Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 729
Filter
2.
Hum Immunol ; 82(11): 838-849, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34404545

ABSTRACT

BACKGROUND AND PURPOSE: Currently there are no widely accepted guidelines for chimerism analysis testing in hematopoietic cell transplantation (HCT) patients. The objective of this review is to provide a practical guide to address key aspects of performing and utilizing chimerism testing results. In developing this guide, we conducted a survey of testing practices among laboratories that are accredited for performing engraftment monitoring/chimerism analysis by either the American Society for Histocompatibility & Immunogenetics (ASHI) and/or the European Federation of Immunogenetics (EFI). We interpreted the survey results in the light of pertinent literature as well as the experience in the laboratories of the authors. RECENT DEVELOPMENTS: In recent years there has been significant advances in high throughput molecular methods such as next generation sequencing (NGS) as well as growing access to these technologies in histocompatibility and immunogenetics laboratories. These methods have the potential to improve the performance of chimerism testing in terms of sensitivity, availability of informative genetic markers that distinguish donors from recipients as well as cost. SUMMARY: The results of the survey revealed a great deal of heterogeneity in chimerism testing practices among participating laboratories. The most consistent response indicated monitoring of engraftment within the first 30 days. These responses are reflective of published literature. Additional clinical indications included early detection of impending relapse as well as identification of cases of HLA-loss relapse.


Subject(s)
Hematopoietic Stem Cell Transplantation , High-Throughput Nucleotide Sequencing/statistics & numerical data , Histocompatibility Testing/statistics & numerical data , Laboratories, Clinical/statistics & numerical data , Practice Patterns, Physicians'/statistics & numerical data , Chimerism , High-Throughput Nucleotide Sequencing/standards , Histocompatibility Testing/methods , Histocompatibility Testing/standards , Humans , Laboratories, Clinical/standards , Practice Guidelines as Topic , Practice Patterns, Physicians'/standards , Surveys and Questionnaires/statistics & numerical data , Transplantation Chimera/genetics , Transplantation Chimera/immunology , Transplantation, Homologous
3.
Arch Immunol Ther Exp (Warsz) ; 69(1): 13, 2021 May 10.
Article in English | MEDLINE | ID: mdl-33970329

ABSTRACT

This study evaluated the efficacy of donor recipient chimeric cell (DRCC) therapy created by fusion of donor and recipient derived bone marrow cells (BMC) in chimerism and tolerance induction in a rat vascularized composite allograft (VCA) model. Twenty-four VCA (groin flaps) from MHC-mismatched ACI (RT1a) donors were transplanted to Lewis (RT1l) recipients. Rats were randomly divided into (n = 6/group): Group 1-untreated controls, Groups 2-7-day immunosuppression controls, Group 3-DRCC, and Group 4-DRCC with 7-day anti-αßTCR monoclonal antibody and cyclosporine A protocol. DRCC created by polyethylene glycol-mediated fusion of ACI and Lewis BMC were cultured and transplanted (2-4 × 106) to VCA recipients via intraosseous delivery route. Flow cytometry assessed peripheral blood chimerism while fluorescent microscopy and PCR tested the presence of DRCC in the recipient's blood, bone marrow (BM), and lymphoid organs at the study endpoint (VCA rejection). No complications were observed after DRCC intraosseous delivery. Group 4 presented the longest average VCA survival (79.3 ± 30.9 days) followed by Group 2 (53.3 ± 13.6 days), Group 3 (18 ± 7.5 days), and Group 1 (8.5 ± 1 days). The highest chimerism level was detected in Group 4 (57.9 ± 6.2%) at day 7 post-transplant. The chimerism declined at day 21 post-transplant and remained at 10% level during the entire follow-up period. Single dose of DRCC therapy induced long-term multilineage chimerism and extended VCA survival. DRCC introduces a novel concept of customized donor-recipient cell-based therapy supporting solid organ and VCA transplants.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow Transplantation/methods , Composite Tissue Allografts/transplantation , Graft Rejection/therapy , Transplantation Chimera/immunology , Animals , Composite Tissue Allografts/immunology , Disease Models, Animal , Graft Rejection/immunology , Graft Survival/immunology , Humans , Male , Rats , Tissue Donors , Transplant Recipients
4.
Front Immunol ; 12: 651515, 2021.
Article in English | MEDLINE | ID: mdl-33815412

