Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 3724, 2023 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-37349288

RESUMO

Cancers are often associated with hypoxia and metabolic reprogramming, resulting in enhanced tumor progression. Here, we aim to study breast cancer hypoxia responses, focusing on secreted proteins from low-grade (luminal-like) and high-grade (basal-like) cell lines before and after hypoxia. We examine the overlap between proteomics data from secretome analysis and laser microdissected human breast cancer stroma, and we identify a 33-protein stromal-based hypoxia profile (33P) capturing differences between luminal-like and basal-like tumors. The 33P signature is associated with metabolic differences and other adaptations following hypoxia. We observe that mRNA values for 33P predict patient survival independently of molecular subtypes and basic prognostic factors, also among low-grade luminal-like tumors. We find a significant prognostic interaction between 33P and radiation therapy.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/patologia , Proteoma/metabolismo , Perfilação da Expressão Gênica , Linhagem Celular Tumoral , Hipóxia/genética , Regulação Neoplásica da Expressão Gênica
2.
bioRxiv ; 2023 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-37205344

RESUMO

Tumor neurogenesis, a process by which new nerves invade tumors, is a growing area of interest in cancer research. Nerve presence has been linked to aggressive features of various solid tumors, including breast and prostate cancer. A recent study suggested that the tumor microenvironment may influence cancer progression through recruitment of neural progenitor cells from the central nervous system. However, the presence of neural progenitors in human breast tumors has not been reported. Here, we investigate the presence of Doublecortin (DCX) and Neurofilament-Light (NFL) co-expressing (DCX+/NFL+) cells in patient breast cancer tissue using Imaging Mass Cytometry. To map the interaction between breast cancer cells and neural progenitor cells further, we created an in vitro model mimicking breast cancer innervation, and characterized using mass spectrometry-based proteomics on the two cell types as they co- evolved in co-culture. Our results indicate stromal presence of DCX+/NFL+ cells in breast tumor tissue from a cohort of 107 patient cases, and that neural interaction contribute to drive a more aggressive breast cancer phenotype in our co-culture models. Our results support that neural involvement plays an active role in breast cancer and warrants further studies on the interaction between nervous system and breast cancer progression.

3.
Cancer Genomics Proteomics ; 19(2): 130-144, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35181583

RESUMO

BACKGROUND/AIM: Better stratification of the risk of relapse will help select the right patients for adjuvant treatment and improve targeted therapies for patients with colon cancer. MATERIALS AND METHODS: To understand why a subset of tumors relapse, we compared the proteome of two groups of patients with colon cancer with similar stage, stratified based on the presence or absence of recurrence. RESULTS: Using tumor biopsies from the primary operation, we identified dissimilarity between recurrent and nonrecurrent mismatch satellite stable colon cancer and found that signaling related to immune activation and inflammation was associated with relapse. CONCLUSION: Immune modulation may have an effect on mismatch satellite stable colon cancer. At present, immune therapy is offered primarily to microsatellite instable colon cancer. Hopefully, immune therapy in mismatch satellite stable colon cancer beyond PD-1 and PD-L1 inhibitors can be implemented.


Assuntos
Neoplasias do Colo , Sistema Imunitário , Proteoma , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Humanos , Repetições de Microssatélites , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/imunologia , Prognóstico
4.
Stem Cell Reports ; 15(5): 1067-1079, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33125875

RESUMO

The role of leptin receptor (OB-R) signaling in linking pluripotency with growth and development and the consequences of dysfunctional leptin signaling on progression of metabolic disease is poorly understood. Using a global unbiased proteomics approach we report that embryonic fibroblasts (MEFs) carrying the db/db mutation exhibit metabolic abnormalities, while their reprogrammed induced pluripotent stem cells (iPSCs) show altered expression of proteins involved in embryonic development. An upregulation in expression of eukaryotic translation initiation factor 4e (Eif4e) and Stat3 binding to the Eif4e promoter was supported by enhanced protein synthesis in mutant iPSCs. Directed differentiation of db/db iPSCs toward the neuronal lineage showed defects. Gene editing to correct the point mutation in db/db iPSCs using CRISPR-Cas9, restored expression of neuronal markers and protein synthesis while reversing the metabolic defects. These data imply a direct role for OB-R in regulating metabolism in embryonic fibroblasts and key developmental pathways in iPSCs.


