Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Biomater Sci ; 11(11): 4022-4031, 2023 May 30.
Article in English | MEDLINE | ID: mdl-37129566

ABSTRACT

Vascularized composite allotransplantation (VCA) is a restorative option for patients suffering from severe tissue defects not amenable to conventional reconstruction. However, the toxicities associated with life-long multidrug immunosuppression to enable allograft survival and induce immune tolerance largely limit the broader application of VCA. Here, we investigate the potential of targeted immunomodulation using CTLA4-Ig combined with a biological porcine-derived extracellular matrix (ECM) scaffold that elicits a pro-regenerative Th2 response to promote allograft survival and regulate the inflammatory microenvironment in a stringent mouse orthotopic hind limb transplantation model (BALB/c to C57BL/6). The median allograft survival time (MST) increased significantly from 15.0 to 24.5 days (P = 0.0037; Mantel-Cox test) after adding ECM to the CTLA4-Ig regimen. Characterization of the immune infiltration shows a pro-regenerative phenotype prevails over those associated with inflammation and rejection including macrophages (F4/80hi+CD206hi+MHCIIlow), eosinophils (F4/80lowSiglec-F+), and T helper 2 (Th2) T cells (CD4+IL-4+). This was accompanied by an increased expression of genes associated with a Type 2 polarized immune state such as Il4, Ccl24, Arg1 and Ym1 within the graft. Furthermore, when ECM was applied along with a clinically relevant combination of CTLA4-Ig and Rapamycin, allograft survival was prolonged from 33.0 to 72.5 days (P = 0.0067; Mantel-Cox test). These studies implicate the clinical exploration of combined regimens involving local application of pro-regenerative, immunomodulatory biomaterials in surgical wound sites with targeted co-stimulatory blockade to reduce adverse effects of immunosuppression and enhance graft survival in VCA.


Subject(s)
Composite Tissue Allografts , Mice , Swine , Animals , Abatacept , Mice, Inbred C57BL , Transplantation, Homologous , Immunomodulation
2.
J Vis Exp ; (193)2023 03 31.
Article in English | MEDLINE | ID: mdl-37067285

ABSTRACT

Central venous catheters (CVCs) are invaluable devices in large animal research as they facilitate a wide range of medical applications, including blood monitoring and reliable intravenous fluid and drug administration. Specifically, the tunneled multi-lumen Hickman catheter (HC) is commonly used in swine models due to its lower extrication and complication rates. Despite fewer complications relative to other CVCs, HC-related morbidity presents a significant challenge, as it can significantly delay or otherwise negatively impact ongoing studies. The proper insertion and maintenance of HCs is paramount in preventing these complications, but there is no consensus on best practices. The purpose of this protocol is to comprehensively describe an approach for the insertion and maintenance of a tunneled HC in swine that mitigates HC-related complications and morbidity. The use of these techniques in >100 swine has resulted in complication-free patent lines up to 8 months and no catheter-related mortality or infection of the ventral surgical site. This protocol offers a method to optimize the lifespan of the HC and guidance for approaching issues during use.


Subject(s)
Catheterization, Central Venous , Central Venous Catheters , Animals , Swine , Central Venous Catheters/adverse effects , Catheters, Indwelling
3.
Transpl Int ; 34(5): 894-905, 2021 05.
Article in English | MEDLINE | ID: mdl-33626223

ABSTRACT

In vascularized composite allotransplantation (VCA), invasive tissue biopsies remain the gold standard in diagnosing rejection carrying significant morbidity. We aimed to show feasibility of tape-stripping for noninvasive immune monitoring in VCA. Tape-stripping was performed on allografts and native skin of upper extremity transplant recipients. Healthy nontransplanted individuals served as controls. The technique was also used in swine on naïve skin in nontransplanted animals, native skin of treated, transplanted swine, nonrejecting VCAs, and rejecting VCAs. Extracted protein was analyzed for differences in cytokine expression using Luminex technology. Significantly decreased levels of INFγ and IL-1Ra were seen between human allograft samples and native skin. In swine, rejecting grafts had increased IL-1Ra compared to naïve and native skin, decreased levels of GM-CSF compared to native skin, and decreased IL-10 compared to nonrejecting grafts. Unsupervised hierarchical clustering revealed rejecting grafts separated from the nonrejecting (P = 0.021). Variable importance in projection scores identified GM-CSF, IL-1Ra, and IL-2 as the most important profiles for group discrimination. Differences in cytokine expression are detectable in human VCA patient native skin and VCA graft skin using a noninvasive tape-stripping method. Swine studies suggest that differences in cytokines between rejecting and nonrejecting grafts are discernable.


