Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.489
Filtrar
1.
World J Gastroenterol ; 30(23): 2927-2930, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38946872

RESUMEN

In this editorial, we focus specifically on the mechanisms by which pancreatic inflammation affects pancreatic cancer. Cancer of the pancreas remains one of the deadliest cancer types. The highest incidence and mortality rates of pancreatic cancer are found in developed countries. Trends of pancreatic cancer incidence and mortality vary considerably worldwide. A better understanding of the etiology and identification of the risk factors is essential for the primary prevention of this disease. Pancreatic tumors are characterized by a complex microenvironment that orchestrates metabolic alterations and supports a milieu of interactions among various cell types within this niche. In this editorial, we highlight the foundational studies that have driven our understanding of these processes. In our experimental center, we have carefully studied the mechanisms of that link pancreatic inflammation and pancreatic cancer. We focused on the role of mast cells (MCs). MCs contain pro-angiogenic factors, including tryptase, that are associated with increased angiogenesis in various tumors. In this editorial, we address the role of MCs in angiogenesis in both pancreatic ductal adenocarcinoma tissue and adjacent normal tissue. The assessment includes the density of c-Kit receptor-positive MCs, the density of tryptase-positive MCs, the area of tryptase-positive MCs, and angiogenesis in terms of microvascularization density.


Asunto(s)
Mastocitos , Neovascularización Patológica , Neoplasias Pancreáticas , Microambiente Tumoral , Humanos , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/inmunología , Mastocitos/metabolismo , Mastocitos/inmunología , Microambiente Tumoral/inmunología , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/metabolismo , Páncreas/patología , Páncreas/inmunología , Páncreas/metabolismo , Animales , Pancreatitis/metabolismo , Pancreatitis/patología , Pancreatitis/inmunología , Factores de Riesgo , Mediadores de Inflamación/metabolismo , Triptasas/metabolismo , Inflamación/metabolismo
3.
Immun Inflamm Dis ; 12(7): e1351, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39023414

RESUMEN

BACKGROUND: Severe acute pancreatitis (SAP) is a potentially lethal inflammatory pancreatitis condition that is usually linked to multiple organ failure. When it comes to SAP, the lung is the main organ that is frequently involved. Many SAP patients experience respiratory failure following an acute lung injury (ALI). Clinicians provide insufficient care for compounded ALI since the underlying pathophysiology is unknown. The mortality rate of SAP patients is severely impacted by it. OBJECTIVE: The study aims to provide insight into immune cells, specifically their roles and modifications during SAP and ALI, through a comprehensive literature review. The emphasis is on immune cells as a therapeutic approach for treating SAP and ALI. FINDINGS: Immune cells play an important role in the complicated pathophysiology ofSAP and ALI by maintaining the right balance of pro- and anti-inflammatory responses. Immunomodulatory drugs now in the market have low thepeutic efficacy because they selectively target one immune cell while ignoring immune cell interactions. Accurate management of dysregulated immune responses is necessary. A critical initial step is precisely characterizing the activity of the immune cells during SAP and ALI. CONCLUSION: Given the increasing incidence of SAP, immunotherapy is emerging as a potential treatment option for these patients. Interactions among immune cells improve our understanding of the intricacy of concurrent ALI in SAP patients. Acquiring expertise in these domains will stimulate the development of innovative immunomodulation therapies that will improve the outlook for patients with SAP and ALI.


Asunto(s)
Lesión Pulmonar Aguda , Pancreatitis , Humanos , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/etiología , Pancreatitis/inmunología , Pancreatitis/terapia , Pancreatitis/patología , Pancreatitis/complicaciones , Páncreas/inmunología , Páncreas/patología , Pulmón/inmunología , Pulmón/patología , Animales , Inmunoterapia/métodos
4.
Int J Mol Sci ; 25(11)2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38891952

RESUMEN

The pancreas is an organ with both exocrine and endocrine functions, comprising a highly organized and complex tissue microenvironment composed of diverse cellular and non-cellular components. The impairment of microenvironmental homeostasis, mediated by the dysregulation of cell-to-cell crosstalk, can lead to pancreatic diseases such as pancreatitis, diabetes, and pancreatic cancer. Macrophages, key immune effector cells, can dynamically modulate their polarization status between pro-inflammatory (M1) and anti-inflammatory (M2) modes, critically influencing the homeostasis of the pancreatic microenvironment and thus playing a pivotal role in the pathogenesis of the pancreatic disease. This review aims to summarize current findings and provide detailed mechanistic insights into how alterations mediated by macrophage polarization contribute to the pathogenesis of pancreatic disorders. By analyzing current research comprehensively, this article endeavors to deepen our mechanistic understanding of regulatory molecules that affect macrophage polarity and the intricate crosstalk that regulates pancreatic function within the microenvironment, thereby facilitating the development of innovative therapeutic strategies that target perturbations in the pancreatic microenvironment.


Asunto(s)
Macrófagos , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Animales , Enfermedades Pancreáticas/patología , Enfermedades Pancreáticas/inmunología , Enfermedades Pancreáticas/metabolismo , Microambiente Celular/inmunología , Páncreas/inmunología , Páncreas/patología , Páncreas/metabolismo , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Activación de Macrófagos/inmunología
5.
Nat Commun ; 15(1): 4971, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38871688

RESUMEN

Human type 1 diabetes (T1D) is caused by autoimmune attack on the insulin-producing pancreatic beta cells by islet antigen-reactive T cells. How human islet antigen-reactive (IAR) CD4+ memory T cells from peripheral blood affect T1D progression in the pancreas is poorly understood. Here, we aim to determine if IAR T cells in blood could be detected in pancreas. We identify paired αß (TRA/TRB) T cell receptors (TCRs) in IAR T cells from the blood of healthy, at-risk, new-onset, and established T1D donors, and measured sequence overlap with TCRs in pancreata from healthy, at risk and T1D organ donors. We report extensive TRA junction sharing between IAR T cells and pancreas-infiltrating T cells (PIT), with perfect-match or single-mismatch TRA junction amino acid sequences comprising ~29% total unique IAR TRA junctions (942/3,264). PIT-matched TRA junctions were largely public and enriched for TRAV41 usage, showing significant nucleotide sequence convergence, increased use of germline-encoded versus non-templated residues in epitope engagement, and a potential for cross-reactivity. Our findings thus link T cells with distinctive germline-like TRA chains in the peripheral blood with T cells in the pancreas.


Asunto(s)
Diabetes Mellitus Tipo 1 , Páncreas , Receptores de Antígenos de Linfocitos T alfa-beta , Humanos , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/sangre , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Páncreas/inmunología , Masculino , Femenino , Adulto , Linfocitos T CD4-Positivos/inmunología , Linfocitos T/inmunología , Células Germinativas/inmunología , Células Germinativas/metabolismo , Autoantígenos/inmunología
6.
J Clin Lab Anal ; 38(10): e25076, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38853390

RESUMEN

BACKGROUND: Severe acute pancreatitis (SAP) is associated with tremendous systemic inflammation, T-helper 17 (Th17) cells, and regulatory T (Treg) cells play an essential role in the inflammatory responses. Meanwhile, soluble fibrinogen-like protein 2 (Sfgl2) is a critical immunosuppressive effector cytokine of Treg cells and modulates immune responses. However, the impact of SAP induction on Sfgl2 expression and the role of Sfgl2 in immunomodulation under SAP conditions are largely unknown. METHODS: A taurocholate-induced mouse SAP model was established. The ratios of CD4+CD25+Foxp3+ Treg cells or CD4+IL-17+ Th17 cells in blood and pancreatic tissues as well as surface expression of CD80, CD86, and major histocompatibility complex class II (MHC-II) were determined by flow cytometry. Gene mRNA expression was determined by qPCR. Serum amylase and soluble factors were quantitated by commercial kits. Bone marrow-derived dendritic cells (DCs) were generated, and NF-κB/p65 translocation was measured by immunofluorescence staining. RESULTS: SAP induction in mice decreased the Th17/Treg ratio in the pancreatic tissue and increased the Th17/Treg ratio in the peripheral blood. In addition, SAP was associated with a reduced level of Sfgl2 in the pancreatic tissue and blood: higher levels of serum IL-17, IL-2, IFN-α, and TNF-α, and lower levels of serum IL-4 and IL-10. Furthermore, the SAP-induced reduction in Sfgl2 expression was accompanied by dysregulated maturation of bone marrow-derived DCs. CONCLUSIONS: SAP causes reduced Sfgl2 expression and Th17/Treg imbalance, thus providing critical insights for the development of Sfgl2- and Th17/Treg balance-targeted immunotherapies for patients with SAP.


Asunto(s)
Modelos Animales de Enfermedad , Fibrinógeno , Pancreatitis , Linfocitos T Reguladores , Ácido Taurocólico , Células Th17 , Animales , Células Th17/inmunología , Linfocitos T Reguladores/inmunología , Pancreatitis/inmunología , Pancreatitis/inducido químicamente , Pancreatitis/metabolismo , Ratones , Fibrinógeno/metabolismo , Masculino , Ratones Endogámicos C57BL , Regulación hacia Abajo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Enfermedad Aguda , Páncreas/inmunología , Páncreas/patología , Páncreas/metabolismo
7.
Int Immunopharmacol ; 136: 112284, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38823179

RESUMEN

Cathepsin B (CTSB) and inflammatory cytokines are critical in initiating and developing pancreatitis. Calcineurin, a central calcium (Ca2+)-responsive signaling molecule, mediates acinar cell death and inflammatory responses leading to pancreatitis. However, the detailed mechanisms for regulating CTSB activity and inflammatory cytokine production are unknown. Myricetin (MC) exhibits various biological activities, including anti-inflammatory effects. Here, we aimed to investigate MC effects on pancreatitis and the underlying mechanisms. Prophylactic and therapeutic MC treatment ameliorated the severity of cerulein-, L-arginine-, and PDL-induced acute pancreatitis (AP). The inhibition of CTSB activity by MC was mediated via decreased calcineurin activity and macrophage infiltration, not neutrophils infiltration, into the pancreas. Additionally, calcineurin activity inhibition by MC prevented the phosphorylation of Ca2+/CaM-dependent protein kinase kinase 2 (CaMKK2) during AP, resulting in the inhibition of CaMKIV phosphorylation and adenosine monophosphate-activated protein kinase (AMPK) dephosphorylation. Furthermore, MC reduced nuclear factor-κB activation by modulating the calcineurin-CaMKIV-IKKα/ß-Iκ-Bα and calcineurin-AMPK-sirtuin1 axes, resulting in reduced production of tumor necrosis factor-α, interleukin (IL)-1ß, and IL-6. Our results showed that MC alleviated AP severity by inhibiting acinar cell death and inflammatory responses, suggesting that MC as a calcineurin and CaMKK2 signaling modulator may be a potential treatment for AP.


Asunto(s)
Calcineurina , Catepsina B , Citocinas , Flavonoides , Ratones Endogámicos C57BL , Pancreatitis , Animales , Pancreatitis/tratamiento farmacológico , Pancreatitis/inmunología , Pancreatitis/patología , Pancreatitis/inducido químicamente , Flavonoides/farmacología , Flavonoides/uso terapéutico , Citocinas/metabolismo , Catepsina B/metabolismo , Ratones , Masculino , Calcineurina/metabolismo , Antiinflamatorios/uso terapéutico , Antiinflamatorios/farmacología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Ceruletida , FN-kappa B/metabolismo , Páncreas/patología , Páncreas/efectos de los fármacos , Páncreas/inmunología , Transducción de Señal/efectos de los fármacos , Arginina/metabolismo , Modelos Animales de Enfermedad , Proteínas Quinasas Activadas por AMP/metabolismo
8.
Pancreas ; 53(7): e617-e626, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38696351

RESUMEN

BACKGROUND: Although the involvement of intestinal microbiota in innate immunity has been reported recently, the pathogenicity of autoimmune pancreatitis (AIP) remains unclear. This study aimed to investigate whether probiotics ameliorate inflammation in AIP through interactions with innate immunity. MATERIALS AND METHODS: The AIP mouse model was generated by intraperitoneal administration of Escherichia coli to C56BL/6 female mice. Alterations in the intestinal microbiota in the AIP group were evaluated using high-throughput sequencing. Peritoneal macrophages (PMs) were collected and cocultured in vitro with Lactobacillus gasseri (LG) or ligands of Toll-like receptors (TLRs). LG was administered intraperitoneally to AIP model mice, and pancreatitis activity was evaluated to examine the ameliorative effects of LG. RESULTS: In the AIP model mice, inflammation was significantly induced in the pancreas, and the intestinal microbiota was altered with decreased LG. Antimicrobial treatment suppressed pancreatitis. In vitro, E. coli stimulation increased inflammatory cytokine expression, which was significantly decreased when the LG or TLR7 ligand was cocultured with PMs. Intraperitoneal administration of LG to AIP model mice significantly suppressed pancreatitis. CONCLUSION: The mouse model demonstrated the involvement of intestinal microbiota in pancreatitis, and LG administration suppressed pancreatitis, possibly through TLR7 signaling in PMs. LG may be a helpful probiotic for treating AIP.