ABSTRACT

Staphylococcus aureus is the predominant pathogen causing osteomyelitis. Unfortunately, no immunotherapy exists to treat these very challenging and costly infections despite decades of research, and numerous vaccine failures in clinical trials. This lack of success can partially be attributed to an overreliance on murine models where the immune correlates of protection often diverge from that of humans. Moreover, S. aureus secretes numerous immunotoxins with unique tropism to human leukocytes, which compromises the targeting of immune cells in murine models. To study the response of human immune cells during chronic S. aureus bone infections, we engrafted non-obese diabetic (NOD)-scid IL2Rγnull (NSG) mice with human hematopoietic stem cells (huNSG) and analyzed protection in an established model of implant-associated osteomyelitis. The results showed that huNSG mice have increases in weight loss, osteolysis, bacterial dissemination to internal organs, and numbers of Staphylococcal abscess communities (SACs), during the establishment of implant-associated MRSA osteomyelitis compared to NSG controls (p < 0.05). Flow cytometry and immunohistochemistry demonstrated greater human T cell numbers in infected versus uninfected huNSG mice (p < 0.05), and that T-bet+ human T cells clustered around the SACs, suggesting S. aureus-mediated activation and proliferation of human T cells in the infected bone. Collectively, these proof-of-concept studies underscore the utility of huNSG mice for studying an aggressive form of S. aureus osteomyelitis, which is more akin to that seen in humans. We have also established an experimental system to investigate the contribution of specific human T cells in controlling S. aureus infection and dissemination.


Subject(s)
Abscess/immunology , Osteolysis/immunology , Osteomyelitis/immunology , Prosthesis-Related Infections/immunology , Staphylococcal Infections/immunology , Abscess/microbiology , Abscess/pathology , Animals , Disease Models, Animal , Female , Hematopoietic Stem Cell Transplantation , Humans , Mice , Osteolysis/microbiology , Osteolysis/pathology , Osteomyelitis/microbiology , Osteomyelitis/pathology , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/pathology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/immunology , Transplantation Chimera/immunology
5.
Front Immunol ; 12: 622604, 2021.
Article in English | MEDLINE | ID: mdl-33732244

ABSTRACT

Induction of immune tolerance for solid organ and vascular composite allografts is the Holy Grail for transplantation medicine. This would obviate the need for life-long immunosuppression which is associated with serious adverse outcomes, such as infections, cancers, and renal failure. Currently the most promising means of tolerance induction is through establishing a mixed chimeric state by transplantation of donor hematopoietic stem cells; however, with the exception of living donor renal transplantation, the mixed chimerism approach has not achieved durable immune tolerance on a large scale in preclinical or clinical trials with other solid organs or vascular composite allotransplants (VCA). Ossium Health has established a bank of cryopreserved bone marrow (BM), termed "hematopoietic progenitor cell (HPC), Marrow," recovered from deceased organ donor vertebral bodies. This new source for hematopoietic cell transplant will be a valuable resource for treating hematological malignancies as well as for inducing transplant tolerance. In addition, we have discovered and developed a large source of mesenchymal stem (stromal) cells (MSC) tightly associated with the vertebral body bone fragment byproduct of the HPC, Marrow recovery process. Thus, these vertebral bone adherent MSC (vBA-MSC) are matched to the banked BM obtained from each donor, as opposed to third-party MSC, which enhances safety and potentially efficacy. Isolation and characterization of vBA-MSC from over 30 donors has demonstrated that the cells are no different than traditional BM-MSC; however, their abundance is >1,000-fold higher than obtainable from living donor BM aspirates. Based on our own unpublished data as well as reports published by others, MSC facilitate chimerism, especially at limiting hematopoietic stem and progenitor cell (HSPC) numbers and increase safety by controlling and/or preventing graft-vs.-host-disease (GvHD). Thus, vBA-MSC have the potential to facilitate mixed chimerism, promote complementary peripheral immunomodulatory functions and increase safety of BM infusions. Both HPC, Marrow and vBA-MSC have potential use in current VCA and solid organ transplant (SOT) tolerance clinical protocols that are amenable to "delayed tolerance." Current trials with HPC, Marrow are planned with subsequent phases to include vBA-MSC for tolerance of both VCA and SOT.


Subject(s)
Biological Specimen Banks , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Tissue Donors , Transplantation Tolerance , Animals , Bone Marrow Transplantation/adverse effects , Donor Selection , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival , Hematopoietic Stem Cell Transplantation/adverse effects , Histocompatibility , Humans , Immunosuppressive Agents/therapeutic use , Mesenchymal Stem Cell Transplantation/adverse effects , Phenotype , Transplantation Chimera/immunology , Treatment Outcome
6.
J Clin Invest ; 131(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33630757