Assuntos
Fator de Iniciação 4E em Eucariotos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Biossíntese de Proteínas , Receptores para Leptina/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Animais , Sistemas CRISPR-Cas , Diferenciação Celular , Linhagem da Célula , Fator de Iniciação 4E em Eucariotos/genética , Fibroblastos/metabolismo , Edição de Genes , Regulação da Expressão Gênica no Desenvolvimento , Metaboloma , Camundongos , Camundongos Knockout , Neurogênese , Proteínas , Proteômica , Receptores para Leptina/genética
5.
Int J Mol Sci ; 21(21)2020 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-33105604

RESUMO

Abnormal hepatic insulin signaling is a cause or consequence of hepatic steatosis. DPP-4 inhibitors might be protective against fatty liver. We previously reported that the systemic inhibition of insulin receptor (IR) and IGF-1 receptor (IGF1R) by the administration of OSI-906 (linsitinib), a dual IR/IGF1R inhibitor, induced glucose intolerance, hepatic steatosis, and lipoatrophy in mice. In the present study, we investigated the effects of a DPP-4 inhibitor, linagliptin, on hepatic steatosis in OSI-906-treated mice. Unlike high-fat diet-induced hepatic steatosis, OSI-906-induced hepatic steatosis is not characterized by elevations in inflammatory responses or oxidative stress levels. Linagliptin improved OSI-906-induced hepatic steatosis via an insulin-signaling-independent pathway, without altering glucose levels, free fatty acid levels, gluconeogenic gene expressions in the liver, or visceral fat atrophy. Hepatic quantitative proteomic and phosphoproteomic analyses revealed that perilipin-2 (PLIN2), major urinary protein 20 (MUP20), cytochrome P450 2b10 (CYP2B10), and nicotinamide N-methyltransferase (NNMT) are possibly involved in the process of the amelioration of hepatic steatosis by linagliptin. Thus, linagliptin improved hepatic steatosis induced by IR and IGF1R inhibition via a previously unknown mechanism that did not involve gluconeogenesis, lipogenesis, or inflammation, suggesting the non-canonical actions of DPP-4 inhibitors in the treatment of hepatic steatosis under insulin-resistant conditions.


Assuntos
Imidazóis/efeitos adversos , Linagliptina/farmacologia , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Pirazinas/efeitos adversos , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor de Insulina/antagonistas & inibidores , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Glicemia/metabolismo , Família 2 do Citocromo P450/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Imidazóis/farmacologia , Insulina/sangue , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Perilipina-2/metabolismo , Pirazinas/farmacologia , Esteroide Hidroxilases/metabolismo , Triglicerídeos/sangue
6.
Biomedicines ; 8(7)2020 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-32605028

RESUMO

Mutations in the hepatocyte nuclear factor 4α (HNF4α) gene affect prenatal and postnatal pancreas development, being characterized by insulin-producing ß-cell dysfunction. Little is known about the cellular and molecular mechanisms leading to ß-cell failure as result of HNF4α mutation. In this study, we compared the miRNA profile of differentiating human induced pluripotent stem cells (hiPSC) derived from HNF4α+/Δ mutation carriers and their family control along the differentiation timeline. Moreover, we associated this regulation with the corresponding transcriptome profile to isolate transcript-miRNA partners deregulated in the mutated cells. This study uncovered a steep difference in the miRNA regulation pattern occurring during the posterior foregut to pancreatic endoderm transition, defining early and late differentiation regulatory windows. The pathway analysis of the miRNAome-transcriptome interactions revealed a likely gradual involvement of HNF4α+/Δ mutation in p53-mediated cell cycle arrest, with consequences for the proliferation potential, survival and cell fate acquisition of the differentiating cells. The present study is based on bioinformatics approaches and we expect that, pending further experimental validation, certain miRNAs deregulated in the HNF4α+/Δ cells would prove useful for therapy.