Subject(s)
Graft Rejection , Vascularized Composite Allotransplantation , Animals , Humans , Immunity , Skin Transplantation , Swine , Upper Extremity
4.
Small ; 16(38): e2002791, 2020 09.
Article in English | MEDLINE | ID: mdl-32812339

ABSTRACT

Combination therapies that target multiple pathways involved in immune rejection of transplants hold promise for patients in need of restorative surgery. Herein, a noninteracting multiphase molecular assembly approach is developed to crystallize tofacitinib, a potent JAK1/3 inhibitor, within a shear-thinning self-assembled fibrillar peptide hydrogel network. The resulting microcrystalline tofacitinib hydrogel (MTH) can be syringe-injected directly to the grafting site during surgery to locally deliver the small molecule. The rate of drug delivered from MTH is largely controlled by the dissolution of the encapsulated microcrystals. A single application of MTH, in combination with systemically delivered CTLA4-Ig, a co-stimulation inhibitor, affords significant graft survival in mice receiving heterotopic heart transplants. Locoregional studies indicate that the local delivery of tofacitinib at the graft site enabled by MTH is required for the observed enhanced graft survival.


Subject(s)
Heart Transplantation , Hydrogels , Animals , Humans , Immunomodulation , Mice , Peptides
5.
Front Immunol ; 11: 572, 2020.
Article in English | MEDLINE | ID: mdl-32328063

ABSTRACT

Transplant tolerance in the absence of long-term immunosuppression has been an elusive goal for solid organ transplantation. Recently, it has become clear that metabolic reprogramming plays a critical role in promoting T cell activation, differentiation, and function. Targeting metabolism can preferentially inhibit T cell effector generation while simultaneously promoting the generation of T regulatory cells. We hypothesized that costimulatory blockade with CTLA4Ig in combination with targeting T cell metabolism might provide a novel platform to promote the induction of transplant tolerance.


Subject(s)
Abatacept/pharmacology , Deoxyglucose/pharmacology , Diazooxonorleucine/pharmacology , Immunosuppression Therapy/methods , Metformin/pharmacology , Transplantation Tolerance/drug effects , Allografts , Animals , Glycolysis/drug effects , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Mice , T-Lymphocytes/drug effects
6.
Transpl Int ; 33(8): 948-957, 2020 08.
Article in English | MEDLINE | ID: mdl-32299127

ABSTRACT

We herein investigate the safety and efficacy of single-agent anti-rejection regimens in a mouse vascularized composite allotransplantation (VCA) model. Orthotopic hind-limb transplantations (Balb/c â†’ C57BL/6) were performed using 6- to 8-week-old mice. A thirty-day regimen of either rapamycin, tacrolimus (both 1, 3, 5 mg/kg/day) or cyclosporine (25, 35, 50 mg/kg/day) was used. Primary endpoints were animal and graft survival, and secondary chimerism and regulatory T-cell levels. For rapamycin and tacrolimus given at 1, 3, and 5 mg/kg/day, median graft survival time (MST) was 23 days (18-28 days), 30 days (23-30 days), and 30 d (30-30 days) and 14 days (13-18 days), 30 days (16-30 days), and 30 days (30-30 days), respectively. For cyclosporine dosed at 25 and 35 mg/kg/day, MST was 15 days (12-18 days) and 21 days (14-27 days). Toxicity from CsA 50 mg/kg led to 100% mortality. Mixed chimerism levels were higher in rapamycin-treated animals than in tacrolimus-treated recipients (P = 0.029). Tacrolimus was superior in preventing leukocyte recruitment to the allograft. In murine VCA, no standardized immunosuppressive regimen exists, limiting comparability of outcomes and survival. Our data demonstrate that rapamycin and tacrolimus maintenance treatment at 5 mg/kg/day both yielded allograft survival (

