1
|
Tani K, Kimura D, Asano Y, Song CY, Shimoda H, Minakawa M. Isogenic Transplantation of Hybrid Artificial Pleural Tissue Consisting of Rat Cells and Polyglycolic Acid Nanofiber Sheet Induces Restoration of Mesothelial Defects in Rat Model. Artif Organs 2025; 49:778-789. [PMID: 39817871 DOI: 10.1111/aor.14947] [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: 04/05/2024] [Revised: 12/12/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Impairment of the visceral pleura following thoracic surgery often leads to air leaks and intrathoracic adhesions. For preventing such complications, mesothelial cell proliferation at the pleural defects can be effective. To develop new materials for pleural defects restoration, we constructed a hybrid artificial pleural tissue (H-APLT) combining polyglycolic acid (PGA) nanofiber sheets with a three-dimensional culture of mesothelial cells and fibroblasts and evaluated its therapeutic efficacy in a rat pleural defect model. METHODS After rat lungs were harvested, pleural mesothelial cells and lung fibroblasts were cultured separately. To construct H-APLT, the cells were then coated with multiple layers of fibronectin and gelatin, followed by a single layer of mesothelial cells on top of multiple layers of fibroblasts accumulated onto a collagen-coated PGA nanofiber sheet. Left lateral thoracotomy was performed, and H-APLTs were transplanted into a rat model with pleural defects (N = 8). After 2-12 weeks of transplantation, lung resection and histological analyses were performed. RESULTS H-APLTs exhibited a pleural structure with a highly integrated mesothelial layer in vitro. After transplantation, all eight rats survived until sacrifice. At 12 weeks post-transplantation, the mesothelial layer on the lung surface was observed to be without defects with no intrathoracic adhesions detected. CONCLUSION Successful isogenic engraftment of H-APLTs was achieved in a rat model of pleural defects. The combination of accumulated fibroblasts and collagen-coated PGA nanofiber sheets contributed to the maintenance of the mesothelial layer's structure and function, potentially preventing air leaks and intrathoracic adhesions.
Collapse
Affiliation(s)
- Kengo Tani
- Department of Thoracic and Cardiovascular Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Daisuke Kimura
- Department of Thoracic and Cardiovascular Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yoshiya Asano
- Department of Neuroanatomy, Cell Biology and Histology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Cheng-Yang Song
- Department of Thoracic and Cardiovascular Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Thoracic Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hiroshi Shimoda
- Department of Neuroanatomy, Cell Biology and Histology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Anatomical Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Masahito Minakawa
- Department of Thoracic and Cardiovascular Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| |
Collapse
|
2
|
Hara D, Sasaki K, Doi S, Ike T, Maeda K, Yoshida M, Takahashi A, Osaki Y, Ishiuchi N, Maeoka Y, Doi T, Chiba T, Nakashima A, Masaki T. Targeting MLL1/WDR5-Mediated Epigenetic Regulation Mitigates Peritoneal Fibrosis by Reducing p16 INK4a. FASEB J 2025; 39:e70543. [PMID: 40232893 DOI: 10.1096/fj.202402382r] [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: 10/07/2024] [Revised: 02/24/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
Peritoneal fibrosis poses a significant challenge to the long-term efficacy of peritoneal dialysis (PD), with emerging evidence highlighting the role of cellular senescence in its pathogenesis. p16INK4a is a cell cycle regulator that has been implicated in cellular senescence. Mixed-lineage leukemia 1 (MLL1) forms a complex with WD-40 repeat protein 5 (WDR5) and exhibits histone H3K4 methyltransferase activity. We have previously shown that inhibition of the MLL1/WDR5 complex reduces p16INK4a expression and attenuates renal senescence after injury in mice. This study aimed to investigate whether inhibiting MLL1/WDR5 attenuates peritoneal senescence, inflammation, and fibrosis in both human samples and in mice with methylglyoxal (MGO)-induced peritoneal fibrosis (MGO-injected mice), while also exploring the associated underlying mechanisms. MLL1/WDR5, histone 3 lysine 4 trimethylation (H3K4me3), and p16INK4a expression were elevated in TGF-β1-stimulated human peritoneal mesothelial cells (HPMCs), non-adherent cells obtained from patients undergoing PD, and the submesothelial compact zones of MGO-injected mice. Notably, p16INK4a expression in these cells was positively correlated with the dialysate/plasma creatinine ratio. Treatment with the MLL1/WDR5 protein-protein interaction inhibitors MM-102 and OICR-9429 reduced H3K4me3 levels and p16INK4a expression, suppressing fibrosis in HPMCs as well as peritoneal fibrosis and inflammation in MGO-injected mice. These inhibitors also improved peritoneal function in MGO-injected mice. Additionally, we demonstrated that MLL1/WDR5-induced H3K4me3 directly regulates p16INK4a gene transcription, and that inhibiting MLL1/WDR5 reduces H3K4me3, thereby suppressing p16INK4a gene transcription. These findings suggest that targeting MLL1/WDR5 activation alleviates peritoneal senescence, inflammation, and fibrosis, highlighting its potential as a promising therapeutic strategy for peritoneal fibrosis.
Collapse
Affiliation(s)
- Daisuke Hara
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kensuke Sasaki
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Shigehiro Doi
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Takeshi Ike
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kazuya Maeda
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Maria Yoshida
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Akira Takahashi
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Yosuke Osaki
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Naoki Ishiuchi
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Yujiro Maeoka
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Toshiki Doi
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
- Division of Nephrology, Ichiyokai Harada Hospital, Hiroshima, Japan
| | - Takuto Chiba
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
3
|
Neyroud D, D'Lugos A, Trevino E, Callaway C, Lamm J, Laitano O, Poole B, Deyhle M, Brantley J, Le L, Judge A, Judge S. Local Inflammation Precedes Diaphragm Wasting and Fibrotic Remodelling in a Mouse Model of Pancreatic Cancer. J Cachexia Sarcopenia Muscle 2025; 16:e13668. [PMID: 39810606 PMCID: PMC11733308 DOI: 10.1002/jcsm.13668] [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: 07/29/2024] [Revised: 10/16/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Cancer cachexia represents a debilitating muscle wasting condition that is highly prevalent in gastrointestinal cancers, including pancreatic ductal adenocarcinoma (PDAC). Cachexia is estimated to contribute to ~30% of cancer-related deaths, with deterioration of respiratory muscles suspected to be a key contributor to cachexia-associated morbidity and mortality. In recent studies, we identified fibrotic remodelling of respiratory accessory muscles as a key feature of human PDAC cachexia. METHODS To gain insight into mechanisms driving respiratory muscle wasting and fibrotic remodelling in response to PDAC, we conducted temporal histological and transcriptomic analyses on diaphragm muscles harvested from mice-bearing orthotopic murine pancreatic (KPC) tumours at time points reflective of precachexia (D8 and D10), mild-moderate cachexia (D12 and D14) and advanced cachexia (endpoint). RESULTS During the precachexia phase, diaphragms showed significant leukocyte infiltration (+3-fold to +13-fold; D8-endpoint vs. Sham, p < 0.05) and transcriptomic enrichment of inflammatory processes associated with tissue injury that remained increased through endpoint. Diaphragm inflammation was followed by increases in PDGFR-ɑ+ fibroadipogenic progenitors (+2.5 to +3.8-fold; D10-endpoint vs. Sham, p < 0.05), fibre atrophy (-16% to -24%, D12 to endpoint vs. Sham, p < 0.05), ECM expansion (+1.5 to +1.8-fold; D14-endpoint vs. Sham, p < 0.05), collagen accumulation (+3.8-fold; endpoint vs. Sham, p = 0.0013) and reductions in breathing frequency (-55%, p = 0.0074) and diaphragm excursion (-43%, p = 0.0006). These biological processes were supported by changes in the diaphragm transcriptome. Ingenuity pathway analysis predicted factors involved in inflammatory responses to tissue injury, including TGF-β1, angiotensin and PDGF BB, as top upstream regulators activated in diaphragms prior to and throughout cachexia progression, while PGC-1α and the insulin receptor were among the top upstream regulators predicted to be suppressed. The transcriptomic dataset further revealed progressive disturbances to networks involved in lipid, glucose and oxidative metabolism, activation of the unfolded protein response and neuromuscular junction remodelling associated with denervation. CONCLUSIONS In summary, our data support leukocyte infiltration and expansion of PDGFRα mesenchymal progenitors as early events that precede wasting and fibrotic remodelling of the diaphragm in response to PDAC that may also underlie metabolic disturbances, weakness and respiratory complications.
Collapse
Affiliation(s)
- Daria Neyroud
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
- Institute of Sports SciencesUniversity of LausanneLausanneSwitzerland
| | - Andrew C. D'Lugos
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Enrique J. Trevino
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
| | - Chandler S. Callaway
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Jacqueline Lamm
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
| | - Orlando Laitano
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFloridaUSA
| | - Brittney Poole
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Physiology and Aging, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Michael R. Deyhle
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Justina Brantley
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
| | - Lam Le
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
| | - Andrew R. Judge
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Physiology and Aging, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Sarah M. Judge
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Physiology and Aging, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
4
|
Hausburg MA, Banton KL, Cassidy CD, Madayag RM, Palacio CH, Williams JS, Bar-Or R, Ryznar RJ, Bar-Or D. Mesothelial cell responses to acute appendicitis or small bowel obstruction reactive ascites: Insights into immunoregulation of abdominal adhesion. PLoS One 2025; 20:e0317056. [PMID: 39775680 PMCID: PMC11709316 DOI: 10.1371/journal.pone.0317056] [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: 10/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Previous abdominal surgery (PAS) increases risk of small bowel obstruction (SBO) due to adhesions, and appendectomy (appy) is an independent risk factor for abdominal adhesion-related complications. Peritoneal inflammation, e.g., acute appendicitis (AA), causes formation of reactive ascitic fluid (rA) that activates peritoneum surface mesothelial cells (MCs) to form adhesions. Pathologic adhesions may arise if restoration of MC-regulated fibrinolysis and secretion of glycocalyx (GCX) are disrupted. Proteins affecting these processes may originate from peritoneal rA. This is a prospective observational IRB-approved study at three Level 1 trauma centers where rA is collected prior to surgical intervention for non-perforated AA or adhesiolysis for SBO. Samples from 48 appy and 15 SBO patients were used to treat human MCs and subjected to quantification of 85 inflammatory mediators. Results were compared between patients with surgically naïve abdomens (naïve) and patients with >1 PAS. Select rA caused MCs to form clusters of fibroblastic cells, extracellular matrix fibers (FIB), and secretion of GCX. PAS and naïve patient rA fluids were clustered into "fiber-GCX" (FIB-GCX) groups: highFIB-highGCX, highFIB-lowGCX, noFIB-highGCX, noFIB-lowGCX, and noFIB-noGCX. Between groups, 26 analytes were differentially abundant including innate immune response, wound healing, and mucosal defense proteins. Factors that contributed to the differences between groups were rA-induced highFIB and history of PAS. Overall, PAS patient rA showed a muted immune response compared to rA from naïve patients. Our data suggest that abdominal surgery may negatively impact future immune responses in the abdomen. Further, quantifying immunomodulators in peritoneal rA may lead to the development a personalized approach to post-surgical adhesion treatment and prevention.
Collapse
Affiliation(s)
- Melissa A. Hausburg
- Trauma Research, Swedish Medical Center, Englewood, Colorado, United States of America
- Trauma Research, Wesley Medical Center, Wichita, Kansas, United States of America
- Trauma Services, Lutheran Hospital, Wheat Ridge, Colorado, United States of America
- Trauma Research, South Texas Health System McAllen, McAllen, Texas, United States of America
| | - Kaysie L. Banton
- Trauma Services, Swedish Medical Center, Englewood, Colorado, United States of America
| | | | - Robert M. Madayag
- Trauma Services, Lutheran Hospital, Wheat Ridge, Colorado, United States of America
- Trauma Services, St. Anthony Hospital, Lakewood, Colorado, United States of America
| | - Carlos H. Palacio
- Trauma Services, South Texas Health System McAllen, McAllen, Texas, United States of America
| | - Jason S. Williams
- Trauma Research, Swedish Medical Center, Englewood, Colorado, United States of America
- Trauma Research, Wesley Medical Center, Wichita, Kansas, United States of America
- Trauma Services, Lutheran Hospital, Wheat Ridge, Colorado, United States of America
- Trauma Research, South Texas Health System McAllen, McAllen, Texas, United States of America
| | - Raphael Bar-Or
- Trauma Research, Swedish Medical Center, Englewood, Colorado, United States of America
- Trauma Research, Wesley Medical Center, Wichita, Kansas, United States of America
- Trauma Services, Lutheran Hospital, Wheat Ridge, Colorado, United States of America
- Trauma Research, South Texas Health System McAllen, McAllen, Texas, United States of America
| | - Rebecca J. Ryznar
- Department of Molecular Biology, Rocky Vista University, Parker, Colorado, United States of America
| | - David Bar-Or
- Trauma Research, Swedish Medical Center, Englewood, Colorado, United States of America
- Trauma Research, Wesley Medical Center, Wichita, Kansas, United States of America
- Trauma Services, Lutheran Hospital, Wheat Ridge, Colorado, United States of America
- Trauma Research, South Texas Health System McAllen, McAllen, Texas, United States of America
| |
Collapse
|
5
|
Liang S, Qian Y, Liu Y, Wang Y, Su L, Yan S. Ligustrazine nanoparticles inhibits epithelial-mesenchymal transition and alleviates postoperative abdominal adhesion. Biochem Biophys Res Commun 2024; 739:150994. [PMID: 39547120 DOI: 10.1016/j.bbrc.2024.150994] [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: 08/13/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Following abdominal surgery, the occurrence of postoperative abdominal adhesion (PAA) is highly prevalent and stands out as one of the most frequently encountered complications. The effect and molecular mechanisms of Ligustrazine nanoparticles (LN) underlying epithelial-mesenchymal transition (EMT) in PAA still remain elusive. Adhesions were induced in Male Sprague-Dawley rats by injuring the cecum (cecal abrasion model), followed by administration of LN and hyaluronate acid (HA). The mechanism was further verified by enzyme-linked immunosorbent assay, wound healing assay, si-RNA and Western blot. Animal experiments revealed that LN effectively ameliorated adhesions, notably decreased tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-8, and fibrosis, and reduced the expression of TGF-β1 and EMT related markers (Fibronectin and E-cadherin). Furthermore, in vitro experiments demonstrated that LN might inhibit the TGF-β1 FOXC2 pathway through suppressing the expression of Fibronectin, P120, and E-cadherin and ameliorating peritoneal adhesion. Collectively, our findings indicate that LN inhibits PAA formation by reducing inflammation, decreasing EMT and promoting peritoneal mesothelial cell repair. Therefore, LN might be considered a potential candidate for the treatment of PPA. However, further clinical studies are required to approve the effectiveness of LN.
