1
|
Swetha K, Indumathi MC, Siddappa S, Chen CH, Marathe GK. Comparative Study of Non-invasive Mouse Models of Pancreatitis. Dig Dis Sci 2025; 70:233-244. [PMID: 39604666 DOI: 10.1007/s10620-024-08771-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND AND AIMS Although a relevant animal model is essential for studying human diseases, one has yet to be established for mouse pancreatitis. Early non-invasive models of mouse pancreatitis have serious limitations. METHODS In this study, we compared the efficiency, consistency, and reproducibility of inducing pancreatitis in 3 non-invasive mouse models of pancreatitis in Wistar albino mice: (1) L-arginine-induced model (2 intraperitoneal injections of 4 g/kg body weight of L-arginine spaced 1 h apart), (2) caerulein-induced model (6 intraperitoneal injections of 50 µg/kg body weight of caerulein at hourly intervals), and (3) caerulein + LPS (lipopolysaccharide)-induced model (6 intraperitoneal doses of 50 µg/kg body weight of caerulein at hourly intervals, along with an LPS [10 mg/kg body weight] injection immediately after the last caerulein injection). RESULTS Our findings showed that the L-arginine-induced model was inconsistent. The levels of the pancreatic enzymes, amylase and lipase, were higher in the caerulein and caerulein + LPS groups. Histological examination showed tissue destruction in the induced groups, with varying degrees of fibrosis in the caerulein + LPS group. CONCLUSIONS The caerulein + LPS model was the most reliable model in Wistar albino mice. Our findings may be useful in helping investigators choose the most appropriate animal model for pancreatitis research.
Collapse
Affiliation(s)
- Kamatam Swetha
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore-06, India
| | | | - Shiva Siddappa
- Division of Biochemistry, School of Life Sciences, JSS Academy of Higher Education and Research, Mysore-15, India
| | - Chu-Huang Chen
- Vascular and Medicinal Research, The Texas Heart Institute, Houston, TX, 77030, USA
| | - Gopal K Marathe
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore-06, India.
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore-06, India.
| |
Collapse
|
2
|
Jahangir S, Khatua B, Smichi N, Rajalingamgari P, Narayana Pillai A, Summers MJ, McFayden B, Kostenko S, Gades NM, Singh VP. Buprenorphine affects the initiation and severity of interleukin-induced acute pancreatitis in mice. Am J Physiol Gastrointest Liver Physiol 2024; 327:G16-G24. [PMID: 38651230 DOI: 10.1152/ajpgi.00083.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Acute pancreatitis (AP) is a common disease with no targeted therapy and has varied outcomes ranging from spontaneous resolution to being lethal. Although typically painful, AP can also be painless. Various agents, including opioids, are used for pain control in AP; the risks and benefits of which are often debated. As experimental AP in mice is used to study the efficacy of potential therapies, we studied the effect of a commonly used opioid, buprenorphine, on the initiation and progression of AP. For this, we administered extended-release buprenorphine subcutaneously before inducing the previously established severe AP model that uses interleukins 12 and 18 (IL12,18) in genetically obese (ob/ob) mice and compared this to mice with AP but without the drug. Mice were monitored over 3 days, and parameters of AP induction and progression were compared. Buprenorphine significantly reduced serum amylase, lipase, pancreatic necrosis, and AP-associated fat necrosis, which is ubiquitous in obese mice and humans. Buprenorphine delayed the AP-associated reduction of carotid artery pulse distention and the development of hypothermia, hastened renal injury, and muted the early increase in respiratory rate versus IL12,18 alone. The site of buprenorphine injection appeared erythematous, inflamed, and microscopically showed thinning, loss of epidermal layers that had increased apoptosis. In summary, subcutaneous extended-release buprenorphine interfered with the induction of AP by reducing serum amylase, lipase, pancreatic and fat necrosis, the worsening of AP by delaying hypotension, hypothermia, while hastening renal injury, respiratory depression, and causing cutaneous injury at the site of injection.NEW & NOTEWORTHY Extended-release buprenorphine interferes with the initiation and progression of acute pancreatitis at multiple levels.
Collapse
Affiliation(s)
- Sarah Jahangir
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Biswajit Khatua
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Nabil Smichi
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | | | | | - Megan J Summers
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Bryce McFayden
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Sergiy Kostenko
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Naomi M Gades
- Department of Comparative Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| | - Vijay P Singh
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, United States
| |
Collapse
|
3
|
Chung MJ, Park SW, Lee KJ, Park DH, Koh DH, Lee J, Lee HS, Park JY, Bang S, Min S, Park JH, Kim SJ, Park CH. Clinical impact of pancreatic steatosis measured by CT on the risk of post-ERCP pancreatitis: a multicenter prospective trial. Gastrointest Endosc 2024; 99:214-223.e4. [PMID: 37598866 DOI: 10.1016/j.gie.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND AND AIMS Pancreatic steatosis (PS) may be a risk factor for acute pancreatitis. Whether it is also a risk factor for post-ERCP pancreatitis (PEP) has not been evaluated. This study aimed to determine the impact of PS on PEP development. METHODS This multicenter prospective trial enrolled 786 consecutive patients who underwent contrast-enhanced abdominal CT and subsequent first-time ERCP. PS was evaluated based on pancreatic attenuation on unenhanced CT images. The risk of PS for the development of PEP was evaluated using a logistic regression model. RESULTS Of 527 patients included in the study, 157 (29.8%) had PS and 370 (70.2%) did not. At 24 hours after ERCP, there was a significant difference in the PEP identified in 22 patients (14.0%) in the PS group and 23 patients (6.2%) in the "no PS" (NPS) group (P = .017). Diabetes and hypertension were more common in the PS group than in the NPS group; no differences in dyslipidemia were found. Patients with PS had a higher risk for the development of PEP than those with NPS (odds ratio, 2.09; 95% confidence interval, 1.08-4.03). No other variables were identified as risk factors for PEP. CONCLUSIONS PS is a significant risk factor for PEP for which preventive measures should be considered. Standardized measurement protocols to assess PS by CT are needed. (Clinical trial registration number: KCT0006068.).
Collapse
Affiliation(s)
- Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Kyong Joo Lee
- Division of Gastroenterology, Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Da Hae Park
- Division of Gastroenterology, Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Dong Hee Koh
- Division of Gastroenterology, Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Jin Lee
- Division of Gastroenterology, Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Hee Seung Lee
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seonjeong Min
- Department of Radiology, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Ji Hoon Park
- Division of Gastroenterology, Department of Internal Medicine, CHA Ilsan Medical Center, CHA University, Goyang, Republic of Korea
| | - So Jeong Kim
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chan Hyuk Park
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| |
Collapse
|
4
|
Bannone E, Pulvirenti A, Marchegiani G, Vacca PG, Marchetti A, Cattelani A, Salvia R, Bassi C. No role for protease inhibitors as a mitigation strategy for postpancreatectomy acute pancreatitis (PPAP): Propensity score matching analysis. Pancreatology 2023; 23:904-910. [PMID: 37839921 DOI: 10.1016/j.pan.2023.09.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND While the use of protease inhibitor gabexate mesylate (GM) is still controversial in acute pancreatitis, it has never been tested for postpancreatectomy acute pancreatitis (PPAP). This study aims to assess the impact of GM on postoperative serum hyperamylasaemia (POH) or PPAP after pancreatoduodenectomy (PD). METHODS Consecutive patients developing POH after PD between 2016 and 2021 were included. According to GM administration, patients were divided into GM-treated and control (CTR) groups. GM was administered from postoperative day 1-3 in POH patients who underwent surgery before 2017. A 2:1 propensity matching was used to minimize the risk of bias. RESULTS Overall, 264 patients with POH were stratified in the GM (59 patients) and CTR (104 patients) cohorts, which showed balanced baseline characteristics after matching. No difference in postoperative complications was observed between the groups (all p > 0.05), except for PPAP occurrence, which was significantly higher in the GM group (37% vs. 22%, p = 0.037). A total of 45 patients (28%) evolved to PPAP. Comparing PPAP patients in the GM and CTR groups, no significant differences in POPF, relaparotomy, and mortality (all p > 0.09) were found. No difference in intravenous crystalloid administration was found in patients with PPAP, whether or not they developed major complications or pancreatic fistula (p > 0.05) CONCLUSION: Protease inhibitor seems ineffective in preventing a PPAP after PD once a POH has occurred. Further studies are needed to achieve benchmarks for treating PPAP and identify mitigation strategies to prevent the evolution of POH into additional morbidity.
Collapse
Affiliation(s)
- Elisa Bannone
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona Hospital Trust, Verona, Italy; Department of General Surgery, Poliambulanza Foundation Hospital, Brescia, Italy. https://twitter.com/PancreasVerona
| | - Alessandra Pulvirenti
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona Hospital Trust, Verona, Italy
| | - Giovanni Marchegiani
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona Hospital Trust, Verona, Italy
| | - Pier Giuseppe Vacca
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona Hospital Trust, Verona, Italy
| | - Alessio Marchetti
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona Hospital Trust, Verona, Italy
| | - Alice Cattelani
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona Hospital Trust, Verona, Italy
| | - Roberto Salvia
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona Hospital Trust, Verona, Italy
| | - Claudio Bassi
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona Hospital Trust, Verona, Italy
| |
Collapse
|
5
|
Kostenko S, Khatua B, Trivedi S, Pillai AN, McFayden B, Morsy M, Rajalingamgari P, Sharma V, Noel P, Patel K, El-Kurdi B, Borges da Silva H, Chen X, Chandan V, Navina S, Vela S, Cartin-Ceba R, Snozek C, Singh VP. Amphipathic Liponecrosis Impairs Bacterial Clearance and Causes Infection During Sterile Inflammation. Gastroenterology 2023; 165:999-1015. [PMID: 37263302 DOI: 10.1053/j.gastro.2023.05.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND & AIMS Although transient bacteremia is common during dental and endoscopic procedures, infections developing during sterile diseases like acute pancreatitis (AP) can have grave consequences. We examined how impaired bacterial clearance may cause this transition. METHODS Blood samples from patients with AP, normal controls, and rodents with pancreatitis or those administered different nonesterified fatty acids (NEFAs) were analyzed for albumin-unbound NEFAs, microbiome, and inflammatory cell injury. Macrophage uptake of unbound NEFAs using a novel coumarin tracer were done and the downstream effects-NEFA-membrane phospholipid (phosphatidylcholine) interactions-were studied on isothermal titration calorimetry. RESULTS Patients with infected AP had higher circulating unsaturated NEFAs; unbound NEFAs, including linoleic acid (LA) and oleic acid (OA); higher bacterial 16S DNA; mitochondrial DNA; altered β-diversity; enrichment in Pseudomonadales; and increased annexin V-positive myeloid (CD14) and CD3-positive T cells on admission. These, and increased circulating dead inflammatory cells, were also noted in rodents with unbound, unsaturated NEFAs. Isothermal titration calorimetry showed progressively stronger unbound LA interactions with aqueous media, phosphatidylcholine, cardiolipin, and albumin. Unbound NEFAs were taken into protein-free membranes, cells, and mitochondria, inducing voltage-dependent anion channel oligomerization, reducing ATP, and impairing phagocytosis. These were reversed by albumin. In vivo, unbound LA and OA increased bacterial loads and impaired phagocytosis, causing infection. LA and OA were more potent for these amphipathic interactions than the hydrophobic palmitic acid. CONCLUSIONS Release of stored LA and OA can increase their circulating unbound levels and cause amphipathic liponecrosis of immune cells via uptake by membrane phospholipids. This impairs bacterial clearance and causes infection during sterile inflammation.