7.
Front Cell Dev Biol ; 8: 109, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32161757

RESUMO

Generating insulin-producing ß-cells from human induced pluripotent stem cells is a promising cell replacement therapy for improving or curing insulin-dependent diabetes. The transplantation of end-stages differentiating cells into living hosts was demonstrated to improve ß-cell maturation. Nevertheless, the cellular and molecular mechanisms outlining the transplanted cells' response to the in vivo environment are still to be properly characterized. Here we use global proteomics and large-scale imaging techniques to demultiplex and filter the cellular processes and molecular signatures modulated by the immediate in vivo effect. We show that in vivo exposure swiftly confines in vitro generated human pancreatic progenitors to single hormone expression. The global proteome landscape of the transplanted cells was closer to native human islets, especially in regard to energy metabolism and redox balance. Moreover, our study indicates a possible link between these processes and certain epigenetic regulators involved in cell identity. Pathway analysis predicted HNF1A and HNF4A as key regulators controlling the in vivo islet-promoting response, with experimental evidence suggesting their involvement in confining islet cell fate following xeno-transplantation.

8.
Sci Rep ; 10(1): 414, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31942009

RESUMO

Cell replacement therapies hold great therapeutic potential. Nevertheless, our knowledge of the mechanisms governing the developmental processes is limited, impeding the quality of differentiation protocols. Generating insulin-expressing cells in vitro is no exception, with the guided series of differentiation events producing heterogeneous cell populations that display mixed pancreatic islet phenotypes and immaturity. The achievement of terminal differentiation ultimately requires the in vivo transplantation of, usually, encapsulated cells. Here we show the impact of cell confinement on the pancreatic islet signature during the guided differentiation of alginate encapsulated human induced pluripotent stem cells (hiPSCs). Our results show that encapsulation improves differentiation by significantly reshaping the proteome landscape of the cells towards an islet-like signature. Pathway analysis is suggestive of integrins transducing the encapsulation effect into intracellular signalling cascades promoting differentiation. These analyses provide a molecular framework for understanding the confinement effects on hiPSCs differentiation while confirming its importance for this process.


Assuntos
Alginatos/farmacologia , Biomarcadores/metabolismo , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Integrinas/metabolismo , Ilhotas Pancreáticas/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Fenótipo , Transdução de Sinais
9.
Acta Physiol (Oxf) ; 228(4): e13433, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31872528

RESUMO

AIM: The loss of insulin-secreting ß-cells, ultimately characterizing most diabetes forms, demands the development of cell replacement therapies. The common endpoint for all ex vivo strategies is transplantation into diabetic patients. However, the effects of hyperglycaemia environment on the transplanted cells were not yet properly assessed. Thus, the main goal of this study was to characterize global effect of brief and prolonged in vivo hyperglycaemia exposure on the cell fate acquisition and maintenance of transplanted human pancreatic progenitors. METHODS: To rigorously study the effect of hyperglycaemia, in vitro differentiated human-induced pluripotent stem cells (hiPSC)-derived pancreatic progenitors were xenotransplanted in normoglycaemic and diabetic NSG rat insulin promoter (RIP)-diphtheria toxin receptor (DTR) mice. The transplants were retrieved after 1-week or 1-month exposure to overt hyperglycaemia and analysed by large-scale microscopy or global proteomics. For this study we pioneer the use of the NSG RIP-DTR system in the transplantation of hiPSC, making use of its highly reproducible specific and absolute ß-cell ablation property in the absence of inflammation or other organ toxicity. RESULTS: Here we show for the first time that besides the presence of an induced oxidative stress signature, the cell fate and proteome landscape response to hyperglycaemia was different, involving largely different mechanisms, according to the period spent in the hyperglycaemic environment. Surprisingly, brief hyperglycaemia exposure increased the bihormonal cell number by impeding the activity of specific islet lineage determinants. Moreover, it activated antioxidant and inflammation protection mechanisms signatures in the transplanted cells. In contrast, the prolonged exposure was characterized by decreased numbers of hormone + cells, low/absent detoxification signature, augmented production of oxygen reactive species and increased apoptosis. CONCLUSION: Hyperglycaemia exposure induced distinct, period-dependent, negative effects on xenotransplanted human pancreatic progenitor, affecting their energy homeostasis, cell fate acquisition and survival.