ABSTRACT

In humans receiving intestinal transplantation (ITx), long-term multilineage blood chimerism often develops. Donor T cell macrochimerism (≥4%) frequently occurs without graft-versus-host disease (GVHD) and is associated with reduced rejection. Here we demonstrate that patients with macrochimerism had high graft-versus-host (GvH) to host-versus-graft (HvG) T cell clonal ratios in their allografts. These GvH clones entered the circulation, where their peak levels were associated with declines in HvG clones early after transplant, suggesting that GvH reactions may contribute to chimerism and control HvG responses without causing GVHD. Consistently, donor-derived T cells, including GvH clones, and CD34+ hematopoietic stem and progenitor cells (HSPCs) were simultaneously detected in the recipients' BM more than 100 days after transplant. Individual GvH clones appeared in ileal mucosa or PBMCs before detection in recipient BM, consistent with an intestinal mucosal origin, where donor GvH-reactive T cells expanded early upon entry of recipient APCs into the graft. These results, combined with cytotoxic single-cell transcriptional profiles of donor T cells in recipient BM, suggest that tissue-resident GvH-reactive donor T cells migrated into the recipient circulation and BM, where they destroyed recipient hematopoietic cells through cytolytic effector functions and promoted engraftment of graft-derived HSPCs that maintain chimerism. These mechanisms suggest an approach to achieving intestinal allograft tolerance.


Subject(s)
Graft vs Host Disease/immunology , Intestines/transplantation , Lymphopoiesis/immunology , Organ Transplantation , T-Lymphocytes/immunology , Transplantation Chimera/immunology , Allografts , Female , Graft vs Host Disease/pathology , Humans , Intestines/immunology , Intestines/pathology , Male , T-Lymphocytes/pathology
7.
FASEB J ; 35(3): e21413, 2021 03.
Article in English | MEDLINE | ID: mdl-33570785

ABSTRACT

Successful intrauterine hematopoietic cell transplantation (IUT) for congenital hemoglobinopathies is hampered by maternal alloresponsiveness. We investigate these interactions in semi-allogenic murine IUT. E14 fetuses (B6 females × BALB/c males) were each treated with 5E+6 maternal (B6) or paternal (BALB/c) bone marrow cells and serially monitored for chimerism (>1% engraftment), trafficked maternal immune cells, and immune responsiveness to donor cells. A total of 41.0% of maternal IUT recipients (mIUT) were chimeras (mean donor chimerism 3.0 ± 1.3%) versus 75.0% of paternal IUT recipients (pIUT, 3.6 ± 1.1%). Chimeras showed higher maternal microchimerism of CD4, CD8, and CD19 than non-chimeras. These maternal cells showed minimal responsiveness to B6 or BALB/c stimulation. To interrogate tolerance, mIUT were injected postnatally with 5E+6 B6 cells/pup; pIUT received BALB/c cells. IUT-treated pups showed no changes in trafficked maternal or fetal immune cell levels compared to controls. Donor-specific IgM and IgG were expressed by 1%-3% of recipients. mIUT splenocytes showed greater proliferation of regulatory T cells (Treg) upon BALB/c stimulation, while B6 stimulation upregulated the pro-inflammatory cytokines more than BALB/c. pIUT splenocytes produced identical Treg and cytokine responses to BALB/c and B6 cells, with higher Treg activity and lower pro-inflammatory cytokine expression upon exposure to BALB/c. In contrast, naïve fetal splenocytes demonstrated greater alloresponsiveness to BALB/c compared to B6 cells. Thus pIUT, associated with increased maternal cell trafficking, modulates fetal Treg, and cytokine responsiveness to donor cells more efficiently than mIUT, resulting in improved engraftment. Paternal donor cells may be considered alternatively to maternal donor cells for intrauterine and postnatal transplantation to induce tolerance and maintain engraftment.


Subject(s)
Bone Marrow Transplantation , Graft Survival/immunology , Immune Tolerance/immunology , Transplantation, Homologous , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Transplantation/methods , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation/methods , Mice , Mice, Inbred BALB C , Transplantation Chimera/immunology , Transplantation, Homologous/methods
8.
Front Immunol ; 12: 791725, 2021.
Article in English | MEDLINE | ID: mdl-35069574

ABSTRACT

Chronic rejection and immunosuppression-related toxicity severely affect long-term outcomes of kidney transplantation. The induction of transplantation tolerance - the lack of destructive immune responses to a transplanted organ in the absence of immunosuppression - could potentially overcome these limitations. Immune tolerance to kidney allografts from living donors has been successfully achieved in humans through clinical protocols based on chimerism induction with hematopoietic cell transplantation after non-myeloablative conditioning. Notably, two of these protocols have led to immune tolerance in a significant fraction of HLA-mismatched donor-recipient combinations, which represent the large majority of cases in clinical practice. Studies in mice and large animals have been critical in dissecting tolerance mechanisms and in selecting the most promising approaches for human translation. However, there are several key differences in tolerance induction between these models and humans, including the rate of success and stability of donor chimerism, as well as the relative contribution of different mechanisms in inducing donor-specific unresponsiveness. Kidney allograft tolerance achieved through durable full-donor chimerism may be due to central deletion of graft-reactive donor T cells, even though mechanistic data from patient series are lacking. On the other hand, immune tolerance attained with transient mixed chimerism-based protocols initially relies on Treg-mediated suppression, followed by peripheral deletion of donor-reactive recipient T-cell clones under antigenic pressure from the graft. These conclusions were supported by data deriving from novel high-throughput T-cell receptor sequencing approaches that allowed tracking of alloreactive repertoires over time. In this review, we summarize the most important mechanistic studies on tolerance induction with combined kidney-bone marrow transplantation in humans, discussing open issues that still need to be addressed and focusing on techniques developed in recent years to efficiently monitor the alloresponse in tolerance trials. These cutting-edge methods will be instrumental for the development of immune tolerance protocols with improved efficacy and to identify patients amenable to safe immunosuppression withdrawal.