Collapse
Affiliation(s)
| | | | | | | | - Bryce McFayden
- Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Mahmoud Morsy
- Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Vijeta Sharma
- Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Pawan Noel
- Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Krutika Patel
- Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Bara El-Kurdi
- Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Xianfeng Chen
- Department of Research Services, Mayo Clinic, Rochester, Minnesota
| | - Vishal Chandan
- Department of Pathology, School of Medicine, University of California, Irvine, California
| | | | - Stacie Vela
- Gastroenterology Section, Carl T. Hayden Veterans' Administration Medical Center, Phoenix, Arizona
| | - Rodrigo Cartin-Ceba
- Department of Pulmonary and Critical Care Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Christine Snozek
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona
| | - Vijay P Singh
- Department of Medicine, Mayo Clinic, Rochester, Minnesota; Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, Arizona.
| |
Collapse
|
6
|
Mareninova OA, Gretler SR, Lee GE, Pimienta M, Qin Y, Elperin JM, Ni J, Razga Z, Gukovskaya AS, Gukovsky I. Ethanol inhibits pancreatic acinar cell autophagy through upregulation of ATG4B, mediating pathological responses of alcoholic pancreatitis. Am J Physiol Gastrointest Liver Physiol 2023; 325:G265-G278. [PMID: 37431575 PMCID: PMC10511161 DOI: 10.1152/ajpgi.00053.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/23/2023] [Accepted: 07/01/2023] [Indexed: 07/12/2023]
Abstract
Excessive alcohol intake is a major risk factor for pancreatitis, sensitizing the exocrine pancreas to stressors by mechanisms that remain obscure. Impaired autophagy drives nonalcoholic pancreatitis, but the effects of ethanol (EtOH) and alcoholic pancreatitis on autophagy are poorly understood. Here, we find that ethanol reduces autophagosome formation in pancreatic acinar cells, both in a mouse model of alcoholic pancreatitis induced by a combination of EtOH diet and cerulein (a CCK ortholog) and in EtOH+CCK-treated acinar cells (ex vivo model). Ethanol treatments decreased pancreatic level of LC3-II, a key mediator of autophagosome formation. This was caused by ethanol-induced upregulation of ATG4B, a cysteine protease that, cell dependently, regulates the balance between cytosolic LC3-I and membrane-bound LC3-II. We show that ATG4B negatively regulates LC3-II in acinar cells subjected to EtOH treatments. Ethanol raised ATG4B level by inhibiting its degradation, enhanced ATG4B enzymatic activity, and strengthened its interaction with LC3-II. We also found an increase in ATG4B and impaired autophagy in a dissimilar, nonsecretagogue model of alcoholic pancreatitis induced by EtOH plus palmitoleic acid. Adenoviral ATG4B overexpression in acinar cells greatly reduced LC3-II and inhibited autophagy. Furthermore, it aggravated trypsinogen activation and necrosis, mimicking key responses of ex vivo alcoholic pancreatitis. Conversely, shRNA Atg4B knockdown enhanced autophagosome formation and alleviated ethanol-induced acinar cell damage. The results reveal a novel mechanism, whereby ethanol inhibits autophagosome formation and thus sensitizes pancreatitis, and a key role of ATG4B in ethanol's effects on autophagy. Enhancing pancreatic autophagy, particularly by downregulating ATG4B, could be beneficial in mitigating the severity of alcoholic pancreatitis.NEW & NOTEWORTHY Ethanol sensitizes mice and humans to pancreatitis, but the underlying mechanisms remain obscure. Autophagy is important for maintaining pancreatic acinar cell homeostasis, and its impairment drives pancreatitis. This study reveals a novel mechanism, whereby ethanol inhibits autophagosome formation through upregulating ATG4B, a key cysteine protease. ATG4B upregulation inhibits autophagy in acinar cells and aggravates pathological responses of experimental alcoholic pancreatitis. Enhancing pancreatic autophagy, particularly by down-regulating ATG4B, could be beneficial for treatment of alcoholic pancreatitis.
Collapse
Affiliation(s)
- Olga A Mareninova
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, California, United States
| | - Sophie R Gretler
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, California, United States
| | - Grace E Lee
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
| | - Michael Pimienta
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
| | - Yueqiu Qin
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- Division of Gastroenterology and Hepatology, Youjiang Medical University for Nationalities, Baise, China
| | - Jason M Elperin
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
| | - Jinliang Ni
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Zsolt Razga
- Institute of Pathology, University of Szeged, Szeged, Hungary
| | - Anna S Gukovskaya
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, California, United States
| | - Ilya Gukovsky
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, California, United States
| |
Collapse
|
7
|
Nitesh P, Biju P, Kalayarasan R, Krishna PS. Necrotising Inflammation of Peri-Pancreatic Tissue With Normal Appearance of Pancreas Is a Distinct Entity of Acute Pancreatitis: Report of Two Cases With Review of Literature. Cureus 2023; 15:e43075. [PMID: 37680405 PMCID: PMC10481991 DOI: 10.7759/cureus.43075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
Improved insights into the pathophysiology of acute pancreatitis have paved the way for identification of distinct entities in the spectrum of the disease. The presence of necrotising inflammation limited to peripancreatic tissue with a normal appearance of pancreas is one such entity. This entity, described as extrapancreatic necrotising pancreatitis (EPN), is considered a less aggressive form of acute necrotising pancreatitis. This entity needs to be recognized precisely and managed accordingly among patients with acute pancreatitis. However, EPN has not been highlighted in the revised classification of acute pancreatitis. Here we report two patients with EPN with varied presentations and diverse management and outcome.
Collapse
Affiliation(s)
- Pagadala Nitesh
- Surgical Gastroenterology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Pottakkat Biju
- Surgical Gastroenterology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Raja Kalayarasan
- Surgical Gastroenterology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Pothugunta S Krishna
- Surgical Gastroenterology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| |
Collapse
|
8
|
Vela S, Guerra A, Farrell G, Trivedi S, Chaffin H, Rood C, Singh R, Kostenko S, Chang YH, Snozek C, Patel K, Khatua B, Singh VP. Pathophysiology and Biomarker Potential of Fatty Acid Ethyl Ester Elevation During Alcoholic Pancreatitis. Gastroenterology 2021; 161:1513-1525. [PMID: 34303660 PMCID: PMC9318056 DOI: 10.1053/j.gastro.2021.07.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/07/2021] [Accepted: 07/16/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS The role of fatty acid ethyl esters (FAEEs) during human alcoholic pancreatitis is unknown. We compared FAEEs levels with their nonesterified fatty acids (NEFAs) precursors during alcohol intoxication and clinical alcoholic pancreatitis. The pathophysiology underlying FAEEs increase and their role as diagnostic biomarkers for alcoholic pancreatitis was investigated. METHODS A prospective blinded study compared FAEEs, NEFAs, and ethanol blood levels on hospitalization for alcoholic pancreatitis (n = 31), alcohol intoxication (n = 25), and in normal controls (n = 43). Serum FAEEs were measured at admission for nonalcoholic pancreatitis (n = 75). Mechanistic cell and animal studies were done. RESULTS Median FAEEs were similarly elevated during alcohol intoxication (205 nmol/L; 95% confidence interval [CI], 71.8-515 nmol/L, P < .001) and alcoholic pancreatitis (103.1 nmol/L; 95% CI, 53-689 nmol/L, P < .001) vs controls (1.7 nmol/L; 95% CI, 0.02-4.3 nmol/L) or nonalcoholic pancreatitis (8 nmol/L; 95% CI, 1.1-11.5 nmol/L). Alcoholic pancreatitis increased serum NEFAs (1024 ± 710 μmol/L vs 307 ± 185 μmol/L in controls, P < .05). FAEEs comprised 0.1% to 2% of the parent NEFA concentrations. FAEES correlated strongly with NEFAs independent of ethanol levels in alcoholic pancreatitis but not during alcohol intoxication. On receiver operating characteristic curve analysis for diagnosing alcoholic pancreatitis, the area under the curve for serum FAEEs was 0.87 (95% CI, 0.78-0.95, P < .001). In mice and cells, alcohol administration transiently increased all FAEEs. Oleic acid ethyl ester was the only FAEE with a sustained increase up to 24 hours after intraperitoneal oleic acid plus ethanol administration. CONCLUSIONS The sustained, alcohol-independent, large (20- to 50-fold) increase in circulating FAEEs during alcoholic pancreatitis results from their visceral release and mirrors the 2- to 4-fold increase in parent NEFA. The large areas under the curve of FAEEs on receiver operating characteristic curve analysis supports their role as alcoholic pancreatitis biomarkers.
Collapse
Affiliation(s)
- Stacie Vela
- Gastroenterology Section, Carl T. Hayden Veterans’ Administration Medical Center, Phoenix, AZ
| | - Andre Guerra
- Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Gail Farrell
- Gastroenterology Section, Carl T. Hayden Veterans’ Administration Medical Center, Phoenix, AZ
| | | | | | | | - Ravinder Singh
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Yu-Hui Chang
- Department of Biostatistics, Mayo Clinic, Scottsdale, AZ
| | - Christine Snozek
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ
| | | | | | - Vijay P. Singh
- Department of Medicine, Mayo Clinic, Scottsdale, AZ,Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona
| |
Collapse
|
9
|
de Oliveira C, Khatua B, Noel P, Kostenko S, Bag A, Balakrishnan B, Patel KS, Guerra AA, Martinez MN, Trivedi S, McCullough A, Lam-Himlin DM, Navina S, Faigel DO, Fukami N, Pannala R, Phillips AE, Papachristou GI, Kershaw EE, Lowe ME, Singh VP. Pancreatic triglyceride lipase mediates lipotoxic systemic inflammation. J Clin Invest 2020; 130:1931-1947. [PMID: 31917686 DOI: 10.1172/jci132767] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 01/03/2020] [Indexed: 12/22/2022] Open
Abstract
Visceral adipose tissue plays a critical role in numerous diseases. Although imaging studies often show adipose involvement in abdominal diseases, their outcomes may vary from being a mild self-limited illness to one with systemic inflammation and organ failure. We therefore compared the pattern of visceral adipose injury during acute pancreatitis and acute diverticulitis to determine its role in organ failure. Acute pancreatitis-associated adipose tissue had ongoing lipolysis in the absence of adipocyte triglyceride lipase (ATGL). Pancreatic lipase injected into mouse visceral adipose tissue hydrolyzed adipose triglyceride and generated excess nonesterified fatty acids (NEFAs), which caused organ failure in the absence of acute pancreatitis. Pancreatic triglyceride lipase (PNLIP) increased in adipose tissue during pancreatitis and entered adipocytes by multiple mechanisms, hydrolyzing adipose triglyceride and generating excess NEFAs. During pancreatitis, obese PNLIP-knockout mice, unlike obese adipocyte-specific ATGL knockouts, had lower visceral adipose tissue lipolysis, milder inflammation, less severe organ failure, and improved survival. PNLIP-knockout mice, unlike ATGL knockouts, were protected from adipocyte-induced pancreatic acinar injury without affecting NEFA signaling or acute pancreatitis induction. Therefore, during pancreatitis, unlike diverticulitis, PNLIP leaking into visceral adipose tissue can cause excessive visceral adipose tissue lipolysis independently of adipocyte-autonomous ATGL, and thereby worsen organ failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Ann McCullough
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Dora M Lam-Himlin
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona, USA
| | | | | | | | | | - Anna Evans Phillips
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Erin E Kershaw
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mark E Lowe
- Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, USA
| | | |
Collapse
|
10
|
Mareninova OA, Jia W, Gretler SR, Holthaus CL, Thomas DDH, Pimienta M, Dillon DL, Gukovskaya AS, Gukovsky I, Groblewski GE. Transgenic expression of GFP-LC3 perturbs autophagy in exocrine pancreas and acute pancreatitis responses in mice. Autophagy 2020; 16:2084-2097. [PMID: 31942816 PMCID: PMC7595606 DOI: 10.1080/15548627.2020.1715047] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 12/25/2019] [Accepted: 01/08/2020] [Indexed: 12/20/2022] Open
Abstract
Pancreatitis is a common, sometimes fatal, disease of exocrine pancreas, initiated by damaged acinar cells. Recent studies implicate disordered macroautophagy/autophagy in pancreatitis pathogenesis. ATG8/LC3 protein is critical for autophagosome formation and a widely used marker of autophagic vacuoles. Transgenic GFP-LC3 mice are a valuable tool to investigate autophagy ; however, comparison of homeostatic and disease responses between GFP-LC3 and wild-type (WT) mice has not been done. We examined the effects of GFP-LC3 expression on autophagy, acinar cell function, and experimental pancreatitis. Unexpectedly, GFP-LC3 expression markedly increased endogenous LC3-II level in pancreas, caused by downregulation of ATG4B, the protease that deconjugates/delipidates LC3-II. By contrast, GFP-LC3 expression had lesser or no effect on autophagy in liver, lung and spleen. Autophagic flux analysis showed that autophagosome formation in GFP-LC3 acinar cells increased 3-fold but was not fully counterbalanced by increased autophagic degradation. Acinar cell (ex vivo) pancreatitis inhibited autophagic flux in WT and essentially blocked it in GFP-LC3 cells. In vivo pancreatitis caused autophagy impairment in WT mice, manifest by upregulation of LC3-II and SQSTM1/p62, increased number and size of autophagic vacuoles, and decreased level of TFEB, all of which were exacerbated in GFP-LC3 mice. GFP-LC3 expression affected key pancreatitis responses; most dramatically, it worsened increases in serum AMY (amylase), a diagnostic marker of acute pancreatitis, in several mouse models. The results emphasize physiological importance of autophagy for acinar cell function, demonstrate organ-specific effects of GFP-LC3 expression, and indicate that application of GFP-LC3 mice in disease models should be done with caution.Abbreviations: AP: acute pancreatitis; Arg-AP: L-arginine-induced acute pancreatitis; ATG: autophagy-related (protein); AVs: autophagic vacuoles; CCK: cholecystokinin-8; CDE: choline-deficient, D,L-ethionine supplemented diet; CER: caerulein (ortholog of CCK); CTSB: cathepsin B; CTSD: cathepsin D; CTSL: cathepsin L; ER: endoplasmic reticulum; LAMP: lysosomal-associated membrane protein; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; TEM: transmission electron microscopy; TFEB: transcription factor EB; ZG: zymogen granule(s).
Collapse
Affiliation(s)
- Olga A. Mareninova
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
- Pancreatic Research Group, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Wenzhuo Jia
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
- Department of General Surgery, Beijing Hospital, National Centre of Gerontology, Beijing, China
| | - Sophie R. Gretler
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
- Pancreatic Research Group, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Conner L. Holthaus
- Department of Nutritional Sciences, University of Wisconsin, Madison, WI, USA
| | - Diana D. H. Thomas
- Department of Nutritional Sciences, University of Wisconsin, Madison, WI, USA
| | - Michael Pimienta
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Dustin L. Dillon
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
- Pancreatic Research Group, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Anna S. Gukovskaya
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
- Pancreatic Research Group, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Ilya Gukovsky
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
- Pancreatic Research Group, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Guy E. Groblewski
- Department of Nutritional Sciences, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
11
|
Nonalcoholic fatty pancreas disease is related independently to the severity of acute pancreatitis. Eur J Gastroenterol Hepatol 2019; 31:973-978. [PMID: 31233410 DOI: 10.1097/meg.0000000000001477] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND This study aimed to investigate the association between nonalcoholic fatty pancreas disease and the severity of acute pancreatitis (AP). PATIENTS AND METHODS Among the 1662 AP patients admitted between August 2010 and August 2017, 82 eligible patients with moderately severe acute pancreatitis (SAP) and SAP were selected. Meanwhile, 164 mild AP patients were age-matched, sex-matched, and BMI-matched at a ratio of 1 : 2. Nonalcoholic fatty pancreas disease was estimated by mean pancreas attenuation by unenhanced computed tomography. Finally, 1662 patients were screened and 246 patients were analyzed. RESULTS For the 246 patients, the mean pancreatic attenuation and pancreas-to-spleen attenuation ratio (P/S ratio) were significantly lower in the moderately SAP and SAP groups compared with those in the mild AP group (both, P<0.001). Pancreatic attenuation decreased with an increase in the rate of ICU transfer, AP severity, systemic complications, and prognostic factors of AP (Acute Physiology and Chronic Health Evaluation II score≥8; P<0.001). A decreased P/S ratio was correlated positively with the increased mortality of patients with AP (hazard ratio: 0.000; 95% confidence interval: 0.000-0.012; P<0.001), as determined by Cox proportional regression analysis adjusted for creatinine, calcium, and albumin levels. CONCLUSION The pancreatic attenuation level and P/S ratio are correlated independently to severity, mortality, and systemic complications in patients with AP.
Collapse
|
12
|
Zhan X, Wan J, Zhang G, Song L, Gui F, Zhang Y, Li Y, Guo J, Dawra RK, Saluja AK, Haddock AN, Zhang L, Bi Y, Ji B. Elevated intracellular trypsin exacerbates acute pancreatitis and chronic pancreatitis in mice. Am J Physiol Gastrointest Liver Physiol 2019; 316:G816-G825. [PMID: 30943050 PMCID: PMC6620583 DOI: 10.1152/ajpgi.00004.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 01/31/2023]
Abstract
Intra-acinar trypsinogen activation occurs in the earliest stages of pancreatitis and is believed to play important roles in pancreatitis pathogenesis. However, the exact role of intra-acinar trypsin activity in pancreatitis remains elusive. Here, we aimed to examine the specific effects of intra-acinar trypsin activity on the development of pancreatitis using a transgenic mouse model. This transgenic mouse model allowed for the conditional expression of a mutant trypsinogen that can be activated specifically inside pancreatic acinar cells. We found that expression of this active mutated trypsin had no significant effect on triggering spontaneous pancreatitis. Instead, several protective compensatory mechanisms, including SPINK1 and heat shock proteins, were upregulated. Notably, these transgenic mice developed much more severe acute pancreatitis, compared with control mice, when challenged with caerulein. Elevated tissue edema, serum amylase, inflammatory cell infiltration and acinar cell apoptosis were dramatically associated with increased trypsin activity. Furthermore, chronic pathological changes were observed in the pancreas of all transgenic mice, including inflammatory cell infiltration, parenchymal atrophy and cell loss, fibrosis, and fatty replacement. These changes were not observed in control mice treated with caerulein. The alterations in pancreata from transgenic mice mimicked the histological changes common to human chronic pancreatitis. Taken together, we provided in vivo evidence that increased intra-acinar activation of trypsinogen plays an important role in the initiation and progression of both acute and chronic pancreatitis. NEW & NOTEWORTHY Trypsinogen is activated early in pancreatitis. However, the roles of trypsin in the development of pancreatitis have not been fully addressed. Using a genetic approach, we showed trypsin activity is critical for the severity of both acute and chronic pancreatitis.
Collapse
Affiliation(s)
- Xianbao Zhan
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
- Department of Oncology, Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Jianhua Wan
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Guowei Zhang
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University , Guangzhou , China
| | - Lele Song
- Department of Oncology, Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Fu Gui
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Yuebo Zhang
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Yinghua Li
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Jia Guo
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Rajinder K Dawra
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami , Miami, Florida
| | - Ashok K Saluja
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami , Miami, Florida
| | - Ashley N Haddock
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| | - Lizhi Zhang
- Department of Pathology, Mayo Clinic , Rochester, Minnesota
| | - Yan Bi
- Department of Gastroenterology and Hepatology, Mayo Clinic , Jacksonville, Florida
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic , Jacksonville, Florida
| |
Collapse
|
13
|
Saluja A, Dudeja V, Dawra R, Sah RP. Early Intra-Acinar Events in Pathogenesis of Pancreatitis. Gastroenterology 2019; 156:1979-1993. [PMID: 30776339 DOI: 10.1053/j.gastro.2019.01.268] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/09/2019] [Accepted: 01/21/2019] [Indexed: 12/11/2022]
Abstract
Premature activation of digestive enzymes in the pancreas has been linked to development of pancreatitis for more than a century. Recent development of novel models to study the role of pathologic enzyme activation has led to advances in our understanding of the mechanisms of pancreatic injury. Colocalization of zymogen and lysosomal fraction occurs early after pancreatitis-causing stimulus. Cathepsin B activates trypsinogen in these colocalized organelles. Active trypsin increases permeability of these organelles resulting in leakage of cathepsin B into the cytosol leading to acinar cell death. Although trypsin-mediated cell death leads to pancreatic injury in early stages of pancreatitis, multiple parallel mechanisms, including activation of inflammatory cascades, endoplasmic reticulum stress, autophagy, and mitochondrial dysfunction in the acinar cells are now recognized to be important in driving the profound systemic inflammatory response and extensive pancreatic injury seen in acute pancreatitis. Chymotrypsin, another acinar protease, has recently been shown be play critical role in clearance of pathologically activated trypsin protecting against pancreatic injury. Mutations in trypsin and other genes thought to be associated with pathologic enzyme activation (such as serine protease inhibitor 1) have been found in familial forms of pancreatitis. Sustained intra-acinar activation of nuclear factor κB pathway seems to be key pathogenic mechanism in chronic pancreatitis. Better understanding of these mechanisms will hopefully allow us to improve treatment strategies in acute and chronic pancreatitis.
Collapse
|
14
|
Vrolyk V, Schneberger D, Le K, Wobeser BK, Singh B. Mouse model to study pulmonary intravascular macrophage recruitment and lung inflammation in acute necrotizing pancreatitis. Cell Tissue Res 2019; 378:97-111. [DOI: 10.1007/s00441-019-03023-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 03/27/2019] [Indexed: 12/18/2022]
|
15
|
Khatua B, Trivedi RN, Noel P, Patel K, Singh R, de Oliveira C, Trivedi S, Mishra V, Lowe M, Singh VP. Carboxyl Ester Lipase May Not Mediate Lipotoxic Injury during Severe Acute Pancreatitis. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1226-1240. [PMID: 30954473 DOI: 10.1016/j.ajpath.2019.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/19/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
Abstract
Acute lipolysis of visceral fat or circulating triglycerides may worsen acute pancreatitis (AP)-associated local and systemic injury. The pancreas expresses pancreatic triacylglycerol lipase (PNLIP), pancreatic lipase-related protein 2 (PNLIPRP2), and carboxyl ester lipase (CEL), which may leak into the visceral fat or systemic circulation during pancreatitis. We, thus, aimed to determine the pancreatic lipase(s) regulating lipotoxicity during AP. For this AP, associated fat necrosis was analyzed using Western blot analysis. Bile acid (using liquid chromatography-tandem mass spectrometry) and fatty acid (using gas chromatography) concentrations were measured in human fat necrosis. The fat necrosis milieu was simulated in vitro using glyceryl trilinoleate because linoleic acid is increased in fat necrosis. Bile acid requirements to effectively hydrolyze glyceryl trilinoleate were studied using exogenous or overexpressed lipases. The renal cell line (HEK 293) was used to study lipotoxic injury. Because dual pancreatic lipase knockouts are lethal, exocrine parotid acini lacking lipases were used to verify the results. PNLIP, PNLIPRP2, and CEL were increased in fat necrosis. Although PNLIP and PNLIPRP2 were equipotent in inducing lipolysis and lipotoxic injury, CEL required bile acid concentrations higher than in human fat necrosis. The high bile acid requirements for effective lipolysis make CEL an unlikely mediator of lipotoxic injury in AP. It remains to be explored whether PNLIP or PNLIPRP2 worsens AP severity in vivo.
Collapse
Affiliation(s)
| | - Ram N Trivedi
- Department of Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Pawan Noel
- Department of Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Krutika Patel
- Department of Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Ravinder Singh
- Department of Lab Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Vivek Mishra
- Department of Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Mark Lowe
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Vijay P Singh
- Department of Medicine, Mayo Clinic, Scottsdale, Arizona.
| |
Collapse
|
16
|
Quan S, Principe DR, Dean AE, Park SH, Grippo PJ, Gius D, Horikoshi N. Loss of Sirt2 increases and prolongs a caerulein-induced pancreatitis permissive phenotype and induces spontaneous oncogenic Kras mutations in mice. Sci Rep 2018; 8:16501. [PMID: 30405152 PMCID: PMC6220268 DOI: 10.1038/s41598-018-34792-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
Mice lacking Sirt2 spontaneously develop tumors in multiple organs, as well as when expressed in combination with oncogenic KrasG12D, leading to pancreatic tumors. Here, we report that after caerulein-induced pancreatitis, Sirt2-deficient mice exhibited an increased inflammatory phenotype and delayed pancreatic tissue recovery. Seven days post injury, the pancreas of Sirt2-/- mice display active inflammation, whereas wild-type mice had mostly recovered. In addition, the pancreas from the Sirt2-/- mice exhibited extensive tissue fibrosis, which was still present at six weeks after exposure. The mice lacking Sirt2 also demonstrated an enhanced whole body pro-inflammatory phenotype that was most obvious with increasing age. Importantly, an accumulation of a cell population with spontaneous cancerous KrasG12D mutations was observed in the Sirt2-/- mice that is enhanced in the recovering pancreas after exposure to caerulein. Finally, transcriptome analysis of the pancreas of the Sirt2-/- mice exhibited a pro-inflammatory genomic signature. These results suggest that loss of Sirt2, as well as increased age, enhanced the immune response to pancreatic injury and induced an inflammatory phenotype permissive for the accumulation of cells carrying oncogenic Kras mutations.
Collapse
Affiliation(s)
- Songhua Quan
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Daniel R Principe
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Angela E Dean
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Seong-Hoon Park
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of General and Applied Toxicology, Genetic Toxicology Research Group, Korea Institute of Toxicology (KIT), Daejeon, South Korea
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - David Gius
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Pharmacology, Robert Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Nobuo Horikoshi
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
17
|
Boggs K, Wang T, Orabi AI, Mukherjee A, Eisses JF, Sun T, Wen L, Javed TA, Esni F, Chen W, Husain SZ. Pancreatic gene expression during recovery after pancreatitis reveals unique transcriptome profiles. Sci Rep 2018; 8:1406. [PMID: 29362419 PMCID: PMC5780441 DOI: 10.1038/s41598-018-19392-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 12/29/2017] [Indexed: 02/07/2023] Open
Abstract
It is well known that pancreatic recovery after a single episode of injury such as an isolated bout of pancreatitis occurs rapidly. It is unclear, however, what changes are inflicted in such conditions to the molecular landscape of the pancreas. In the caerulein hyperstimulation model of pancreatitis, the murine pancreas has the ability to recover within one week based on histological appearance. In this study, we sought to characterize by RNA-sequencing (RNA-seq) the transcriptional profile of the recovering pancreas up to two weeks post-injury. We found that one week after injury there were 319 differentially expressed genes (DEGs) compared with baseline and that after two weeks there were 53 DEGs. Forty (12.5%) of the DEGs persisted from week one to week two, and another 13 DEGs newly emerged in the second week. Amongst the top up-regulated DEGs were several trypsinogen genes (trypsinogen 4, 5, 12, 15, and 16). To our knowledge, this is the first characterization of the transcriptome during pancreatic recovery by deep sequencing, and it reveals on a molecular basis that there is an ongoing recovery of the pancreas even after apparent histological resolution. The findings also raise the possibility of an emerging novel transcriptome upon pancreatic recovery.
Collapse
Affiliation(s)
- Kristy Boggs
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Ting Wang
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Abrahim I Orabi
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Amitava Mukherjee
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - John F Eisses
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Tao Sun
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Li Wen
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Tanveer A Javed
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Farzad Esni
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Wei Chen
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Sohail Z Husain
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
18
|
Abstract
The metabolic consequences of visceral fat deposition are well known, and the presence of intrapancreatic fat (IPF) has been recognized for decades. However, our knowledge about the distribution of fat in the pancreas and its clinical implications is in a nascent stage. Various terms have been proposed to describe IPF; for the purpose of this narrative review, we chose the general term fatty pancreas. Herein, we describe the radiologic, endoscopic, and histopathologic aspects of diagnosing fatty pancreas and provide an overview of the diseases associated with this condition. Our purpose is to highlight diagnostic challenges and identify specific clinical questions that would benefit from further study. As evident in this review, IPF is associated with various metabolic diseases, pancreatitis, pancreatic cancer, and precancer-yet establishing causality needs careful, further study.
Collapse
|
19
|
Noel P, Patel K, Durgampudi C, Trivedi RN, de Oliveira C, Crowell MD, Pannala R, Lee K, Brand R, Chennat J, Slivka A, Papachristou GI, Khalid A, Whitcomb DC, DeLany JP, Cline RA, Acharya C, Jaligama D, Murad FM, Yadav D, Navina S, Singh VP. Peripancreatic fat necrosis worsens acute pancreatitis independent of pancreatic necrosis via unsaturated fatty acids increased in human pancreatic necrosis collections. Gut 2016; 65:100-11. [PMID: 25500204 PMCID: PMC4869971 DOI: 10.1136/gutjnl-2014-308043] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 11/17/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Peripancreatic fat necrosis occurs frequently in necrotising pancreatitis. Distinguishing markers from mediators of severe acute pancreatitis (SAP) is important since targeting mediators may improve outcomes. We evaluated potential agents in human pancreatic necrotic collections (NCs), pseudocysts (PCs) and pancreatic cystic neoplasms and used pancreatic acini, peripheral blood mononuclear cells (PBMC) and an acute pancreatitis (AP) model to determine SAP mediators. METHODS We measured acinar and PBMC injury induced by agents increased in NCs and PCs. Outcomes of caerulein pancreatitis were studied in lean rats coadministered interleukin (IL)-1β and keratinocyte chemoattractant/growth-regulated oncogene, triolein alone or with the lipase inhibitor orlistat. RESULTS NCs had higher fatty acids, IL-8 and IL-1β versus other fluids. Lipolysis of unsaturated triglyceride and resulting unsaturated fatty acids (UFA) oleic and linoleic acids induced necro-apoptosis at less than half the concentration in NCs but other agents did not do so at more than two times these concentrations. Cytokine coadministration resulted in higher pancreatic and lung inflammation than caerulein alone, but only triolein coadministration caused peripancreatic fat stranding, higher cytokines, UFAs, multisystem organ failure (MSOF) and mortality in 97% animals, which were prevented by orlistat. CONCLUSIONS UFAs, IL-1β and IL-8 are elevated in NCs. However, UFAs generated via peripancreatic fat lipolysis causes worse inflammation and MSOF, converting mild AP to SAP.
Collapse
Affiliation(s)
- Pawan Noel
- Departments of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Krutika Patel
- Departments of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Chandra Durgampudi
- Departments of Medicine, University of Pittsburgh Medical Center, Pasavant, Pennsylvania, USA
| | - Ram N Trivedi
- Departments of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | | | | | - Rahul Pannala
- Departments of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Kenneth Lee
- Departments of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Randall Brand
- Departments of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jennifer Chennat
- Departments of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adam Slivka
- Departments of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Asif Khalid
- Departments of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David C Whitcomb
- Departments of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - James P DeLany
- Departments of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rachel A Cline
- Departments of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chathur Acharya
- Departments of Medicine, University of Pittsburgh Medical Center, Pasavant, Pennsylvania, USA
| | - Deepthi Jaligama
- Departments of Medicine, University of Pittsburgh Medical Center, Pasavant, Pennsylvania, USA
| | - Faris M Murad
- Departments of Medicine, Washington University, Saint Louis, Missouri, USA
| | - Dhiraj Yadav
- Departments of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sarah Navina
- Departments of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vijay P Singh
- Departments of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| |
Collapse
|
20
|
Patel K, Trivedi RN, Durgampudi C, Noel P, Cline RA, DeLany JP, Navina S, Singh VP. Lipolysis of visceral adipocyte triglyceride by pancreatic lipases converts mild acute pancreatitis to severe pancreatitis independent of necrosis and inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:808-19. [PMID: 25579844 DOI: 10.1016/j.ajpath.2014.11.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/13/2014] [Accepted: 11/20/2014] [Indexed: 02/06/2023]
Abstract
Visceral fat necrosis has been associated with severe acute pancreatitis (SAP) for over 100 years; however, its pathogenesis and role in SAP outcomes are poorly understood. Based on recent work suggesting that pancreatic fat lipolysis plays an important role in SAP, we evaluated the role of pancreatic lipases in SAP-associated visceral fat necrosis, the inflammatory response, local injury, and outcomes of acute pancreatitis (AP). For this, cerulein pancreatitis was induced in lean and obese mice, alone or with the lipase inhibitor orlistat and parameters of AP induction (serum amylase and lipase), fat necrosis, pancreatic necrosis, and multisystem organ failure, and inflammatory response were assessed. Pancreatic lipases were measured in fat necrosis and were overexpressed in 3T3-L1 cells. We noted obesity to convert mild cerulein AP to SAP with greater cytokines, unsaturated fatty acids (UFAs), and multisystem organ failure, and 100% mortality without affecting AP induction or pancreatic necrosis. Increased pancreatic lipase amounts and activity were noted in the extensive visceral fat necrosis of dying obese mice. Lipase inhibition reduced fat necrosis, UFAs, organ failure, and mortality but not the parameters of AP induction. Pancreatic lipase expression increased lipolysis in 3T3-L1 cells. We conclude that UFAs generated via lipolysis of visceral fat by pancreatic lipases convert mild AP to SAP independent of pancreatic necrosis and the inflammatory response.
Collapse
Affiliation(s)
- Krutika Patel
- Department of Medicine, University of Pittsburgh Medical Center and the University of Pittsburgh, Pittsburgh, Pennsylvania; Mayo Clinic, Scottsdale, Arizona
| | - Ram N Trivedi
- Department of Medicine, University of Pittsburgh Medical Center and the University of Pittsburgh, Pittsburgh, Pennsylvania; Mayo Clinic, Scottsdale, Arizona
| | - Chandra Durgampudi
- Department of Medicine, University of Pittsburgh Medical Center and the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pawan Noel
- Department of Medicine, University of Pittsburgh Medical Center and the University of Pittsburgh, Pittsburgh, Pennsylvania; Mayo Clinic, Scottsdale, Arizona
| | - Rachel A Cline
- Department of Medicine, University of Pittsburgh Medical Center and the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - James P DeLany
- Department of Medicine, University of Pittsburgh Medical Center and the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sarah Navina
- Department of Pathology, University of Pittsburgh Medical Center and the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vijay P Singh
- Department of Medicine, University of Pittsburgh Medical Center and the University of Pittsburgh, Pittsburgh, Pennsylvania; Mayo Clinic, Scottsdale, Arizona.
| |
Collapse
|
21
|
Durgampudi C, Noel P, Patel K, Cline R, Trivedi RN, DeLany JP, Yadav D, Papachristou GI, Lee K, Acharya C, Jaligama D, Navina S, Murad F, Singh VP. Acute lipotoxicity regulates severity of biliary acute pancreatitis without affecting its initiation. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1773-84. [PMID: 24854864 DOI: 10.1016/j.ajpath.2014.02.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 02/07/2014] [Accepted: 02/27/2014] [Indexed: 02/07/2023]
Abstract
Obese patients have worse outcomes during acute pancreatitis (AP). Previous animal models of AP have found worse outcomes in obese rodents who may have a baseline proinflammatory state. Our aim was to study the role of acute lipolytic generation of fatty acids on local severity and systemic complications of AP. Human postpancreatitis necrotic collections were analyzed for unsaturated fatty acids (UFAs) and saturated fatty acids. A model of biliary AP was designed to replicate the human variables by intraductal injection of the triglyceride glyceryl trilinoleate alone or with the chemically distinct lipase inhibitors orlistat or cetilistat. Parameters of AP etiology and outcomes of local and systemic severity were measured. Patients with postpancreatitis necrotic collections were obese, and 13 of 15 had biliary AP. Postpancreatitis necrotic collections were enriched in UFAs. Intraductal glyceryl trilinoleate with or without the lipase inhibitors resulted in oil red O-positive areas, resembling intrapancreatic fat. Both lipase inhibitors reduced the glyceryl trilinoleate-induced increase in serum lipase, UFAs, pancreatic necrosis, serum inflammatory markers, systemic injury, and mortality but not serum alanine aminotransferase, bilirubin, or amylase. We conclude that UFAs are enriched in human necrotic collections and acute UFA generation via lipolysis worsens pancreatic necrosis, systemic inflammation, and injury associated with severe AP. Inhibition of lipolysis reduces UFA generation and improves these outcomes of AP without interfering with its induction.