Assuntos
Diferenciação Celular/fisiologia , Hiperglicemia/fisiopatologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células Secretoras de Insulina/fisiologia , Estresse Oxidativo/fisiologia , Adulto , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/transplante , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , Ratos , Transplante Heterólogo
10.
Stem Cells ; 38(4): 542-555, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31828876

RESUMO

A comprehensive characterization of the molecular processes controlling cell fate decisions is essential to derive stable progenitors and terminally differentiated cells that are functional from human pluripotent stem cells (hPSCs). Here, we report the use of quantitative proteomics to describe early proteome adaptations during hPSC differentiation toward pancreatic progenitors. We report that the use of unbiased quantitative proteomics allows the simultaneous profiling of numerous proteins at multiple time points, and is a valuable tool to guide the discovery of signaling events and molecular signatures underlying cellular differentiation. We also monitored the activity level of pathways whose roles are pivotal in the early pancreas differentiation, including the Hippo signaling pathway. The quantitative proteomics data set provides insights into the dynamics of the global proteome during the transition of hPSCs from a pluripotent state toward pancreatic differentiation.


Assuntos
Pâncreas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Diferenciação Celular , Humanos , Pâncreas/citologia
11.
Stem Cells Int ; 2019: 8036035, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31827534

RESUMO

Human induced pluripotent stem cells (hiPSCs) are of high interest because they can be differentiated into a vast range of different cell types. Ideally, reprogrammed cells should sustain long-term culturing in an undifferentiated state. However, some reprogrammed cell lines represent an unstable state by spontaneously differentiating and changing their cellular phenotype and colony morphology. This phenomenon is not fully understood, and no method is available to predict it reliably. In this study, we analyzed and compared the proteome landscape of 20 reprogrammed cell lines classified as stable and unstable based on long-term colony morphology. We identified distinct proteomic signatures associated with stable colony morphology and with unstable colony morphology, although the typical pluripotency markers (POU5F1, SOX2) were present with both morphologies. Notably, epithelial to mesenchymal transition (EMT) protein markers were associated with unstable colony morphology, and the transforming growth factor beta (TGFB) signalling pathway was predicted as one of the main regulator pathways involved in this process. Furthermore, we identified specific proteins that separated the stable from the unstable state. Finally, we assessed both spontaneous embryonic body (EB) formation and directed differentiation and showed that reprogrammed lines with an unstable colony morphology had reduced differentiation capacity. To conclude, we found that different defined patterns of colony morphology in reprogrammed cells were associated with distinct proteomic profiles and different outcomes in differentiation capacity.

12.
Nat Metab ; 1(5): 509-518, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31423480

RESUMO

Type 1 diabetes (T1D) is characterized by pancreatic islet infiltration by autoreactive immune cells and a near-total loss of ß-cells1. Restoration of insulin-producing ß-cells coupled with immunomodulation to suppress the autoimmune attack has emerged as a potential approach to counter T1D2-4. Here we report that enhancing ß-cell mass early in life, in two models of female NOD mice, results in immunomodulation of T-cells, reduced islet infiltration and lower ß-cell apoptosis, that together protect them from developing T1D. The animals displayed altered ß-cell antigens, and islet transplantation studies showed prolonged graft survival in the NOD-LIRKO model. Adoptive transfer of splenocytes from the NOD-LIRKOs prevented development of diabetes in pre-diabetic NOD mice. A significant increase in the splenic CD4+CD25+FoxP3+ regulatory T-cell (Treg) population was observed to underlie the protected phenotype since Treg depletion rendered NOD-LIRKO mice diabetic. The increase in Tregs coupled with activation of TGF-ß/SMAD3 signaling pathway in pathogenic T-cells favored reduced ability to kill ß-cells. These data support a previously unidentified observation that initiating ß-cell proliferation, alone, prior to islet infiltration by immune cells alters the identity of ß-cells, decreases pathologic self-reactivity of effector cells and increases Tregs to prevent progression of T1D.


Assuntos
Proliferação de Células , Diabetes Mellitus Tipo 1/patologia , Sistema Imunitário/imunologia , Células Secretoras de Insulina/patologia , Animais , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Progressão da Doença , Humanos , Camundongos
13.
Artigo em Inglês | MEDLINE | ID: mdl-31139151

RESUMO

Current published protocols for targeted differentiation of human stem cells toward pancreatic ß-cells fail to deliver sufficiently mature cells with functional properties comparable to human islet ß-cells. We aimed to assess whether Wnt-modulation could promote the final protocol stages of ß-cell maturation, building our hypothesis on our previous findings of Wnt activation in immature hiPSC-derived stage 7 (S7) cells compared to adult human islets and with recent data reporting a link between Wnt/PCP and in vitro ß-cell maturation. In this study, we stimulated canonical and non-canonical Wnt signaling in hiPSC-derived S7 cells using syntetic proteins including WNT3A, WNT4, WNT5A and WNT5B, and we inhibited endogenous Wnt signaling with the Tankyrase inhibitor G007-LK (TKi). Whereas neither canonical nor non-canonical Wnt stimulation alone was able to mature hiPSC-derived S7 cells, WNT-inhibition with TKi increased the fraction of monohormonal cells and global proteomics of TKi-treated S7 cells showed a proteomic signature more similar to adult human islets, suggesting that inhibition of endogenous Wnt contributes toward final ß-cell maturation.

14.
Nature ; 567(7746): 43-48, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30760930

RESUMO

Cell-identity switches, in which terminally differentiated cells are converted into different cell types when stressed, represent a widespread regenerative strategy in animals, yet they are poorly documented in mammals. In mice, some glucagon-producing pancreatic α-cells and somatostatin-producing δ-cells become insulin-expressing cells after the ablation of insulin-secreting ß-cells, thus promoting diabetes recovery. Whether human islets also display this plasticity, especially in diabetic conditions, remains unknown. Here we show that islet non-ß-cells, namely α-cells and pancreatic polypeptide (PPY)-producing γ-cells, obtained from deceased non-diabetic or diabetic human donors, can be lineage-traced and reprogrammed by the transcription factors PDX1 and MAFA to produce and secrete insulin in response to glucose. When transplanted into diabetic mice, converted human α-cells reverse diabetes and continue to produce insulin even after six months. Notably, insulin-producing α-cells maintain expression of α-cell markers, as seen by deep transcriptomic and proteomic characterization. These observations provide conceptual evidence and a molecular framework for a mechanistic understanding of in situ cell plasticity as a treatment for diabetes and other degenerative diseases.


Assuntos
Diabetes Mellitus/patologia , Diabetes Mellitus/terapia , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/patologia , Animais , Biomarcadores/análise , Linhagem da Célula/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Diabetes Mellitus/imunologia , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Feminino , Glucagon/metabolismo , Células Secretoras de Glucagon/efeitos dos fármacos , Células Secretoras de Glucagon/transplante , Glucose/farmacologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Fatores de Transcrição Maf Maior/genética , Fatores de Transcrição Maf Maior/metabolismo , Masculino , Camundongos , Especificidade de Órgãos/efeitos dos fármacos , Polipeptídeo Pancreático/metabolismo , Células Secretoras de Polipeptídeo Pancreático/citologia , Células Secretoras de Polipeptídeo Pancreático/efeitos dos fármacos , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Proteômica , Análise de Sequência de RNA , Transativadores/genética , Transativadores/metabolismo , Transcriptoma , Transdução Genética
15.
PLoS One ; 13(11): e0206475, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30419021

RESUMO

Patients with bladder cancer need frequent controls over long follow-up time due to high recurrence rate and risk of conversion to muscle invasive cancer with poor prognosis. We identified cancer-related molecular signatures in apparently healthy bladder in patients with subsequent muscular invasiveness during follow-up. Global proteomics of the normal tissue biopsies revealed specific proteome fingerprints in these patients prior to subsequent muscular invasiveness. In these presumed normal samples, we detected modulations of proteins previously associated with different cancer types. This study indicates that analyzing apparently healthy tissue of a cancer-invaded organ may suggest disease progression.


Assuntos
Progressão da Doença , Músculos/patologia , Proteômica , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Prognóstico , Neoplasias da Bexiga Urinária/diagnóstico , Neoplasias da Bexiga Urinária/genética
16.
Sci Rep ; 7(1): 4780, 2017 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-28684784

RESUMO

MODY1 is a maturity-onset monogenic diabetes, caused by heterozygous mutations of the HNF4A gene. To date the cellular and molecular mechanisms leading to disease onset remain largely unknown. In this study, we demonstrate that insulin-positive cells can be generated in vitro from human induced pluripotent stem cells (hiPSCs) derived from patients carrying a non-sense HNF4A mutation, proving for the first time, that a human HNF4A mutation is neither blocking the expression of the insulin genes nor the development of insulin-producing cells in vitro. However, regardless of the mutation or diabetes status, these insulin-producing cells are immature, a common downfall off most current ß-cell differentiation protocols. To further address the immature state of the cells, in vitro differentiated cells and adult human islets were compared by global proteomic analysis. We report the predicted upstream regulators and signalling pathways characterizing the proteome landscape of each entity. Subsequently, we focused on the molecular components absent or misregulated in the in vitro differentiated cells, to probe the components involved in the deficient in vitro maturation towards fully functional ß-cells. This analysis identified the modulation of key developmental signalling pathways representing potential targets for improving the efficiency of the current differentiation protocols.


Assuntos
Diferenciação Celular , Diabetes Mellitus Tipo 2/genética , Células-Tronco Pluripotentes Induzidas/fisiologia , Células Secretoras de Insulina/fisiologia , Proteoma/análise , Adulto , Técnicas de Cultura de Células , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas , Masculino , Modelos Genéticos
17.
Nephron ; 132(1): 70-80, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26745798

RESUMO

BACKGROUND: Tubular atrophy and interstitial fibrosis mark the final stage in most forms of progressive kidney diseases. Little is known regarding changes in the tubular proteome. In this study, we investigated changes in the tubular proteome of normal or minimally damaged tubular tissue in the non-clipped kidney from rats with two-kidney one-clip (2K1C) hypertension. METHODS: Formalin-fixed paraffin-embedded kidney sections from four 2K1C rats with hypertensive kidney damage and 6 sham rats were used. Tubulointerstitial tissue without discernable interstitial expansion or pronounced tubular alterations was microdissected and this was assumed to represent an early stage of chronic tubular damage in 2K1C. Samples were analyzed by mass spectrometry and relative protein abundances were compared between 2K1C and sham. RESULTS: A total of 1,160 proteins were identified with at least 2 unique peptides, allowing for relative quantitation between samples. Among these, 151 proteins were more abundant, and 192 proteins were less abundant in 2K1C compared with sham. Transgelin, vimentin and creatine kinase B-type were among the proteins that were most increased in 2K1C. Ingenuity Pathway Analysis showed increased abundance of proteins related to Rho signaling and protein turnover (eIF2 signaling and protein ubiquitination), and decreased abundance of proteins related to fatty acid ß-oxidation. CONCLUSION: Tubular tissue from normal or minimally damaged hypertensive kidney damage demonstrate extensive proteomic changes with upregulation of pathways associated with progressive kidney damage, such as Rho signaling and protein turnover. Thus, proteomics presents itself to be a promising tool for the discovery of early damage markers from not yet morphologically visible tubular damage.


Assuntos
Hipertensão Renovascular/genética , Hipertensão Renovascular/metabolismo , Rim/patologia , Proteômica , Animais , Pressão Sanguínea , Túbulos Renais/patologia , Masculino , Tamanho do Órgão , Proteinúria/genética , Proteinúria/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/genética , Regulação para Cima
18.
Proteomics ; 15(19): 3361-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26152395

RESUMO

Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS with unknown cause. Proteins with different abundance in the cerebrospinal fluid (CSF) from relapsing-remitting MS (RRMS) patients and neurological controls could give novel insight to the MS pathogenesis and be used to improve diagnosis, predict prognosis and disease course, and guide in therapy decisions. We combined iTRAQ labeling and Orbitrap mass spectrometry to discover proteins with different CSF abundance between six RRMS patients and 18 neurological disease controls. From 777 quantified proteins seven were selected as biomarker candidates, namely chitinase-3-like protein 1, secretogranin-1 (Sg1), cerebellin-1, neuroserpin, cell surface glycoprotein MUC18, testican-2 and glutamate receptor 4. An independent sample set of 13 early-MS patients, 13 RRMS patients and 13 neurological controls was used in a multiple reaction monitoring verification study. We found the intracellular calcium binding protein Sg1 to be increased in early-MS patients compared to RRMS and neurological controls. Sg1 should be included in further studies to elucidate its role in the early phases of MS pathogenesis and its potential as a biomarker for this disease.