Subject(s)
Kidney Transplantation , Kidney/immunology , Transplantation Chimera/immunology , Transplantation Conditioning , Transplantation Tolerance , Allografts , Animals , Hematopoietic Stem Cell Transplantation , Humans
9.
J Clin Invest ; 130(12): 6457-6476, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32817590

ABSTRACT

Clinical trials are currently testing whether induction of haploidentical mixed chimerism (Haplo-MC) induces organ transplantation tolerance. Whether Haplo-MC can be used to treat established autoimmune diseases remains unknown. Here, we show that established autoimmunity in euthymic and adult-thymectomized NOD (H-2g7) mice was cured by induction of Haplo-MC under a non-myeloablative anti-thymocyte globulin-based conditioning regimen and infusion of CD4+ T cell-depleted hematopoietic graft from H-2b/g7 F1 donors that expressed autoimmune-resistant H-2b or from H-2s/g7 F1 donors that expressed autoimmune-susceptible H-2s. The cure was associated with enhanced thymic negative selection, increased thymic Treg (tTreg) production, and anergy or exhaustion of residual host-type autoreactive T cells in the periphery. The peripheral tolerance was accompanied by expansion of donor- and host-type CD62L-Helios+ tTregs as well as host-type Helios-Nrp1+ peripheral Tregs (pTregs) and PD-L1hi plasmacytoid DCs (pDCs). Depletion of donor- or host-type Tregs led to reduction of host-type PD-L1hi pDCs and recurrence of autoimmunity, whereas PD-L1 deficiency in host-type DCs led to reduction of host-type pDCs and Helios-Nrp1+ pTregs. Thus, induction of Haplo-MC reestablished both central and peripheral tolerance through mechanisms that depend on allo-MHC+ donor-type DCs, PD-L1hi host-type DCs, and the generation and persistence of donor- and host-type tTregs and pTregs.


Subject(s)
Bone Marrow Transplantation , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Transplantation Chimera/immunology , Allografts , Animals , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Mice , Mice, Inbred NOD
10.
Transplantation ; 104(8): 1580-1590, 2020 08.
Article in English | MEDLINE | ID: mdl-32732835

ABSTRACT

BACKGROUND: Although short-term outcomes for liver transplantation have improved, patient and graft survival are limited by infection, cancer, and other complications of immunosuppression. Rapid induction of tolerance after liver transplantation would decrease these complications, improving survival and quality of life. Tolerance to kidneys, but not thoracic organs or islets, has been achieved in nonhuman primates and humans through the induction of transient donor chimerism. Since the liver is considered to be tolerogenic, we tested the hypothesis that the renal transplant transient chimerism protocol would induce liver tolerance. METHODS: Seven cynomolgus macaques received immune conditioning followed by simultaneous donor bone marrow and liver transplantation. The more extensive liver surgery required minor adaptations of the kidney protocol to decrease complications. All immunosuppression was discontinued on postoperative day (POD) 28. Peripheral blood chimerism, recipient immune reconstitution, liver function tests, and graft survival were determined. RESULTS: The level and duration of chimerism in liver recipients were comparable to those previously reported in renal transplant recipients. However, unlike in the kidney model, the liver was rejected soon after immunosuppression withdrawal. Rejection was associated with proliferation of recipient CD8 T effector cells in the periphery and liver, increased serum interleukin (IL)-6 and IL-2, but peripheral regulatory T cell (Treg) numbers did not increase. Antidonor antibody was also detected. CONCLUSIONS: These data show the transient chimerism protocol does not induce tolerance to livers, likely due to greater CD8 T cell responses than in the kidney model. Successful tolerance induction may depend on greater control or deletion of CD8 T cells in this model.


Subject(s)
Bone Marrow Transplantation/adverse effects , Graft Rejection/prevention & control , Liver Transplantation/adverse effects , Transplantation Chimera/immunology , Transplantation Conditioning/methods , Allografts/immunology , Animals , Bone Marrow/immunology , Bone Marrow Transplantation/methods , Disease Models, Animal , Graft Rejection/immunology , Graft Survival/immunology , Humans , Liver/immunology , Liver Transplantation/methods , Macaca fascicularis , T-Lymphocytes, Cytotoxic/immunology , Transplantation Tolerance , Transplantation, Homologous/adverse effects
11.
Front Immunol ; 11: 1028, 2020.
Article in English | MEDLINE | ID: mdl-32536926

ABSTRACT

B cell adaptor molecule of 32 kDa (Bam32), known as dual adapter for phosphotyrosine and 3-phosphoinositides 1 (DAPP1), has been implicated in regulating lymphocyte proliferation and recruitment during inflammation. However, its role in neutrophils during inflammation remains unknown. Using intravital microscopy, we examined the role of Bam32 in formyl peptide receptor agonist WKYMVm-induced permeability changes in post-capillary venules and assessed simultaneously neutrophil adhesion and emigration in cremaster muscles of Bam32-deficient (Bam32-/-) and wild-type (WT) control mice. We observed significantly reduced WKYMVm-induced microvascular hyperpermeability accompanied by markedly decreased neutrophil emigration in Bam32-/- mice. The Bam32-specific decrease in WKYMVm-induced hyperpermeability was neutrophil-dependent as this was verified in bone marrow transplanted chimeric mice. We discovered that Bam32 was critically required for WKYMVm-induced intracellular and extracellular production of reactive oxygen species (ROS) in neutrophils. Pharmacological scavenging of ROS eliminated the differences in WKYMVm-induced hyperpermeability between Bam32-/- and WT mice. Deficiency of Bam32 decreased WKYMVm-induced ERK1/2 but not p38 or JNK phosphorylation in neutrophils. Inhibition of ERK1/2 signaling cascade suppressed WKYMVm-induced ROS generation in WT neutrophils and microvascular hyperpermeability in WT mice. In conclusion, our study reveals that Bam32-dependent, ERK1/2-involving ROS generation in neutrophils is critical in WKYMVm-induced microvascular hyperpermeability during neutrophil recruitment.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Capillary Permeability/drug effects , Capillary Permeability/physiology , Lipoproteins/metabolism , Neutrophils/metabolism , Oligopeptides/pharmacology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Marrow Transplantation , Capillary Permeability/immunology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Adhesion/physiology , Lipoproteins/deficiency , Lipoproteins/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Neutrophil Infiltration/physiology , Neutrophils/drug effects , Neutrophils/immunology , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/agonists , Transplantation Chimera/immunology , Transplantation Chimera/physiology , Venules/drug effects , Venules/immunology , Venules/physiology
12.
PLoS Pathog ; 16(5): e1008544, 2020 05.
Article in English | MEDLINE | ID: mdl-32407390

ABSTRACT

Beyond their canonical roles in hemostasis and thrombosis, platelets function in the innate immune response by interacting directly with pathogens and by regulating the recruitment and activation of immune effector cells. Thrombocytopenia often coincides with neutropenia in patients with hematologic malignancies and in allogeneic hematopoietic cell transplant recipients, patient groups at high risk for invasive fungal infections. While neutropenia is well established as a major clinical risk factor for invasive fungal infections, the role of platelets in host defense against human fungal pathogens remains understudied. Here, we examined the role of platelets in murine Aspergillus fumigatus infection using two complementary approaches to induce thrombocytopenia without concurrent neutropenia. Thrombocytopenic mice were highly susceptible to A. fumigatus challenge and rapidly succumbed to infection. Although platelets regulated early conidial phagocytosis by neutrophils in a spleen tyrosine kinase (Syk)-dependent manner, platelet-regulated conidial phagocytosis was dispensable for host survival. Instead, our data indicated that platelets primarily function to maintain hemostasis and lung integrity in response to exposed fungal antigens, since thrombocytopenic mice exhibited severe hemorrhage into the airways in response to fungal challenge in the absence of overt angioinvasion. Challenge with swollen, heat-killed, conidia was lethal in thrombocytopenic hosts and could be reversed by platelet transfusion, consistent with the model that fungus-induced inflammation in platelet-depleted mice was sufficient to induce lethal hemorrhage. These data provide new insights into the role of platelets in the anti-Aspergillus host response and expand their role to host defense against filamentous molds.


Subject(s)
Aspergillus fumigatus/immunology , Blood Platelets/immunology , Hematopoietic Stem Cell Transplantation , Neutropenia/immunology , Pulmonary Aspergillosis/immunology , Transplantation Chimera/immunology , Allografts , Animals , Mice , Neutropenia/microbiology , Neutropenia/pathology , Pulmonary Aspergillosis/pathology , Transplantation Chimera/microbiology
13.
J Immunol ; 205(1): 272-281, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32444392

ABSTRACT

Acute graft-versus-host disease (GVHD) is a frequent complication of hematopoietic transplantation, yet patient risk stratification remains difficult, and prognostic biomarkers to guide early clinical interventions are lacking. We developed an approach to evaluate the potential of human T cells from hematopoietic grafts to produce GVHD. Nonconditioned NBSGW mice transplanted with titrated doses of human bone marrow developed GVHD that was characterized by widespread lymphocyte infiltration and organ pathology. Interestingly, GVHD was not an inevitable outcome in our system and was influenced by transplant dose, inflammatory status of the host, and type of graft. Mice that went on to develop GVHD showed signs of rapid proliferation in the human T cell population during the first 1-3 wk posttransplant and had elevated human IFN-γ in plasma that correlated negatively with the expansion of the human hematopoietic compartment. Furthermore, these early T cell activation metrics were predictive of GVHD onset 3-6 wk before phenotypic pathology. These results reveal an early window of susceptibility for pathological T cell activation following hematopoietic transplantation that is not simply determined by transient inflammation resulting from conditioning-associated damage and show that T cell parameters during this window can serve as prognostic biomarkers for risk of later GVHD development.


