Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 41(10): 1507-1517, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35082384

RESUMO

Molecular events occurring in stepwise progression from pre-malignant lesions (pancreatic intraepithelial neoplasia; PanIN) to the development of pancreatic ductal adenocarcinoma (PDAC) are poorly understood. Thus, characterization of early PanIN lesions may reveal markers that can help in diagnosing PDAC at an early stage and allow understanding the pathology of the disease. We performed the molecular and histological assessment of patient-derived PanINs, tumor tissues and pancreas from mouse models with PDAC (KC mice that harbor K-RAS mutation in pancreatic tissue), where we noted marked upregulation of gastrokine (GKN) proteins. To further understand the role of gastrokine proteins in PDAC development, GKN-deficient KC mice were developed by intercrossing gastrokine-deficient mice with KC mice. Panc-02 (pancreatic cancer cells of mouse origin) were genetically modified to express GKN1 for further in vitro and in vivo analysis. Our results show that gastrokine proteins were absent in healthy pancreas and invasive cancer, while its expression was prominent in low-grade PanINs. We could detect these proteins in pancreatic juice and serum of KC mice. Furthermore, accelerated PanIN and tumor development were noted in gastrokine deficient KC mice. Loss of gastrokine 1 protein delayed apoptosis during carcinogenesis leading to the development of desmoplastic stroma while loss of gastrokine 2 increased the proliferation rate in precursor lesions. In summary, we identified gastrokine proteins in early pancreatic precursor lesions, where gastrokine proteins delay pancreatic carcinogenesis.


Assuntos
Carcinoma in Situ , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Hormônios Peptídicos , Animais , Carcinogênese , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patologia , Carcinoma Ductal Pancreático/patologia , Humanos , Camundongos , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas
2.
Immunohorizons ; 4(11): 688-700, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33154038

RESUMO

Autoimmune pancreatitis (AIP) is a rare form of chronic pancreatitis, for which treatment options, especially the long-term management, are limited. The only therapy that has been established and accepted so far is corticosteroids, but the relapse rate is significant. In the current study, we discern the effector mechanisms of targeted LTßR pathway inhibition using LTßR-Ig. Furthermore, the efficacy of LTßR-Ig therapy is compared with the depletion of immune cell subsets (CD4+ and CD20+), which are suggested to play a pathological role in AIP development. Three well-established mouse models of AIP were used to examine treatment efficacies and mechanisms. Tg(Ela1-Lta,b) mice represent a genetic model, in which AIP develops spontaneously. In MRL/Mp and IL-10-/- mice, AIP is induced by repeated polyinosinic:polycytidylic acid injection. Mice with AIP were treated with anti-CD20, anti-CD4 mAbs, or targeted LTßR-Ig. LTßR-Ig and anti-CD20 treatment led to significant improvement of AIP, including a decrease in autoantibody production and pancreatic inflammation in Tg(Ela1-Lta,b) and IL-10-/- mice. The molecular mechanism of this beneficial effect possibly involves the downregulation of Stat3 and noncanonical NF-κb activation. Anti-CD4 treatment reduced Th1 and Th2 signature but did not alleviate AIP. Additionally, in contrast to anti-CD20 or anti-CD4 treatments, blocking LTßR signaling disrupted tertiary lymphoid organs in all three models. We demonstrate that treatment with LTßR-Ig or anti-CD20 Ab alleviated murine AIP. LTßR-Ig treatment for AIP was effective in both lymphotoxin-dependent and lymphotoxin-independent AIP models, possibly because of its dual anti-inflammatory and antiautoimmune mechanisms.


Assuntos
Anticorpos Monoclonais/farmacologia , Pancreatite Autoimune/tratamento farmacológico , Imunoglobulina G/farmacologia , Interleucina-10/metabolismo , Receptor beta de Linfotoxina/efeitos dos fármacos , Animais , Antígenos CD20/imunologia , Pancreatite Autoimune/induzido quimicamente , Pancreatite Autoimune/patologia , Antígenos CD4/imunologia , Modelos Animais de Doenças , Feminino , Interleucina-10/genética , Receptor beta de Linfotoxina/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Poli I-C/administração & dosagem , Transdução de Sinais/imunologia
3.
Cancer Cell ; 36(3): 250-267.e9, 2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31526758

