1
|
Bertola L, Pepe G, Dolce A, Lecchi C, Borroni EM, Savino B, Canesi S, Sala L, Moretti P, Giordano A, Ressel L, Scanziani E, Vegeto E, Recordati C. Sex-dependent modulation of caerulein-induced acute pancreatitis in C57BL/6J mice. Vet Pathol 2025:3009858241312606. [PMID: 39878085 DOI: 10.1177/03009858241312606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Acute pancreatitis (AP) is a life-threatening condition, with a higher mortality rate in men than women and in which estrogens might play a protective role. This study aimed to investigate sex-dependent differences in a mouse model of caerulein-induced AP. Thirty-six C57BL/6J mice (19 females and 17 males) were treated intraperitoneally with phosphate-buffered saline or caerulein, and sacrificed 12 hours, 2 days, or 7 days after the last injection. Blood was collected for amylase, lipase, and glucose determination. Severity and extent of inflammation, apoptosis, and acinar to ductal metaplasia (ADM) in pancreatic tissue were scored histologically and total macrophages, major histocompatibility complex (MHC)-II+ cells, M2 macrophages, T and B cells, neutrophils, apoptosis, and ADM were marked immunohistochemically and quantified by digital image analysis. Serum amylase had a peak at 12 hours, without differences between the sexes. In females, pancreatitis reached a peak at 12 hours with a fast recovery while, in males, the peak was delayed to day 2 with residual apoptosis still present. Macrophages were the main inflammatory cell population, followed by T cells, B cells and neutrophils, without differences between sexes. In males, CD206+ cells and apoptosis were higher at both days 2 and 7, and cytokeratin-19+ (CK19+) ADM was higher at day 7 compared with females. The results of this study revealed a faster onset and resolution of caerulein-induced AP in female mice compared with male mice, supporting a sex-dependent modulation of acute pancreatitis.
Collapse
Affiliation(s)
- Luca Bertola
- University of Milan, Lodi, Italy
- Fondazione UNIMI, Milan, Italy
| | | | | | | | - Elena Monica Borroni
- University of Milan, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Benedetta Savino
- University of Milan, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Simone Canesi
- University of Milan, Lodi, Italy
- Fondazione UNIMI, Milan, Italy
| | - Laura Sala
- University of Milan, Lodi, Italy
- Fondazione UNIMI, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
2
|
Li B, Chu Y, Wang X, Meng P, Fang L, Tian ZB, Li X. Risk factors associated with pain and pain relief in patients with chronic pancreatitis. Postgrad Med J 2025:qgae185. [PMID: 39841129 DOI: 10.1093/postmj/qgae185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 10/08/2024] [Indexed: 01/23/2025]
Abstract
BACKGROUND Abdominal pain is one of the most prominent symptoms in patients with chronic pancreatitis (CP) and can manifest intermittently or persistently. The mechanism of pain is not yet clear, and no effective treatment is currently available. This study aimed to explore the risk factors for pain in patients with CP, which may provide new insights for developing effective pain control modalities. METHODS This clinical study was based on a single-centre research database that included 570 patients with CP. We compared the differences in baseline data, clinical characteristics, and psychophysiology traits between patients with and without pain. Subsequently, patients will be followed up to assess changes in their risk factors and explore their relationship with pain. RESULTS In the final risk factor model, young age (P = .031; odds ratio [OR] = 0.986 [0.973, 0.999]), prolonged disease duration (P < .001; OR = 1.307 [1.127, 1.516]), heavy smoking (P = .014; OR = 1.331 [1.060, 1.617]), alcohol consumption (P = .003; OR = 1.419 [1.127, 1.788]), low body mass index (P < .001; OR = 0.786 [0.703, 0.879]), pancreatic exocrine insufficiency (P = .040; OR = 1.683 [1.024, 2.767]), acute pancreatitis attacks (P = .027; OR = 1.759 [1.067, 2.902]), anxiety, and depression (P = .016; OR = 1.047 [1.009, 1.088]; P = .014; OR = 1.068 [1.013, 1.126]) were associated with CP pain. Reducing tobacco and alcohol intake (P = .001; OR = 2.367 [1.525, 4.637]; P = .024; OR = 2.011 [1.085, 3.199]), increasing the body mass index (P = .005; OR = 1.968 [1.265, 3.805]), and improving anxiety (P = .001; OR = 1.164 [1.081, 1.340]) were identified to be beneficial for pain relief. Compared to the effects on persistent pain, pancreatic enzyme supplementation (P = .004; OR = 1.794 [1.186, 2.502]) had a clear effect on intermittent pain in patients with CP. CONCLUSION We identified a multifactorial model of pain risk factors for CP and confirmed that modifying these risk factors could influence patient pain symptoms.
Collapse
Affiliation(s)
- Bingqing Li
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao 266075, China
| | - Yuning Chu
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao 266075, China
| | - Xiaowei Wang
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao 266075, China
| | - Pin Meng
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao 266075, China
| | - Liang Fang
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao 266075, China
| | - Zi-Bin Tian
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao 266075, China
| | - Xiaoyu Li
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao 266075, China
| |
Collapse
|
3
|
Trinh VQH, Ankenbauer KE, Torbit SM, Liu J, Batardiere M, Kumar B, Maurer HC, Revetta F, Chen Z, Kruse A, Judd A, Copeland C, Wong J, Ben-Levy O, Jarvis B, Brown M, Brown JW, Das K, Makino Y, Spraggins JM, Lau K, Azadi P, Maitra A, Tan MCB, DelGiorno KE. Mutant GNAS drives a pyloric metaplasia with tumor suppressive glycans in intraductal papillary mucinous neoplasia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.25.581948. [PMID: 38464029 PMCID: PMC10925208 DOI: 10.1101/2024.02.25.581948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
BACKGROUND & AIMS Intraductal Papillary Mucinous Neoplasms (IPMNs) are cystic lesions and bona fide precursors for pancreatic ductal adenocarcinoma (PDAC). Recent studies have shown that pancreatic precancer is characterized by a transcriptomic program similar to gastric metaplasia. The aims of this study were to assay IPMN for pyloric markers, to identify molecular drivers, and to determine a functional role for this program in the pancreas. METHODS Pyloric marker expression was evaluated by RNA-seq and multiplex immunostaining in patient samples. Cell lines and organoids expressing KrasG12D +/- GNASR201C underwent RNA sequencing. A PyScenic-based regulon analysis was performed to identify molecular drivers, and candidates were evaluated by RNA-seq, immunostaining, and small interfering RNA knockdown. Glycosylation profiling was performed to identify GNASR201C-driven changes. Glycan abundance was evaluated in patient samples. RESULTS Pyloric markers were identified in human IPMN. GNASR201C drove expression of this program as well as an indolent phenotype characterized by distinct glycosyltransferase changes. Glycan profiling identified an increase in LacdiNAcs and loss of pro-tumorigenic Lewis antigens. Knockdown of transcription factors Spdef or Creb3l1 or chitinase treatment reduced LacdiNAc deposition and reversed the indolent phenotype. LacdiNAc and 3-sulfoLeA/C abundance discriminated low from high grade patient IPMN. CONCLUSION GNASR201C drives an indolent phenotype in IPMN by amplifying a differentiated, pyloric phenotype through SPDEF/CREB3L1 which is characterized by distinct glycans. Acting as a glycan rheostat, mutant GNAS elevates LacdiNAcs at the expense of pro-tumorigenic acidic Lewis epitopes, inhibiting cancer cell invasion and disease progression. LacdiNAc and 3-Sulfo-LeA/C are mutually exclusive and may serve as markers of disease progression.
Collapse
|
4
|
Góes VCD, Brandão-Bezerra L, Neves RH, Oliveira AVD, Machado-Silva JR. Impact of acute schistosomiasis mansoni and concurrent type 1 diabetes on pancreatic architecture in mice. Exp Parasitol 2024; 268:108885. [PMID: 39725378 DOI: 10.1016/j.exppara.2024.108885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/13/2024] [Accepted: 12/24/2024] [Indexed: 12/28/2024]
Abstract
It is not well understood how type 1 diabetes (T1D) and concomitant acute schistosomiasis mansoni affect pancreatic architecture. Male Swiss mice were administered streptozotocin (single 100 mg/kg i.p.) and thirty days later infected with 80 Schistosoma mansoni cercariae. Mice were divided into groups (n = 5): A (healthy control), B (infected), C (uninfected diabetic), and D (diabetic + infected) and euthanized at week 9 post-infection. Blood glucose levels, biometry, stereology, and pancreatic histology were evaluated. Groups C and D showed hyperglycemia (>200 mg/dL). Group B had a higher (+79%) pancreatic mass than A. The endocrine pancreas showed fewer islets of Langerhans (-62%; -50%) and a smaller islet area (-36%; -30%) in C and D, respectively, compared to A. Group D had a smaller (-37%) islet area than B. The volume density of the islets was reduced (-33%) in group C compared to A. Within the exocrine pancreas, the volume density of the pancreatic parenchyma was reduced in groups B (-29%) and D (-26%), and increased in C (+15%) compared to A. Group D was reduced (-35%) compared to C. Group D showed generalized pancreatitis, including disrupted tissue with multiple nuclei of destroyed acinar cells and lost connective tissue and acinar cells with a paucity of zymogen granules. Pancreatic stellate cells were found around areas of distorted architecture. Paired adult worms were found within the pancreatic vessels. In conclusion, concomitant T1D and schistosomiasis mansoni promote extensive exocrine and endocrine changes in the pancreas, whereas pancreatic involvement begins in acute schistosomiasis.
Collapse
Affiliation(s)
- Vanessa Coelho de Góes
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Luciana Brandão-Bezerra
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Renata Heisler Neves
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil.
| | - Albanita Viana de Oliveira
- Department of Pathology and Laboratories, Medical Sciences College (FCM), Rio de Janeiro State University, Brazil
| | - José Roberto Machado-Silva
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Dai C, Zhang Y, Gong Y, Bradley A, Tang Z, Sellick K, Shrestha S, Spears E, Covington BA, Stanley J, Jenkins R, Richardson TM, Brantley RA, Coate K, Saunders DC, Wright JJ, Brissova M, Dean ED, Powers AC, Chen W. Hyperaminoacidemia from interrupted glucagon signaling increases pancreatic acinar cell proliferation and size via mTORC1 and YAP pathways. iScience 2024; 27:111447. [PMID: 39720531 PMCID: PMC11667045 DOI: 10.1016/j.isci.2024.111447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/28/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024] Open
Abstract
Increased blood amino acid levels (hyperaminoacidemia) stimulate pancreas expansion by unclear mechanisms. Here, by genetic and pharmacological disruption of glucagon receptor (GCGR) in mice and zebrafish, we found that the ensuing hyperaminoacidemia promotes pancreatic acinar cell proliferation and cell hypertrophy, which can be mitigated by a low protein diet in mice. In addition to mammalian target of rapamycin complex 1 (mTORC1) signaling, acinar cell proliferation required slc38a5, the most highly expressed amino acid transporter gene in both species. Transcriptomics data revealed the activation signature of yes-associated protein (YAP) in acinar cells of mice with hyperaminoacidemia, consistent with the observed increase in YAP-expressing acinar cells. Yap1 activation also occurred in acinar cells in gcgr-/- zebrafish, which was reversed by rapamycin. Knocking down yap1 in gcgr-/- zebrafish decreased mTORC1 activity and acinar cell proliferation and hypertrophy. Thus, the study discovered a previously unrecognized role of the YAP/Taz pathway in hyperaminoacidemia-induced acinar cell hypertrophy and hyperplasia.
Collapse
Affiliation(s)
- Chunhua Dai
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yue Zhang
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Yulong Gong
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Amber Bradley
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zihan Tang
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Katelyn Sellick
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shristi Shrestha
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erick Spears
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brittney A. Covington
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jade Stanley
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Regina Jenkins
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tiffany M. Richardson
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Rebekah A. Brantley
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katie Coate
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Diane C. Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jordan J. Wright
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Marcela Brissova
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - E. Danielle Dean
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Alvin C. Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Wenbiao Chen
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
6
|
Alver CG, Dominguez-Bendala J, Agarwal A. Engineered tools to study endocrine dysfunction of pancreas. BIOPHYSICS REVIEWS 2024; 5:041303. [PMID: 39449867 PMCID: PMC11498943 DOI: 10.1063/5.0220396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Pancreas, a vital organ with intricate endocrine and exocrine functions, is central to the regulation of the body's glucose levels and digestive processes. Disruptions in its endocrine functions, primarily regulated by islets of Langerhans, can lead to debilitating diseases such as diabetes mellitus. Murine models of pancreatic dysfunction have contributed significantly to the understanding of insulitis, islet-relevant immunological responses, and the optimization of cell therapies. However, genetic differences between mice and humans have severely limited their clinical translational relevance. Recent advancements in tissue engineering and microfabrication have ushered in a new era of in vitro models that offer a promising solution. This paper reviews the state-of-the-art engineered tools designed to study endocrine dysfunction of the pancreas. Islet on a chip devices that allow precise control of various culture conditions and noninvasive readouts of functional outcomes have led to the generation of physiomimetic niches for primary and stem cell derived islets. Live pancreatic slices are a new experimental tool that could more comprehensively recapitulate the complex cellular interplay between the endocrine and exocrine parts of the pancreas. Although a powerful tool, live pancreatic slices require more complex control over their culture parameters such as local oxygenation and continuous removal of digestive enzymes and cellular waste products for maintaining experimental functionality over long term. The combination of islet-immune and slice on chip strategies can guide the path toward the next generation of pancreatic tissue modeling for better understanding and treatment of endocrine pancreatic dysfunctions.
