1
|
Abdolmohammadi K, Mahmoudi T, Alimohammadi M, Tahmasebi S, Zavvar M, Hashemi SM. Mesenchymal stem cell-based therapy as a new therapeutic approach for acute inflammation. Life Sci 2023; 312:121206. [PMID: 36403645 DOI: 10.1016/j.lfs.2022.121206] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
Acute inflammatory diseases such as acute colitis, kidney injury, liver failure, lung injury, myocardial infarction, pancreatitis, septic shock, and spinal cord injury are significant causes of death worldwide. Despite advances in the understanding of its pathophysiology, there are many restrictions in the treatment of these diseases, and new therapeutic approaches are required. Mesenchymal stem cell-based therapy due to immunomodulatory and regenerative properties is a promising candidate for acute inflammatory disease management. Based on preclinical results, mesenchymal stem cells and their-derived secretome improved immunological and clinical parameters. Furthermore, many clinical trials of acute kidney, liver, lung, myocardial, and spinal cord injury have yielded promising results. In this review, we try to provide a comprehensive view of mesenchymal stem cell-based therapy in acute inflammatory diseases as a new treatment approach.
Collapse
Affiliation(s)
- Kamal Abdolmohammadi
- Department of Immunology, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Tayebeh Mahmoudi
- 17 Shahrivar Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Zavvar
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanothechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Improving the Efficacy of Mesenchymal Stem/Stromal-Based Therapy for Treatment of Inflammatory Bowel Diseases. Biomedicines 2021; 9:biomedicines9111507. [PMID: 34829736 PMCID: PMC8615066 DOI: 10.3390/biomedicines9111507] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel diseases (IBD) consisting of persistent and relapsing inflammatory processes of the intestinal mucosa are caused by genetic, environmental, and commensal microbiota factors. Despite recent advances in clinical treatments aiming to decrease inflammation, nearly 30% of patients treated with biologicals experienced drawbacks including loss of response, while others can develop severe side effects. Hence, novel effective treatments are highly needed. Mesenchymal stem/stromal cell (MSCs) therapy is an innovative therapeutic alternative currently under investigation for IBD. MSCs have the inherent capacity of modulating inflammatory immune responses as well as regenerating damaged tissues and are therefore a prime candidate to use as cell therapy in patients with IBD. At present, MSC-based therapy has been shown preclinically to modulate intestinal inflammation, whilst the safety of MSC-based therapy has been demonstrated in clinical trials. However, the successful results in preclinical studies have not been replicated in clinical trials. In this review, we will summarize the protocols used in preclinical and clinical trials and the novel approaches currently under investigation which aim to increase the beneficial effects of MSC-based therapy for IBD.
Collapse
|
3
|
Abstract
AbstractInflammatory bowel disease, which mainly involves Crohn's disease and ulcerative rectocolitis, is an inflammatory condition of the mucosa that can afflict any segment of the gastrointestinal tract. Despite the fact that the existing therapies result in improvement in patient's symptomatology and quality of life, there is no curative treatment. Surgical treatment involves complex procedures associated with high morbidity and mortality rates. In this context, cell therapy with stem cells has emerged as a treatment with broad potential applicability. In this study, we intended to verify the efficacy of transplantation of adipose tissue-derived stem cells in rats with intestinal inflammation induced by trinitrobenzenesulfonic acid. The cell population was isolated from the adipose tissue of inguinal region of rats and processed for culture by mechanical dissociation. The animals were evaluated with respect to clinical and biochemical aspects, as well as by macroscopic, microscopic and histological analyses. In the experimental model of bowel inflammation by 2,4,6-trinitrobenzenesulfonic acid, the infusion of adipose tissue significantly reduced the presence of adhesions in the colon and adjacent organs and decreased the activity of myeloperoxidase, a marker of neutrophil infiltration in the injured mucosa. The results suggest that cell therapy with adipose tissue can promote and/or accelerate the regeneration of damaged intestinal mucosa. It is concluded that the presence of adhesions and the determination of myeloperoxidase activity provide indications that adipose tissue can promote and/or accelerate the regeneration of inflammatory bowel mucosa.
Collapse
|
4
|
Hosseini-Asl SK, Mehrabani D, Karimi-Busheri F. Therapeutic Effect of Mesenchymal Stem Cells in Ulcerative Colitis: A Review on Achievements and Challenges. J Clin Med 2020; 9:E3922. [PMID: 33287220 PMCID: PMC7761671 DOI: 10.3390/jcm9123922] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
The worldwide epidemiology of inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), still shows an increasing trend in Asia and Iran. Despite an improvement in the treatment landscape focused on symptomatic control, long-term colectomies have not decreased over the last 10-year period. Thus, novel therapies are urgently needed in clinics to supplement the existing treatments. Mesenchymal stem cells (MSCs) are multipotent adult stem cells with immunosuppressive effects, targeting IBD as a new treatment strategy. They have recently received global attention for their use in cell transplantation due to their easy expansion and wide range of activities to be engrafted, and because they are home to the mucosa of the intestine. Moreover, MSCs are able to differentiate into epithelial and other cells that can directly promote repair in the mucosal damages in UC. It seems that there is a need to deepen our understanding to target MSCs as a promising treatment option for UC patients who are refractory to conventional therapies. Here, we overviewed the therapeutic effects of MSCs in UC and discussed the achievements and challenges in the cell transplantation of UC.
Collapse
Affiliation(s)
- Seyed-Kazem Hosseini-Asl
- Department of Internal Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran
| | - Davood Mehrabani
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71987-74731, Iran
- Comparative and Experimental Medicine Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran
| | - Feridoun Karimi-Busheri
- Department of Oncology, Faculty of Medicine, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
5
|
Zhang J, Yuan Z, Zhong W, Wei Y. Stem Cell as Vehicles of Antibody in Treatment of Lymphoma: a Novel and Potential Targeted Therapy. Stem Cell Rev Rep 2020; 17:829-841. [PMID: 33205352 DOI: 10.1007/s12015-020-10080-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2020] [Indexed: 02/06/2023]
Abstract
Lymphoma is a heterogeneous malignancy and its incidence is increasing in the past decades all over the world. Although more than half of lymphoma patients achieve complete or partial remission from the standard first-line ABVD or R-CHOP based therapy, patients who fail to respond to these regimens will give rise to relapsed or refractory (R/R) lymphoma and may lead to a worse prognosis. Developing novel agents is important for R/R lymphoma. Based on the homing ability and being genetically modified easily, stem cells are usually used as vehicles in cell-based anti-tumor therapy, which can not only retain their own biological characteristics, but also make anti-tumor agents secrete constantly in tumor environment, to eventually kill the tumor cells more effectively. In this review, we will briefly introduce the properties of antibody therapy carried by stem cells, especially the hopes and hurdles of stem cell-mediated antibody secretion in the treatment of lymphoma.
Collapse
Affiliation(s)
- Jiayi Zhang
- Department of Blood Transfusion, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou, Guangdong, China
| | - Zhaohu Yuan
- Department of Blood Transfusion, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Weijie Zhong
- Department of Geriatrics, Hematology & Oncology ward, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yaming Wei
- Department of Blood Transfusion, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China. .,Guangdong Engineering Research Center of Precise Transfusion, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
Xiang H, Zhang X, Yang C, Xu W, Ge X, Zhang R, Qiu Y, Sun W, Li F, Xiang T, Chen H, Wang Z, Zeng Q. Autologous bone marrow stem cell transplantation for the treatment of ulcerative colitis complicated with herpes zoster: a case report. Front Med 2016; 10:522-526. [PMID: 27896624 DOI: 10.1007/s11684-016-0485-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 08/17/2016] [Indexed: 12/27/2022]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease with continuous or recurrent symptoms. A 42-year-old male patient with intermittent diarrhea accompanied by bloody mucopurulent stools was admitted to our hospital. The diagnosis of UC was confirmed by a combination of laboratory examination, colonoscopy, and histological assay. The patient developed herpes zoster in the hospital, which challenged traditional treatments. Therefore, we performed an autologous bone marrow cells to modulate the immune system with his permission. Autologous bone marrow mononuclear cells were collected and injected locally into the bowel mucosa, and subsequently injected systemically through a peripheral vein. After the patient underwent auto bone marrow mononuclear cells transplantations twice, the patient's symptoms were alleviated. Furthermore, he recovered from hematochezia, and his hypersensitive C reactive protein decreased. Colonoscopy results showed reduced lesions and decreased areas with bleeding and edema in the sigmoid colon and rectum. No recurrence occurred in the subsequent two years, but long-time monitoring is still necessary for the prophylaxis of colorectal cancer.
Collapse
Affiliation(s)
- Hang Xiang
- Institute of Health Management, Institute of Geriatrics, Beijing Key Laboratory of Normal Aging and Geriatrics, Department of Gerontal Gastroenterology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiaomei Zhang
- Institute of Health Management, Institute of Geriatrics, Beijing Key Laboratory of Normal Aging and Geriatrics, Department of Gerontal Gastroenterology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Chao Yang
- Department of Blood Transfusion, Department of Hematology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Wenhuan Xu
- Institute of Health Management, Institute of Geriatrics, Beijing Key Laboratory of Normal Aging and Geriatrics, Department of Gerontal Gastroenterology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xin Ge
- Galactophore Department of the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Rong Zhang
- Department of Blood Transfusion, Department of Hematology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Ya Qiu
- Institute of Health Management, Institute of Geriatrics, Beijing Key Laboratory of Normal Aging and Geriatrics, Department of Gerontal Gastroenterology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wanjun Sun
- Department of Blood Transfusion, Department of Hematology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Fan Li
- Institute of Health Management, Institute of Geriatrics, Beijing Key Laboratory of Normal Aging and Geriatrics, Department of Gerontal Gastroenterology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Tianyuan Xiang
- Geriatrics Institute of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Haixu Chen
- Institute of Health Management, Institute of Geriatrics, Beijing Key Laboratory of Normal Aging and Geriatrics, Department of Gerontal Gastroenterology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Zheng Wang
- Department of Biotherapy of PLA 455 Hospital, Shanghai, 200052, China.
| | - Qiang Zeng
- Institute of Health Management, Institute of Geriatrics, Beijing Key Laboratory of Normal Aging and Geriatrics, Department of Gerontal Gastroenterology, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
7
|
Legaki E, Roubelakis MG, Theodoropoulos GE, Lazaris A, Kollia A, Karamanolis G, Marinos E, Gazouli M. Therapeutic Potential of Secreted Molecules Derived from Human Amniotic Fluid Mesenchymal Stem/Stroma Cells in a Mice Model of Colitis. Stem Cell Rev Rep 2016; 12:604-612. [PMID: 27503204 DOI: 10.1007/s12015-016-9677-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammatory bowel diseases (IBDs) are the result of pathological immune responses due to environmental factors or microbial antigens into a genetically predisposed individual. Mainly due to their trophic properties, a mounting interest is focused on the use of human mesenchymal stem/stromal cells (hMSCs) to treat IBD disease in animal models. The aim of the study is to test whether the secreted molecules, derived from a specific population of second trimester amniotic fluid mesenchymal stem/stromal cells, the spindle-shaped MSCs (SS-AF-MSCs), could be utilized as a novel therapeutic, cell free approach for IBD therapy. Induction of colitis was achieved by oral administration of dextran sulphate sodium (DSS) (3 % w/v in tap water), for 5 days, to 8-week-old NOD/SCID mice. The progression of colitis was assessed on a daily basis through recording the body weight, stool consistency and bleeding. Conditioned media (CM) derived from SS-AF-MSCs were collected, concentrated and then delivered intraperitoneally into DSS treated mice. To evaluate and determine the inflammatory cytokine levels, histopathological approach was applied. Administration of CM derived from SS-AF-MSCs cells reduced the severity of colitis in mice. More importantly, TGFb1 protein levels were increased in the mice received CM, while TNFa and MMP2 protein levels were decreased, respectively. Accordingly, IL-10 was significantly increased in mice received CM, whereas TNFa and IL-1b were decreased at mRNA level. Our results demonstrated that CM derived from SS-AF-MSCs cells is able to ameliorate DSS-induced colitis in immunodeficient colitis mouse model, and thus, it has a potential for use in IBD therapy.
Collapse
Affiliation(s)
- E Legaki
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Michalakopoulou, 176, Athens, Greece
| | - M G Roubelakis
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Michalakopoulou, 176, Athens, Greece
| | - G E Theodoropoulos
- First Propaedeutic Surgical Department, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - A Lazaris
- Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - A Kollia
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Michalakopoulou, 176, Athens, Greece
| | - G Karamanolis
- Gastroenterology Unit, 2nd Surgical Department, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - E Marinos
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Michalakopoulou, 176, Athens, Greece
| | - M Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Michalakopoulou, 176, Athens, Greece.
| |
Collapse
|
8
|
Ciccocioppo R, Gallia A, Avanzini MA, Betti E, Picone C, Vanoli A, Paganini C, Biagi F, Maccario R, Corazza GR. A Refractory Celiac Patient Successfully Treated With Mesenchymal Stem Cell Infusions. Mayo Clin Proc 2016; 91:812-9. [PMID: 27087453 DOI: 10.1016/j.mayocp.2016.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 02/20/2016] [Accepted: 03/01/2016] [Indexed: 02/07/2023]
Abstract
Type II refractory celiac disease (RCD), as defined according to the amount of aberrant intraepithelial lymphocytes, is a condition characterized by severe malabsorption syndrome and poor prognosis, with no effective treatment. Based on the regenerative and immunomodulatory properties of mesenchymal stem cells (MSCs), we investigated the feasibility, safety, and efficacy of serial infusions of autologous bone marrow-derived MSCs in a 51-year-old woman with type II RCD. Mesenchymal stem cells were isolated, expanded, and characterized following standard protocols. Monitoring of the patient's malabsorption indexes, mucosal architecture, and percentage of aberrant intraepithelial lymphocytes was scheduled for the time of enrollment, at each infusion, and after 6 months. Determination of mucosal expression of interleukin (IL)-15 and its receptor was also performed. Expansion of MSCs was feasible, and the patient underwent 4 systemic infusions of 2 × 10(6) MSCs/kg body weight 4 months apart, without adverse effects. During the treatment period, she experienced gradual and durable amelioration of her general condition, with normalization of stool frequency, body mass index, laboratory test results, and mucosal architecture. Remarkably, the expression of IL-15 and its receptor almost completely disappeared. Thus, treatment of RCD with serial MSC infusions seems promising, leading to recovery from the life-threatening condition while blocking the IL-15 pathogenic pathway.
Collapse
Affiliation(s)
- Rachele Ciccocioppo
- Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy.
| | - Alessandra Gallia
- Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Maria A Avanzini
- Cell Factory and Research Laboratory-Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Elena Betti
- Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Cristina Picone
- Clinic Cytometry Laboratory-Department of Hematology, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Alessandro Vanoli
- Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Chiara Paganini
- Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Federico Biagi
- Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Rita Maccario
- Cell Factory and Research Laboratory-Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Gino R Corazza
- Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| |
Collapse
|
9
|
Ciccocioppo R, Cangemi GC, Kruzliak P, Corazza GR. Concise Review: Cellular Therapies: The Potential to Regenerate and Restore Tolerance in Immune-Mediated Intestinal Diseases. Stem Cells 2016; 34:1474-86. [PMID: 27016400 DOI: 10.1002/stem.2367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/01/2016] [Accepted: 03/10/2016] [Indexed: 12/18/2022]
Abstract
Chronic inflammatory enteropathies, including celiac disease, Crohn's disease, and ulcerative colitis, are lifelong disabling conditions whose cure is still an unmet need, despite the great strides made in understanding their complex pathogenesis. The advent of cellular therapies, mainly based on the use of stem cells, represents a great step forward thanks to their multitarget strategy. Both hematopoietic stem cells (HSC) and mesenchymal stem/stromal cells (MSC) have been employed in the treatment of refractory cases with promising results. The lack of immunogenicity makes MSC more suitable for therapeutic purposes as their infusion may be performed across histocompatibility locus antigen barriers without risk of rejection. The best outcome has been obtained when treating fistulizing Crohn's disease with local injections of MSC. In addition, both HSC and MSC proved successful in promoting regeneration of intestinal mucosa, and favoring the expansion of a T-cell regulatory subset. By virtue of the ability to favor mucosal homeostasis, this last cell population has been exploited in clinical trials, with inconsistent results. Finally, the recent identification of the epithelial stem cell marker has opened up the possibility of tissue engineering, with an array of potential applications for intestinal diseases. However, the underlying mechanisms of action of these interconnected therapeutic strategies are still poorly understood. It is conceivable that over the next few years their role will become clearer as the biological interactions with injured tissues and the hierarchy by which they deliver their action are unraveled through a continuous moving from bench to bedside and vice versa. Stem Cells 2016;34:1474-1486.
Collapse
Affiliation(s)
- Rachele Ciccocioppo
- Clinica Medica I, Department of Internal Medicine Fondazione IRCCS Policlinico San Matteo, Università degli Studi di Pavia, Italy
| | - Giuseppina Cristina Cangemi
- Clinica Medica I, Department of Internal Medicine Fondazione IRCCS Policlinico San Matteo, Università degli Studi di Pavia, Italy
| | - Peter Kruzliak
- Laboratory of Structural Biology and Proteomics, Central Laboratories, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Gino Roberto Corazza
- Clinica Medica I, Department of Internal Medicine Fondazione IRCCS Policlinico San Matteo, Università degli Studi di Pavia, Italy
| |
Collapse
|
10
|
Yemm A, Adams D, Kalia N. Targeting the delivery of systemically administered haematopoietic stem/progenitor cells to the inflamed colon using hydrogen peroxide and platelet microparticle pre-treatment strategies. Stem Cell Res 2015; 15:569-580. [PMID: 26479027 DOI: 10.1016/j.scr.2015.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/30/2015] [Accepted: 10/02/2015] [Indexed: 12/13/2022] Open
Abstract
Haematopoietic stem and progenitor cell (HSC) therapy may be promising for the treatment of inflammatory bowel disorders (IBDs). However, clinical success remains poor, partly explained by limited HSC recruitment following systemic delivery. The mechanisms governing HSC adhesion within inflamed colon, and whether this event can be enhanced, are not known. An immortalised HSC-like line (HPC7) was pre-treated with hydrogen peroxide (H2O2), activated platelet releasate enriched supernatant (PES) or platelet microparticles (PMPs). Subsequent adhesion was monitored using adhesion assays or in vivo ischaemia-reperfusion (IR) and colitis injured mouse colon intravitally. Integrin clustering was determined confocally and cell morphology using scanning electron microscopy. Both injuries resulted in increased HPC7 adhesion within colonic mucosal microcirculation. H2O2 and PES significantly enhanced adhesion in vitro and in the colitis, but not IR injured, colon. PMPs had no effect on adhesion. PES and PMPs induced clustering of integrins on the HPC7 surface, but did not alter their expression. Adhesion to the colon is modulated by injury but only in colitis injury can this recruitment be enhanced. The enhanced adhesion induced by PES is likely through integrin distribution changes on the HPC7 surface. Improving local HSC presence in injured colon may result in better therapeutic efficacy for treatment of IBD.
Collapse
Affiliation(s)
- Adrian Yemm
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - David Adams
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; School of Immunity and Infection, Institute of Biomedical Research, The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Neena Kalia
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
11
|
Forte D, Ciciarello M, Valerii MC, De Fazio L, Cavazza E, Giordano R, Parazzi V, Lazzari L, Laureti S, Rizzello F, Cavo M, Curti A, Lemoli RM, Spisni E, Catani L. Human cord blood-derived platelet lysate enhances the therapeutic activity of adipose-derived mesenchymal stromal cells isolated from Crohn's disease patients in a mouse model of colitis. Stem Cell Res Ther 2015; 6:170. [PMID: 26353774 PMCID: PMC4564981 DOI: 10.1186/s13287-015-0166-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 04/03/2015] [Accepted: 08/20/2015] [Indexed: 02/07/2023] Open
Abstract
Introduction Due to their immunomodulatory properties, mesenchymal stromal cells (MSCs) have been used for auto-immune disease treatment. Crohn disease (CD) and ulcerative colitis are two major inflammatory bowel diseases (IBDs), resulting from pathological immune responses to environmental or microbial antigens. Preclinical and clinical studies have suggested that MSC-based cellular therapy hold promising potential for IBD treatment. However, open issues include the selection of the proper cell dose, the source and the optimal route of administration of MSCs for more effective results. Platelet lysate has gained clinical interest due to its efficacy in accelerating wound healing. Thus, we propose to combine the administration of MSCs with a human umbilical cord blood-derived platelet lysate (hCBPL) as a novel strategy to improve MSC-based therapy for IBD resolution. Methods Colitis was induced in 8-week-old C57BL/6J mice by daily oral administration of dextran sulphate sodium (DSS) (1.5 % w/v in tap water) for 9 days. MSCs were isolated from adipose tissue of CD patients (adCD-MSCs), expanded in proliferation medium, resuspended in hCBPL or PBS and administrated via enema for three times (1 × 106 cells/mouse/time) every other day starting on day +7 from DSS induction. The colitis evolution was evaluated by daily monitoring of body weight, stool consistency and bleeding. Histopathological analysis was performed. Inflammatory cytokine plasma levels were determined. adCD-MSCs stained with lipophilic membrane dye Nile Red, were injected in DSS mice as described above. Colon section of mice sacrificed 24 hours after last cell administration, were analyzed by confocal microscopy. Results We found that adCD-MSCs could be easily isolated and expanded from CD patients. Upon injection, adCD-MSCs exerted a therapeutic effect on DSS-induced colitis. Moreover, hCBPL increased adCD-MSCs efficacy by significantly reducing colitis scores, extension of the colon inflamed area and plasma levels of inflammatory mediators. Finally, Nile Red staining of MSCs is very efficient, stable and does not impair their vitality and function. Nile Red-labelling was clearly detected in the colitic area of adCD-MSCs injected mice and it was significantly brighter in the colon sections of mice that had received adCD-MSCs/hCBPL. Conclusions In summary, with this study we propose a novel and promising adCD-MSC/hCBPL-based therapy for refractory IBDs. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0166-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dorian Forte
- Institute of Hematology "L. & A. Seràgnoli", Department of Experimental, Diagnostic and Specialty Medicine, Policlinico S. Orsola-Malpighi, University of Bologna, via G. Massarenti 9, 40138, Bologna, Italy.
| | - Marilena Ciciarello
- Institute of Hematology "L. & A. Seràgnoli", Department of Experimental, Diagnostic and Specialty Medicine, Policlinico S. Orsola-Malpighi, University of Bologna, via G. Massarenti 9, 40138, Bologna, Italy.
| | - Maria Chiara Valerii
- Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Via Selmi 3, 40126, Bologna, Italy.
| | - Luigia De Fazio
- Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Via Selmi 3, 40126, Bologna, Italy.
| | - Elena Cavazza
- Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Via Selmi 3, 40126, Bologna, Italy.
| | - Rosaria Giordano
- Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via F. Sforza 35, 20122, Milano, Italy.
| | - Valentina Parazzi
- Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via F. Sforza 35, 20122, Milano, Italy.
| | - Lorenza Lazzari
- Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via F. Sforza 35, 20122, Milano, Italy.
| | - Silvio Laureti
- Inflammatory Bowel Disease Unit, Policlinico S. Orsola-Malpighi, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Fernando Rizzello
- Inflammatory Bowel Disease Unit, Policlinico S. Orsola-Malpighi, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Michele Cavo
- Institute of Hematology "L. & A. Seràgnoli", Department of Experimental, Diagnostic and Specialty Medicine, Policlinico S. Orsola-Malpighi, University of Bologna, via G. Massarenti 9, 40138, Bologna, Italy.
| | - Antonio Curti
- Institute of Hematology "L. & A. Seràgnoli", Department of Experimental, Diagnostic and Specialty Medicine, Policlinico S. Orsola-Malpighi, University of Bologna, via G. Massarenti 9, 40138, Bologna, Italy.
| | - Roberto M Lemoli
- Chair of Hematology, Department of Internal and Specialty Medicine (DiMI), University of Genoa, Viale Benedetto XV 6, 16132, Genoa, Italy.
| | - Enzo Spisni
- Department of Biological, Geological and Environmental Sciences, Biology Unit, University of Bologna, Via Selmi 3, 40126, Bologna, Italy.
| | - Lucia Catani
- Institute of Hematology "L. & A. Seràgnoli", Department of Experimental, Diagnostic and Specialty Medicine, Policlinico S. Orsola-Malpighi, University of Bologna, via G. Massarenti 9, 40138, Bologna, Italy.
| |
Collapse
|
12
|
Xiang H, Chen H, Li F, Liu J, Su Y, Hao L, Wang F, Wang Z, Zeng Q. Predictive factors for prolonged remission after autologous hematopoietic stem cell transplantation in young patients with type 1 diabetes mellitus. Cytotherapy 2015; 17:1638-45. [PMID: 26318272 DOI: 10.1016/j.jcyt.2015.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/27/2015] [Accepted: 07/04/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND AIMS Autologous hematopoietic stem cell transplantation (auto-HSCT) followed by immunoablation is a promising therapy for type 1 diabetes mellitus (T1DM) treatment due to the immunosuppression and immunomodulation mechanisms. Indeed, a considerable number of patients have been able to discontinue insulin use with this treatment. However, nonresponse and relapse occur after auto-HSCT. It is important to select the patients who can potentially benefit from this treatment, but the factors that might influence the therapeutic outcome are unclear. The objective of this study was to explore the predictors for prolonged remission after auto-HSCT therapy. METHODS The data for this study were extracted from an open-label prospective study, which was performed to treat new-onset T1DM patients with auto-HSCT. The 128 patients were categorized into insulin-free (IF) or insulin-dependent (ID) groups according to their response to treatment during the follow-up. We compared the baseline data of the two groups and explored possible prognostic factors and their odd ratios (ORs) with univariate analysis and multivariate logistic regression. Receiver operating characteristic curves (ROC) were performed to test the model discrimination function. RESULTS During a follow-up of 28.5 ± 8.3 months, 71 of 128 patients in the IF group discontinued insulin use, whereas 57 of 128 patients in the ID group did not decrease their insulin dose or resumed insulin treatment after a transient remission. Multivariate logistic regression analysis demonstrated that prolonged remission was positively correlated with fasting C-peptide level (OR = 2.60, 95% confidence interval [CI]: 1.16-5.85) but negatively correlated with onset age (OR = 0.36, 95% CI: 0.14-0.88) and tumor necrosis factor-α levels (OR = 0.32, 95% CI: 0.14-0.73). ROC analysis confirmed the combined predictive function of these three variables (AUC = 0.739, 95% CI: 0.655-0.824). CONCLUSIONS Age and fasting C-peptide and tumor necrosis factor-α levels were identified as possible predictors for prolonged remission following auto-HSCT therapy.
Collapse
Affiliation(s)
- Hang Xiang
- Health Management Institute of Chinese PLA General Hospital, Beijing, China
| | - Haixu Chen
- Beijing Key Laboratory of Normal Aging and Geriatrics, Geriatrics Institute of Chinese PLA General Hospital, Beijing, China
| | - Fan Li
- Health Management Institute of Chinese PLA General Hospital, Beijing, China
| | - Jing Liu
- Beijing Key Laboratory of Normal Aging and Geriatrics, Geriatrics Institute of Chinese PLA General Hospital, Beijing, China
| | - Yuehan Su
- Department of Biotherapy of PLA 455 Hospital, Shanghai, China
| | - Lu Hao
- Medical Administration Office, Surgical Department of Chinese PLA General Hospital, Beijing, China
| | - Fei Wang
- Health Management Institute of Chinese PLA General Hospital, Beijing, China
| | - Zheng Wang
- Department of Biotherapy of PLA 455 Hospital, Shanghai, China.
| | - Qiang Zeng
- Health Management Institute of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
13
|
Algeri M, Conforti A, Pitisci A, Starc N, Tomao L, Bernardo ME, Locatelli F. Mesenchymal stromal cells and chronic inflammatory bowel disease. Immunol Lett 2015; 168:191-200. [PMID: 26170204 DOI: 10.1016/j.imlet.2015.06.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 06/12/2015] [Indexed: 12/21/2022]
Abstract
Recent experimental findings have shown the ability of mesenchymal stromal cells (MSCs) to home to damaged tissues and to produce paracrine factors with anti-inflammatory properties, potentially resulting in reduction of inflammation and functional recovery of the damaged tissues. Prompted by these intriguing properties and on the basis of encouraging preclinical data, MSCs are currently being studied in several immune-mediated disorders. Inflammatory bowel diseases (IBD) represent a setting in which MSCs-based therapy has been extensively investigated. Phase I and II studies have documented the safety and feasibility of MSCs. However, efficacy results have so far been conflicting. In this review, we will discuss the biologic rationale that makes MSCs a promising therapeutic tool for IBD, and analyze recent experimental and clinical findings, highlighting current limitations and future perspectives of MSCs-related immunotherapy for IBD.
Collapse
Affiliation(s)
- M Algeri
- Department of Pediatric Hematology-Oncology, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - A Conforti
- Department of Pediatric Hematology-Oncology, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - A Pitisci
- Department of Pediatric Hematology-Oncology, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - N Starc
- Department of Pediatric Hematology-Oncology, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy; Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - L Tomao
- Department of Pediatric Hematology-Oncology, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - M E Bernardo
- Department of Pediatric Hematology-Oncology, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - F Locatelli
- Department of Pediatric Hematology-Oncology, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy; Department of Pediatrics, University of Pavia, Italy.
| |
Collapse
|
14
|
Sala E, Genua M, Petti L, Anselmo A, Arena V, Cibella J, Zanotti L, D'Alessio S, Scaldaferri F, Luca G, Arato I, Calafiore R, Sgambato A, Rutella S, Locati M, Danese S, Vetrano S. Mesenchymal Stem Cells Reduce Colitis in Mice via Release of TSG6, Independently of Their Localization to the Intestine. Gastroenterology 2015; 149:163-176.e20. [PMID: 25790743 DOI: 10.1053/j.gastro.2015.03.013] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 02/19/2015] [Accepted: 03/06/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Mesenchymal stem cells (MSCs) are pluripotent cells that can promote expansion of immune regulatory cells and might be developed for the treatment of immune disorders, including inflammatory bowel diseases. MSCs were reported to reduce colitis in mice; we investigated whether MSC localization to the intestine and production of paracrine factors, including tumor necrosis factor-induced protein 6 (TSG6), were required for these effects. METHODS MSCs were isolated from bone marrow (BM-MSCs) of 4- to 6-week-old C57BL/6, C57BL/6-green fluorescent protein, or Balb/c Tsg6-/- male mice. Colitis was induced by ad libitum administration of dextran sulfate sodium for 10 days; after 5 days the mice were given intraperitoneal injections of BM-MSCs or saline (controls). Blood samples and intestinal tissues were collected 24, 48, 96, and 120 hours later; histologic and flow cytometry analyses were performed. RESULTS Injection of BM-MSCs reduced colitis in mice, increasing body weight and reducing markers of intestinal inflammation, compared with control mice. However, fewer than 1% of MSCs reached the inflamed colon. Most of the BM-MSCs formed aggregates in the peritoneal cavity. The aggregates contained macrophages and B and T cells, and produced immune-regulatory molecules including FOXP3, interleukin (IL)10, transforming growth factor-β, arginase type II, chemokine (C-C motif) ligand 22 (CCL22), heme oxygenase-1, and TSG6. Serum from mice given BM-MSCs, compared with mice given saline, had increased levels of TSG6. Injection of TSG6 reduced the severity of colitis in mice, along with the numbers of CD45+ cells, neutrophils and metalloproteinase activity in the mucosa, while increasing the percentage of Foxp3CD45+ cells. TSG6 injection also promoted the expansion of regulatory macrophages that expressed IL10 and inducible nitric oxide synthase, and reduced serum levels of interferon-γ, IL6, and tumor necrosis factor. Tsg6-/- MSCs did not suppress the mucosal inflammatory response in mice with colitis. CONCLUSIONS BM-MSCs injected into mice with colitis do not localize to the intestine but instead form aggregates in the peritoneum where they produce immunoregulatory molecules, including TSG6, that reduce intestinal inflammation. TSG6 is sufficient to reduce intestinal inflammation in mice with colitis.
Collapse
Affiliation(s)
- Emanuela Sala
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy
| | - Marco Genua
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy
| | - Luciana Petti
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy
| | - Achille Anselmo
- Department of Immunology, Humanitas Clinical and Research Center, Milan, Italy
| | - Vincenzo Arena
- Department of Pathology, Catholic University of Rome, Rome, Italy
| | - Javier Cibella
- Department of Immunology, Humanitas Clinical and Research Center, Milan, Italy
| | - Lucia Zanotti
- Department of Immunology, Humanitas Clinical and Research Center, Milan, Italy
| | - Silvia D'Alessio
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy
| | - Franco Scaldaferri
- Internal Medicine, Gastroenterology Division, Catholic University of the Sacred Heart, Rome, Italy
| | - Giovanni Luca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Alessandro Sgambato
- Institute of General Pathology, Catholic University of the Sacred Heart, Rome, Italy
| | - Sergio Rutella
- Clinical Research Center, Division of Translational Medicine, Sidra Medical & Research Center, Doha, Qatar
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Silvio Danese
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy
| | - Stefania Vetrano
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy.
| |
Collapse
|
15
|
Xing Y, Chen X, Cao Y, Huang J, Xie X, Wei Y. Expression of Wnt and Notch signaling pathways in inflammatory bowel disease treated with mesenchymal stem cell transplantation: evaluation in a rat model. Stem Cell Res Ther 2015; 6:101. [PMID: 25998108 PMCID: PMC4487973 DOI: 10.1186/s13287-015-0092-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 03/31/2015] [Accepted: 05/11/2015] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION The purpose of this study was to investigate the expression of Wnt and Notch signaling pathway-related genes in inflammatory bowel disease (IBD) treated with mesenchymal stem cell transplantation (MSCT). METHODS TNBS (2,4,6-trinitrobenzene sulfonic acid) was used to establish IBD in a rat model. Mesenchymal stem cells (MSCs) were transplanted via tail vein transfusion. Saline water was used in a control group. The expression of Wnt and Notch main signaling molecules was screened by gene chips and verified by quantitative reverse transcription-polymerase chain reaction in the IBD rat model on day 14 and day 28 after transplantation. RESULTS The IBD rat models were successfully established and MSCs were transplanted into those models. Genome-wide expression profile chips identified a total of 388 differentially expressive genes, of which 191 were upregulated and 197 were downregulated in the MSC-transplanted group in comparison with the IBD control group. Real-time quantitative polymerase chain reaction results showed that the level of Olfm4 mRNA expression in the IBD group (2.54±0.20) was significantly increased compared with the MSCT group (1.39±0.54) and the normal group (1.62±0.25) (P <0.05). The Wnt3a mRNA was more highly expressed in IBD rats (2.92±0.94) and decreased in MSCT rats (0.17±0.63, P <0.05). The expression of GSK-3β mRNA was decreased in the setting of inflammation (0.65±0.04 versus 1.00±0.01 in normal group, P <0.05) but returned to normal levels after MSCT (0.81±0.17). The expression of β-catenin was observed to increase in IBD tissues (1.76±0.44) compared with normal tissues (1.00±0.01, P <0.05), but no difference was found in the MSCT group (1.12±0.36). Wnt11 declined at 14 days and returned to normal levels at 28 days in the IBD group; in comparison, a significantly lower expression was found in MSCT rats. There were no differences in the expression of Fzd3, c-myc, TCF4, and Wnt5a in inflammation, but all of those genes declined after MSCT treatment. CONCLUSIONS The canonical Wnt and Notch signaling pathways are activated in IBD and may be suppressed by stem cell transplantation to differentiate into intestinal epithelium after MSCT. Moreover, the non-canonical Wnt signaling may be inhibited by canonical Wnt signaling in the setting of inflammation and may also be suppressed by MSCT.
Collapse
Affiliation(s)
- Yanfen Xing
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| | - Xiaojie Chen
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| | - Yanwen Cao
- Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Department of Gastroenterology, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| | - Jianyun Huang
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| | - Xuhong Xie
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| | - Yaming Wei
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| |
Collapse
|
16
|
Ciccocioppo R, Cangemi GC, Roselli EA, Kruzliak P. Are stem cells a potential therapeutic tool in coeliac disease? Cell Mol Life Sci 2015; 72:1317-29. [PMID: 25511197 PMCID: PMC11113911 DOI: 10.1007/s00018-014-1797-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/02/2014] [Accepted: 12/01/2014] [Indexed: 02/08/2023]
Abstract
Despite the growing understanding of its pathogenesis, the treatment of coeliac disease is still based on a lifelong gluten-free diet that, although efficacious, is troublesome for affected patients, and a definitive cure is still an unmet need. In this regard, the development of new chemical- and biological-derived agents has often resulted in unsatisfactory effects when tested in vivo, probably because of their ability to target only a single pathway, whilst the immunological cascade responsible for tissue injury is complex and redundant. The advent of cellular therapies, mainly based on the use of stem cells, is an emerging area of interest since it has the advantage of a multi-target strategy. Both haematopoietic and mesenchymal stem cells have been employed in the treatment of refractory patients suffering from autoimmune diseases, with promising results. However, the lack of immunogenicity makes mesenchymal stem cells more suitable than their haematopoietic counterpart, since their transplantation may be performed in the absence of a myeloablative conditioning regimen. In addition, mesenchymal stem cells have been shown to harbour strong modulatory effects on almost all cells involved in immune response, together with a potent regenerative action. It is therefore conceivable that over the next few years their therapeutic use will increase as their biological interactions with injured tissues become clearer.
Collapse
Affiliation(s)
- Rachele Ciccocioppo
- Center for the Study and Cure of Coeliac Disease, Clinica Medica I, IRCCS San Matteo Hospital Foundation, University of Pavia, Piazzale Golgi, 19, 27100 Pavia, Italy
| | - Giuseppina Cristina Cangemi
- Center for the Study and Cure of Coeliac Disease, Clinica Medica I, IRCCS San Matteo Hospital Foundation, University of Pavia, Piazzale Golgi, 19, 27100 Pavia, Italy
| | - Emanuela Anna Roselli
- Center for the Study and Cure of Coeliac Disease, Clinica Medica I, IRCCS San Matteo Hospital Foundation, University of Pavia, Piazzale Golgi, 19, 27100 Pavia, Italy
| | - Peter Kruzliak
- International Clinical Research Center, St. Anne’s University Hospital and Masaryk University, Pekarska 53, 656 91 Brno, Czech Republic
| |
Collapse
|
17
|
Abstract
Advanced mucosal healing (MH) after intestinal mucosal inflammation coincides with sustained clinical remission and reduced rates of hospitalization and surgical resection, explaining why MH is increasingly considered as a full therapeutic goal and as an endpoint for clinical trials. Intestinal MH is a complex phenomenon viewed as a succession of steps necessary to restore tissue structure and function. These steps include epithelial cell migration and proliferation, cell differentiation, restoration of epithelial barrier functions, and modulation of cell apoptosis. Few clinical studies have evaluated the needs for specific macronutrients and micronutrients and their effects on intestinal MH, most data having been obtained from animal and cell studies. These data suggest that supplementation with specific amino acids including arginine, glutamine, glutamate, threonine, methionine, serine, proline, and the amino acid-derived compounds, polyamines can favorably influence MH. Short-chain fatty acids, which are produced by the microbiota from undigested polysaccharides and protein-derived amino acids, also exert beneficial effects on the process of intestinal MH in experimental models. Regarding supplementation with lipids, although the effects of ω-3 and ω-6 fatty acids remain controversial, endogenous prostaglandin synthesis seems to be necessary for MH. Finally, among micronutrients, several vitamin and mineral deficiencies with different frequencies have been observed in patients with inflammatory bowel diseases and supplementation with some of them (vitamin A, vitamin D3, vitamin C, and zinc) are presumed to favor MH. Future work, including clinical studies, should evaluate the efficiency of supplementation with combination of dietary compounds as adjuvant nutritional intervention for MH of the inflamed intestinal mucosa.
Collapse
|
18
|
Abstract
Current treatment approaches fail to address the underlying factors responsible for the pathogenesis of Crohn's disease (CD). Mesenchymal stromal cells (MSCs) are fibroblastic plastic adherent cells with specific surface markers, and may demonstrate trilineage differentiation ability. MSCs can be isolated from either the adipose tissue, bone marrow or umbilical cord. In refractory fistulizing CD, both autologous and allogeneic MSC therapy with repeated administrations is a feasible and safe therapeutic option with suggestion of efficacy, and several phase 3 trials are currently underway to determine its efficacy. In this review, we discuss the potential therapeutic role of MSC therapy for CD, which is predominantly related to its immunomodulatory role. The characteristics of MSCs derived from patients with CD and its pharmacological interaction are outlined. Preclinical studies using experimental models of colitis and clinical studies using MSCs for the treatment of fistulizing CD are highlighted. Finally, the current perspective and potential limitations of this novel therapy with recommendations for future studies are discussed.
Collapse
Affiliation(s)
- Aaron Liew
- Regenerative Medicine Institute (REMEDI), National Centre for Biomedical Engineering Science (NCBES), Galway, Ireland
| | | | | |
Collapse
|
19
|
Yi JX, Fei SJ, Pang XL, Wu KJ. Therapeutic effects of curcumin combined with MSCs against ulcerative colitis in rats. Shijie Huaren Xiaohua Zazhi 2014; 22:1642-1650. [DOI: 10.11569/wcjd.v22.i12.1642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To assess the therapeutic effects of curcumin combined with mesenchymal stem cells (MSCs) in the treatment of ulcerative colitis in rats and to explore the underlying mechanisms.
METHODS: Ulcerative colitis was induced in rats with 2,4,6-trinitrobenzene sulfonic acid (TNBS). 40 male Sprague-Dawley rats were randomly divided into 5 groups: a normal group, a model group, an MSCs group, a curcumin group, and a curcumin plus MSCs group. The signs and symptoms of rats in each group were observed, including vitality, diet, property of stool and bloody stool. The severity of colitis was assessed using gross morphological score (GMS) and histopathologic damage score (HDS). The percentages of Th17 and Treg lymphocytes in peripheral blood were determined by flow cytometry. Expression of IL-17 and Foxp3 in the colonic mucosa was detected by immunohistochemistry.
RESULTS: Compared with the normal group, the GMS and HDS were significantly higher in the model group (0.00 ± 0.00 vs 4.63 ± 0.74; 0.25 ± 0.46 vs 4.00 ± 0.53, P < 0.01); the levels of Treg in the peripheral blood and Foxp3 in the colon tissue were significant decreased, and the levels of Th17 in the peripheral blood and IL-17 in the colon tissue were significantly increased in the model group (8.01 ± 0.29 vs 1.16 ± 0.18; 9.50 ± 1.60 vs 0.88 ± 0.64; 0.63 ± 0.13 vs 4.56 ± 0.21; 0.88 ± 0.64 vs 7.75 ± 1.16, P < 0.01 for all). The GMS and HDS were significantly lower in the treatment groups than in the model group (4.63 ± 0.74 vs 2.12 ± 0.64, 2.00 ± 0.53, 1.00 ± 0.53; 4.00 ± 0.53 vs 2.00 ± 0.53, 1.88 ± 0.83, 1.25 ± 0.46, P < 0.01 for all). The levels of Treg and Foxp3 were significantly increased, and the levels of Th17 and IL-17 were significantly decreased in the treatment groups compared with the model group (1.16 ± 0.18 vs 3.29 ± 0.18, 3.19 ± 0.20, 5.00 ± 0.19; 0.88 ± 0.64 vs 3.75 ± 0.46, 4.00 ± 0.92, 6.88 ± 1.25; 4.56 ± 0.21 vs 2.60 ± 0.15, 2.59 ± 0.23, 1.52 ± 0.21; 0.88 ± 0.64 vs 3.75 ± 0.46, 4.00 ± 0.92, 6.88 ± 1.25, P < 0.01 for all). Compared with the MSCs group and curcumin group, the GMS and HDS were significantly decreased in the curcumin plus MSCs group (1.00 ± 0.53 vs 2.12 ± 0.64, 2.00 ± 0.53; 1.25 ± 0.46 vs 2.00 ± 0.53, 1.88 ± 0.83, P < 0.05 for all). The levels of Treg and Foxp3 in the combination treatment group were significantly increased, and the levels of Th17 and IL-17 were significantly decreased compared with monotherapy groups (5.00 ± 0.19 vs 3.29 ± 0.18, 3.19 ± 0.20; 6.88 ± 1.25 vs 3.75 ± 0.46, 4.00 ± 0.92; 1.52 ± 0.21 vs 2.60 ± 0.15, 2.59 ± 0.23; 2.38 ± 0.92 vs 5.00 ± 1.07, 4.62 ± 1.19, P < 0.01 for all).
CONCLUSION: Curcumin together with MSCs can decrease colonic mucosal damage and promote the repair of impaired mucosa in UC rats possibly via mechanisms associated with regulating the balance of Treg and Th17 1ymphocytes and the relevant cytokines in colonic tissue.
Collapse
|
20
|
Watanabe S, Arimura Y, Nagaishi K, Isshiki H, Onodera K, Nasuno M, Yamashita K, Idogawa M, Naishiro Y, Murata M, Adachi Y, Fujimiya M, Imai K, Shinomura Y. Conditioned mesenchymal stem cells produce pleiotropic gut trophic factors. J Gastroenterol 2014; 49:270-282. [PMID: 24217964 DOI: 10.1007/s00535-013-0901-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/09/2013] [Indexed: 02/04/2023]
Abstract
BACKGROUND Although mounting evidence implicates mesenchymal stem cells (MSCs) in intestinal tissue repair, controversy remains regarding the engraftment, proliferation, and differentiation for repopulating MSCs in recipient tissues. Therefore, we investigated the paracrine and/or endocrine role of MSCs in experimental colitis. METHODS We analyzed the therapeutic effects of MSC-conditioned medium (MSC-CM) on dextran sulfate sodium (DSS)- or 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis. We investigated the effects of MSC-CM on the epithelial cell viability, mobility, cell cycle, and cytokine production in ex vivo lamina propria/mesenteric lymphocytes, a macrophage cell line, and the mixed lymphocyte reaction. An optimal regimen against colitis was explored. The contents of MSC-CM were analyzed using a WNT signaling pathway polymerase chain reaction array, an inflammatory cytokines antibody array, and liquid chromatography-tandem mass spectrometry analysis. RESULTS Independent of the systemic administration route, MSC-CM concentrates were effective for the inductive phase of TNBS-induced colitis and for the recovery phase of DSS-induced colitis. Hypoxia appeared to be one of the optimal preconditioning factors assessed by cell motility and viability through activating the PI3K-Akt pathway in rat small intestine epithelial cells, IEC-6. Thus, Hypoxia had profound effects on the contents of MSC-CM, which comprised pleiotropic gut trophic factors involved in each wound healing process, including the anti-inflammatory, proliferative, and tissue remodeling phases. CONCLUSIONS Identification and optimization of potential gut trophic factors in MSC-CM is urgently needed to form the basis for new drug discovery and for optimizing cell-based therapies for inflammatory bowel disease.
Collapse
Affiliation(s)
- Shuhei Watanabe
- Department of Gastroenterology, Rheumatology, and Clinical Immunology, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Fawzy SA, El-Din Abo-Elnou RK, Abd-El-Maksoud El-Deeb DF, Yousry Abd-Elkader MM. The possible role of mesenchymal stem cells therapy in the repair of experimentally induced colitis in male albino rats. Int J Stem Cells 2014; 6:92-103. [PMID: 24386553 DOI: 10.15283/ijsc.2013.6.2.92] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Colitis is inflammation of the colon which can be transmural or confined to the mucosa. Colitis may be acute or chronic. In case of serious intestinal discontinuity of epithelium, the regeneration capacity of local stem cells is not enough to complete tissue repair. Bone marrow mesenchymal stem cells (BM-MSCs) migrate into the gastrointestinal wall, where they may contribute to the repair progress. The present study aimed at evaluating the possible therapeutic effect of MSCs on induced colitis in albino rat. METHODS AND RESULTS Twenty male albino rats were divided into 3 groups (control, colitis, MSCs), control group (4 rats), colitis group (8 rats) received once intra-rectal injection of 2 ml of 3% acetic acid. MSCs therapy group (8 rats) injected with MSCs 24 hours after colitis induction. In each group, rats were subdivided into subgroups (a & b). Subgroup (a) corresponds to rats sacrificed 3 days and subgroup (b) corresponds to rats sacrificed 10 days after colitis induction. Isolation and culture of MSCs from rat bone marrow were performed. Colon sections were examined using light and fluorescent microscopy. Colon specimens were subjected to histological, morphometric and statistical studies. In colitis group, ulceration, loss of surface columnar epithelium, disturbed crypts architecture with few goblet cells and huge lymphatic nodule piercing the muscularis mucosa were reported. In stem cell therapy group, MSCs stimulate colonic repair through differentiation into several cells and dampen the inflammation. CONCLUSIONS MSCs represent future therapeutic hopes for intestinal injury and chronic intestinal inflammatory states.
Collapse
Affiliation(s)
- Sohair Ahmed Fawzy
- Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | | | |
Collapse
|
22
|
Kavanagh DPJ, Yemm AI, Alexander JS, Frampton J, Kalia N. Enhancing the adhesion of hematopoietic precursor cell integrins with hydrogen peroxide increases recruitment within murine gut. Cell Transplant 2012; 22:1485-99. [PMID: 22889470 DOI: 10.3727/096368912x653192] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem cells (HSCs) migrate to injury sites and aid in tissue repair. However, clinical success is poor and is partially due to limited HSC recruitment. We hypothesized that HSC pretreatment with H2O2 would enhance their recruitment to injured gut. As HSCs are rare cells, the number of primary cells obtained from donors is often inadequate for functional experiments. To circumvent this, in this study we utilized a functionally relevant cell line, HPC-7. Anesthetized mice were subjected to intestinal ischemia-reperfusion (IR) injury, and HPC-7 recruitment was examined intravitally. Adhesion to endothelial cells (ECs), injured gut sections, and ICAM-1/VCAM-1 protein were also quantitated in vitro. H2O2 pretreatment significantly enhanced HPC-7 recruitment to injured gut in vivo. A concomitant reduction in pulmonary adhesion was also observed. Enhanced adhesion was also observed in all in vitro models. Increased clustering of α4 and β2 integrins, F-actin polymerization, and filopodia formation were observed in pretreated HPC-7s. Importantly, H2O2 did not reduce HPC-7 viability or proliferative ability. HPC-7 recruitment to injured gut can be modulated by H2O2 pretreatment. This may be through increasing the affinity or avidity of surface integrins that mediate HPC-7 homing to injured sites or through stimulating the migratory apparatus. Strategies that enhance hematopoietic stem/progenitor cell recruitment may ultimately affect their therapeutic efficacy.
Collapse
Affiliation(s)
- Dean P J Kavanagh
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, The Medical School, University of Birmingham, Edgbaston, Birmingham, UK
| | | | | | | | | |
Collapse
|
23
|
Gurudutta GU, Satija NK, Singh VK, Verma YK, Gupta P, Tripathi RP. Stem cell therapy: a novel & futuristic treatment modality for disaster injuries. Indian J Med Res 2012; 135:15-25. [PMID: 22382178 PMCID: PMC3307178 DOI: 10.4103/0971-5916.93419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Stem cell therapy hold the potential to meet the demand for transplant cells/tissues needed for treating damages resulting from both natural and man-made disasters. Pluripotency makes embryonic stem cells and induced pluripotent stem cells ideal for use, but their teratogenic character is a major hindrance. Therapeutic benefits of bone marrow transplantation are well known but characterizing the potentialities of haematopoietic and mesenchymal cells is essential. Haematopoietic stem cells (HSCs) have been used for treating both haematopoietic and non-haematopoietic disorders. Ease of isolation, in vitro expansion, and hypoimmunogenecity have brought mesenchymal stem cells (MSCs) into limelight. Though differentiation of MSCs into tissue-specific cells has been reported, differentiation-independent mechanisms seem to play a more significant role in tissue repair which need to be addressed further. The safety and feasibility of MSCs have been demonstrated in clinical trials, and their use in combination with HSC for radiation injury treatment seems to have extended benefit. Therefore, using stem cells for treatment of disaster injuries along with the conventional medical practice would likely accelerate the repair process and improve the quality of life of the victim.
Collapse
Affiliation(s)
- G U Gurudutta
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences, Delhi, India.
| | | | | | | | | | | |
Collapse
|
24
|
Alison MR, Lin WR, Lee CY, Poulsom R, Wright NA, Otto WR. The ailing gut: a therapeutic role for bone marrow cells? Transplantation 2012; 93:565-571. [PMID: 22222785 DOI: 10.1097/tp.0b013e3182419893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
It is obvious that the BM does more than simply supply the GIT with cells of the innate and adaptive immune system. A growing number of studies suggest that BMCs can differentiate into ISEMFs (Lee et al., PLOS ONE 2011;6:e26082) and in the setting of inflammation can be contributors to all lineages of the neovasculature. The role of BMCs in epithelial turnover is more problematic; their contribution after transient mucosal injury seems negligible, but a number of studies in both rodents and man suggest that small numbers of BMCs can be incorporated into the epithelial compartment with more chronic injury (e.g., GvHD in man and chemically induced colitis in rodents); commonly cell fusion seems to be responsible for this. Significantly, this engraftment does not seem to occur in the stem-cell compartment, with the notable single report of the chronically infected murine gastric mucosa, where the BM origin of the stem cells can be the only rational explanation for the complete colonization of the mucosa by BMDCs. In the clinical setting, a role for MSCs in ameliorating colitis seems promising, though the mechanisms by which this is achieved remain somewhat unclear, though both immunomodulatory and regenerative effects of BMCs are likely to be important.
Collapse
Affiliation(s)
- Malcolm R Alison
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Newark Street, London, UK.
| | | | | | | | | | | |
Collapse
|
25
|
Sipos F, Valcz G, Molnár B. Physiological and pathological role of local and immigrating colonic stem cells. World J Gastroenterol 2012; 18:295-301. [PMID: 22294835 PMCID: PMC3261524 DOI: 10.3748/wjg.v18.i4.295] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 06/27/2011] [Accepted: 07/04/2011] [Indexed: 02/06/2023] Open
Abstract
The latest avenue of research is revealing the existence of and role for the colonic stem cells in the physiological renewal of the mucosa and in pathological circumstances where they have both positive and negative effects. In the case of human colon, different levels of stem cell compartments exist. First, the crypt epithelial stem cells, which have a role in the normal crypt epithelial cell dynamics and in colorectal carcinogenesis. Close to the crypts, the second layer of stem cells can be found; the local subepithelial stem cell niche, including the pericryptic subepithelial myofibroblasts that regulate the epithelial cell differentiation and have a crucial role in cancer progression and chronic inflammation-related fibrosis. The third level of stem cell compartment is the immigrating bone-marrow-derived stem cells, which have an important role in wound healing after severe mucosal inflammation, but are also involved in cancer invasion. This paper focuses on stem cell biology in the context of physiological and pathological processes in the human colon.
Collapse
Affiliation(s)
- Ferenc Sipos
- Ferenc Sipos, Gábor Valcz, 2nd Department of Internal Medicine, Semmelweis University, 1088 Budapest, Hungary.
| | | | | |
Collapse
|
26
|
Lee CY, Jeffery R, Hutchinson G, Alison MR, Poulsom R, Wright NA, Otto WR. Bone marrow cells in murine colitis: multi-signal analysis confirms pericryptal myofibroblast engraftment without epithelial involvement. PLoS One 2011; 6:e26082. [PMID: 22022515 PMCID: PMC3192776 DOI: 10.1371/journal.pone.0026082] [Citation(s) in RCA: 4] [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: 01/21/2011] [Accepted: 09/19/2011] [Indexed: 01/30/2023] Open
Abstract
Background The contribution of bone marrow-derived cells to epithelial tissues in the inflamed gut remains controversial. Recent reports have suggested that cell fusion between bone marrow-derived cells and the intestinal epithelium takes place in inflammatory conditions. Methods In attempts to confirm this, we have undertaken gender mis-matched bone marrow (BM) transplants from male Swiss Webster (SWR) mice to B and T cell-deficient female Rag2 KO mice which, 4 weeks later, were given 5% dextran sodium sulphate in drinking water to induce acute colitis. A further BM-treated group of animals with a graft versus host-like condition was also studied. We developed a new method to combine up to three brightfield or fluorescent lectin- or immuno-histochemical signals with fluorescent in situ hybridisation for the Y and X chromosomes to enable us unequivocally to identify BM-derived male cells which presented as different cell types in the gastrointestinal tract. Principal Findings In rolled preparations of whole intestines we scanned around 1.5 million crypts at many tissue levels. In no instance did we see a Y chromosome-positive cell in the epithelial compartment, which was not also CD45-positive. We saw no evidence of cell fusion, based on combined X and Y chromosome analysis. Levels of CD45-positive stromal and lymphoid cells and pericryptal myfibroblasts (positive for α-smooth muscle actin) increased with time up to a plateau, which resembled the level seen in untreated control grafted animals. We saw very few Y chromosome-positive endothelial cells in intestinal stromal vessels. Conclusions We conclude that whole BM transplantation does not result in intestinal epithelial engraftment in this model. Our new methods can usefully assist in multi-signal analyses of cell phenotypes following BM transplant and in models of chimaerism and regenerative medicine.
Collapse
Affiliation(s)
- Chung-Yin Lee
- Histopathology Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
| | - Rosemary Jeffery
- Histopathology Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
| | - Gillian Hutchinson
- Biological Resources Unit, Cancer Research UK, London Research Institute, London, United Kingdom
| | - Malcolm R. Alison
- Centre for Digestive Diseases, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Richard Poulsom
- Histopathology Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
- Centre for Digestive Diseases, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Nicholas A. Wright
- Histopathology Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
- Centre for Digestive Diseases, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - William R. Otto
- Histopathology Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
- Centre for Digestive Diseases, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
- * E-mail:
| |
Collapse
|
27
|
Trebol Lopez J, Georgiev Hristov T, García-Arranz M, García-Olmo D. Stem Cell Therapy for Digestive Tract Diseases: Current State and Future Perspectives. Stem Cells Dev 2011; 20:1113-29. [DOI: 10.1089/scd.2010.0277] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Jacobo Trebol Lopez
- General and Digestive Tract Surgery Department, University Hospital “La Paz”, Madrid, Spain
- Cell Therapy Laboratory, Investigation Institute IdiPAZ, University Hospital “La Paz”, Madrid, Spain
| | - Tihomir Georgiev Hristov
- General and Digestive Tract Surgery Department, University Hospital “La Paz”, Madrid, Spain
- Cell Therapy Laboratory, Investigation Institute IdiPAZ, University Hospital “La Paz”, Madrid, Spain
| | - Mariano García-Arranz
- Cell Therapy Laboratory, Investigation Institute IdiPAZ, University Hospital “La Paz”, Madrid, Spain
| | - Damián García-Olmo
- General and Digestive Tract Surgery Department, University Hospital “La Paz”, Madrid, Spain
- Cell Therapy Laboratory, Investigation Institute IdiPAZ, University Hospital “La Paz”, Madrid, Spain
- Surgery Department, Autonomous University of Madrid, Madrid, Spain
| |
Collapse
|
28
|
Klemmt PAB, Vafaizadeh V, Groner B. The potential of amniotic fluid stem cells for cellular therapy and tissue engineering. Expert Opin Biol Ther 2011; 11:1297-314. [DOI: 10.1517/14712598.2011.587800] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
29
|
Valcz G, Krenács T, Sipos F, Leiszter K, Tóth K, Balogh Z, Csizmadia A, Muzes G, Molnár B, Tulassay Z. The role of the bone marrow derived mesenchymal stem cells in colonic epithelial regeneration. Pathol Oncol Res 2011; 17:11-16. [PMID: 20405350 DOI: 10.1007/s12253-010-9262-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 03/22/2010] [Indexed: 02/08/2023]
Abstract
Bone marrow derived mesenchymal stem cells (BM-MSCs) take part in the colonic mucosal regeneration. They are multipotent cells, which can be identified with both negative (i.e. CD13, CD 14, CD45, c-Kit, major histocompatibility complex /MHC class I and II) and positive (i.e. CD54 (ICAM1), CD133, CD146 (MCAM), CD166, Flk-1, Sca-1, Thy-1, stage-specific antigen I /SSEA-I and Musashi-1, HLA class I) markers. These cells can repopulate the gastrointestinal mucosa as they may differentiate into stromal- (i.e. myofi-broblast) or epithelial-like (Paneth-, epithel-, goblet or enteroendocrin) cells without proliferation. During the mesenchymal to epithelial transition (MET) stem cells enter the epithelial layer and take up epithelial cell-like properties. Rarely BM-MSCs may retain their stem cell characteristics and are capable of producing progeny. The isolated lymphoid aggregates may serve as a platform from where BM-MSCs migrate to the nearby crypts as mediated by several chemoattractant proteins, which are expressed in injured tissue. The number of BM-MSCs is influenced by the degree of inflammation. In this review we summarize the current information about the role of BM-MSCs in the repair progress of injured colonic epithelium and their potential clinical applications.
Collapse
Affiliation(s)
- Gábor Valcz
- 2nd Department of Internal Medicine, Cell Analysis Laboratory, Semmelweis University, Szentkiralyi Street 46, 1088 Budapest, Hungary.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Myogenic lineage differentiated mesenchymal stem cells enhance recovery from dextran sulfate sodium-induced colitis in the rat. J Gastroenterol 2011; 46:143-52. [PMID: 20848145 DOI: 10.1007/s00535-010-0320-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Accepted: 08/13/2010] [Indexed: 02/04/2023]
Abstract
BACKGROUND Although mounting evidence implicates mesenchymal stem cells (MSCs) in intestinal tissue repair, uncertainty remains concerning the distribution, function, and fate of repopulating MSCs in recipient colonic tissues. Therefore, we investigated the role of transplanted MSCs in the repair phase of DSS colitis. METHODS LacZ-labeled rat MSCs were transplanted into rats with colitis induced by 4% DSS on day 2. Regular water replaced the DSS solution on day 6. Therapeutic effect was evaluated on day 9 by clinicopathologic and growth factor/cytokine expression profiles. We analyzed the Notch signaling pathway by Western blotting and characterized immunofluorescence of lacZ-labeled MSCs with confocal laser microscopy. In vivo differentiation of MSC was confirmed by transmission electron microscopy (TEM). RESULTS Recovery of colitis was modestly but significantly promoted by MSC transplantation due to proceeding cell cycle and inhibiting apoptosis in the epithelia. Tgfa mRNA expression increased significantly, while Notch signaling was inhibited in the colonic tissues with MSC transplantation. β-Galactosidase-positive cells, which expressed α-SMA, desmin, and vimentin, were infrequently detected in the lamina propria stroma. DSS exposure in vitro proved to be the most potent inducer for α-SMA in MSCs where TEM demonstrated myogenic lineage differentiation. CONCLUSIONS We found that MSCs transplantation modestly promoted the repair of DSS colitis. The donor-derived MSCs were likely reprogrammed to differentiate to myogenic lineage cells by cues from the micro milieu. Further characterization of these cells is warranted as a basis for applying cell-based therapy for inflammatory bowel disease.
Collapse
|
31
|
Klemmt PAB, Vafaizadeh V, Groner B. Murine amniotic fluid stem cells contribute mesenchymal but not epithelial components to reconstituted mammary ducts. Stem Cell Res Ther 2010; 1:20. [PMID: 20609228 PMCID: PMC2941112 DOI: 10.1186/scrt20] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/17/2010] [Accepted: 07/07/2010] [Indexed: 01/06/2023] Open
Abstract
Introduction Amniotic fluid harbors cells indicative of all three germ layers, and pluripotent fetal amniotic fluid stem cells (AFSs) are considered potentially valuable for applications in cellular therapy and tissue engineering. We investigated whether it is possible to direct the cell fate of AFSs in vivo by transplantation experiments into a particular microenvironment, the mammary fat pad. This microenvironment provides the prerequisites to study stem cell function and the communication between mesenchymal and epithelial cells. On clearance of the endogenous epithelium, the ductal tree can be reconstituted by the transfer of exogenously provided mammary stem cells. Analogously, exogenously provided stem cells from other tissues can be investigated for their potential to contribute to mammary gland regeneration. Methods We derived pluripotent murine AFSs, measured the expression of stem cell markers, and confirmed their in vitro differentiation potential. AFSs were transplanted into cleared and non cleared fat pads of immunocompromised mice to evaluate their ability to assume particular cell fates under the instructive conditions of the fat-pad microenvironment and the hormonal stimulation during pregnancy. Results Transplantation of AFSs into cleared fat pads alone or in the presence of exogenous mammary epithelial cells caused their differentiation into stroma and adipocytes and replaced endogenous mesenchymal components surrounding the ducts in co-transplantation experiments. Similarly, transplantation of AFSs into fat pads that had not been previously cleared led to AFS-derived stromal cells surrounding the elongating endogenous ducts. AFSs expressed the marker protein α-SMA, but did not integrate into the myoepithelial cell layer of the ducts in virgin mice. With pregnancy, a small number of AFS-derived cells were present in acinar structures. Conclusions Our data demonstrate that the microenvironmental cues of the mammary fat pad cause AFSs to participate in mammary gland regeneration by providing mesenchymal components to emerging glandular structures, but do not incorporate or differentiate into ductal epithelial cells.
Collapse
Affiliation(s)
- Petra A B Klemmt
- Georg-Speyer-Haus, Institute for Biomedical Research, Paul-Ehrlich-Str, 42-44, 60596 Frankfurt, Germany.
| | | | | |
Collapse
|
32
|
Singh UP, Singh NP, Singh B, Mishra MK, Nagarkatti M, Nagarkatti PS, Singh SR. Stem cells as potential therapeutic targets for inflammatory bowel disease. Front Biosci (Schol Ed) 2010; 2:993-1008. [PMID: 20515838 DOI: 10.2741/s115] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The incidence and prevalence of Crohn's disease and ulcerative colitis, the two major forms of inflammatory bowel disease (IBD), are rising. According to some estimates >1 million new cases of IBD arise in the United States annually. The conventional therapies available for IBD range from anti-inflammatory drugs to immunosuppressive agents, but these therapies generally fail to achieve satisfactory results due to their side effects. Interest in a new therapeutic option, that is, biological therapy, has gained much momentum recently due to its focus on different stages of the inflammatory process. Stem cell (SC) research has become a new direction for IBD therapy due to our recent understanding of cell populations involved in the pathogenic process. To this end, hematopoietic and mesenchymal stem cells are receiving more attention from IBD investigators. The intestinal environment, with its crypts and niches, supports incoming embryonic and hematopoietic stem cells and allows them to engraft and differentiate. The above findings suggest that, in the future, SC-based therapy will be a promising alternative to conventional therapy for IBD. In this review, we discuss SCs as potential therapeutic targets for future treatment of IBD.
Collapse
Affiliation(s)
- Udai P Singh
- Pathology and Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Intestinal stem cells and their roles during mucosal injury and repair. J Surg Res 2010; 167:1-8. [PMID: 20599211 DOI: 10.1016/j.jss.2010.04.037] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 04/13/2010] [Accepted: 04/20/2010] [Indexed: 01/10/2023]
Abstract
The ability of the host to respond to intestinal injury requires the regeneration of native tissue through a highly orchestrated response from the intestinal stem cells, a population of cells located within the intestinal crypts that have the capability to repopulate the entire villous. The field of intestinal stem cell biology is thus of great interest to surgeons and non-surgeons alike, given its relevance to diseases of intestinal injury and inflammation such as inflammatory bowel disease, trauma, and necrotizing enterocolitis. The field of intestinal stem cell research has been advanced recently by the identification of the putative marker, Lgr5, which has allowed for the isolation and further characterization of the intestinal stem cell. Under the control of the WNT signaling pathway, Lgr5 marks the rapidly dividing cells of the intestinal crypt, and identifies a population of cells that is capable of regenerating the entire villous. We now review the identification of Lgr5 as an intestinal stem cell marker, identify controversies in the intestinal stem cell field, and highlight the response of the intestinal stem cell to injury within the intestinal mucosa that may occur clinically.
Collapse
|