Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 789
Filter
1.
Arch Oral Biol ; 168: 106080, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39217919

ABSTRACT

OBJECTIVE: Sox2 plays crucial roles in tissues homeostasis and regeneration. However, there are lack of a comprehensive examination of Sox2 expression and its functional role in submandibular gland regeneration. Therefore, we aimed to elucidate the impact of Sox2 on submandibular gland regeneration. MATERIALS AND METHODS: A Sprague-Dawley rat submandibular gland duct ligation/de-ligation regeneration model was conducted in this study. Sox2-shRNA vectors were retro-ductally administered into the submandibular gland to establish a stable Sox2 knockdown model. Conventional histopathological and molecular biological methods were used to investigate phenotypic changes. RESULTS: The submandibular gland normalized completely 28 days after ligature removal (following 7 days of duct ligation). AQP5 expression gradually increased after ligation removal until returning to normal levels. In submandibular gland regeneration, Sox2 re-expressed and co-expressed with AQP5+ acinar cells, and Sox2 expression peaked on day 14, recovered to normal on day 28, reproducing the developmental pattern. Sox2 knockdown hindered gland regeneration and induced irreversible fibrosis. The AQP5 expression was significantly lower than the contemporaneous solely ligated group, while the blue collagen deposition and the Vimentin expression increased prominently. The expression of CD68, IL-1ß, TNF-α and IL-17A increased significantly, and epithelial cells in the Sox2 knockdown group expressed higher levels of IL-17A. CONCLUSIONS: These findings highlight Sox2 as a crucial regulator of the acinar cell lineage. Sox2+ progenitor cells are pivotal for acinar cell maintenance, which is indispensable for submandibular gland regeneration. Collectively, our findings may help develop targeted interventions for enhancing tissue repair and preventing irreversible fibrosis in salivary gland disorders.


Subject(s)
Aquaporin 5 , Rats, Sprague-Dawley , Regeneration , SOXB1 Transcription Factors , Stem Cells , Submandibular Gland , Animals , Submandibular Gland/metabolism , Rats , Regeneration/physiology , Aquaporin 5/metabolism , SOXB1 Transcription Factors/metabolism , Stem Cells/metabolism , Male , Ligation , Acinar Cells/metabolism , Vimentin/metabolism , RNA, Small Interfering , CD68 Molecule
2.
Mol Ther ; 32(8): 2624-2640, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38956871

ABSTRACT

Chronic pancreatitis (CP) is marked by progressive fibrosis and the activation of pancreatic stellate cells (PSCs), accompanied by the destruction of pancreatic parenchyma, leading to the loss of acinar cells (ACs). Few research studies have explored the mechanism by which damaged ACs (DACs) contribute to PSCs activation and pancreatic fibrosis. Currently, there are no effective drugs for curing CP or limiting the progression of pancreatic fibrosis. In this research, co-culture with intact acinar cells (IACs) suppressed PSC activation, while co-culture with DACs did the opposite. Krüppel-like factor 4 (KLF4) was significantly upregulated in DACs and was established as the key molecule that switches ACs from PSCs-suppressor to PSCs-activator. We revealed the exosomes of IACs contributed to the anti-activated function of IACs-CS on PSCs. MiRNome profiling showed that let-7 family is significantly enriched in IAC-derived exosomes (>30% miRNome), which partially mediates IACs' suppressive impacts on PSCs. Furthermore, it has been observed that the enrichment of let-7 in exosomes was influenced by the expression level of KLF4. Mechanistic studies demonstrated that KLF4 in ACs upregulated Lin28A, thereby decreasing let-7 levels in AC-derived exosomes, and thus promoting PSCs activation. We utilized an adeno-associated virus specifically targeting KLF4 in ACs (shKLF4-pAAV) to suppress PSCs activation in CP, resulting in reduced pancreatic fibrosis. IAC-derived exosomes hold potential as potent weapons against PSCs activation via let-7s, while activated KLF4/Lin28A signaling in DACs diminished such functions. ShKLF4-pAAV holds promise as a novel therapeutic approach for CP.


Subject(s)
Acinar Cells , Exosomes , Fibrosis , Kruppel-Like Factor 4 , MicroRNAs , Pancreatic Stellate Cells , Pancreatitis, Chronic , Kruppel-Like Factor 4/metabolism , Animals , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Exosomes/metabolism , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/pathology , MicroRNAs/genetics , Acinar Cells/metabolism , Acinar Cells/pathology , Dependovirus/genetics , Mice , Humans , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Male , Coculture Techniques , Pancreas/metabolism , Pancreas/pathology , Genetic Therapy/methods
3.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999930

ABSTRACT

Although SARS-CoV-2 induces mucin hypersecretion in the respiratory tract, hyposalivation/xerostomia has been reported by COVID-19 patients. We evaluate the submandibular gland (SMGs) pathogenesis in SARS-CoV-2-infected K18-hACE2 mice, focusing on the impact of infection on the mucin production and structural integrity of acini, ductal system, myoepithelial cells (MECs) and telocytes. The spike protein, the nucleocapsid protein, hACE2, actin, EGF, TNF-α and IL-1ß were detected by immunofluorescence, and the Egfr and Muc5b expression was evaluated. In the infected animals, significant acinar hypertrophy was observed in contrast to ductal atrophy. Nucleocapsid proteins and/or viral particles were detected in the SMG cells, mainly in the nuclear membrane-derived vesicles, confirming the nuclear role in the viral formation. The acinar cells showed intense TNF-α and IL-1ß immunoexpression, and the EGF-EGFR signaling increased, together with Muc5b upregulation. This finding explains mucin hypersecretion and acinar hypertrophy, which compress the ducts. Dying MECs and actin reduction were also observed, indicating failure of contraction and acinar support, favoring acinar hypertrophy. Viral assembly was found in the dying telocytes, pointing to these intercommunicating cells as viral transmitters in SMGs. Therefore, EGF-EGFR-induced mucin hypersecretion was triggered by SARS-CoV-2 in acinar cells, likely mediated by cytokines. The damage to telocytes and MECs may have favored the acinar hypertrophy, leading to ductal obstruction, explaining xerostomia in COVID-19 patients. Thus, acinar cells, telocytes and MECs may be viral targets, which favor replication and cell-to-cell viral transmission in the SMG, corroborating the high viral load in saliva of infected individuals.


Subject(s)
COVID-19 , ErbB Receptors , SARS-CoV-2 , Submandibular Gland , Xerostomia , COVID-19/pathology , COVID-19/virology , COVID-19/metabolism , Animals , Submandibular Gland/virology , Submandibular Gland/pathology , Submandibular Gland/metabolism , SARS-CoV-2/physiology , Mice , Xerostomia/etiology , Xerostomia/pathology , Xerostomia/virology , Xerostomia/metabolism , ErbB Receptors/metabolism , Humans , Angiotensin-Converting Enzyme 2/metabolism , Mucin-5B/metabolism , Acinar Cells/pathology , Acinar Cells/metabolism , Acinar Cells/virology , Interleukin-1beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Disease Models, Animal
4.
Cancer Res ; 84(14): 2297-2312, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39005053

ABSTRACT

Metabolic reprogramming is a hallmark of cancer and is crucial for cancer progression, making it an attractive therapeutic target. Understanding the role of metabolic reprogramming in cancer initiation could help identify prevention strategies. To address this, we investigated metabolism during acinar-to-ductal metaplasia (ADM), the first step of pancreatic carcinogenesis. Glycolytic markers were elevated in ADM lesions compared with normal tissue from human samples. Comprehensive metabolic assessment in three mouse models with pancreas-specific activation of KRAS, PI3K, or MEK1 using Seahorse measurements, nuclear magnetic resonance metabolome analysis, mass spectrometry, isotope tracing, and RNA sequencing analysis revealed a switch from oxidative phosphorylation to glycolysis in ADM. Blocking the metabolic switch attenuated ADM formation. Furthermore, mitochondrial metabolism was required for de novo synthesis of serine and glutathione (GSH) but not for ATP production. MYC mediated the increase in GSH intermediates in ADM, and inhibition of GSH synthesis suppressed ADM development. This study thus identifies metabolic changes and vulnerabilities in the early stages of pancreatic carcinogenesis. Significance: Metabolic reprogramming from oxidative phosphorylation to glycolysis mediated by MYC plays a crucial role in the development of pancreatic cancer, revealing a mechanism driving tumorigenesis and potential therapeutic targets. See related commentary by Storz, p. 2225.


Subject(s)
Metaplasia , Pancreatic Neoplasms , Animals , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Mice , Metaplasia/metabolism , Metaplasia/pathology , Glycolysis , Carcinogenesis/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Oxidative Phosphorylation , Glutathione/metabolism , Cellular Reprogramming , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Male , Mitochondria/metabolism , Mitochondria/pathology , Metabolic Reprogramming
5.
Front Immunol ; 15: 1418703, 2024.
Article in English | MEDLINE | ID: mdl-39044831

ABSTRACT

Introduction: Salivary gland dysfunction, often resulting from salivary gland obstruction-induced inflammation, is a prevalent condition. Corticosteroid, known for its anti-inflammatory and immunomodulatory properties, is commonly prescribed in clinics. This study investigates the therapeutic implications and potential side effects of dexamethasone on obstructive sialadenitis recovery using duct ligation mice and salivary gland organoid models. Methods: Functional and pathological changes were assessed after administering dexamethasone to the duct following deligation 2 weeks after maintaining ligation of the mouse submandibular duct. Additionally, lipopolysaccharide- and tumor necrosis factor-induced salivary gland organoid inflammation models were established to investigate the effects and underlying mechanisms of action of dexamethasone. Results: Dexamethasone administration facilitated SG function restoration, by increasing salivary gland weight and saliva volume while reducing saliva lag time. Histological evaluation revealed, reduced acinar cell atrophy and fibrosis with dexamethasone treatment. Additionally, dexamethasone suppressed pro-inflammatory cytokines IL-1ß and TNF expression. In a model of inflammation in salivary gland organoids induced by inflammatory substances, dexamethasone restored acinar markers such as AQP5 gene expression levels, while inhibiting pro-inflammatory cytokines TNF and IL6, as well as chemokines CCL2, CXCL5, and CXCL12 induction. Macrophages cultured in inflammatory substance-treated media from salivary gland organoid cultures exhibited pro-inflammatory polarization. However, treatment with dexamethasone shifted them towards an anti-inflammatory phenotype by reducing M1 markers (Tnf, Il6, Il1b, and Cd86) and elevating M2 markers (Ym1, Il10, Cd163, and Klf4). However, high-dose or prolonged dexamethasone treatment induced acino-ductal metaplasia and had side effects in both in vivo and in vitro models. Conclusions: Our findings suggest the effectiveness of corticosteroids in treating obstructive sialadenitis-induced salivary gland dysfunction by regulating pro-inflammatory cytokines.