Collapse
Affiliation(s)
- Chandra Durgampudi
- Department of Medicine, University of Pittsburgh Medical Center Pasavant, Pittsburgh, Pennsylvania
| | - Pawan Noel
- Department of Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Krutika Patel
- Department of Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Rachel Cline
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ram N Trivedi
- Department of Medicine, Mayo Clinic, Scottsdale, Arizona
| | - James P DeLany
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dhiraj Yadav
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Kenneth Lee
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Chathur Acharya
- Department of Medicine, University of Pittsburgh Medical Center Pasavant, Pittsburgh, Pennsylvania
| | - Deepthi Jaligama
- Department of Medicine, University of Pittsburgh Medical Center Pasavant, Pittsburgh, Pennsylvania
| | - Sarah Navina
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Faris Murad
- Department of Medicine, Washington University, St. Louis, Missouri
| | - Vijay P Singh
- Department of Medicine, Mayo Clinic, Scottsdale, Arizona.
| |
Collapse
|
22
|
Binker MG, Cosen-Binker LI. Acute pancreatitis: The stress factor. World J Gastroenterol 2014; 20:5801-5807. [PMID: 24914340 PMCID: PMC4024789 DOI: 10.3748/wjg.v20.i19.5801] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/12/2014] [Accepted: 04/09/2014] [Indexed: 02/06/2023] Open
Abstract
Acute pancreatitis is an inflammatory disorder of the pancreas that may cause life-threatening complications. Etiologies of pancreatitis vary, with gallstones accounting for the majority of all cases, followed by alcohol. Other causes of pancreatitis include trauma, ischemia, mechanical obstruction, infections, autoimmune, hereditary, and drugs. The main events occurring in the pancreatic acinar cell that initiate and propagate acute pancreatitis include inhibition of secretion, intracellular activation of proteases, and generation of inflammatory mediators. Small cytokines known as chemokines are released from damaged pancreatic cells and attract inflammatory cells, whose systemic action ultimately determined the severity of the disease. Indeed, severe forms of pancreatitis may result in systemic inflammatory response syndrome and multiorgan dysfunction syndrome, characterized by a progressive physiologic failure of several interdependent organ systems. Stress occurs when homeostasis is threatened, and stressors can include physical or mental forces, or combinations of both. Depending on the timing and duration, stress can result in beneficial or harmful consequences. While it is well established that a previous acute-short-term stress decreases the severity of experimentally-induced pancreatitis, the worsening effects of chronic stress on the exocrine pancreas have received relatively little attention. This review will focus on the influence of both prior acute-short-term and chronic stress in acute pancreatitis.
Collapse
|
23
|
Acharya C, Navina S, Singh VP. Role of pancreatic fat in the outcomes of pancreatitis. Pancreatology 2014; 14:403-8. [PMID: 25278311 PMCID: PMC4185152 DOI: 10.1016/j.pan.2014.06.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 06/19/2014] [Accepted: 06/19/2014] [Indexed: 12/11/2022]
Abstract
The role of obesity in relation to various disease processes is being increasingly studied, with reports over the last several years increasingly mentioning its association with worse outcomes in acute disease. Obesity has also gained recognition as a risk factor for severe acute pancreatitis (SAP).The mortality in SAP may be as high as 30% and is usually attributable to multi system organ failure (MSOF) earlier in the disease, and complications of necrotizing pancreatitis later [9-11]. To date there is no specific treatment for acute pancreatitis (AP) and the management is largely expectant and supportive. Obesity in general has also been associated with poor outcomes in sepsis and other pathological states including trauma and burns. With the role of unsaturated fatty acids (UFA) as propagators in SAP having recently come to light and with the recognition of acute lipotoxicity, there is now an opportunity to explore different strategies to reduce the mortality and morbidity in SAP and potentially other disease states associated with such a pathophysiology. In this review we will discuss the role of fat and implications of the consequent acute lipotoxicity on the outcomes of acute pancreatitis in lean and obese states and during acute on chronic pancreatitis.
Collapse
Affiliation(s)
- Chathur Acharya
- Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | | | | |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW In this article, we review important advances in our understanding of the mechanisms of pancreatitis. RECENT FINDINGS The relative contributions of intrapancreatic trypsinogen activation and nuclear factor kappa B (NFκB) activation, the two major early independent cellular events in pancreatitis, have been investigated using novel genetic models. Trypsinogen activation has traditionally held the spotlight for many decades as the central pathogenic event of pancreatitis. However, recent experimental evidence points to the role of trypsin activation in early acinar cell damage but not in the inflammatory response of acute pancreatitis, which was shown to be induced by NFκB activation. Further, chronic pancreatitis developed independently of trypsinogen activation in the caerulein model. Sustained NFκB activation, but not persistent intra-acinar expression of active trypsin, was shown to result in chronic pancreatitis. Calcineurin-NFAT (nuclear factor of activated T-cells) signaling was shown to mediate downstream effects of pathologic rise in intracellular calcium. Interleukin-6 was identified as a key cytokine mediating pancreatitis-associated lung injury. SUMMARY Recent advances challenge the long-believed trypsin-centered understanding of pancreatitis. It is becoming increasingly clear that activation of intense inflammatory signaling mechanisms in acinar cells is crucial to the pathogenesis of pancreatitis, which may explain the strong systemic inflammatory response in pancreatitis.
Collapse
|
25
|
Acharya C, Cline RA, Jaligama D, Noel P, Delany JP, Bae K, Furlan A, Baty CJ, Karlsson JM, Rosario BL, Patel K, Mishra V, Durgampudi C, Yadav D, Navina S, Singh VP. Fibrosis reduces severity of acute-on-chronic pancreatitis in humans. Gastroenterology 2013; 145:466-75. [PMID: 23684709 PMCID: PMC3964816 DOI: 10.1053/j.gastro.2013.05.012] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Revised: 04/15/2013] [Accepted: 05/06/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Acute pancreatitis (AP) and chronic pancreatitis (CP) share etiologies, but AP can be more severe and is associated with a higher rate of mortality. We investigated features of CP that protect against severe disease. The amount of intrapancreatic fat (IPF) is increased in obese patients and fibrosis is increased in patients with CP, so we studied whether fibrosis or fat regulate severity of AP attacks in patients with CP. METHODS We reviewed records from the University of Pittsburgh Medical Center/Presbyterian Hospital Autopsy Database (1998-2008) for patients with a diagnosis of AP (n = 23), CP (n = 35), or both (AP-on-CP; n = 15). Pancreatic histology samples from these patients and 50 randomly selected controls (no pancreatic disease) were analyzed, and IPF data were correlated with computed tomography data. An adipocyte and acinar cell Transwell coculture system, with or without collagen type I, was used to study the effects of fibrosis on acinar-adipocyte interactions. We studied the effects of nonesterified fatty acids (NEFAs) and adipokines on acinar cells in culture. RESULTS Levels of IPF were significantly higher in nonobese patients with CP than in nonobese controls. In patients with CP or AP-on-CP, areas of IPF were surrounded by significantly more fibrosis than in controls or patients with AP. Fat necrosis-associated peri-fat acinar necrosis (PFAN, indicated by NEFA spillage) contributed to most of the necrosis observed in samples from patients with AP; however, findings of peri-fat acinar necrosis and total necrosis were significantly lower in samples from patients with CP or AP-on-CP. Fibrosis appeared to wall off the fat necrosis and limit peri-fat acinar necrosis, reducing acinar necrosis. In vitro, collagen I limited the lipolytic flux between acinar cells and adipocytes and prevented increases in adipokines in the acinar compartment. This was associated with reduced acinar cell necrosis. However, NEFAs, but not adipokines, caused acinar cell necrosis. CONCLUSIONS Based on analysis of pancreatic samples from patients with CP, AP, or AP-on-CP and in vitro studies, fibrosis reduces the severity of acute exacerbations of CP by reducing lipolytic flux between adipocytes and acinar cells.
Collapse
Affiliation(s)
- Chathur Acharya
- Department of Medicine University of Pittsburgh Medical Center
Passavant
| | - Rachel A. Cline
- Department of Medicine, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| | - Deepthi Jaligama
- Department of Medicine University of Pittsburgh Medical Center
Passavant
| | - Pawan Noel
- Department of Medicine, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| | - James P. Delany
- Department of Medicine, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| | - Kyongtae Bae
- Department of Radiology, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| | - Alessandro Furlan
- Department of Radiology, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| | - Catherine J. Baty
- Department of Cell Biology & Physiology, University of
Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15206
| | - Jenny M. Karlsson
- Department of Cell Biology & Physiology, University of
Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15206
| | - Bedda L Rosario
- Department of Epidemology, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| | - Krutika Patel
- Department of Medicine, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| | - Vivek Mishra
- Department of Medicine, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| | - Chandra Durgampudi
- Department of Medicine University of Pittsburgh Medical Center
Passavant
| | - Dhiraj Yadav
- Department of Medicine, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| | - Sarah Navina
- Department of Pathology, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| | - Vijay P. Singh
- Department of Medicine, University of Pittsburgh, 200 Lothrop
Street, Pittsburgh, PA 15206
| |
Collapse
|
26
|
Sah RP, Dudeja V, Dawra RK, Saluja AK. Cerulein-induced chronic pancreatitis does not require intra-acinar activation of trypsinogen in mice. Gastroenterology 2013; 144:1076-1085.e2. [PMID: 23354015 PMCID: PMC3928043 DOI: 10.1053/j.gastro.2013.01.041] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 01/02/2013] [Accepted: 01/07/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Premature activation of trypsinogen activation can cause pancreatic injury and has been associated with chronic pancreatitis (CP). Mice that lack intra-acinar activation of trypsinogen, such as trypsinogen-7-null (T(-/-)) and cathepsin B-null (CB(-/-)) mice, have been used to study trypsin-independent processes of CP development. We compared histologic features and inflammatory responses of pancreatic tissues from these mice with those from wild-type mice after the development of CP. METHODS CP was induced in wild-type, T(-/-), and CB(-/-) mice by twice-weekly induction of acute pancreatitis for 10 weeks; acute pancreatitis was induced by hourly intraperitoneal injections of cerulein (50 μg/kg × 6). Pancreatic samples were collected and evaluated by histologic and immunohistochemical analyses. Normal human pancreas samples, obtained from the islet transplant program at the University of Minnesota, were used as controls and CP samples were obtained from surgical resections. RESULTS Compared with pancreatic tissues from wild-type mice, those from T(-/-) and CB(-/-) mice had similar levels of atrophy, histomorphologic features of CP, and chronic inflammation. All samples had comparable intra-acinar activation of nuclear factor (NF)-κB, a transcription factor that regulates the inflammatory response, immediately after injection of cerulein. Pancreatic tissue samples from patients with CP had increased activation of NF-κB (based on nuclear translocation of p65 in acinar cells) compared with controls. CONCLUSIONS Induction of CP in mice by cerulein injection does not require intra-acinar activation of trypsinogen. Pancreatic acinar cells of patients with CP have increased levels of NF-κB activation compared with controls; regulation of the inflammatory response by this transcription factor might be involved in the pathogenesis of CP.
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW In this article, recent advances in the pathogenesis of acute pancreatitis have been reviewed. RECENT FINDINGS Pathologic intra-acinar trypsinogen activation had been hypothesized to be the central mechanism of pancreatitis for over a century. This hypothesis could be explored for the first time with the development of a novel mouse model lacking pathologic intra-acinar trypsinogen activation. It became clear that intra-acinar trypsinogen activation contributes to early acinar injury, but local and systemic inflammation progress independently during pancreatitis. Early intra-acinar nuclear factor kappa B (NFκB) activation, which occurs parallel to but independent of trypsinogen activation, may be crucial in pancreatitis. Although the mechanism of NFκB and trypsinogen activation is not entirely clear, further insights have been made into key pathogenic cellular events such as calcium signaling, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, autophagy and impaired trafficking, and lysosomal and secretory responses. Cellular intrinsic damage-sensing mechanisms that lead to activation of the inflammatory response aimed at repair, but lead to disease when overwhelmed, are beginning to be understood. SUMMARY New findings necessitate a paradigm shift in our understanding of acute pancreatitis. Intra-acinar trypsinogen activation leads to early pancreatic injury, but the inflammatory response of acute pancreatitis develops independently, driven by early activation of inflammatory pathways.
Collapse
|
28
|
Navina S, Acharya C, DeLany JP, Orlichenko LS, Baty CJ, Shiva SS, Durgampudi C, Karlsson JM, Lee K, Bae KT, Furlan A, Behari J, Liu S, McHale T, Nichols L, Papachristou GI, Yadav D, Singh VP. Lipotoxicity causes multisystem organ failure and exacerbates acute pancreatitis in obesity. Sci Transl Med 2012; 3:107ra110. [PMID: 22049070 DOI: 10.1126/scitranslmed.3002573] [Citation(s) in RCA: 312] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity increases the risk of adverse outcomes during acute critical illnesses such as burns, severe trauma, and acute pancreatitis. Although individuals with more body fat and higher serum cytokines and lipase are more likely to experience problems, the roles that these characteristics play are not clear. We used severe acute pancreatitis as a representative disease to investigate the effects of obesity on local organ function and systemic processes. In obese humans, we found that an increase in the volume of intrapancreatic adipocytes was associated with more extensive pancreatic necrosis during acute pancreatitis and that acute pancreatitis was associated with multisystem organ failure in obese individuals. In vitro studies of pancreatic acinar cells showed that unsaturated fatty acids were proinflammatory, releasing intracellular calcium, inhibiting mitochondrial complexes I and V, and causing necrosis. Saturated fatty acids had no such effects. Inhibition of lipolysis in obese (ob/ob) mice with induced pancreatitis prevented a rise in serum unsaturated fatty acids and prevented renal injury, lung injury, systemic inflammation, hypocalcemia, reduced pancreatic necrosis, and mortality. Thus, therapeutic approaches that target unsaturated fatty acid-mediated lipotoxicity may reduce adverse outcomes in obese patients with critical illnesses such as severe acute pancreatitis.
Collapse
Affiliation(s)
- Sarah Navina
- Department of Pathology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Orlichenko L, Stolz DB, Noel P, Behari J, Liu S, Singh VP. ADP-ribosylation factor 1 protein regulates trypsinogen activation via organellar trafficking of procathepsin B protein and autophagic maturation in acute pancreatitis. J Biol Chem 2012; 287:24284-93. [PMID: 22570480 DOI: 10.1074/jbc.m111.328815] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Several studies have suggested that autophagy might play a deleterious role in acute pancreatitis via intra-acinar activation of digestive enzymes. The prototype for this phenomenon is cathepsin B-mediated trypsin generation. To determine the organellar basis of this process, we investigated the subcellular distribution of the cathepsin B precursor, procathepsin B. We found that procathepsin B is enriched in Golgi-containing microsomes, suggesting a role for the ADP-ribosylation (ARF)-dependent trafficking of cathepsin B. Indeed, caerulein treatment increased processing of procathepsin B, whereas a known ARF inhibitor brefeldin A (BFA) prevented this. Similar treatment did not affect processing of procathepsin L. BFA-mediated ARF1 inhibition resulted in reduced cathepsin B activity and consequently reduced trypsinogen activation. However, formation of light chain 3 (LC3-II) was not affected, suggesting that BFA did not prevent autophagy induction. Instead, sucrose density gradient centrifugation and electron microscopy showed that BFA arrested caerulein-induced autophagosomal maturation. Therefore, ARF1-dependent trafficking of procathepsin B and the maturation of autophagosomes results in cathepsin B-mediated trypsinogen activation induced by caerulein.
Collapse
Affiliation(s)
- Lidiya Orlichenko
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | |
Collapse
|
30
|
Sakai A, Nishiumi S, Shiomi Y, Kobayashi T, Izumi Y, Kutsumi H, Hayakumo T, Azuma T, Yoshida M. Metabolomic analysis to discover candidate therapeutic agents against acute pancreatitis. Arch Biochem Biophys 2012; 522:107-20. [PMID: 22483684 DOI: 10.1016/j.abb.2012.03.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 03/21/2012] [Accepted: 03/21/2012] [Indexed: 01/22/2023]
Abstract
Novel and effective drugs against acute pancreatitis are required. Therefore, we examined the changes in the metabolite levels in the serum and pancreatic tissue of mice with cerulein- and arginine-induced pancreatitis using gas-chromatography/mass-spectrometry (GC/MS) and investigated whether these alterations affected the severity of acute pancreatitis. In the cerulein-induced pancreatitis model, 93 and 129 metabolites were detected in the serum and pancreatic tissue, respectively. In the L-arginine-induced acute pancreatitis model, 120 and 133 metabolites were detected in the serum and pancreatic tissue, respectively. Among the metabolites, the concentrations of tricarboxylic acid (TCA) cycle intermediates and amino acids were altered in pancreatitis, and in pancreatic tissue, the levels of the intermediates involved in the initial part of the TCA cycle were increased and those of the intermediates involved in the latter part of the TCA cycle were decreased. Some metabolites exhibited similar changes in both pancreatitis mouse models, e.g., the levels of glutamic acid and O-phosphoethanolamine were significantly decreased in the pancreatic tissue. Supplementation with glutamic acid and O-phosphoethanolamine attenuated the severity of cerulein-induced acute pancreatitis. Our results suggest that GC/MS-based metabolomics is capable of accurately representing the status of acute pancreatitis, leading to the discovery of therapeutic agents for pancreatitis.
Collapse
Affiliation(s)
- Aya Sakai
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chu-o-ku, Kobe, Hyogo 650-0017, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Dawra R, Sah RP, Dudeja V, Rishi L, Saluja AK, Garg P, Saluja AK. Intra-acinar trypsinogen activation mediates early stages of pancreatic injury but not inflammation in mice with acute pancreatitis. Gastroenterology 2011; 141:2210-2217.e2. [PMID: 21875495 PMCID: PMC3587766 DOI: 10.1053/j.gastro.2011.08.033] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 08/11/2011] [Accepted: 08/18/2011] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS The role of trypsinogen activation in the pathogenesis of acute pancreatitis (AP) has not been clearly established. METHODS We generated and characterized mice lacking trypsinogen isoform 7 (T7) gene (T(-/-)). The effects of pathologic activation of trypsinogen were studied in these mice during induction of AP with cerulein. Acinar cell death, tissue damage, early intra-acinar activation of the transcription factor nuclear factor κB (NF-κB), and local and systemic inflammation were compared between T(-/-) and wild-type mice with AP. RESULTS Deletion of T7 reduced the total trypsinogen content by 60% but did not affect physiologic function. T(-/-) mice lacked pathologic activation of trypsinogen, which occurs within acinar cells during early stages of AP progression. Absence of trypsinogen activation in T(-/-) mice led to near complete inhibition of acinar cell death in vitro and a 50% reduction in acinar necrosis during AP progression. However, T(-/-) mice had similar degrees of local and systemic inflammation during AP progression and comparable levels of intra-acinar NF-κB activation, which was previously shown to occur concurrently with trypsinogen activation during early stages of pancreatitis. CONCLUSIONS T7 is activated during pathogenesis of AP in mice. Intra-acinar trypsinogen activation leads to acinar death during early stages of pancreatitis, which accounts for 50% of the pancreatic damage in AP. However, progression of local and systemic inflammation in AP does not require trypsinogen activation. NF-κB is activated early in acinar cells, independently of trypsinogen activation, and might be responsible for progression of AP.
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Despite being a subject of much scientific scrutiny, the pathogenesis of acute pancreatitis is still not well understood. This article reviews recent advances in our understanding of acute pancreatitis. RECENT FINDINGS Zymogen activation, observed within acini early during acute pancreatitis for a long time, was shown to be sufficient to induce acute pancreatitis. Another key early event, NFκB activation, has previously been shown to induce acute pancreatitis. The relationship between these two key early steps is beginning to be clarified. Mechanisms of zymogen activation - pathologic calcium signaling, pH changes, colocalization and autophagy, and of NFκB activation have been investigated intensively along with potential therapeutic targets both upstream and downstream of these key events. Additional key findings have been elucidation of the role of bioenergetics and the dual role of oxidative stress in acute pancreatitis, recognition of endoplasmic reticulum stress as an early step and the status of duct cells as important entities in pancreatic injury. SUMMARY Current findings have provided further insight into the roles and mechanisms of zymogen activation and inflammatory pathways in pancreatic injury. Future studies, which will be of great importance in identifying therapeutic targets, are being undertaken to establish the relative contributions of these pathways during acute pancreatitis.
Collapse
|
33
|
Binker MG, Binker-Cosen AA, Richards D, Gaisano HY, de Cosen RH, Cosen-Binker LI. Chronic stress sensitizes rats to pancreatitis induced by cerulein: Role of TNF-α. World J Gastroenterol 2010; 16:5565-81. [PMID: 21105189 PMCID: PMC2992674 DOI: 10.3748/wjg.v16.i44.5565] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate chronic stress as a susceptibility factor for developing pancreatitis, as well as tumor necrosis factor-α (TNF-α) as a putative sensitizer.
METHODS: Rat pancreatic acini were used to analyze the influence of TNF-α on submaximal (50 pmol/L) cholecystokinin (CCK) stimulation. Chronic restraint (4 h every day for 21 d) was used to evaluate the effects of submaximal (0.2 μg/kg per hour) cerulein stimulation on chronically stressed rats.
RESULTS: In vitro exposure of pancreatic acini to TNF-α disorganized the actin cytoskeleton. This was further increased by TNF-α/CCK treatment, which additionally reduced amylase secretion, and increased trypsin and nuclear factor-κB activities in a protein-kinase-C δ and ε-dependent manner. TNF-α/CCK also enhanced caspases’ activity and lactate dehydrogenase release, induced ATP loss, and augmented the ADP/ATP ratio. In vivo, rats under chronic restraint exhibited elevated serum and pancreatic TNF-α levels. Serum, pancreatic, and lung inflammatory parameters, as well as caspases’activity in pancreatic and lung tissue, were substantially enhanced in stressed/cerulein-treated rats, which also experienced tissues’ ATP loss and greater ADP/ATP ratios. Histological examination revealed that stressed/cerulein-treated animals developed abundant pancreatic and lung edema, hemorrhage and leukocyte infiltrate, and pancreatic necrosis. Pancreatitis severity was greatly decreased by treating animals with an anti-TNF-α-antibody, which diminished all inflammatory parameters, histopathological scores, and apoptotic/necrotic markers in stressed/cerulein-treated rats.
CONCLUSION: In rats, chronic stress increases susceptibility for developing pancreatitis, which involves TNF-α sensitization of pancreatic acinar cells to undergo injury by physiological cerulein stimulation.
Collapse
|
34
|
Fletcher PL, Fletcher MD, Weninger K, Anderson TE, Martin BM. Vesicle-associated membrane protein (VAMP) cleavage by a new metalloprotease from the Brazilian scorpion Tityus serrulatus. J Biol Chem 2009; 285:7405-16. [PMID: 20026600 DOI: 10.1074/jbc.m109.028365] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We present evidence that venom from the Brazilian scorpion Tityus serrulatus and a purified fraction selectively cleave essential SNARE proteins within exocrine pancreatic tissue. Western blotting for vesicle-associated membrane protein type v-SNARE proteins (or synaptobrevins) reveals characteristic alterations to venom-treated excised pancreatic lobules in vitro. Immunocytochemistry by electron microscopy confirms both the SNARE identity as VAMP2 and the proteolysis of VAMP2 as a marked decrease in secondary antibody-conjugated colloidal gold particles that are predominantly associated with mature zymogen granules. Studies with recombinant SNARE proteins were used to determine the specific cleavage site in VAMP2 and the susceptibility of VAMP8 (endobrevin). The VAMP2 cleavage site is between the transmembrane anchor and the SNARE motif that assembles into the ternary SNARE complex. Inclusion of divalent chelating agents (EDTA) with fraction nu, an otherwise active purified component from venom, eliminates SNARE proteolysis, suggesting the active protein is a metalloprotease. The unique cleavages of VAMP2 and VAMP8 may be linked to pancreatitis that develops following scorpion envenomation as both of these v-SNARE proteins are associated with zymogen granule membranes in pancreatic acinar cells. We have isolated antarease, a metalloprotease from fraction nu that cleaves VAMP2, and report its amino acid sequence.
Collapse
Affiliation(s)
- Paul L Fletcher
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina 27834, USA
| | | | | | | | | |
Collapse
|
35
|
Binker MG, Binker-Cosen AA, Gaisano HY, Cosen-Binker LI. Inhibition of Rac1 decreases the severity of pancreatitis and pancreatitis-associated lung injury in mice. Exp Physiol 2008; 93:1091-103. [PMID: 18567599 DOI: 10.1113/expphysiol.2008.043141] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pancreatitis is a disease with high morbidity and mortality. In vitro experiments on pancreatic acini showed that supramaximal but not submaximal cholecystokinin (CCK) stimulation induces effects in the acinar cell that can be correlated with acinar morphological changes observed in the in vivo experimental model of cerulein-induced pancreatitis. The GTPase Rac1 was previously reported to be involved in CCK-evoked amylase release from pancreatic acinar cells. Here, we demonstrate that pretreatment with the Rac1 inhibitor NSC23766 (100 microM, 2 h) effectively blocked Rac1 translocation and activation in CCK-stimulated pancreatic acini, without affecting activation of its closely related GTPase, RhoA. This specific Rac1 inhibition decreased supramaximal (10 nM) CCK-stimulated acinar amylase release (27.% reduction), which seems to be connected to the reduction observed in serum amylase (46.6% reduction) and lipase levels (46.1% reduction) from cerulein-treated mice receiving NSC23766 (100 nmol h(-1)). The lack of Rac1 activation also reduced formation of reactive oxygen species (ROS; 20.8% reduction) and lactate dehydrogenase release (LDH; 24.3% reduction), but did not alter calcium signaling or trypsinogen activation in 10 nM CCK-stimulated acini. In the in vivo model, the cerulein-treated mice receiving NSC23766 also presented a decrease in both pancreatic and lung histopathological scores (reduction in oedema, 32.4 and 66.4%; haemorrhage, 48.3 and 60.2%; and leukocyte infiltrate, 53.5 and 43.6%, respectively; reduction in pancreatic necrosis, 65.6%) and inflammatory parameters [reduction in myeloperoxidase, 52.2 and 38.9%; nuclear factor kappaB (p65), 61.3 and 48.6%; and nuclear factor kappaB (p50), 46.9 and 44.9%, respectively], together with lower serum levels for inflammatory (TNF-alpha, 40.4% reduction) and cellular damage metabolites (LDH, 52.7% reduction). Collectively, these results suggest that pharmacological Rac1 inhibition ameliorates the severity of pancreatitis and pancreatitis-associated lung injury through the reduction of pancreatic acinar damage induced by pathological digestive enzyme secretion and overproduction of ROS.
Collapse
Affiliation(s)
- Marcelo G Binker
- CBRHC Research Center, Arribenos 1697, P.1, Buenos Aires, 1426, Argentina
| | | | | | | |
Collapse
|
36
|
Abstract
OBJECTIVE To test the hypothesis that disruption of acinar cell membranes is the earliest event that takes place after the onset of acute pancreatitis. METHODS Cerulein and taurocholate pancreatitis were induced in rats. Furthermore, stimulation with different doses of bombesin, pilocarpine, and cerulein was performed. Five to 180 minutes after initiation of treatment, animals were killed. Disruption of cell membranes was detected by the penetration of the experimental animal's own albumin or immunoglobulin G (IgG) into acinar cells by immunocytological localization. Tissue was further analyzed by electron microscopy and electron microscopic immunostaining. RESULTS Animals with pancreatitis displayed significantly greater antialbumin and anti-IgG immunostaining in the cytoplasm of acinar cells and in vacuoles in comparison with controls, confirming membrane disruption. This was not detectable after stimulation with bombesin, pilocarpine, and nonsupramaximal doses of cerulein. The first changes were seen after 5 minutes of induction of pancreatitis. Results were verified by electron microscopy and electron microscopic immunohistochemistry. CONCLUSIONS The penetration of albumin and IgG into acinar cells indicates that wounding of their plasma membrane occurs at the onset of acute pancreatitis. Disruption of the membranes could be expected to allow the influx of calcium ions, causing massive intracellular alterations, and exit of molecules, such as enzymes from acinar cells.
Collapse
|
37
|
Cosen-Binker LI, Gaisano HY. Recent insights into the cellular mechanisms of acute pancreatitis. CANADIAN JOURNAL OF GASTROENTEROLOGY = JOURNAL CANADIEN DE GASTROENTEROLOGIE 2007; 21:19-24. [PMID: 17225878 PMCID: PMC2656626 DOI: 10.1155/2007/930424] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In acute pancreatitis, initiating cellular events causing acinar cell injury includes co-localization of zymogens with lysosomal hydrolases, leading to premature enzyme activation and pathological exocytosis of zymogens into the interstitial space. This is followed by processes that accentuate cell injury; triggering acute inflammatory mediators, intensifying oxidative stress, compromising the microcirculation and activating a neurogenic feedback. Such localized events then progress to a systemic inflammatory response leading to multiorgan dysfunction syndrome with resulting high morbidity and mortality. The present review discusses some of the most recent insights into each of these cellular processes postulated to cause or propagate the process of acute pancreatitis, and also the role of alcohol and genetics.
Collapse
Affiliation(s)
| | - Herbert Y Gaisano
- Correspondence: Dr Herbert Y Gaisano, University of Toronto, Room 7226, Medical Science Building, 1 King’s College Circle, Toronto, Ontario M5S 1A8. Telephone 416-978-1526, fax 416-978-8765, e-mail
| |
Collapse
|
38
|
Abstract
Many animal models are available to investigate the pathogenesis of pancreatitis, an inflammatory disorder of the pancreas. However, the secretagogue hyperstimulation model of pancreatitis is the most commonly used. Animals infused with high doses of cholecystokinin (CCK) exhibit hyperamylasemia, pancreatic edema, and acinar cell injury, which closely mimic pancreatitis in humans. Intra-acinar zymogen activation is an essential early event in the pathogenesis of secretagogue-induced pancreatitis. Early in the course of pancreatitis, lysosomal hydrolases colocalize with digestive zymogens and activate them. These activated zymogens then cause acinar cell injury and necrosis, a characteristic of pancreatitis. Besides being the site of initiation of injury in pancreatitis, acinar cells also synthesize and release cytokines and chemokines very early in the course of pancreatitis, which then attract and activate inflammatory cells and initiate the disease's systemic phase.
Collapse
Affiliation(s)
- Ashok K Saluja
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
39
|
Abstract
Chronic pancreatitis is a fibroinflammatory disease of the pancreas. Etiologically, most cases are related to alcohol abuse and smoking. Recently, gene mutations have been identified as the cause of hereditary pancreatitis. Other chronic pancreatitis types that were defined in recent years are autoimmune pancreatitis (lymphoplasmacytic sclerosing pancreatitis) and paraduodenal pancreatitis ('groove pancreatitis', 'cystic dystrophy of heterotopic pancreas'). This review describes and discusses the main histological findings, the pathogenesis and the clinical features of the various types of chronic pancreatitis. In addition, pseudotumors and other tumor-like lesions are briefly mentioned.
Collapse
Affiliation(s)
- Günter Klöppel
- Department of Pathology, University of Kiel, Kiel, Schleswig-Holstein, Germany.
| |
Collapse
|
40
|
Zhang XM, Feng ZS, Zhao QH, Xiao CM, Mitchell DG, Shu J, Zeng NL, Xu XX, Lei JY, Tian XB. Acute interstitial edematous pancreatitis: Findings on non-enhanced MR imaging. World J Gastroenterol 2006; 12:5859-65. [PMID: 17007053 PMCID: PMC4100668 DOI: 10.3748/wjg.v12.i36.5859] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the appearances of acute interstitial edematous pancreatitis (IEP) on non-enhanced MR imaging.
METHODS: A total of 53 patients with IEP diagnosed by clinical features and laboratory findings were underwent MR imaging. MR imaging sequences included fast spoiled gradient echo (FSPGR) fat saturation axial T1-weighted imaging, gradient echo T1-weighted (in phase), single shot fast spin echo (SSFSE) T2-weighted, respiratory triggered (R-T) T2-weighted with fat saturation, and MR cholangiopancreatography. Using the MR severity score index, pancreatitis was graded as mild (0-2 points), moderate (3-6 points) and severe (7-10 points).
RESULTS: Among the 53 patients, IEP was graded as mild in 37 patients and as moderate in 16 patients. Forty-seven of 53 (89%) patients had at least one abnormality on MR images. Pancreas was hypointense relative to liver on FSPGR T1-weighted images in 18.9% of patients, and hyperintense in 25% and 30% on SSFSE T2-weighted and R-T T2-weighted images, respectively. The prevalences of the findings of IEP on R-T T2-weighted images were, respectively, 85% for pancreatic fascial plane, 77% for left renal fascial plane, 55% for peripancreatic fat stranding, 42% for right renal fascial plane, 45% for perivascular fluid, 40% for thickened pancreatic lobular septum and 25% for peripancreatic fluid, which were markedly higher than those on in-phase or SSFSE T2-weighted images (P < 0.001).
CONCLUSION: IEP primarily manifests on non-enhanced MR images as thickened pancreatic fascial plane, left renal fascial plane, peripancreatic fat stranding, and peripancreatic fluid. R-T T2-weighted imaging is more sensitive than in-phase and SSFSE T2-weighted imaging for depicting IEP.
Collapse
Affiliation(s)
- Xiao-Ming Zhang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong 637000, Sichuan Province, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
More than 100 years ago it was proposed that pancreatitis essentially is a disease in which the pancreas undergoes autodigestion by its own prematurely activated digestive enzymes. Why and how digestive zymogens autoactivate within the pancreas early in the disease process has been a matter of controversy and debate. Some of the mechanisms that are considered to be involved indigestive protease activation are inherited and as of recently can be tested for clinically. Here we review the most recent progress in elucidating the mechanisms involved in the onset of pancreatitis. We specifically focus on serine and cysteine proteases in the autodigestive cascade that precedes acinar cell injury and the biochemical processes involved in their activation.
Collapse
Affiliation(s)
- Manuel Ruthenbürger
- Department of Gastroenterology, Endocrinology and Nutrition, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Loeffler-Str 23A, 17487 Greifswald, Germany
| | | | | |
Collapse
|
42
|
Abstract
The underlying mechanisms involved in the pathogenesis of acute pancreatitis are ill understood. The mortality rate of this disease has not significantly improved over the past few decades. Current treatment options are limited, and predominantly aimed at supportive therapy. A key feature of severe acute pancreatitis is the presence of extensive tissue necrosis with both local and systemic manifestations of inflammatory response syndromes. A better understanding of the underlying pathophysiology of severe acute pancreatitis may lead to more targeted therapeutic options, potentially leading to improved survival. Animal models of acute pancreatitis are therefore an essential investigative tool for these aims to be achieved. This review discusses the suitability of recent non-invasive models of acute pancreatitis such as hormone-induced, alcohol-induced, immune-mediated, diet-induced, gene knockout and L-arginine; and invasive models including closed duodenal loop, antegrade pancreatic duct perfusion, biliopancreatic duct injection, combination of secretory hyperstimulation with minimal intraductal bile acid exposure, vascular-induced, ischaemia/reperfusion and duct ligation.
Collapse
Affiliation(s)
- Kim Hue Su
- Department of Surgery, University of Melbourne, Austin HospitalMelbourne VictoriaAustralia
| | - Christine Cuthbertson
- Department of Surgery, University of Melbourne, Austin HospitalMelbourne VictoriaAustralia
| | - Christopher Christophi
- Department of Surgery, University of Melbourne, Austin HospitalMelbourne VictoriaAustralia
| |
Collapse
|
43
|
Abstract
Acute pancreatitis is characterized by the occurrence of necroinflammatory changes in the pancreas. Three types of necrosis may be distinguished: (1) interstitial tissue necrosis, which subsequently may also involve acinar and ductal cells, (2) ductal necrosis, and (3) acinar necrosis. The first type of necrosis is autodigestive in nature and is typical of the most common forms of acute pancreatitis, which are associated with alcohol, bile duct disease, metabolic conditions, and other rare factors. Clinically, these types of pancreatitis may be either mild or severe (Atlanta classification). The mild form is also known as edematous pancreatitis, because there is edematous swelling of the pancreas combined with tiny foci of interstitial (fat) necrosis. Severe or necrotizing pancreatitis shows large areas of often hemorrhagic necrosis of the pancreatic and particularly the peripancreatic tissue. The ductal type of necrosis is rare and may be seen in pancreatitis associated with prolonged circulatory failure. The acinar type of necrosis is caused by infectious agents. Complications of acute pancreatitis, such as pseudocyst, bleeding, and infection, determine the course of the disease.
Collapse
Affiliation(s)
- Günter Klöppel
- Department of Pathology, University of Kiel, Kiel, Germany.
| |
Collapse
|
44
|
Abstract
Fibroinflammatory changes to the pancreatic tissue characterize chronic pancreatitis. This article summarizes the current state of knowledge on the pathology and pathogenesis of chronic pancreatitis associated with alcohol, hereditary factors, metabolic conditions, and anatomical abnormalities. Specifically, the pathogenetic mechanisms that lead to chronic pancreatitis in patients with alcohol abuse will be discussed. In addition, brief descriptions of the features of chronic pancreatitis of nonalcoholic origin and of the pancreatic fibrosis that is not associated with symptoms of chronic pancreatitis will be given.
Collapse
Affiliation(s)
- Günter Klöppel
- Department of Pathology, University of Kiel, Kiel, Germany.
| |
Collapse
|
45
|
Chebli JMF, Gaburri PD, De Souza AFM, Junior EVM, Gaburri AK, Felga GEG, De Paula EA, Forn CG, De Almeida GV, De Castro Nehme F. Oral refeeding in patients with mild acute pancreatitis: prevalence and risk factors of relapsing abdominal pain. J Gastroenterol Hepatol 2005; 20:1385-1389. [PMID: 16105125 DOI: 10.1111/j.1440-1746.2005.03986.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM In acute pancreatitis (AP), oral refeeding may stimulate pancreatic secretion, increasing the inflammation of the glandular tissue causing relapse of abdominal pain or even exacerbation of the disease. This study aimed to assess the prevalence and risk factors of abdominal pain relapse over oral refeeding in patients convalescing with AP as well as the impact of pain recurrence on the hospital stay. METHODS Inclusion criteria were AP confirmed by biochemical and/or radiological data in the absence of severe disease or extensive necrosis. The same diet was offered to all patients during oral refeeding. Demographic, clinical, biochemical and radiological data were prospectively recorded and analyzed. RESULTS A total of 130 patients were included. During the oral refeeding period, 32 (24.6%) patients had pain relapse, which was more common on days 1 (68.8%) and 2 (28.1%). Pain relapse was related to higher serum levels of lipase on the day before refeeding, higher serum levels of C-reactive protein on the fourth day, and presence of peripancreatic fluid collections (P < 0.01). Pain relapse significantly increased total hospital stay (P < 0.01). CONCLUSIONS In patients with mild AP, pain relapse during oral refeeding was relatively high (24.6%), particularly on the first or second day. Their risk appeared be associated with more intense or persistent pancreatic inflammation on the day before refeeding, and presence of peripancreatic fluid collections. Pain relapse increased hospital stay, and likely overall costs on disease treatment.
Collapse
Affiliation(s)
- Julio Maria Fonseca Chebli
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of the Federal University of Juiz de Fora, Minas Gerais, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Considerable progress in the understanding of the pathogenesis of acute pancreatitis is based on the conclusive finding that the initiation of the disease occurs within the acinar cell. Two lines of evidence have contributed to the progress in understanding the disease process: (1) the identification of patients with a hereditary form of pancreatitis as carriers of germline-mutations in the genes for cationic trypsinogen and the pancreatic secretory trypsin inhibitor and (2) the use of various transgenic and knock-out mouse strains in experimental models of acute pancreatitis. On the other hand, these studies have delivered several unexpected results that appear to be incompatible with long-standing dogmas and paradigms of pancreatic research. Further progress in knowledge will result if the well-characterized enzymatic properties of human enzymes that are involved in the initial activation cascade can be investigated under in vivo conditions in transgenic animals or in permanent acinar cell lines. Such studies will permit the development of effective strategies for the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Walter Halangk
- Division of Experimental Surgery, Department of Surgery, Otto-von-Guericke-Universität, Magdeburg, Leipziger Strasse, 44 D-39120 Magdeburg, Germany.
| | | |
Collapse
|
47
|
Abstract
Considerable progress in the understanding of the pathogenesis of acute pancreatitis is based on the conclusive finding that the initiation of the disease occurs within the acinar cell. Two lines of evidence have contributed to the progress in understanding the disease process: (1) the identification of patients with a hereditary form of pancreatitis as carriers of germline-mutations in the genes for cationic trypsinogen and the pancreatic secretory trypsin inhibitor and (2) the use of various transgenic and knock-out mouse strains in experimental models of acute pancreatitis. On the other hand, these studies have delivered several unexpected results that appear to be incompatible with long-standing dogmas and paradigms of pancreatic research. Further progress in knowledge will result if the well-characterized enzymatic properties of human enzymes that are involved in the initial activation cascade can be investigated under in vivo conditions in transgenic animals or in permanent acinar cell lines. Such studies will permit the development of effective strategies for the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Walter Halangk
- Division of Experimental Surgery, Department of Surgery, Otto-von-Guericke-Universität, Magdeburg, Leipziger Strasse, 44 D-39120 Magdeburg, Germany.
| | | |
Collapse
|
48
|
Bimmler D, Schiesser M, Perren A, Scheele G, Angst E, Meili S, Ammann R, Graf R. Coordinate regulation of PSP/reg and PAP isoforms as a family of secretory stress proteins in an animal model of chronic pancreatitis. J Surg Res 2004; 118:122-35. [PMID: 15100001 DOI: 10.1016/s0022-4804(03)00342-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2003] [Indexed: 11/22/2022]
Abstract
BACKGROUND PSP/reg and PAP are secretory stress proteins (SSP) and may be part of a protective mechanism. They share structural homologies and form insoluble fibrils after tryptic activation. To further explore the regulation of these proteins, we investigated the male WBN/Kob rat, a model of pancreatic inflammatory and fibrotic disease similar to chronic pancreatitis. MATERIALS AND METHODS Expression of PSP/reg and PAP I, II and III in the WBN/Kob rat pancreas was evaluated on the mRNA and protein level, by immunohistochemistry and by highly sensitive isoform specific ELISAs. RESULTS The SSPs are constitutively secreted, PAP in nanomolar, PSP/reg in micromolar concentrations. Before conventional morphological changes are detectable in the WBN/Kob rat, focally increased expression of secretory stress protein is visible. SSP levels in pancreatic juice of WBN/Kob rats reach peak values 10- to 50-fold higher than in Wistar control rats. The highest expression was localized to acini with inflammatory infiltration. CONCLUSIONS There is a tight spatial and temporal association between pre-inflammatory changes or inflammation and SSP-expression. These results support our concept that PSP/reg and PAP are coordinately regulated SSP.
Collapse
Affiliation(s)
- Daniel Bimmler
- Pancreatitis Research Laboratory, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Gaisano HY, Sheu L, Whitcomb D. Alcoholic chronic pancreatitis involves displacement of Munc18c from the pancreatic acinar basal membrane surface. Pancreas 2004; 28:395-400. [PMID: 15097857 DOI: 10.1097/00006676-200405000-00008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The minimal machinery for fusion of secretory vesicles with the cell membrane is a cognate set of v- and t-SNAREs on opposing membranes. Spontaneous SNARE complex assembly leading to unregulated membrane fusion is prevented by Munc18 proteins that bind membrane SNAREs syntaxins. Munc18 blocks syntaxin interactions with cognate SNARE proteins and thereby act as an inhibitor of exocytosis. The pancreatic acinar cell contains several sets of cognate SNAREs and Munc18 proteins that mediate the distinct exocytic events. We had reported that in the rat pancreas, Munc18c co-localizes with t-SNAREs syntaxin4 and SNAP23 on the acinar cell basolateral plasma membrane. Under conditions that induce pancreatitis in vivo, displacement of Munc18c from the basolateral plasma membrane relieved its blockade of SNARE-mediated membrane fusion in this region and thereby redirected apical exocytosis to the basal membrane surface. Here we show in a case of human mild alcoholic chronic pancreatitis that Munc18c is also displaced from the plasma membrane of intact acinar cells, which would render these cells receptive to pathologic basolateral exocytosis and further episodes of pancreatitis.
Collapse
Affiliation(s)
- Herbert Y Gaisano
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | | | | |
Collapse
|
50
|
Abstract
Excessive ethanol consumption is a common risk factor for acute and chronic pancreatitis. Ethanol could lead to the onset of pancreatitis in a number of ways; the most recently discovered is its effect on intrapancreatic digestive enzyme activation, by either sensitizing acinar cells to pathologic stimuli or stimulating the release of a secretagogue (cholecystokinin) from duodenal I cells. Recent advances in cell biologic and molecular techniques have permitted us to address the intracellular events involved in digestive enzyme activation in a manner that was previously considered impossible. Investigations that used these novel techniques found that (a) trypsin is, in contrast to its role in the small intestine, not necessarily involved in the premature intracellular activation of other digestive proteases such as proelastase; (b) trypsinogen does not autoactivate intracellularly but is instead largely activated by the lysosomal hydrolase cathepsin B; and (c) the role of trypsin in the intrapancreatic protease cascade is most likely one that involves the degradation, rather than the activation, of active digestive proteases including trypsin itself. These studies, as well as investigations that have addressed the role of mutant trypsin in the disease onset of hereditary pancreatitis, suggest that trypsin may not be critical for triggering pancreatitis but might have a protective role against the action of some of the other digestive proteases. While the specific role of different digestive enzymes in initiating pancreatitis is still a matter of debate and the topic of ongoing investigations, experimental evidence suggests that ethanol can directly interfere with the processes involved in digestive zymogen activation.
Collapse
Affiliation(s)
- Markus M Lerch
- Department of Medicine A, Ernst-Moritz-Arndt Universität, Greifswald, Germany.
| | | | | | | | | | | |
Collapse
|