RESUMO

How lymphoma cells (LCs) invade the brain during the development of central nervous system lymphoma (CNSL) is unclear. We found that NF-κB-induced gliosis promotes CNSL in immunocompetent mice. Gliosis elevated cell-adhesion molecules, which increased LCs in the brain but was insufficient to induce CNSL. Astrocyte-derived CCL19 was required for gliosis-induced CNSL. Deleting CCL19 in mice or CCR7 from LCs abrogated CNSL development. Two-photon microscopy revealed LCs transiently entering normal brain parenchyma. Astrocytic CCL19 enhanced parenchymal CNS retention of LCs, thereby promoting CNSL formation. Aged, gliotic wild-type mice were more susceptible to forming CNSL than young wild-type mice, and astrocytic CCL19 was observed in both human gliosis and CNSL. Therefore, CCL19-CCR7 interactions may underlie an increased age-related risk for CNSL.


Assuntos
Envelhecimento/patologia , Neoplasias do Sistema Nervoso Central/patologia , Quimiocina CCL19/metabolismo , Gliose/patologia , Linfoma/patologia , Adolescente , Adulto , Idoso , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/diagnóstico por imagem , Barreira Hematoencefálica/patologia , Linhagem Celular Tumoral/transplante , Neoplasias do Sistema Nervoso Central/diagnóstico por imagem , Neoplasias do Sistema Nervoso Central/cirurgia , Quimiocina CCL19/genética , Quimiocina CXCL12 , Modelos Animais de Doenças , Feminino , Gliose/diagnóstico por imagem , Humanos , Microscopia Intravital , Linfoma/diagnóstico por imagem , Linfoma/cirurgia , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência por Excitação Multifotônica , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Receptores CCR7/genética , Receptores CCR7/metabolismo , Imagem com Lapso de Tempo , Adulto Jovem
4.
J Pathol ; 246(3): 352-365, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30058725

RESUMO

Pancreatic ductal adenocarcinoma (PDAC), which is the primary cause of pancreatic cancer mortality, is poorly responsive to currently available interventions. Identifying new targets that drive PDAC formation and progression is critical for developing alternative therapeutic strategies to treat this lethal malignancy. Using genetic and pharmacological approaches, we investigated in vivo and in vitro whether uptake of the monoamine serotonin [5-hydroxytryptamine (5-HT)] is required for PDAC development. We demonstrated that pancreatic acinar cells have the ability to readily take up 5-HT in a transport-mediated manner. 5-HT uptake promoted activation of the small GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1), which is required for transdifferentiation of acinar cells into acinar-to-ductal metaplasia (ADM), a key determinant in PDAC development. Consistent with the central role played by Rac1 in ADM formation, inhibition of the 5-HT transporter Sert (Slc6a4) with fluoxetine reduced ADM formation both in vitro and in vivo in a cell-autonomous manner. In addition, fluoxetine treatment profoundly compromised the stromal reaction and affected the proliferation and lipid metabolism of malignant PDAC cells. We propose that Sert is a promising therapeutic target to counteract the early event of ADM, with the potential to stall the initiation and progression of pancreatic carcinogenesis. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Carcinoma Ductal Pancreático/enzimologia , Proliferação de Células , Genes ras , Neuropeptídeos/metabolismo , Pâncreas/enzimologia , Neoplasias Pancreáticas/enzimologia , Serotonina/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/prevenção & controle , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transdiferenciação Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Embrião de Galinha , Modelos Animais de Doenças , Ativação Enzimática , Fluoxetina/farmacologia , Predisposição Genética para Doença , Humanos , Metaplasia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neovascularização Patológica , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/prevenção & controle , Fenótipo , Ratos , Proteínas da Membrana Plasmática de Transporte de Serotonina/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Transdução de Sinais , Triptofano Hidroxilase/genética , Triptofano Hidroxilase/metabolismo
5.
Sci Rep ; 8(1): 9391, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29925922

RESUMO

Adult pancreatic acinar cells have the ability to re-enter the cell cycle and proliferate upon injury or tissue loss. Despite this mitotic ability, the extent of acinar proliferation is often limited and unable to completely regenerate the injured tissue or restore the initial volume of the organ, thus leading to pancreatic dysfunction. Identifying molecular determinants of enhanced proliferation is critical to overcome this issue. In this study, we discovered that Murphy Roths Large (MRL/MpJ) mice can be exploited to identify molecular effectors promoting acinar proliferation upon injury, with the ultimate goal to develop therapeutic regimens to boost pancreatic regeneration. Our results show that, upon cerulein-induced acinar injury, cell proliferation was enhanced and cell cycle components up-regulated in the pancreas of MRL/MpJ mice compared to the control strain C57BL/6. Initial damage of acinar cells was exacerbated in these mice, manifested by increased serum levels of pancreatic enzymes, intra-pancreatic trypsinogen activation and acinar cell apoptosis. In addition, MRL/MpJ pancreata presented enhanced inflammation, de-differentiation of acinar cells and acinar-to-ductal metaplasia. Manipulation of inflammatory levels and mitogenic stimulation with the thyroid hormone 5,3-L-tri-iodothyronine revealed that factors derived from initial acinar injury rather than inflammatory injury promote the replicative advantage in MRL/MpJ mice.


Assuntos
Células Acinares/citologia , Inflamação/metabolismo , Pâncreas/metabolismo , Células Acinares/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Ceruletídeo/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pâncreas/efeitos dos fármacos , Pâncreas/imunologia
6.
Gut ; 67(9): 1663-1673, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-28774888

RESUMO

OBJECTIVE: Chronic pancreatitis (CP) and autoimmune pancreatitis (AIP) are characterised by different inflammatory processes. If pancreatic inflammation is a prerequisite for autoimmunity is still unclear. AIP is considered mostly a T cell-mediated disease; however, in induction of CP, macrophages play a pivotal role. p21-a member of cyclin-dependent kinase inhibitors-can influence inflammatory processes, in particular can regulate T cell activation and promote macrophage development. We therefore examined the role of p21-mediated inflammation in AIP. DESIGN: We intercrossed lymphotoxin (LT) overexpressing mice (Tg(Ela1-LTa,b))-a model to study AIP development-with p21-deficient mice. Furthermore, we characterised p21 expression in human AIP and non-AIP specimens. RESULTS: p21 deficiency in LT mice (LTp21-/-) prevented early pancreatic injury and reduced inflammation. In acinar cells, diminished proliferation and abrogated activation of non-canonical nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) pathway was observed. In contrast, 12-month-old LT mice with and without p21 had similar inflammatory signatures and T-B cell infiltration. Interestingly, LT and LTp21-/- mice had comparable tertiary lymphoid organs (TLOs), autoantibodies and elevated IgG levels. However, acinar cell proliferation, acinar-to-ductal metaplasia and acinar non-canonical NF-κB pathway activation remained impaired in LTp21-/- pancreata. CONCLUSIONS: Our findings indicate that p21 is crucial for pancreatic inflammation in LT-driven pancreatic injury. p21 is involved in early acinar secretion of inflammatory mediators that attract innate immune cells. However, p21 is not essential for humoral immune response, accountable for autoimmunity. Remarkably, p21 renders acinar cells less susceptible to proliferation and transdifferentiation. We therefore suggest that AIP can also develop independent of chronic inflammatory processes.


Assuntos
Doenças Autoimunes/genética , Mutagênese , Pancreatite Crônica/genética , Linfócitos T/metabolismo , Quinases Ativadas por p21/genética , Animais , Doenças Autoimunes/complicações , Biomarcadores/sangue , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Pancreatite Crônica/complicações
7.
Br J Pharmacol ; 174(21): 3865-3880, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28832971

RESUMO

BACKGROUND AND PURPOSE: Pancreatitis is a common inflammation of the pancreas with rising incidence in many countries. Despite improvements in diagnostic techniques, the disease is associated with high risk of severe morbidity and mortality and there is an urgent need for new therapeutic interventions. In this study, we evaluated whether histone deacetylases (HDACs), key epigenetic regulators of gene transcription, are involved in the development of the disease. EXPERIMENTAL APPROACH: We analysed HDAC regulation during cerulein-induced acute, chronic and autoimmune pancreatitis using different transgenic mouse models. The functional relevance of class I HDACs was tested with the selective inhibitor MS-275 in vivo upon pancreatitis induction and in vitro in activated macrophages and primary acinar cell explants. KEY RESULTS: HDAC expression and activity were up-regulated in a time-dependent manner following induction of pancreatitis, with the highest abundance observed for class I HDACs. Class I HDAC inhibition did not prevent the initial acinar cell damage. However, it effectively reduced the infiltration of inflammatory cells, including macrophages and T cells, in both acute and chronic phases of the disease, and directly disrupted macrophage activation. In addition, MS-275 treatment reduced DNA damage in acinar cells and limited acinar de-differentiation into acinar-to-ductal metaplasia in a cell-autonomous manner by impeding the EGF receptor signalling axis. CONCLUSIONS AND IMPLICATIONS: These results demonstrate that class I HDACs are critically involved in the development of acute and chronic forms of pancreatitis and suggest that blockade of class I HDAC isoforms is a promising target to improve the outcome of the disease.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/efeitos dos fármacos , Pancreatite/tratamento farmacológico , Células Acinares/metabolismo , Doença Aguda , Animais , Doenças Autoimunes/fisiopatologia , Benzamidas/farmacologia , Modelos Animais de Doenças , Receptores ErbB/metabolismo , Histona Desacetilases/metabolismo , Leucócitos/metabolismo , Masculino , Metaplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pancreatite/fisiopatologia , Pancreatite Crônica/tratamento farmacológico , Pancreatite Crônica/fisiopatologia , Piridinas/farmacologia , Fatores de Tempo
8.
Oncotarget ; 8(18): 30162-30174, 2017 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-28415799

RESUMO

INTRODUCTION: In patients with infection and sepsis serum levels of Pancreatic Stone protein/regenerating protein I (PSP) are highly elevated. The origin of PSP during these conditions is presumably the pancreas, however, an intestinal origin cannot be excluded. Similarly, pancreatitis-associated protein (PAP) was identified in the pancreas. These proteins were also localized in intestinal organs. Here we aim to elucidate the bio-distribution of PSP and PAP in animal models of sepsis and in healthy humans. RESULTS: PSP and PAP responded to remote lesions in rats although the pancreatic response was much more pronounced than the intestinal. Tissue distribution of PSP demonstrated a 100-fold higher content in the pancreas compared to any other organ while PAP was most abundant in the small intestine. Both proteins responded to CLP or sham operation in the pancreas. PSP also increased in the intestine during CLP. The distribution of PSP and PAP in human tissue mirrored the distribution in the murine models. MATERIALS AND METHODS: Distribution of PSP and PAP was visualized by immunohistochemistry. Rats and mice underwent midline laparotomies followed by mobilization of tissue and incision of the pancreatic duct or duodenum. Standard cecum-ligation-puncture (CLP) procedures or sham laparotomies were performed. Human tissue extracts were analyzed for PSP and PAP. CONCLUSIONS: The pancreas reacts to remote lesions and septic insults in mice and rats with increased PSP synthesis, while PAP is selectively responsive to septic events. Furthermore, our results suggest that serum PSP in septic patients is predominantly derived through an acute phase response of the pancreas.


Assuntos
Litostatina/metabolismo , Pâncreas/metabolismo , Proteínas Associadas a Pancreatite/metabolismo , Estresse Fisiológico , Animais , Biomarcadores , Humanos , Masculino , Camundongos , Transporte Proteico , Ratos , Sepse/sangue , Sepse/etiologia , Sepse/metabolismo
9.
J Pathol ; 238(3): 434-45, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26510396

RESUMO

Determining signalling pathways that regulate pancreatic regeneration following pancreatitis is critical for implementing therapeutic interventions. In this study we elucidated the molecular mechanisms underlying the effects of transforming growth factor-ß (TGFß) in pancreatic epithelial cells during tissue regeneration. To this end, we conditionally inactivated TGFß receptor II (TGFß-RII) using a Cre-LoxP system under the control of pancreas transcription factor 1a (PTF1a) promoter, specific for the pancreatic epithelium, and evaluated the molecular and cellular changes in a mouse model of cerulein-induced pancreatitis. We show that TGFß-RII signalling does not mediate the initial acinar cell damage observed at the onset of pancreatitis. However, TGFß-RII signalling not only restricts acinar cell replication during the regenerative phase of the disease but also limits ADM formation in vivo and in vitro in a cell-autonomous manner. Analyses of molecular mechanisms underlying the observed phenotype revealed that TGFß-RII signalling stimulates the expression of cyclin-dependent kinase inhibitors and intersects with the EGFR signalling axis. Finally, TGFß-RII ablation in epithelial cells resulted in increased infiltration of inflammatory cells in the early phases of pancreatitis and increased activation of pancreatic stellate cells in the later stages of pancreatitis, thus highlighting a TGFß-based crosstalk between epithelial and stromal cells regulating the development of pancreatic inflammation and fibrosis. Collectively, our data not only contribute to clarifying the cellular processes governing pancreatic tissue regeneration, but also emphasize the conserved role of TGFß as a tumour suppressor, both in the regenerative process following pancreatitis and in the initial phases of pancreatic cancer.