Subject(s)
Dexamethasone , Kruppel-Like Factor 4 , Sialadenitis , Animals , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Dexamethasone/administration & dosage , Mice , Sialadenitis/drug therapy , Sialadenitis/pathology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Organoids/drug effects , Cytokines/metabolism , Mice, Inbred C57BL , Salivary Glands/drug effects , Salivary Glands/pathology , Salivary Glands/metabolism , Salivary Glands/immunology , Aquaporin 5/metabolism , Aquaporin 5/genetics , Male , Macrophages/drug effects , Macrophages/immunology , Acinar Cells/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Humans
6.
ACS Nano ; 18(29): 19283-19302, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38990194

ABSTRACT

Developing strategies to target injured pancreatic acinar cells (PACs) in conjunction with primary pathophysiology-specific pharmacological therapy presents a challenge in the management of acute pancreatitis (AP). We designed and synthesized a trypsin-cleavable organosilica precursor bridged by arginine-based amide bonds, leveraging trypsin's ability to selectively identify guanidino groups on arginine via Asp189 at the active S1 pocket and cleave the carboxy-terminal (C-terminal) amide bond via catalytic triads. The precursors were incorporated into the framework of mesoporous silica nanoparticles (MSNs) for encapsulating the membrane-permeable Ca2+ chelator BAPTA-AM with a high loading content (∼43.9%). Mesenchymal stem cell membrane coating and surface modification with PAC-targeting ligands endow MSNs with inflammation recruitment and precise PAC-targeting abilities, resulting in the highest distribution at 3 h in the pancreas with 4.7-fold more accumulation than that of naked MSNs. The outcomes transpired as follows: After bioinspired MSNs' skeleton biodegradation by prematurely and massively activated trypsin, BAPTA-AM was on-demand released in injured PACs, thereby effectively eliminating intracellular calcium overload (reduced Ca2+ level by 81.3%), restoring cellular redox status, blocking inflammatory cascades, and inhibiting cell necrosis by impeding the IκBα/NF-κB/TNF-α/IL-6 and CaMK-II/p-RIP3/p-MLKL/caspase-8,9 signaling pathways. In AP mice, a single dose of the formulation significantly restored pancreatic function (lipase and amylase reduced more by 60%) and improved the survival rate from 50 to 91.6%. The formulation offers a potentially effective strategy for clinical translation in AP treatment.


Subject(s)
Pancreatitis , Trypsin , Animals , Pancreatitis/drug therapy , Pancreatitis/pathology , Pancreatitis/metabolism , Trypsin/metabolism , Trypsin/chemistry , Mice , Porosity , Nanomedicine , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Organosilicon Compounds/chemistry , Organosilicon Compounds/pharmacology , Male , Humans , Acinar Cells/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Mice, Inbred C57BL
7.
Diabetes ; 73(9): 1447-1461, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38905124

ABSTRACT

Exocrine-to-endocrine cross talk in the pancreas is crucial to maintain ß-cell function. However, the molecular mechanisms underlying this cross talk are largely undefined. Trefoil factor 2 (Tff2) is a secreted factor known to promote the proliferation of ß-cells in vitro, but its physiological role in vivo in the pancreas is unknown. Also, it remains unclear which pancreatic cell type expresses Tff2 protein. We therefore created a mouse model with a conditional knockout of Tff2 in the murine pancreas. We find that the Tff2 protein is preferentially expressed in acinar but not ductal or endocrine cells. Tff2 deficiency in the pancreas reduces ß-cell mass on embryonic day 16.5. However, homozygous mutant mice are born without a reduction of ß-cells and with acinar Tff3 compensation by day 7. When mice are aged to 1 year, both male and female homozygous and male heterozygous mutants develop impaired glucose tolerance without affected insulin sensitivity. Perifusion analysis reveals that the second phase of glucose-stimulated insulin secretion from islets is reduced in aged homozygous mutant compared with controls. Collectively, these results demonstrate a previously unknown role of Tff2 as an exocrine acinar cell-derived protein required for maintaining functional endocrine ß-cells in mice.


Subject(s)
Acinar Cells , Aging , Insulin-Secreting Cells , Mice, Knockout , Trefoil Factor-2 , Animals , Insulin-Secreting Cells/metabolism , Mice , Trefoil Factor-2/metabolism , Trefoil Factor-2/genetics , Male , Acinar Cells/metabolism , Female , Aging/metabolism , Aging/physiology , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/cytology , Insulin Secretion/physiology , Insulin Secretion/genetics , Trefoil Factors/metabolism , Trefoil Factors/genetics , Peptides/metabolism
8.
Cell Rep ; 43(6): 114346, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38850534

ABSTRACT

Histopathological heterogeneity in the human pancreas is well documented; however, functional evidence at the tissue level is scarce. Herein, we investigate in situ glucose-stimulated islet and carbachol-stimulated acinar cell secretion across the pancreas head (PH), body (PB), and tail (PT) regions in donors without diabetes (ND; n = 15), positive for one islet autoantibody (1AAb+; n = 7), and with type 1 diabetes (T1D; <14 months duration, n = 5). Insulin, glucagon, pancreatic amylase, lipase, and trypsinogen secretion along with 3D tissue morphometrical features are comparable across regions in ND. In T1D, insulin secretion and beta-cell volume are significantly reduced within all regions, while glucagon and enzymes are unaltered. Beta-cell volume is lower despite normal insulin secretion in 1AAb+, resulting in increased volume-adjusted insulin secretion versus ND. Islet and acinar cell secretion in 1AAb+ are consistent across the PH, PB, and PT. This study supports low inter-regional variation in pancreas slice function and, potentially, increased metabolic demand in 1AAb+.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin , Islets of Langerhans , Humans , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Insulin/metabolism , Female , Insulin Secretion/drug effects , Adult , Middle Aged , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Acinar Cells/metabolism , Acinar Cells/pathology , Glucagon/metabolism , Glucose/metabolism , Autoantibodies/immunology , Amylases/metabolism
9.
Biomolecules ; 14(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38927047

ABSTRACT

Acute pancreatitis (AP) entails pancreatic inflammation, tissue damage and dysregulated enzyme secretion, including pancreatic lipase (PL). The role of irisin, an anti-inflammatory and anti-apoptotic cytokine, in AP and exocrine pancreatic stress is unclear. We have previously shown that irisin regulates PL through the PPARγ-PGC1α-FNDC5 pathway. In this study, we investigated irisin and irisin's pathway on AP in in vitro (AR42J-B13) and ex vivo (rat primary acinar) models using molecular, biochemical and immunohistochemistry methodology. Pancreatitis induction (cerulein (cer)) resulted in a significant up-regulation of the PPARγ-PGC1α-FNDC5 axis, PL expression and secretion and endoplasmic reticulum (ER) stress unfolded protein response (UPR) signal-transduction markers (CHOP, XBP-1 and ATF6). Irisin addition in the cer-pancreatitis state resulted in a significant down-regulation of the PPARγ-PGC1α-FNDC5 axis, PPARγ nucleus-translocation and inflammatory state (TNFα and IL-6) in parallel to diminished PL expression and secretion (in vitro and ex vivo models). Irisin addition up-regulated the expression of pro-survival UPR markers (ATF6 and XBP-1) and reduced UPR pro-apoptotic markers (CHOP) under cer-pancreatitis and induced ER stress (tunicamycin), consequently increasing cells viability. Irisin's pro-survival effect under cer-pancreatitis state was abolished under PPARγ inhibition. Our findings suggest irisin as a potential therapeutic option for AP via its ability to up-regulate pro-survival UPR signals and activate the PPARγ-PGC1α-FNDC5 pathway.


Subject(s)
Acinar Cells , Cell Survival , Fibronectins , PPAR gamma , Pancreatitis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction , Unfolded Protein Response , Fibronectins/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis/drug therapy , PPAR gamma/metabolism , Unfolded Protein Response/drug effects , Rats , Cell Survival/drug effects , Acinar Cells/metabolism , Acinar Cells/drug effects , Acinar Cells/pathology , Signal Transduction/drug effects , Endoplasmic Reticulum Stress/drug effects , Ceruletide , Male , Cell Line , Lipase/metabolism
10.
Discov Med ; 36(185): 1162-1168, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38926102

ABSTRACT

BACKGROUND: Atypical acinar cell foci (AACF) seen in pancreatic cancer are fatal and have been studied with some causative agents. However, for the first time, the effect of acetylsalicylic acid with nitric oxide (NO-ASA) on AACF was examined in this study. Although NO-ASA has very successful inhibitory effects against some types of cancer, it has not been investigated whether they can exert their inhibition effects on AACFs. METHODS: For experimental purposes, 21 14-day-old male Wistar albino rats were used. Azaserine (30 mg/kg) was dissolved in 0.9% NaCl solution and injected intraperitoneally (i.p.) into 14 rats, except for the Control group (Cont) rats, for three weeks. Rats that were injected with azaserine once a week for three weeks and those that did not receive treatment were divided into experimental groups. 15 days after the end of the azaserine injection protocol, NO-ASA was applied to azaserine with NO-ASA (Az+NO-ASA) group rats three consecutive times with an interval of 15 days by gavage. At the end of the 5-month period, pancreatic tissue was dissected and weighed. Pancreas preparations prepared from histological sections were examined for AACF burden and analyzed via a video image analyzer. One-way analysis of variance (ANOVA) non-parametric statistical analyses were performed to test whether there was a difference between the averages of the experimental and Control groups. RESULTS: AACF burden in both groups injected with azaserine was found to be statistically significant in all categories compared to that of the Control group (p < 0.05). The average Calculated Estimated average AACF volume (mm3) values, the Calculated estimated average AACF diameter (µm), the Estimated average number of AACF per unit volume, AACF rate as a % of Calculated Organ Volume were higher in the AzCont group rats than in the Az+NO-ASA group, when compared, and there was an important level statistical difference between the groups (p < 0.05). It was determined that for all parameters AACFs load in Az+NO-ASA group rats were significantly reduced compared to that of AzCont group rats (p < 0.05). CONCLUSIONS: We observed that, as a result of the NO-ASA application, the experimental AACF focus ratio created by azaserine injection was significantly inhibited. The inhibitory effect of AACFs in Az+NO-ASA group rats may have resulted from the significant and independent chemopreventive and/or chemotherapeutic activity of NO-ASA against exocrine pancreatic AACF foci.


Subject(s)
Acinar Cells , Aspirin , Nitric Oxide , Pancreas, Exocrine , Pancreatic Neoplasms , Rats, Wistar , Animals , Male , Aspirin/pharmacology , Aspirin/therapeutic use , Aspirin/administration & dosage , Nitric Oxide/metabolism , Rats , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Acinar Cells/drug effects , Acinar Cells/pathology , Acinar Cells/metabolism , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/pathology
11.
J Pathol ; 263(4-5): 466-481, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38924548

ABSTRACT

The E3 ubiquitin ligase thyroid hormone receptor interacting protein 12 (TRIP12) has been implicated in pancreatic adenocarcinoma (PDAC) through its role in mediating the degradation of pancreas transcription factor 1a (PTF1a). PTF1a is a transcription factor essential for the acinar differentiation state that is notably diminished during the early steps of pancreatic carcinogenesis. Despite these findings, the direct involvement of TRIP12 in the onset of pancreatic cancer has yet to be established. In this study, we demonstrated that TRIP12 protein was significantly upregulated in human pancreatic preneoplastic lesions. Furthermore, we observed that TRIP12 overexpression varied within PDAC samples and PDAC-derived cell lines. We further demonstrated that TRIP12 was required for PDAC-derived cell growth and for the expression of E2F-targeted genes. Acinar-to-ductal cell metaplasia (ADM) is a reversible process that reflects the high plasticity of acinar cells. ADM becomes irreversible in the presence of oncogenic Kras mutations and leads to the formation of preneoplastic lesions. Using two genetically modified mouse models, we showed that a loss of TRIP12 prevented acini from developing ADM in response to pancreatic injury. With two additional mouse models, we further discovered that a depletion of TRIP12 prevented the formation of KrasG12D-induced preneoplastic lesions and impaired metastasis formation in the presence of mutated KrasG12D and Trp53R172H genes. In summary our study identified an overexpression of TRIP12 from the early stages of pancreatic carcinogenesis and proposed this E3 ubiquitin ligase as a novel regulator of acinar plasticity with an important dual role in initiation and metastatic steps of PDAC. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Acinar Cells , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Ubiquitin-Protein Ligases , Animals , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/enzymology , Humans , Acinar Cells/pathology , Acinar Cells/metabolism , Acinar Cells/enzymology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/enzymology , Metaplasia/pathology , Metaplasia/metabolism , Cell Plasticity , Carcinogenesis/genetics , Carcinogenesis/metabolism , Mice , Cell Line, Tumor , Cell Proliferation , Mice, Knockout , Gene Expression Regulation, Neoplastic , Precancerous Conditions/pathology , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/enzymology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/metabolism , Carrier Proteins
12.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167310, 2024 10.
Article in English | MEDLINE | ID: mdl-38901651

ABSTRACT

The pancreas is a glandular organ with both endocrine and exocrine functions. Researchers have investigated the roles of several Rab proteins, which are major regulators of membrane trafficking, in pancreatic exocytosis of zymogen granules in exocrine cells, also known as acinar cells. However, detailed molecular mechanisms mediated by Rab proteins are not fully understood. RASEF/Rab45 is an atypical Rab GTPase that contains N-terminal EF-hand and coiled-coil domains, as well as a C-terminal Rab-GTPase domain. In this study, we investigated the in vivo role of RASEF in pancreatic acinar cells using RASEF-knockout (KO) mice. Morphological analyses revealed that pancreatic acinar cells in RASEF-KO mice had an increased number of zymogen granules and abnormal formations of organelles, such as the endoplasmic reticulum (ER) and lysosomes. Biochemical analyses showed that ER proteins were decreased, but digestive enzymes were increased in the RASEF-KO pancreas. Moreover, trypsinogen was activated and co-localized with the endo-lysosomal marker LAMP1 in RASEF-KO pancreas. Upon cerulein administration to induce acute pancreatitis, impaired enzyme release from the pancreas was observed in the serum of RASEF-KO mice. These findings suggest that RASEF likely regulates the formation and sorting of zymogen granules and secretion of digestive enzymes by pancreatic acinar cells.


Subject(s)
Acinar Cells , Secretory Vesicles , ras Guanine Nucleotide Exchange Factors , Animals , Male , Mice , Acinar Cells/metabolism , Endoplasmic Reticulum/metabolism , Exocytosis , Lysosomes/metabolism , Mice, Knockout , Pancreas/metabolism , Pancreas, Exocrine/metabolism , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis/chemically induced , Protein Transport , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Secretory Vesicles/metabolism , Trypsinogen/metabolism , ras Guanine Nucleotide Exchange Factors/genetics , ras Guanine Nucleotide Exchange Factors/metabolism
13.
Gastroenterology ; 167(4): 718-732.e18, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38729450

ABSTRACT

BACKGROUND & AIMS: Acinar-to-ductal metaplasia (ADM) is crucial in the development of pancreatic ductal adenocarcinoma. However, our understanding of the induction and resolution of ADM remains limited. We conducted comparative transcriptome analyses to identify conserved mechanisms of ADM in mouse and human. METHODS: We identified Sox4 among the top up-regulated genes. We validated the analysis by RNA in situ hybridization. We performed experiments in mice with acinar-specific deletion of Sox4 (Ptf1a: CreER; Rosa26-LSL-YFPLSL-YFP; Sox4fl/fl) with and without an activating mutation in Kras (KrasLSL-G12D/+). Mice were given caerulein to induce pancreatitis. We performed phenotypic analysis by immunohistochemistry, tissue decellularization, and single-cell RNA sequencing. RESULTS: We demonstrated that Sox4 is reactivated in ADM and pancreatic intraepithelial neoplasias. Contrary to findings in other tissues, Sox4 actually counteracts cellular dedifferentiation and helps maintain tissue homeostasis. Moreover, our investigations unveiled the indispensable role of Sox4 in the specification of mucin-producing cells and tuft-like cells from acinar cells. We identified Sox4-dependent non-cell-autonomous mechanisms regulating the stromal reaction during disease progression. Notably, Sox4-inferred targets are activated upon KRAS inactivation and tumor regression. CONCLUSIONS: Our results indicate that our transcriptome analysis can be used to investigate conserved mechanisms of tissue injury. We demonstrate that Sox4 restrains acinar dedifferentiation and is necessary for the specification of acinar-derived metaplastic cells in pancreatic injury and cancer initiation and is activated upon Kras ablation and tumor regression in mice. By uncovering novel potential strategies to promote tissue homeostasis, our findings offer new avenues for preventing the development of pancreatic ductal adenocarcinoma.


Subject(s)
Acinar Cells , Carcinoma, Pancreatic Ductal , Cell Dedifferentiation , Ceruletide , Metaplasia , Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Animals , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Acinar Cells/pathology , Acinar Cells/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Metaplasia/genetics , Metaplasia/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Mice , Humans , Pancreatitis/pathology , Pancreatitis/genetics , Pancreatitis/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Disease Models, Animal , Pancreas/pathology , Pancreas/metabolism , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Gene Expression Profiling , Carcinoma in Situ/pathology , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Transcriptome
14.
Pancreas ; 53(8): e662-e669, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38696385

ABSTRACT

OBJECTIVES: Amino acids play an essential role in protein synthesis, metabolism, and survival of pancreatic acini. Adequate nutritional support is important for acute pancreatitis treatment. However, high concentrations of arginine and lysine may induce acute pancreatitis. The study aimed to identify the most suitable l -amino acids as safe energy sources for pancreatic acinar cells. MATERIALS AND METHODS: Pancreatic acini were isolated from male Wistar rats. Effects of amino acids (0.1-20 mM) on uncoupled respiration of isolated acini were studied with a Clark electrode. Cell death was evaluated with fluorescent microscopy and DNA gel electrophoresis. RESULTS: Among the tested amino acids, glutamate, glutamine, alanine, lysine, and aspartate were able to stimulate the uncoupled respiration rate of isolated pancreatic acini, whereas arginine, histidine, and asparagine were not. Lysine, arginine, and glutamine (20 mM) caused complete loss of plasma membrane integrity of acinar cells after 24 hours of incubation. Glutamine also caused early (2-4 hours) cell swelling and blebbing. Aspartate, asparagine, and glutamate only moderately decreased the number of viable cells, whereas alanine and histidine were not toxic. DNA fragmentation assay and microscopic analysis of nuclei showed no evidence of apoptosis in cells treated with amino acids. CONCLUSIONS: Alanine and glutamate are safe and effective energy sources for mitochondria of pancreatic acinar cells.


Subject(s)
Acinar Cells , Amino Acids , Rats, Wistar , Animals , Male , Amino Acids/metabolism , Amino Acids/pharmacology , Acinar Cells/metabolism , Acinar Cells/drug effects , Rats , Pancreas/metabolism , Pancreas/drug effects , Energy Metabolism/drug effects , DNA Fragmentation/drug effects , Glutamine/metabolism , Glutamine/pharmacology , Arginine/pharmacology , Cell Survival/drug effects , Glutamic Acid/metabolism , Glutamic Acid/toxicity , Glutamic Acid/pharmacology , Mitochondria/metabolism , Mitochondria/drug effects , Apoptosis/drug effects , Cell Membrane/metabolism , Cell Membrane/drug effects , Oxygen Consumption/drug effects , Histidine/pharmacology
15.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731942

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) can originate from acinar-to-ductal metaplasia (ADM). Pancreatic acini harboring oncogenic Kras mutations are transdifferentiated to a duct-like phenotype that further progresses to become pancreatic intraepithelial neoplasia (PanIN) lesions, giving rise to PDAC. Although ADM formation is frequently observed in KrasG12D transgenic mouse models of PDAC, the exact mechanisms of how oncogenic KrasG12D regulates this process remain an enigma. Herein, we revealed a new downstream target of oncogenic Kras, cytokine CCL9, during ADM formation. Higher levels of CCL9 and its receptors, CCR1 and CCR3, were detected in ADM regions of the pancreas in p48cre:KrasG12D mice and human PDAC patients. Knockdown of CCL9 in KrasG12D-expressed pancreatic acini reduced KrasG12D-induced ADM in a 3D organoid culture system. Moreover, exogenously added recombinant CCL9 and overexpression of CCL9 in primary pancreatic acini induced pancreatic ADM. We also showed that, functioning as a downstream target of KrasG12D, CCL9 promoted pancreatic ADM through upregulation of the intracellular levels of reactive oxygen species (ROS) and metalloproteinases (MMPs), including MMP14, MMP3 and MMP2. Blockade of MMPs via its generic inhibitor GM6001 or knockdown of specific MMP such as MMP14 and MMP3 decreased CCL9-induced pancreatic ADM. In p48cre:KrasG12D transgenic mice, blockade of CCL9 through its specific neutralizing antibody attenuated pancreatic ADM structures and PanIN lesion formation. Furthermore, it also diminished infiltrating macrophages and expression of MMP14, MMP3 and MMP2 in the ADM areas. Altogether, our results provide novel mechanistic insight into how oncogenic Kras enhances pancreatic ADM through its new downstream target molecule, CCL9, to initiate PDAC.


Subject(s)
Acinar Cells , Carcinoma, Pancreatic Ductal , Metaplasia , Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Reactive Oxygen Species , Animals , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Mice , Reactive Oxygen Species/metabolism , Humans , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Metaplasia/metabolism , Metaplasia/genetics , Acinar Cells/metabolism , Acinar Cells/pathology , Mice, Transgenic , Chemokines, CC/metabolism , Chemokines, CC/genetics , Macrophage Inflammatory Proteins/metabolism , Macrophage Inflammatory Proteins/genetics , Pancreas/metabolism , Pancreas/pathology
16.
JCI Insight ; 9(13)2024 May 23.
Article in English | MEDLINE | ID: mdl-38781030

ABSTRACT

Acute pancreatitis (AP) is among the most common hospital gastrointestinal diagnoses; understanding the mechanisms underlying the severity of AP is critical for development of new treatment options for this disease. Here, we evaluate the biological function of phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) in AP pathogenesis in 2 independent genetically engineered mouse models of AP. PFKFB3 was elevated in AP and severe AP (SAP), and KO of Pfkfb3 abrogated the severity of alcoholic SAP (FAEE-SAP). Using a combination of genetic, pharmacological, and molecular studies, we defined the interaction of PFKFB3 with inositol 1,4,5-trisphosphate receptor (IP3R) as a key event mediating this phenomenon. Further analysis demonstrated that the interaction between PFKFB3 and IP3R promotes FAEE-SAP severity by altering intracellular calcium homeostasis in acinar cells. Together, our results support a PFKFB3-driven mechanism controlling AP pathobiology and define this enzyme as a therapeutic target to ameliorate the severity of this condition.


Subject(s)
Acinar Cells , Calcium , Inositol 1,4,5-Trisphosphate Receptors , Pancreatitis , Phosphofructokinase-2 , Animals , Phosphofructokinase-2/metabolism , Phosphofructokinase-2/genetics , Mice , Pancreatitis/metabolism , Pancreatitis/genetics , Pancreatitis/pathology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Calcium/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Mice, Knockout , Disease Models, Animal , Severity of Illness Index , Male , Humans , Calcium Signaling/genetics
17.
Biochem Pharmacol ; 225: 116279, 2024 07.
Article in English | MEDLINE | ID: mdl-38740221

ABSTRACT

Berberine, a natural isoquinoline alkaloid, exhibits a variety of pharmacological effects, but the pharmacological targets and mechanisms remain elusive. Here, we report a novel finding that berberine inhibits acetylcholine (ACh)-induced intracellular Ca2+ oscillations, mediated through an inhibition of the muscarinic subtype 3 (M3) receptor. Patch-clamp recordings and confocal Ca2+ imaging were applied to acute dissociated pancreatic acinar cells prepared from CD1 mice to examine the effects of berberine on ACh-induced Ca2+ oscillations. Whole-cell patch-clamp recordings showed that berberine (from 0.1 to 10 µM) reduced ACh-induced Ca2+ oscillations in a concentration-dependent manner, and this inhibition also depended on ACh concentrations. The inhibitory effect of berberine neither occurred in intracellular targets nor extracellular cholecystokinin (CCK) receptors, chloride (Cl-) channels, and store-operated Ca2+ channels. Together, the results demonstrate that berberine directly inhibits the muscarinic M3 receptors, further confirmed by evidence of the interaction between berberine and M3 receptors in pancreatic acinar cells.


Subject(s)
Acinar Cells , Berberine , Calcium Signaling , Receptor, Muscarinic M3 , Animals , Berberine/pharmacology , Receptor, Muscarinic M3/metabolism , Receptor, Muscarinic M3/antagonists & inhibitors , Mice , Acinar Cells/drug effects , Acinar Cells/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Pancreas/drug effects , Pancreas/metabolism , Male , Acetylcholine/metabolism , Calcium/metabolism , Dose-Response Relationship, Drug
18.
Nat Commun ; 15(1): 3740, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702347

ABSTRACT

Insufficient functional ß-cell mass causes diabetes; however, an effective cell replacement therapy for curing diabetes is currently not available. Reprogramming of acinar cells toward functional insulin-producing cells would offer an abundant and autologous source of insulin-producing cells. Our lineage tracing studies along with transcriptomic characterization demonstrate that treatment of adult mice with a small molecule that specifically inhibits kinase activity of focal adhesion kinase results in trans-differentiation of a subset of peri-islet acinar cells into insulin producing ß-like cells. The acinar-derived insulin-producing cells infiltrate the pre-existing endocrine islets, partially restore ß-cell mass, and significantly improve glucose homeostasis in diabetic mice. These findings provide evidence that inhibition of the kinase activity of focal adhesion kinase can convert acinar cells into insulin-producing cells and could offer a promising strategy for treating diabetes.


Subject(s)
Acinar Cells , Diabetes Mellitus, Experimental , Insulin-Secreting Cells , Animals , Insulin-Secreting Cells/metabolism , Mice , Acinar Cells/metabolism , Male , Insulin/metabolism , Cell Transdifferentiation , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Mice, Inbred C57BL , Protein Kinase Inhibitors/pharmacology , Islets of Langerhans/metabolism
19.
ACS Nano ; 18(21): 13885-13898, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38757565

ABSTRACT

Severe acute pancreatitis (SAP), characterized by pancreatic acinar cell death, currently lacks effective targeted therapies. Ellagic acid (EA), rich in pomegranate, shows promising anti-inflammatory and antioxidant effects in SAP treatment. However, the roles of other forms of EA, such as plant extracellular vesicles (EVs) extracted from pomegranate, and Urolithin A (UA), converted from EA through gut microbiota metabolism in vivo, have not been definitively elucidated. Our research aimed to compare the effects of pomegranate-derived EVs (P-EVs) and UA in the treatment of SAP to screen an effective formulation and to explore its mechanisms in protecting acinar cells in SAP. By comparing the protective effects of P-EVs and UA on injured acinar cells, UA showed superior therapeutic effects than P-EVs. Subsequently, we further discussed the mechanism of UA in alleviating SAP inflammation. In vivo animal experiments found that UA could not only improve the inflammatory environment of pancreatic tissue and peripheral blood circulation in SAP mice but also revealed that the mechanism of UA in improving SAP might be related to mitochondria and endoplasmic reticulum (ER) through the results including pancreatic tissue transcriptomics and transmission electron microscopy. Further research found that UA could regulate ER-mitochondrial calcium channels and reduce pancreatic tissue necroptosis. In vitro experiments of mouse pancreatic organoids and acinar cells also confirmed that UA could improve pancreatic inflammation by regulating the ER-mitochondrial calcium channel and necroptosis pathway proteins. This study not only explored the therapeutic effect of plant EVs on SAP but also revealed that UA could alleviate SAP by regulating ER-mitochondrial calcium channel and reducing acinar cell necroptosis, providing insights into the pathogenesis and potential treatment of SAP.


Subject(s)
Coumarins , Endoplasmic Reticulum , Mitochondria , Pancreatitis , Animals , Coumarins/pharmacology , Coumarins/chemistry , Pancreatitis/drug therapy , Pancreatitis/metabolism , Pancreatitis/pathology , Mice , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Calcium Channels/metabolism , Male , Mice, Inbred C57BL , Pomegranate/chemistry , Acinar Cells/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Extracellular Vesicles/metabolism , Extracellular Vesicles/chemistry
20.
Cell Mol Life Sci ; 81(1): 207, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38709385

ABSTRACT

The co-localization of the lysosomal protease cathepsin B (CTSB) and the digestive zymogen trypsinogen is a prerequisite for the initiation of acute pancreatitis. However, the exact molecular mechanisms of co-localization are not fully understood. In this study, we investigated the role of lysosomes in the onset of acute pancreatitis by using two different experimental approaches. Using an acinar cell-specific genetic deletion of the ras-related protein Rab7, important for intracellular vesicle trafficking and fusion, we analyzed the subcellular distribution of lysosomal enzymes and the severity of pancreatitis in vivo and ex vivo. Lysosomal permeabilization was performed by the lysosomotropic agent Glycyl-L-phenylalanine 2-naphthylamide (GPN). Acinar cell-specific deletion of Rab7 increased endogenous CTSB activity and despite the lack of re-distribution of CTSB from lysosomes to the secretory vesicles, the activation of CTSB localized in the zymogen compartment still took place leading to trypsinogen activation and pancreatic injury. Disease severity was comparable to controls during the early phase but more severe at later time points. Similarly, GPN did not prevent CTSB activation inside the secretory compartment upon caerulein stimulation, while lysosomal CTSB shifted to the cytosol. Intracellular trypsinogen activation was maintained leading to acute pancreatitis similar to controls. Our results indicate that initiation of acute pancreatitis seems to be independent of the presence of lysosomes and that fusion of lysosomes and zymogen granules is dispensable for the disease onset. Intact lysosomes rather appear to have protective effects at later disease stages.


Subject(s)
Cathepsin B , Lysosomes , Pancreatitis , Secretory Vesicles , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins , Animals , Lysosomes/metabolism , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis/genetics , Cathepsin B/metabolism , Cathepsin B/genetics , Mice , Secretory Vesicles/metabolism , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins/metabolism , Acute Disease , Acinar Cells/metabolism , Acinar Cells/pathology , Trypsinogen/metabolism , Trypsinogen/genetics , Ceruletide , Enzyme Precursors/metabolism , Enzyme Precursors/genetics , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL