1
|
Interferon-γ enhances the immunosuppressive ability of canine bone marrow-derived mesenchymal stem cells by activating the TLR3-dependent IDO/kynurenine pathway. Mol Biol Rep 2022; 49:8337-8347. [PMID: 35690960 DOI: 10.1007/s11033-022-07648-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/25/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND The immunomodulatory function of mesenchymal stem cells (MSCs) has been considered to be vital for MSC-based therapies. Many works have been devoted to excavate effective strategies for enhancing the immunomodulation effect of MSCs. Nonetheless, canine MSC-mediated immunomodulation is still poorly understood. METHODS AND RESULTS The inflammatory microenvironment was simulated through the employment of interferon-γ (IFN-γ) in a culture system. Compared with unstimulated cBMSCs, IFN-γ stimulation increased the mRNA levels of Toll-like receptor 3 (TLR3) and indoleamine 2, 3-dioxygenase 1 (IDO-1), and simultaneously enhanced the secretion of immunosuppressive molecules, including interleukin (IL)-10, hepatocyte growth factor (HGF), and kynurenine in cBMSCs. IFN-γ stimulation significantly enhanced the ability of cBMSCs and their supernatant to suppress the proliferation of murine spleen lymphocytes. Lymphocyte subtyping evaluation revealed that cBMSCs and their supernatant diminished the percentage of CD3+CD4+ and CD3+CD8+ lymphocytes compared with the control group, with a decreasing CD4+/CD8+ ratio. Notably, exposure to IFN-γ decreased the CD4+/CD8+ ratio more effectively than unstimulated cells or supernatant. Additionally, IFN-γ-stimulation increased the mRNA levels of the Th1 cytokines TNF-α, and remarkably decreased the mRNA level of the Th2 cytokine IL-4 and IL-10. CONCLUSION Our findings substantiate that IFN-γ stimulation can enhance the immunomodulatory properties of cBMSCs by promoting TLR3-dependent activation of the IDO/kynurenine pathway, increasing the secretion of immunoregulatory molecules and strengthening interactions with T lymphocytes, which may provide a meaningful strategy for the clinical application of cBMSCs in immune-related diseases.
Collapse
|
2
|
Buote NJ. Laparoscopic adipose-derived stem cell harvest technique with bipolar sealing device: Outcome in 12 dogs. Vet Med Sci 2022; 8:1421-1428. [PMID: 35537084 PMCID: PMC9297765 DOI: 10.1002/vms3.816] [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: 11/16/2022] Open
Abstract
Objective This study aimed to describe the technique and clinical outcomes in dogs undergoing Laparoscopic Adipose‐Derived Stem Cell Harvest via bipolar sealing device (LADSCHB) for degenerative orthopaedic and neurologic disease. Study Design Descriptive retrospective case series. Animals Eleven dogs with orthopaedic disease and one dog with degenerative spinal disease were enrolled in the study. Methods Medical records of dogs undergoing LADSCHB were reviewed for signalment, weight, reason for the procedure, anaesthesia time, surgery time, other procedures performed, post‐operative pain protocols, incision size, amount of adipose tissue collected, number of viable cells collected, days to discharge, short‐term complications, and owner satisfaction. Results The median weight of the population was 34.2 kg (range 9.2–62 kg), the median surgery time was 39 min (range 15–45 min), mean incision length was 2.5 cm, the median amount of adipose collected was 60 g, and the median number of viable stem cells was 21 million cells. Conversion to open laparotomy was not needed. The most common reason for the harvest was osteoarthritis of the elbow (8/12 cases). Nine cases had other procedures performed at the same time as the harvest. No complications were noted during the procedure or within the post‐operative period. All owners surveyed were satisfied with the laparoscopic harvest procedure. Conclusions LADSCHB was technically feasible, productive, and not associated with any complications. This procedure was performed rapidly and was paired with other surgical procedures. Clinical Significance LADSCHB allows for stem cell harvest with commonly utilized laparoscopic equipment. This surgical technique could lead to the increased ability to treat patients with diseases that benefit from stem cell therapy.
Collapse
Affiliation(s)
- Nicole J Buote
- VCA West Los Angeles Animal Hospital, Los Angeles, California, USA
| |
Collapse
|
3
|
Park SG, An JH, Li Q, Chae HK, Park SM, Lee JH, Ahn JO, Song WJ, Youn HY. Feline adipose tissue-derived mesenchymal stem cells pretreated with IFN-γ enhance immunomodulatory effects through the PGE₂ pathway. J Vet Sci 2021; 22:e16. [PMID: 33774932 PMCID: PMC8007449 DOI: 10.4142/jvs.2021.22.e16] [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: 06/05/2020] [Revised: 12/12/2020] [Accepted: 01/07/2021] [Indexed: 01/03/2023] Open
Abstract
Background Preconditioning with inflammatory stimuli is used to improve the secretion of anti-inflammatory agents in stem cells from variant species such as mouse, human, and dog. However, there are only few studies on feline stem cells. Objectives This study aimed to evaluate the immune regulatory capacity of feline adipose tissue-derived (fAT) mesenchymal stem cells (MSCs) pretreated with interferon-gamma (IFN-γ). Methods To assess the interaction of lymphocytes and macrophages with IFN-γ-pretreated fAT-MSCs, mouse splenocytes and RAW 264.7 cells were cultured with the conditioned media from IFN-γ-pretreated MSCs. Results Pretreatment with IFN-γ increased the gene expression levels of cyclooxygenase-2, indoleamine 2,3-dioxygenase, hepatocyte growth factor, and transforming growth factor-beta 1 in the MSCs. The conditioned media from IFN-γ-pretreated MSCs increased the expression levels of M2 macrophage markers and regulatory T-cell markers compared to those in the conditioned media from naive MSCs. Further, prostaglandin E2 (PGE2) inhibitor NS-398 attenuated the immunoregulatory potential of MSCs, suggesting that the increased PGE2 levels induced by IFN-γ stimulation is a crucial factor in the immune regulatory capacity of MSCs pretreated with IFN-γ. Conclusions IFN-γ pretreatment improves the immune regulatory profile of fAT-MSCs mainly via the secretion of PGE2, which induces macrophage polarization and increases regulatory T-cell numbers.
Collapse
Affiliation(s)
- Seol Gi Park
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Ju Hyun An
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Qiang Li
- Department of Veterinary Medicine, College of Agriculture, Yanbian University, Yanji, Jilin 133000, China
| | - Hyung Kyu Chae
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Su Min Park
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Jeong Hwa Lee
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Jin Ok Ahn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Woo Jin Song
- Department of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute of Veterinary Science, Jeju National University, Jeju 63243, Korea.
| | - Hwa Young Youn
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
4
|
Ma Z, Zhou J, Yang T, Xie W, Song G, Song Z, Chen J. Mesenchymal stromal cell therapy for pancreatitis: Progress and challenges. Med Res Rev 2021; 41:2474-2488. [PMID: 33840113 DOI: 10.1002/med.21801] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/17/2020] [Accepted: 03/23/2021] [Indexed: 12/15/2022]
Abstract
Pancreatitis is a common gastrointestinal disease with no effective therapeutic options, particularly for cases of severe acute and chronic pancreatitis (CP). Mesenchymal stromal cells (MSCs) are multipotent cells with diverse biological properties, including directional migration, paracrine, immunosuppressive, and antiinflammatory effects, which are considered an ideal candidate cell type for repairing tissue damage caused by various pathogenies. Several researchers have reported significant therapeutic efficacy of MSCs in animal models of acute and CP. However, the specific underlying mechanisms are yet to be clarified and clinical application of MSCs as pancreatitis therapy has rarely been reported. This review mainly focuses on the potential and challenges in clinical application of MSCs for treatment of acute and CP, along with discussion of the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Zhilong Ma
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia Zhou
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tingsong Yang
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wangcheng Xie
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guodong Song
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhenshun Song
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ji Chen
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Rhew SY, Park SM, Li Q, An JH, Chae HK, Lee JH, Ahn JO, Song WJ, Youn HY. Efficacy and safety of allogenic canine adipose tissue-derived mesenchymal stem cell therapy for insulin-dependent diabetes mellitus in four dogs: A pilot study. J Vet Med Sci 2021; 83:592-600. [PMID: 33551441 PMCID: PMC8111340 DOI: 10.1292/jvms.20-0195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess regenerative and immunomodulatory properties and
can control the immune dysregulation that leads to β-cell destruction. Stem-cell
transplantation could thus manage insulin-dependent diabetes mellitus (IDDM) in dogs. In
this pilot study, we aimed to assess canine adipose tissue-derived MSCs (cAT-MSCs)
transplantation as a treatment for canine diabetes mellitus. This study included four dogs
with over a year of insulin treatment for IDDM, following diagnosis at the Veterinary
Medicine Teaching Hospital of Seoul National University. Allogenic cAT-MSCs were infused
intravenously three or five times monthly to dogs with IDDM. Blood and urine samples were
obtained monthly. General clinical symptoms, including changes in body weight, vitality,
appetite, and water intake were assessed. Three of the four owners observed improvement of
vitality after stem cell treatment. Two of the four dogs showed improvement in appetite
and body weight, polyuria, and polydipsia. C-peptide has increased by about 5–15% in three
of the cases, and fructosamine and HbA1c levels have improved in two of the cases.
Hyperlipidemia was resolved in two of the dogs, and there was no concurrent bacterial
cystitis in any of the dogs. C-peptide secretion and lipid metabolism are associated with
diabetic complications. Improvement in these parameters following the treatment suggests
that cAT-MSC transplantation in dogs with IDDM might help to improve their insulin
secretory capacity and prevent diabetic complications.
Collapse
Affiliation(s)
- Sung-Yong Rhew
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Su-Min Park
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Qiang Li
- Department of Veterinary Medicine, College of Agriculture, YanBian University, YanJi, JiLin, 133000, China
| | - Ju-Hyun An
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyung-Kyu Chae
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jung-Hwa Lee
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin-Ok Ahn
- Laboratory of Veterinary Internal Medicine, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Kangwon National University, Gangwon-do 24341, Republic of Korea
| | - Woo-Jin Song
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
6
|
Chela H, Romana BS, Madabattula M, Albarrak AA, Yousef MH, Samiullah S, Tahan V. Stem cell therapy: a potential for the perils of pancreatitis. TURKISH JOURNAL OF GASTROENTEROLOGY 2020; 31:415-424. [PMID: 32721912 DOI: 10.5152/tjg.2020.19143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acute and chronic pancreatitis carry a significant disease burden and there is no definite treatment that exists for either. They are associated with local and systemic inflammation and lead to numerous complications. Stem cell therapy has been explored for other disease processes and is a topic of research that has gained momentum with regards to implications for acute and chronic pancreatitis. They not only carry the potential to aid in regeneration but also prevent pancreatic injury as well as injury of other organs and hence the resultant complications. Stem cells appear to have immunomodulatory properties and clinical potential as evidenced by numerous studies in animal models. This review article discusses the types of stem cells commonly used and the properties that show promise in the field of pancreatitis.
Collapse
Affiliation(s)
- Harleen Chela
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Bhupinder S Romana
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Markandeya Madabattula
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Abdulmajeed A Albarrak
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Mohamad H Yousef
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Sami Samiullah
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Veysel Tahan
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
7
|
Xu X, Yu H, Sun L, Zheng C, Shan Y, Zhou Z, Wang C, Chen B. Adipose‑derived mesenchymal stem cells ameliorate dibutyltin dichloride‑induced chronic pancreatitis by inhibiting the PI3K/AKT/mTOR signaling pathway. Mol Med Rep 2020; 21:1833-1840. [PMID: 32319628 PMCID: PMC7057804 DOI: 10.3892/mmr.2020.10995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/17/2020] [Indexed: 12/20/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) play a positive role in tissue injury repair and regeneration. The aim of this study was to determine whether ASCs could ameliorate chronic pancreatitis (CP) induced by the injection of dibutyltin dichloride (DBTC) and to elucidate its potential mechanisms. Furthermore, this study also explored whether there was a significant difference if the ASCs were injected via the inferior vena cava or the left gastric artery. CP was induced in rats by a single intravenous administration of DBTC, and the accumulation of collagen and apoptotic rates of pancreatic acinar cells were analyzed. According to the results, ASCs markedly reduced DBTC-induced pancreatic damage and collagen deposition in the rat model of CP. Moreover, ASCs significantly decreased pancreatic cell apoptosis by regulating the expression levels of caspase-3, BAX and Bcl-2. These effects were observed regardless of whether the injection was in the inferior vena cava or the left gastric artery. It was also found that the expression levels of phosphorylated PI3K, AKT and mTOR in pancreatic tissues of the DBTC-induced CP model group were significantly increased, while the expression levels of phosphorylated PI3K, AKT and mTOR in the two treatment groups were markedly decreased. ASCs noticeably suppressed the PI3K/AKT/mTOR pathway in the pancreatic tissue of DBTC-induced CP. This study indicated that ASCs protect against pancreatic fibrosis by modulating the PI3K/AKT/mTOR pathway, and have the potential to be a new strategy for the treatment of CP in the future.
Collapse
Affiliation(s)
- Xiangxiang Xu
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Huajun Yu
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Linxiao Sun
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Chenlei Zheng
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yunfeng Shan
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhenxu Zhou
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Cheng Wang
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Bicheng Chen
- Department of Surgery, Key Laboratory of Diagnosis and Treatment of Severe Hepato‑Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
8
|
Li HY, He HC, Song JF, Du YF, Guan M, Wu CY. Bone marrow-derived mesenchymal stem cells repair severe acute pancreatitis by secreting miR-181a-5p to target PTEN/Akt/TGF-β1 signaling. Cell Signal 2020; 66:109436. [DOI: 10.1016/j.cellsig.2019.109436] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/02/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022]
|
9
|
Roch AM, Maatman TK, Cook TG, Wu HH, Merfeld-Clauss S, Traktuev DO, March KL, Zyromski NJ. Therapeutic Use of Adipose-Derived Stromal Cells in a Murine Model of Acute Pancreatitis. J Gastrointest Surg 2020; 24:67-75. [PMID: 31745900 DOI: 10.1007/s11605-019-04411-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/11/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND No specific therapy exists for acute pancreatitis (AP), and current treatment remains entirely supportive. Adipose stem cells (ASCs) have significant immunomodulatory and regenerative activities. We hypothesized that systemic administration of ASCs would mitigate inflammation in AP. METHODS AP was induced in mice by 6 hourly intraperitoneal injections of cerulein. Twenty-four hours after AP induction, mice were randomized into four systemic treatment groups: sham group (no acute pancreatitis), vehicle, human ASCs, and human ASC-conditioned media. Mice were sacrificed at 48 h, and blood and organs were collected and analyzed. Pancreatic injury was quantified histologically using a published score (edema, inflammation, and necrosis). Pancreatic inflammation was also studied by immunohistochemistry and PCR. RESULTS When using IV infusion of Hoechst-labeled ASCs, ASCs were found to localize to inflamed tissues: lungs and pancreas. Mice treated with ASCs had less severe AP, as shown by a significantly decreased histopathology score (edema, inflammation, and necrosis) (p = 0.001). ASCs infusion polarized pancreatic macrophages toward an anti-inflammatory M2 phenotype. ASC-conditioned media reduced pancreatic inflammation similarly to ASCs only, highlighting the importance of ASCs secreted factors in modulating inflammation. CONCLUSION Intravenous delivery of human ASCs markedly reduces pancreatic inflammation in a murine model of AP ASCs which represent an effective therapy for AP.
Collapse
Affiliation(s)
- Alexandra M Roch
- Department of Surgery, Indiana University School of Medicine, 545 Barnhill Drive EH 519, Indianapolis, IN, 46202, USA
| | - Thomas K Maatman
- Department of Surgery, Indiana University School of Medicine, 545 Barnhill Drive EH 519, Indianapolis, IN, 46202, USA
| | - Todd G Cook
- Department of Surgery, Indiana University School of Medicine, 545 Barnhill Drive EH 519, Indianapolis, IN, 46202, USA
| | - Howard H Wu
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephanie Merfeld-Clauss
- Department of Medicine, Division of Cardiovascular Medicine, Center for Regenerative medicine, University of Florida, Gainesville, FL, USA
| | - Dmitry O Traktuev
- Department of Medicine, Division of Cardiovascular Medicine, Center for Regenerative medicine, University of Florida, Gainesville, FL, USA
| | - Keith L March
- Department of Medicine, Division of Cardiovascular Medicine, Center for Regenerative medicine, University of Florida, Gainesville, FL, USA
| | - Nicholas J Zyromski
- Department of Surgery, Indiana University School of Medicine, 545 Barnhill Drive EH 519, Indianapolis, IN, 46202, USA.
| |
Collapse
|
10
|
KIM SM, LI Q, AN JH, CHAE HK, YANG JI, RYU MO, NAM A, SONG WJ, YOUN HY. Enhanced angiogenic activity of dimethyloxalylglycine-treated canine adipose tissue-derived mesenchymal stem cells. J Vet Med Sci 2019; 81:1663-1670. [PMID: 31582601 PMCID: PMC6895634 DOI: 10.1292/jvms.19-0337] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/19/2019] [Indexed: 12/13/2022] Open
Abstract
The paracrine function of mesenchymal stem cells (MSCs) during transplantation has been recently studied due to its poor differentiation ratio. Dimethyloxalylglycine (DMOG) has been used to promote angiogenesis in experimental animal models, however, comparable approaches for canine MSCs are not sufficient. In the present study, we assessed whether DMOG improves angiogenesis in canine adipose tissue-derived mesenchymal stem cells (cAT-MSCs). cAT-MSCs were treated with DMOG and their effect on angiogenesis was investigated by cell proliferation assay, western blotting, and tube formation assay. Dimethyloxalylglycine preconditioning enhanced the expression of vascular endothelial growth factor (VEGF) among pro-angiogenic factors in cAT-MSCs via hypoxia-inducible factor-1α stabilization. Dimethyloxalylglycine primed-cAT-MSC-conditioned media increased angiogenesis in human umbilical vein endothelial cells. These results suggest that DMOG conditioning of cAT-MSCs augmented the secretion of VEGF, which acted as a prominent pro-angiogenic factor during angiogenesis. DMOG-primed cAT-MSCs may have the potential to induce beneficial effects in ischemic diseases in clinical trials.
Collapse
Affiliation(s)
- Sang-Min KIM
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Qiang LI
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Ju-Hyun AN
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyung-Kyu CHAE
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji-In YANG
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Min-Ok RYU
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Aryung NAM
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Woo-Jin SONG
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hwa-Young YOUN
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
11
|
DePompeo CM, Giassetti MI, Elnaggar MM, Oatley JM, Davis WC, Fransson BA. Isolation of canine adipose-derived mesenchymal stem cells from falciform tissue obtained via laparoscopic morcellation: A pilot study. Vet Surg 2019; 49 Suppl 1:O28-O37. [PMID: 31222769 DOI: 10.1111/vsu.13267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/22/2019] [Accepted: 05/25/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To evaluate the feasibility of stem cell isolation from falciform fat harvested via laparoscopic morcellation. STUDY DESIGN Pilot study. ANIMALS Eleven client-owned dogs. METHODS Falciform was harvested traditionally via laparotomy and laparoscopically via tissue morcellation. Harvested tissue was processed with a commercially available adipose tissue dissociation kit to obtain a stromal vascular fraction (SVF). Cells were subsequently labeled for CD90, CD45, and CD44 cell surface antigens by using magnetic-activated cell sorting (MACS) and fluorescence-activated cell sorting flow cytometry. CD90+ cells were quantitated, and their viability was assessed with a hemocytometer and a trypan blue exclusion test of cell viability. RESULTS No perioperative complications occurred in dogs undergoing laparoscopic morcellation. Laparoscopically and traditionally harvested samples yielded an average of 0.39 (±0.1) × 106 and 0.33 (±0.1) × 106 CD90+ cells, respectively, per 10 million SVF cells. CD90+ cell viability after MACS was 89% (±11%) for morcellated and 86% (±7%) for traditionally harvested samples. Neither CD90+ cell quantity nor viability was different between samples obtained via traditional laparotomy vs laparoscopic morcellation (P = .38 and P = .63, respectively). Populations of CD90+ cells isolated with each harvest technique had similar CD44 and CD45 expression profiles. CONCLUSION Viable populations of CD90+ cells with similar CD44/CD45 expression profiles were isolated from laparoscopically morcellated and traditionally harvested falciform tissue. No appreciable morbidity was associated with laparoscopic falciform morcellation. CLINICAL SIGNIFICANCE Laparoscopic morcellation is a safe and effective minimally invasive approach to falciform tissue harvest for adipose-derived mesenchymal stem cell isolation.
Collapse
Affiliation(s)
- Christine M DePompeo
- Veterinary Teaching Hospital, Small Animal Surgery Department, Washington State University, Pullman, Washington
| | - Mariana Ianello Giassetti
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, Pullman, Washington
| | - Mahmoud M Elnaggar
- Veterinary Microbiology and Pathology, Monoclonal Antibody Center and Flow Cytometry Facility, Washington State University, Pullman, Washington
| | - Jon M Oatley
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, Pullman, Washington
| | - William C Davis
- Veterinary Microbiology and Pathology, Monoclonal Antibody Center and Flow Cytometry Facility, Washington State University, Pullman, Washington
| | - Boel A Fransson
- Veterinary Teaching Hospital, Small Animal Surgery Department, Washington State University, Pullman, Washington
| |
Collapse
|
12
|
Song WJ, Li Q, Ryu MO, Nam A, An JH, Jung YC, Ahn JO, Youn HY. Canine adipose tissue-derived mesenchymal stem cells pre-treated with TNF-alpha enhance immunomodulatory effects in inflammatory bowel disease in mice. Res Vet Sci 2019; 125:176-184. [PMID: 31247473 PMCID: PMC7111869 DOI: 10.1016/j.rvsc.2019.06.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 06/12/2019] [Accepted: 06/18/2019] [Indexed: 12/17/2022]
Abstract
Canine inflammatory bowel disease (IBD) is an intractable autoimmune disorder that results in various gastrointestinal and systemic symptoms. Mesenchymal stem cells (MSCs), which release immunomodulatory factors such as tumor necrosis factor-α (TNF-α)-induced gene/protein 6 (TSG-6) and prostaglandin E2 (PGE2), have been suggested as an alternative therapeutic option for IBD treatment in veterinary medicine. Furthermore, although it is known that MSCs pre-treated with pro-inflammatory cytokines show enhanced anti-inflammatory properties via the secretion of soluble factors, the underlying mechanisms of IBD remain unclear. The aim of this study was to demonstrate the therapeutic effects and corresponding mechanisms of canine adipose tissue-derived (cAT)-MSCs stimulated with TNF-α in mouse models of IBD. Mice with dextran sulfate sodium (DSS)- or dinitrobenzene sulfonic acid (DNBS)-induced colitis were injected intraperitoneally with cAT-MSCs pre-treated with TNF-α. Colitis severity was assessed and colon tissues were collected for histopathological, enzyme-linked immunosorbent assay, and flow cytometry analysis. cAT-MSCs stimulated with TNF-α secreted higher concentrations of immunomodulatory factors such as TSG-6 and PGE2, which play a key role in inducing phenotypic alterations in macrophages. Consequently, TNF-α-pre-treated cAT-MSCs further regulated colonic inflammatory cytokines such as interleukin (IL)-1β, IL-6, and IL-10, and ameliorated DSS- or DNBS-induced colitis in mice. Additionally, we demonstrated that M1 macrophages (F4/80+/iNOS+ cells) were decreased in colon tissues from mice treated with TNF-α-pre-treated cAT-MSCs, whereas M2 macrophages (F4/80+/CD206+ cells) were increased. These results may suggest a new cell-based therapeutic option for treating IBD. Canine AT-MSCs stimulated with TNF-α enhanced immunomodulatory factor secretion. TNF-α-stimulated cAT-MSCs showed enhanced anti-inflammatory effects during experimental colitis. TNF-α-stimulated cAT-MSCs induced M2 macrophage phenotypic alterations in the colon. Preconditioning canine AT-MSCs with TNF-α could be applicable to dogs with IBD.
Collapse
Affiliation(s)
- Woo-Jin Song
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 08826 Seoul, Republic of Korea
| | - Qiang Li
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 08826 Seoul, Republic of Korea
| | - Min-Ok Ryu
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 08826 Seoul, Republic of Korea
| | - Aryung Nam
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 08826 Seoul, Republic of Korea
| | - Ju-Hyun An
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 08826 Seoul, Republic of Korea
| | - Yun Chan Jung
- Chaon, A-301-3, 240, Pangyoyeok-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Jin-Ok Ahn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Kangwon National University, 24341 Chuncheon, Gangwondo, Republic of Korea
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 08826 Seoul, Republic of Korea.
| |
Collapse
|
13
|
Silva-Vaz P, Abrantes AM, Castelo-Branco M, Gouveia A, Botelho MF, Tralhão JG. Murine Models of Acute Pancreatitis: A Critical Appraisal of Clinical Relevance. Int J Mol Sci 2019; 20:E2794. [PMID: 31181644 PMCID: PMC6600324 DOI: 10.3390/ijms20112794] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/15/2022] Open
Abstract
Acute pancreatitis (AP) is a severe disease associated with high morbidity and mortality. Clinical studies can provide some data concerning the etiology, pathophysiology, and outcomes of this disease. However, the study of early events and new targeted therapies cannot be performed on humans due to ethical reasons. Experimental murine models can be used in the understanding of the pancreatic inflammation, because they are able to closely mimic the main features of human AP, namely their histologic glandular changes and distant organ failure. These models continue to be important research tools for the reproduction of the etiological, environmental, and genetic factors associated with the pathogenesis of this inflammatory pathology and the exploration of novel therapeutic options. This review provides an overview of several murine models of AP. Furthermore, special focus is made on the most frequently carried out models, the protocols used, and their advantages and limitations. Finally, examples are provided of the use of these models to improve knowledge of the mechanisms involved in the pathogenesis, identify new biomarkers of severity, and develop new targeted therapies.
Collapse
Affiliation(s)
- Pedro Silva-Vaz
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), 6200-506 Covilhã, Portugal.
- General Surgery Department, Unidade Local de Saúde de Castelo Branco, 6000-085 Castelo Branco, Portugal.
- Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Ana Margarida Abrantes
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Biophysics and Biomathematics Institute, IBILI-Faculty of Medicine of University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Miguel Castelo-Branco
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), 6200-506 Covilhã, Portugal.
- Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - António Gouveia
- General Surgery Department, Unidade Local de Saúde de Castelo Branco, 6000-085 Castelo Branco, Portugal.
- Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Maria Filomena Botelho
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Biophysics and Biomathematics Institute, IBILI-Faculty of Medicine of University of Coimbra, 3000-548 Coimbra, Portugal.
| | - José Guilherme Tralhão
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Biophysics and Biomathematics Institute, IBILI-Faculty of Medicine of University of Coimbra, 3000-548 Coimbra, Portugal.
- Surgery Department, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal.
| |
Collapse
|
14
|
Goodman RR, Jong MK, Davies JE. Concise review: The challenges and opportunities of employing mesenchymal stromal cells in the treatment of acute pancreatitis. Biotechnol Adv 2019; 42:107338. [PMID: 30639517 DOI: 10.1016/j.biotechadv.2019.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/02/2019] [Accepted: 01/07/2019] [Indexed: 02/08/2023]
Abstract
To date only small animal models have been employed to assess the effect of mesenchymal stromal cell (MSC) therapy on acute pancreatitis (AP), the most common cause of hospitalization for gastrointestinal diseases worldwide. We outline the challenges inherent in the small animal models of AP. We also point to specific benefits afforded by the adoption of larger animal models. The potential for MSC therapeutics in the treatment of AP was recognized over a decade ago. With sharper focus on the form of AP and development of new MSC delivery routes in larger animals, we believe the challenge can be engaged.
Collapse
Affiliation(s)
- Robbie R Goodman
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto M5S 3G9, Canada
| | - Madelaine K Jong
- Faculty of Dentistry, University of Toronto, Toronto M5G 1G6, Canada
| | - John E Davies
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto M5S 3G9, Canada; Faculty of Dentistry, University of Toronto, Toronto M5G 1G6, Canada.
| |
Collapse
|
15
|
Ayala-Cuellar AP, Kang JH, Jeung EB, Choi KC. Roles of Mesenchymal Stem Cells in Tissue Regeneration and Immunomodulation. Biomol Ther (Seoul) 2019; 27:25-33. [PMID: 29902862 PMCID: PMC6319543 DOI: 10.4062/biomolther.2017.260] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/27/2018] [Accepted: 04/16/2018] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells are classified as multipotent stem cells, due to their capability to transdifferentiate into various lineages that develop from mesoderm. Their popular appeal as cell-based therapy was initially based on the idea of their ability to restore tissue because of their differentiation potential in vitro; however, the lack of evidence of their differentiation to target cells in vivo led researchers to focus on their secreted trophic factors and their role as potential powerhouses on regulation of factors under different immunological environments and recover homeostasis. To date there are more than 800 clinical trials on humans related to MSCs as therapy, not to mention that in animals is actively being applied as therapeutic resource, though it has not been officially approved as one. But just as how results from clinical trials are important, so is to reveal the biological mechanisms involved on how these cells exert their healing properties to further enhance the application of MSCs on potential patients. In this review, we describe characteristics of MSCs, evaluate their benefits as tissue regenerative therapy and combination therapy, as well as their immunological properties, activation of MSCs that dictate their secreted factors, interactions with other immune cells, such as T cells and possible mechanisms and pathways involved in these interactions.
Collapse
Affiliation(s)
| | - Ji-Houn Kang
- Laboratory of Internal Medicine, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, Republic of Korea.,Institute of Life Science and Bio-Engineering, TheraCell Bio & Science, Cheongju 28644, Republic of Korea
| |
Collapse
|
16
|
An JH, Song WJ, Li Q, Kim SM, Yang JI, Ryu MO, Nam AR, Bhang DH, Jung YC, Youn HY. Prostaglandin E 2 secreted from feline adipose tissue-derived mesenchymal stem cells alleviate DSS-induced colitis by increasing regulatory T cells in mice. BMC Vet Res 2018; 14:354. [PMID: 30453939 PMCID: PMC6245895 DOI: 10.1186/s12917-018-1684-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/01/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is an intractable autoimmune disease, relatively common in cats, with chronic vomiting and diarrhea. Previous studies have reported that mesenchymal stem cells (MSCs) alleviate inflammation by modulating immune cells. However, there is a lack of research on cross-talk mechanism between feline adipose tissue-derived mesenchymal stem cells (fAT-MSCs) and immune cells in IBD model. Hence, this study aimed to evaluate the therapeutic effects of fAT-MSC on mice model of colitis and to clarify the therapeutic mechanism of fAT-MSCs. RESULTS Intraperitoneal infusion of fAT-MSC ameliorated the clinical and histopathologic severity of colitis, including body weight loss, diarrhea, and inflammation in the colon of Dextran sulfate sodium (DSS)-treated mice (C57BL/6). Since regulatory T cells (Tregs) are pivotal in modulating immune responses and maintaining tolerance in colitis, the relation of Tregs with fAT-MSC-secreted factor was investigated in vitro. PGE2 secreted from fAT-MSC was demonstrated to induce elevation of FOXP3 mRNA expression and adjust inflammatory cytokines in Con A-induced feline peripheral blood mononuclear cells (PBMCs). Furthermore, in vivo, FOXP3+ cells of the fAT-MSC group were significantly increased in the inflamed colon, relative to that in the PBS group. CONCLUSION Our results suggest that PGE2 secreted from fAT-MSC can reduce inflammation by increasing FOXP3+ Tregs in mice model of colitis. Consequently, these results propose the possibility of administration of fAT-MSC to cats with not only IBD but also other immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Ju-Hyun An
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Woo-Jin Song
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Qiang Li
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Sang-Min Kim
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Ji-In Yang
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Min-Ok Ryu
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - A Ryung Nam
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Dong Ha Bhang
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Yun-Chan Jung
- Chaon Corporation, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13493, Republic of Korea
| | - Hwa-Young Youn
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
17
|
Kim HJ, Li Q, Song WJ, Yang HM, Kim SY, Park SC, Ahn JO, Youn HY. Fibroblast growth factor-1 as a mediator of paracrine effects of canine adipose tissue-derived mesenchymal stem cells on in vitro-induced insulin resistance models. BMC Vet Res 2018; 14:351. [PMID: 30445954 PMCID: PMC6240186 DOI: 10.1186/s12917-018-1671-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/25/2018] [Indexed: 12/13/2022] Open
Abstract
Background In the field of diabetes research, many studies on cell therapy have been conducted using mesenchymal stem cells. This research was intended to shed light on the influence of canine adipose-tissue-derived mesenchymal stem cell conditioned medium (cAT-MSC CM) on in vitro insulin resistance models that were induced in differentiated 3T3-L1 adipocytes and the possible mechanisms involved in the phenomenon. Results Gene expression levels of insulin receptor substrate-1 (IRS-1) and glucose transporter type 4 (GLUT4) were used as indicators of insulin resistance. Relative protein expression levels of IRS-1 and GLUT4 were augmented in the cAT-MSC CM treatment group compared to insulin resistance models, indicating beneficial effects of cAT-MSC to DM, probably by actions of secreting factors. With reference to previous studies on fibroblast growth factor-1 (FGF1), we proposed FGF1 as a key contributing factor to the mechanism of action. We added anti-FGF1 neutralizing antibody to the CM-treated insulin resistance models. As a result, significantly diminished protein levels of IRS-1 and GLUT4 were observed, supporting our assumption. Similar results were observed in glucose uptake assay. Conclusions Accordingly, this study advocated the potential of FGF-1 from cAT-MSC CM as an alternative insulin sensitizer and discovered a signalling factor associated with the paracrine effects of cAT-MSC. Electronic supplementary material The online version of this article (10.1186/s12917-018-1671-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyeon-Jin Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Qiang Li
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Woo-Jin Song
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Mi Yang
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Su-Yeon Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang-Chul Park
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jin-Ok Ahn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.,Current Address: Department of Veterinary Internal Medicine, College of Veterinary Medicine, Kangwon National University, Chuncheon, 24341, Gangwondo, Republic of Korea
| | - Hwa-Young Youn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
18
|
Canine mesenchymal stem cells treated with TNF-α and IFN-γ enhance anti-inflammatory effects through the COX-2/PGE 2 pathway. Res Vet Sci 2018; 119:19-26. [PMID: 29783120 DOI: 10.1016/j.rvsc.2018.05.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 04/16/2018] [Accepted: 05/12/2018] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) have been used in studies on treatment of various diseases, and their application to immune-mediated diseases has garnered interest. Various methods for enhancing the immunomodulation effect of human MSCs have been used; however, similar approaches for canine MSCs are relatively unexplored. Accordingly, we evaluated immunomodulatory effects and mechanisms in canine MSCs treated with TNF-α and IFN-γ. Lipopolysaccharide (LPS)-stimulated RAW 264.7 cells were incubated with the conditioned media (CM) from canine MSCs for 48 h. Expression of RNA was assessed by quantitative reverse transcription PCR (qRT-PCR), and protein levels were assessed by western blot. Expression of inducible nitric oxide synthase (iNOS), IL-6 and IL-1β was significantly (one-way ANOVA) decreased in LPS-stimulated RAW 264.7 cells incubated with CM from canine MSCs compared to that in LPS-stimulated RAW 264.7 cells alone. Furthermore, anti-inflammatory effects of TNF-α- and IFN-γ-primed canine MSCs were significantly increased compared with those of naïve canine MSCs. Expression of cyclooxygenase 2 (COX-2) and prostaglandin E2 (PGE2) were likewise significantly increased in primed canine MSCs. The level of iNOS protein in LPS-stimulated RAW 264.7 cells incubated with CM from the primed canine MSCs was decreased, but it increased when the cells were treated with NS-398(PGE2 inhibitor). In conclusion, compared with naïve canine MSCs, cells primed with TNF-α and IFN-γ cause a greater reduction in release of anti-inflammatory cytokines from LPS-stimulated RAW 264.7 cells; the mechanism is upregulation of the COX-2/PGE2 pathway.
Collapse
|
19
|
Song WJ, Li Q, Ryu MO, Ahn JO, Bhang DH, Jung YC, Youn HY. TSG-6 released from intraperitoneally injected canine adipose tissue-derived mesenchymal stem cells ameliorate inflammatory bowel disease by inducing M2 macrophage switch in mice. Stem Cell Res Ther 2018; 9:91. [PMID: 29625582 PMCID: PMC5889600 DOI: 10.1186/s13287-018-0841-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/11/2018] [Accepted: 03/14/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is an intractable autoimmune disorder that markedly deteriorates one's quality of life. Mesenchymal stem cells (MSCs) alleviate inflammation by modulating inflammatory cytokines in inflamed tissues, and have been suggested as a promising alternative for IBD treatment in human and veterinary cases. Furthermore, tumor necrosis factor-α-induced gene/protein 6 (TSG-6) is a key factor influencing MSC immunomodulatory properties; however, the precise mechanism of TSG-6 release from canine MSCs in IBD remains unclear. This study aimed to assess the therapeutic effects of canine adipose tissue-derived (cAT)-MSC-produced TSG-6 in an IBD mouse model and to explore the mechanisms underlying the immunomodulatory properties. METHODS Mice with dextran sulfate sodium-induced colitis were administered cAT-MSCs intraperitoneally; colon tissues were collected on day 10 for histopathological, quantitative real-time polymerase chain reaction, and immunofluorescence analyses. RESULTS cAT-MSC-secreted TSG-6 ameliorated IBD and regulated colonic expression of pro- and anti-inflammatory cytokines such as tumor necrosis factor-α, interleukin-6, and interleukin-10. To investigate the effect of cAT-MSC-secreted TSG-6 on activated macrophages in vitro, a transwell coculture system was used; TSG-6 released by cAT-MSCs induced a macrophage phenotypic switch from M1 to M2. The cAT-MSC-secreted TSG-6 increased M2 macrophages in the inflamed colon in vivo. CONCLUSIONS TSG-6 released from cAT-MSCs can alleviate dextran sulfate sodium-induced colitis by inducing a macrophage phenotypic switch to M2 in mice.
Collapse
Affiliation(s)
- Woo-Jin Song
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Qiang Li
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min-Ok Ryu
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jin-Ok Ahn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dong Ha Bhang
- Department of Molecular and Cellular Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Yun Chan Jung
- KPC Corporation, Gwangju, Gyeonggi, 12773, Republic of Korea
| | - Hwa-Young Youn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
20
|
Mesenchymal Stromal Cell Therapy for Pancreatitis: A Systematic Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3250864. [PMID: 29743979 PMCID: PMC5878867 DOI: 10.1155/2018/3250864] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/31/2017] [Indexed: 12/19/2022]
Abstract
Background Based on animal studies, adult mesenchymal stromal cells (MSCs) are promising for the treatment of pancreatitis. However, the best type of this form of cell therapy and its mechanism of action remain unclear. Methods We searched the PubMed, Web of Science, Scopus, Google Scholar, and Clinical Trials.gov websites for studies using MSCs as a therapy for both acute and chronic pancreatitis published until September 2017. Results We identified 276 publications; of these publications, 18 met our inclusion criteria. In animal studies, stem cell therapy was applied more frequently for acute pancreatitis than for chronic pancreatitis. No clinical trials were identified. MSC therapy ameliorated pancreatic inflammation in acute pancreatitis and pancreatic fibrosis in chronic pancreatitis. Bone marrow and umbilical cord MSCs were the most frequently administered cell types. Due to the substantial heterogeneity among the studies regarding the type, source, and dose of MSCs used, conducting a meta-analysis was not feasible to determine the best type of MSCs. Conclusion The available data were insufficient for determining the best type of MSCs for the treatment of acute or chronic pancreatitis; therefore, clinical trials investigating the use of MSCs as therapy for pancreatitis are not warranted.
Collapse
|
21
|
Abstract
Mesenchymal stem cells (MSCs) have attracted attention as a cell source for regenerative medicine. In particular, MSCs have an anti-inflammatory effect by secreting several kinds of bioactive molecules. MSC therapy is now being applied to various gastrointestinal diseases, such as graft-versus-host disease, inflammatory bowel disease, and liver cirrhosis. Therefore, MSC therapy has the potential to be a novel treatment for acute and chronic pancreatitis by suppressing inflammation. Several studies have investigated the effect of MSC therapy on acute and chronic pancreatitis, but the underlying mechanisms remain unknown. In this review, we summarize the present status of MSC therapy for acute and chronic pancreatitis.
Collapse
Affiliation(s)
- Kazumichi Kawakubo
- Department of Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita15 Nishi7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Shunsuke Ohnishi
- Department of Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita15 Nishi7, Kita-ku, Sapporo, 060-8638, Japan
| | - Masaki Kuwatani
- Division of Endoscopy, Hokkaido University Hospital, Kita14 Nishi5, Kita-ku, Sapporo, 060-8648, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita15 Nishi7, Kita-ku, Sapporo, 060-8638, Japan
| |
Collapse
|
22
|
Sun Z, Gou W, Kim DS, Dong X, Strange C, Tan Y, Adams DB, Wang H. Adipose Stem Cell Therapy Mitigates Chronic Pancreatitis via Differentiation into Acinar-like Cells in Mice. Mol Ther 2017; 25:2490-2501. [PMID: 28784560 PMCID: PMC5675167 DOI: 10.1016/j.ymthe.2017.06.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 06/10/2017] [Accepted: 06/17/2017] [Indexed: 12/11/2022] Open
Abstract
The objective of this study was to assess the capacity of adipose-derived mesenchymal stem cells (ASCs) to mitigate disease progression in an experimental chronic pancreatitis mouse model. Chronic pancreatitis (CP) was induced in C57BL/6 mice by repeated ethanol and cerulein injection, and mice were then infused with 4 × 105 or 1 × 106 GFP+ ASCs. Pancreas morphology, fibrosis, inflammation, and presence of GFP+ ASCs in pancreases were assessed 2 weeks after treatment. We found that ASC infusion attenuated pancreatic damage, preserved pancreas morphology, and reduced pancreatic fibrosis and cell death. GFP+ ASCs migrated to pancreas and differentiated into amylase+ cells. In further confirmation of the plasticity of ASCs, ASCs co-cultured with acinar cells in a Transwell system differentiated into amylase+ cells with increased expression of acinar cell-specific genes including amylase and chymoB1. Furthermore, culture of acinar or pancreatic stellate cell lines in ASC-conditioned medium attenuated ethanol and cerulein-induced pro-inflammatory cytokine production in vitro. Our data show that a single intravenous injection of ASCs ameliorated CP progression, likely by directly differentiating into acinar-like cells and by suppressing inflammation, fibrosis, and pancreatic tissue damage. These results suggest that ASC cell therapy has the potential to be a valuable treatment for patients with pancreatitis.
Collapse
Affiliation(s)
- Zhen Sun
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; Qingdao Agricultural University, Qingdao, Shandong 266109, China
| | - Wenyu Gou
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Do-Sung Kim
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Xiao Dong
- Qingdao Agricultural University, Qingdao, Shandong 266109, China
| | - Charlie Strange
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yu Tan
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - David B Adams
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
23
|
Jin XR, Xu BR, Hou GF, Sun B, Bai XW. Mesenchymal stem cell transplantation for treatment of pancreatitis. Shijie Huaren Xiaohua Zazhi 2017; 25:2714-2720. [DOI: 10.11569/wcjd.v25.i30.2714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are one of the main cell resources of regenerative medicine. Recently, MSCs have been used to treat many diseases, such as Alzheimer's disease, inflammatory bowel disease and cirrhosis, with certain curative effects achieved. MSCs can not only secrete a variety of anti-inflammatory cytokines, but also reduce the secretion of inflammatory factors. Therefore, acute pancreatitis (AP) and chronic pancreatitis (CP) can be treated with MSCs. Several studies have investigated the effect of MSC therapy on acute and CP. MSCs exert a therapeutic effect on AP perhaps via two pathways: anti-inflammatory pathway and anti-apoptotic pathway. However, the mechanism for the therapeutic effect of MSCs on CP is unclear. In this review, we will summarize the progress in MSC treatment of AP and CP.
Collapse
Affiliation(s)
- Xiang-Ren Jin
- Department of Pancreatic and Biliary Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Bo-Ran Xu
- Department of Pancreatic and Biliary Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Guo-Fang Hou
- Department of Pancreatic and Biliary Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Xue-Wei Bai
- Department of Pancreatic and Biliary Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
24
|
Jackson E, Shoemaker R, Larian N, Cassis L. Adipose Tissue as a Site of Toxin Accumulation. Compr Physiol 2017; 7:1085-1135. [PMID: 28915320 DOI: 10.1002/cphy.c160038] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We examine the role of adipose tissue, typically considered an energy storage site, as a potential site of toxicant accumulation. Although the production of most persistent organic pollutants (POPs) was banned years ago, these toxicants persist in the environment due to their resistance to biodegradation and widespread distribution in various environmental forms (e.g., vapor, sediment, and water). As a result, human exposure to these toxicants is inevitable. Largely due to their lipophilicity, POPs bioaccumulate in adipose tissue, resulting in greater body burdens of these environmental toxicants with obesity. POPs of major concern include polychlorinated biphenyls (PCBs), polychlorinated dibenzo-p-dioxins and furans (PCDDs/PCDFs), and polybrominated biphenyls and diphenyl ethers (PBBs/PBDEs), among other organic compounds. In this review, we (i) highlight the physical characteristics of toxicants that enable them to partition into and remain stored in adipose tissue, (ii) discuss the specific mechanisms of action by which these toxicants act to influence adipocyte function, and (iii) review associations between POP exposures and the development of obesity and diabetes. An area of controversy relates to the relative potential beneficial versus hazardous health effects of toxicant sequestration in adipose tissue. © 2017 American Physiological Society. Compr Physiol 7:1085-1135, 2017.
Collapse
Affiliation(s)
- Erin Jackson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Robin Shoemaker
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Nika Larian
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Lisa Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
25
|
Nam A, Han SM, Go DM, Kim DY, Seo KW, Youn HY. Long-Term Management with Adipose Tissue-Derived Mesenchymal Stem Cells and Conventional Treatment in a Dog with Hepatocutaneous Syndrome. J Vet Intern Med 2017; 31:1514-1519. [PMID: 28782844 PMCID: PMC5598886 DOI: 10.1111/jvim.14798] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 04/24/2017] [Accepted: 06/29/2017] [Indexed: 12/20/2022] Open
Abstract
Hepatocutaneous syndrome (HS) is an uncommon skin disorder that occurs in conjunction with liver disease and is diagnosed based on decreased plasma concentrations of amino acids and the histopathology of skin lesions. The survival period generally is <6 months. A 10-year-old castrated male Maltese dog was presented for evaluation of lethargy, polyuria, polydipsia, and skin lesions including alopecia, erythema, and crusts. Based on increased liver enzyme activity, low plasma amino acid concentrations, and findings from liver cytology and skin biopsy, the dog was diagnosed with HS. In addition to administration of antioxidants, hepatoprotective agents, and amino acids IV, allogenic adipose tissue-derived mesenchymal stem cells were infused 46 times over a 30-month period: 8 times directly into the liver parenchyma guided by ultrasonography and the remainder of the times into peripheral veins. After commencing stem cell therapy, the dog's hair re-grew and the skin lesions disappeared or became smaller. During ongoing management, the patient suddenly presented with anorexia and uncontrolled vomiting, and severe azotemia was observed. The dog died despite intensive care. On necropsy, severe liver fibrosis and superficial necrolytic dermatitis were observed. The dog survived for 32 months after diagnosis. A combination of amino acid and stem cell therapy may be beneficial for patients with HS.
Collapse
Affiliation(s)
- A Nam
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - S-M Han
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - D-M Go
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - D-Y Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - K-W Seo
- College of Veterinary Medicine, Chungnam National University, Daejeon, Korea
| | - H-Y Youn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
26
|
Qian D, Wei G, Xu C, He Z, Hua J, Li J, Hu Q, Lin S, Gong J, Meng H, Zhou B, Teng H, Song Z. Bone marrow-derived mesenchymal stem cells (BMSCs) repair acute necrotized pancreatitis by secreting microRNA-9 to target the NF-κB1/p50 gene in rats. Sci Rep 2017; 7:581. [PMID: 28373667 PMCID: PMC5428835 DOI: 10.1038/s41598-017-00629-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 03/08/2017] [Indexed: 02/07/2023] Open
Abstract
Acute pancreatitis (AP) is a common acute abdominal disease, 10-20% of which can evolve into severe AP (SAP) causing significant morbidity and mortality. Bone marrow-derived mesenchymal stem cells (BMSCs) have the potential of repairing SAP, but the detailed mechanism remains unknown. We demonstrate here that microRNA-9 (miR-9) modified BMSCs (pri-miR-9-BMSCs) can significantly reduce the pancreatic edema, infiltration, hemorrhage, necrosis, the release of amylase and lipase. Meanwhile, decreased local/systemic inflammatory response (TNF-α↓, IL-1β↓, IL-6↓, HMGB1↓, MPO↓, CD68↓, IL-4↑, IL-10↑, and TGF-β↑) and enhanced regeneration of damaged pancreas (Reg4↑, PTF1↑, and PDX1↑) are also promoted. But these effects diminish or disappear after antagonizing miR-9 (TuD). Besides, we find that miR-9 is negatively correlated with AP and miR-9 agomir which can mimic the effects of pri-miR-9-BMSCs and protect injured pancreas. Furthermore, we investigate that BMSCs deliver miR-9 to the injured pancreas or peripheral blood mononuclear cell (PBMC), which can target the NF-κB1/p50 gene and inhibit the NF-κB signaling pathway (p-P65↓, NF-κB1/p50↓, IκBα↑, IκBβ↑). Taken together, these results show that miR-9 is a key paracrine factor of BMSCs attenuating SAP targeting the NF-κB1/p50 gene and suppressing the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Daohai Qian
- Department of General Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, 241001, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China.,Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California, 90089, USA
| | - Ge Wei
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Chenglei Xu
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhigang He
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Jie Hua
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Jian Li
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Qili Hu
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Shengping Lin
- Intensive Care Unit, Sir Run Run Shaw Hospital, Affiliated to Zhejiang University of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Jian Gong
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Hongbo Meng
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Bo Zhou
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Hongfei Teng
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhenshun Song
- Department of General Surgery, Shanghai Tenth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|