Assuntos
Células Acinares/patologia , Pâncreas/patologia , Pancreatite/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Amilases/metabolismo , Animais , Carcinoma Ductal Pancreático/patologia , Pontos de Checagem do Ciclo Celular/fisiologia , Proliferação de Células/fisiologia , Transformação Celular Neoplásica/patologia , Células Cultivadas , Ceruletídeo/toxicidade , Células Epiteliais/patologia , Fibrose/patologia , Irritantes/toxicidade , Lipase/metabolismo , Masculino , Metaplasia/patologia , Camundongos Knockout , Camundongos Transgênicos , Pâncreas/enzimologia , Neoplasias Pancreáticas/patologia , Pancreatite/enzimologia , Receptor do Fator de Crescimento Transformador beta Tipo II
10.
Kidney Int ; 89(1): 113-26, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26398497

RESUMO

Accumulation of inflammatory cells in different renal compartments is a hallmark of progressive kidney diseases including glomerulonephritis (GN). Lymphotoxin ß receptor (LTßR) signaling is crucial for the formation of lymphoid tissue, and inhibition of LTßR signaling has ameliorated several non-renal inflammatory models. Therefore, we tested whether LTßR signaling could also have a role in renal injury. Renal biopsies from patients with GN were found to express both LTα and LTß ligands, as well as LTßR. The LTßR protein and mRNA were localized to tubular epithelial cells, parietal epithelial cells, crescents, and cells of the glomerular tuft, whereas LTß was found on lymphocytes and tubular epithelial cells. Human tubular epithelial cells, mesangial cells, and mouse parietal epithelial cells expressed both LTα and LTß mRNA upon stimulation with TNF in vitro. Several chemokine mRNAs and proteins were expressed in response to LTßR signaling. Importantly, in a murine lupus model, LTßR blockade improved renal function without the reduction of serum autoantibody titers or glomerular immune complex deposition. Thus, a preclinical mouse model and human studies strongly suggest that LTßR signaling is involved in renal injury and may be a suitable therapeutic target in renal diseases.


Assuntos
Glomerulonefrite por IGA/metabolismo , Nefrite Lúpica/metabolismo , Receptor beta de Linfotoxina/antagonistas & inibidores , Receptor beta de Linfotoxina/metabolismo , RNA Mensageiro/análise , Transdução de Sinais , Adulto , Animais , Linhagem Celular , Quimiocinas/genética , Quimiocinas/metabolismo , Modelos Animais de Doenças , Células Epiteliais/química , Células Epiteliais/metabolismo , Feminino , Glomerulonefrite por IGA/genética , Humanos , Imunoglobulinas/farmacologia , Glomérulos Renais/química , Glomérulos Renais/patologia , Túbulos Renais/química , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Ligantes , Nefrite Lúpica/genética , Linfócitos/química , Receptor beta de Linfotoxina/análise , Receptor beta de Linfotoxina/genética , Linfotoxina-alfa/análise , Linfotoxina-alfa/genética , Linfotoxina-alfa/metabolismo , Linfotoxina-beta/análise , Linfotoxina-beta/genética , Linfotoxina-beta/metabolismo , Masculino , Células Mesangiais/metabolismo , Camundongos , Pessoa de Meia-Idade , Transdução de Sinais/efeitos dos fármacos , Transcriptoma
11.
J Pathol ; 237(4): 495-507, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26235267

RESUMO

The exocrine pancreas exhibits a distinctive capacity for tissue regeneration and renewal following injury. This regenerative ability has important implications for a variety of disorders, including pancreatitis and pancreatic cancer, diseases associated with high morbidity and mortality. Thus, understanding its underlying mechanisms may help in developing therapeutic interventions. Serotonin has been recognized as a potent mitogen for a variety of cells and tissues. Here we investigated whether serotonin exerts a mitogenic effect in pancreatic acinar cells in three regenerative models, inflammatory tissue injury following pancreatitis, tissue loss following partial pancreatectomy, and thyroid hormone-stimulated acinar proliferation. Genetic and pharmacological techniques were used to modulate serotonin levels in vivo. Acinar dedifferentiation and cell cycle progression during the regenerative phase were investigated over the course of 2 weeks. By comparing acinar proliferation in the different murine models of regeneration, we found that serotonin did not affect the clonal regeneration of mature acinar cells. Serotonin was, however, required for acinar dedifferentiation following inflammation-mediated tissue injury. Specifically, lack of serotonin resulted in delayed up-regulation of progenitor genes and delayed the formation of acinar-to-ductal metaplasia and defective acinar cell proliferation. We identified serotonin-dependent acinar secretion as a key step in progenitor-based regeneration, as it promoted acinar cell dedifferentiation and the recruitment of type 2 macrophages. Finally, we identified a regulatory Hes1-Ptfa axis in the uninjured adult pancreas, activated by zymogen secretion. Our findings indicated that serotonin plays a critical role in the regeneration of the adult pancreas following pancreatitis by promoting the dedifferentiation of acinar cells.


Assuntos
Células Acinares/citologia , Desdiferenciação Celular/fisiologia , Pâncreas Exócrino/fisiologia , Serotonina/metabolismo , Envelhecimento , Animais , Modelos Animais de Doenças , Immunoblotting , Imuno-Histoquímica , Metaplasia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Pancreatite/patologia , Regeneração
12.
J Pathol ; 235(3): 502-14, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25212177

RESUMO

Trans-differentiation of pancreatic acinar cells into ductal-like lesions, a process defined as acinar-to-ductal metaplasia (ADM), is observed in the course of organ regeneration following pancreatitis. In addition, ADM is found in association with pre-malignant PanIN lesions and correlates with an increased risk of pancreatic adenocarcinoma (PDAC). Human PDAC samples show down-regulation of p21(WAF1) (/Cip1) , a key regulator of cell cycle and cell differentiation. Here we investigated whether p21 down-regulation is implicated in controlling the early events of acinar cell trans-differentiation and ADM formation. p21-mediated regulation of ADM formation and regression was analysed in vivo during the course of cerulein-induced pancreatitis, using wild-type (WT) and p21-deficient (p21(-/-) ) mice. Biochemical and immunohistochemical methods were used to evaluate disease progression over 2 weeks of the disease and during a recovery phase. We found that p21 was strongly up-regulated in WT acinar cells during pancreatitis, while it was absent in ADM areas, suggesting that p21 down-regulation is associated with ADM formation. In support of this hypothesis, p21(-/-) mice showed a significant increase in number and size of metaplasia. In addition, p21 over-expression in acinar cells reduced ADM formation in vitro, suggesting that the protein regulates the metaplastic transition in a cell-autonomous manner. p21(-/-) mice displayed increased expression and relocalization of ß-catenin both during pancreatitis and in the subsequent recovery phase. Finally, loss of p21 was accompanied by increased DNA damage and development of senescence. Our findings are consistent with a gate-keeper role of p21 in acinar cells to limit senescence activation and ADM formation during pancreatic regeneration.


Assuntos
Diferenciação Celular/fisiologia , Senescência Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Pâncreas/patologia , Pâncreas/fisiopatologia , Pancreatite/patologia , Pancreatite/fisiopatologia , Animais , Ciclo Celular/fisiologia , Proliferação de Células/fisiologia , Ceruletídeo/efeitos adversos , Inibidor de Quinase Dependente de Ciclina p21/deficiência , Inibidor de Quinase Dependente de Ciclina p21/genética , Modelos Animais de Doenças , Regulação para Baixo/fisiologia , Técnicas In Vitro , Metaplasia , Camundongos , Camundongos Knockout , Pancreatite/induzido quimicamente , Regeneração/fisiologia , beta Catenina/fisiologia
13.
Cytokine Growth Factor Rev ; 25(2): 125-37, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24508087

RESUMO

The pathogenic mechanisms of autoimmune pancreatitis (AIP), an increasingly recognized, immune-mediated form of chronic pancreatitis, have so far remained elusive. Treatment options for AIP are currently limited and disease relapse is frequent. Still, AIP can be characterized by specific clinical and histologic features. It has turned out that as described in other autoimmune diseases the generation of tertiary lymphoid organs is also a hallmark of patients with AIP. We have recently demonstrated that pancreata derived from human AIP patients display overexpression of lymphotoxin (LT) α and ß and LTßR-target genes expressed by immune cells but also by irradiation resistant cells of the pancreas (e.g. acinar cells). Expression of LT α and ß on acinar cells in murine pancreata Tg(Ela1-Lta,b) mice led to chronic pancreatitis and sufficed to reproduce key features of human AIP including the development of autoimmunity and AIP associated secondary extra pancreatic pathologies. Here, we review how aberrant and ectopic expression of LT α and ß can induce inflammation and autoimmune diseases in general and how this knowledge might specifically lead to an alternative treatment for patients suffering from autoimmune pancreatitis.