Subject(s)
Graft vs Host Disease/diagnosis , Hematopoietic Stem Cell Transplantation/adverse effects , T-Lymphocytes/immunology , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Graft vs Host Disease/blood , Graft vs Host Disease/immunology , Humans , Interferon-gamma/blood , Interferon-gamma/immunology , Lymphocyte Activation , Male , Mice , Postoperative Period , Primary Cell Culture , Prognosis , Time Factors , Transplantation Chimera/immunology , Transplantation Conditioning/adverse effects , Transplantation, Heterologous/adverse effects
14.
PLoS One ; 15(5): e0233497, 2020.
Article in English | MEDLINE | ID: mdl-32442182

ABSTRACT

Mixed hematopoietic chimerism enables donor-specific tolerance for solid organ grafts. This study evaluated the influence of different serological major histocompatibility complex disparities on chimerism development, graft-versus-host disease incidence and subsequently on solid organ tolerance in a rat model. For bone marrow transplantation conditioning total body irradiation was titrated using 10, 8 or 6 Gray. Bone marrow transplantation was performed across following major histocompatibility complex mismatched barriers: complete disparity, MHC class II, MHC class I or non-MHC mismatch. Recipients were clinically monitored for graft-versus-host disease and analyzed for chimerism using flow cytometry. After a reconstitution of 100 days, composition of peripheral leukocytes was determined. Mixed chimeras were challenged with heart grafts from allogeneic donor strains to define the impact of donor MHC class disparities on solid organ tolerance on the basis of stable chimerism. After myeloablation with 10 Gray of total body irradiation, chimerism after bone marrow transplantation was induced independent of MHC disparity. MHC class II disparity increased the incidence of graft-versus-host disease and reduced induction of stable chimerism upon myelosuppressive total body irradiation with 8 and 6 Gray, respectively. Stable mixed chimeras showed tolerance towards heart grafts from donors with MHC matched to either bone marrow donors or recipients. Isolated matching of MHC class II with bone marrow donors likewise led to stable tolerance as opposed to matching of MHC class I. In summary, MHC class II disparity was critically associated with the onset of graft-versus host disease and was identified as obstacle for successful development of chimerism after bone marrow transplantation and subsequent donor-specific solid organ tolerance.


Subject(s)
Bone Marrow Transplantation , Histocompatibility Antigens Class II/immunology , Transplantation Chimera/immunology , Transplantation Tolerance/immunology , Allografts , Animals , Graft vs Host Disease/immunology , Heart Transplantation , Humans , Male , Models, Animal , Models, Immunological , Organ Transplantation , Rats , Rats, Inbred BN , Rats, Inbred Lew , Tissue Donors , Transplantation Conditioning , Whole-Body Irradiation
15.
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
16.
Immunology ; 160(2): 198-208, 2020 06.
Article in English | MEDLINE | ID: mdl-32145062

ABSTRACT

Nuclear factor (NF)-κB-inducing kinase (NIK) is known to be a critical regulator of multiple aspects of the immune response. Although the role of NIK in the development of medullary thymic epithelial cells (mTECs) has been well documented, the impact of NIK on the differentiation and function of cortical thymic epithelial cells (cTECs) remains ambiguous. To investigate the possible involvement of NIK in cTEC differentiation, we have compared the gene expression and function of cTECs from a NIK-mutant mouse, alymphoplasia (aly/aly) with those of cTECs from wild-type (WT) mice. Flow cytometric analyses revealed that expression levels of MHC class II, but not MHC class I or other TEC markers, were higher in aly/aly cells than in WT cells. Notably, the proportion of MHC class IIhi+ cTECs was elevated in aly/aly mice. We also demonstrated that expression of Ccl5 mRNA in the MHC class IIhi+ subset of aly/aly cTECs was decreased compared with that in WT cells, implying an abnormal pattern of gene expression in aly/aly cTECs. Analyses of bone marrow chimera using aly/aly or aly/+ mice as hosts suggested that Vß usage and CD5 expression on WT T-cells were altered when they matured in aly/aly thymi. These results collectively indicate that NIK may be involved in controlling the function of cTEC in selecting a proper T-cell repertoire.