Collapse
Affiliation(s)
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Ashutosh Agarwal
- Author to whom correspondence should be addressed:. Tel.: +1 305 243-8925
| |
Collapse
|
7
|
Dai Y, Guan X, Guo F, Kong X, Ji S, Shang D, Bai C, Zhang Q, Zhao L. Botanical drugs and their natural compounds: a neglected treasury for inhibiting the carcinogenesis of pancreatic ductal adenocarcinoma. PHARMACEUTICAL BIOLOGY 2024; 62:853-873. [PMID: 39520705 PMCID: PMC11552278 DOI: 10.1080/13880209.2024.2421759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/25/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
CONTEXT Pancreatic ductal adenocarcinoma (PDAC), which is characterized by its malignant nature, presents challenges for early detection and is associated with a poor prognosis. Any strategy that can interfere with the beginning or earlier stage of PDAC greatly delays disease progression. In response to this intractable problem, the exploration of new drugs is critical to reduce the incidence of PDAC. OBJECTIVE In this study, we summarize the mechanisms of pancreatitis-induced PDAC and traditional Chinese medicine (TCM) theory and review the roles and mechanisms of botanical drugs and their natural compounds that can inhibit the process of pancreatitis-induced PDAC. METHODS With the keywords 'chronic pancreatitis', 'TCM', 'Chinese medicinal formulae', 'natural compounds', 'PDAC' and 'pancreatic cancer', we conducted an extensive literature search of the PubMed, Web of Science, and other databases to identify studies that effectively prevent PDAC in complex inflammatory microenvironments. RESULTS We summarized the mechanism of pancreatitis-induced PDAC. Persistent inflammatory microenvironments cause multiple changes in the pancreas itself, including tissue damage, abnormal cell differentiation, and even gene mutation. According to TCM, pancreatitis-induced PDAC is the process of 'dampness-heat obstructing the spleen and deficiency due to stagnation' induced by a variety of pathological factors. A variety of botanical drugs and their natural compounds, such as Chaihu classical formulae, flavonoids, phenolics, terpenoids, etc., may be potential drugs to interfere with the development of PDAC via reshaping the inflammatory microenvironment by improving tissue injury and pancreatic fibrosis. CONCLUSIONS Botanical drugs and their natural compounds show great potential for preventing PDAC in complex inflammatory microenvironments.
Collapse
Affiliation(s)
- Yunfei Dai
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xi Guan
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fangyue Guo
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xin Kong
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of pharmacy, Dalian Medical University, Dalian, China
| | - Shuqi Ji
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Changchuan Bai
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingkai Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Liang Zhao
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
8
|
Li AL, Sugiura K, Nishiwaki N, Suzuki K, Sadeghian D, Zhao J, Maitra A, Falvo D, Chandwani R, Pitarresi JR, Sims PA, Rustgi AK. FRA1 controls acinar cell plasticity during murine Kras G12D-induced pancreatic acinar to ductal metaplasia. Dev Cell 2024; 59:3025-3042.e7. [PMID: 39178842 PMCID: PMC11576252 DOI: 10.1016/j.devcel.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 04/17/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024]
Abstract
Acinar cells have been proposed as a cell-of-origin for pancreatic ductal adenocarcinoma (PDAC) after undergoing acinar-to-ductal metaplasia (ADM). ADM can be triggered by pancreatitis, causing acinar cells to de-differentiate to a ductal-like state. We identify FRA1 (gene name Fosl1) as the most active transcription factor during KrasG12D acute pancreatitis-mediated injury, and we have elucidated a functional role of FRA1 by generating an acinar-specific Fosl1 knockout mouse expressing KrasG12D. Using a gene regulatory network and pseudotime trajectory inferred from single-nuclei ATAC-seq and bulk RNA sequencing (RNA-seq), we hypothesized a regulatory model of the acinar-ADM-pancreatic intraepithelial neoplasia (PanIN) continuum and experimentally validated that Fosl1 knockout mice are delayed in the onset of ADM and neoplastic transformation. Our study also identifies that pro-inflammatory cytokines, such as granulocyte colony stimulating factor (G-CSF), can regulate FRA1 activity to modulate ADM. Our findings identify that FRA1 is a mediator of acinar cell plasticity and is critical for acinar cell de-differentiation and transformation.
Collapse
Affiliation(s)
- Alina L Li
- Divison of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kensuke Sugiura
- Divison of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Noriyuki Nishiwaki
- Divison of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kensuke Suzuki
- Divison of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of General Surgery, Chiba University, Chiba 260-0856, Japan
| | - Dorsay Sadeghian
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Zhao
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David Falvo
- Department of Surgery and of Cell and Developmental Biology, Meyer Cancer Center, Weill-Cornell Medicine, New York, NY 10065, USA
| | - Rohit Chandwani
- Department of Surgery and of Cell and Developmental Biology, Meyer Cancer Center, Weill-Cornell Medicine, New York, NY 10065, USA
| | - Jason R Pitarresi
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan School of Medicine, Worchester, MA 01655, USA
| | - Peter A Sims
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Anil K Rustgi
- Divison of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
9
|
Wang H, Gao J, Wen L, Huang K, Liu H, Zeng L, Zeng Z, Liu Y, Mo Z. Ion channels in acinar cells in acute pancreatitis: crosstalk of calcium, iron, and copper signals. Front Immunol 2024; 15:1444272. [PMID: 39606246 PMCID: PMC11599217 DOI: 10.3389/fimmu.2024.1444272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
The initial stages of acute pancreatitis (AP) are characterized by a significant event - acinar ductal metaplasia (ADM). This process is a crucial feature of both acute and chronic pancreatitis, serving as the first step in the development of pancreatic cancer. Ion channels are integral transmembrane proteins that play a pivotal role in numerous biological processes by modulating ion flux. In many diseases, the expression and activity of ion channels are often dysregulated. Metal ions, including calcium ions (Ca2+), ferrous ions (Fe2+), and Copper ions (Cu2+), assume a distinctive role in cellular metabolism. These ions possess specific biological properties relevant to cellular function. However, the interactions among these ions exacerbate the imbalance within the intracellular environment, resulting in cellular damage and influencing the progression of AP. A more in-depth investigation into the mechanisms by which these ions interact with acinar cells is essential for elucidating AP's pathogenesis and identifying novel therapeutic strategies. Currently, treatment for AP primarily focuses on pain relief, complications prevention, and prognosis improvement. There are limited specific treatments targeting acinous cell dedifferentiation or ion imbalance. This study aims to investigate potential therapeutic strategies by examining ion crosstalk within acinar cells in the context of acute pancreatitis.
Collapse
Affiliation(s)
- Hanli Wang
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Jianhua Gao
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Lingling Wen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kejun Huang
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Huixian Liu
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Linsheng Zeng
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Zhongyi Zeng
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yuxiang Liu
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Zhizhun Mo
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
10
|
Shields MA, Metropulos AE, Spaulding C, Alzahrani KA, Hirose T, Ohno S, Pham TND, Munshi HG. BET Inhibition Rescues Acinar-Ductal-Metaplasia and Ciliogenesis and Ameliorates Chronic Pancreatitis-Driven Changes in Mice With Loss of the Polarity Protein Par3. Cell Mol Gastroenterol Hepatol 2024; 18:101389. [PMID: 39128653 PMCID: PMC11437875 DOI: 10.1016/j.jcmgh.2024.101389] [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: 12/22/2023] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND & AIMS The apical-basal polarity of pancreatic acinar cells is essential for maintaining tissue architecture. However, the mechanisms by which polarity proteins regulate acinar pancreas injury and regeneration are poorly understood. METHODS Cerulein-induced pancreatitis was induced in mice with conditional deletion of the polarity protein Par3 in the pancreas. The impact of Par3 loss on pancreas injury and regeneration was assessed by histologic analyses and transcriptional profiling by RNA sequencing. Mice were pretreated with the bromodomain and extraterminal domain (BET) inhibitor JQ1 before cotreatment with cerulein to determine the effect of BET inhibition on pancreas injury and regeneration. RESULTS Initially, we show that Par3 is increased in acinar-ductal metaplasia (ADM) lesions present in human and mouse chronic pancreatitis specimens. Although Par3 loss disrupts tight junctions, Par3 is dispensable for pancreatogenesis. However, with aging, Par3 loss results in low-grade inflammation, acinar degeneration, and pancreatic lipomatosis. Par3 loss exacerbates acute pancreatitis-induced injury and chronic pancreatitis-induced acinar cell loss, promotes pancreatic lipomatosis, and prevents regeneration. Par3 loss also results in suppression of chronic pancreatitis-induced ADM and primary ciliogenesis. Notably, targeting BET proteins attenuates chronic pancreatitis-induced loss of primary cilia and promotes ADM in mice lacking pancreatic Par3. Targeting BET proteins also attenuates cerulein-induced acinar cell loss and enhances recovery of acinar cell mass and body weight of mice lacking pancreatic Par3. CONCLUSIONS Combined, this study demonstrates how Par3 restrains chronic pancreatitis-induced changes in the pancreas and identifies a potential role for BET inhibitors to attenuate pancreas injury and facilitate regeneration.
Collapse
Affiliation(s)
- Mario A Shields
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; The Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois.
| | - Anastasia E Metropulos
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Jesse Brown VA Medical Center, Chicago, Illinois
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Jesse Brown VA Medical Center, Chicago, Illinois
| | - Khulood A Alzahrani
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Tomonori Hirose
- Department of Molecular Biology, Yokohama City University School of Medicine, Yokohama, Japan; Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shigeo Ohno
- Department of Molecular Biology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Thao N D Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; The Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois
| | - Hidayatullah G Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; The Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois; Jesse Brown VA Medical Center, Chicago, Illinois.
| |
Collapse
|
11
|
Neuß T, Chen MC, Wirges N, Usluer S, Oellinger R, Lier S, Dudek M, Madl T, Jastroch M, Steiger K, Schmitz W, Einwächter H, Schmid RM. Metabolic Reprogramming Is an Initial Step in Pancreatic Carcinogenesis That Can Be Targeted to Inhibit Acinar-to-Ductal Metaplasia. Cancer Res 2024; 84:2297-2312. [PMID: 39005053 DOI: 10.1158/0008-5472.can-23-2213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 02/03/2024] [Accepted: 05/01/2024] [Indexed: 07/16/2024]
Abstract
Metabolic reprogramming is a hallmark of cancer and is crucial for cancer progression, making it an attractive therapeutic target. Understanding the role of metabolic reprogramming in cancer initiation could help identify prevention strategies. To address this, we investigated metabolism during acinar-to-ductal metaplasia (ADM), the first step of pancreatic carcinogenesis. Glycolytic markers were elevated in ADM lesions compared with normal tissue from human samples. Comprehensive metabolic assessment in three mouse models with pancreas-specific activation of KRAS, PI3K, or MEK1 using Seahorse measurements, nuclear magnetic resonance metabolome analysis, mass spectrometry, isotope tracing, and RNA sequencing analysis revealed a switch from oxidative phosphorylation to glycolysis in ADM. Blocking the metabolic switch attenuated ADM formation. Furthermore, mitochondrial metabolism was required for de novo synthesis of serine and glutathione (GSH) but not for ATP production. MYC mediated the increase in GSH intermediates in ADM, and inhibition of GSH synthesis suppressed ADM development. This study thus identifies metabolic changes and vulnerabilities in the early stages of pancreatic carcinogenesis. Significance: Metabolic reprogramming from oxidative phosphorylation to glycolysis mediated by MYC plays a crucial role in the development of pancreatic cancer, revealing a mechanism driving tumorigenesis and potential therapeutic targets. See related commentary by Storz, p. 2225.
Collapse
Affiliation(s)
- Thorsten Neuß
- Department of Clinical Medicine-Clinical Department for Internal Medicine II, TUM School of Medicine and Health, University Medical Center, Technical University of Munich, Munich, Germany
| | - Min-Chun Chen
- Department of Clinical Medicine-Clinical Department for Internal Medicine II, TUM School of Medicine and Health, University Medical Center, Technical University of Munich, Munich, Germany
| | - Nils Wirges
- Technical University of Munich, TUM School of Medicine and Health, Institute of Pathology, Comparative Experimental Pathology, Munich, Germany
| | - Sinem Usluer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, BioTechMed-Graz, Graz, Austria
| | - Rupert Oellinger
- TUM School of Medicine and Health, Institute of Molecular Oncology and Functional Genomics, Technical University of Munich, Munich, Germany
| | - Svenja Lier
- Department of Clinical Medicine-Clinical Department for Internal Medicine II, TUM School of Medicine and Health, University Medical Center, Technical University of Munich, Munich, Germany
| | - Michael Dudek
- TUM School of Medicine and Health, Institute of Molecular Immunology and Experimental Oncology, University Medical Center, Technical University of Munich, Munich, Germany
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, BioTechMed-Graz, Graz, Austria
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Katja Steiger
- Technical University of Munich, TUM School of Medicine and Health, Institute of Pathology, Comparative Experimental Pathology, Munich, Germany
| | - Werner Schmitz
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Henrik Einwächter
- Department of Clinical Medicine-Clinical Department for Internal Medicine II, TUM School of Medicine and Health, University Medical Center, Technical University of Munich, Munich, Germany
| | - Roland M Schmid
- Department of Clinical Medicine-Clinical Department for Internal Medicine II, TUM School of Medicine and Health, University Medical Center, Technical University of Munich, Munich, Germany
| |
Collapse
|
12
|
Bejjani J, Ramsey ML, Lee PJ, Phillips AE, Singh VK, Yadav D, Papachristou GI, Hart PA. Alterations in exocrine pancreatic function after acute pancreatitis. Pancreatology 2024; 24:505-510. [PMID: 38485543 PMCID: PMC11215795 DOI: 10.1016/j.pan.2024.03.003] [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: 01/03/2024] [Revised: 02/22/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
Exocrine pancreatic dysfunction (EPD) is a malabsorptive complication of pancreatic disorders that can lead to a host of symptoms ranging from flatulence to diarrhea and contribute to weight loss and metabolic bone disease. It is increasingly recognized to occur after acute pancreatitis (AP), including episodes with mild severity. The risk of developing EPD after AP is influenced by a range of factors, including the degree of acinar cell destruction and inflammation during AP, and persistent structural derangements following AP. In this article, we discuss the epidemiology, pathophysiology, and clinical management of EPD after AP while highlighting key knowledge gaps.
Collapse
Affiliation(s)
- Joseph Bejjani
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mitchell L Ramsey
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Peter J Lee
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Anna Evans Phillips
- Division of Gastroenterology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Vikesh K Singh
- Division of Gastroenterology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Dhiraj Yadav
- Division of Gastroenterology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Georgios I Papachristou
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
13
|
Waldron RT, Lugea A, Chang HH, Su HY, Quiros C, Lewis MS, Che M, Ramanujan VK, Rozengurt E, Eibl G, Pandol SJ. Upregulated Matrisomal Proteins and Extracellular Matrix Mechanosignaling Underlie Obesity-Associated Promotion of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:1593. [PMID: 38672675 PMCID: PMC11048773 DOI: 10.3390/cancers16081593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Diet-induced obesity (DIO) promotes pancreatic ductal adenocarcinoma (PDAC) in mice expressing KRasG12D in the pancreas (KC mice), but the precise mechanisms remain unclear. Here, we performed multiplex quantitative proteomic and phosphoproteomic analysis by liquid chromatography-tandem mass spectrometry and further bioinformatic and spatial analysis of pancreas tissues from control-fed versus DIO KC mice after 3, 6, and 9 months. Normal pancreatic parenchyma and associated proteins were steadily eliminated and the novel proteins, phosphoproteins, and signaling pathways associated with PDAC tumorigenesis increased until 6 months, when most males exhibited cancer, but females did not. Differentially expressed proteins and phosphoproteins induced by DIO revealed the crucial functional role of matrisomal proteins, which implies the roles of upstream regulation by TGFβ, extracellular matrix-receptor signaling to downstream PI3K-Akt-mTOR-, MAPK-, and Yap/Taz activation, and crucial effects in the tumor microenvironment such as metabolic alterations and signaling crosstalk between immune cells, cancer-associated fibroblasts (CAFs), and tumor cells. Staining tissues from KC mice localized the expression of several prognostic PDAC biomarkers and elucidated tumorigenic features, such as robust macrophage infiltration, acinar-ductal metaplasia, mucinous PanIN, distinct nonmucinous atypical flat lesions (AFLs) surrounded by smooth muscle actin-positive CAFs, invasive tumors with epithelial-mesenchymal transition arising close to AFLs, and expanding deserted areas by 9 months. We next used Nanostring GeoMX to characterize the early spatial distribution of specific immune cell subtypes in distinct normal, stromal, and PanIN areas. Taken together, these data richly contextualize DIO promotion of Kras-driven PDAC tumorigenesis and provide many novel insights into the signaling pathways and processes involved.
Collapse
Affiliation(s)
- Richard T. Waldron
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Aurelia Lugea
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hui-Hua Chang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Hsin-Yuan Su
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Crystal Quiros
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Michael S. Lewis
- Department of Medicine and Department of Pathology & Laboratory Medicine, VA Greater Los Angeles Health System, Cedars-Sinai Medical Center, Los Angeles, CA 90073, USA;
| | - Mingtian Che
- Biobank and Research Pathology Resource, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - V. Krishnan Ramanujan
- Biobank and Research Pathology Resource, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Stephen J. Pandol
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
14
|
Fleming Martinez AK, Storz P. Protein kinase D1 - A targetable mediator of pancreatic cancer development. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119646. [PMID: 38061566 PMCID: PMC10872883 DOI: 10.1016/j.bbamcr.2023.119646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/17/2023] [Accepted: 11/30/2023] [Indexed: 01/14/2024]
Abstract
Members of the Protein kinase D (PKD) kinase family each play important cell-specific roles in the regulation of normal pancreas functions. In pancreatic diseases PKD1 is the most widely characterized isoform with roles in pancreatitis and in induction of pancreatic cancer and its progression. PKD1 expression and activation increases in pancreatic acinar cells through macrophage secreted factors, Kirsten rat sarcoma viral oncogene homolog (KRAS) signaling, and reactive oxygen species (ROS), driving the formation of precancerous lesions. In precancerous lesions PKD1 regulates cell survival, growth, senescence, and generation of doublecortin like kinase 1 (DCLK1)-positive cancer stem cells (CSCs). Within tumors, regulation by PKD1 includes chemoresistance, apoptosis, proliferation, CSC features, and the Warburg effect. Thus, PKD1 plays a critical role throughout pancreatic disease initiation and progression.
Collapse
Affiliation(s)
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
15
|
Torborg SR, Grbovic-Huezo O, Singhal A, Holm M, Wu K, Han X, Ho YJ, Haglund C, Mitchell MJ, Lowe SW, Dow LE, Pitter KL, Sanchez-Rivera FJ, Levchenko A, Tammela T. Solid tumor growth depends on an intricate equilibrium of malignant cell states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.30.573100. [PMID: 38234855 PMCID: PMC10793475 DOI: 10.1101/2023.12.30.573100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Control of cell identity and number is central to tissue function, yet principles governing organization of malignant cells in tumor tissues remain poorly understood. Using mathematical modeling and candidate-based analysis, we discover primary and metastatic pancreatic ductal adenocarcinoma (PDAC) organize in a stereotypic pattern whereby PDAC cells responding to WNT signals (WNT-R) neighbor WNT-secreting cancer cells (WNT-S). Leveraging lineage-tracing, we reveal the WNT-R state is transient and gives rise to the WNT-S state that is highly stable and committed to organizing malignant tissue. We further show that a subset of WNT-S cells expressing the Notch ligand DLL1 form a functional niche for WNT-R cells. Genetic inactivation of WNT secretion or Notch pathway components, or cytoablation of the WNT-S state disrupts PDAC tissue organization, suppressing tumor growth and metastasis. This work indicates PDAC growth depends on an intricately controlled equilibrium of functionally distinct cancer cell states, uncovering a fundamental principle governing solid tumor growth and revealing new opportunities for therapeutic intervention.
Collapse
|
16
|
Doke M, Álvarez-Cubela S, Klein D, Altilio I, Schulz J, Mateus Gonçalves L, Almaça J, Fraker CA, Pugliese A, Ricordi C, Qadir MMF, Pastori RL, Domínguez-Bendala J. Dynamic scRNA-seq of live human pancreatic slices reveals functional endocrine cell neogenesis through an intermediate ducto-acinar stage. Cell Metab 2023; 35:1944-1960.e7. [PMID: 37898119 DOI: 10.1016/j.cmet.2023.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 08/23/2023] [Accepted: 10/03/2023] [Indexed: 10/30/2023]
Abstract
Human pancreatic plasticity is implied from multiple single-cell RNA sequencing (scRNA-seq) studies. However, these have been invariably based on static datasets from which fate trajectories can only be inferred using pseudotemporal estimations. Furthermore, the analysis of isolated islets has resulted in a drastic underrepresentation of other cell types, hindering our ability to interrogate exocrine-endocrine interactions. The long-term culture of human pancreatic slices (HPSs) has presented the field with an opportunity to dynamically track tissue plasticity at the single-cell level. Combining datasets from same-donor HPSs at different time points, with or without a known regenerative stimulus (BMP signaling), led to integrated single-cell datasets storing true temporal or treatment-dependent information. This integration revealed population shifts consistent with ductal progenitor activation, blurring of ductal/acinar boundaries, formation of ducto-acinar-endocrine differentiation axes, and detection of transitional insulin-producing cells. This study provides the first longitudinal scRNA-seq analysis of whole human pancreatic tissue, confirming its plasticity in a dynamic fashion.
Collapse
Affiliation(s)
- Mayur Doke
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Dagmar Klein
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Isabella Altilio
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joseph Schulz
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Luciana Mateus Gonçalves
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Christopher A Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alberto Pugliese
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mirza M F Qadir
- Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ricardo L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
17
|
Yin H, Zhang Z, Zhang D, Peng L, Xia C, Yang X, Wang X, Li Z, Chang J, Huang H. A new method for treating chronic pancreatitis and preventing fibrosis using bioactive calcium silicate ion solution. J Mater Chem B 2023; 11:9163-9178. [PMID: 37642526 DOI: 10.1039/d3tb01287e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Chronic pancreatitis (CP) is a multifactorial fibroinflammatory syndrome. At present, there is no effective way to treat it clinically. In this study, we proposed a new approach by application of a highly active calcium silicate ion solution derived from calcium silicate (CS) bioceramics, which effectively inhibited the development of CP. This bioceramic derived bioactive ionic solution mainly regulated pancreatic acinar cells (PACs), macrophages and pancreatic stellate cells (PSCs) by SiO32- ions to inhibit inflammation and fibrosis and promote acinar regeneration. The possible mechanism of the therapeutic effect of CS ion solution mainly includes the inhibition of PAC apoptosis by down-regulating the c-caspase3 signal pathway and promotion of the regeneration of PACs by up-regulating the WNT/β-catenin signaling pathway. In addition, the CS ion solution also effectively down-regulated the NF-κB signaling pathway to reduce macrophage infiltration and PAC inflammatory factor secretion, thereby reducing PSC mediated pancreatic fibrosis. This bioceramics-based ion solution provides a new idea for disease treatment using biomaterials, which may have the potential for the development of new therapy for CP.
Collapse
Affiliation(s)
- Hua Yin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, People's Republic of China.
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Ningxia, 750004, People's Republic of China
| | - Zhaowenbin Zhang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People's Republic of China
| | - Deyu Zhang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, People's Republic of China.
| | - Lisi Peng
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, People's Republic of China.
| | - Chuanchao Xia
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, People's Republic of China.
| | - Xiaoli Yang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, People's Republic of China.
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Ningxia, 750004, People's Republic of China
| | - Xinyue Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, People's Republic of China.
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, People's Republic of China.
| | - Jiang Chang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People's Republic of China
| | - Haojie Huang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, People's Republic of China.
| |
Collapse
|
18
|
Azevedo-Pouly A, Hale MA, Swift GH, Hoang CQ, Deering TG, Xue J, Wilkie TM, Murtaugh LC, MacDonald RJ. Key transcriptional effectors of the pancreatic acinar phenotype and oncogenic transformation. PLoS One 2023; 18:e0291512. [PMID: 37796967 PMCID: PMC10553828 DOI: 10.1371/journal.pone.0291512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/30/2023] [Indexed: 10/07/2023] Open
Abstract
Proper maintenance of mature cellular phenotypes is essential for stable physiology, suppression of disease states, and resistance to oncogenic transformation. We describe the transcriptional regulatory roles of four key DNA-binding transcription factors (Ptf1a, Nr5a2, Foxa2 and Gata4) that sit at the top of a regulatory hierarchy controlling all aspects of a highly differentiated cell-type-the mature pancreatic acinar cell (PAC). Selective inactivation of Ptf1a, Nr5a2, Foxa2 and Gata4 individually in mouse adult PACs rapidly altered the transcriptome and differentiation status of PACs. The changes most emphatically included transcription of the genes for the secretory digestive enzymes (which conscript more than 90% of acinar cell protein synthesis), a potent anabolic metabolism that provides the energy and materials for protein synthesis, suppressed and properly balanced cellular replication, and susceptibility to transformation by oncogenic KrasG12D. The simultaneous inactivation of Foxa2 and Gata4 caused a greater-than-additive disruption of gene expression and uncovered their collaboration to maintain Ptf1a expression and control PAC replication. A measure of PAC dedifferentiation ranked the effects of the conditional knockouts as Foxa2+Gata4 > Ptf1a > Nr5a2 > Foxa2 > Gata4. Whereas the loss of Ptf1a or Nr5a2 greatly accelerated Kras-mediated transformation of mature acinar cells in vivo, the absence of Foxa2, Gata4, or Foxa2+Gata4 together blocked transformation completely, despite extensive dedifferentiation. A lack of correlation between PAC dedifferentiation and sensitivity to oncogenic KrasG12D negates the simple proposition that the level of differentiation determines acinar cell resistance to transformation.
Collapse
Affiliation(s)
- Ana Azevedo-Pouly
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Michael A. Hale
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Galvin H. Swift
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Chinh Q. Hoang
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Tye G. Deering
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jumin Xue
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Thomas M. Wilkie
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - L. Charles Murtaugh
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States of America
| | - Raymond J. MacDonald
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
19
|
Fujiwara S, Moroki T, Hitomi M, Sato M, Terayama Y, Yoshikawa T. Pathological analysis of lesions in the exocrine pancreas of rats induced by Zinc Maltol. J Toxicol Pathol 2023; 36:205-211. [PMID: 37868114 PMCID: PMC10585244 DOI: 10.1293/tox.2023-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/27/2023] [Indexed: 10/24/2023] Open
Abstract
The pancreas plays an important role in the homeostasis of zinc (Zn), a nutritionally essential metal. In several previous studies, Zn ions induced inflammatory changes in the exocrine pancreas; however, little is known about Zn complexes. In this study, we microscopically, immunohistochemically, and ultrastructurally examined pancreatic lesions in Sprague-Dawley (SD) rats induced by a 4-week repeated oral dose toxicity study of Zinc Maltol (ZM), a zinc (II) complex. ZM induces acinar atrophy and increases the number of duct-like structures. Immunohistochemistry revealed a decrease in the number of trypsin-positive cells, and an increase in the number of SOX9-positive cells. Interstitial fibrosis and macrophage infiltration also correlated with the degree of acinar atrophy. Electron microscopic evaluation revealed that the acinar cells that lost granules were surrounded by fibroblasts and collagen fibers. In conclusion, we provided a detailed description of ZM-induced pancreatic lesions in SD rats.
Collapse
Affiliation(s)
- Sakura Fujiwara
- Department of Drug Safety Research, Preclinical Research,
Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., 463-10
Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima 771-0192, Japan
| | - Takayasu Moroki
- Department of Drug Safety Research, Preclinical Research,
Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., 463-10
Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima 771-0192, Japan
| | - Masaya Hitomi
- Department of Drug Safety Research, Preclinical Research,
Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., 463-10
Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima 771-0192, Japan
| | - Makoto Sato
- Department of Drug Safety Research, Preclinical Research,
Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., 463-10
Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima 771-0192, Japan
| | - Yui Terayama
- Department of Drug Safety Research, Preclinical Research,
Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., 463-10
Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima 771-0192, Japan
| | - Tsuyoshi Yoshikawa
- Department of Drug Safety Research, Preclinical Research,
Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., 463-10
Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima 771-0192, Japan
| |
Collapse
|
20
|
Shields MA, Metropulos AE, Spaulding C, Hirose T, Ohno S, Pham TN, Munshi HG. BET inhibition rescues ciliogenesis and ameliorates pancreatitis-driven phenotypic changes in mice with Par3 loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.14.557654. [PMID: 37745543 PMCID: PMC10515915 DOI: 10.1101/2023.09.14.557654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The apical-basal polarity of pancreatic acinar cells is essential for maintaining tissue architecture. However, the mechanisms by which polarity proteins regulate acinar pancreas tissue homeostasis are poorly understood. Here, we evaluate the role of Par3 in acinar pancreas injury and homeostasis. While Par3 loss in the mouse pancreas disrupts tight junctions, Par3 loss is dispensable for pancreatogenesis. However, with aging, Par3 loss results in low-grade inflammation, acinar degeneration, and pancreatic lipomatosis. Par3 loss also exacerbates pancreatitis-induced acinar cell loss, resulting in pronounced pancreatic lipomatosis and failure to regenerate. Moreover, Par3 loss in mice harboring mutant Kras causes extensive pancreatic intraepithelial neoplastic (PanIN) lesions and large pancreatic cysts. We also show that Par3 loss restricts injury-induced primary ciliogenesis. Significantly, targeting BET proteins enhances primary ciliogenesis during pancreatitis-induced injury and, in mice with Par3 loss, limits pancreatitis-induced acinar loss and facilitates acinar cell regeneration. Combined, this study demonstrates how Par3 restrains pancreatitis- and Kras-induced changes in the pancreas and identifies a potential role for BET inhibitors to attenuate pancreas injury and facilitate pancreas tissue regeneration.
Collapse
Affiliation(s)
- Mario A. Shields
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Anastasia E. Metropulos
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Tomonori Hirose
- Department of Molecular Biology, Yokohama City University School of Medicine, Yokohama, Japan
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shigeo Ohno
- Department of Molecular Biology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Thao N.D. Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Hidayatullah G. Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
21
|
Lopez K, Deng JJ, Xu Y, Sharkey FE, Wang P, Liu J. Exploring the Role of YAP1 and TAZ in Pancreatic Acinar Cells and the Therapeutic Potential of VT-104 in Pancreatic Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558321. [PMID: 37781601 PMCID: PMC10541090 DOI: 10.1101/2023.09.18.558321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Increasing evidences have linked the hippo pathway with the fibroinflammatory diseases. We generated a series of genetic knockout mice for targeting the key components of Hippo pathway to examine the individual effects of YAP1 and TAZ on pancreatic inflammation and evaluated the therapeutic potential of the YAP1/TAZ inhibitor VT-104. Mice with acinar-specific knockout of YAP1/TAZ did not exhibit any histological abnormalities in the pancreas. LATS1/2 deficiency induced acinar-to-ductal metaplasia, immune cell infiltration and fibroblast activation, which were rescued by the homozygous knockout YAP1, but not TAZ. Additionally, treatment with VT-104 also decreased pathological alterations induced by deletions of LATS1 and LATS2 in acinar cells. Our findings highlight the critical role of YAP1 in modulating pancreatic inflammation and demonstrate that VT-104 holds therapeutic potential to mitigate pancreatitis-associated pathological manifestations. Further exploration is necessary to unravel the underlying mechanisms and translate these insights into clinical applications.
Collapse
|
22
|
Liu X, Liu X, Du Y, Zou D, Tian C, Li Y, Lan X, David CJ, Sun Q, Chen M. Aberrant accumulation of Kras-dependent pervasive transcripts during tumor progression renders cancer cells dependent on PAF1 expression. Cell Rep 2023; 42:112979. [PMID: 37572321 DOI: 10.1016/j.celrep.2023.112979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/05/2023] [Accepted: 07/31/2023] [Indexed: 08/14/2023] Open
Abstract
KRAS is the most commonly mutated oncogene in human cancer, and mutant KRAS is responsible for over 90% of pancreatic ductal adenocarcinoma (PDAC), the most lethal cancer. Here, we show that RNA polymerase II-associated factor 1 complex (PAF1C) is specifically required for survival of PDAC but not normal adult pancreatic cells. We show that PAF1C maintains cancer cell genomic stability by restraining overaccumulation of enhancer RNAs (eRNAs) and promoter upstream transcripts (PROMPTs) driven by mutant Kras. Loss of PAF1C leads to cancer-specific lengthening and accumulation of pervasive transcripts on chromatin and concomitant aberrant R-loop formation and DNA damage, which, in turn, trigger cell death. We go on to demonstrate that the global transcriptional hyperactivation driven by Kras signaling during tumorigenesis underlies the specific demand for PAF1C by cancer cells. Our work provides insights into how enhancer transcription hyperactivation causes general transcription factor addiction during tumorigenesis.
Collapse
Affiliation(s)
- Xinhong Liu
- State Key Laboratory of Molecular Oncology, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiangzheng Liu
- State Key Laboratory of Molecular Oncology, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yingxue Du
- Tsinghua University School of Life Sciences, Beijing 100084, China
| | - Di Zou
- State Key Laboratory of Molecular Oncology, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Chen Tian
- State Key Laboratory of Molecular Oncology, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yong Li
- State Key Laboratory of Molecular Oncology, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xun Lan
- State Key Laboratory of Molecular Oncology, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, School of Medicine, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Charles J David
- State Key Laboratory of Molecular Oncology, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, School of Medicine, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Qianwen Sun
- Tsinghua University School of Life Sciences, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Mo Chen
- State Key Laboratory of Molecular Oncology, SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, School of Medicine, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
23
|
Pan Z, Van den Bossche JL, Rodriguez-Aznar E, Janssen P, Lara O, Ates G, Massie A, De Paep DL, Houbracken I, Mambretti M, Rooman I. Pancreatic acinar cell fate relies on system x C- to prevent ferroptosis during stress. Cell Death Dis 2023; 14:536. [PMID: 37604805 PMCID: PMC10442358 DOI: 10.1038/s41419-023-06063-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/28/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023]
Abstract
Acinar cell dedifferentiation is one of the most notable features of acute and chronic pancreatitis. It can also be the initial step that facilitates pancreatic cancer development. In the present study, we further decipher the precise mechanisms and regulation using primary human cells and murine experimental models. Our RNAseq analysis indicates that, in both species, early acinar cell dedifferentiation is accompanied by multiple pathways related to cell survival that are highly enriched, and where SLC7A11 (xCT) is transiently upregulated. xCT is the specific subunit of the cystine/glutamate antiporter system xC-. To decipher its role, gene silencing, pharmacological inhibition and a knock-out mouse model were used. Acinar cells with depleted or reduced xCT function show an increase in ferroptosis relating to lipid peroxidation. Lower glutathione levels and more lipid ROS accumulation could be rescued by the antioxidant N-acetylcysteine or the ferroptosis inhibitor ferrostatin-1. In caerulein-induced acute pancreatitis in mice, xCT also prevents lipid peroxidation in acinar cells. In conclusion, during stress, acinar cell fate seems to be poised for avoiding several forms of cell death. xCT specifically prevents acinar cell ferroptosis by fueling the glutathione pool and maintaining ROS balance. The data suggest that xCT offers a druggable tipping point to steer the acinar cell fate in stress conditions.
Collapse
Affiliation(s)
- Zhaolong Pan
- Laboratory for Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jan-Lars Van den Bossche
- Laboratory for Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Rodriguez-Aznar
- Laboratory for Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pauline Janssen
- Laboratory for Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Neuro-Aging & Viro-Immunotherapy Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Olaya Lara
- Laboratory for Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Neuro-Aging & Viro-Immunotherapy Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Gamze Ates
- Neuro-Aging & Viro-Immunotherapy Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ann Massie
- Neuro-Aging & Viro-Immunotherapy Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Diedert Luc De Paep
- Beta Cell Bank, Universitair Ziekenhuis Brussel and Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Houbracken
- Laboratory for Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marco Mambretti
- Laboratory for Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ilse Rooman
- Laboratory for Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium.
- Visual and Spatial Tissue Analysis (VSTA) Core Facility, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
24
|
Jiang Z, Wu F, Laise P, Takayuki T, Na F, Kim W, Kobayashi H, Chang W, Takahashi R, Valenti G, Sunagawa M, White RA, Macchini M, Renz BW, Middelhoff M, Hayakawa Y, Dubeykovskaya ZA, Tan X, Chu TH, Nagar K, Tailor Y, Belin BR, Anand A, Asfaha S, Finlayson MO, Iuga AC, Califano A, Wang TC. Tff2 defines transit-amplifying pancreatic acinar progenitors that lack regenerative potential and are protective against Kras-driven carcinogenesis. Cell Stem Cell 2023; 30:1091-1109.e7. [PMID: 37541213 PMCID: PMC10414754 DOI: 10.1016/j.stem.2023.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 04/06/2023] [Accepted: 07/07/2023] [Indexed: 08/06/2023]
Abstract
While adult pancreatic stem cells are thought not to exist, it is now appreciated that the acinar compartment harbors progenitors, including tissue-repairing facultative progenitors (FPs). Here, we study a pancreatic acinar population marked by trefoil factor 2 (Tff2) expression. Long-term lineage tracing and single-cell RNA sequencing (scRNA-seq) analysis of Tff2-DTR-CreERT2-targeted cells defines a transit-amplifying progenitor (TAP) population that contributes to normal homeostasis. Following acute and chronic injury, Tff2+ cells, distinct from FPs, undergo depopulation but are eventually replenished. At baseline, oncogenic KrasG12D-targeted Tff2+ cells are resistant to PDAC initiation. However, KrasG12D activation in Tff2+ cells leads to survival and clonal expansion following pancreatitis and a cancer stem/progenitor cell-like state. Selective ablation of Tff2+ cells prior to KrasG12D activation in Mist1+ acinar or Dclk1+ FP cells results in enhanced tumorigenesis, which can be partially rescued by adenoviral Tff2 treatment. Together, Tff2 defines a pancreatic TAP population that protects against Kras-driven carcinogenesis.
Collapse
Affiliation(s)
- Zhengyu Jiang
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Feijing Wu
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA; The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Pasquale Laise
- Department of Systems Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA; DarwinHealth Inc., New York, NY, USA
| | - Tanaka Takayuki
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Fu Na
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Woosook Kim
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Hiroki Kobayashi
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Wenju Chang
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Ryota Takahashi
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Giovanni Valenti
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Masaki Sunagawa
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Ruth A White
- Division of Hematology and Oncology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Marina Macchini
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Bernhard W Renz
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of General, Visceral, and Transplantation Surgery, LMU University Hospital, LMU Munich, Germany
| | - Moritz Middelhoff
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Division of Digestive and Liver Diseases, CU and Klinikum rechts der Isar, Technical University, Munich, Germany
| | - Yoku Hayakawa
- Graduate School of Medicine, Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Zinaida A Dubeykovskaya
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Xiangtian Tan
- Department of Systems Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Timothy H Chu
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Karan Nagar
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Yagnesh Tailor
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Bryana R Belin
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Akanksha Anand
- Division of Digestive and Liver Diseases, Department of Medicine and Department of Gastroenterology II, Klinikum rechts der Isar, Technical University, Munich, Germany
| | - Samuel Asfaha
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Michael O Finlayson
- Department of Systems Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Alina C Iuga
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Andrea Califano
- Department of Systems Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA; DarwinHealth Inc., New York, NY, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
25
|
Melzer MK, Kleger A. Pancreatic acinar heterogeneity hijacks carcinogenesis and homeostasis. Cell Stem Cell 2023; 30:1003-1005. [PMID: 37541205 DOI: 10.1016/j.stem.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 08/06/2023]
Abstract
In this issue, Jiang and colleagues employ multiple lineage-tracing approaches to elaborate on the role of Tff2+ transit-amplifying progenitor cells in the pancreatic acinar compartment of mice. This work provides insights into the steady-state homeostasis and tumor-suppressive features of certain progenitor cells and presents findings on acinar cell heterogeneity.
Collapse
Affiliation(s)
- Michael Karl Melzer
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany; Department of Urology, Ulm University Hospital, Ulm, Germany
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany; Division of Interdisciplinary Pancreatology, Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany; Core Facility Organoids, Ulm University, Ulm, Germany.
| |
Collapse
|
26
|
Marstrand-Daucé L, Lorenzo D, Chassac A, Nicole P, Couvelard A, Haumaitre C. Acinar-to-Ductal Metaplasia (ADM): On the Road to Pancreatic Intraepithelial Neoplasia (PanIN) and Pancreatic Cancer. Int J Mol Sci 2023; 24:9946. [PMID: 37373094 PMCID: PMC10298625 DOI: 10.3390/ijms24129946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Adult pancreatic acinar cells show high plasticity allowing them to change in their differentiation commitment. Pancreatic acinar-to-ductal metaplasia (ADM) is a cellular process in which the differentiated pancreatic acinar cells transform into duct-like cells. This process can occur as a result of cellular injury or inflammation in the pancreas. While ADM is a reversible process allowing pancreatic acinar regeneration, persistent inflammation or injury can lead to the development of pancreatic intraepithelial neoplasia (PanIN), which is a common precancerous lesion that precedes pancreatic ductal adenocarcinoma (PDAC). Several factors can contribute to the development of ADM and PanIN, including environmental factors such as obesity, chronic inflammation and genetic mutations. ADM is driven by extrinsic and intrinsic signaling. Here, we review the current knowledge on the cellular and molecular biology of ADM. Understanding the cellular and molecular mechanisms underlying ADM is critical for the development of new therapeutic strategies for pancreatitis and PDAC. Identifying the intermediate states and key molecules that regulate ADM initiation, maintenance and progression may help the development of novel preventive strategies for PDAC.
Collapse
Affiliation(s)
- Louis Marstrand-Daucé
- INSERM UMR1149, Inflammation Research Center (CRI), Université Paris Cité, 75018 Paris, France; (L.M.-D.); (D.L.); (A.C.); (P.N.); (A.C.)
| | - Diane Lorenzo
- INSERM UMR1149, Inflammation Research Center (CRI), Université Paris Cité, 75018 Paris, France; (L.M.-D.); (D.L.); (A.C.); (P.N.); (A.C.)
| | - Anaïs Chassac
- INSERM UMR1149, Inflammation Research Center (CRI), Université Paris Cité, 75018 Paris, France; (L.M.-D.); (D.L.); (A.C.); (P.N.); (A.C.)
- Department of Pathology, Bichat Hospital, Université Paris Cité, 75018 Paris, France
| | - Pascal Nicole
- INSERM UMR1149, Inflammation Research Center (CRI), Université Paris Cité, 75018 Paris, France; (L.M.-D.); (D.L.); (A.C.); (P.N.); (A.C.)
| | - Anne Couvelard
- INSERM UMR1149, Inflammation Research Center (CRI), Université Paris Cité, 75018 Paris, France; (L.M.-D.); (D.L.); (A.C.); (P.N.); (A.C.)
- Department of Pathology, Bichat Hospital, Université Paris Cité, 75018 Paris, France
| | - Cécile Haumaitre
- INSERM UMR1149, Inflammation Research Center (CRI), Université Paris Cité, 75018 Paris, France; (L.M.-D.); (D.L.); (A.C.); (P.N.); (A.C.)
| |
Collapse
|
27
|
Cancedda R, Mastrogiacomo M. Transit Amplifying Cells (TACs): a still not fully understood cell population. Front Bioeng Biotechnol 2023; 11:1189225. [PMID: 37229487 PMCID: PMC10203484 DOI: 10.3389/fbioe.2023.1189225] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
Maintenance of tissue homeostasis and tissue regeneration after an insult are essential functions of adult stem cells (SCs). In adult tissues, SCs proliferate at a very slow rate within "stem cell niches", but, during tissue development and regeneration, before giving rise to differentiated cells, they give rise to multipotent and highly proliferative cells, known as transit-amplifying cells (TACs). Although differences exist in diverse tissues, TACs are not only a transitory phase from SCs to post-mitotic cells, but they also actively control proliferation and number of their ancestor SCs and proliferation and differentiation of their progeny toward tissue specific functional cells. Autocrine signals and negative and positive feedback and feedforward paracrine signals play a major role in these controls. In the present review we will consider the generation and the role played by TACs during development and regeneration of lining epithelia characterized by a high turnover including epidermis and hair follicles, ocular epithelial surfaces, and intestinal mucosa. A comparison between these different tissues will be made. There are some genes and molecular pathways whose expression and activation are common to most TACs regardless their tissue of origin. These include, among others, Wnt, Notch, Hedgehog and BMP pathways. However, the response to these molecular signals can vary in TACs of different tissues. Secondly, we will consider cultured cells derived from tissues of mesodermal origin and widely adopted for cell therapy treatments. These include mesenchymal stem cells and dedifferentiated chondrocytes. The possible correlation between cell dedifferentiation and reversion to a transit amplifying cell stage will be discussed.
Collapse
Affiliation(s)
- Ranieri Cancedda
- Emeritus Professor, Università degli Studi di Genova, Genoa, Italy
| | - Maddalena Mastrogiacomo
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università Degli Studi di Genova, Genova, Italy
| |
Collapse
|
28
|
Melzer MK, Schirge S, Gout J, Arnold F, Srinivasan D, Burtscher I, Allgöwer C, Mulaw M, Zengerling F, Günes C, Lickert H, Christoffels VM, Liebau S, Wagner M, Seufferlein T, Bolenz C, Moon AM, Perkhofer L, Kleger A. TBX3 is dynamically expressed in pancreatic organogenesis and fine-tunes regeneration. BMC Biol 2023; 21:55. [PMID: 36941669 PMCID: PMC10029195 DOI: 10.1186/s12915-023-01553-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 02/27/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND The reactivation of genetic programs from early development is a common mechanism for injury-induced organ regeneration. T-box 3 (TBX3) is a member of the T-box family of transcription factors previously shown to regulate pluripotency and subsequent lineage commitment in a number of tissues, including limb and lung. TBX3 is also involved in lung and heart organogenesis. Here, we provide a comprehensive and thorough characterization of TBX3 and its role during pancreatic organogenesis and regeneration. RESULTS We interrogated the level and cell specificity of TBX3 in the developing and adult pancreas at mRNA and protein levels at multiple developmental stages in mouse and human pancreas. We employed conditional mutagenesis to determine its role in murine pancreatic development and in regeneration after the induction of acute pancreatitis. We found that Tbx3 is dynamically expressed in the pancreatic mesenchyme and epithelium. While Tbx3 is expressed in the developing pancreas, its absence is likely compensated by other factors after ablation from either the mesenchymal or epithelial compartments. In an adult model of acute pancreatitis, we found that a lack of Tbx3 resulted in increased proliferation and fibrosis as well as an enhanced inflammatory gene programs, indicating that Tbx3 has a role in tissue homeostasis and regeneration. CONCLUSIONS TBX3 demonstrates dynamic expression patterns in the pancreas. Although TBX3 is dispensable for proper pancreatic development, its absence leads to altered organ regeneration after induction of acute pancreatitis.
Collapse
Affiliation(s)
- Michael Karl Melzer
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Clinic of Urology, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Silvia Schirge
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Johann Gout
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Frank Arnold
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Dharini Srinivasan
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Chantal Allgöwer
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Medhanie Mulaw
- Unit for Single-cell Genomics, Ulm University, 89081, Ulm, Germany
| | | | - Cagatay Günes
- Clinic of Urology, Ulm University Hospital, Ulm, 89081, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
- Chair of b-Cell Biology, Technische Universität München, School of Medicine, Klinikum Rechts der Isar, 81675, München, Germany
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 15, 1105AZ, Amsterdam, The Netherlands
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074, Tübingen, Germany
| | - Martin Wagner
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
| | - Thomas Seufferlein
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
| | | | - Anne M Moon
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA
- Department of Human Genetics (adjunct), University of Utah, Salt Lake City, UT, USA
- The Mindich Child Health and Development Institute, Hess Center for Science and Medicine at Mount Sinai, New York, NY, USA
| | - Lukas Perkhofer
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Alexander Kleger
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany.
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany.
- Core Facility Organoids, Ulm University, 89081, Ulm, Germany.
| |
Collapse
|
29
|
Marcucio R, Miclau T, Bahney C. A Shifting Paradigm: Transformation of Cartilage to Bone during Bone Repair. J Dent Res 2023; 102:13-20. [PMID: 36303415 PMCID: PMC9791286 DOI: 10.1177/00220345221125401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
While formation and regeneration of the skeleton have been studied for a long period of time, significant scientific advances in this field continue to emerge based on an unmet clinical need to improve options to promote bone repair. In this review, we discuss the relationship between mechanisms of bone formation and bone regeneration. Data clearly show that regeneration is not simply a reinduction of the molecular and cellular programs that were used for development. Instead, the mechanical environment exerts a strong influence on the mode of repair, while during development, cell-intrinsic processes drive the mode of skeletal formation. A major advance in the field has shown that cell fate is flexible, rather than terminal, and that chondrocytes are able to differentiate into osteoblasts and other cell types during development and regeneration. This is discussed in a larger context of regeneration in vertebrates as well as the clinical implication that this shift in understanding presents.
Collapse
Affiliation(s)
- R.S. Marcucio
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
| | - T. Miclau
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
| | - C.S. Bahney
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
- Steadman Philippon Research Institute, Vail, CO, USA
| |
Collapse
|
30
|
Mainz L, Sarhan MAFE, Roth S, Sauer U, Kalogirou C, Eckstein M, Gerhard-Hartmann E, Seibert HD, Voelker HU, Geppert C, Rosenwald A, Eilers M, Schulze A, Diefenbacher M, Rosenfeldt MT. Acute systemic knockdown of Atg7 is lethal and causes pancreatic destruction in shRNA transgenic mice. Autophagy 2022; 18:2880-2893. [PMID: 35343375 PMCID: PMC9673934 DOI: 10.1080/15548627.2022.2052588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The notion that macroautophagy/autophagy is a potentially attractive therapeutic target for a variety of diseases, including cancer, largely stems from pre-clinical mouse studies. Most of these examine the effects of irreversible and organ confined autophagy deletion using site specific Cre-loxP recombination of the essential autophagy regulating genes Atg7 or Atg5. Model systems with the ability to impair autophagy systemically and reversibly at all disease stages would allow a more realistic approach to evaluate the consequences of authophagy inhibition as a therapeutic concept and its potential side effects. Here, we present shRNA transgenic mice that via doxycycline (DOX) regulable expression of a highly efficient miR30-E-based shRNA enabled knockdown of Atg7 simultaneously in the majority of organs, with the brain and spleen being noteable exceptions. Induced animals deteriorated rapidly and experienced profound destruction of the exocrine pancreas, severe hypoglycemia and depletion of hepatic glycogen storages. Cessation of DOX application restored apparent health, glucose homeostasis and pancreatic integrity. In a similar Atg5 knockdown model we neither observed loss of pancreatic integrity nor diminished survival after DOX treatment, but identified histological changes consistent with steatohepatitis and hepatic fibrosis in the recovery period after termination of DOX. Regulable Atg7-shRNA mice are valuable tools that will enable further studies on the role of autophagy impairment at various disease stages and thereby help to evaluate the consequences of acute autophagy inhibition as a therapeutic concept.Abbreviations: ACTB: actin, beta; AMY: amylase complex; ATG4B: autophagy related 4B, cysteine peptidase; ATG5: autophagy related 5; ATG7: autophagy related 7; Cag: CMV early enhancer/chicken ACTB promoter; Col1a1: collagen, type I, alpha 1; Cre: cre recombinase; DOX: doxycycline; GCG: glucagon; GFP: green fluorescent protein; INS: insulin; LC3: microtubule-associated protein 1 light chain 3; miR30-E: optimized microRNA backbone; NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; PNLIP: pancreatic lipase; rtTA: reverse tetracycline transactivator protein; SQSTM1/p62: sequestome 1; TRE: tetracycline responsive element.
Collapse
Affiliation(s)
- Laura Mainz
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Mohamed A. F. E. Sarhan
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Sabine Roth
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Ursula Sauer
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Charis Kalogirou
- Department of Urology and Pediatric Urology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elena Gerhard-Hartmann
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Helen-Desiree Seibert
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Hans-Ulrich Voelker
- Department of Pathology, Leopoldina Medizinisches Versorgungszentrum, Schweinfurt, Germany
| | - Carol Geppert
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Martin Eilers
- Biocenter, Department of Biochemistry and Molecular Biology, Julius-Maximilians-University of Würzburg, Germany
| | - Almut Schulze
- Division of Metabolism and Microenvironment, Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), Germany
| | - Markus Diefenbacher
- Biocenter, Department of Biochemistry and Molecular Biology, Julius-Maximilians-University of Würzburg, Germany
| | - Mathias T. Rosenfeldt
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany,CONTACT Mathias T. Rosenfeldt Institute of Pathology – University of Würzburg, Josef-Schneider-Str. 2,97080Würzburg, Germany
| |
Collapse
|
31
|
Hashimoto A, Handa H, Hata S, Hashimoto S. Orchestration of mesenchymal plasticity and immune evasiveness via rewiring of the metabolic program in pancreatic ductal adenocarcinoma. Front Oncol 2022; 12:1005566. [PMID: 36408139 PMCID: PMC9669439 DOI: 10.3389/fonc.2022.1005566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most fatal cancer in humans, due to its difficulty of early detection and its high metastatic ability. The occurrence of epithelial to mesenchymal transition in preinvasive pancreatic lesions has been implicated in the early dissemination, drug resistance, and cancer stemness of PDAC. PDAC cells also have a reprogrammed metabolism, regulated by driver mutation-mediated pathways, a desmoplastic tumor microenvironment (TME), and interactions with stromal cells, including pancreatic stellate cells, fibroblasts, endothelial cells, and immune cells. Such metabolic reprogramming and its functional metabolites lead to enhanced mesenchymal plasticity, and creates an acidic and immunosuppressive TME, resulting in the augmentation of protumor immunity via cancer-associated inflammation. In this review, we summarize our recent understanding of how PDAC cells acquire and augment mesenchymal features via metabolic and immunological changes during tumor progression, and how mesenchymal malignancies induce metabolic network rewiring and facilitate an immune evasive TME. In addition, we also present our recent findings on the interesting relevance of the small G protein ADP-ribosylation factor 6-based signaling pathway driven by KRAS/TP53 mutations, inflammatory amplification signals mediated by the proinflammatory cytokine interleukin 6 and RNA-binding protein ARID5A on PDAC metabolic reprogramming and immune evasion, and finally discuss potential therapeutic strategies for the quasi-mesenchymal subtype of PDAC.
Collapse
Affiliation(s)
- Ari Hashimoto
- Department of Molecular Biology, Hokkaido University Faculty of Medicine, Sapporo, Japan
- *Correspondence: Ari Hashimoto, ; Shigeru Hashimoto,
| | - Haruka Handa
- Department of Molecular Biology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Soichiro Hata
- Department of Molecular Biology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
- *Correspondence: Ari Hashimoto, ; Shigeru Hashimoto,
| |
Collapse
|
32
|
Xiang H, Yu H, Zhou Q, Wu Y, Ren J, Zhao Z, Tao X, Dong D. Macrophages: A rising star in immunotherapy for chronic pancreatitis. Pharmacol Res 2022; 185:106508. [DOI: 10.1016/j.phrs.2022.106508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 10/10/2022] [Indexed: 11/29/2022]
|
33
|
Targeting PI3K/AKT/mTOR Signaling Pathway in Pancreatic Cancer: From Molecular to Clinical Aspects. Int J Mol Sci 2022; 23:ijms231710132. [PMID: 36077529 PMCID: PMC9456549 DOI: 10.3390/ijms231710132] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023] Open
Abstract
Although pancreatic cancer (PC) was considered in the past an orphan cancer type due to its low incidence, it may become in the future one of the leading causes of cancer death. Pancreatic ductal adenocarcinoma (PDAC) is the most frequent type of PC, being a highly aggressive malignancy and having a 5-year survival rate of less than 10%. Non-modifiable (family history, age, genetic susceptibility) and modifiable (smoking, alcohol, acute and chronic pancreatitis, diabetes mellitus, intestinal microbiota) risk factors are involved in PC pathogenesis. Chronic inflammation induced by various factors plays crucial roles in PC development from initiation to metastasis. In multiple malignant conditions such as PC, cytokines, chemokines, and growth factors activate the class I phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) (PI3K/AKT/mTOR) signaling pathway, which plays key roles in cell growth, survival, proliferation, metabolism, and motility. Currently, mTOR, AKT, and PI3K inhibitors are used in clinical studies. Moreover, PI3K/mTOR dual inhibitors are being tested in vitro and in vivo with promising results for PC patients. The main aim of this review is to present PC incidence, risk factors, tumor microenvironment development, and PI3K/AKT/mTOR dysregulation and inhibitors used in clinical, in vivo, and in vitro studies.
Collapse
|
34
|
Takeo M, Nishio A, Masuda M, Aoi K, Okazaki T, Fukui T, Uchida K, Naganuma M, Okazaki K. Repeated Stimulation of Toll-Like Receptor 2 and Dectin-1 Induces Chronic Pancreatitis in Mice Through the Participation of Acquired Immunity. Dig Dis Sci 2022; 67:3783-3796. [PMID: 34424458 DOI: 10.1007/s10620-021-07186-w] [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: 12/10/2020] [Accepted: 07/20/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Stimulation of Toll-like receptor 3 (TLR3) induces autoimmune-mediated pancreatitis in susceptible mice, whereas stimulation of TLR4 causes nonautoimmune-mediated pancreatitis. However, the effects of TLR2 stimulation on the pancreas are unknown. AIMS We investigated the role of TLR2 stimulation on pancreatic damage by repeatedly stimulating mice with TLR2 ligands. METHODS Wild-type (WT) and interleukin 10-deficient (IL-10-knockout (KO)) mice were administered zymosan and lipoteichoic acid (LTA) intraperitoneally at various doses twice weekly for 4 weeks. Syngeneic T-cell-deficient mice, B-cell-deficient mice, recombination activating gene 2-deficient (RAG2-KO) mice and RAG2-KO mice that had been reconstituted with CD4+ or CD8+ T cells isolated from WT mice were treated with zymosan similarly. Mice were killed, the severity of pancreatitis was graded histologically, and serum cytokine levels were measured. RESULTS Repeated administration of zymosan induced pancreatitis dose dependently in both WT and IL-10-KO mice. Administration of LTA induced pancreatitis only in IL-10-KO mice. Adoptive transfer of splenocytes obtained from IL-10-KO mice with pancreatitis did not cause pancreatitis in recipient RAG2-KO mice. Pancreatitis was scarcely observed in RAG2-KO mice and was attenuated in T-cell-deficient and B-cell-deficient mice compared with WT mice. A single administration of zymosan significantly increased the serum level of monocyte chemoattractant protein 1 (MCP-1) in WT mice. CONCLUSIONS Repeated stimulation of TLR2 and dectin-1 induced nonautoimmune-mediated pancreatitis in mice. Participation of acquired immunity seems to play an important role in the pathogenesis of pancreatitis in association with the increase in serum MCP-1 level.
Collapse
Affiliation(s)
- Masahiro Takeo
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, Japan
| | - Akiyoshi Nishio
- Department of Gastroenterology and Hepatology, Kansai Medical University Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka, 570-8507, Japan.
| | - Masataka Masuda
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, Japan
| | - Kazunori Aoi
- Department of Gastroenterology and Hepatology, Kansai Medical University Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka, 570-8507, Japan
| | - Takashi Okazaki
- Department of Gastroenterology and Hepatology, Kansai Medical University Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka, 570-8507, Japan
| | - Toshiro Fukui
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, Japan
| | - Kazushige Uchida
- Department of Gastroenterology and Hepatology, Kochi Medical School, 185-1 Kohasu Okocho, Nankoku, Kochi, Japan
| | - Makoto Naganuma
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, Japan
| | - Kazuichi Okazaki
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, Japan
| |
Collapse
|
35
|
Yang H, Messina-Pacheco J, Corredor ALG, Gregorieff A, Liu JL, Nehme A, Najafabadi HS, Riazalhosseini Y, Gao B, Gao ZH. An integrated model of acinar to ductal metaplasia-related N7-methyladenosine regulators predicts prognosis and immunotherapy in pancreatic carcinoma based on digital spatial profiling. Front Immunol 2022; 13:961457. [PMID: 35979350 PMCID: PMC9377277 DOI: 10.3389/fimmu.2022.961457] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 06/24/2022] [Indexed: 12/14/2022] Open
Abstract
Acinar-to-ductal metaplasia (ADM) is a recently recognized, yet less well-studied, precursor lesion of pancreatic ductal adenocarcinoma (PDAC) developed in the setting of chronic pancreatitis. Through digital spatial mRNA profiling, we compared ADM and adjacent PDAC tissues from patient samples to unveil the bridging genes during the malignant transformation of pancreatitis. By comparing the bridging genes with the 7-methylguanosine (m7G)-seq dataset, we screened 19 m7G methylation genes for a subsequent large sample analysis. We constructed the “m7G score” model based on the RNA-seq data for pancreatic cancer in The Cancer Genome Atlas (TCGA) database and The Gene Expression Omnibus (GEO) database. Tumors with a high m7G score were characterized by increased immune cell infiltration, increased genomic instability, higher response rate to combined immune checkpoint inhibitors (ICIs), and overall poor survival. These findings indicate that the m7G score is associated with tumor invasiveness, immune cell infiltration, ICI treatment response, and overall patients’ survival. We also identified FN1 and ITGB1 as core genes in the m7Gscore model, which affect immune cell infiltration and genomic instability not only in pancreatic cancer but also in pan-cancer. FN1 and ITGB1 can inhibit immune T cell activition by upregulation of macrophages and neutrophils, thereby leading to immune escape of pancreatic cancer cells and reducing the response rate of ICI treatment.
Collapse
Affiliation(s)
- Hao Yang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Julia Messina-Pacheco
- Department of Pathology, McGill University and the Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - Andrea Liliam Gomez Corredor
- Department of Pathology, McGill University and the Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - Alex Gregorieff
- Department of Pathology, McGill University and the Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - Jun-li Liu
- MeDic Program, The Research Institute of McGill University Health Centre, & Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Ali Nehme
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
| | - Hamed S. Najafabadi
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
| | - Yasser Riazalhosseini
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
| | - Bo Gao
- Department of General Surgery, Peking University People’s Hospital, Beijing, China
- *Correspondence: Zu-hua Gao, ; Bo Gao,
| | - Zu-hua Gao
- Department of Pathology and Laboratory Medicine, British Columbia (BC) Cancer Research Center, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Zu-hua Gao, ; Bo Gao,
| |
Collapse
|
36
|
Pérez S, Rius-Pérez S. Macrophage Polarization and Reprogramming in Acute Inflammation: A Redox Perspective. Antioxidants (Basel) 2022; 11:antiox11071394. [PMID: 35883885 PMCID: PMC9311967 DOI: 10.3390/antiox11071394] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 12/12/2022] Open
Abstract
Macrophage polarization refers to the process by which macrophages can produce two distinct functional phenotypes: M1 or M2. The balance between both strongly affects the progression of inflammatory disorders. Here, we review how redox signals regulate macrophage polarization and reprogramming during acute inflammation. In M1, macrophages augment NADPH oxidase isoform 2 (NOX2), inducible nitric oxide synthase (iNOS), synaptotagmin-binding cytoplasmic RNA interacting protein (SYNCRIP), and tumor necrosis factor receptor-associated factor 6 increase oxygen and nitrogen reactive species, which triggers inflammatory response, phagocytosis, and cytotoxicity. In M2, macrophages down-regulate NOX2, iNOS, SYNCRIP, and/or up-regulate arginase and superoxide dismutase type 1, counteract oxidative and nitrosative stress, and favor anti-inflammatory and tissue repair responses. M1 and M2 macrophages exhibit different metabolic profiles, which are tightly regulated by redox mechanisms. Oxidative and nitrosative stress sustain the M1 phenotype by activating glycolysis and lipid biosynthesis, but by inhibiting tricarboxylic acid cycle and oxidative phosphorylation. This metabolic profile is reversed in M2 macrophages because of changes in the redox state. Therefore, new therapies based on redox mechanisms have emerged to treat acute inflammation with positive results, which highlights the relevance of redox signaling as a master regulator of macrophage reprogramming.
Collapse
|
37
|
Shibata O, Kamimura K, Tanaka Y, Ogawa K, Owaki T, Oda C, Morita S, Kimura A, Abe H, Ikarashi S, Hayashi K, Yokoo T, Terai S. Establishment of a pancreatic cancer animal model using the pancreas-targeted hydrodynamic gene delivery method. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:342-352. [PMID: 35474735 PMCID: PMC9018811 DOI: 10.1016/j.omtn.2022.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/27/2022] [Indexed: 11/29/2022]
Abstract
This research developed an easy-to-use, reproducible pancreatic cancer animal model utilizing pancreas-targeted hydrodynamic gene delivery to deliver human pancreatic cancer-related genes to the pancreas of wild-type rats. KRASG12D-induced pancreatic intraepithelial neoplasia lesions showed malignant transformation in the main pancreatic duct at 4 weeks and developed acinar-to-ductal metaplasia, which led to pancreatic ductal adenocarcinoma within 5 weeks, and the gene combination of KRASG12D and YAP enhanced these effects. The repeat hydrodynamic gene delivery of KRASG12D + YAP combination at 4 weeks showed acinar-to-ductal metaplasia in all rats and pancreatic ductal adenocarcinoma in 80% of rats 1 week later. Metastatic tumors in the liver, lymph nodes, and subcutaneous lesions and nervous invasion were confirmed. KRASG12D and YAP combined transfer contributes to the E- to N-cadherin switch in pancreatic ductal adenocarcinoma cells and to tumor metastases. This pancreatic cancer model will speed up pancreatic cancer research for novel treatments and biomarkers for early diagnosis.
Collapse
Affiliation(s)
- Osamu Shibata
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
- Department of General Medicine, Niigata University School of Medicine, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
- Corresponding author. Kenya Kamimura, Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan.
| | - Yuto Tanaka
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Kohei Ogawa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Takashi Owaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Chiyumi Oda
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Shinichi Morita
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Atsushi Kimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Satoshi Ikarashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Kazunao Hayashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 1–757 Asahimachi–dori Chuo–ku, Niigata 951-8510, Japan
| |
Collapse
|
38
|
Abstract
The pancreatic β-cells are essential for regulating glucose homeostasis through the coordinated release of the insulin hormone. Dysfunction of the highly specialized β-cells results in diabetes mellitus, a growing global health epidemic. In this review, we describe the development and function of β-cells the emerging concept of heterogeneity within insulin-producing cells, and the potential of other cell types to assume β-cell functionality via transdifferentiation. We also discuss emerging routes to design cells with minimal β-cell properties and human stem cell differentiation efforts that carry the promise to restore normoglycemia in patients suffering from diabetes.
Collapse
Affiliation(s)
- Natanya Kerper
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | - Sudipta Ashe
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
39
|
Li X, He J, Xie K. Molecular signaling in pancreatic ductal metaplasia: emerging biomarkers for detection and intervention of early pancreatic cancer. Cell Oncol (Dordr) 2022; 45:201-225. [PMID: 35290607 DOI: 10.1007/s13402-022-00664-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 11/27/2022] Open
Abstract
Pancreatic ductal metaplasia (PDM) is the transformation of potentially various types of cells in the pancreas into ductal or ductal-like cells, which eventually replace the existing differentiated somatic cell type(s). PDM is usually triggered by and manifests its ability to adapt to environmental stimuli and genetic insults. The development of PDM to atypical hyperplasia or dysplasia is an important risk factor for pancreatic intraepithelial neoplasia (PanIN) and pancreatic ductal adenocarcinoma (PDA). Recent studies using genetically engineered mouse models, cell lineage tracing, single-cell sequencing and others have unraveled novel cellular and molecular insights in PDM formation and evolution. Those novel findings help better understand the cellular origins and functional significance of PDM and its regulation at cellular and molecular levels. Given that PDM represents the earliest pathological changes in PDA initiation and development, translational studies are beginning to define PDM-associated cell and molecular biomarkers that can be used to screen and detect early PDA and to enable its effective intervention, thereby truly and significantly reducing the dreadful mortality rate of PDA. This review will describe recent advances in the understanding of PDM biology with a focus on its underlying cellular and molecular mechanisms, and in biomarker discovery with clinical implications for the management of pancreatic regeneration and tumorigenesis.
Collapse
Affiliation(s)
- Xiaojia Li
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, 510006, China
- Department of Pathology, The South China University of Technology School of Medicine, Guangzhou, China
| | - Jie He
- Institute of Digestive Diseases Research, The South China University of Technology School of Medicine, Guangzhou, China
| | - Keping Xie
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, 510006, China.
- Department of Pathology, The South China University of Technology School of Medicine, Guangzhou, China.
| |
Collapse
|
40
|
Fukase M, Sakata N, Kushida Y, Wakao S, Unno M, Dezawa M. Intravenous injection of human multilineage-differentiating stress-enduring cells alleviates mouse severe acute pancreatitis without immunosuppressants. Surg Today 2022; 52:603-615. [PMID: 34687364 DOI: 10.1007/s00595-021-02382-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/16/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION We examined the effect of intravenously injected human multilineage-differentiating stress-enduring (Muse) cells, non-tumorigenic endogenous reparative stem cells already used in clinical trials, on a severe acute pancreatitis (SAP) mouse model without immunosuppressants. METHODS Human Muse cells (1.0 × 105 cells) collected from mesenchymal stem cells (MSCs) as SSEA-3(+) were injected into a C57BL/6 mouse model via the jugular vein 6 h after SAP-induction with taurocholate. The control group received saline or the same number of SSEA-3(-)-non-Muse MSCs. RESULTS Edematous parameters, F4/80(+) macrophage infiltration and terminal deoxynucleotidyl transferase dUTP nick-end labeling positivity was the lowest and the number of proliferating endogenous pancreatic progenitors (CK18(+)/Ki67(+) cells) the highest in the Muse group among the three groups, with statistical significance, at 72 h. An enzyme-linked immunosorbent assay and quantitative polymerase chain reaction demonstrated that in vitro production of VEGF, HGF, IGF-1, and MMP-2, which are relevant to tissue protection, anti-inflammation, and anti-fibrosis, were higher in Muse cells than in non-Muse MSCs, particularly when cells were cultured in SAP mouse serum. Consistently, the pancreas of animals in the Muse group contained higher amounts of those factors according to Western blotting at 18 h than that in the non-Muse MSCs and control groups. CONCLUSIONS Intravenous injection of human Muse cells was suggested to be effective for attenuating edema, inflammation and apoptosis in the acute phase of SAP.
Collapse
Affiliation(s)
- Masahiko Fukase
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, Miyagi, 980-8575, Japan.
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryomachi, Aobaku, Sendai, Miyagi, 980-8574, Japan.
| | - Naoaki Sakata
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryomachi, Aobaku, Sendai, Miyagi, 980-8574, Japan
| | - Yoshihiro Kushida
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, Miyagi, 980-8575, Japan
| | - Shohei Wakao
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, Miyagi, 980-8575, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryomachi, Aobaku, Sendai, Miyagi, 980-8574, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
41
|
Hagen CM, Roth E, Graf TR, Verrey F, Graf R, Gupta A, Pellegrini G, Poncet N, Camargo SMR. Loss of LAT1 sex-dependently delays recovery after caerulein-induced acute pancreatitis. World J Gastroenterol 2022; 28:1024-1054. [PMID: 35431492 PMCID: PMC8968515 DOI: 10.3748/wjg.v28.i10.1024] [Citation(s) in RCA: 2] [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] [Received: 08/22/2021] [Revised: 10/08/2021] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The expression of amino acid transporters is known to vary during acute pancreatitis (AP) except for LAT1 (slc7a5), the expression of which remains stable. LAT1 supports cell growth by importing leucine and thereby stimulates mammalian target of rapamycin (mTOR) activity, a phenomenon often observed in cancer cells. The mechanisms by which LAT1 influences physiological and pathophysiological processes and affects disease progression in the pancreas are not yet known.
AIM To evaluate the role of LAT1 in the development of and recovery from AP.
METHODS AP was induced with caerulein (cae) injections in female and male mice expressing LAT1 or after its knockout (LAT1 Cre/LoxP). The development of the initial AP injury and its recovery were followed for seven days after cae injections by daily measuring body weight, assessing microscopical tissue architecture, mRNA and protein expression, protein synthesis, and enzyme activity levels, as well as by testing the recruitment of immune cells by FACS and ELISA.
RESULTS The initial injury, evaluated by measurements of plasma amylase, lipase, and trypsin activity, as well as the gene expression of dedifferentiation markers, did not differ between the groups. However, early metabolic adaptations that support regeneration at later stages were blunted in LAT1 knockout mice. Especially in females, we observed less mTOR reactivation and dysfunctional autophagy. The later regeneration phase was clearly delayed in female LAT1 knockout mice, which did not regain normal expression of the pancreas-specific differentiation markers recombining binding protein suppressor of hairless-like protein (rbpjl) and basic helix-loop-helix family member A15 (mist1). Amylase mRNA and protein levels remained lower, and, strikingly, female LAT1 knockout mice presented signs of fibrosis lasting until day seven. In contrast, pancreas morphology had returned to normal in wild-type littermates.
CONCLUSION LAT1 supports the regeneration of acinar cells after AP. Female mice lacking LAT1 exhibited more pronounced alterations than male mice, indicating a sexual dimorphism of amino acid metabolism.
Collapse
Affiliation(s)
- Cristina M Hagen
- Institute of Physiology, University of Zurich, Zurich 8057, ZH, Switzerland
| | - Eva Roth
- Institute of Physiology, University of Zurich, Zurich 8057, ZH, Switzerland
| | - Theresia Reding Graf
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, Zurich University Hospital, Zurich 8091, ZH, Switzerland
| | - François Verrey
- Institute of Physiology, University of Zurich, Zurich 8057, ZH, Switzerland
| | - Rolf Graf
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, Zurich University Hospital, Zurich 8091, ZH, Switzerland
| | - Anurag Gupta
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, Zurich University Hospital, Zurich 8091, ZH, Switzerland
| | - Giovanni Pellegrini
- Institute of Veterinary Pathology, University of Zurich, Zurich 8057, ZH, Switzerland
| | - Nadège Poncet
- Institute of Physiology, University of Zurich, Zurich 8057, ZH, Switzerland
| | | |
Collapse
|
42
|
Li S, Xie K. Ductal metaplasia in pancreas. Biochim Biophys Acta Rev Cancer 2022; 1877:188698. [DOI: 10.1016/j.bbcan.2022.188698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 02/07/2023]
|
43
|
Ma Z, Lytle NK, Chen B, Jyotsana N, Novak SW, Cho CJ, Caplan L, Ben-Levy O, Neininger AC, Burnette DT, Trinh VQ, Tan MCB, Patterson EA, Arrojo E Drigo R, Giraddi RR, Ramos C, Means AL, Matsumoto I, Manor U, Mills JC, Goldenring JR, Lau KS, Wahl GM, DelGiorno KE. Single-Cell Transcriptomics Reveals a Conserved Metaplasia Program in Pancreatic Injury. Gastroenterology 2022; 162:604-620.e20. [PMID: 34695382 PMCID: PMC8792222 DOI: 10.1053/j.gastro.2021.10.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/15/2021] [Accepted: 10/09/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Acinar to ductal metaplasia (ADM) occurs in the pancreas in response to tissue injury and is a potential precursor for adenocarcinoma. The goal of these studies was to define the populations arising from ADM, the associated transcriptional changes, and markers of disease progression. METHODS Acinar cells were lineage-traced with enhanced yellow fluorescent protein (EYFP) to follow their fate post-injury. Transcripts of more than 13,000 EYFP+ cells were determined using single-cell RNA sequencing (scRNA-seq). Developmental trajectories were generated. Data were compared with gastric metaplasia, KrasG12D-induced neoplasia, and human pancreatitis. Results were confirmed by immunostaining and electron microscopy. KrasG12D was expressed in injury-induced ADM using several inducible Cre drivers. Surgical specimens of chronic pancreatitis from 15 patients were evaluated by immunostaining. RESULTS scRNA-seq of ADM revealed emergence of a mucin/ductal population resembling gastric pyloric metaplasia. Lineage trajectories suggest that some pyloric metaplasia cells can generate tuft and enteroendocrine cells (EECs). Comparison with KrasG12D-induced ADM identifies populations associated with disease progression. Activation of KrasG12D expression in HNF1B+ or POU2F3+ ADM populations leads to neoplastic transformation and formation of MUC5AC+ gastric-pit-like cells. Human pancreatitis samples also harbor pyloric metaplasia with a similar transcriptional phenotype. CONCLUSIONS Under conditions of chronic injury, acinar cells undergo a pyloric-type metaplasia to mucinous progenitor-like populations, which seed disparate tuft cell and EEC lineages. ADM-derived EEC subtypes are diverse. KrasG12D expression is sufficient to drive neoplasia when targeted to injury-induced ADM populations and offers an alternative origin for tumorigenesis. This program is conserved in human pancreatitis, providing insight into early events in pancreas diseases.
Collapse
Affiliation(s)
- Zhibo Ma
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California
| | - Nikki K Lytle
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California
| | - Bob Chen
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Nidhi Jyotsana
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Sammy Weiser Novak
- Waitt Advanced Biophotonics Center, Salk Insitute for Biological Studies, La Jolla, California
| | - Charles J Cho
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas
| | - Leah Caplan
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Olivia Ben-Levy
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Abigail C Neininger
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Dylan T Burnette
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Vincent Q Trinh
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Marcus C B Tan
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Emilee A Patterson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Rafael Arrojo E Drigo
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Rajshekhar R Giraddi
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California
| | - Cynthia Ramos
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California
| | - Anna L Means
- Vanderbilt Ingram Cancer Center, Nashville, Tennessee; Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Uri Manor
- Waitt Advanced Biophotonics Center, Salk Insitute for Biological Studies, La Jolla, California
| | - Jason C Mills
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas
| | - James R Goldenring
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Vanderbilt Ingram Cancer Center, Nashville, Tennessee; Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee; Nashville VA Medical Center, Nashville, Tennessee
| | - Ken S Lau
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Vanderbilt Ingram Cancer Center, Nashville, Tennessee; Vanderbilt Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Geoffrey M Wahl
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California
| | - Kathleen E DelGiorno
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Vanderbilt Ingram Cancer Center, Nashville, Tennessee; Vanderbilt Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
44
|
Falcomatà C, Saur D. Self-renewal equality in pancreas homeostasis, regeneration, and cancer. Cell Rep 2021; 37:110135. [PMID: 34910913 DOI: 10.1016/j.celrep.2021.110135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Two studies by Lodestijn et al. in Cell Stem Cell and Cell Reports reveal a lack of stem cell hierarchies in acinar cell-derived tissue renewal and host instructed clonogenic growth of pancreatic cancer, thereby elucidating determinants of pancreas regeneration and cancer.
Collapse
Affiliation(s)
- Chiara Falcomatà
- Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, Klinikum rechts der Isar, School of Medicine, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany.
| | - Dieter Saur
- Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, Klinikum rechts der Isar, School of Medicine, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany; Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany.
| |
Collapse
|
45
|
Malinova A, Veghini L, Real FX, Corbo V. Cell Lineage Infidelity in PDAC Progression and Therapy Resistance. Front Cell Dev Biol 2021; 9:795251. [PMID: 34926472 PMCID: PMC8675127 DOI: 10.3389/fcell.2021.795251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/15/2021] [Indexed: 12/23/2022] Open
Abstract
Infidelity to cell fate occurs when differentiated cells lose their original identity and either revert to a more multipotent state or transdifferentiate into a different cell type, either within the same embryonic lineage or in an entirely different one. Whilst in certain circumstances, such as in wound repair, this process is beneficial, it can be hijacked by cancer cells to drive disease initiation and progression. Cell phenotype switching has been shown to also serve as a mechanism of drug resistance in some epithelial cancers. In pancreatic ductal adenocarcinoma (PDAC), the role of lineage infidelity and phenotype switching is still unclear. Two consensus molecular subtypes of PDAC have been proposed that mainly reflect the existence of cell lineages with different degrees of fidelity to pancreatic endodermal precursors. Indeed, the classical subtype of PDAC is characterised by the expression of endodermal lineage specifying transcription factors, while the more aggressive basal-like/squamous subtype is defined by epigenetic downregulation of endodermal genes and alterations in chromatin modifiers. Here, we summarise the current knowledge of mechanisms (genetic and epigenetic) of cell fate switching in PDAC and discuss how pancreatic organoids might help increase our understanding of both cell-intrinsic and cell-extrinsic factors governing lineage infidelity during the distinct phases of PDAC evolution.
Collapse
Affiliation(s)
- Antonia Malinova
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Lisa Veghini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Francisco X. Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre, Madrid, Spain
- CIBERONC, Madrid, Spain
- Department de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- ARC-Net Research Centre, University of Verona, Verona, Italy
| |
Collapse
|
46
|
Dai J, He Y, Jiang M, Niu M, Li B, Wu Z, Bao J, Wen L, Wang X, Hu G. Reg4 regulates pancreatic regeneration following pancreatitis via modulating the Notch signaling. J Cell Physiol 2021; 236:7565-7577. [PMID: 33899235 DOI: 10.1002/jcp.30397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/25/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
Pancreatic regeneration after acute pancreatitis is critical in the normal restoration of pancreatic exocrine function, the inhibition of which can cause severe complications including pancreatic exocrine insufficiency. However, the regulators of pancreatic regeneration and the underlying mechanisms remain uncovered. Here, using the inducible Tet-on system, we found that regenerating family member 4 (Reg4) knockdown significantly impaired pancreatic regeneration after pancreatitis. Both acinar-to-ductal metaplasia and the resolution of pancreatitis during regeneration were affected by Reg4 knockdown. Further investigations confirmed that Reg4 exerted its function through regulating Notch activation both in vitro and in vivo. Our study revealed Reg4 as a new regulator and potential therapeutic target for pancreatic regeneration.
Collapse
Affiliation(s)
- Juanjuan Dai
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Intensive Care Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yan He
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingjie Jiang
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Head and Neck, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Mengya Niu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zengkai Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingpiao Bao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingpeng Wang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoyong Hu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
47
|
Sandhu N, Rana S, Meena K. Nuclear receptor subfamily 5 group A member 2 (NR5A2): role in health and diseases. Mol Biol Rep 2021; 48:8155-8170. [PMID: 34643922 DOI: 10.1007/s11033-021-06784-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/22/2021] [Indexed: 10/20/2022]
Abstract
Nuclear receptors are the regulatory molecules that mediate cellular signals as they interact with specific DNA sequences. NR5A2 is a member of NR5A subfamily having four members (Nr5a1-Nr5a4). NR5A2 shows involvement in diverse biological processes like reverse cholesterol transport, embryonic stem cell pluripotency, steroidogenesis, development and differentiation of embryo, and adult homeostasis. NR5A2 haploinsufficiency has been seen associated with chronic pancreatitis, pancreatic and gastrointestinal cancer. There is a close relationship between the progression of pancreatic cancer from chronic pancreatitis, NR5A2 serving a common link. NR5A2 activity is regulated by intracellular phospholipids, transcriptional coregulators and post-translational modifications. The specific ligand of NR5A2 is unknown hence called an orphan receptor, but specific phospholipids such as dilauroyl phosphatidylcholine and diundecanoyl phosphatidylcholine act as a ligand and they are established drug targets in various diseases. This review will focus on the NR5A2 structure, regulation of its activity, and role in biological processes and diseases. In future, need more emphasis on discovering small molecule agonists and antagonist, which act as a drug target for therapeutic applications.
Collapse
Affiliation(s)
- Nikita Sandhu
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, Uttarakhand, India
| | - Satyavati Rana
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, Uttarakhand, India
| | - Kiran Meena
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, Uttarakhand, India.
| |
Collapse
|
48
|
Neuhöfer P, Roake CM, Kim SJ, Lu RJ, West RB, Charville GW, Artandi SE. Acinar cell clonal expansion in pancreas homeostasis and carcinogenesis. Nature 2021; 597:715-719. [PMID: 34526722 DOI: 10.1038/s41586-021-03916-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 08/16/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer deaths worldwide1. Studies in human tissues and in mouse models have suggested that for many cancers, stem cells sustain early mutations driving tumour development2,3. For the pancreas, however, mechanisms underlying cellular renewal and initiation of PDAC remain unresolved. Here, using lineage tracing from the endogenous telomerase reverse transcriptase (Tert) locus, we identify a rare TERT-positive subpopulation of pancreatic acinar cells dispersed throughout the exocrine compartment. During homeostasis, these TERThigh acinar cells renew the pancreas by forming expanding clones of acinar cells, whereas randomly marked acinar cells do not form these clones. Specific expression of mutant Kras in TERThigh acinar cells accelerates acinar clone formation and causes transdifferentiation to ductal pre-invasive pancreatic intraepithelial neoplasms by upregulating Ras-MAPK signalling and activating the downstream kinase ERK (phospho-ERK). In resected human pancreatic neoplasms, we find that foci of phospho-ERK-positive acinar cells are common and frequently contain activating KRAS mutations, suggesting that these acinar regions represent an early cancer precursor lesion. These data support a model in which rare TERThigh acinar cells may sustain KRAS mutations, driving acinar cell expansion and creating a field of aberrant cells initiating pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Patrick Neuhöfer
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Caitlin M Roake
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Stewart J Kim
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Ryan J Lu
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert B West
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gregory W Charville
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven E Artandi
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
49
|
Wan J, Wang J, Wagner LE, Wang OH, Gui F, Chen J, Zhu X, Haddock AN, Edenfield BH, Haight B, Mukhopadhyay D, Wang Y, Yule DI, Bi Y, Ji B. Pancreas-specific CHRM3 activation causes pancreatitis in mice. JCI Insight 2021; 6:132585. [PMID: 34314386 PMCID: PMC8492327 DOI: 10.1172/jci.insight.132585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 07/22/2021] [Indexed: 12/26/2022] Open
Abstract
Hyperstimulation of the cholecystokinin 1 receptor (CCK1R), a G protein-coupled receptor (GPCR), in pancreatic acinar cells is commonly used to induce pancreatitis in rodents. Human pancreatic acinar cells lack CCK1R but express cholinergic receptor muscarinic 3 (M3R), another GPCR. To test whether M3R activation is involved in pancreatitis, a mutant M3R was conditionally expressed in pancreatic acinar cells in mice. This mutant receptor loses responsiveness to its native ligand, acetylcholine, but can be activated by an inert small molecule, clozapine-N-oxide (CNO). Intracellular calcium and amylase were elicited by CNO in pancreatic acinar cells isolated from mutant M3R mice but not WT mice. Similarly, acute pancreatitis (AP) could be induced by a single injection of CNO in the transgenic mice but not WT mice. Compared with the cerulein-induced AP, CNO caused more widespread acinar cell death and inflammation. Furthermore, chronic pancreatitis developed at 4 weeks after 3 episodes of CNO-induced AP. In contrast, in mice with 3 recurrent episodes of cerulein-included AP, pancreas histology was restored in 4 weeks. Furthermore, the M3R antagonist ameliorated the severity of cerulein-induced AP in WT mice. We conclude that M3R activation can cause the pathogenesis of pancreatitis. This model may provide an alternative approach for pancreatitis research.
Collapse
Affiliation(s)
- Jianhua Wan
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Jiale Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Larry E. Wagner
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - Oliver H. Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Fu Gui
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jiaxiang Chen
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Xiaohui Zhu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Ashley N. Haddock
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Brian Haight
- Prodo Laboratories Inc., Aliso Viejo, California, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Ying Wang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - David I. Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - Yan Bi
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
50
|
Arnold F, Mahaddalkar PU, Kraus JM, Zhong X, Bergmann W, Srinivasan D, Gout J, Roger E, Beutel AK, Zizer E, Tharehalli U, Daiss N, Russell R, Perkhofer L, Oellinger R, Lin Q, Azoitei N, Weiss F, Lerch MM, Liebau S, Katz S, Lechel A, Rad R, Seufferlein T, Kestler HA, Ott M, Sharma AD, Hermann PC, Kleger A. Functional Genomic Screening During Somatic Cell Reprogramming Identifies DKK3 as a Roadblock of Organ Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2100626. [PMID: 34306986 PMCID: PMC8292873 DOI: 10.1002/advs.202100626] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Indexed: 05/06/2023]
Abstract
Somatic cell reprogramming and tissue repair share relevant factors and molecular programs. Here, Dickkopf-3 (DKK3) is identified as novel factor for organ regeneration using combined transcription-factor-induced reprogramming and RNA-interference techniques. Loss of Dkk3 enhances the generation of induced pluripotent stem cells but does not affect de novo derivation of embryonic stem cells, three-germ-layer differentiation or colony formation capacity of liver and pancreatic organoids. However, DKK3 expression levels in wildtype animals and serum levels in human patients are elevated upon injury. Accordingly, Dkk3-null mice display less liver damage upon acute and chronic failure mediated by increased proliferation in hepatocytes and LGR5+ liver progenitor cell population, respectively. Similarly, recovery from experimental pancreatitis is accelerated. Regeneration onset occurs in the acinar compartment accompanied by virtually abolished canonical-Wnt-signaling in Dkk3-null animals. This results in reduced expression of the Hedgehog repressor Gli3 and increased Hedgehog-signaling activity upon Dkk3 loss. Collectively, these data reveal Dkk3 as a key regulator of organ regeneration via a direct, previously unacknowledged link between DKK3, canonical-Wnt-, and Hedgehog-signaling.
Collapse
Affiliation(s)
- Frank Arnold
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Pallavi U Mahaddalkar
- Institute for Diabetes and RegenerationHelmholtz Zentrum MünchenIngolstädter Landstraße 185764 NeuherbergGermany
| | - Johann M. Kraus
- Institute of Medical Systems BiologyUlm UniversityAlbert‐Einstein Allee 1189081 UlmGermany
| | - Xiaowei Zhong
- Department of GastroenterologyHepatology and EndocrinologyHannover Medical SchoolFeodor‐Lynen‐Str. 730625 HannoverGermany
| | - Wendy Bergmann
- Core Facility for Cell Sorting and Cell AnalysisUniversity Medical Center RostockSchillingallee 7018057 RostockGermany
| | - Dharini Srinivasan
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Johann Gout
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Elodie Roger
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Alica K. Beutel
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Eugen Zizer
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Umesh Tharehalli
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Nora Daiss
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Ronan Russell
- Diabetes CenterUniversity of CaliforniaSan FranciscoCA94143USA
| | - Lukas Perkhofer
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Rupert Oellinger
- Institute of Molecular Oncology and Functional GenomicsTranslaTUM Cancer CenterTechnical University of MunichIsmaninger Str. 2281675 MunichGermany
| | - Qiong Lin
- Bayer AG Research & DevelopmentPharmaceuticalsMüllerstraße 17813353 BerlinGermany
| | - Ninel Azoitei
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Frank‐Ulrich Weiss
- Department of Medicine AUniversity Medicine GreifswaldFerdinand‐Sauerbruch‐Straße17475 GreifswaldGermany
| | - Markus M. Lerch
- Department of Medicine AUniversity Medicine GreifswaldFerdinand‐Sauerbruch‐Straße17475 GreifswaldGermany
- Klinikum der Ludwig‐Maximilians‐Universität München‐GroßhadernMarchioninistraße 1581377 MünchenGermany
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology INDBEberhard Karls University TübingenÖsterbergstr. 372074 TübingenGermany
| | - Sarah‐Fee Katz
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - André Lechel
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Roland Rad
- Institute of Molecular Oncology and Functional GenomicsTranslaTUM Cancer CenterTechnical University of MunichIsmaninger Str. 2281675 MunichGermany
| | - Thomas Seufferlein
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Hans A. Kestler
- Institute of Medical Systems BiologyUlm UniversityAlbert‐Einstein Allee 1189081 UlmGermany
| | - Michael Ott
- Department of GastroenterologyHepatology and EndocrinologyHannover Medical SchoolFeodor‐Lynen‐Str. 730625 HannoverGermany
| | - Amar Deep Sharma
- Department of GastroenterologyHepatology and EndocrinologyHannover Medical SchoolFeodor‐Lynen‐Str. 730625 HannoverGermany
| | - Patrick C. Hermann
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| | - Alexander Kleger
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein Allee 2389081 UlmGermany
| |
Collapse
|