Subject(s)
Vascularized Composite Allotransplantation , Animals , Disease Models, Animal , Graft Rejection/prevention & control , Graft Survival , Immunosuppressive Agents , Mice , Mice, Inbred C57BL , Tacrolimus
7.
JCI Insight ; 5(7)2020 04 09.
Article in English | MEDLINE | ID: mdl-32271163

ABSTRACT

Vascularized composite allotransplantation (VCA) has become a valid therapeutic option to restore form and function after devastating tissue loss. However, the need for high-dose multidrug immunosuppression to maintain allograft survival is still hampering more widespread application of VCA. In this study, we investigated the immunoregulatory potential of costimulation blockade (CoB; CTLA4-Ig and anti-CD154 mAb) combined with nonmyeoablative total body irradiation (TBI) to promote allograft survival of VCA in a fully MHC-mismatched mouse model of orthotopic hind limb transplantation. Compared with untreated controls (median survival time [MST] 8 days) and CTLA4-Ig treatment alone (MST 17 days), CoB treatment increased graft survival (MST 82 days), and the addition of nonmyeloablative TBI led to indefinite graft survival (MST > 210 days). Our analysis suggests that VCA-derived BM induced mixed chimerism in animals treated with CoB and TBI + CoB, promoting gradual deletion of alloreactive T cells as the underlying mechanism of long-term allograft survival. Acceptance of donor-matched secondary skin grafts, decreased ex vivo T cell responsiveness, and increased graft-infiltrating Tregs further indicated donor-specific tolerance induced by TBI + CoB. In summary, our data suggest that vascularized BM-containing VCAs are immunologically favorable grafts promoting chimerism induction and long-term allograft survival in the context of CoB.


Subject(s)
Abatacept/pharmacology , Graft Survival/drug effects , Immunosuppressive Agents/pharmacology , Transplantation Chimera/immunology , Transplantation Tolerance , Vascularized Composite Allotransplantation , Allografts , Animals , Graft Survival/immunology , Male , Mice , Mice, Inbred BALB C
8.
Transpl Int ; 33(7): 796-805, 2020 07.
Article in English | MEDLINE | ID: mdl-32145119

ABSTRACT

Penis transplantation represents an exciting new avenue for restoration of male genitalia and function after devastating tissue loss. This animal model is designed to fill a critical void to study immunologic aspects related to reconstructive transplantation of male genitalia. A rat penile graft dissection was designed based on the internal pudendal arteries and dorsal penile vein and includes the skin of the prepuce. A nonsuture cuff technique was used to anastomose the graft vessels to the recipient superficial epigastric and femoral vessels. Seventy-seven penile transplantations were performed. Graft design yields suitable caliber and length of vessels at the radix of the penis. Anastomosis of the dorsal penile vein and the internal pudendal arteries insures optimal graft perfusion. The nonsuture cuff technique allows for successful microvascular anastomosis by a single surgeon with an average overall operative time of 2.5 h. Long-term graft survival (>30 days) was observed in syngeneic transplants. We have established a robust murine model with ideal vascular perfusion of penile tissue to study the unique immunobiology of male genitourinary allotransplantation. Heterotopic inset further allows for visual monitoring of graft viability, while the native penis serves as an optimal control.


Subject(s)
Plastic Surgery Procedures , Vascularized Composite Allotransplantation , Anastomosis, Surgical , Animals , Male , Mice , Penis/surgery , Rats , Transplantation, Homologous
9.
Transplantation ; 103(11): 2245-2254, 2019 11.
Article in English | MEDLINE | ID: mdl-31574039

ABSTRACT

BACKGROUND: Penis transplantation represents an exciting new avenue for restoration of male urogenitalia. However, little is known about the specific immunological features of penile transplants, limiting their application in complex urogenital reconstruction. To properly study this emerging form of transplantation, adequate preclinical models are a necessity. The purpose of this study is to establish a clinical and histological rejection classification of urogenital tissue transplants using a new rat heterotopic penile transplant model that includes preputial skin. METHODS: Syngeneic and allogeneic heterotopic penile transplantations were performed on Lewis and Brown Norway rats using a new model designed by our group. Grafts were clinically and histologically monitored at postoperative days (POD) 3-30. RESULTS: Six syngeneic and 25 allogeneic transplants were performed. All syngeneic and tacrolimus-treated grafts survived until endpoint. Allogeneic graft rejection is shown to follow a 4-stage clinical progression with all untreated allografts developing epidermal sloughing at POD7 and full rejecting between POD14 and POD16. Histological samples were used to develop a specific 4-grade rejection classification analogous to the 2007 Banff Criteria for skin-containing allografts. CONCLUSIONS: Graft skin and urethral lining tissue are first rejection targets followed by tunica albuginea and corpora cavernosa in a distal to proximal pattern. We established a robust and reproducible murine model to study the immunobiology of male genital tissue in the context of transplantation and developed a novel 4-grade clinical and histological rejection scale based on graft skin and urethral lining as the main targets of rejection.


Subject(s)
Graft Rejection/immunology , Graft Survival/immunology , Microsurgery/methods , Organ Transplantation/methods , Penile Transplantation , Animals , Inflammation , Male , Models, Animal , Postoperative Period , Rats , Rats, Inbred BN , Rats, Inbred Lew , Transplantation, Heterotopic , Transplantation, Homologous , Transplantation, Isogeneic
10.
Brain ; 142(11): 3456-3472, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31529023

ABSTRACT

The immunological barrier currently precludes the clinical utilization of allogeneic stem cells. Although glial-restricted progenitors have become attractive candidates to treat a wide variety of neurological diseases, their survival in immunocompetent recipients is limited. In this study, we adopted a short-term, systemically applicable co-stimulation blockade-based strategy using CTLA4-Ig and anti-CD154 antibodies to modulate T-cell activation in the context of allogeneic glial-restricted progenitor transplantation. We found that co-stimulation blockade successfully prevented rejection of allogeneic glial-restricted progenitors from immunocompetent mouse brains. The long-term engrafted glial-restricted progenitors myelinated dysmyelinated adult mouse brains within one month. Furthermore, we identified a set of plasma miRNAs whose levels specifically correlated to the dynamic changes of immunoreactivity and as such could serve as biomarkers for graft rejection or tolerance. We put forward a successful strategy to induce alloantigen-specific hyporesponsiveness towards stem cells in the CNS, which will foster effective therapeutic application of allogeneic stem cells.


Subject(s)
Immune Tolerance , Microglia/immunology , Microglia/transplantation , Myelin Sheath , Neural Stem Cells/immunology , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , Adoptive Transfer , Allografts , Animals , Cytokines/biosynthesis , Graft Rejection , Lymphocyte Culture Test, Mixed , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , T-Lymphocytes/immunology , Transplantation, Homologous
11.
J Clin Invest ; 128(11): 4770-4786, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30277476

ABSTRACT

The targeted delivery of therapeutic drugs to lymph nodes (LNs) provides an unprecedented opportunity to improve the outcomes of transplantation and immune-mediated diseases. The high endothelial venule is a specialized segment of LN vasculature that uniquely expresses peripheral node addressin (PNAd) molecules. PNAd is recognized by MECA79 mAb. We previously generated a MECA79 mAb-coated microparticle (MP) that carries tacrolimus. Although this MP trafficked to LNs, it demonstrated limited therapeutic efficacy in our transplant model. Here, we have synthesized a nanoparticle (NP) as a carrier of anti-CD3, and optimized the conjugation strategy to coat the NP surface with MECA79 mAb (MECA79-anti-CD3-NP) to enhance LN accumulation. As compared with nonconjugated NPs, a significantly higher quantity of MECA79-NPs accumulated in the draining lymph node (DLN). Many MECA79-NPs underwent internalization by T cells and dendritic cells within the LNs. Short-term treatment of murine cardiac allograft recipients with MECA79-anti-CD3-NP resulted in significantly prolonged allograft survival in comparison with the control groups. Prolonged graft survival following treatment with MECA79-anti-CD3-NP was characterized by a significant increase in intragraft and DLN Treg populations. Treg depletion abrogated the prolongation of heart allograft survival. We believe this targeted approach of drug delivery could redefine the methods of administering immune therapeutics in transplantation.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Drug Carriers/pharmacology , Graft Survival/drug effects , Heart Transplantation , Lymph Nodes/immunology , Nanoparticles/therapeutic use , Tacrolimus/pharmacology , Allografts , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Dendritic Cells/immunology , Dendritic Cells/pathology , Drug Carriers/chemistry , Graft Survival/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Nanoparticles/chemistry , T-Lymphocytes/immunology , Tacrolimus/chemistry
12.
Transplantation ; 102(4): 593-600, 2018 04.
Article in English | MEDLINE | ID: mdl-29298238

