated in lung cancer patients and significantly correlated with C5a levels. In conclusion, C5a induces the formation of NETs from PMN-MDSCs in the presence of cancer cells, which may facilitate cancer cell dissemination and metastasis.


Assuntos
Complemento C5a/imunologia , Armadilhas Extracelulares/imunologia , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Xenoenxertos , Humanos , Imunofenotipagem , Camundongos , Modelos Biológicos , Metástase Neoplásica , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Receptor da Anafilatoxina C5a/metabolismo
13.
Front Immunol ; 12: 767376, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34868021

RESUMO

Evidence supports a role of complement anaphylatoxin C5a in the pathophysiology of COVID-19. However, information about the evolution and impact of C5a levels after hospital discharge is lacking. We analyzed the association between circulating C5a levels and the clinical evolution of hospitalized patients infected with SARS-CoV-2. Serum C5a levels were determined in 32 hospitalized and 17 non-hospitalized patients from Clinica Universidad de Navarra. One hundred and eighty eight serial samples were collected during the hospitalization stay and up to three months during the follow-up. Median C5a levels were 27.71 ng/ml (25th to 75th percentile: 19.35-34.96) for samples collected during hospitalization, versus 16.76 ng/ml (12.90-25.08) for samples collected during the follow-up (p<0.001). There was a negative correlation between serum C5a levels and the number of days from symptom onset (p<0.001). C5a levels also correlated with a previously validated clinical risk score (p<0.001), and was associated with the severity of the disease (p<0.001). An overall reduction of C5a levels was observed after hospital discharge. However, elevated C5a levels persisted in those patients with high COVID-19 severity (i.e. those with a longest stay in the hospital), even after months from hospital discharge (p=0.020). Moreover, high C5a levels appeared to be associated with the presence of long-term respiratory symptoms (p=0.004). In conclusion, serum C5a levels remain high in severe cases of COVID-19, and are associated with the presence of respiratory symptoms after hospital discharge. These results may suggest a role for C5a in the long-term effects of COVID-19 infection.


Assuntos
COVID-19/sangue , Complemento C5a/metabolismo , Alta do Paciente/estatística & dados numéricos , Idoso , COVID-19/complicações , COVID-19/imunologia , Feminino , Seguimentos , Hospitalização , Humanos , Imunidade Inata , Masculino , Pessoa de Meia-Idade , Transtornos Respiratórios/sangue , Transtornos Respiratórios/etiologia , Transtornos Respiratórios/imunologia , Fatores de Risco , SARS-CoV-2 , Índice de Gravidade de Doença
14.
Cancers (Basel) ; 13(15)2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34359708

RESUMO

Ovarian cancer is one of the most lethal gynecological cancers. Current therapeutic strategies allow temporary control of the disease, but most patients develop resistance to treatment. Moreover, although successful in a range of solid tumors, immunotherapy has yielded only modest results in ovarian cancer. Emerging evidence underscores the relevance of the components of innate and adaptive immunity in ovarian cancer progression and response to treatment. Particularly, over the last decade, the complement system, a pillar of innate immunity, has emerged as a major regulator of the tumor microenvironment in cancer immunity. Tumor-associated complement activation may support chronic inflammation, promote an immunosuppressive microenvironment, induce angiogenesis, and activate cancer-related signaling pathways. Recent insights suggest an important role of complement effectors, such as C1q or anaphylatoxins C3a and C5a, and their receptors C3aR and C5aR1 in ovarian cancer progression. Nevertheless, the implication of these factors in different clinical contexts is still poorly understood. Detailed knowledge of the interplay between ovarian cancer cells and complement is required to develop new immunotherapy combinations and biomarkers. In this context, we discuss the possibility of targeting complement to overcome some of the hurdles encountered in the treatment of ovarian cancer.

15.
Endocrinology ; 162(7)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33963396

RESUMO

CONTEXT: Healthy hyperplasic (many but smaller fat cells) white adipose tissue (WAT) expansion is mediated by recruitment, proliferation and/or differentiation of new fat cells. This process (adipogenesis) is controlled by transcriptional programs that have been mostly identified in rodents. OBJECTIVE: A systemic investigation of adipogenic human transcription factors (TFs) that are relevant for metabolic conditions has not been revealed previously. METHODS: TFs regulated in WAT by obesity, adipose morphology, cancer cachexia, and insulin resistance were selected from microarrays. Their role in differentiation of human adipose tissue-derived stem cells (hASC) was investigated by RNA interference (RNAi) screen. Lipid accumulation, cell number, and lipolysis were measured for all screened factors (148 TFs). RNA (RNAseq), protein (Western blot) expression, insulin, and catecholamine responsiveness were examined in hASC following siRNA treatment of selected target TFs. RESULTS: Analysis of TFs regulated by metabolic conditions in human WAT revealed that many of them belong to adipogenesis-regulating pathways. The RNAi screen identified 39 genes that affected fat cell differentiation in vitro, where 11 genes were novel. Of the latter JARID2 stood out as being necessary for formation of healthy fat cell metabolic phenotype by regulating expression of multiple fat cell phenotype-specific genes. CONCLUSION: This comprehensive RNAi screening in hASC suggests that a large proportion of WAT TFs that are impacted by metabolic conditions might be important for hyperplastic adipose tissue expansion. The screen also identified JARID2 as a novel TF essential for the development of functional adipocytes.


Assuntos
Adipócitos/metabolismo , Adipogenia/genética , Complexo Repressor Polycomb 2/genética , Interferência de RNA/fisiologia , Fatores de Transcrição/análise , Fatores de Transcrição/genética , Adipócitos/química , Adipócitos/patologia , Tecido Adiposo Branco/química , Tecido Adiposo Branco/patologia , Adolescente , Sequência de Bases , Diferenciação Celular/genética , Células Cultivadas , Feminino , Neoplasias Gastrointestinais , Regulação da Expressão Gênica , Humanos , Hiperplasia/genética , Resistência à Insulina/genética , Masculino , Obesidade/genética , Complexo Repressor Polycomb 2/fisiologia , Células-Tronco/química , Fatores de Transcrição/fisiologia
16.
Front Immunol ; 11: 564921, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33162978

RESUMO

In spite of intensive treatment Type 1 diabetes leads to serious complications. Preservation of residual beta cell function makes the disease milder, facilitates treatment, prevents complications and increase survival. So far immune interventions have had limited effect, and some serious adverse events and risks. In an open pilot trial we aimed to improve efficacy of GAD-alum treatment using lymph-node administration in combination with oral vitamin D. Here we report the clinical effect and focus on biomarkers for response to treatment. Patients (n = 12) aged 12 to 24 years with recent onset of Type 1 diabetes received 4 µg GAD-alum into lymph-node at day 30, 60, and 90, and oral Vitamin D 2000 U/d, days 1 to 120. Beta cell function was estimated by Mixed Meal Tolerance Tests. GADA, GADA subclasses, GAD65-induced cytokines and proliferation, and T cells markers were analyzed. The treatment was tolerable with no adverse events. Fasting C-peptide and insulin requirement remained stable at 15 months, while HbA1c was lower than baseline. Stimulated C-peptide showed no change at 6 months but declined after 15 months (81% of baseline). Eleven patients remained in partial remission (IDAAC < 9). Patients (n = 9) with better clinical outcome had reduced proportion of IgG1 and increased IgG2, IgG3, and IgG4, increased IL-10 secretion, and reduction of proliferation and CD8+ T cells activation. Patients with poorer clinical response had higher baseline levels of GAD65-induced cytokines and T-cell activation, and an increased ratio of effector/central memory T cells. Intra-lymphatic GAD treatment combined with Vitamin D might preserve beta cell function and improve clinical course in T1D. Patients with less benefit have a different quality of immune response both before and after treatment. Clinical Trial Registration: clinicaltrials.gov, identifier NCT02352974.


Assuntos
Hidróxido de Alumínio/administração & dosagem , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/imunologia , Glutamato Descarboxilase/administração & dosagem , Imunidade/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Linfonodos/imunologia , Vitamina D/administração & dosagem , Vitaminas/administração & dosagem , Administração Oral , Adolescente , Peptídeo C/metabolismo , Linfócitos T CD8-Positivos/imunologia , Criança , Feminino , Humanos , Imunoglobulina G/metabolismo , Injeções Intralinfáticas , Interleucina-10/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Masculino , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento , Adulto Jovem
17.
Cell Metab ; 31(2): 375-390.e11, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31866443

RESUMO

While obesity and associated metabolic complications are linked to inflammation of white adipose tissue (WAT), the causal factors remain unclear. We hypothesized that the local metabolic environment could be an important determinant. To this end, we compared metabolites released from WAT of 81 obese and non-obese women. This identified glutamine to be downregulated in obesity and inversely associated with a pernicious WAT phenotype. Glutamine administration in vitro and in vivo attenuated both pro-inflammatory gene and protein levels in adipocytes and WAT and macrophage infiltration in WAT. Metabolomic and bioenergetic analyses in human adipocytes suggested that glutamine attenuated glycolysis and reduced uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) levels. UDP-GlcNAc is the substrate for the post-translational modification O-linked ß-N-acetylglucosamine (O-GlcNAc) mediated by the enzyme O-GlcNAc transferase. Functional studies in human adipocytes established a mechanistic link between reduced glutamine, O-GlcNAcylation of nuclear proteins, and a pro-inflammatory transcriptional response. Altogether, glutamine metabolism is linked to WAT inflammation in obesity.


Assuntos
Adipócitos , Tecido Adiposo Branco , Glutamina , Inflamação/metabolismo , Obesidade/metabolismo , Acetilglucosamina , Adipócitos/metabolismo , Adipócitos/patologia , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Adulto , Animais , Células Cultivadas , Estudos de Coortes , Feminino , Glucose/metabolismo , Glutamina/metabolismo , Glutamina/farmacologia , Glicosilação/efeitos dos fármacos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , N-Acetilglucosaminiltransferases/metabolismo
18.
J Clin Endocrinol Metab ; 104(10): 4552-4562, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31132124

RESUMO

OBJECTIVE: Although IL-10 is generally considered as an anti-inflammatory cytokine, it was recently shown to have detrimental effects on insulin sensitivity and fat cell metabolism in rodents. Whether this also pertains to human white adipose tissue (hWAT) is unclear. We therefore determined the main cellular source and effects of IL-10 on human adipocytes and hWAT-resident immune cells and its link to insulin resistance. METHODS: Associations between hWAT IL-10 production and metabolic parameters were investigated in 216 participants with large interindividual variations in body mass index and insulin sensitivity. Adipose cells expressing or secreting IL-10 and the cognate IL-10 receptor α (IL10RA) were identified by flow cytometry sorting. Effects on adipogenesis, lipolysis, and inflammatory/metabolic gene expression were measured in two human primary adipocyte models. Secretion of inflammatory cytokines was investigated in cultures of IL-10-treated hWAT macrophages and leukocytes by Luminex analysis (Luminex Corp.). RESULTS: IL-10 gene expression and protein secretion in hWAT correlated positively with body mass index (BMI) and homeostasis model assessment-insulin resistance (HOMA-IR). Gene expression analyses in mature fat cells and flow cytometry-sorted hWAT-resident adipocyte progenitors, macrophages, and leukocytes demonstrated that the expression of IL-10 and the IL10RA were significantly enriched in proinflammatory M1 macrophages. In contrast to murine data, functional studies showed that recombinant IL-10 had no effect on adipocyte phenotype. In hWAT-derived macrophages and leukocytes, it induced an anti-inflammatory profile. CONCLUSION: In hWAT, IL-10 is upregulated in proinflammatory macrophages of obese and insulin-resistant persons. However, in contrast to findings in mice, IL-10 does not directly affect human adipocyte function.