Collapse
Affiliation(s)
- Shasha Liang
- Teaching and Research Office of Obstetrics and Gynecology, Medical College of Zhengzhou University of Industrial Technology, Xinzheng, 451100, Henan, China
| | - Yifei Qian
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Ying Liu
- Department of Gynaecology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215009, Jiangsu, China
| | - Yahui Wang
- Department of Anorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215009, Jiangsu, China
| | - Lianlin Su
- Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Shuai Yan
- Department of Anorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215009, Jiangsu, China.
| |
Collapse
|
6
|
Wu T, Yu Z, Dai J, Li J, Ning F, Liu X, Zhu N, Zhang X. JPH203 alleviates peritoneal fibrosis via inhibition of amino acid-mediated mTORC1 signaling. Biochem Biophys Res Commun 2024; 734:150656. [PMID: 39362029 DOI: 10.1016/j.bbrc.2024.150656] [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: 07/11/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND AND AIMS The mesothelial-mesenchymal transition (MMT) of mesothelial cells has been recognized as a critical process during progression of peritoneal fibrosis (PF). Despite its crucial role in amino acid transport and metabolism, the involvement of L-type amino acid transporter 1 (LAT1) and the potential therapeutic role of its inhibitor, JPH203, in fibrotic diseases remain unexplored. Considering the paucity of research on amino acid-mediated mTORC1 activation in PF, our study endeavors to elucidate the protective effects of JPH203 against PF and explore the involvement of amino acid-mediated mTORC1 signaling in this context. METHODS We established the transforming growth factor beta 1 (TGF-β1) induced MMT model in primary human mesothelial cells and the peritoneal dialysis fluid (PDF) induced PF model in mice. The therapeutic effects of JPH203 on PF were then examined on these two models by real-time quantitative polymerase chain reaction, western blotting, immunofluorescence staining, Masson's trichrome staining, H&E staining, picro-sirius red staining, and immunohistochemistry. The involvement of amino acid-mediated mTORC1 signaling was screened by RNA sequencing and further verified by western blotting in vitro. RESULTS LAT1 was significantly upregulated and JPH203 markedly attenuated fibrotic phenotype both in vitro and in vivo. RNA-seq unveiled a significant enrichment of mTOR signaling pathway in response to JPH203 treatment. Western blotting results indicated that JPH203 alleviates PF by inhibiting amino acid-mediated mTORC1 signaling, which differs from the direct inhibition observed with rapamycin. CONCLUSION JPH203 alleviates PF by inhibiting amino acid-mediated mTORC1 signaling.
Collapse
Affiliation(s)
- Tiangang Wu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Zanzhe Yu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Junhao Dai
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiayang Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Fengling Ning
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xin Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Nan Zhu
- Department of Nephrology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China; School of Pharmacy, East China Normal University, Shanghai, China.
| |
Collapse
|
7
|
Niayesh-Mehr R, Kalantar M, Bontempi G, Montaldo C, Ebrahimi S, Allameh A, Babaei G, Seif F, Strippoli R. The role of epithelial-mesenchymal transition in pulmonary fibrosis: lessons from idiopathic pulmonary fibrosis and COVID-19. Cell Commun Signal 2024; 22:542. [PMID: 39538298 PMCID: PMC11558984 DOI: 10.1186/s12964-024-01925-y] [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: 08/16/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Despite the tremendous advancements in the knowledge of the pathophysiology and clinical aspects of SARS-CoV-2 infection, still many issues remain unanswered, especially in the long-term effects. Mounting evidence suggests that pulmonary fibrosis (PF) is one of the most severe complications associated with COVID-19. Therefore, understanding the molecular mechanisms behind its development is helpful to develop successful therapeutic strategies. Epithelial to mesenchymal transition (EMT) and its cell specific variants endothelial to mesenchymal transition (EndMT) and mesothelial to mesenchymal transition (MMT) are physio-pathologic cellular reprogramming processes induced by several infectious, inflammatory and biomechanical stimuli. Cells undergoing EMT acquire invasive, profibrogenic and proinflammatory activities by secreting several extracellular mediators. Their activity has been implicated in the pathogenesis of PF in a variety of lung disorders, including idiopathic pulmonary fibrosis (IPF) and COVID-19. Aim of this article is to provide an updated survey of the cellular and molecular mechanisms, with emphasis on EMT-related processes, implicated in the genesis of PF in IFP and COVID-19.
Collapse
Affiliation(s)
- Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojtaba Kalantar
- Department of Occupational Health, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Giulio Bontempi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Claudia Montaldo
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Saeedeh Ebrahimi
- Department of Medical Microbiology (Bacteriology and Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ghader Babaei
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Faezeh Seif
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy.
| |
Collapse
|
8
|
Moran HR, Nyarko OO, O’Rourke R, Ching RCK, Riemslagh FW, Peña B, Burger A, Sucharov CC, Mosimann C. The pericardium forms as a distinct structure during heart formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613484. [PMID: 39345600 PMCID: PMC11429720 DOI: 10.1101/2024.09.18.613484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The heart integrates diverse cell lineages into a functional unit, including the pericardium, a mesothelial sac that supports heart movement, homeostasis, and immune responses. However, despite its critical roles, the developmental origins of the pericardium remain uncertain due to disparate models. Here, using live imaging, lineage tracking, and single-cell transcriptomics in zebrafish, we find the pericardium forms within the lateral plate mesoderm from dedicated anterior mesothelial progenitors and distinct from the classic heart field. Imaging of transgenic reporters in zebrafish documents lateral plate mesoderm cells that emerge lateral of the classic heart field and among a continuous mesothelial progenitor field. Single-cell transcriptomics and trajectories of hand2-expressing lateral plate mesoderm reveal distinct populations of mesothelial and cardiac precursors, including pericardial precursors that are distinct from the cardiomyocyte lineage. The mesothelial gene expression signature is conserved in mammals and carries over to post-natal development. Light sheet-based live-imaging and machine learning-supported cell tracking documents that during heart tube formation, pericardial precursors that reside at the anterior edge of the heart field migrate anteriorly and medially before fusing, enclosing the embryonic heart to form a single pericardial cavity. Pericardium formation proceeds even upon genetic disruption of heart tube formation, uncoupling the two structures. Canonical Wnt/β-catenin signaling modulates pericardial cell number, resulting in a stretched pericardial epithelium with reduced cell number upon canonical Wnt inhibition. We connect the pathological expression of secreted Wnt antagonists of the SFRP family found in pediatric dilated cardiomyopathy to increased pericardial stiffness: sFRP1 in the presence of increased catecholamines causes cardiomyocyte stiffness in neonatal rats as measured by atomic force microscopy. Altogether, our data integrate pericardium formation as an independent process into heart morphogenesis and connect disrupted pericardial tissue properties such as pericardial stiffness to pediatric cardiomyopathies.
Collapse
Affiliation(s)
- Hannah R. Moran
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Obed O. Nyarko
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rebecca O’Rourke
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Ryenne-Christine K. Ching
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Frederike W. Riemslagh
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Brisa Peña
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Cardiovascular Institute, Division of Cardiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Bioengineering Department, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Alexa Burger
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Carmen C. Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
9
|
Weber MC, Clees Z, Buck A, Fischer A, Steffani M, Wilhelm D, Martignoni M, Friess H, Rinkevich Y, Neumann PA. Role of the serosa in intestinal anastomotic healing: insights from in-depth histological analysis of human and murine anastomoses. BJS Open 2024; 8:zrae108. [PMID: 39230923 PMCID: PMC11373408 DOI: 10.1093/bjsopen/zrae108] [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: 06/21/2024] [Accepted: 07/27/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Anastomotic leakage following colorectal surgery remains a significant complication despite advances in surgical techniques. Recent findings on serosal injury repair in coelomic cavities, such as the peritoneum, challenge the current understanding of the cellular origins and mechanisms underlying intestinal anastomotic healing. Understanding the contribution of each layer of the intestinal wall during anastomotic healing is needed to find new therapeutic strategies to prevent anastomotic leakage. The aim of this experimental study was to investigate the role of the serosal layer of the intestinal wall in anastomotic healing. MATERIALS AND METHODS Comprehensive histologic analysis of human and murine anastomoses was performed to elucidate histologic changes in the different intestinal layers during anastomotic healing. In vivo staining of the extracellular matrix (ECM) in the serosal layer was performed using a fluorophore-conjugated N-hydroxysuccinimide-ester before anastomosis surgery in a murine model. RESULTS Histological examination of both human and murine anastomoses revealed that closure of the serosal layer occurred first during the healing process. In vivo serosal ECM staining demonstrated that a significant portion of the newly formed ECM within the anastomosis was indeed deposited onto the serosal layer. Furthermore, mesenchymal cells within the anastomotic scar were positive for mesothelial cell markers, podoplanin and Wilms tumour protein. CONCLUSIONS In this experimental study, the results suggest that serosal scar formation is an important mechanism for anastomotic integrity in intestinal anastomoses. Mesothelial cells may significantly contribute to scar formation during anastomotic healing through epithelial-to-mesenchymal transition, potentially suggesting a novel therapeutic target to prevent anastomotic leakage by enhancing physiological healing processes.
Collapse
Affiliation(s)
- Marie-Christin Weber
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Zoé Clees
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Annalisa Buck
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Munich, Germany
| | - Adrian Fischer
- Institute of Regenerative Biology and Medicine, Helmholtz Munich, Munich, Germany
| | - Marcella Steffani
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Dirk Wilhelm
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Marc Martignoni
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Munich, Munich, Germany
| | - Philipp-Alexander Neumann
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Munich, Germany
| |
Collapse
|
10
|
Daley AD, Bénézech C. Fat-associated lymphoid clusters: Supporting visceral adipose tissue B cell function in immunity and metabolism. Immunol Rev 2024; 324:78-94. [PMID: 38717136 DOI: 10.1111/imr.13339] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/09/2024] [Indexed: 07/23/2024]
Abstract
It is now widely understood that visceral adipose tissue (VAT) is a highly active and dynamic organ, with many functions beyond lipid accumulation and storage. In this review, we discuss the immunological role of this tissue, underpinned by the presence of fat-associated lymphoid clusters (FALCs). FALC's distinctive structure and stromal cell composition support a very different immune cell mix to that found in classical secondary lymphoid organs, which underlies their unique functions of filtration, surveillance, innate-like immune responses, and adaptive immunity within the serous cavities. FALCs are important B cell hubs providing B1 cell-mediated frontline protection against infection and supporting B2 cell-adaptative immune responses. Beyond these beneficial immune responses orchestrated by FALCs, immune cells within VAT play important homeostatic role. Dysregulation of immune cells during obesity and aging leads to chronic pathological "metabolic inflammation", which contributes to the development of cardiometabolic diseases. Here, we examine the emerging and complex functions of B cells in VAT homeostasis and the metabolic complications of obesity, highlighting the potential role that FALCs play and emphasize the areas where further research is needed.
Collapse
Affiliation(s)
- Alexander D Daley
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
11
|
Lu Y, Elrod J, Herrmann M, Knopf J, Boettcher M. Neutrophil Extracellular Traps: A Crucial Factor in Post-Surgical Abdominal Adhesion Formation. Cells 2024; 13:991. [PMID: 38891123 PMCID: PMC11171752 DOI: 10.3390/cells13110991] [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: 04/07/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Post-surgical abdominal adhesions, although poorly understood, are highly prevalent. The molecular processes underlying their formation remain elusive. This review aims to assess the relationship between neutrophil extracellular traps (NETs) and the generation of postoperative peritoneal adhesions and to discuss methods for mitigating peritoneal adhesions. A keyword or medical subject heading (MeSH) search for all original articles and reviews was performed in PubMed and Google Scholar. It included studies assessing peritoneal adhesion reformation after abdominal surgery from 2003 to 2023. After assessing for eligibility, the selected articles were evaluated using the Critical Appraisal Skills Programme checklist for qualitative research. The search yielded 127 full-text articles for assessment of eligibility, of which 7 studies met our criteria and were subjected to a detailed quality review using the Critical Appraisal Skills Programme (CASP) checklist. The selected studies offer a comprehensive analysis of adhesion pathogenesis with a special focus on the role of neutrophil extracellular traps (NETs) in the development of peritoneal adhesions. Current interventional strategies are examined, including the use of mechanical barriers, advances in regenerative medicine, and targeted molecular therapies. In particular, this review emphasizes the potential of NET-targeted interventions as promising strategies to mitigate postoperative adhesion development. Evidence suggests that in addition to their role in innate defense against infections and autoimmune diseases, NETs also play a crucial role in the formation of peritoneal adhesions after surgery. Therefore, therapeutic strategies that target NETs are emerging as significant considerations for researchers. Continued research is vital to fully elucidate the relationship between NETs and post-surgical adhesion formation to develop effective treatments.
Collapse
Affiliation(s)
- Yuqing Lu
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Julia Elrod
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Martin Herrmann
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Jasmin Knopf
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
12
|
Lecoutre S, Rebière C, Marcelin G, Clément K. How does bariatric surgery remodel adipose tissue? ANNALES D'ENDOCRINOLOGIE 2024; 85:175-178. [PMID: 38871506 DOI: 10.1016/j.ando.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
This lecture delves into the pivotal role of adipose tissue in obesity and its response to weight loss, particularly via bariatric surgery. Adipose tissue, responsible for storing excess energy, undergoes significant changes during obesity, marked by inflammation and fibrosis. Bariatric surgery, serving as a model, allow the exploration of adipose tissue remodeling post-weight loss, inducing metabolic and fibro-inflammatory shifts. Despite successful weight loss, inflammation and fibrosis persist, as evidenced by changes in immune cells, altered cytokine profiles and the accumulation of extracellular matrix (ECM). Unfortunately, these lingering effects impair the normal adipose tissue function. In this context, adipose progenitors, an heterogenous resident population of mesenchymal stromal cells, display functions important to fibrosis development, capable of differentiating into myofibroblasts and contributing to ECM deposition. Particularly, a distinct subpopulation of adipose progenitors with high CD9 expression (CD9high) is associated with fibrosis and insulin resistance in human obesity. The persistence of fibrosis post-weight loss poses challenges, correlating with metabolic dysfunction despite improved glucose tolerance. A comprehensive understanding of the mechanisms driving adipose tissue remodeling and fibrosis post-weight loss is imperative for the development of effective treatments for obesity. The intricate interplay between adipose tissue, inflammation, and fibrosis underscores the necessity for further in-depth research to elucidate these mechanisms and formulate targeted therapies for obesity-related complications.
Collapse
Affiliation(s)
- Simon Lecoutre
- Research Unit: Nutrition and Obesities; Systemic Approaches, NutriOmics, Inserm, Sorbonne université, Paris, France
| | - Clémentine Rebière
- Research Unit: Nutrition and Obesities; Systemic Approaches, NutriOmics, Inserm, Sorbonne université, Paris, France; Nutrition Department, Pitié-Salpêtrière Hospital, Paris Public Hospitals, Paris, France
| | - Geneviève Marcelin
- Nutrition Department, Pitié-Salpêtrière Hospital, Paris Public Hospitals, Paris, France
| | - Karine Clément
- Research Unit: Nutrition and Obesities; Systemic Approaches, NutriOmics, Inserm, Sorbonne université, Paris, France.
| |
Collapse
|
13
|
Alcocer B, Bou P, Bosch L, Torrente C. Presumptive spontaneous hemothorax associated to thymic involution in a dog with steroid responsive meningitis-arteritis (SRMA). Top Companion Anim Med 2024; 60:100863. [PMID: 38513795 DOI: 10.1016/j.tcam.2024.100863] [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: 10/26/2023] [Revised: 01/29/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024]
Abstract
OBJECTIVE To describe an unusual case of spontaneous hemothorax resulting from thymic involution in a dog with suspected acquired bleeding dyscrasia associated with steroid-responsive meningitis-arteritis (SRMA). CASE DESCRIPTION A 6-month-old spayed female Golden Retriever was referred due to the sudden onset of lethargy, fever (pyrexia), loss of appetite (anorexia), and moderate neck pain. These symptoms emerged six days after an ovariohysterectomy performed by the primary veterinarian. Upon admission, the patient exhibited pale mucous membranes, tachycardia (180 bpm), bilateral muffled heart sounds and tachypnea. Abdominal and thoracic point-of-care ultrasound (POCUS) were performed and revealed bilateral pleural effusion. Due to the patient's unstable condition, emergent thoracocentesis and transfusion of packed red blood cells was required. The initial work-up performed included a complete blood cell count (CBC), biochemistry profile, venous blood gas and coagulation panel (PT, APTT, fibrinogen). Pleural effusion analysis was compatible with hemothorax. Bloodwork was unremarkable including the initial coagulation panel. Further coagulation test was performed including buccal mucosal bleeding time, viscoelastic-based clot detection tests (TEG) and Von Willebrand factor antigen measurement. TEG revealed marked hyperfibrinolysis. Angiostrongylus vasorum and 4DX snap test were performed and yielded a negative result. Thoracic CT scan revealed the presence of a soft tissue-attenuating mass in the ventral mediastinum, thymic involution, and enlargement of the sternal and mediastinal lymph nodes. Therapy with tranexamic acid and corticosteroids at anti-inflammatory doses was initiated. Marked clinical improvement was observed within 24 hours, and after three days of hospitalization the patient was discharged. One month later, the dog was referred again for acute pyrexia, hyporexia, and neck pain which progressed to non-ambulatory tetraparesis. Neurological examination was compatible with C6-T2 lesion. MRI and cerebrospinal fluid analysis were performed and revealed a final diagnosis of steroid-responsive meningitis-arteritis (SRMA) with associated intramedullary hemorrhage. Corticosteroids were started again, and the patient showed a dramatic improvement over the next 24 hours. Three weeks after the diagnosis, the dog returned to a clinically normal state. The treatment was gradually tapered over the following months, guided by regular neurological and clinical examinations and CRP measurements, without any relapses. NEW OR UNIQUE INFORMATION To the best of the author's knowledge, this is the first documented case of a dog experiencing spontaneous hemothorax as a result of thymic hemorrhage/involution which, in the absence of other identifiable diseases, was attributed to a hyperfibrinolytic state induced by a severe inflammatory disease such as SRMA.
Collapse
Affiliation(s)
- B Alcocer
- Emergency and Critical Care Service, Fundació Hospital Clínic Veterinari-UAB, Universitat Autònoma de Barcelona (UAB), Campus, Carrer de l'Hospital, s/n, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - P Bou
- Emergency and Critical Care Service, Fundació Hospital Clínic Veterinari-UAB, Universitat Autònoma de Barcelona (UAB), Campus, Carrer de l'Hospital, s/n, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - L Bosch
- Emergency and Critical Care Service, Fundació Hospital Clínic Veterinari-UAB, Universitat Autònoma de Barcelona (UAB), Campus, Carrer de l'Hospital, s/n, 08193 Cerdanyola del Vallès, Barcelona, Spain; Animal Medicine and Surgery Department, Facultat de Veterinària de la UAB, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
| | - C Torrente
- Emergency and Critical Care Service, Fundació Hospital Clínic Veterinari-UAB, Universitat Autònoma de Barcelona (UAB), Campus, Carrer de l'Hospital, s/n, 08193 Cerdanyola del Vallès, Barcelona, Spain; Animal Medicine and Surgery Department, Facultat de Veterinària de la UAB, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
| |
Collapse
|
14
|
Choo YY, Sakai T, Ikebe R, Jeffers A, Idell S, Tucker TA, Ikebe M. Role of ZIP kinase in development of myofibroblast differentiation from HPMCs. Am J Physiol Lung Cell Mol Physiol 2024; 326:L353-L366. [PMID: 38252666 PMCID: PMC11281797 DOI: 10.1152/ajplung.00251.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/14/2023] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
During the development of pleural fibrosis, pleural mesothelial cells (PMCs) undergo phenotypic switching from differentiated mesothelial cells to mesenchymal cells (MesoMT). Here, we investigated how external stimuli such as TGF-β induce HPMC-derived myofibroblast differentiation to facilitate the development of pleural fibrosis. TGF-β significantly increased di-phosphorylation but not mono-phosphorylation of myosin II regulatory light chain (RLC) in HPMCs. An increase in RLC di-phosphorylation was also found at the pleural layer of our carbon black bleomycin (CBB) pleural fibrosis mouse model, where it showed filamentous localization that coincided with alpha smooth muscle actin (αSMA) in the cells in the pleura. Among the protein kinases that can phosphorylate myosin II RLC, ZIPK (zipper-interacting kinase) protein expression was significantly augmented after TGF-β stimulation. Furthermore, ZIPK gene silencing attenuated RLC di-phosphorylation, suggesting that ZIPK is responsible for di-phosphorylation of myosin II in HPMCs. Although TGF-β significantly increased the expression of ZIP kinase protein, the change in ZIP kinase mRNA was marginal, suggesting a posttranscriptional mechanism for the regulation of ZIP kinase expression by TGF-β. ZIPK gene knockdown (KD) also significantly reduced TGF-β-induced upregulation of αSMA expression. This finding suggests that siZIPK attenuates myofibroblast differentiation of HPMCs. siZIPK diminished TGF-β-induced contractility of HPMCs consistent with siZIPK-induced decrease in the di-phosphorylation of myosin II RLC. The present results implicate ZIPK in the regulation of the contractility of HPMC-derived myofibroblasts, phenotype switching, and myofibroblast differentiation of HPMCs.NEW & NOTEWORTHY Here, we highlight that ZIP kinase is responsible for di-phosphorylation of myosin light chain, which facilitates stress fiber formation and actomyosin-based cell contraction during mesothelial to mesenchymal transition in human pleural mesothelial cells. This transition has a significant impact on tissue remodeling and subsequent stiffness of the pleura. This study provides insight into a new therapeutic strategy for the treatment of pleural fibrosis.
Collapse
Affiliation(s)
- Young-Yeon Choo
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Tsuyoshi Sakai
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Reiko Ikebe
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Ann Jeffers
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Torry A Tucker
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| |
Collapse
|
15
|
Zhang J, Li H, Zhong H, Chen X, Hu ZX. Omega-3 polyunsaturated fatty acids protect peritoneal mesothelial cells from hyperglycolysis and mesothelial-mesenchymal transition through the FFAR4/CaMKKβ/AMPK/mTOR signaling pathway. Int Immunopharmacol 2024; 128:111561. [PMID: 38262160 DOI: 10.1016/j.intimp.2024.111561] [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: 12/24/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/25/2024]
Abstract
Peritoneal fibrosis is a severe clinical complication associated with peritoneal dialysis (PD) and impacts its efficacy and patient outcomes. The process of mesothelial-mesenchymal transition (MMT) in peritoneal mesothelial cells plays a pivotal role in fibrogenesis, whereas metabolic reprogramming, characterized by excessive glycolysis, is essential in MMT development. No reliable therapies are available despite substantial progress made in understanding the mechanisms underlying peritoneal fibrosis. Protective effect of omega-3 polyunsaturated fatty acids (ω3 PUFAs) has been described in PD-induced peritoneal fibrosis, although the detailed mechanisms remain unknown. It is known that ω3 PUFAs bind to and activate the free fatty acid receptor 4 (FFAR4). However, the expression and role of FFAR4 in the peritoneum have not been investigated. Thus, we hypothesized that ω3 PUFAs would alleviate peritoneal fibrosis by inhibiting hyperglycolysis and MMT through FFAR4 activation. First, we determined FFAR4 expression in peritoneal mesothelium in humans and mice. FFAR4 expression was abnormally decreased in patients on PD and mice and HMrSV5 mesothelial cells exposed to PD fluid (PDF); this change was restored by the ω3 PUFAs (EPA and DHA). ω3 PUFAs significantly inhibited peritoneal hyperglycolysis, MMT, and fibrosis in PDF-treated mice and HMrSV5 mesothelial cells; these changes induced by ω3 PUFAs were blunted by treatment with the FFAR4 antagonist AH7614 and FFAR4 siRNA. Additionally, ω3 PUFAs induced FFAR4, Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ), and AMPK and suppressed mTOR, leading to the inhibition of hyperglycolysis, demonstrating that the ω3 PUFAs-mediated FFAR4 activation ameliorated peritoneal fibrosis by inhibiting hyperglycolysis and MMT via CaMKKβ/AMPK/mTOR signaling. As natural FFAR4 agonists, ω3 PUFAs may be considered for the treatment of PD-associated peritoneal fibrosis.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Li
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Zhong
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhang-Xue Hu
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China; National Clinical Research Center for Geriatrics and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
Pitaraki E, Jagirdar RM, Rouka E, Bartosova M, Sinis SI, Gourgoulianis KI, Eleftheriadis T, Stefanidis I, Liakopoulos V, Hatzoglou C, Schmitt CP, Zarogiannis SG. 2-Deoxy-glucose ameliorates the peritoneal mesothelial and endothelial barrier function perturbation occurring due to Peritoneal Dialysis fluids exposure. Biochem Biophys Res Commun 2024; 693:149376. [PMID: 38104523 DOI: 10.1016/j.bbrc.2023.149376] [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: 10/12/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Peritoneal dialysis (PD) and prolonged exposure to PD fluids (PDF) induce peritoneal membrane (PM) fibrosis and hypervascularity, leading to functional PM degeneration. 2-deoxy-glucose (2-DG) has shown potential as PM antifibrotic by inhibiting hyper-glycolysis induced mesothelial-to-mesenchymal transition (MMT). We investigated whether administration of 2-DG with several PDF affects the permeability of mesothelial and endothelial barrier of the PM. The antifibrotic effect of 2-DG was confirmed by the gel contraction assay with embedded mesothelial (MeT-5A) or endothelial (EA.hy926) cells cultured in Dianeal® 2.5 % (CPDF), BicaVera® 2.3 % (BPDF), Balance® 2.3 % (LPDF) with/without 2-DG addition (0.2 mM), and qPCR for αSMA, CDH2 genes. Moreover, 2-DG effect was tested on the permeability of monolayers of mesothelial and endothelial cells by monitoring the transmembrane resistance (RTM), FITC-dextran (10, 70 kDa) diffusion and mRNA expression levels of CLDN-1 to -5, ZO1, SGLT1, and SGLT2 genes. Contractility of MeT-5A cells in CPDF/2-DG was decreased, accompanied by αSMA (0.17 ± 0.03) and CDH2 (2.92 ± 0.29) gene expression fold changes. Changes in αSMA, CDH2 were found in EA.hy926 cells, though αSMA also decreased under LPDF/2-DG incubation (0.42 ± 0.02). Overall, 2-DG mitigated the PDF-induced alterations in mesothelial and endothelial barrier function as shown by RTM, dextran transport and expression levels of the CLDN-1 to -5, ZO1, and SGLT2. Thus, supplementation of PDF with 2-DG not only reduces MMT but also improves functional permeability characteristics of the PM mesothelial and endothelial barrier.
Collapse
Affiliation(s)
- Eleanna Pitaraki
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Rajesh M Jagirdar
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Erasmia Rouka
- Department of Nursing, School of Health Sciences, University of Thessaly, GAIOPOLIS, 41500, Larissa, Greece
| | - Maria Bartosova
- Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, 69120, Heidelberg, Germany
| | - Sotirios I Sinis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece; Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Konstantinos I Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Theodoros Eleftheriadis
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Vassilios Liakopoulos
- 2(nd) Department of Nephrology, AHEPA Hospital, Aristotle University of Thessaloniki, 54636, Thessaloniki, Greece
| | - Chrissi Hatzoglou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Claus Peter Schmitt
- Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, 69120, Heidelberg, Germany
| | - Sotirios G Zarogiannis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece.
| |
Collapse
|
17
|
Dai H, Shan Y, Yu M, Wang F, Zhou Z, Sun J, Sheng L, Huang L, Sheng M. Network pharmacology, molecular docking and experimental verification of the mechanism of huangqi-jixuecao herb pair in treatment of peritoneal fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116874. [PMID: 37437794 DOI: 10.1016/j.jep.2023.116874] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/30/2023] [Accepted: 07/01/2023] [Indexed: 07/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Huangqi-Jixuecao herb pair (HQJXCHP) is a traditional herbal formula composed of two widely applied TCM prescriptions, Huangqi (Astragalus membranaceus (Fisch.) Bunge) and Jixuecao (Centella asiatica (L.) Urb.), used for hundreds of years to replenish qi and clear away heat. However, the therapeutic effects of HQJXCHP against peritoneal fibrosis (PF) and potential targets are currently unclear. AIMS OF THE STUDY The main objective of this study was preliminary prediction and validation of the effects and molecular mechanisms of action of HQJXCHP against PF based on network pharmacology analysis and experimental verification. MATERIALS AND METHODS The ingredients of HQJXCHP were analyzed via HPLC-Q-TOF/MS. Bioactive compounds of HQJXCHP used for network pharmacology analysis were obtained from the TCMSP database. HQJXCHP-related therapeutic targets in PF were obtained from the GeneCards, OMIM, Therapeutic Targets and PharmGkb databases. Therapeutic target-related signaling pathways were predicted via GO and KEGG pathway enrichment analyses. The targets of HQJXCHO were further validated in a PDS-induced PF mouse model in vivo and PMCs MMT model in vitro. RESULTS A total of 23 bioactive compounds of HQJXCHP related 188 target genes were retrieved. The HQJXCHP compound-target and PF-related target networks identified 131 common target genes. Subsequent protein-protein interaction (PPI) network analysis results disclosed Akt1, TP53, TNF, VEGFA and CASP3 as the top five key targets of HQJXCHP. Further molecular docking data revealed strong affinity of the two key compounds of HQJXCHP, quercetin and kaempferol, for these key targets. GO and KEGG pathway enrichment analyses further showed that PI3K/Akt, IL-17, TNF and TLR pathways contribute to the therapeutic effects of HQJXCHP on PF. An in vivo PDS-induced PF mouse model and in vitro PMCs mesothelial-to-mesenchymal transition (MMT) model with or without HQJXCHP intervention were used to confirm the effects and mechanisms of action of HQJXCHP. Western blot and qRT-PCR results showed that HQ, JXC and HQJXCHP reduced PDS-induced inflammatory cell aggregation and peritoneal thickening through suppressing the MMT process, among which HQJXCHP exerted the greatest therapeutic effect. Moreover, HQJXCHP inhibited activation of the PI3K/Akt, IL-17, TNF and TLR signaling pathways induced by PDS. CONCLUSIONS This is the first study to employ network pharmacology and molecular docking analyses to predict the targets of HQJXCHP with therapeutic effects on PDS-related PF. Data from in vivo and in vitro validation experiments collectively showed that HQJXCHP delays the PF process through inhibiting PI3K/Akt, IL-17, TNF and TLR signaling pathways. Overall, our findings highlight the successful application of network pharmacology theory to provide a scientific basis for clinical utility of HQJXCHP against PF.
Collapse
Affiliation(s)
- Huibo Dai
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Shan
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Manshu Yu
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Funing Wang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziren Zhou
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinyi Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyan Huang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meixiao Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
18
|
Tao M, Shi Y, Chen H, Li J, Wang Y, Ma X, Du L, Wang Y, Yang X, Hu Y, Zhou X, Zhong Q, Yan D, Qiu A, Zhuang S, Liu N. The disruptor of telomeric silencing 1-like (DOT1L) promotes peritoneal fibrosis through the upregulation and activation of protein tyrosine kinases. MOLECULAR BIOMEDICINE 2024; 5:3. [PMID: 38172378 PMCID: PMC10764708 DOI: 10.1186/s43556-023-00161-z] [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: 08/12/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
The disruptor of telomeric silencing 1-like (DOT1L), a specific histone methyltransferase that catalyzed methylation of histone H3 on lysine 79, was associated with the pathogenesis of many diseases, but its role in peritoneal fibrosis remained unexplored. Here, we examined the role of DOT1L in the expression and activation of protein tyrosine kinases and development of peritoneal fibrosis. We found that a significant rise of DOT1L expression in the fibrotic peritoneum tissues from long-term PD patients and mice. Inhibition of DOT1L significantly attenuated the profibrotic phenotypic differentiation of mesothelial cells and macrophages, and alleviated peritoneal fibrosis. Mechanistically, RNA sequencing and proteomic analysis indicated that DOT1L was mainly involved in the processes of protein tyrosine kinase binding and extracellular matrix structural constituent in the peritoneum. Chromatin immunoprecipitation (ChIP) showed that intranuclear DOT1L guided H3K79me2 to upregulate EGFR in mesothelial cells and JAK3 in macrophages. Immunoprecipitation and immunofluorescence showed that extranuclear DOT1L could interact with EGFR and JAK3, and maintain the activated signaling pathways. In summary, DOT1L promoted the expression and activation of tyrosine kinases (EGFR in mesothelial cells and JAK3 in macrophages), promoting cells differentiate into profibrotic phenotype and thus peritoneal fibrosis. We provide the novel mechanism of dialysis-related peritoneal fibrosis (PF) and the new targets for clinical drug development. DOT1L inhibitor had the PF therapeutic potential.
Collapse
Affiliation(s)
- Min Tao
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yingfeng Shi
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Hui Chen
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Jinqing Li
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yi Wang
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Xiaoyan Ma
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Lin Du
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yishu Wang
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Xinyu Yang
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yan Hu
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Xun Zhou
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Qin Zhong
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Danying Yan
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Andong Qiu
- Shanghai Key Laboratory of Maternal and Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Na Liu
- Department of Nephrology, Pudong New District, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| |
Collapse
|
19
|
Mutsaers SE, Krymskaya VP. Pleural Fibrosis: Now That's What mTORC(ing) About. Am J Respir Cell Mol Biol 2024; 70:8-10. [PMID: 37788451 PMCID: PMC10768835 DOI: 10.1165/rcmb.2023-0327ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/03/2023] [Indexed: 10/05/2023] Open
Affiliation(s)
- Steven E Mutsaers
- Institute for Respiratory Health and School of Biomedical Sciences The University of Western Australia Nedlands, Western Australia, Australia
| | - Vera P Krymskaya
- Department of Medicine, Lung Biology Institute Perelman School of Medicine at the University of Pennsylvania Philadelphia, Pennsylvania
| |
Collapse
|
20
|
Kargozar S, Gorgani S, Nazarnezhad S, Wang AZ. Biocompatible Nanocomposites for Postoperative Adhesion: A State-of-the-Art Review. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 14:4. [PMID: 38202459 PMCID: PMC10780749 DOI: 10.3390/nano14010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024]
Abstract
To reduce and prevent postsurgical adhesions, a variety of scientific approaches have been suggested and applied. This includes the use of advanced therapies like tissue-engineered (TE) biomaterials and scaffolds. Currently, biocompatible antiadhesive constructs play a pivotal role in managing postoperative adhesions and several biopolymer-based products, namely hyaluronic acid (HA) and polyethylene glycol (PEG), are available on the market in different forms (e.g., sprays, hydrogels). TE polymeric constructs are usually associated with critical limitations like poor biocompatibility and mechanical properties. Hence, biocompatible nanocomposites have emerged as an advanced therapy for postoperative adhesion treatment, with hydrogels and electrospun nanofibers among the most utilized antiadhesive nanocomposites for in vitro and in vivo experiments. Recent studies have revealed that nanocomposites can be engineered to generate smart three-dimensional (3D) scaffolds that can respond to different stimuli, such as pH changes. Additionally, nanocomposites can act as multifunctional materials for the prevention of adhesions and bacterial infections, as well as tissue healing acceleration. Still, more research is needed to reveal the clinical potential of nanocomposite constructs and the possible success of nanocomposite-based products in the biomedical market.
Collapse
Affiliation(s)
- Saeid Kargozar
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Sara Gorgani
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran; (S.G.); (S.N.)
| | - Simin Nazarnezhad
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran; (S.G.); (S.N.)
| | - Andrew Z. Wang
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| |
Collapse
|
21
|
Steitz AM, Schröder C, Knuth I, Keber CU, Sommerfeld L, Finkernagel F, Jansen JM, Wagner U, Müller-Brüsselbach S, Worzfeld T, Huber M, Beutgen VM, Graumann J, Pogge von Strandmann E, Müller R, Reinartz S. TRAIL-dependent apoptosis of peritoneal mesothelial cells by NK cells promotes ovarian cancer invasion. iScience 2023; 26:108401. [PMID: 38047087 PMCID: PMC10692662 DOI: 10.1016/j.isci.2023.108401] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/04/2023] [Accepted: 11/03/2023] [Indexed: 12/05/2023] Open
Abstract
A crucial requirement for metastasis formation in ovarian high-grade serous carcinoma (HGSC) is the disruption of the protective peritoneal mesothelium. Using co-culture systems of primary human cells, we discovered that tumor-associated NK cells induce TRAIL-dependent apoptosis in mesothelial cells via death receptors DR4 and DR5 upon encounter with activated T cells. Upregulation of TRAIL expression in NK cells concomitant with enhanced cytotoxicity toward mesothelial cells was driven predominantly by T-cell-derived TNFα, as shown by affinity proteomics-based analysis of the T cell secretome in conjunction with functional studies. Consistent with these findings, we detected apoptotic mesothelial cells in the peritoneal fluid of HGSC patients. In contrast to mesothelial cells, HGSC cells express negligible levels of both DR4 and DR5 and are TRAIL resistant, indicating cell-type-selective killing by NK cells. Our data point to a cooperative action of T and NK in breaching the mesothelial barrier in HGSC patients.
Collapse
Affiliation(s)
- Anna Mary Steitz
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Clarissa Schröder
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Isabel Knuth
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Corinna U. Keber
- Institute for Pathology, Philipps University, 35043 Marburg, Germany
| | - Leah Sommerfeld
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Florian Finkernagel
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Julia M. Jansen
- Clinic for Gynecology, Gynecological Oncology, Gynecological Endocrinology, University Hospital (UKGM), 35043 Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology, Gynecological Endocrinology, University Hospital (UKGM), 35043 Marburg, Germany
| | - Sabine Müller-Brüsselbach
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Thomas Worzfeld
- Institute of Pharmacology, Biochemical-Pharmacological Center (BPC), Philipps University, 35043 Marburg, Germany
| | - Magdalena Huber
- Institute of Systems Immunology, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Vanessa M. Beutgen
- Institute of Translational Proteomics, Philipps University, 35043 Marburg, Germany
- Core Facility Translational Proteomics, Philipps University, 35043 Marburg, Germany
| | - Johannes Graumann
- Institute of Translational Proteomics, Philipps University, 35043 Marburg, Germany
- Core Facility Translational Proteomics, Philipps University, 35043 Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology (ZTI), Clinic for Hematology, Oncology and Immunology, Philipps University, 35043 Marburg, Germany
| | - Rolf Müller
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Silke Reinartz
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| |
Collapse
|
22
|
Kaesler N, Cheng M, Nagai J, O’Sullivan J, Peisker F, Bindels EM, Babler A, Moellmann J, Droste P, Franciosa G, Dugourd A, Saez-Rodriguez J, Neuss S, Lehrke M, Boor P, Goettsch C, Olsen JV, Speer T, Lu TS, Lim K, Floege J, Denby L, Costa I, Kramann R. Mapping cardiac remodeling in chronic kidney disease. SCIENCE ADVANCES 2023; 9:eadj4846. [PMID: 38000021 PMCID: PMC10672229 DOI: 10.1126/sciadv.adj4846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023]
Abstract
Patients with advanced chronic kidney disease (CKD) mostly die from sudden cardiac death and recurrent heart failure. The mechanisms of cardiac remodeling are largely unclear. To dissect molecular and cellular mechanisms of cardiac remodeling in CKD in an unbiased fashion, we performed left ventricular single-nuclear RNA sequencing in two mouse models of CKD. Our data showed a hypertrophic response trajectory of cardiomyocytes with stress signaling and metabolic changes driven by soluble uremia-related factors. We mapped fibroblast to myofibroblast differentiation in this process and identified notable changes in the cardiac vasculature, suggesting inflammation and dysfunction. An integrated analysis of cardiac cellular responses to uremic toxins pointed toward endothelin-1 and methylglyoxal being involved in capillary dysfunction and TNFα driving cardiomyocyte hypertrophy in CKD, which was validated in vitro and in vivo. TNFα inhibition in vivo ameliorated the cardiac phenotype in CKD. Thus, interventional approaches directed against uremic toxins, such as TNFα, hold promise to ameliorate cardiac remodeling in CKD.
Collapse
Affiliation(s)
- Nadine Kaesler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Mingbo Cheng
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James Nagai
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James O’Sullivan
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Fabian Peisker
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Eric M. J. Bindels
- Department of Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Anne Babler
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Julia Moellmann
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Patrick Droste
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Giulia Franciosa
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Aurelien Dugourd
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Sabine Neuss
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Thimoteus Speer
- Department of Medicine (Nephrology), Goethe University Frankfurt, Frankfurt, Germany
| | - Tzong-Shi Lu
- Brigham and Women’s Hospital, Renal Division, Boston, MA, USA
| | - Kenneth Lim
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jürgen Floege
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Laura Denby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Ivan Costa
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
23
|
Akasaka H, Naora H. Revisiting the Use of Normal Saline for Peritoneal Washing in Ovarian Cancer. Int J Mol Sci 2023; 24:16449. [PMID: 38003636 PMCID: PMC10671679 DOI: 10.3390/ijms242216449] [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: 10/16/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
The omentum is the predominant site of ovarian cancer metastasis, but it is difficult to remove the omentum in its entirety. There is a critical need for effective approaches that minimize the risk of colonization of preserved omental tissues by occult cancer cells. Normal saline (0.9% sodium chloride) is commonly used to wash the peritoneal cavity during ovarian cancer surgery. The omentum has a prodigious ability to absorb fluid in the peritoneal cavity, but the impact of normal saline on the omentum is poorly understood. In this review article, we discuss why normal saline is not a biocompatible solution, drawing insights from clinical investigations of normal saline in fluid resuscitation and from the cytopathologic evaluation of peritoneal washings. We integrate these insights with the unique biology of the omentum and omental metastasis, highlighting the importance of considering the absorptive ability of the omentum when administering agents into the peritoneal cavity. Furthermore, we describe insights from preclinical studies regarding the mechanisms by which normal saline might render the omentum conducive for colonization by cancer cells. Importantly, we discuss the possibility that the risk of colonization of preserved omental tissues might be minimized by using balanced crystalloid solutions for peritoneal washing.
Collapse
Affiliation(s)
| | - Honami Naora
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
24
|
Levai E, Marinovic I, Bartosova M, Zhang C, Schaefer B, Jenei H, Du Z, Drozdz D, Klaus G, Arbeiter K, Romero P, Schwenger V, Schwab C, Szabo AJ, Zarogiannis SG, Schmitt CP. Human peritoneal tight junction, transporter and channel expression in health and kidney failure, and associated solute transport. Sci Rep 2023; 13:17429. [PMID: 37833387 PMCID: PMC10575882 DOI: 10.1038/s41598-023-44466-z] [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] [Received: 04/30/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023] Open
Abstract
Next to the skin, the peritoneum is the largest human organ, essentially involved in abdominal health and disease states, but information on peritoneal paracellular tight junctions and transcellular channels and transporters relative to peritoneal transmembrane transport is scant. We studied their peritoneal localization and quantity by immunohistochemistry and confocal microscopy in health, in chronic kidney disease (CKD) and on peritoneal dialysis (PD), with the latter allowing for functional characterizations, in a total of 93 individuals (0-75 years). Claudin-1 to -5, and -15, zonula occludens-1, occludin and tricellulin, SGLT1, PiT1/SLC20A1 and ENaC were consistently detected in mesothelial and arteriolar endothelial cells, with age dependent differences for mesothelial claudin-1 and arteriolar claudin-2/3. In CKD mesothelial claudin-1 and arteriolar claudin-2 and -3 were more abundant. Peritonea from PD patients exhibited increased mesothelial and arteriolar claudin-1 and mesothelial claudin-2 abundance and reduced mesothelial and arteriolar claudin-3 and arteriolar ENaC. Transperitoneal creatinine and glucose transport correlated with pore forming arteriolar claudin-2 and mesothelial claudin-4/-15, and creatinine transport with mesothelial sodium/phosphate cotransporter PiT1/SLC20A1. In multivariable analysis, claudin-2 independently predicted the peritoneal transport rates. In conclusion, tight junction, transcellular transporter and channel proteins are consistently expressed in peritoneal mesothelial and endothelial cells with minor variations across age groups, specific modifications by CKD and PD and distinct associations with transperitoneal creatinine and glucose transport rates. The latter deserve experimental studies to demonstrate mechanistic links.Clinical Trial registration: The study was performed according to the Declaration of Helsinki and is registered at www.clinicaltrials.gov (NCT01893710).
Collapse
Affiliation(s)
- Eszter Levai
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
- HUNREN SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Iva Marinovic
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Conghui Zhang
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Betti Schaefer
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Hanna Jenei
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Zhiwei Du
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Dorota Drozdz
- Jagiellonian University Medical College, Krakow, Poland
| | | | - Klaus Arbeiter
- Department of Pediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria
| | - Philipp Romero
- Division of Pediatric Surgery, Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Vedat Schwenger
- Department of Nephrology, Klinikum der Landeshauptstadt Stuttgart, Stuttgart, Germany
| | | | - Attila J Szabo
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
- HUNREN SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Sotirios G Zarogiannis
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Department of Physiology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Claus Peter Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.
| |
Collapse
|
25
|
Koukorava C, Ward K, Ahmed K, Almaghrabi S, Dauleh S, Pereira SM, Taylor A, Haddrick M, Cross MJ, Wilm B. Mesothelial Cells Exhibit Characteristics of Perivascular Cells in an In Vitro Angiogenesis Assay. Cells 2023; 12:2436. [PMID: 37887280 PMCID: PMC10605208 DOI: 10.3390/cells12202436] [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: 08/11/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/28/2023] Open
Abstract
Mesothelial cells have been shown to have remarkable plasticity towards mesenchymal cell types during development and in disease situations. Here, we have characterized the potential of mesothelial cells to undergo changes toward perivascular cells using an in vitro angiogenesis assay. We demonstrate that GFP-labeled mesothelial cells (GFP-MCs) aligned closely and specifically with endothelial networks formed when human dermal microvascular endothelial cells (HDMECs) were cultured in the presence of VEGF-A165 on normal human dermal fibroblasts (NHDFs) for a 7-day period. The co-culture with GFP-MCs had a positive effect on branch point formation indicating that the cells supported endothelial tube formation. We interrogated the molecular response of the GFP-MCs to the angiogenic co-culture by qRT-PCR and found that the pericyte marker Ng2 was upregulated when the cells were co-cultured with HDMECs on NHDFs, indicating a change towards a perivascular phenotype. When GFP-MCs were cultured on the NHDF feeder layer, they upregulated the epithelial-mesenchymal transition marker Zeb1 and lost their circularity while increasing their size, indicating a change to a more migratory cell type. We analyzed the pericyte-like behavior of the GFP-MCs in a 3D cardiac microtissue (spheroid) with cardiomyocytes, cardiac fibroblasts and cardiac endothelial cells where the mesothelial cells showed alignment with the endothelial cells. These results indicate that mesothelial cells have the potential to adopt a perivascular phenotype and associate with endothelial cells to potentially support angiogenesis.
Collapse
Affiliation(s)
- Chrysa Koukorava
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Kelly Ward
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Katie Ahmed
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Shrouq Almaghrabi
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Sumaya Dauleh
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Sofia M. Pereira
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Arthur Taylor
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
- Medicines Discovery Catapult, Alderley Park, Macclesfield SK10 4ZF, UK
| | - Malcolm Haddrick
- Medicines Discovery Catapult, Alderley Park, Macclesfield SK10 4ZF, UK
| | - Michael J. Cross
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Bettina Wilm
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
- Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3GE, UK
| |
Collapse
|
26
|
Huang W, Xia D, Bi W, Lai X, Yu B, Chen W. Advances in stem cell therapy for peritoneal fibrosis: from mechanisms to therapeutics. Stem Cell Res Ther 2023; 14:293. [PMID: 37817212 PMCID: PMC10566108 DOI: 10.1186/s13287-023-03520-3] [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] [Received: 12/25/2022] [Accepted: 09/26/2023] [Indexed: 10/12/2023] Open
Abstract
Peritoneal fibrosis (PF) is a pathophysiological condition caused by a variety of pathogenic factors. The most important features of PF are mesothelial-mesenchymal transition and accumulation of activated (myo-)fibroblasts, which hinder effective treatment; thus, it is critical to identify other practical approaches. Recently, stem cell (SC) therapy has been indicated to be a potential strategy for this disease. Increasing evidence suggests that many kinds of SCs alleviate PF mainly by differentiating into mesothelial cells; secreting cytokines and extracellular vesicles; or modulating immune cells, particularly macrophages. However, there are relatively few articles summarizing research in this direction. In this review, we summarize the risk factors for PF and discuss the therapeutic roles of SCs from different sources. In addition, we outline effective approaches and potential mechanisms of SC therapy for PF. We hope that our review of articles in this area will provide further inspiration for research on the use of SCs in PF treatment.
Collapse
Affiliation(s)
- Weiyan Huang
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Demeng Xia
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wendi Bi
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xueli Lai
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Bing Yu
- Department of Cell Biology, Center for Stem Cell and Medicine, Naval Medical University (Second Military Medical University), Shanghai, China.
| | - Wei Chen
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
27
|
Whitfield HJ, Berthelet J, Mangiola S, Bell C, Anderson RL, Pal B, Yeo B, Papenfuss AT, Merino D, Davis MJ. Single-cell RNA sequencing captures patient-level heterogeneity and associated molecular phenotypes in breast cancer pleural effusions. Clin Transl Med 2023; 13:e1356. [PMID: 37691350 PMCID: PMC10493486 DOI: 10.1002/ctm2.1356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND Malignant pleural effusions (MPEs) are a common complication of advanced cancers, particularly those adjacent to the pleura, such as lung and breast cancer. The pathophysiology of MPE formation remains poorly understood, and although MPEs are routinely used for the diagnosis of breast cancer patients, their composition and biology are poorly understood. It is difficult to distinguish invading malignant cells from resident mesothelial cells and to identify the directionality of interactions between these populations in the pleura. There is a need to characterize the phenotypic diversity of breast cancer cell populations in the pleural microenvironment, and investigate how this varies across patients. METHODS Here, we used single-cell RNA-sequencing to study the heterogeneity of 10 MPEs from seven metastatic breast cancer patients, including three Miltenyi-enriched samples using a negative selection approach. This dataset of almost 65 000 cells was analysed using integrative approaches to compare heterogeneous cell populations and phenotypes. RESULTS We identified substantial inter-patient heterogeneity in the composition of cell types (including malignant, mesothelial and immune cell populations), in expression of subtype-specific gene signatures and in copy number aberration patterns, that captured variability across breast cancer cell populations. Within individual MPEs, we distinguished mesothelial cell populations from malignant cells using key markers, the presence of breast cancer subtype expression patterns and copy number aberration patterns. We also identified pleural mesothelial cells expressing a cancer-associated fibroblast-like transcriptomic program that may support cancer growth. CONCLUSIONS Our dataset presents the first unbiased assessment of breast cancer-associated MPEs at a single cell resolution, providing the community with a valuable resource for the study of MPEs. Our work highlights the molecular and cellular diversity captured in MPEs and motivates the potential use of these clinically relevant biopsies in the development of targeted therapeutics for patients with advanced breast cancer.
Collapse
Affiliation(s)
- Holly J. Whitfield
- Department of Medical Biology, The Faculty of MedicineDentistry and Health Science, The University of MelbourneCarltonVictoriaAustralia
- Bioinformatics DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| | - Jean Berthelet
- Olivia Newton‐John Cancer Research InstituteHeidelbergVictoriaAustralia
- School of Cancer MedicineLa Trobe UniversityBundooraVictoriaAustralia
| | - Stefano Mangiola
- Department of Medical Biology, The Faculty of MedicineDentistry and Health Science, The University of MelbourneCarltonVictoriaAustralia
- Bioinformatics DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| | - Caroline Bell
- Olivia Newton‐John Cancer Research InstituteHeidelbergVictoriaAustralia
- School of Cancer MedicineLa Trobe UniversityBundooraVictoriaAustralia
| | - Robin L. Anderson
- Olivia Newton‐John Cancer Research InstituteHeidelbergVictoriaAustralia
- School of Cancer MedicineLa Trobe UniversityBundooraVictoriaAustralia
- Peter MacCallum Cancer CentreParkvilleVictoriaAustralia
- Department of Clinical Pathology, Faculty of MedicineDentistry and Health Science, The University of MelbourneCarltonVictoriaAustralia
| | - Bhupinder Pal
- Olivia Newton‐John Cancer Research InstituteHeidelbergVictoriaAustralia
- School of Cancer MedicineLa Trobe UniversityBundooraVictoriaAustralia
| | - Belinda Yeo
- Olivia Newton‐John Cancer Research InstituteHeidelbergVictoriaAustralia
- School of Cancer MedicineLa Trobe UniversityBundooraVictoriaAustralia
- Austin HealthHeidelbergVictoriaAustralia
| | - Anthony T. Papenfuss
- Department of Medical Biology, The Faculty of MedicineDentistry and Health Science, The University of MelbourneCarltonVictoriaAustralia
- Bioinformatics DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Clinical Pathology, Faculty of MedicineDentistry and Health Science, The University of MelbourneCarltonVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneCarltonVictoriaAustralia
| | - Delphine Merino
- Department of Medical Biology, The Faculty of MedicineDentistry and Health Science, The University of MelbourneCarltonVictoriaAustralia
- Olivia Newton‐John Cancer Research InstituteHeidelbergVictoriaAustralia
- School of Cancer MedicineLa Trobe UniversityBundooraVictoriaAustralia
- Immunology DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| | - Melissa J. Davis
- Department of Medical Biology, The Faculty of MedicineDentistry and Health Science, The University of MelbourneCarltonVictoriaAustralia
- Bioinformatics DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Clinical Pathology, Faculty of MedicineDentistry and Health Science, The University of MelbourneCarltonVictoriaAustralia
- The University of Queensland Diamantina InstituteThe University of QueenslandBrisbaneQueenslandAustralia
- The South Australian Immunogenomics Cancer InstituteThe University of AdelaideAdelaideSouth AustraliaAustralia
| |
Collapse
|
28
|
Kreienbühl J, Changkhong S, Orlowski V, Kirschner MB, Opitz I, Meerang M. Cullin 4B Ubiquitin Ligase Is Important for Cell Survival and Regulates TGF-β1 Expression in Pleural Mesothelioma. Int J Mol Sci 2023; 24:13410. [PMID: 37686215 PMCID: PMC10487616 DOI: 10.3390/ijms241713410] [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] [Received: 08/07/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
We previously demonstrated that cullin 4B (CUL4B) upregulation was associated with worse outcomes of pleural mesothelioma (PM) patients, while the overexpression of its paralog CUL4A was not associated with clinical outcomes. Here, we aimed to identify the distinct roles of CUL4B and CUL4A in PM using an siRNA approach in PM cell lines (ACC Meso-1 and Mero82) and primary culture. The knockdown of CUL4B and CUL4A resulted in significantly reduced colony formation, increased cell death, and delayed cell proliferation. Furthermore, similar to the effect of CUL4A knockdown, downregulation of CUL4B led to reduced expression of Hippo pathway genes including YAP1, CTGF, and survivin. Interestingly, CUL4B and not CUL4A knockdown reduced TGF-β1 and MMP2 expression, suggesting a unique association of CUL4B with this pathway. However, the treatment of PM cells with exogenous TGF-β1 following CUL4B knockdown did not rescue PM cell growth. We further analyzed ACC Meso-1 xenograft tumor tissues treated with the cullin inhibitor, pevonedistat, which targets protein neddylation, and observed the downregulation of human TGF-β1 and MMP2. In summary, our data suggest that CUL4B overexpression is important for tumor cell growth and survival and may drive PM aggressiveness via the regulation of TGF-β1 expression and, furthermore, reveal a new mechanism of action of pevonedistat.
Collapse
Affiliation(s)
| | | | | | | | | | - Mayura Meerang
- Department of Thoracic Surgery, University Hospital Zürich, 8091 Zürich, Switzerland (V.O.); (M.B.K.); (I.O.)
| |
Collapse
|
29
|
Meng M, Lu M, Feng J, Zhou X, Meng P, Chen L, Zou X, Liu X, Liu L, Gao X, Zhang Y. Exosomal PPARγ derived from macrophages suppresses LPS-induced peritonitis by negative regulation of CD14/TLR4 axis. Inflamm Res 2023; 72:1567-1581. [PMID: 37438583 DOI: 10.1007/s00011-023-01765-5] [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] [Received: 03/06/2023] [Revised: 06/13/2023] [Accepted: 06/30/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Intercellular communication between macrophages and peritoneal mesothelial cells (PMCs) has been suggested as a key factor regulating peritonitis development. Here, we explored whether PPARγ (peroxisome proliferator-activated receptor gamma) can be packaged into macrophage exosomes to mediate intercellular communication and regulate peritonitis. METHODS Macrophage exosomes were isolated by ultracentrifugation and identified by nanoparticle tracking analysis and transmission electron microscopy. Proteomic analysis of macrophage-derived exosomes was performed using mass spectrometry. Co-culture models of supernatants or exosomes with PMCs, as well as a mouse peritonitis model induced by lipopolysaccharide (LPS), were employed. RESULTS In this study, using stable Raw264.7 cells overexpressing GFP-FLAG-PPARγ (OE-PPARγ), we found that PPARγ inhibited LPS-induced inflammatory responses in Raw264.7 cells and that PPARγ was incorporated into macrophage exosomes during this process. Overexpression of PPARγ mainly regulated the secretion of differentially expressed exosomal proteins involved in the biological processes of protein transport, lipid metabolic process, cell cycle, apoptotic process, DNA damage stimulus, as well as the KEGG pathway of salmonella infection. Using co-culture models and mouse peritonitis model, we showed that exosomes from Raw264.7 cells overexpressing PPARγ inhibited LPS-induced inflammation in co-cultured human PMCs and in mice through downregulating CD14 and TLR4, two key regulators of the salmonella infection pathway. Pretreatment of the PPARγ inhibitor GW9662 abolished the anti-inflammatory effect of exosomes from Raw264.7 OE-PPARγ cells on human PMCs. CONCLUSIONS These results suggested that overexpression of PPARγ largely altered the proteomic profile of macrophage exosomes and that exosomal PPARγ from macrophages acted as a regulator of intercellular communication to suppress LPS-induced inflammatory responses in vitro and in vivo via negatively regulating the CD14/TLR4 axis.
Collapse
Affiliation(s)
- Meng Meng
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Meizhi Lu
- Department of Nephrology, Affiliated Huadu Hospital, Southern Medical University (People's Hospital of Huadu District), Guangzhou, China
| | - Junxia Feng
- Department of Nephrology, Affiliated Huadu Hospital, Southern Medical University (People's Hospital of Huadu District), Guangzhou, China
| | - Xiaoying Zhou
- Department of Nephrology, Affiliated Huadu Hospital, Southern Medical University (People's Hospital of Huadu District), Guangzhou, China
| | - Ping Meng
- Department of Nephrology, Affiliated Huadu Hospital, Southern Medical University (People's Hospital of Huadu District), Guangzhou, China
| | - Luxuan Chen
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xunliang Zou
- Department of Nephrology, The Fifth Affiliated (Zhuhai) Hospital of Zunyi Medical University, Zhuhai, 519100, China
| | - Xiaohui Liu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Langxia Liu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Xuejuan Gao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Yunfang Zhang
- Department of Nephrology, Affiliated Huadu Hospital, Southern Medical University (People's Hospital of Huadu District), Guangzhou, China.
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
30
|
Chauvin M, Meinsohn MC, Dasari S, May P, Iyer S, Nguyen NMP, Oliva E, Lucchini Z, Nagykery N, Kashiwagi A, Mishra R, Maser R, Wells J, Bult CJ, Mitra AK, Donahoe PK, Pépin D. Cancer-associated mesothelial cells are regulated by the anti-Müllerian hormone axis. Cell Rep 2023; 42:112730. [PMID: 37453057 DOI: 10.1016/j.celrep.2023.112730] [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: 11/23/2022] [Revised: 04/27/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Cancer-associated mesothelial cells (CAMCs) in the tumor microenvironment are thought to promote growth and immune evasion. We find that, in mouse and human ovarian tumors, cancer cells express anti-Müllerian hormone (AMH) while CAMCs express its receptor AMHR2, suggesting a paracrine axis. Factors secreted by cancer cells induce AMHR2 expression during their reprogramming into CAMCs in mouse and human in vitro models. Overexpression of AMHR2 in the Met5a mesothelial cell line is sufficient to induce expression of immunosuppressive cytokines and growth factors that stimulate ovarian cancer cell growth in an AMH-dependent way. Finally, syngeneic cancer cells implanted in transgenic mice with Amhr2-/- CAMCs grow significantly slower than in wild-type hosts. The cytokine profile of Amhr2-/- tumor-bearing mice is altered and their tumors express less immune checkpoint markers programmed-cell-death 1 (PD1) and cytotoxic T lymphocyte-associated protein 4 (CTLA4). Taken together, these data suggest that the AMH/AMHR2 axis plays a critical role in regulating the pro-tumoral function of CAMCs in ovarian cancer.
Collapse
Affiliation(s)
- M Chauvin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - M-C Meinsohn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - S Dasari
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
| | - P May
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - S Iyer
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - N M P Nguyen
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - E Oliva
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Z Lucchini
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - N Nagykery
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - A Kashiwagi
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - R Mishra
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - R Maser
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA
| | - J Wells
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA
| | - C J Bult
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA
| | - A K Mitra
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - D Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA; Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA.
| |
Collapse
|
31
|
Kiniger C, Janssen JN, Lederer KA, Lipnik K, Doulidis PG. Sclerosing encapsulating peritonitis in cats: a two-case report and literature review. JFMS Open Rep 2023; 9:20551169231178447. [PMID: 37434990 PMCID: PMC10331345 DOI: 10.1177/20551169231178447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023] Open
Abstract
Case series summary Two castrated male domestic shorthair cats (aged 8 months [case 1] and 13 years [case 2]) were presented at the Small Animal Clinic of the Veterinary Medicine University of Vienna, Austria, both with acute vomiting and distended abdomen, as well as a history of chronic apathy, recurrent vomiting and diarrhoea. Both cats underwent invasive diagnostic procedures approximately 1 month before the diagnosis of sclerosing encapsulating peritonitis (SEP), namely an exploratory laparotomy and a bronchoscopy, respectively. Abdominal ultrasound revealed severely corrugated intestinal loops and, in case 2, the presence of peritoneal effusion. A thick and diffuse fibrous capsule around the intestine was detected and removed surgically, and biopsies were taken from the affected organs confirming the SEP. Case 1 recovered well, was discharged some days after surgery and was clinically unremarkable for the next 2 years. Case 2 showed unsatisfactory improvement directly after surgery and was euthanased a few days later, as the owner declined any further therapy. Relevance and novel information SEP is a very rare condition of unclear origins in cats. Here we describe the clinical and diagnostic imaging features, surgical treatment, and outcome of SEP in two cats. The results indicate that prompt diagnosis and appropriate interventions may improve the outcome.
Collapse
Affiliation(s)
- Caterina Kiniger
- Clinical Unit of Small Animal Internal Medicine, Department for Small Animals and Horses, University of Veterinary Medicine, Vienna, Austria
| | - Janina N Janssen
- Clinical Unit of Small Animal Surgery, Department for Small Animals and Horses, University of Veterinary Medicine, Vienna, Austria
| | - Kristina A Lederer
- Diagnostic Imaging, Department for Small Animals and Horses, University of Veterinary Medicine, Vienna, Austria
| | - Karoline Lipnik
- Institute of Pathology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Pavlos G Doulidis
- Clinical Unit of Small Animal Internal Medicine, Department for Small Animals and Horses, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
32
|
Peeney D, Fan Y, Gurung S, Lazaroff C, Ratnayake S, Warner A, Karim B, Meerzaman D, Stetler-Stevenson WG. Whole organism profiling of the Timp gene family. Matrix Biol Plus 2023; 18:100132. [PMID: 37095886 PMCID: PMC10121480 DOI: 10.1016/j.mbplus.2023.100132] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023] Open
Abstract
Tissue inhibitor of metalloproteinases (TIMPs/Timps) are an endogenous family of widely expressed matrisome-associated proteins that were initially identified as inhibitors of matrix metalloproteinase activity (Metzincin family proteases). Consequently, TIMPs are often considered simply as protease inhibitors by many investigators. However, an evolving list of new metalloproteinase-independent functions for TIMP family members suggests that this concept is outdated. These novel TIMP functions include direct agonism/antagonism of multiple transmembrane receptors, as well as functional interactions with matrisome targets. While the family was fully identified over two decades ago, there has yet to be an in-depth study describing the expression of TIMPs in normal tissues of adult mammals. An understanding of the tissues and cell-types that express TIMPs 1 through 4, in both normal and disease states are important to contextualize the growing functional capabilities of TIMP proteins, which are often dismissed as non-canonical. Using publicly available single cell RNA sequencing data from the Tabula Muris Consortium, we analyzed approximately 100,000 murine cells across eighteen tissues from non-diseased organs, representing seventy-three annotated cell types, to define the diversity in Timp gene expression across healthy tissues. We describe the unique expression profiles across tissues and organ-specific cell types that all four Timp genes display. Within annotated cell-types, we identify clear and discrete cluster-specific patterns of Timp expression, particularly in cells of stromal and endothelial origins. RNA in-situ hybridization across four organs expands on the scRNA sequencing analysis, revealing novel compartments associated with individual Timp expression. These analyses emphasize a need for specific studies investigating the functional significance of Timp expression in the identified tissues and cell sub-types. This understanding of the tissues, specific cell types and microenvironment conditions in which Timp genes are expressed adds important physiological context to the growing array of novel functions for TIMP proteins.
Collapse
Affiliation(s)
- David Peeney
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Yu Fan
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institute of Health, Rockville, MD, USA
| | - Sadeechya Gurung
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Carolyn Lazaroff
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Shashikala Ratnayake
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institute of Health, Rockville, MD, USA
| | - Andrew Warner
- Molecular Histopathology Laboratory, Frederick National Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Daoud Meerzaman
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institute of Health, Rockville, MD, USA
| | - William G. Stetler-Stevenson
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| |
Collapse
|
33
|
Kauschke V, Philipp-Gehlhaar M, Schneider J. Expression of microRNAs in leukocytes and serum of asbestosis patients. Eur J Med Res 2023; 28:175. [PMID: 37189132 PMCID: PMC10184414 DOI: 10.1186/s40001-023-01129-z] [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: 08/04/2022] [Accepted: 04/26/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Although asbestos use is banned in many countries, long latency of asbestos-related diseases like pleural plaques or asbestosis mean it is still a public health issue. People suffering from these diseases have a higher risk of developing mesothelioma or lung cancer, which can progress quickly and aggressively. MicroRNAs were suggested as potential biomarkers in several diseases. However, in asbestosis, blood microRNAs are less explored. Since miR-32-5p, miR-143-3p, miR-145-5p, miR-146b-5p, miR-204-5p and miR-451a are involved in fibrotic processes and in cancer, expression of these microRNAs was analyzed in leukocytes and serum of asbestosis patients. METHODS MicroRNA expression was analyzed in leukocytes and serum of 36 patients (26 affected by pleural plaques and 10 by asbestosis) and 15 healthy controls by real-time RT-PCR. Additionally, data analyses were performed regarding disease severity based on ILO classification. RESULTS MicroRNA miR-146b-5p was significantly down-regulated in leukocytes of patients suffering from pleural plaques with a large effect indicated by η2p = 0.150 and Cohen's f = 0.42, a value of difference of 0.725 and a 95% confidence interval of 0.070-1.381. In patients suffering from asbestosis miR-146b-5p was not significantly regulated. However, data analyses considering disease severity only, revealed that miR-146b-5p was significantly down-regulated in leukocytes of mildly diseased patients compared to controls with a large effect indicated by η2p = 0.178 and Cohen's f = 0.465, a value of difference of 0.848 and a 95% confidence interval of 0.097-1.599. Receiver operating characteristic (ROC) curve and an area under the ROC curve value of 0.757 for miR-146b-5p indicated acceptable discrimination ability between patients suffering from pleural plaques and healthy controls. Less microRNAs were detectable in serum than in leukocytes, showing no significant expression differences in all participants of this study. Moreover, miR-145-5p was regulated significantly differently in leukocytes and serum. An R2 value of 0.004 for miR-145-5p indicated no correlation in microRNA expression between leukocytes and serum. CONCLUSION Leukocytes seem more suitable than serum for microRNA analyses regarding disease and potentially cancer risk assessment of patients suffering from asbestos-related pleural plaques or asbestosis. Long-term studies may reveal whether down-regulation of miR-146b-5p in leukocytes might be an early indicator for an increased cancer risk.
Collapse
Affiliation(s)
- Vivien Kauschke
- Institute and Outpatient Clinic of Occupational and Social Medicine, University Hospital of Giessen and Marburg, Aulweg 129, 35392, Giessen, Germany.
| | - Monika Philipp-Gehlhaar
- Institute and Outpatient Clinic of Occupational and Social Medicine, University Hospital of Giessen and Marburg, Aulweg 129, 35392, Giessen, Germany
| | - Joachim Schneider
- Institute and Outpatient Clinic of Occupational and Social Medicine, University Hospital of Giessen and Marburg, Aulweg 129, 35392, Giessen, Germany
| |
Collapse
|
34
|
McElhinney K, Irnaten M, O’Brien C. p53 and Myofibroblast Apoptosis in Organ Fibrosis. Int J Mol Sci 2023; 24:ijms24076737. [PMID: 37047710 PMCID: PMC10095465 DOI: 10.3390/ijms24076737] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023] Open
Abstract
Organ fibrosis represents a dysregulated, maladaptive wound repair response that results in progressive disruption of normal tissue architecture leading to detrimental deterioration in physiological function, and significant morbidity/mortality. Fibrosis is thought to contribute to nearly 50% of all deaths in the Western world with current treatment modalities effective in slowing disease progression but not effective in restoring organ function or reversing fibrotic changes. When physiological wound repair is complete, myofibroblasts are programmed to undergo cell death and self-clearance, however, in fibrosis there is a characteristic absence of myofibroblast apoptosis. It has been shown that in fibrosis, myofibroblasts adopt an apoptotic-resistant, highly proliferative phenotype leading to persistent myofibroblast activation and perpetuation of the fibrotic disease process. Recently, this pathological adaptation has been linked to dysregulated expression of tumour suppressor gene p53. In this review, we discuss p53 dysregulation and apoptotic failure in myofibroblasts and demonstrate its consistent link to fibrotic disease development in all types of organ fibrosis. An enhanced understanding of the role of p53 dysregulation and myofibroblast apoptosis may aid in future novel therapeutic and/or diagnostic strategies in organ fibrosis.
Collapse
Affiliation(s)
- Kealan McElhinney
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| | - Mustapha Irnaten
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| | - Colm O’Brien
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| |
Collapse
|
35
|
Trionfetti F, Marchant V, González-Mateo GT, Kawka E, Márquez-Expósito L, Ortiz A, López-Cabrera M, Ruiz-Ortega M, Strippoli R. Novel Aspects of the Immune Response Involved in the Peritoneal Damage in Chronic Kidney Disease Patients under Dialysis. Int J Mol Sci 2023; 24:5763. [PMID: 36982834 PMCID: PMC10059714 DOI: 10.3390/ijms24065763] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Chronic kidney disease (CKD) incidence is growing worldwide, with a significant percentage of CKD patients reaching end-stage renal disease (ESRD) and requiring kidney replacement therapies (KRT). Peritoneal dialysis (PD) is a convenient KRT presenting benefices as home therapy. In PD patients, the peritoneum is chronically exposed to PD fluids containing supraphysiologic concentrations of glucose or other osmotic agents, leading to the activation of cellular and molecular processes of damage, including inflammation and fibrosis. Importantly, peritonitis episodes enhance peritoneum inflammation status and accelerate peritoneal injury. Here, we review the role of immune cells in the damage of the peritoneal membrane (PM) by repeated exposure to PD fluids during KRT as well as by bacterial or viral infections. We also discuss the anti-inflammatory properties of current clinical treatments of CKD patients in KRT and their potential effect on preserving PM integrity. Finally, given the current importance of coronavirus disease 2019 (COVID-19) disease, we also analyze here the implications of this disease in CKD and KRT.
Collapse
Affiliation(s)
- Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| | - Vanessa Marchant
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
- REDINREN/RICORS2040, 28029 Madrid, Spain
| | - Guadalupe T. González-Mateo
- Cell-Cell Communication & Inflammation Unit, Centre for Molecular Biology “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain
- Premium Research, S.L., 19005 Guadalajara, Spain
| | - Edyta Kawka
- Department of Pathophysiology, Poznan University of Medical Sciences, 10 Fredry St., 61-701 Poznan, Poland
| | - Laura Márquez-Expósito
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
- REDINREN/RICORS2040, 28029 Madrid, Spain
| | - Alberto Ortiz
- IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Manuel López-Cabrera
- Cell-Cell Communication & Inflammation Unit, Centre for Molecular Biology “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
- REDINREN/RICORS2040, 28029 Madrid, Spain
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| |
Collapse
|
36
|
De Lazari MGT, Viana CTR, Pereira LX, Orellano LAA, Ulrich H, Andrade SP, Campos PP. Sodium butyrate attenuates peritoneal fibroproliferative process in mice. Exp Physiol 2023; 108:146-157. [PMID: 36459573 PMCID: PMC10103766 DOI: 10.1113/ep090559] [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: 05/12/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022]
Abstract
NEW FINDINGS What is the central question of this study? Peritoneal injury can result in a persistent fibroproliferative process in the abdominal cavity, causing pain and loss of function of internal organs. This study aimed to demonstrate the use of sodium butyrate (NaBu) as a potential agent to attenuate peritoneal fibrosis induced by a synthetic matrix. What is the main finding and its importance? Our findings provide the first evidence that NaBu attenuates the inflammatory, angiogenesis and fibrogenesis axes involved in the formation of peritoneal fibrovascular tissue, indicating the potential of this compound to ameliorate peritoneal fibrosis. ABSTRACT The aim of this study was to identify the bio-efficacy of sodium butyrate (NaBu) on preventing the development of peritoneal fibrovascular tissue induced by implantation of a synthetic matrix in the abdominal cavity. Polyether-polyurethane sponge discs were implanted in the peritoneal cavity of mice, which were treated daily with oral administration of NaBu (100 mg/kg). Control animals received water (100 μl). After 7 days, the implants were removed for assessment of inflammatory, angiogenic and fibrogenic markers. Compared with control values, NaBu treatment decreased mast cell recruitment/activation, inflammatory enzyme activities, levels of pro-inflammatory cytokines, and the proteins p65 and p50 of the nuclear factor-κB pathway. Angiogenesis, as determined by haemoglobin content, vascular endothelial growth factor levels and the number of blood vessels in the implant, was reduced by the treatment. In NaBu-treated animals, the predominant collagen present in the abdominal fibrovascular tissue was thin collagen, whereas in control implants it was thick collagen. Transforming growth factor-β1 levels were also lower in implants of treated animals. Sodium butyrate downregulated the inflammatory, angiogenesis and fibrogenesis axes of the fibroproliferative tissue induced by the intraperitoneal synthetic matrix. This compound has potential to control/regulate chronic inflammation and adverse healing processes in the abdominal cavity.
Collapse
Affiliation(s)
| | | | - Luciana Xavier Pereira
- Department of Experimental PathologyUniversidade Federal de São João del‐ReiDivinópolisMinas GeraisBrazil
| | | | - Henning Ulrich
- Department of BiochemistryInstitute of ChemistryUniversity of São PauloSão PauloSão PauloBrazil
| | - Silvia Passos Andrade
- Department of Physiology and BiophysicsInstitute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Paula Peixoto Campos
- Department of General PathologyInstitute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| |
Collapse
|
37
|
Li D, Guo X, Zhao W, Jingyu J, Xia C, Yu G. Genome-wide DNA methylation dynamics in carbon tetrachloride-induced mice liver fibrosis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:85-92. [PMID: 36594057 PMCID: PMC9790058 DOI: 10.22038/ijbms.2022.66256.14555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/22/2022] [Indexed: 01/04/2023]
Abstract
Objectives Many persistent harmful stimuli can result in chronic liver diseases, which lead to about 2 million deaths per year in the whole world. Liver fibrosis was found to exist in all kinds of chronic liver diseases. Many studies suggested that DNA methylation was associated with the pathogenesis of liver fibrosis. This study aimed to quantitatively detect DNA methylation changes in the whole genome in fibrotic liver tissues of mice. Materials and Methods Liver fibrosis was induced by intraperitoneal injection of carbon tetrachloride (CCl4) for 4 weeks. A genome-wide methylome analysis was performed using 850K BeadChips assays. The methylation status of 27 CpG dinucleotides located in 3 genes was detected by pyrosequencing to confirm chip data accuracy, and mRNA expressions of these 3 genes were examined by RT-qPCR methods. Results A total of 130,068 differentially methylated sites (DMS, 58,474 hypermethylated, and 71,594 hypomethylated) between fibrotic liver tissues and control mice liver tissues were identified by the 850k BeadChips array. Consistency between pyrosequencing data and 850k BeadChips array data was observed (R=0.928; P<0.01). Apoptosis, positive regulation of transcription of Notch receptor target, and negative regulation of p38MAPK signal cascade activities were significantly enriched in the Gene Ontology (GO) analyses. Cholesterol metabolism, bile secretion, and more biosynthesis and metabolism pathways were enriched in KEGG pathway analyses. Ten key genes were identified by the Cytoscape plugin cytoHubba. Conclusion 7850 genes were found to have methylation change in fibrotic liver tissues of mice, which facilitates future research for clinical application.
Collapse
Affiliation(s)
- Deming Li
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China,These authors contributed eqully to this work
| | - Xiaoshu Guo
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China,Department of Physiology, Changzhi Medical College, Shanxi, China,These authors contributed eqully to this work
| | - Wenyu Zhao
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Jingyu Jingyu
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Cong Xia
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Guoying Yu
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China,Corresponding author: Guoying Yu. State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China. Tel: +86-03733326340; Fax: +86-0373 3326524;
| |
Collapse
|
38
|
Nagai H, Tanoue Y, Nakamura T, Chan CJJ, Yamada S, Saitou M, Fukuda T, Sheng G. Mesothelial fusion mediates chorioallantoic membrane formation. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210263. [PMID: 36252211 PMCID: PMC9574633 DOI: 10.1098/rstb.2021.0263] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 05/29/2022] [Indexed: 12/23/2022] Open
Abstract
In amniotic vertebrates (birds, reptiles and mammals), an extraembryonic structure called the chorioallantoic membrane (CAM) functions as respiratory organ for embryonic development. The CAM is derived from fusion between two pre-existing membranes, the allantois, a hindgut diverticulum and a reservoir for metabolic waste, and the chorion which marks the embryo's external boundary. Modified CAM in eutherian mammals, including humans, gives rise to chorioallantoic placenta. Despite its importance, little is known about cellular and molecular mechanisms mediating CAM formation and maturation. In this work, using the avian model, we focused on the early phase of CAM morphogenesis when the allantois and chorion meet and initiate fusion. We report here that chicken chorioallantoic fusion takes place when the allantois reaches the size of 2.5-3.0 mm in diameter and in about 6 hours between E3.75 and E4. Electron microscopy and immunofluorescence analyses suggested that before fusion, in both the allantois and chorion, an epithelial-shaped mesothelial layer is present, which dissolves after fusion, presumably by undergoing epithelial-mesenchymal transition. The fusion process per se, however, is independent of allantoic growth, circulation, or its connection to the developing mesonephros. Mesoderm cells derived from the allantois and chorion can intermingle post-fusion, and chorionic ectoderm cells exhibit a specialized sub-apical intercellular interface, possibly to facilitate infiltration of allantois-derived vascular progenitors into the chorionic ectoderm territory for optimal oxygen transport. Finally, we investigated chorioallantoic fusion-like process in primates, with limited numbers of archived human and fresh macaque samples. We summarize the similarities and differences of CAM formation among different amniote groups and propose that mesothelial epithelial-mesenchymal transition mediates chorioallantoic fusion in most amniotic vertebrates. Further study is needed to clarify tissue morphogenesis leading to chorioallantoic fusion in primates. Elucidating molecular mechanisms regulating mesothelial integrity and epithelial-mesenchymal transition will also help understand mesothelial diseases in the adult, including mesothelioma, ovarian cancer and fibrosis. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.
Collapse
Affiliation(s)
- Hiroki Nagai
- International Research Center for Medical Sciences (IRCMS), Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yuki Tanoue
- International Research Center for Medical Sciences (IRCMS), Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Tomonori Nakamura
- Institute for the Advanced Study of Human Biology (WPI-ASHBI), Kyoto University, Kyoto 606-8501, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- The Hakubi Center for Advanced Research, Kyoto University, Kyoto 606-8501, Japan
| | - Christopher J. J. Chan
- International Research Center for Medical Sciences (IRCMS), Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Shigehito Yamada
- Congenital Anomaly Research Center, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Mitinori Saitou
- Institute for the Advanced Study of Human Biology (WPI-ASHBI), Kyoto University, Kyoto 606-8501, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Takaichi Fukuda
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Guojun Sheng
- International Research Center for Medical Sciences (IRCMS), Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
39
|
Chua JW, Thangaveloo M, Lim DXE, Madden LE, Phillips ARJ, Becker DL. Connexin43 in Post-Surgical Peritoneal Adhesion Formation. Life (Basel) 2022; 12:1734. [PMID: 36362888 PMCID: PMC9697983 DOI: 10.3390/life12111734] [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: 09/02/2022] [Revised: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 09/10/2024] Open
Abstract
OBJECTIVE Post-surgical peritoneal adhesions are a serious problem for the quality of life and fertility. Yet there are no effective ways of preventing their occurrence. The gap junction protein Cx43 is known to be involved in fibrosis in several different organs and disease conditions often associated with inflammation. Here we examined the Cx43 dynamic expression in an ischemic button model of surgical adhesions. METHODS Using the mouse ischemic button model, Cx43 antisense was delivered in Pluronic gel to attenuate Cx43 expression. The severity of button formation and immunofluorescence analysis of Cx43 and TGF-β1 were performed. The concentration of tissue plasminogen activator via ELISA was also performed. RESULTS As early as 6 h after button formation, the Cx43 levels were elevated in and around the button and some weak adhesions were formed. By 24 h Cx43 levels had increased further and adhesions were more defined. At 7 days the adhesions were much more robust, opaque, and vascularized, requiring blunt or sharp dissection to break them. Cx43 antisense attenuated its upregulation and, reduced the number and severity of adhesions that formed. CONCLUSION Targeting Cx43 after surgical procedures may be a potential therapeutic strategy for preventing adhesion formation or at least reducing their severity.
Collapse
Affiliation(s)
- Jia Wang Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- Skin Research Institute Singapore, Singapore 308232, Singapore
| | - Moogaambikai Thangaveloo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- Skin Research Institute Singapore, Singapore 308232, Singapore
| | - Debbie Xiu En Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- Skin Research Institute Singapore, Singapore 308232, Singapore
| | - Leigh E. Madden
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- Skin Research Institute Singapore, Singapore 308232, Singapore
| | | | - David L. Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- Skin Research Institute Singapore, Singapore 308232, Singapore
| |
Collapse
|
40
|
Carrillo-Rodríguez P, Robles-Guirado JÁ, Cruz-Palomares A, Palacios-Pedrero MÁ, González-Paredes E, Más-Ciurana A, Franco-Herrera C, Ruiz-de-Castroviejo-Teba PA, Lario A, Longobardo V, Montosa-Hidalgo L, Pérez-Sánchez-Cañete MM, Corzo-Corbera MM, Redondo-Sánchez S, Jodar AB, Blanco FJ, Zumaquero E, Merino R, Sancho J, Zubiaur M. Extracellular vesicles from pristane-treated CD38-deficient mice express an anti-inflammatory neutrophil protein signature, which reflects the mild lupus severity elicited in these mice. Front Immunol 2022; 13:1013236. [DOI: 10.3389/fimmu.2022.1013236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
In CD38-deficient (Cd38-/-) mice intraperitoneal injection of pristane induces a lupus-like disease, which is milder than that induced in WT mice, showing significant differences in the inflammatory and autoimmune processes triggered by pristane. Extracellular vesicles (EV) are present in all body fluids. Shed by cells, their molecular make-up reflects that of their cell of origin and/or tissue pathological situation. The aim of this study was to analyze the protein composition, protein abundance, and functional clustering of EV released by peritoneal exudate cells (PECs) in the pristane experimental lupus model, to identify predictive or diagnostic biomarkers that might discriminate the autoimmune process in lupus from inflammatory reactions and/or normal physiological processes. In this study, thanks to an extensive proteomic analysis and powerful bioinformatics software, distinct EV subtypes were identified in the peritoneal exudates of pristane-treated mice: 1) small EV enriched in the tetraspanin CD63 and CD9, which are likely of exosomal origin; 2) small EV enriched in CD47 and CD9, which are also enriched in plasma-membrane, membrane-associated proteins, with an ectosomal origin; 3) small EV enriched in keratins, ECM proteins, complement/coagulation proteins, fibrin clot formation proteins, and endopetidase inhibitor proteins. This enrichment may have an inflammation-mediated mesothelial-to-mesenchymal transition origin, representing a protein corona on the surface of peritoneal exudate EV; 4) HDL-enriched lipoprotein particles. Quantitative proteomic analysis allowed us to identify an anti-inflammatory, Annexin A1-enriched pro-resolving, neutrophil protein signature, which was more prominent in EV from pristane-treated Cd38-/- mice, and quantitative differences in the protein cargo of the ECM-enriched EV from Cd38-/- vs WT mice. These differences are likely to be related with the distinct inflammatory outcome shown by Cd38-/- vs WT mice in response to pristane treatment. Our results demonstrate the power of a hypothesis-free and data-driven approach to transform the heterogeneity of the peritoneal exudate EV from pristane-treated mice in valuable information about the relative proportion of different EV in a given sample and to identify potential protein markers specific for the different small EV subtypes, in particular those proteins defining EV involved in the resolution phase of chronic inflammation.
Collapse
|
41
|
Polycaprolactone/Chitosan Composite Nanofiber Membrane as a Preferred Scaffold for the Culture of Mesothelial Cells and the Repair of Damaged Mesothelium. Int J Mol Sci 2022; 23:ijms23179517. [PMID: 36076916 PMCID: PMC9455682 DOI: 10.3390/ijms23179517] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/26/2022] Open
Abstract
Mesothelial cells are specific epithelial cells lining the serosal cavity and internal organs. Nonetheless, few studies have explored the possibility to culture mesothelial cells in a nanostructure scaffold for tissue engineering applications. Therefore, this study aims to fabricate nanofibers from a polycaprolactone (PCL) and PCL/chitosan (CS) blend by electrospinning, and to elucidate the effect of CS on the cellular response of mesothelial cells. The results demonstrate that a PCL and PCL/CS nanofiber membrane scaffold could be prepared with a comparable fiber diameter (~300 nm) and porosity for cell culture. Blending CS with PCL influenced the mechanical properties of the scaffold due to interference of PCL crystallinity in the nanofibers. However, CS substantially improves scaffold hydrophilicity and results in a ~6-times-higher cell attachment rate in PCL/CS. The mesothelial cells maintain high viability in both nanofiber membranes, but PCL/CS provides better maintenance of cobblestone-like mesothelial morphology. From gene expression analysis and immunofluorescence staining, the incorporation of CS also results in the upregulated expression of mesothelial marker genes and the enhanced production of key mesothelial maker proteins, endorsing PCL/CS to better maintain the mesothelial phenotype. The PCL/CS scaffold was therefore chosen for the in vivo studies, which involved transplanting a cell/scaffold construct containing allograft mesothelial cells for mesothelium reconstruction in rats. In the absence of mesothelial cells, the mesothelium wound covered with PCL/CS showed an inflammatory response. In contrast, a mesothelium layer similar to native mesothelium tissue could be obtained by implanting the cell/scaffold construct, based on hematoxylin and eosin (H&E) and immunohistochemical staining.
Collapse
|
42
|
Busato F, Drigo M, Zoia A. Reduced risk of arterial thromboembolism in cats with pleural effusion due to congestive heart failure. J Feline Med Surg 2022; 24:e142-e152. [PMID: 35549930 PMCID: PMC10812273 DOI: 10.1177/1098612x221094663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 12/29/2022]
Abstract
OBJECTIVES The aim of the study was to determine whether cardiogenic pleural effusion in cats is associated with a lower risk of arterial thromboembolism (ATE) compared with cats with cardiac disease without evidence of pleural effusion. METHODS A cross-sectional study was conducted on owned cats with natural occurring cardiac diseases. Cats included were classified in three groups: those with cardiac disease but no evidence of congestive heart failure (CHF); those with evidence of cardiogenic pulmonary oedema; and those with evidence of cardiogenic pleural effusion. Prevalence of ATE was calculated and the variables analysed for an association with this outcome were the presence and type of CHF, sex and neuter status, age, breed, type of cardiac diseases and left atrial (LA) dimension. A multivariable logistic regression model was used to fit the association between ATE and these variables. RESULTS A total of 366 cats with cardiac disease met the inclusion criteria: 179 were included in the group with cardiac disease but no evidence of CHF, 66 in the group with evidence of cardiogenic pulmonary oedema and 121 in the group with evidence of cardiogenic pleural effusion. Prevalence of ATE (58/366 [15.8%]) was significantly different among groups (with no evidence of CHF, 28/179 [15.6%]; with evidence of cardiogenic pulmonary oedema, 22/66 [33.3%]; with evidence of cardiogenic pleural effusion, 8/121 [6.6%]; P <0.001). Cats with ATE had a significantly higher LA to aortic root ratio (2.30 ± 0.46) than those without ATE (2.04 ± 0.46; P <0.001). Multivariable logistic regression analysis indicated that the group with evidence of cardiogenic pleural effusion was associated with a lower risk of developing ATE compared with groups with cardiac disease but no evidence of CHF and with evidence of cardiogenic pulmonary oedema (P = 0.005 and P <0.001, respectively). CONCLUSIONS AND RELEVANCE Presence of cardiogenic pleural effusion is associated with a lower risk of developing ATE, while LA enlargement is a risk factor for ATE.
Collapse
Affiliation(s)
- Francesca Busato
- Division of Internal Medicine, San Marco Veterinary Clinic, Veggiano (PD), Italy
| | - Michele Drigo
- Department of Medicina Animale, Produzione e Salute, Padua University, Legnaro (PD), Italy
| | - Andrea Zoia
- Division of Internal Medicine, San Marco Veterinary Clinic, Veggiano (PD), Italy
| |
Collapse
|
43
|
Kawata M, Kondo J, Onuma K, Ito Y, Yokoi T, Hamanishi J, Mandai M, Kimura T, Inoue M. Polarity switching of ovarian cancer cell clusters via SRC family kinase is involved in the peritoneal dissemination. Cancer Sci 2022; 113:3437-3448. [PMID: 35848881 PMCID: PMC9530866 DOI: 10.1111/cas.15493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022] Open
Abstract
Peritoneal dissemination is a predominant pattern of metastasis in patients with advanced ovarian cancer. Despite recent progress in the management strategy, peritoneal dissemination remains a determinant of poor ovarian cancer prognosis. Using various histological types of patient‐derived ovarian cancer organoids, the roles of the apicobasal polarity of ovarian cancer cell clusters in peritoneal dissemination were studied. First, it was found that both ovarian cancer tissues and ovarian organoids showed apicobasal polarity, where zonula occludens‐1 (ZO‐1) and integrin beta 4 (ITGB4) served as markers for apical and basal sides, respectively. The organoids in suspension culture, as a model of cancer cell cluster floating in ascites, showed apical‐out/basal‐in polarity status, while once embedded in extracellular matrix (ECM), the organoids switched their polarity to apical‐in/basal‐out. This polarity switch was accompanied by the SRC kinase family (SFK) phosphorylation and was inhibited by SFK inhibitors. SFK inhibitors abrogated the adherence of the organoids onto the ECM‐coated plastic surface. When the organoids were seeded on a mesothelial cell layer, they cleared and invaded mesothelial cells. In vivo, dasatinib, an SFK inhibitor, suppressed peritoneal dissemination of ovarian cancer organoids in immunodeficient mice. These results suggest SFK‐mediated polarity switching is involved in peritoneal metastasis. Polarity switching would be a potential therapeutic target for suppressing peritoneal dissemination in ovarian cancer.
Collapse
Affiliation(s)
- Mayuko Kawata
- Department of Clinical Bioresource Research and Development, Kyoto University Graduate School of Medicine.,Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine
| | - Jumpei Kondo
- Department of Clinical Bioresource Research and Development, Kyoto University Graduate School of Medicine.,Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine.,Department of Molecular Biochemistry and Clinical Investigation, Division of Health Science, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita City, Osaka, Japan
| | - Kunishige Onuma
- Department of Clinical Bioresource Research and Development, Kyoto University Graduate School of Medicine
| | - Yu Ito
- Department of Clinical Bioresource Research and Development, Kyoto University Graduate School of Medicine.,Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine
| | - Takeshi Yokoi
- Department of Obstetrics and Gynecology, Kaizuka, City Hospital
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine
| | - Masahiro Inoue
- Department of Clinical Bioresource Research and Development, Kyoto University Graduate School of Medicine
| |
Collapse
|
44
|
Huang H, Wang Z, Zhang Y, Pradhan RN, Ganguly D, Chandra R, Murimwa G, Wright S, Gu X, Maddipati R, Müller S, Turley SJ, Brekken RA. Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer. Cancer Cell 2022; 40:656-673.e7. [PMID: 35523176 PMCID: PMC9197998 DOI: 10.1016/j.ccell.2022.04.011] [Citation(s) in RCA: 271] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/25/2022] [Accepted: 04/14/2022] [Indexed: 12/11/2022]
Abstract
Recent studies have identified a unique cancer-associated fibroblast (CAF) population termed antigen-presenting CAFs (apCAFs), characterized by the expression of major histocompatibility complex class II molecules, suggesting a function in regulating tumor immunity. Here, by integrating multiple single-cell RNA-sequencing studies and performing robust lineage-tracing assays, we find that apCAFs are derived from mesothelial cells. During pancreatic cancer progression, mesothelial cells form apCAFs by downregulating mesothelial features and gaining fibroblastic features, a process induced by interleukin-1 and transforming growth factor β. apCAFs directly ligate and induce naive CD4+ T cells into regulatory T cells (Tregs) in an antigen-specific manner. Moreover, treatment with an antibody targeting the mesothelial cell marker mesothelin can effectively inhibit mesothelial cell to apCAF transition and Treg formation induced by apCAFs. Taken together, our study elucidates how mesothelial cells may contribute to immune evasion in pancreatic cancer and provides insight on strategies to enhance cancer immune therapy.
Collapse
Affiliation(s)
- Huocong Huang
- Department of Surgery, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA.
| | - Zhaoning Wang
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Yuqing Zhang
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA; Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | | | - Debolina Ganguly
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA; Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Raghav Chandra
- Department of Surgery, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Gilbert Murimwa
- Department of Surgery, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Steven Wright
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Xiaowu Gu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Ravikanth Maddipati
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | | | | | - Rolf A Brekken
- Department of Surgery, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA; Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA.
| |
Collapse
|
45
|
Ditzig Z, Wilson CM, Salas J, Serve KM. Plasminogen Binding and Activation at the Mesothelial Cell Surface Promotes Invasion through a Collagen Matrix. Int J Mol Sci 2022; 23:ijms23115984. [PMID: 35682663 PMCID: PMC9180734 DOI: 10.3390/ijms23115984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 01/04/2023] Open
Abstract
Plasminogen (Plg) activation to the serine protease plasmin (Pla) plays a key role in regulating wound healing and fibrotic responses, particularly when bound to cell surface receptors. Our previous work suggested that mesothelial cells bind Plg at the cell surface, though no Plg receptors were described for these cells. Since mesothelial cells contribute to injury responses, including cellular differentiation to a mesenchymal-like phenotype and extracellular matrix remodeling, we hypothesized that Plg binding would promote these responses. Here, we confirm that Plg binds to both pleural and peritoneal mesothelial cells via the lysine-binding domain present in Plg, and we demonstrate the presence of three Plg receptors on the mesothelial cell surface: α-Enolase, Annexin A2, and Plg-RKT. We further show that bound-Plg is activated to Pla on the cell surface and that activation is blocked by an inhibitor of urokinase plasminogen activator or by the presence of animal-derived FBS. Lastly, we demonstrate that Plg promotes mesothelial cell invasion through a type I collagen matrix but does not promote cellular differentiation or proliferation. These data demonstrate for the first time that mesothelial cells bind and activate Plg at the cell surface and that active Pla is involved in mesothelial cell invasion without cell differentiation.
Collapse
|
46
|
Prummel KD, Crowell HL, Nieuwenhuize S, Brombacher EC, Daetwyler S, Soneson C, Kresoja-Rakic J, Kocere A, Ronner M, Ernst A, Labbaf Z, Clouthier DE, Firulli AB, Sánchez-Iranzo H, Naganathan SR, O'Rourke R, Raz E, Mercader N, Burger A, Felley-Bosco E, Huisken J, Robinson MD, Mosimann C. Hand2 delineates mesothelium progenitors and is reactivated in mesothelioma. Nat Commun 2022; 13:1677. [PMID: 35354817 PMCID: PMC8967825 DOI: 10.1038/s41467-022-29311-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/04/2022] [Indexed: 01/27/2023] Open
Abstract
The mesothelium lines body cavities and surrounds internal organs, widely contributing to homeostasis and regeneration. Mesothelium disruptions cause visceral anomalies and mesothelioma tumors. Nonetheless, the embryonic emergence of mesothelia remains incompletely understood. Here, we track mesothelial origins in the lateral plate mesoderm (LPM) using zebrafish. Single-cell transcriptomics uncovers a post-gastrulation gene expression signature centered on hand2 in distinct LPM progenitor cells. We map mesothelial progenitors to lateral-most, hand2-expressing LPM and confirm conservation in mouse. Time-lapse imaging of zebrafish hand2 reporter embryos captures mesothelium formation including pericardium, visceral, and parietal peritoneum. We find primordial germ cells migrate with the forming mesothelium as ventral migration boundary. Functionally, hand2 loss disrupts mesothelium formation with reduced progenitor cells and perturbed migration. In mouse and human mesothelioma, we document expression of LPM-associated transcription factors including Hand2, suggesting re-initiation of a developmental program. Our data connects mesothelium development to Hand2, expanding our understanding of mesothelial pathologies.
Collapse
Affiliation(s)
- Karin D Prummel
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
- Structural and Computational Biology Unit, EMBL, Heidelberg, Germany
| | - Helena L Crowell
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zürich, Switzerland
| | - Susan Nieuwenhuize
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Eline C Brombacher
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stephan Daetwyler
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Charlotte Soneson
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zürich, Switzerland
| | - Jelena Kresoja-Rakic
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Laboratory of Molecular Oncology, Department of Thoracic Surgery, University Hospital Zurich, Zürich, Switzerland
| | - Agnese Kocere
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Manuel Ronner
- Laboratory of Molecular Oncology, Department of Thoracic Surgery, University Hospital Zurich, Zürich, Switzerland
| | | | - Zahra Labbaf
- Institute for Cell Biology, ZMBE, Muenster, Germany
| | - David E Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anthony B Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN, USA
| | - Héctor Sánchez-Iranzo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
- Institute of Biological and Chemical System - Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Sundar R Naganathan
- Institute of Bioengineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Rebecca O'Rourke
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Erez Raz
- Institute for Cell Biology, ZMBE, Muenster, Germany
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern, Switzerland
- Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Alexa Burger
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, Department of Thoracic Surgery, University Hospital Zurich, Zürich, Switzerland
| | - Jan Huisken
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Morgridge Institute for Research, Madison, WI, USA
| | - Mark D Robinson
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zürich, Switzerland
| | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
47
|
Hausburg MA, Bocker JM, Madayag RM, Mains CW, Banton KL, Liniewicz TE, Tanner A, Sercy E, Bar-Or R, Williams JS, Ryznar RJ, Bar-Or D. Characterization of Peritoneal Reactive Ascites Collected from Acute Appendicitis and Small Bowel Obstruction Patients. Clin Chim Acta 2022; 531:126-136. [PMID: 35346646 DOI: 10.1016/j.cca.2022.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 11/03/2022]
|
48
|
Wilson AC, Chiles J, Ashish S, Chanda D, Kumar PL, Mobley JA, Neptune ER, Thannickal VJ, McDonald MLN. Integrated bioinformatics analysis identifies established and novel TGFβ1-regulated genes modulated by anti-fibrotic drugs. Sci Rep 2022; 12:3080. [PMID: 35197532 PMCID: PMC8866468 DOI: 10.1038/s41598-022-07151-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/14/2022] [Indexed: 11/29/2022] Open
Abstract
Fibrosis is a leading cause of morbidity and mortality worldwide. Although fibrosis may involve different organ systems, transforming growth factor-β (TGFβ) has been established as a master regulator of fibrosis across organs. Pirfenidone and Nintedanib are the only currently-approved drugs to treat fibrosis, specifically idiopathic pulmonary fibrosis, but their mechanisms of action remain poorly understood. To identify novel drug targets and uncover potential mechanisms by which these drugs attenuate fibrosis, we performed an integrative 'omics analysis of transcriptomic and proteomic responses to TGFβ1-stimulated lung fibroblasts. Significant findings were annotated as associated with pirfenidone and nintedanib treatment in silico via Coremine. Integrative 'omics identified a co-expressed transcriptomic and proteomic module significantly correlated with TGFβ1 treatment that was enriched (FDR-p = 0.04) with genes associated with pirfenidone and nintedanib treatment. While a subset of genes in this module have been implicated in fibrogenesis, several novel TGFβ1 signaling targets were identified. Specifically, four genes (BASP1, HSD17B6, CDH11, and TNS1) have been associated with pirfenidone, while five genes (CLINT1, CADM1, MTDH, SYDE1, and MCTS1) have been associated with nintedanib, and MYDGF has been implicated with treatment using both drugs. Using the Clue Drug Repurposing Hub, succinic acid was highlighted as a metabolite regulated by the protein encoded by HSD17B6. This study provides new insights into the anti-fibrotic actions of pirfenidone and nintedanib and identifies novel targets for future mechanistic studies.
Collapse
Affiliation(s)
- Ava C. Wilson
- grid.265892.20000000106344187Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL USA ,grid.265892.20000000106344187Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA
| | - Joe Chiles
- grid.265892.20000000106344187Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA
| | - Shah Ashish
- grid.265892.20000000106344187Department of Orthopedic Surgery, University of Alabama at Birmingham, Birmingham, AL USA
| | - Diptiman Chanda
- grid.265892.20000000106344187Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA
| | - Preeti L. Kumar
- grid.265892.20000000106344187Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA
| | - James A. Mobley
- grid.265892.20000000106344187Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL USA
| | - Enid R. Neptune
- grid.21107.350000 0001 2171 9311Department of Medicine, Johns Hopkins University, Baltimore, MD USA
| | - Victor J. Thannickal
- grid.265892.20000000106344187Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,grid.265219.b0000 0001 2217 8588John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA USA
| | - Merry-Lynn N. McDonald
- grid.265892.20000000106344187Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL USA ,grid.265892.20000000106344187Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,grid.265892.20000000106344187Department of Genetics, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
49
|
Wei G, Wang Z, Liu R, Zhou C, Li E, Shen T, Wang X, Wu Y, Li X. A combination of hybrid polydopamine-human keratinocyte growth factor nanoparticles and sodium hyaluronate for the efficient prevention of postoperative abdominal adhesion formation. Acta Biomater 2022; 138:155-167. [PMID: 34653692 DOI: 10.1016/j.actbio.2021.10.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/12/2022]
Abstract
Postoperative abdominal adhesion (PAA) is one of the more universal complications of abdominal surgery with a frequent incidence. Currently available keratinocyte growth factor (KGF)-based glues for the prevention of adhesions remain a great bottleneck since their long-term biological activity in vivo is insufficient. In this study, we fabricated hybrid polydopamine (PDA)-KGF nanoparticles (PDA-KGF NPs) by using an in situ self-assembly and polymerization method. The physicochemical properties of the PDA-KGF nanoparticles were systematically characterized. The effect of preventing PAA in rats was evaluated by using hybrid PDA-KGF NPs combined with hyaluronate (Ha). The expression levels of inflammatory factors and the degree of inflammatory cell infiltration in the injured peritoneum were evaluated by enzyme-linked immunosorbent assays and hematoxylin-eosin staining, respectively. The levels of phospho-Src expression were revealed by Western blotting. The degree of fibrosis and the density of deposited collagen fibers were measured with real-time reverse-transcription polymerase chain reaction and picrosirius red staining. The results indicated that the PDA-KGF NPs combined with Ha greatly prevented the incidence of abdominal adhesion s and promoted the repair of mesothelial cells in injured peritoneum. More importantly, the PDA-KGF NPs combined with Ha obviously reduced collagen deposition and fibrosis and inhibited the inflammatory response. Our results suggest that PDA-KGF NPs combined with Ha are promising barrier-like biomaterials for the effective prevention of postoperative tissue adhesion. STATEMENT OF SIGNIFICANCE: Postoperative abdominal adhesion (PAA) as an inevitable postoperative complication affected the quality of life of patients. Currently available methods for preventing adhesions mainly employ degradable biomaterials. Previous research demonstrated that a hybrid keratinocyte growth factor (KGF)-sodium hyaluronate (Ha) gel could prevent the formation of PAAs. However, its clinical outcomes are not satisfactory since their bioactivity in vivo is too short. In this article, we fabricated hybrid polydopamine (PDA)-KGF nanoparticles (PDA-KGF NPs), which extend KGF bioactivity, effectively prevent PAA. Moreover, PDA-KGF NPs could remarkably reduce both collagen deposition and fibrosis, inhibit the inflammatory response, and promote mesothelial regeneration. Overall, the PDA-KGF NPs combined with Ha exhibit efficient antiadhesion properties, may provide a promising clinical protocol for the prevention of PAA.
Collapse
Affiliation(s)
- Guangbing Wei
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Zijun Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Ruilin Liu
- College of Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China; College of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Cancan Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Enmeng Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Tianli Shen
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Xingjie Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Yunhua Wu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of General Surgery, Shaanxi Provincial People's Hospital, Xi' an 710068, PR China
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| |
Collapse
|
50
|
Zindel J, Mittner J, Bayer J, April-Monn SL, Kohler A, Nusse Y, Dosch M, Büchi I, Sanchez-Taltavull D, Dawson H, Gomez de Agüero M, Asahina K, Kubes P, Macpherson AJ, Stroka D, Candinas D. Intraperitoneal microbial contamination drives post-surgical peritoneal adhesions by mesothelial EGFR-signaling. Nat Commun 2021; 12:7316. [PMID: 34916513 PMCID: PMC8677808 DOI: 10.1038/s41467-021-27612-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/01/2021] [Indexed: 12/19/2022] Open
Abstract
Abdominal surgeries are lifesaving procedures but can be complicated by the formation of peritoneal adhesions, intra-abdominal scars that cause intestinal obstruction, pain, infertility, and significant health costs. Despite this burden, the mechanisms underlying adhesion formation remain unclear and no cure exists. Here, we show that contamination of gut microbes increases post-surgical adhesion formation. Using genetic lineage tracing we show that adhesion myofibroblasts arise from the mesothelium. This transformation is driven by epidermal growth factor receptor (EGFR) signaling. The EGFR ligands amphiregulin and heparin-binding epidermal growth factor, are sufficient to induce these changes. Correspondingly, EGFR inhibition leads to a significant reduction of adhesion formation in mice. Adhesions isolated from human patients are enriched in EGFR positive cells of mesothelial origin and human mesothelium shows an increase of mesothelial EGFR expression during bacterial peritonitis. In conclusion, bacterial contamination drives adhesion formation through mesothelial EGFR signaling. This mechanism may represent a therapeutic target for the prevention of adhesions after intra-abdominal surgery.
Collapse
Affiliation(s)
- Joel Zindel
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department of Pharmacology and Physiology and Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology & Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Jonas Mittner
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Julia Bayer
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Simon L April-Monn
- Clinical Pathology Division and Translational Research Unit, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Andreas Kohler
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ysbrand Nusse
- Department of Pharmacology and Physiology and Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology & Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Michel Dosch
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Isabel Büchi
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Daniel Sanchez-Taltavull
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Heather Dawson
- Clinical Pathology Division and Translational Research Unit, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Mercedes Gomez de Agüero
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Kinji Asahina
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis and Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
- Central Research Laboratory, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Paul Kubes
- Department of Pharmacology and Physiology and Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology & Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Andrew J Macpherson
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Deborah Stroka
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Daniel Candinas
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|