Asunto(s)
Pancreatitis Autoinmune , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Inmunidad Innata , Macrófagos Peritoneales , Ratones Endogámicos C57BL , Probióticos , Animales , Microbioma Gastrointestinal/inmunología , Probióticos/administración & dosificación , Probióticos/farmacología , Femenino , Pancreatitis Autoinmune/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/microbiología , Citocinas/metabolismo , Ratones , Escherichia coli/inmunología , Receptor Toll-Like 7/inmunología , Páncreas/inmunología , Páncreas/patología , Lactobacillus/inmunología
9.
World J Gastroenterol ; 30(15): 2091-2095, 2024 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-38681985

RESUMEN

In this editorial we comment on the article by Jaber et al. Autoimmune pancreatitis (AIP) represents a distinct form of pancreatitis, categorized into AIP-1 and AIP-2, characterized by obstructive jaundice, lymphoplasmacytic infiltrate, and fibrosis. AIP-1, associated with elevated immunoglobulin G4 (IgG4) levels, exhibits higher relapse rates, affecting older males, while AIP-2 is less common and linked to inflammatory bowel disease. AIP is considered a manifestation of IgG4-related systemic disease, sharing characteristic histological findings. Steroids are the primary treatment, with emerging biomarkers like interferon alpha and interleukin-33. AIP poses an increased risk of various malignancies, and the association with pancreatic cancer is debated. Surgery is reserved for severe cases, necessitating careful evaluation due to diagnostic challenges. AIP patients may have concurrent PanINs but display favorable long-term outcomes compared to pancreatic cancer patients. Thorough diagnostic assessment, including biopsy and steroid response, is crucial for informed surgical decisions in AIP.


Asunto(s)
Pancreatitis Autoinmune , Inmunoglobulina G , Neoplasias Pancreáticas , Humanos , Pancreatitis Autoinmune/diagnóstico , Pancreatitis Autoinmune/inmunología , Pancreatitis Autoinmune/terapia , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Páncreas/patología , Páncreas/inmunología , Páncreas/cirugía , Biomarcadores/sangre , Biopsia , Masculino , Esteroides/uso terapéutico , Resultado del Tratamiento
10.
Int Immunopharmacol ; 133: 112081, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38652963

RESUMEN

Acute pancreatitis (AP) is a prevalent gastrointestinal disorder. The immune response plays a crucial role in AP progression. However, the impact of immune regulatory checkpoint PD-L1 on severe acute pancreatitis (SAP) remains uncertain. Hence, this study aimed to examine the influence of PD-L1 on SAP. We assessed PD-L1 expression in neutrophils and monocytes obtained from SAP patients. We induced SAP in C57BL/6J mice, PD-L1 gene-deficient mice, and PD-L1 humanized mice using intraperitoneal injections of cerulein plus lipopolysaccharide. Prior to the initial cerulein injection, a PD-L1 inhibitor was administered. Pancreatic tissues were collected for morphological and immunohistochemical evaluation, and serum levels of amylase, lipase, and cytokines were measured. Flow cytometry analysis was performed using peripheral blood cells. The expression of PD-L1 in neutrophils and monocytes was significantly higher in SAP patients compared to healthy individuals. Likewise, the expression of PD-L1 in inflammatory cells in the peripheral blood of SAP-induced C57BL/6J mice was notably higher than in the control group. In mice with PD-L1 deficiency, SAP model exhibited lower pancreatic pathology scores, amylase, lipase, and cytokine levels compared to wild-type mice. PD-L1 deletion resulted in reduced neutrophil apoptosis, leading to an earlier peak in neutrophil apoptosis. Furthermore, it decreased early monocyte apoptosis and diminished the peak of T lymphocyte apoptosis. Within the SAP model, administration of a PD-L1 inhibitor reduced pancreatic pathology scores, amylase, lipase, and cytokine levels in both C57BL/6J mice and PD-L1 humanized mice. These findings suggest that inhibiting PD-L1 expression can alleviate the severity of SAP.


Asunto(s)
Apoptosis , Antígeno B7-H1 , Monocitos , Neutrófilos , Páncreas , Pancreatitis , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Amilasas/sangre , Apoptosis/efectos de los fármacos , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Ceruletida , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Lipasa/sangre , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/efectos de los fármacos , Páncreas/efectos de los fármacos , Páncreas/inmunología , Páncreas/patología , Pancreatitis/inmunología , Pancreatitis/inducido químicamente , Pancreatitis/tratamiento farmacológico , Pancreatitis/patología
11.
Acta Pharmacol Sin ; 45(7): 1466-1476, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38514862

RESUMEN

Disturbances in intestinal immune homeostasis predispose susceptible individuals to type 1 diabetes (T1D). G-protein-coupled receptor 41 (GPR41) is a receptor for short-chain fatty acids (SCFAs) mainly produced by gut microbiota, which plays key roles in maintaining intestinal homeostasis. In this study, we investigated the role of GPR41 in the progression of T1D. In non-obese diabetic (NOD) mice, we found that aberrant reduction of GPR41 expression in the pancreas and colons was associated with the development of T1D. GPR41-deficient (Gpr41-/-) mice displayed significantly exacerbated streptozotocin (STZ)-induced T1D compared to wild-type mice. Furthermore, Gpr41-/- mice showed enhanced gut immune dysregulation and increased migration of gut-primed IFN-γ+ T cells to the pancreas. In bone marrow-derived dendritic cells from Gpr41-/- mice, the expression of suppressor of cytokine signaling 3 (SOCS) was significantly inhibited, while the phosphorylation of STAT3 was significantly increased, thus promoting dendritic cell (DC) maturation. Furthermore, adoptive transfer of bone marrow-derived dendritic cells (BMDC) from Gpr41-/- mice accelerated T1D in irradiated NOD mice. We conclude that GPR41 is essential for maintaining intestinal and pancreatic immune homeostasis and acts as a negative regulator of DC maturation in T1D. GPR41 may be a potential therapeutic target for T1D.


Asunto(s)
Células Dendríticas , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Ratones Endogámicos NOD , Ratones Noqueados , Receptores Acoplados a Proteínas G , Estreptozocina , Animales , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ratones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/inmunología , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Interferón gamma/metabolismo , Páncreas/metabolismo , Páncreas/patología , Páncreas/inmunología , Masculino , Femenino , Microbioma Gastrointestinal
12.
BMC Microbiol ; 22(1): 8, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34983374

RESUMEN

BACKGROUND: Type 1 diabetes (T1D) is an autoimmune disease that is increasing in prevalence worldwide. One of the contributing factors to the pathogenesis of T1D is the composition of the intestinal microbiota, as has been demonstrated. in T1D patients, with some studies demonstrating a deficiency in their levels of Prevotella. We have isolated a strain of Prevotella histicola from a duodenal biopsy that has anti-inflammatory properties, and in addition, alters the development of autoimmune diseases in mouse models. Therefore, our hypothesis is that the oral administration of P. histicola might delay the development of T1D in the non-obese diabetic (NOD) mice. To assess this, we used the following materials and methods. Female NOD mice (ages 5-8 weeks) were administered every other day P. histicola that was cultured in-house. Blood glucose levels were measured every other week. Mice were sacrificed at various time points for histopathological analysis of the pancreas. Modulation of immune response by the commensal was tested by analyzing regulatory T-cells and NKp46+ cells using flow cytometry and intestinal cytokine mRNA transcript levels using quantitative RT-PCR. For microbial composition, 16 s rRNA gene analysis was conducted on stool samples collected at various time points. RESULTS: Administration of P. histicola in NOD mice delayed the onset of T1D. Beta diversity in the fecal microbiomes demonstrated that the microbial composition of the mice administered P. histicola was different from those that were not treated. Treatment with P. histicola led to a significant increase in regulatory T cells with a concomitant decrease in NKp46+ cells in the pancreatic lymph nodes as compared to the untreated group after 5 weeks of treatment. CONCLUSIONS: These observations suggest that P. histicola treatment delayed onset of diabetes by increasing the levels of regulatory T cells in the pancreatic lymph nodes. This preliminary work supports the rationale that enteral exposure to a non pathogenic commensal P. histicola be tested as a future therapy for T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/dietoterapia , Microbioma Gastrointestinal/fisiología , Prevotella/fisiología , Probióticos/administración & dosificación , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Citocinas/genética , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/microbiología , Duodeno/inmunología , Duodeno/microbiología , Heces/microbiología , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Páncreas/inmunología , Páncreas/patología
13.
JCI Insight ; 7(2)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35076023

RESUMEN

Platelet-neutrophil aggregates (PNAs) facilitate neutrophil activation and migration and could underpin the recruitment of neutrophils to the pancreas during type 1 diabetes (T1D) pathogenesis. PNAs, measured by flow cytometry, were significantly elevated in the circulation of autoantibody-positive (Aab+) children and new-onset T1D children, as well as in pre-T1D (at 4 weeks and 10-12 weeks) and T1D-onset NOD mice, compared with relevant controls, and PNAs were characterized by activated P-selectin+ platelets. PNAs were similarly increased in pre-T1D and T1D-onset NOD isolated islets/insulitis, and immunofluorescence staining revealed increased islet-associated neutrophil extracellular trap (NET) products (myeloperoxidase [MPO] and citrullinated histones [CitH3]) in NOD pancreata. In vitro, cell-free histones and NETs induced islet cell damage, which was prevented by the small polyanionic drug methyl cellobiose sulfate (mCBS) that binds to histones and neutralizes their pathological effects. Elevated circulating PNAs could, therefore, act as an innate immune and pathogenic biomarker of T1D autoimmunity. Platelet hyperreactivity within PNAs appears to represent a previously unrecognized hematological abnormality that precedes T1D onset. In summary, PNAs could contribute to the pathogenesis of T1D and potentially function as a pre-T1D diagnostic.


Asunto(s)
Plaquetas/inmunología , Agregación Celular/inmunología , Diabetes Mellitus Tipo 1 , Trampas Extracelulares , Neutrófilos/inmunología , Páncreas , Animales , Autoanticuerpos/sangre , Niño , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/inmunología , Diagnóstico Precoz , Trampas Extracelulares/diagnóstico por imagen , Trampas Extracelulares/inmunología , Femenino , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Activación Neutrófila/inmunología , Selectina-P/metabolismo , Páncreas/inmunología , Páncreas/patología
14.
JCI Insight ; 7(2)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-34847076

RESUMEN

Despite decades of research, there is no specific therapy for acute pancreatitis (AP). In the current study, we have evaluated the efficacy of pirfenidone, an antiinflammatory and antifibrotic agent that is approved by the FDA for treatment of idiopathic pulmonary fibrosis (IPF), in ameliorating local and systemic injury in AP. Our results suggest that treatment with pirfenidone in therapeutic settings (e.g., after initiation of injury), even when administered at the peak of injury, reduces severity of local and systemic injury and inflammation in multiple models of AP. In vitro evaluation suggests that pirfenidone decreases cytokine release from acini and macrophages and disrupts acinar-macrophage crosstalk. Therapeutic pirfenidone treatment increases IL-10 secretion from macrophages preceding changes in histology and modulates the immune phenotype of inflammatory cells with decreased levels of inflammatory cytokines. Antibody-mediated IL-10 depletion, use of IL-10-KO mice, and macrophage depletion experiments confirmed the role of IL-10 and macrophages in its mechanism of action, as pirfenidone was unable to reduce severity of AP in these scenarios. Since pirfenidone is FDA approved for IPF, a trial evaluating the efficacy of pirfenidone in patients with moderate to severe AP can be initiated expeditiously.


Asunto(s)
Células Acinares/metabolismo , Fibrosis , Interleucina-10/inmunología , Macrófagos/metabolismo , Páncreas , Pancreatitis , Piridonas/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Células Cultivadas , Citocinas/clasificación , Citocinas/inmunología , Modelos Animales de Enfermedad , Fibrosis/etiología , Fibrosis/prevención & control , Ratones , Páncreas/efectos de los fármacos , Páncreas/inmunología , Páncreas/lesiones , Páncreas/patología , Pancreatitis/tratamiento farmacológico , Pancreatitis/inmunología , Comunicación Paracrina/inmunología , Transducción de Señal/inmunología
16.
J Endocrinol ; 252(2): R41-R57, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34755679

RESUMEN

We review the current knowledge of pancreas pathology in type 1 diabetes. During the last two decades, dedicated efforts toward the recovery of pancreas from deceased patients with type 1 diabetes have promoted significant advances in the characterization of the pathological changes associated with this condition. The implementation of autoantibody screening among organ donors has also allowed examining pancreas pathology in the absence of clinical disease, but in the presence of serological markers of autoimmunity. The assessment of key features of pancreas pathology across various disease stages allows driving parallels with clinical disease stages. The main pathological abnormalities observed in the pancreas with type 1 diabetes are beta-cell loss and insulitis; more recently, hyperexpression of HLA class I and class II molecules have been reproduced and validated. Additionally, there are changes affecting extracellular matrix components, evidence of viral infections, inflammation, and ER stress, which could contribute to beta-cell dysfunction and the stimulation of apoptosis and autoimmunity. The increasing appreciation that beta-cell loss can be less severe at diagnosis than previously estimated, the coexistence of beta-cell dysfunction, and the persistence of key features of pancreas pathology for years after diagnosis impact the perception of the dynamics of this chronic process. The emerging information is helping the identification of novel therapeutic targets and has implications for the design of clinical trials.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Endocrinología/tendencias , Páncreas/patología , Autoinmunidad/fisiología , Autopsia , Diabetes Mellitus Tipo 1/historia , Diabetes Mellitus Tipo 1/inmunología , Progresión de la Enfermedad , Endocrinología/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Páncreas/inmunología
17.
BMC Cancer ; 21(1): 1197, 2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34758773

RESUMEN

BACKGROUND: Both activated tumor-infiltrating lymphocytes (TILs) and immune-suppressive cells, such as regulatory T cells (Tregs), in the tumor microenvironment (TME) play an important role in the prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). METHODS: The densities of TILs, programmed death receptor 1 (PD-1) + T cells, and forkhead box P3 (Foxp3) + T cells were analyzed by immunohistochemical staining. The associations of the immunological status of the PDAC microenvironment with overall survival (OS) time and disease-free survival (DFS) time were evaluated. RESULTS: PDAC patients with a high density of TILs in the TME or PD-1-positive T cells in tertiary lymphoid aggregates (TLAs) demonstrated a significantly better prognosis than those with a low density of TILs or PD-1-negativity, respectively. Moreover, PDAC patients with high levels of Foxp3-expressing T cells showed a worse prognosis than those with low levels of Foxp3-expressing T cells. Importantly, even with a high density of the TILs in TME or PD-1-positive T cells in TLAs, PDAC patients with high levels of Foxp3-expressing T cells showed a worse prognosis than patients with low levels of Foxp3-expressing T cells. A PDAC TME with a high density of TILs/high PD-1 positivity/low Foxp3 expression was an independent predictive marker associated with superior prognosis. CONCLUSION: Combined assessment of TILs, PD-1+ cells, and Foxp3+ T cells in the TME may predict the prognosis of PDAC patients following surgical resection.


Asunto(s)
Carcinoma Ductal Pancreático/mortalidad , Linfocitos Infiltrantes de Tumor/inmunología , Recurrencia Local de Neoplasia/epidemiología , Neoplasias Pancreáticas/inmunología , Microambiente Tumoral/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/cirugía , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Factores de Transcripción Forkhead/metabolismo , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/prevención & control , Páncreas/inmunología , Páncreas/patología , Páncreas/cirugía , Pancreatectomía , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Pronóstico , Receptor de Muerte Celular Programada 1/metabolismo , Estudios Retrospectivos
18.
J Immunol Res ; 2021: 5123823, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34485535

RESUMEN

Acute pancreatitis (AP) is one of the leading causes of hospital admission, 20% of which could progress to the severe type with extensive acinar cell necrosis. Clinical studies have reported that diabetes is an independent risk factor of the incidence of AP and is associated with higher severity than nondiabetic subjects. However, how diabetes participates in AP progression is not well defined. To investigate this question, wild-type (wt) and diabetic db/db mice at the age of 16 weeks were used in the study. AP was induced in wt recipients by 10 injections of 50 µg/kg caerulein with a 1 h interval. One hour after the last caerulein injection, bone marrow cells (BMC) isolated from wt and db/db mice were injected intraperitoneally into the recipients (1 × 107cells/recipient). The recipients with no BMC injection served as controls. Thirteen hours after BMC injection, serum lipase activity was 1.8- and 1.3-folds higher in mice that received db/db BMC, compared with those with no injection and wt BMC injection, respectively (p ≤ 0.02 for both). By H&E staining, the overall severity score was 14.7 for no cell injection and 16.6 for wt BMC injection and increased to 22.6 for db/db BMC injection (p ≤ 0.002 for both). In particular, mice with db/db BMC injection developed more acinar cell necrosis and vacuolization than the other groups (p ≤ 0.03 for both). When sections were stained with an antibody against myeloperoxidase (MPO), the density of MPO+ cells in pancreatitis was 1.9- and 1.6-folds higher than wt BMC and no BMC injection groups, separately (p ≤ 0.02 for both). Quantified by ELISA, db/db BMC produced more IL-6, GM-CSF, and IL-10 compared with wt BMC (p ≤ 0.04 for all). In conclusion, BMC of db/db mice produced more inflammatory cytokines. In response to acinar cell injury, diabetic BMC aggravated the inflammation cascade and acinar cell injury, leading to the progression of acute pancreatitis.