ABSTRACT

BACKGROUND: Candidates for vascularized composite allotransplantation (VCA) are frequently sensitized, putting them at risk for antibody-mediated rejection. Current desensitization strategies are imperfect and require a living-donor setting. Here we investigated the impact of sensitization on and the efficacy of a desensitization protocol utilizing syngeneic hematopoietic stem cell transplantation (HSCT) to prevent antibody-mediated rejection in VCA. METHODS: Skin transplants from Dark Agouti to Lewis rats were performed for sensitization. Orthotopic hind limb transplants from Dark Agouti donors were performed to sensitized and nonsensitized recipients, and the animals were treated with either daily tacrolimus or no immunosuppression. A desensitization protocol consisting of total body irradiation, fludarabine, and syngeneic HSCT was applied to sensitized animals. Graft rejection was monitored by clinical assessment and histological analysis. Serum levels of donor-specific antibodies (DSA IgG) were measured using flow cytometry. RESULTS: Sensitized recipients exhibited accelerated rejection by 5.5 ± 1.2 days without immunosuppression and 10.2 ± 3.6 days with daily tacrolimus compared with 8.7 ± 1.2 days and longer than 30 days in nonsensitized recipients, respectively. Serum levels of DSA IgG were markedly elevated (37.3 ± 3.34-fold from baseline) in sensitized recipients after VCA and correlated with histologic evidence of rejection and C4d deposition. Desensitization significantly reduced DSA compared with sensitized controls (2.6 ± 0.5-fold vs 6.0 ± 1.2-fold, P < 0.01) and along with daily tacrolimus led to improved VCA survival longer than 30 days without evidence of C4d deposition (n = 6). CONCLUSIONS: In summary, sensitization leads to accelerated rejection of VCA, and syngeneic HSCT combined with conventional immunosuppression effectively reduces DSA and improves allograft survival in sensitized rats.


Subject(s)
Composite Tissue Allografts/blood supply , Composite Tissue Allografts/transplantation , Desensitization, Immunologic/methods , Graft Rejection/prevention & control , Hematopoietic Stem Cell Transplantation/methods , Hindlimb/blood supply , Hindlimb/transplantation , Isoantibodies/immunology , Skin Transplantation/methods , Vascularized Composite Allotransplantation/methods , Animals , Complement C4b/immunology , Desensitization, Immunologic/adverse effects , Graft Rejection/blood , Graft Rejection/immunology , Graft Survival , Hematopoietic Stem Cell Transplantation/adverse effects , Immunosuppressive Agents/administration & dosage , Isoantibodies/blood , Male , Models, Animal , Myeloablative Agonists/administration & dosage , Peptide Fragments/immunology , Rats, Inbred Lew , Skin Transplantation/adverse effects , Tacrolimus/administration & dosage , Time Factors , Transplantation, Isogeneic , Vascularized Composite Allotransplantation/adverse effects , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
13.
J Med Chem ; 60(16): 7186-7198, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28759224

ABSTRACT

Aberrant excitatory neurotransmission associated with overproduction of glutamate has been implicated in the development of HIV-associated neurocognitive disorders (HAND). The glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON, 14) attenuates glutamate synthesis in HIV-infected microglia/macrophages, offering therapeutic potential for HAND. We show that 14 prevents manifestation of spatial memory deficits in chimeric EcoHIV-infected mice, a model of HAND. 14 is not clinically available, however, because its development was hampered by peripheral toxicities. We describe the synthesis of several substituted N-(pivaloyloxy)alkoxy-carbonyl prodrugs of 14 designed to circulate inert in plasma and be taken up and biotransformed to 14 in the brain. The lead prodrug, isopropyl 6-diazo-5-oxo-2-(((phenyl(pivaloyloxy)methoxy)carbonyl)amino)hexanoate (13d), was stable in swine and human plasma but liberated 14 in swine brain homogenate. When dosed systemically in swine, 13d provided a 15-fold enhanced CSF-to-plasma ratio and a 9-fold enhanced brain-to-plasma ratio relative to 14, opening a possible clinical path for the treatment of HAND.