Assuntos
Cromogranina B/líquido cefalorraquidiano , Esclerose Múltipla/líquido cefalorraquidiano , Adulto , Biomarcadores , Cromogranina B/genética , Progressão da Doença , Regulação para Baixo , Feminino , Humanos , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Esclerose Múltipla/diagnóstico , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Proteômica
19.
J Proteome Res ; 14(1): 521-30, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25369532

RESUMO

Patients with carboxyl-ester lipase-maturity-onset diabetes of the young (CEL-MODY) display distinct disease stages toward the development of monogenic diabetes and exocrine pancreatic disease. The finding of differentially increased proteins, some related to MAPK signaling, in a discovery proteomics study of secretin-stimulated duodenal juice in three CEL-MODY patients, prompted us to monitor their abundance in an extensive number of CEL-MODY subjects at different disease stages and controls using targeted proteomics. In the current study, we demonstrate the feasibility of selected reaction monitoring assays to quantify protein levels in secretin-stimulated duodenal juice. Furthermore, we define a set of five peptides for potential use as diagnostic tests in CEL-MODY patients. Finally, we propose a further set of seven proteins with a likely pathogenic role in CEL-MODY disease progression.


Assuntos
Biomarcadores/metabolismo , Carboxilesterase/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Duodeno/metabolismo , Secreções Intestinais/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Diabetes Mellitus Tipo 2/diagnóstico , Humanos , Marcação por Isótopo , Espectrometria de Massas , Estatísticas não Paramétricas , alfa-Amilases/metabolismo
20.
J Hypertens ; 33(1): 126-35, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25304470

RESUMO

BACKGROUND: Hypertensive nephrosclerosis is one of the most frequent causes of chronic kidney failure. Proteome analysis potentially improves the pathophysiological understanding and diagnostic precision of this disorder. In the present exploratory study, we investigated experimental nephrosclerosis in the two-kidney, one-clip (2K1C) hypertensive rat model. METHODS: The renal cortex proteome from juxtamedullary cortex and outer cortex of 2K1C male Wistar-Hannover rats (n = 4) was compared with the sham-operated controls (n = 6), using mass spectrometry-based quantitative proteomics. We combined a high abundant plasma protein depletion strategy with an extended liquid chromatographic gradient to improve peptide and protein identification. Immunohistology was used for independent confirmation of abundance. RESULTS: We identified 1724 proteins, of which 1434 were quantified with at least two unique peptides. Comparative proteomics revealed 608 proteins, including the platelet-derived growth factor receptor-ß signalling pathway, with different abundances between the non-clipped kidney of hypertensive 2K1C rats and the corresponding kidney of the normotensive controls (P < 0.05, absolute fold change ≥1.5). Among the most significantly altered proteins in the whole cortex were periostin, transgelin, and creatine kinase B-type. Relative abundance of periostin alone allowed clear classification of 2K1C and controls. Enrichment of periostin in 2K1C rats was verified by immunohistology, showing positivity especially around the fibrotic vessels. CONCLUSION: The proteome is altered in hypertension-induced kidney damage. We propose periostin, especially in combination with transgelin and creatine kinase B-type, as possible proteomic classifier to distinguish hypertensive nephrosclerosis from the normal tissue. This classifier needs to be further validated with respect to early diagnosis of fibrosis, prognosis, and its potential as a novel molecular target for pharmacological interventions.


Assuntos
Hipertensão Renovascular/fisiopatologia , Córtex Renal/fisiopatologia , Rim/fisiopatologia , Proteoma/metabolismo , Proteômica/métodos , Animais , Pressão Sanguínea , Moléculas de Adesão Celular/biossíntese , Creatina Quinase Forma BB/biossíntese , Modelos Animais de Doenças , Masculino , Proteínas dos Microfilamentos/biossíntese , Proteínas Musculares/biossíntese , Nefroesclerose/fisiopatologia , Ratos , Ratos Wistar , Instrumentos Cirúrgicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA