Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(10): e2111537119, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35238643

ABSTRACT

Ischemia reperfusion injury represents a common pathological condition that is triggered by the release of endogenous ligands. While neutrophils are known to play a critical role in its pathogenesis, the tissue-specific spatiotemporal regulation of ischemia-reperfusion injury is not understood. Here, using oxidative lipidomics and intravital imaging of transplanted mouse lungs that are subjected to severe ischemia reperfusion injury, we discovered that necroptosis, a nonapoptotic form of cell death, triggers the recruitment of neutrophils. During the initial stages of inflammation, neutrophils traffic predominantly to subpleural vessels, where their aggregation is directed by chemoattractants produced by nonclassical monocytes that are spatially restricted in this vascular compartment. Subsequent neutrophilic disruption of capillaries resulting in vascular leakage is associated with impaired graft function. We found that TLR4 signaling in vascular endothelial cells and downstream NADPH oxidase 4 expression mediate the arrest of neutrophils, a step upstream of their extravasation. Neutrophil extracellular traps formed in injured lungs and their disruption with DNase prevented vascular leakage and ameliorated primary graft dysfunction. Thus, we have uncovered mechanisms that regulate the initial recruitment of neutrophils to injured lungs, which result in selective damage to subpleural pulmonary vessels and primary graft dysfunction. Our findings could lead to the development of new therapeutics that protect lungs from ischemia reperfusion injury.


Subject(s)
Endothelium, Vascular/metabolism , Lung/metabolism , Necroptosis , Neutrophil Infiltration , Neutrophils/metabolism , Reperfusion Injury/metabolism , Animals , Endothelium, Vascular/injuries , Humans , Lung/blood supply , Mice , Mice, Knockout , Reperfusion Injury/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
2.
J Clin Invest ; 130(12): 6718-6727, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33196461

ABSTRACT

Tertiary lymphoid organs are aggregates of immune and stromal cells including high endothelial venules and lymphatic vessels that resemble secondary lymphoid organs and can be induced at nonlymphoid sites during inflammation. The function of lymphatic vessels within tertiary lymphoid organs remains poorly understood. During lung transplant tolerance, Foxp3+ cells accumulate in tertiary lymphoid organs that are induced within the pulmonary grafts and are critical for the local downregulation of alloimmune responses. Here, we showed that tolerant lung allografts could induce and maintain tolerance of heterotopic donor-matched hearts through pathways that were dependent on the continued presence of the transplanted lung. Using lung retransplantation, we showed that Foxp3+ cells egressed from tolerant lung allografts via lymphatics and were recruited into donor-matched heart allografts. Indeed, survival of the heart allografts was dependent on lymphatic drainage from the tolerant lung allograft to the periphery. Thus, our work indicates that cellular trafficking from tertiary lymphoid organs regulates immune responses in the periphery. We propose that these findings have important implications for a variety of disease processes that are associated with the induction of tertiary lymphoid organs.


Subject(s)
Bronchi/immunology , Lung Transplantation , Lung/immunology , Lymphoid Tissue/immunology , Transplantation Tolerance , Animals , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Transgenic , Transplantation, Homologous
3.
Am J Transplant ; 20(5): 1251-1261, 2020 05.
Article in English | MEDLINE | ID: mdl-31721409

ABSTRACT

Long-term survival after lung transplantation remains profoundly limited by graft rejection. Recent work has shown that bronchus-associated lymphoid tissue (BALT), characterized by the development of peripheral nodal addressin (PNAd)-expressing high endothelial venules and enriched in B and Foxp3+ T cells, is important for the maintenance of allograft tolerance. Mechanisms underlying BALT induction in tolerant pulmonary allografts, however, remain poorly understood. Here, we show that the development of PNAd-expressing high endothelial venules within intragraft lymphoid follicles and the recruitment of B cells, but not Foxp3+ cells depends on IL-22. We identify graft-infiltrating gamma-delta (γδ) T cells and Type 3 innate lymphoid cells (ILC3s) as important producers of IL-22. Reconstitution of IL-22 at late time points through retransplantation into wildtype hosts mediates B cell recruitment into lymphoid follicles within the allograft, resulting in a significant increase in their size, but does not induce PNAd expression. Our work has identified cellular and molecular requirements for the induction of BALT in pulmonary allografts during tolerance induction and may provide a platform for the development of new therapies for lung transplant patients.


Subject(s)
Immunity, Innate , Lymphoid Tissue , Allografts , Bronchi , Graft Rejection/etiology , Humans , Interleukins , Lung , Lymphocytes , Interleukin-22
4.
J Clin Invest ; 129(2): 556-568, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30561386

ABSTRACT

Antibody-mediated rejection (AMR) is a principal cause of acute and chronic failure of lung allografts. However, mechanisms mediating this oftentimes fatal complication are poorly understood. Here, we show that Foxp3+ T cells formed aggregates in rejection-free human lung grafts and accumulated within induced bronchus-associated lymphoid tissue (BALT) of tolerant mouse lungs. Using a retransplantation model, we show that selective depletion of graft-resident Foxp3+ T lymphocytes resulted in the generation of donor-specific antibodies (DSA) and AMR, which was associated with complement deposition and destruction of airway epithelium. AMR was dependent on graft infiltration by B and T cells. Depletion of graft-resident Foxp3+ T lymphocytes resulted in prolonged interactions between B and CD4+ T cells within transplanted lungs, which was dependent on CXCR5-CXCL13. Blockade of CXCL13 as well as inhibition of the CD40 ligand and the ICOS ligand suppressed DSA production and prevented AMR. Thus, we have shown that regulatory Foxp3+ T cells residing within BALT of tolerant pulmonary allografts function to suppress B cell activation, a finding that challenges the prevailing view that regulation of humoral responses occurs peripherally. As pulmonary AMR is largely refractory to current immunosuppression, our findings provide a platform for developing therapies that target local immune responses.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , B-Lymphocytes , Bronchi , Graft Rejection , Lung Transplantation , Lymphocyte Activation , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Bronchi/immunology , Bronchi/pathology , CD40 Ligand/genetics , CD40 Ligand/immunology , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
5.
J Neurosci Methods ; 204(1): 19-27, 2012 02 15.
Article in English | MEDLINE | ID: mdl-22027490

ABSTRACT

BACKGROUND: In order to evaluate nerve regeneration in clinically relevant hindlimb surgical paradigms not feasible in fluorescent mice models, we developed a rat that expresses green fluorescent protein (GFP) in neural tissue. METHODS: Transgenic Sprague-Dawley rat lines were created using pronuclear injection of a transgene expressing GFP under the control of the thy1 gene. Nerves were imaged under fluorescence microscopy and muscles were imaged with confocal microscopy to determine GFP expression following sciatic nerve crush, transection and direct suturing, and transection followed by repair with a nerve isograft from nonexpressing littermates. RESULTS: In each surgical paradigm, fluorescence microscopy demonstrated the loss and reappearance of fluorescence with regeneration of axons following injury. Nerve regeneration was confirmed with imaging of Wallerian degeneration followed by reinnervation of extensor digitorum longus (EDL) muscle motor endplates using confocal microscopy. CONCLUSION: The generation of a novel transgenic rat model expressing GFP in neural tissue allows in vivo imaging of nerve regeneration and visualization of motor endplate reinnervation. This rat provides a new model for studying peripheral nerve injury and regeneration over surgically relevant distances.


Subject(s)
Disease Models, Animal , Green Fluorescent Proteins/metabolism , Nerve Regeneration/physiology , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/physiopathology , Sciatic Neuropathy/pathology , Sciatic Neuropathy/physiopathology , Animals , Hindlimb/injuries , Hindlimb/innervation , Hindlimb/pathology , Rats , Rats, Sprague-Dawley , Rats, Transgenic
6.
Exp Neurol ; 225(2): 310-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20633557

ABSTRACT

Host Schwann cell (SC) migration into nerve allografts is the limiting factor in the duration of immunosuppression following peripheral nerve allotransplantation, and may be affected by different immunosuppressive regimens. Our objective was to compare SC migration patterns between clinical and experimental immunosuppression regimens both over time and at the harvest endpoint. Eighty mice that express GFP under the control of the Schwann cell specific S100 promoter were engrafted with allogeneic, nonfluorescent sciatic nerve grafts. Mice received immunosuppression with either tacrolimus (FK506), or experimental T-cell triple costimulation blockade (CSB), consisting of CTLA4-immunoglobulin fusion protein, anti-CD40 monoclonal antibody, and anti-inducible costimulator monoclonal antibody. Migration of GFP-expressing host SCs into wild-type allografts was assessed in vivo every 3 weeks until 15 weeks postoperatively, and explanted allografts were evaluated for immunohistochemical staining patterns to differentiate graft from host SCs. Immunosuppression with tacrolimus exhibited a plateau of SC migration, characterized by significant early migration (< 3 weeks) followed by a constant level of host SCs in the graft (15 weeks). At the endpoint, graft fluorescence was decreased relative to surrounding host nerve, and donor SCs persisted within the graft. CSB-treated mice displayed gradually increasing migration of host SCs into the graft, without the plateau noted in tacrolimus-treated mice, and also maintained a population of donor SCs at the 15-week endpoint. SC migration patterns are affected by immunosuppressant choice, particularly in the immediate postoperative period, and the use of a single treatment of CSB may allow for gradual population of nerve allografts with host SCs.


Subject(s)
Cell Movement/physiology , Nerve Regeneration/physiology , Schwann Cells/physiology , Sciatic Nerve/transplantation , Analysis of Variance , Animals , Antibodies, Monoclonal/pharmacology , Blotting, Western , CD40 Antigens/immunology , Caspase 3/immunology , Caspase 3/metabolism , Cell Movement/immunology , Immunohistochemistry , Immunosuppression Therapy/methods , Immunosuppressive Agents/pharmacology , Mice , Mice, Transgenic , Nerve Regeneration/immunology , S100 Proteins/immunology , S100 Proteins/metabolism , Schwann Cells/immunology , Sciatic Nerve/immunology , Sciatic Nerve/physiology , Tacrolimus/pharmacology
7.
Arch Facial Plast Surg ; 12(1): 40-7, 2010.
Article in English | MEDLINE | ID: mdl-20083740

ABSTRACT

OBJECTIVE: To determine the Sunderland classification of a bipolar electrocautery injury. METHODS: Twenty-two rats received crush (a reproducible Sunderland second-degree injury) or bipolar electrocautery injury and were evaluated for functional, histomorphometric, and immunohistochemical recovery at 21 or 42 days. Animal experiments were performed between July 3 and December 12, 2007. Axonal regeneration and end plate reinnervation were evaluated in double transgenic cyan fluorescent protein-conjugated Thy1 and green fluorescent protein-conjugated S100 mice. RESULTS: Compared with crush injury, bipolar electrocautery injury caused greater disruption of myelin and neurofilament architecture at the injury site and decreased nerve fiber counts and percentage of neural tissue distal to the injury (P =.007). Complete functional recovery was seen after crush but not bipolar electrocautery injury. Serial live imaging demonstrated axonal regeneration at week 1 after crush and at week 3 after bipolar electrocautery injury. Qualitative assessment of motor end plate reinnervation at 42 days demonstrated complete neuromuscular end plate reinnervation in the crush group and only limited reinnervation in the bipolar electrocautery group. CONCLUSION: Bipolar electrocautery injury in a rodent model resulted in a Sunderland third-degree injury, characterized by gradual, incomplete recovery without intervention.


Subject(s)
Electrocoagulation/methods , Peripheral Nerve Injuries , Peripheral Nerves/surgery , Plastic Surgery Procedures/methods , Animals , Male , Mice , Peripheral Nerves/pathology , Rats , Rats, Inbred Lew , Severity of Illness Index
8.
J Neurosurg ; 113(1): 102-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19943736

ABSTRACT

OBJECT: Glial cell line-derived neurotrophic factor (GDNF) has potent survival effects on central and peripheral nerve populations. The authors examined the differential effects of GDNF following either a sciatic nerve crush injury in mice that overexpressed GDNF in the central or peripheral nervous systems (glial fibrillary acidic protein [GFAP]-GDNF) or in the muscle target (Myo-GDNF). METHODS: Adult mice (GFAP-GDNF, Myo-GDNF, or wild-type [WT] animals) underwent sciatic nerve crush and were evaluated using histomorphometry and muscle force and power testing. Uninjured WT animals served as controls. RESULTS: In the sciatic nerve crush, the Myo-GDNF mice demonstrated a higher number of nerve fibers, fiber density, and nerve percentage (p < 0.05) at 2 weeks. The early regenerative response did not result in superlative functional recovery. At 3 weeks, GFAP-GDNF animals exhibit fewer nerve fibers, decreased fiber width, and decreased nerve percentage compared with WT and Myo-GDNF mice (p < 0.05). By 6 weeks, there were no significant differences between groups. CONCLUSIONS: Peripheral delivery of GDNF resulted in earlier regeneration following sciatic nerve crush injuries than that with central GDNF delivery. Treatment with neurotrophic factors such as GDNF may offer new possibilities for the treatment of peripheral nerve injury.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Motor Endplate/chemistry , Nerve Regeneration/drug effects , Sciatic Nerve/chemistry , Animals , Isometric Contraction/drug effects , Mice , Mice, Transgenic , Nerve Crush
9.
Muscle Nerve ; 39(6): 787-99, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19291791

ABSTRACT

Autografting is the gold standard in the repair of peripheral nerve injuries that are not amenable to end-to-end coaptation. However, because autografts result in donor-site defects and are a limited resource, an effective substitute would be valuable. In a rat model, we compared isografts with Integra NeuraGen (NG) nerve guides, which are a commercially available type I collagen conduit, with processed rat allografts comparable to AxoGen's Avance human decellularized allograft product. In a 14-mm sciatic nerve gap model, isograft was superior to processed allograft, which was in turn superior to NG conduit at 6 weeks postoperatively (P < 0.05 for number of myelinated fibers both at midgraft and distal to the graft). At 12 weeks, these differences were no longer apparent. In a 28-mm graft model, isografts again performed better than processed allografts at both 6 and 22 weeks; regeneration through the NG conduit was often insufficient for analysis in this long graft model. Functional tests confirmed the superiority of isografts, although processed allografts permitted successful reinnervation of distal targets not seen in the NG conduit groups. Processed allografts were inherently non-immunogenic and maintained some internal laminin structure. We conclude that, particularly in a long gap model, nerve graft alternatives fail to confer the regenerative advantages of an isograft. However, AxoGen processed allografts are superior to a currently available conduit-style nerve guide, the Integra NeuraGen. They provide an alternative for reconstruction of short nerve gaps where a conduit might otherwise be used.


Subject(s)
Absorbable Implants , Collagen Type I/pharmacology , Neurosurgical Procedures/methods , Peripheral Nerves/surgery , Peripheral Nerves/transplantation , Transplantation, Homologous/methods , Animals , Collagen Type I/therapeutic use , Disease Models, Animal , Growth Cones/physiology , Laminin/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/ultrastructure , Nerve Fibers, Unmyelinated/metabolism , Nerve Fibers, Unmyelinated/ultrastructure , Nerve Regeneration/physiology , Peripheral Nerve Injuries , Rats , Rats, Inbred Lew , Recovery of Function/physiology , Sciatic Neuropathy/surgery , Transplantation Tolerance/physiology , Treatment Outcome
10.
Exp Neurol ; 211(2): 539-50, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18433746

ABSTRACT

End-to-side (ETS) nerve repair remains an area of intense scrutiny for peripheral nerve surgeon-scientists. In this technique, the transected end of an injured nerve, representing the "recipient" is sutured to the side of an uninjured "donor" nerve. Some works suggest that the recipient limb is repopulated with regenerating collateral axonal sprouts from the donor nerve that go on to form functional synapses. Significant, unresolved questions include whether the donor nerve needs to be injured to facilitate regeneration, and whether a single donor neuron is capable of projecting additional axons capable of differentially innervating disparate targets. We serially imaged living transgenic mice (n=66) expressing spectral variants of GFP in various neuronal subsets after undergoing previously described atraumatic, compressive, or epineurotomy forms of ETS repair (n=22 per group). To evaluate the source, and target innervation of these regenerating axons, nerve morphometry and retrograde labeling were further supplemented by confocal microscopy as well as Western blot analysis. Either compression or epineurotomy with inevitable axotomy were required to facilitate axonal regeneration into the recipient limb. Progressively more injurious models were associated with improved recipient nerve reinnervation (epineurotomy: 184+/-57.6 myelinated axons; compression: 78.9+/-13.8; atraumatic: 0), increased Schwann cell proliferation (epineurotomy: 72.2% increase; compression: 39% increase) and cAMP response-element binding protein expression at the expense of a net deficit in donor axon counts distal to the repair. These differences were manifest by 150 days, at which point quantitative evidence for pruning was obtained. We conclude that ETS repair relies upon injury to the donor nerve.


Subject(s)
Axons/physiology , Nerve Crush/methods , Nerve Regeneration/physiology , Neurosurgical Procedures/methods , Animals , Axotomy/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microsurgery/methods , Tissue Transplantation/methods
11.
Exp Neurol ; 207(1): 128-38, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17628544

ABSTRACT

We propose that double-transgenic thy1-CFP(23)/S100-GFP mice whose Schwann cells constitutively express green fluorescent protein (GFP) and axons express cyan fluorescent protein (CFP) can be used to serially evaluate the temporal relationship between nerve regeneration and Schwann cell migration through acellular nerve grafts. Thy1-CFP(23)/S100-GFP and S100-GFP mice received non-fluorescing cold preserved nerve allografts from immunologically disparate donors. In vivo fluorescent imaging of these grafts was then performed at multiple points. The transected sciatic nerve was reconstructed with a 1-cm nerve allograft harvested from a Balb-C mouse and acellularized via 7 weeks of cold preservation prior to transplantation. The presence of regenerated axons and migrating Schwann cells was confirmed with confocal and electron microscopy on fixed tissue. Schwann cells migrated into the acellular graft (163+/-15 intensity units) from both proximal and distal stumps, and bridged the whole graft within 10 days (388+/-107 intensity units in the central 4-6 mm segment). Nerve regeneration lagged behind Schwann cell migration with 5 or 6 axons imaged traversing the proximal 4 mm of the graft under confocal microcopy within 10 days, and up to 21 labeled axons crossing the distal coaptation site by 15 days. Corroborative electron and light microscopy 5 mm into the graft demonstrated relatively narrow diameter myelinated (431+/-31) and unmyelinated (64+/-9) axons by 28 but not 10 days. Live imaging of the double-transgenic thy1-CFP(23)/S100-GFP murine line enabled serial assessment of Schwann cell-axonal relationships in traumatic nerve injuries reconstructed with acellular nerve allografts.


Subject(s)
Cell Movement , Cell-Free System/transplantation , Mice, Transgenic , Nerve Regeneration , Schwann Cells , Sciatic Nerve/physiopathology , Sciatic Nerve/surgery , Animals , Axons , Cell-Free System/pathology , Fluorescent Dyes , Green Fluorescent Proteins/genetics , Luminescent Agents , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic/genetics , Microscopy, Confocal , Microscopy, Electron , Neuromuscular Junction/pathology , Sciatic Nerve/pathology , Time Factors , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...