Assuntos
Tecido Adiposo Branco/metabolismo , Resistência à Insulina , Interleucina-10/metabolismo , Macrófagos/metabolismo , Adipócitos Brancos/efeitos dos fármacos , Adipócitos Brancos/metabolismo , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Tecido Adiposo/metabolismo , Adulto , Estudos de Casos e Controles , Células Cultivadas , Feminino , Citometria de Fluxo , Expressão Gênica , Humanos , Técnicas In Vitro , Inflamação/genética , Interleucina-10/farmacologia , Subunidade alfa de Receptor de Interleucina-10/metabolismo , Gordura Intra-Abdominal , Lipólise/genética , Células-Tronco Mesenquimais/efeitos dos fármacos , Pessoa de Meia-Idade , Obesidade Metabolicamente Benigna , RNA Mensageiro , Gordura Subcutânea , Células THP-1 , Adulto Jovem
19.
J Diabetes Res ; 2018: 9391845, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30009185

RESUMO

GAD-alum given into lymph nodes to type 1 diabetes patients participating in an open-label pilot trial resulted in preservation of C-peptide similar to promising results from other trials. Here, we compared the immunomodulatory effect of giving GAD-alum directly into lymph nodes versus that induced by subcutaneous administration. Samples from T1D patients (n = 6) who received 4 µg GAD-alum into lymph nodes (LNs), followed by two booster injections one month apart, and from patients (n = 6) who received two subcutaneous injections (SC) (20 µg) given one month apart were compared. GADA, IA-2A, GADA subclasses, IgE, GAD65-induced cytokines, PBMC proliferation, and T cell markers were analyzed. Lower doses of GAD-alum into LN induced higher GADA levels than SC injections and reduced proliferation and IgG1 GADA subclass, while enhancing IgG2, IgG3, and IgG4. The cytokine profile was dominated by the Th2-associated cytokine IL-13, and GAD65 stimulation induced activated CD4 T cells. Patients responding clinically best account for most of the immunological changes. In contrast, SC treatment resulted in predominant IgG1, predominant IFN-γ, higher proliferation, and activated CD4 and CD8 cells. Patients from the LN group with best metabolic outcome seemed to have common immune correlates related to the treatment. This trial is registered with DIAGNODE (NCT02352974, clinicaltrials.gov) and DIABGAD (NCT01785108, clinicaltrials.gov).


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Glutamato Descarboxilase/farmacologia , Células Th2/imunologia , Adolescente , Compostos de Alúmen/farmacologia , Proliferação de Células , Criança , Citocinas/imunologia , Feminino , Humanos , Imunoglobulina G/imunologia , Imunomodulação , Imunofenotipagem , Leucócitos Mononucleares/imunologia , Linfonodos/imunologia , Ativação Linfocitária , Masculino , Projetos Piloto , Resultado do Tratamento , Adulto Jovem
20.
PLoS One ; 13(1): e0191067, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29346396

RESUMO

Micro RNAs (miRNAs) are promising disease biomarkers due to their high stability. Their expression in serum is altered in type 1 diabetes, but whether deviations exist in individuals with high risk for type 1 diabetes remains unexplored. We therefore assessed serum miRNAs in high-risk individuals (n = 21) positive for multiple islet autoantibodies, age-matched healthy children (n = 17) and recent-onset type 1 diabetes patients (n = 8), using Serum/Plasma Focus microRNA PCR Panels from Exiqon. The miRNA levels in the high-risk group were similar to healthy controls, and no specific miRNA profile was identified for the high-risk group. However, serum miRNAs appeared to reflect glycemic status and ongoing islet autoimmunity in high-risk individuals, since several miRNAs were associated to glucose homeostasis and autoantibody titers. High-risk individuals progressing to clinical disease after the sampling could not be clearly distinguished from non-progressors, while miRNA expression in the type 1 diabetes group deviated significantly from high-risk individuals and healthy controls, perhaps explained by major metabolic disturbances around the time of diagnosis.


Assuntos
Autoanticorpos/sangue , Diabetes Mellitus Tipo 1/sangue , Glucose/metabolismo , Ilhotas Pancreáticas/imunologia , MicroRNAs/sangue , Glicemia/metabolismo , Criança , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Feminino , Homeostase , Humanos , Masculino , Reação em Cadeia da Polimerase , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA