Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmacol Res ; 178: 106147, 2022 04.
Article in English | MEDLINE | ID: mdl-35227891

ABSTRACT

CTLA4-Ig is a potent costimulatory blocker that inhibits T cell activation during alloimmune inflammation and increases graft survival and function. CTLA4-Ig-mediated immunosuppression has been demonstrated to support transplant function in various clinical trials and preclinical settings, but its effects on the balance between regulatory T cells (Tregs) and effector T cells (Teffs), as well as complement activation, are less well investigated. In the present study, we proposed to investigate the effects of CTLA4-Ig mediated immunosuppression on the phase of immunotolerance and the subsequent graft microvascular and epithelial repair during the progression of subepithelial fibrosis in a mouse model of orthotopic trachea transplantation. Briefly, CTLA4-Ig treated allografts (2 mg/kg, I.P.), untreated allografts, and syngrafts were serially monitored for peripheral FOXP3+ Tregs, antibody-mediated complement activation (C3d and C4d), tissue oxygenation, donor-recipient microvascular blood flow, and subsequent tissue remodeling following transplantation. Our data demonstrate that CTLA4-Ig mediated immunosuppression significantly results in late increases in both peripheral CD4+/CD8+ FOXP3+ Tregs and serum IL-10, but prevents the microvascular deposition of IgG, complement factor C3d, and epithelial C4d respectively, which proportionally improved blood flow and tissue oxygenation in the graft and, thus, promotes graft repair. Also, it restored the airway lumen, epithelium, and prevented the progression of subepithelial collagen deposition up to 90 days after transplantation. This study demonstrates that CTLA4-Ig-mediated immunosuppression potentially modulates both effector response and a late surge of regulatory activity to preserve graft microvasculature and rescue allograft from sustained hypoxia and ischemia and thereby limits subepithelial fibrosis.


Subject(s)
CTLA-4 Antigen , Graft Rejection , Graft Survival , Abatacept/pharmacology , Abatacept/therapeutic use , Animals , CTLA-4 Antigen/administration & dosage , CTLA-4 Antigen/immunology , Fibrosis , Forkhead Transcription Factors , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival/immunology , Immunosuppression Therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Trachea/transplantation
2.
Int J Mol Sci ; 23(3)2022 Jan 23.
Article in English | MEDLINE | ID: mdl-35163192

ABSTRACT

Interleukin-10 (IL-10) is a vital regulatory cytokine, which plays a constructive role in maintaining immune tolerance during an alloimmune inflammation. Our previous study highlighted that IL-10 mediated immunosuppression established the immune tolerance phase and thereby modulated both microvascular and epithelial integrity, which affected inflammation-associated graft malfunctioning and sub-epithelial fibrosis in rejecting allografts. Here, we further investigated the reparative effects of IL-10 on microvasculature and epithelium in a mouse model of airway transplantation. To investigate the IL-10 mediated microvascular and epithelial repair, we depleted and reconstituted IL-10, and monitored graft microvasculature, airway epithelium, and associated repair proteins. Our data demonstrated that both untreated control allografts and IL-10 (-) allografts showed a significant early (d6) increase in microvascular leakiness, drop-in tissue oxygenation, blood perfusion, and denuded airway epithelium, which is associated with loss of adhesion protein Fascin-1 and ß-catenin on vascular endothelial cells at d10 post-transplantation. However, IL-10 (+) promotes early microvascular and airway epithelial repair, and a proportional increase in endothelial Fascin-1, and ß-catenin at d10 post-transplantation. Moreover, airway epithelial cells also express a significantly higher expression of FOXJ1 and ß-catenin in syngrafts and IL-10 (+) allografts as compared to IL-10 (-) and untreated controls at d10 post-transplantation. Collectively, these findings demonstrated that IL-10 mediated microvascular and epithelial changes are associated with the expression of FOXJ1, ß-catenin, and Fascin-1 proteins on the airway epithelial and vascular endothelial cells, respectively. These findings establish a potential reparative modulation of IL-10 associated microvascular and epithelial repair, which could provide a vital therapeutic strategy to facilitate graft repair in clinical settings.


Subject(s)
Allografts/metabolism , Graft Rejection/immunology , Interleukin-10/metabolism , Animals , Endothelial Cells/immunology , Epithelial Cells/immunology , Epithelium/immunology , Graft Survival/physiology , Immune Tolerance , Immunosuppression Therapy , Interleukin-10/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microvessels/immunology , Microvessels/physiology , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous/methods
3.
Cells ; 10(5)2021 05 19.
Article in English | MEDLINE | ID: mdl-34069395

ABSTRACT

Interleukin-10 plays a vital role in maintaining peripheral immunotolerance and favors a regulatory immune milieu through the suppression of T effector cells. Inflammation-induced microvascular loss has been associated with airway epithelial injury, which is a key pathological source of graft malfunctioning and subepithelial fibrosis in rejecting allografts. The regulatory immune phase maneuvers alloimmune inflammation through various regulatory modulators, and thereby promotes graft microvascular repair and suppresses the progression of fibrosis after transplantation. The present study was designed to investigate the therapeutic impact of IL-10 on immunotolerance, in particular, the reparative microenvironment, which negates airway epithelial injury, and fibrosis in a mouse model of airway graft rejection. Here, we depleted and reconstituted IL-10, and serially monitored the phase of immunotolerance, graft microvasculature, inflammatory cytokines, airway epithelium, and subepithelial collagen in rejecting airway transplants. We demonstrated that the IL-10 depletion suppresses FOXP3+ Tregs, tumor necrosis factor-inducible gene 6 protein (TSG-6), graft microvasculature, and establishes a pro-inflammatory phase, which augments airway epithelial injury and subepithelial collagen deposition while the IL-10 reconstitution facilitates FOXP3+ Tregs, TSG-6 deposition, graft microvasculature, and thereby favors airway epithelial repair and subepithelial collagen suppression. These findings establish a potential reparative modulation of IL-10-associated immunotolerance on microvascular, epithelial, and fibrotic remodeling, which could provide a vital therapeutic option to rescue rejecting transplants in clinical settings.


Subject(s)
Graft Rejection/metabolism , Interleukin-10/metabolism , Re-Epithelialization , Respiratory Mucosa/metabolism , Trachea/transplantation , Animals , Disease Models, Animal , Fibrosis , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival , Inflammation Mediators/metabolism , Interleukin-10/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors , Trachea/immunology , Trachea/metabolism , Trachea/pathology , Transplantation Tolerance
4.
J Transl Med ; 18(1): 147, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32234039

ABSTRACT

BACKGROUND: Complement Regulatory Proteins (CRPs), especially CD55 primarily negate complement factor 3-mediated injuries and maintain tissue homeostasis during complement cascade activation. Complement activation and regulation during alloimmune inflammation contribute to allograft injury and therefore we proposed to investigate a crucial pathological link between vascular expression of CD55, active-C3, T cell immunity and associated microvascular tissue injuries during allograft rejection. METHODS: Balb/c→C57BL/6 allografts were examined for microvascular deposition of CD55, C3d, T cells, and associated tissue microvascular impairments during rejection in mouse orthotopic tracheal transplantation. RESULTS: Our findings demonstrated that hypoxia-induced early activation of HIF-1α favors a cell-mediated inflammation (CD4+, CD8+, and associated proinflammatory cytokines, IL-2 and TNF-α), which proportionally triggers the downregulation of CRP-CD55, and thereby augments the uncontrolled release of active-C3, and Caspase-3 deposition on CD31+ graft vascular endothelial cells. These molecular changes are pathologically associated with microvascular deterioration (low tissue O2 and Blood flow) and subsequent airway epithelial injuries of rejecting allografts as compared to non-rejecting syngrafts. CONCLUSION: Together, these findings establish a pathological correlation between complement dysregulation, T cell immunity, and microvascular associated injuries during alloimmune inflammation in transplantation.


Subject(s)
Endothelial Cells , Graft Rejection , Animals , Hypoxia , Mice , Mice, Inbred C57BL , Trachea
5.
Stem Cell Res Ther ; 10(1): 290, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31547869

ABSTRACT

BACKGROUND: Lung transplantation is a life-saving surgical replacement of diseased lungs in patients with end-stage respiratory malfunctions. Despite remarkable short-term recovery, long-term lung survival continues to face several major challenges, including chronic rejection and severe toxic side effects due to global immunosuppression. Stem cell-based immunotherapy has been recognized as a crucial immunoregulatory regimen in various preclinical and clinical studies. Despite initial therapeutic outcomes, conventional stem cells face key limitations. The novel Cymerus™ manufacturing facilitates production of a virtually limitless supply of consistent human induced pluripotent stem cell (iPSC)-derived mesenchymal stem cells, which could play a key role in selective immunosuppression and graft repair during rejection. METHODS: Here, we demonstrated the impact of iPSC-derived human MSCs on the development of immune tolerance and long-term graft survival in mouse orthotopic airway allografts. BALB/c → C57BL/6 allografts were reconstituted with iPSC-derived MSCs (2 million/transplant/at d0), and allografts were examined for regulatory T cells (Tregs), oxygenation, microvascular blood flow, airway epithelium, and collagen deposition during rejection. RESULTS: We demonstrated that iPSC-derived MSC treatment leads to significant increases in hTSG-6 protein, followed by an upregulation of mouse Tregs and IL-5, IL-10, and IL-15 cytokines, which augments graft microvascular blood flow and oxygenation, and thereby maintained a healthy airway epithelium and prevented the subepithelial deposition of collagen at d90 post transplantation. CONCLUSIONS: Collectively, these data confirmed that iPSC-derived MSC-mediated immunosuppression has potential to establish immune tolerance and rescue allograft from sustained hypoxic/ischemic phase, and subsequently limits long-term airway epithelial injury and collagen progression, which therapeutically warrant a study of Cymerus iPSC-derived MSCs as a potential management option for immunosuppression in transplant recipients.


Subject(s)
Graft Rejection/therapy , Graft Survival , Mesenchymal Stem Cell Transplantation/methods , Organ Transplantation/methods , Trachea/transplantation , Transplantation Tolerance , Animals , Cells, Cultured , Graft Rejection/immunology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Organ Transplantation/adverse effects , T-Lymphocytes, Regulatory/immunology
6.
Front Immunol ; 9: 1010, 2018.
Article in English | MEDLINE | ID: mdl-29881374

ABSTRACT

Microvascular injury during acute rejection has been associated with massive infiltration of CD4+ T effector cells, and the formation of complement products (C3a and C5a). Regulatory T cells (Tregs) are potent immunosuppressors of the adaptive immune system and have proven sufficient to rescue microvascular impairments. Targeting C5a has been linked with improved microvascular recovery, but its effects on the Treg and T effector balance is less well known. Here, we demonstrate the impact of C5a blockade on Treg induction and microvascular restoration in rejecting mouse airway allografts. BALB/c→C57BL/6 allografts were treated with a C5a-neutralizing l-aptamer (10 mg/kg, i.p. at d0 and every second day thereafter), and allografts were serially monitored for Treg infiltration, tissue oxygenation (tpO2), microvascular blood flow, and functional microvasculature between donor and recipients during allograft rejection. We demonstrated that C5a blocking significantly leads to enhanced presence of Tregs in the allograft, reinstates donor-recipient functional microvasculature, improves tpO2, microvascular blood flow, and epithelial repair, followed by an upregulation of IL-5, TGF-ß, IL-10 vascular endothelial growth factor, and ANGPT1 gene expression, while it maintained a healthy epithelium and prevented subepithelial collagen deposition at d28 posttransplantation. Together, these data indicate that inhibition of C5a signaling has potential to preserve microvasculature and rescue allograft from a sustained hypoxic/ischemic phase, limits airway tissue remodeling through the induction of Treg-mediated immune tolerance. These findings may be useful in designing anti-C5a therapy in combination with existing immunosuppressive regimens to rescue tissue/organ rejection.


Subject(s)
Complement C5a/antagonists & inhibitors , Epithelial Cells/immunology , Epithelial Cells/pathology , Graft Rejection/prevention & control , T-Lymphocytes, Regulatory/immunology , Trachea/transplantation , Allografts/immunology , Animals , Complement C5a/immunology , Graft Rejection/immunology , Immune Tolerance , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microvessels/immunology , Respiratory System/cytology , Respiratory System/immunology , Trachea/immunology , Transplantation, Homologous
7.
Clin Immunol ; 174: 84-98, 2017 01.
Article in English | MEDLINE | ID: mdl-27939405

ABSTRACT

Microvascular loss may be a root cause of chronic rejection in lung transplants, which leads to the bronchiolitis obliterans syndrome. Previous research implicates T regulatory cell (Treg) as a key component of immune modulation, however, Treg has never been examined as a reparative mediator to salvage microvasculature during transplantation. Here, we reconstituted purified Tregs in to allografts, and serially monitored allografts for tissue oxygenation, microvascular perfusion for four weeks. We demonstrated that Tregs reconstitution of allografts significantly improve tissue oxygenation, microvascular flow, epithelial repair, number of CD4+CD25highFOXP3+ Tregs, followed by an upregulation of proinflammatory, angiogenic and regulatory genes, while prevented subepithelial deposition of CD4+T cells at d10, and collagen at d28 post-transplantation. Altogether, these findings concluded that Treg-mediated immunotherapy has potential to preserve microvasculature and rescue allograft from sustained hypoxic/ischemic phase, limits airway tissue remodeling, and therefore may be a useful therapeutic tool to prevent chronic rejection after organ transplantation.


Subject(s)
Graft Rejection/immunology , Microvessels/immunology , T-Lymphocytes, Regulatory/immunology , Trachea/transplantation , Animals , Forkhead Transcription Factors/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...