Asunto(s)
Células de la Médula Ósea/inmunología , Complicaciones de la Diabetes/inmunología , Pancreatitis/inmunología , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Ceruletida/administración & dosificación , Ceruletida/toxicidad , Citocinas/metabolismo , Complicaciones de la Diabetes/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Necrosis , Páncreas/efectos de los fármacos , Páncreas/inmunología , Páncreas/patología , Pancreatitis/inducido químicamente , Pancreatitis/patología
19.
Int Immunopharmacol ; 100: 108067, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34481142

RESUMEN

Acute pancreatitis (AP) is an inflammatory disease of the pancreas. Accumulating studies have revealed the involvement of tumor necrosis factor alpha-induced protein 3 (TNFAIP3) in the progression of AP. Here, the current study was conducted to elucidate the role of TNFAIP3 and the underlying molecular mechanisms on the progression of AP. The in vivo animal model and in vitro cell model of AP were generated by retrograde injection of sodium taurocholate and stimulation of cerulein into AR42J cells, respectively. Relationships among TNFAIP3, receptor interacting protein 3 (RIP3) and nod-like receptor protein 3 (NLRP3) were predicted on bioinformatics websites and verified by co-immunoprecipitation. AR42J cells were transfected with overexpressing plasmid or shRNA to study the effects of TNFAIP3/RIP3/NLRP3 axis on cell proliferation and apoptosis, secretion of inflammatory cytokines and production of ROS. The effect of TNFAIP3/RIP3/NLRP3 axis in AP was further confirmed in vivo. High expression of TNFAIP3 was observed in AP pancreatic tissues and AP cell model. TNFAIP3 increased RIP phosphorylation through deubiquitination. RIP activated the NLRP3 inflammasome. Silencing of TNFAIP3 or RIP3T led to elevated proliferation and inhibited apoptosis in AR42J cells, accompanied by decreased inflammatory cytokine levels and ROS production. The protective role of inhibited TNFAIP3 in AP was confirmed evidenced by reduced levels of AMY, LIPA, and ROS in vivo. Collectively, overexpressed TNFAIP3 could contribute to the progression of AP by activating RIP3/NLRP3 axis, providing a potential therapeutic target for AP treatment.


Asunto(s)
Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Pancreatitis/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Masculino , Páncreas/inmunología , Páncreas/patología , Pancreatitis/inducido químicamente , Pancreatitis/patología , Fosforilación/inmunología , Ratas , Ácido Taurocólico/administración & dosificación , Ácido Taurocólico/toxicidad , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Ubiquitinación/inmunología
20.
Genes (Basel) ; 12(9)2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34573354

RESUMEN

Inflammatory bowel diseases (IBDs) are chronic relapsing inflammatory conditions of the gastrointestinal tract, encompassing Crohn's disease (CD), ulcerative colitis (UC) and inflammatory bowel disease unclassified (IBD-U). They are currently considered as systemic disorders determined by a set of genetic predispositions, individual susceptibility and environmental triggers, potentially able to involve other organs and systems than the gastrointestinal tract. A large number of patients experiences one or more extraintestinal manifestations (EIMs), whose sites affected are mostly represented by the joints, skin, bones, liver, eyes, and pancreas. Pancreatic abnormalities are not uncommon and are often underestimated, encompassing acute and chronic pancreatitis, autoimmune pancreatitis, exocrine pancreatic insufficiency and asymptomatic elevation of pancreatic enzymes. In most cases they are the result of environmental triggers. However, several genetic polymorphisms may play a role as precipitating factors or contributing to a more severe course. The aim of this paper is to provide an updated overview on the available evidence concerning the etiology, pathogenesis and clinical presentation of pancreatic diseases in IBD pediatric patients.


Asunto(s)
Enfermedades Inflamatorias del Intestino/complicaciones , Pancreatitis/genética , Niño , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Páncreas/inmunología , Páncreas/patología , Pancreatitis/inmunología , Pancreatitis/patología , Polimorfismo de Nucleótido Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...