Subject(s)
Cell Differentiation/immunology , Epithelial Cells/physiology , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/immunology , Thymus Gland/growth & development , Animals , Bone Marrow Transplantation , Clonal Selection, Antigen-Mediated , Female , Flow Cytometry , Gene Expression Profiling , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Male , Mice , Mice, Transgenic , Mutation , Protein Serine-Threonine Kinases/genetics , Thymus Gland/cytology , Thymus Gland/immunology , Transplantation Chimera/immunology , NF-kappaB-Inducing Kinase
17.
Plast Reconstr Surg ; 145(4): 757e-768e, 2020 04.
Article in English | MEDLINE | ID: mdl-32221215

ABSTRACT

BACKGROUND: Transplantation of vascularized composite allografts is limited mainly by the need for life-long immunosuppression. The consequent side effects and looming specter of chronic rejection portend eventual allograft loss. Development of tolerogenic protocols is thus of utmost importance to the field of vascularized composite allograft transplantation. METHODS: With a modified delayed tolerance induction protocol, 10 cynomolgus macaques received hand (n = 2) or face vascularized composite allografts across both full and haploidentical major histocompatibility complex barriers before donor bone marrow transplantation at a later date. Protocol and for-cause allograft skin biopsies were performed for immunohistochemical analysis and analysis of donor-recipient leukocyte contribution; mixed chimerism in peripheral blood and in vitro immune responses were assessed serially. RESULTS: Before bone marrow transplantation, maintenance immunosuppression for 4 months led to lethal complications, including posttransplant lymphoproliferative disorder (in two of four recipients), which necessitated early study termination. Shortening the maintenance period to 2 months was clinically relevant and allowed all subsequent subjects (n = 6) to complete the delayed tolerance induction protocol. Acute rejection developed within the first 2 to 4 weeks after transplantation, with corresponding near-complete turnover of allograft leukocytes from donor to recipient origin, but donor-specific antibodies remained negative. After bone marrow transplantation, mixed chimerism failed to develop, although carboxyfluorescein succinimidyl ester mixed lymphocyte reaction demonstrated generalized unresponsiveness. However, the accrual of subsequent rejection episodes eventually culminated in graft vasculopathy and irreversible allograft loss. CONCLUSIONS: Despite the various advantages of the delayed tolerance induction protocol, it failed to reliably induce mixed chimerism and thus immunologic tolerance to vascularized composite allografts, given currently available immunosuppression treatment options. Ongoing work shows promise in overcoming these limitations.


Subject(s)
Composite Tissue Allografts/immunology , Graft Rejection/prevention & control , Immune Tolerance , Transplantation Conditioning/methods , Vascularized Composite Allotransplantation/adverse effects , Animals , Biopsy , Bone Marrow Transplantation/methods , Composite Tissue Allografts/pathology , Composite Tissue Allografts/transplantation , Disease Models, Animal , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/immunology , Humans , Immunosuppression Therapy/adverse effects , Immunosuppression Therapy/methods , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Leukocytes/immunology , Lymphocyte Culture Test, Mixed , Lymphoproliferative Disorders/epidemiology , Lymphoproliferative Disorders/etiology , Macaca fascicularis , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Skin/blood supply , Skin/immunology , Skin/pathology , Transplantation Chimera/immunology , Transplantation Conditioning/adverse effects , Transplantation, Homologous/adverse effects , Treatment Failure , Vascularized Composite Allotransplantation/methods
18.
J Immunol ; 204(6): 1474-1485, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31996456

ABSTRACT

An increasing body of evidence suggests that bone marrow-derived myeloid cells play a critical role in the pathophysiology of pulmonary hypertension (PH). However, the true requirement for myeloid cells in PH development has not been demonstrated, and a specific disease-promoting myeloid cell population has not been identified. Using bone marrow chimeras, lineage labeling, and proliferation studies, we determined that, in murine hypoxia-induced PH, Ly6Clo nonclassical monocytes are recruited to small pulmonary arteries and differentiate into pulmonary interstitial macrophages. Accumulation of these nonclassical monocyte-derived pulmonary interstitial macrophages around pulmonary vasculature is associated with increased muscularization of small pulmonary arteries and disease severity. To determine if the sensing of hypoxia by nonclassical monocytes contributes to the development of PH, mice lacking expression of hypoxia-inducible factor-1α in the Ly6Clo monocyte lineage were exposed to hypoxia. In these mice, vascular remodeling and PH severity were significantly reduced. Transcriptome analyses suggest that the Ly6Clo monocyte lineage regulates PH through complement, phagocytosis, Ag presentation, and chemokine/cytokine pathways. Consistent with these murine findings, relative to controls, lungs from pulmonary arterial hypertension patients displayed a significant increase in the frequency of nonclassical monocytes. Taken together, these findings show that, in response to hypoxia, nonclassical monocytes in the lung sense hypoxia, infiltrate small pulmonary arteries, and promote vascular remodeling and development of PH. Our results demonstrate that myeloid cells, specifically cells of the nonclassical monocyte lineage, play a direct role in the pathogenesis of PH.