Assuntos
Doenças Autoimunes/imunologia , Linfotoxina-alfa/imunologia , Pâncreas/patologia , Pancreatite/imunologia , Animais , Artrite Reumatoide/imunologia , Humanos , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/imunologia , Linfotoxina-alfa/biossíntese , Camundongos , Esclerose Múltipla/imunologia , Pâncreas/imunologia , Transdução de Sinais/imunologia , Síndrome de Sjogren/imunologia
15.
Science ; 342(6163): 1243-6, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24311691

RESUMO

Immunoglobulin A (IgA) production at mucosal surfaces contributes to protection against pathogens and controls intestinal microbiota composition. However, mechanisms regulating IgA induction are not completely defined. We show that soluble lymphotoxin α (sLTα3) produced by RORγt(+) innate lymphoid cells (ILCs) controls T cell-dependent IgA induction in the lamina propria via regulation of T cell homing to the gut. By contrast, membrane-bound lymphotoxin ß (LTα1ß2) produced by RORγt(+) ILCs is critical for T cell-independent IgA induction in the lamina propria via control of dendritic cell functions. Ablation of LTα in RORγt(+) cells abrogated IgA production in the gut and altered microbiota composition. Thus, soluble and membrane-bound lymphotoxins produced by ILCs distinctly organize adaptive immune responses in the gut and control commensal microbiota composition.


Assuntos
Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestino Delgado/imunologia , Subpopulações de Linfócitos/imunologia , Linfotoxina-alfa/imunologia , Microbiota/fisiologia , Imunidade Adaptativa , Animais , Linfócitos B/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Homeostase , Imunidade Inata , Imunoglobulina A/biossíntese , Switching de Imunoglobulina , Intestino Delgado/microbiologia , Linfonodos/imunologia , Subpopulações de Linfócitos/metabolismo , Linfotoxina-alfa/metabolismo , Linfotoxina-beta/imunologia , Linfotoxina-beta/metabolismo , Camundongos , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
16.
Gastroenterology ; 143(5): 1361-1374, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22863765

RESUMO

BACKGROUND & AIMS: Little is known about the pathogenic mechanisms of autoimmune pancreatitis (AIP), an increasingly recognized, immune-mediated form of chronic pancreatitis. Current treatment options are limited and disease relapse is frequent. We investigated factors that contribute to the development of AIP and new therapeutic strategies. METHODS: We used quantitative polymerase chain reaction, immunohistochemical, and enzyme-linked immunosorbent analyses to measure the expression of cytokines and chemokines in tissue and serum samples from patients with and without AIP. We created a mouse model of human AIP by overexpressing lymphotoxin (LT)α and ß specifically in acinar cells (Ela1-LTab mice). RESULTS: Messenger RNA levels of LTα and ß were increased in pancreatic tissues from patients with AIP, compared with controls, and expression of chemokines (CXCL13, CCL19, CCL21, CCL1, and B-cell-activating factor) was increased in pancreatic and serum samples from patients. Up-regulation of these factors was not affected by corticosteroid treatment. Acinar-specific overexpression of LTαß (Ela1-LTαß) in mice led to an autoimmune disorder with various features of AIP. Chronic inflammation developed only in the pancreas but was sufficient to cause systemic autoimmunity. Acinar-specific overexpression of LTαß did not cause autoimmunity in mice without lymphocytes (Ela1-LTab/Rag1(-/-)); moreover, lack of proinflammatory monocytes (Ela1-LTab/Ccr2(-/-)) failed to prevent AIP but prevented early pancreatic tissue damage. Administration of corticosteroids reduced pancreatitis but did not affect production of autoantibodies, such as antipancreatic secretory trypsin inhibitor in Ela1-LTab mice. In contrast, inhibition of LTßR signaling reduced chemokine expression, renal immune-complex deposition, and features of AIP in Ela1-LTab mice. CONCLUSIONS: Overexpression of LTαß specifically in acinar cells of mice causes features of AIP. Reagents that neutralize LTßR ligands might be used to treat patients with AIP.


Assuntos
Doenças Autoimunes/metabolismo , Receptor beta de Linfotoxina/metabolismo , Pancreatite Crônica/imunologia , Pancreatite Crônica/metabolismo , Transdução de Sinais , Células Acinares/metabolismo , Corticosteroides/farmacologia , Corticosteroides/uso terapêutico , Análise de Variância , Animais , Autoanticorpos/sangue , Doenças Autoimunes/sangue , Doenças Autoimunes/tratamento farmacológico , Estudos de Casos e Controles , Células Cultivadas , Quimiocinas/efeitos dos fármacos , Quimiocinas/metabolismo , Modelos Animais de Doenças , Glomerulonefrite/imunologia , Glomerulonefrite/patologia , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Contagem de Linfócitos , Receptor beta de Linfotoxina/sangue , Linfotoxina-alfa/efeitos dos fármacos , Linfotoxina-alfa/genética , Linfotoxina-alfa/metabolismo , Linfotoxina-beta/efeitos dos fármacos , Linfotoxina-beta/genética , Linfotoxina-beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Elastase Pancreática/genética , Elastase Pancreática/metabolismo , Pancreatite Crônica/sangue , Pancreatite Crônica/tratamento farmacológico , Regiões Promotoras Genéticas , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Estatísticas não Paramétricas , Subpopulações de Linfócitos T , Regulação para Cima
18.
Cancer Cell ; 17(5): 481-96, 2010 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-20478530

RESUMO

The MAP3-kinase TGF-beta-activated kinase 1 (TAK1) critically modulates innate and adaptive immune responses and connects cytokine stimulation with activation of inflammatory signaling pathways. Here, we report that conditional ablation of TAK1 in liver parenchymal cells (hepatocytes and cholangiocytes) causes hepatocyte dysplasia and early-onset hepatocarcinogenesis, coinciding with biliary ductopenia and cholestasis. TAK1-mediated cancer suppression is exerted through activating NF-kappaB in response to tumor necrosis factor (TNF) and through preventing Caspase-3-dependent hepatocyte and cholangiocyte apoptosis. Moreover, TAK1 suppresses a procarcinogenic and pronecrotic pathway, which depends on NF-kappaB-independent functions of the I kappaB-kinase (IKK)-subunit NF-kappaB essential modulator (NEMO). Therefore, TAK1 serves as a gatekeeper for a protumorigenic, NF-kappaB-independent function of NEMO in parenchymal liver cells.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , MAP Quinase Quinase Quinases/fisiologia , NF-kappa B/metabolismo , Animais , Apoptose , Transformação Celular Neoplásica , Hiperplasia , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Camundongos Transgênicos , Necrose
19.
Cell Transplant ; 19(8): 937-48, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20350355

RESUMO

The therapeutic mechanism of mesenchymal stromal/stem cells (MSC) for the treatment of acute myocardial infarction is not well understood. Our goal was to get insights into this mechanism by analyzing the survival kinetics of allogeneic and syngeneic cell transplants under different tissue conditions. Two MSC cell banks, stably and equally expressing the luciferase reporter construct, were developed for these studies and injected directly to the myocardium of Lewis rat recipients under syngeneic or allogeneic transplantation conditions. Cell survival was monitored by real-time fashion for up to 2 weeks, using optical imaging device (IVIS, Xenogen Corp.). We found that both syngeneic and allogeneic grafts reduced significantly in size during the first week of transplantation, either in the normal or in the late infarcted heart (5 days after MI) and allotransplants became always smaller than syngeneic grafts during this period. Low dose of cyclosporine A treatment had a benefit on both allo- and syngeneic graft sizes, suggesting that multiple mechanisms play a role in early graft reduction. The MSC characteristic factors IL-6, IL-8, MCP-1, and VEGF were well above the control level in the heart tissue at 4 days after cell injection, suggesting that the peak therapeutic effect of MSC can be expected during the first week of the administration. Although allogeneic cells induced immunoglobulin production, their biological effects (cell survival, factor productions) are very similar to the syngeneic transplants and therefore they could deliver the same therapeutic effect as the syngeneic cells. Finally, freshly infarcted tissue (30 min) supported better the survival of MSC than late postischemic tissue (5 days) but only "off the shelf" allogeneic cell transplants fits with this treatment strategy.


Assuntos
Transplante de Células-Tronco Mesenquimais , Miocárdio/citologia , Animais , Sobrevivência Celular , Quimiocina CCL2/metabolismo , Ciclosporina/farmacologia , Genes Reporter , Imageamento Tridimensional , Injeções , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Masculino , Infarto do Miocárdio/terapia , Miocárdio/metabolismo , Ratos , Ratos Endogâmicos Lew , Fatores de Tempo , Transplante Homólogo , Transplante Isogênico , Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...