Subject(s)
Aminocaproates/pharmacology , Azo Compounds/pharmacology , Diazooxonorleucine/pharmacology , Neurocognitive Disorders/drug therapy , Nootropic Agents/pharmacology , Prodrugs/pharmacology , Aminocaproates/administration & dosage , Aminocaproates/chemical synthesis , Animals , Azo Compounds/administration & dosage , Azo Compounds/chemical synthesis , Blood/metabolism , Brain/metabolism , Diazooxonorleucine/administration & dosage , Drug Stability , Female , Glutamic Acid/metabolism , Glutaminase/antagonists & inhibitors , HIV Infections/complications , Humans , Male , Mice, Inbred C57BL , Neurocognitive Disorders/etiology , Nootropic Agents/administration & dosage , Nootropic Agents/chemical synthesis , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Swine , Viral Load/drug effects
14.
J Vis Exp ; (119)2017 01 02.
Article in English | MEDLINE | ID: mdl-28117792

ABSTRACT

Murine full-thickness skin transplantation is a well-established in vivo model to study alloimmune response and graft rejection. Despite its limited application to humans, skin transplantation in mice has been widely employed for transplantation research. The procedure is easy to learn and perform, and it does not require delicate microsurgical techniques nor extensive training. Moreover, graft rejection in this model occurs in a very reproducible immunological reaction and is easily monitored by direct inspection and palpation. In addition, secondary skin transplantation with donor-matched or third-party skin grafts can be performed on more complex transplant models as an alternative and uncomplicated method to assess donor-specific tolerance. The complications are low and are in general limited to anesthesia overdose or respiratory distress after the procedure. Graft failure, on the other hand, occurs commonly as a result of poor preparation of the graft, incorrect positioning in the graft bed, or inappropriate placement of the bandage. In this article, we present a protocol for full-thickness skin transplantation in mice and describe the important steps necessary for a successful procedure.


Subject(s)
Skin Transplantation/methods , Animals , Cyclosporine/pharmacology , Graft Rejection/immunology , Immunosuppressive Agents/pharmacology , Mice, Inbred BALB C , Mice, Inbred C57BL , Sirolimus/pharmacology , Skin Transplantation/instrumentation , Skin Transplantation/mortality , Transplantation, Homologous/methods
15.
Plast Reconstr Surg Glob Open ; 5(12): e1595, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29632774

ABSTRACT

BACKGROUND: Congenital and acquired chest wall deformities represent a significant challenge to functional reconstruction and may impact feasibility of heart transplantation for patients with end-stage organ failure. In the recent past, the concept of replacing like-with-like tissue by using vascularized composite allografts (VCA) has been enthusiastically employed for reconstruction of complex tissue defects. METHODS: In this study, we introduce a novel murine model for en bloc chest wall, heart, and thymus transplantation and thereby the use of complex tissue allografts for reconstruction of both chest wall defects and also end-stage organ failure. Additionally, this model allows us to study the features of combined vascularized bone marrow (VBM), thymus, and heart transplantation on allograft survival and function. Heterotopic chest wall, thymus, and heart transplants were performed in untreated syngeneic and allogeneic combinations and in allogeneic combinations treated with costimulation blockade (CTLA4-Ig and MR-1). RESULTS: Indefinite (ie, 150 d, N = 3) graft survival was observed in syngeneic controls. In untreated recipients of allogeneic grafts, the skin component was rejected after 10 (±1) days, whereas rejection of the heart occurred after 13 (± 1) days (N = 3). Costimulation blockade treatment prolonged survival of the heart and chest wall component (130 d, N = 3) as well as the VBM niche as evidenced by donor-specific chimerism (average: 2.35 ± 1.44%), whereas interestingly, the skin component was rejected after 13 (±1) days. CONCLUSION: Thus, this novel microsurgical model of VCA combined with solid organ transplantation is technically feasible and results in split tolerance when treated with costimulatory blockade.

16.
Sci Rep ; 6: 31306, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27510321

ABSTRACT

This study investigates the efficacy of local and intravenous mesenchymal stem cell (MSC) administration to augment neuroregeneration in both a sciatic nerve cut-and-repair and rat hindlimb transplant model. Bone marrow-derived MSCs were harvested and purified from Brown-Norway (BN) rats. Sciatic nerve transections and repairs were performed in three groups of Lewis (LEW) rats: negative controls (n = 4), local MSCs (epineural) injection (n = 4), and systemic MSCs (intravenous) injection (n = 4). Syngeneic (LEW-LEW) (n = 4) and allogeneic (BN-LEW) (n = 4) hindlimb transplants were performed and assessed for neuroregeneration after local or systemic MSC treatment. Rats undergoing sciatic nerve cut-and-repair and treated with either local or systemic injection of MSCs had significant improvement in the speed of recovery of compound muscle action potential amplitudes and axon counts when compared with negative controls. Similarly, rats undergoing allogeneic hindlimb transplants treated with local injection of MSCs exhibited significantly increased axon counts. Similarly, systemic MSC treatment resulted in improved nerve regeneration following allogeneic hindlimb transplants. Systemic administration had a more pronounced effect on electromotor recovery while local injection was more effective at increasing fiber counts, suggesting different targets of action. Local and systemic MSC injections significantly improve the pace and degree of nerve regeneration after nerve injury and hindlimb transplantation.


Subject(s)
Hindlimb/transplantation , Mesenchymal Stem Cell Transplantation/methods , Nerve Regeneration , Peripheral Nervous System Neoplasms/therapy , Sciatic Nerve/injuries , Animals , Combined Modality Therapy , Disease Models, Animal , Rats , Rats, Inbred BN , Rats, Inbred Lew , Transplantation, Homologous , Treatment Outcome , Wound Healing
17.
J Vis Exp ; (108): 53483, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26967527

ABSTRACT

In vivo animal model systems, and in particular mouse models, have evolved into powerful and versatile scientific tools indispensable to basic and translational research in the field of transplantation medicine. A vast array of reagents is available exclusively in this setting, including mono- and polyclonal antibodies for both diagnostic and interventional applications. In addition, a vast number of genotyped, inbred, transgenic, and knock out strains allow detailed investigation of the individual contributions of humoral and cellular components to the complex interplay of an immune response and make the mouse the gold standard for immunological research. Vascularized Composite Allotransplantation (VCA) delineates a novel field of transplantation using allografts to replace "like with like" in patients suffering traumatic or congenital tissue loss. This surgical methodological protocol shows the use of a non-suture cuff technique for super-microvascular anastomosis in an orthotopic mouse hind limb transplantation model. The model specifically allows for comparison between established paradigms in solid organ transplantation with a novel form of transplants consisting of various different tissue components. Uniquely, this model allows for the transplantation of a viable vascularized bone marrow compartment and niche that have the potential to exert a beneficial effect on the balance of immune acceptance and rejection. This technique provides a tool to investigate alloantigen recognition and allograft rejection and acceptance, as well as enables the pursuit of functional nerve regeneration studies to further advance this novel field of transplantation.


Subject(s)
Hindlimb/transplantation , Plastic Surgery Procedures/methods , Allografts , Anastomosis, Surgical/methods , Animals , Graft Rejection/immunology , Hindlimb/blood supply , Mice , Microsurgery/methods , Models, Biological , Suture Techniques , Transplantation, Homologous/methods
18.
J Vis Exp ; (107): e53442, 2016 Jan 23.
Article in English | MEDLINE | ID: mdl-26863343

ABSTRACT

Exploration of novel strategies in organ transplantation to prolong allograft survival and minimizing the need for long-term maintenance immunosuppression must be pursued. Employing vascularized bone marrow transplantation and co-transplantation of the thymus have shown promise in this regard in various animal models. Vascularized bone marrow transplantation allows for the uninterrupted transfer of donor bone marrow cells within the preserved donor microenvironment, and the incorporation of thymus tissue with vascularized bone marrow transplantation has shown to increase T-cell chimerism ultimately playing a supportive role in the induction of immune regulation. The combination of solid organ and vascularized composite allotransplantation can uniquely combine these strategies in the form of a novel transplant model. Murine models serve as an excellent paradigm to explore the mechanisms of acute and chronic rejection, chimerism, and tolerance induction, thus providing the foundation to propagate superior allograft survival strategies for larger animal models and future clinical application. Herein, we developed a novel heterotopic en bloc chest wall, thymus, and heart transplant model in mice using a cervical non-suture cuff technique. The experience in syngeneic and allogeneic transplant settings is described for future broader immunological investigations via an instructional manuscript and video supplement.


Subject(s)
Heart Transplantation/methods , Models, Animal , Thoracic Wall/transplantation , Thymus Gland/transplantation , Transplantation, Homologous/methods , Animals , Graft Survival , Mice
19.
Nat Nanotechnol ; 11(1): 95-102, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26524396

ABSTRACT

Many surgeries are complicated by the need to anastomose, or reconnect, micrometre-scale vessels. Although suturing remains the gold standard for anastomosing vessels, it is difficult to place sutures correctly through collapsed lumen, making the procedure prone to failure. Here, we report a multiphase transitioning peptide hydrogel that can be injected into the lumen of vessels to facilitate suturing. The peptide, which contains a photocaged glutamic acid, forms a solid-like gel in a syringe and can be shear-thin delivered to the lumen of collapsed vessels (where it distends the vessel) and the space between two vessels (where it is used to approximate the vessel ends). Suturing is performed directly through the gel. Light is used to initiate the final gel-sol phase transition that disrupts the hydrogel network, allowing the gel to be removed and blood flow to resume. This gel adds a new tool to the armamentarium for micro- and supermicrosurgical procedures.


Subject(s)
Hydrogels/chemistry , Peptides/chemistry , Suture Techniques , Sutures , Tissue Adhesives/chemical synthesis , Vascular Surgical Procedures/methods , Animals , Equipment Design , Equipment Failure Analysis , Femoral Artery/drug effects , Femoral Artery/surgery , Hydrogels/administration & dosage , Hydrogels/radiation effects , Light , Materials Testing , Mice , Microsurgery/instrumentation , Microsurgery/methods , Peptides/administration & dosage , Peptides/radiation effects , Phase Transition/radiation effects , Vascular Surgical Procedures/instrumentation , Viscosity
20.
Cell Rep ; 13(4): 760-770, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26489460

ABSTRACT

Upon antigen recognition and co-stimulation, T lymphocytes upregulate the metabolic machinery necessary to proliferate and sustain effector function. This metabolic reprogramming in T cells regulates T cell activation and differentiation but is not just a consequence of antigen recognition. Although such metabolic reprogramming promotes the differentiation and function of T effector cells, the differentiation of regulatory T cells employs different metabolic reprogramming. Therefore, we hypothesized that inhibition of glycolysis and glutamine metabolism might prevent graft rejection by inhibiting effector generation and function and promoting regulatory T cell generation. We devised an anti-rejection regimen involving the glycolytic inhibitor 2-deoxyglucose (2-DG), the anti-type II diabetes drug metformin, and the inhibitor of glutamine metabolism 6-diazo-5-oxo-L-norleucine (DON). Using this triple-drug regimen, we were able to prevent or delay graft rejection in fully mismatched skin and heart allograft transplantation models.


Subject(s)
Graft Rejection/prevention & control , Allografts , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Deoxyglucose/therapeutic use , Diazooxonorleucine/therapeutic use , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glutamine/metabolism , Glycolysis/drug effects , Glycolysis/physiology , Heart Transplantation/methods , Metformin/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Transgenic , Phosphorylation/drug effects , Phosphorylation/physiology , T-Lymphocytes, Regulatory
SELECTION OF CITATIONS
SEARCH DETAIL
...