Subject(s)
Hypertension, Pulmonary/immunology , Hypoxia/complications , Macrophages, Alveolar/immunology , Monocytes/immunology , Vascular Remodeling/immunology , Animals , Antigens, Ly/metabolism , Bone Marrow Transplantation , Cell Differentiation/immunology , Disease Models, Animal , Humans , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/surgery , Hypoxia/immunology , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/blood supply , Lung/immunology , Lung/pathology , Lung Transplantation , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Transgenic , Monocytes/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/immunology , Pulmonary Artery/pathology , Transplantation Chimera/immunology , Vascular Remodeling/genetics
19.
Front Immunol ; 11: 614250, 2020.
Article in English | MEDLINE | ID: mdl-33488624

ABSTRACT

Allogeneic hematopoietic cell transplantation (allo-HCT) is an efficacious and frequently the only treatment option for some hematological malignances. However, it often faces severe morbidities and/or mortalities due to graft versus host disease, and the severity of the conditioning regiment needed, that result in toxicity-related issues poorly tolerable for some patients. These shortcomings have led to the development of less aggressive alternatives like non-myeloablative (NMAC) or reduced-intensity conditioning regiments (RIC). However, these approaches tend to have an increase of cancer relapse and limited persistence of donor-specific chimerism. Thus, strategies that lead towards an accelerated and more durable donor engraftment are still needed. Here, we took advantage of the ability of host-derived unlicensed NK (UnLicNK) cells to favor donor cell engraftment during myeloablative allo-HCT, and evaluated if the adoptive transfer of this cell type can improve donor chimerism in NAMC settings. Indeed, the infusion of these cells significantly increased mixed chimerism in a sublethal allo-HCT mouse model, resulting in a more sustainable donor cell engraftment when compared to the administration of licensed NK cells or HCT controls. We observed an overall increase in the total number and proportion of donor B, NK and myeloid cells after UnLicNK cell infusion. Additionally, the extension and durability of donor chimerism was similar to the one obtained after the tolerogenic Tregs infusion. These results serve as the needed bases for the implementation of the adoptive transfer of UnLicNK cells to upgrade NMAC protocols and enhance allogeneic engraftment during HCT.


Subject(s)
Cytokines/blood , Graft Survival , Hematopoietic Stem Cell Transplantation/methods , Killer Cells, Natural/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation Conditioning/methods , Transplantation, Homologous/methods , Animals , Chimerism , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tissue Donors , Transplantation Chimera/immunology
20.
Front Immunol ; 11: 581111, 2020.
Article in English | MEDLINE | ID: mdl-33519804

ABSTRACT

Postoperative ileus (POI) is triggered by an innate immune response in the muscularis externa (ME) and is accompanied by bacterial translocation. Bacteria can trigger an innate immune response via toll-like receptor (TLR) activation, but the latter's contribution to POI has been disproved for several TLRs, including TLR2 and TLR4. Herein we investigated the role of double-stranded RNA detection via TLR3 and TIR-domain-containing adapter-inducing interferon-ß (TRIF) signaling pathway in POI. POI was induced by small bowel intestinal manipulation in wt, TRIF-/-, TLR3-/-, type I interferon receptor-/- and interferon-ß reporter mice, all on C57BL/6 background, and POI severity was quantified by gene expression analysis, gastrointestinal transit and leukocyte extravasation into the ME. TRIF/TLR3 deficiency reduced postoperative ME inflammation and prevented POI. With bone marrow transplantation, RNA-sequencing, flow cytometry and immunohistochemistry we revealed a distinct TLR3-expressing radio-resistant MHCIIhiCX3CR1- IBA-1+ resident macrophage population within the deep myenteric plexus. TLR3 deficiency in these cells, but not in MHCIIhiCX3CR1+ macrophages, reduced cytokine expression in POI. While this might not be an exclusive macrophage-privileged pathway, the TLR3/TRIF axis contributes to proinflammatory cytokine production in MHCIIhiCX3CR1- IBA-1+ macrophages during POI. Deficiency in TLR3/TRIF protects mice from POI. These data suggest that TLR3 antagonism may prevent POI in humans.


Subject(s)
Ileus/etiology , Macrophages/immunology , Postoperative Complications/etiology , Toll-Like Receptor 3/immunology , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/immunology , Disease Models, Animal , Female , Gene Expression , Ileus/immunology , Ileus/pathology , Immunity, Innate , Macrophages/classification , Macrophages/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myenteric Plexus/immunology , Postoperative Complications/immunology , Postoperative Complications/pathology , Radiation Tolerance/immunology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction/immunology , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/genetics , Transplantation Chimera/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...