79 results on '"Amanda L. Posgai"'
Search Results
2. A genomic data archive from the Network for Pancreatic Organ donors with Diabetes
- Author
-
Daniel J. Perry, Melanie R. Shapiro, Sonya W. Chamberlain, Irina Kusmartseva, Srikar Chamala, Leandro Balzano-Nogueira, Mingder Yang, Jason O. Brant, Maigan Brusko, MacKenzie D. Williams, Kieran M. McGrail, James McNichols, Leeana D. Peters, Amanda L. Posgai, John S. Kaddis, Clayton E. Mathews, Clive H. Wasserfall, Bobbie-Jo M. Webb-Robertson, Martha Campbell-Thompson, Desmond Schatz, Carmella Evans-Molina, Alberto Pugliese, Patrick Concannon, Mark S. Anderson, Michael S. German, Chester E. Chamberlain, Mark A. Atkinson, and Todd M. Brusko
- Subjects
Science - Abstract
Abstract The Network for Pancreatic Organ donors with Diabetes (nPOD) is the largest biorepository of human pancreata and associated immune organs from donors with type 1 diabetes (T1D), maturity-onset diabetes of the young (MODY), cystic fibrosis-related diabetes (CFRD), type 2 diabetes (T2D), gestational diabetes, islet autoantibody positivity (AAb+), and without diabetes. nPOD recovers, processes, analyzes, and distributes high-quality biospecimens, collected using optimized standard operating procedures, and associated de-identified data/metadata to researchers around the world. Herein describes the release of high-parameter genotyping data from this collection. 372 donors were genotyped using a custom precision medicine single nucleotide polymorphism (SNP) microarray. Data were technically validated using published algorithms to evaluate donor relatedness, ancestry, imputed HLA, and T1D genetic risk score. Additionally, 207 donors were assessed for rare known and novel coding region variants via whole exome sequencing (WES). These data are publicly-available to enable genotype-specific sample requests and the study of novel genotype:phenotype associations, aiding in the mission of nPOD to enhance understanding of diabetes pathogenesis to promote the development of novel therapies.
- Published
- 2023
- Full Text
- View/download PDF
3. Human immune phenotyping reveals accelerated aging in type 1 diabetes
- Author
-
Melanie R. Shapiro, Xiaoru Dong, Daniel J. Perry, James M. McNichols, Puchong Thirawatananond, Amanda L. Posgai, Leeana D. Peters, Keshav Motwani, Richard S. Musca, Andrew Muir, Patrick Concannon, Laura M. Jacobsen, Clayton E. Mathews, Clive H. Wasserfall, Michael J. Haller, Desmond A. Schatz, Mark A. Atkinson, Maigan A. Brusko, Rhonda Bacher, and Todd M. Brusko
- Subjects
Autoimmunity ,Immunology ,Medicine - Abstract
The proportions and phenotypes of immune cell subsets in peripheral blood undergo continual and dramatic remodeling throughout the human life span, which complicates efforts to identify disease-associated immune signatures in type 1 diabetes (T1D). We conducted cross-sectional flow cytometric immune profiling on peripheral blood from 826 individuals (stage 3 T1D, their first-degree relatives, those with ≥2 islet autoantibodies, and autoantibody-negative unaffected controls). We constructed an immune age predictive model in unaffected participants and observed accelerated immune aging in T1D. We used generalized additive models for location, shape, and scale to obtain age-corrected data for flow cytometry and complete blood count readouts, which can be visualized in our interactive portal (ImmScape); 46 parameters were significantly associated with age only, 25 with T1D only, and 23 with both age and T1D. Phenotypes associated with accelerated immunological aging in T1D included increased CXCR3+ and programmed cell death 1–positive (PD-1+) frequencies in naive and memory T cell subsets, despite reduced PD-1 expression levels on memory T cells. Phenotypes associated with T1D after age correction were predictive of T1D status. Our findings demonstrate advanced immune aging in T1D and highlight disease-associated phenotypes for biomarker monitoring and therapeutic interventions.
- Published
- 2023
- Full Text
- View/download PDF
4. Modeling cell-mediated immunity in human type 1 diabetes by engineering autoreactive CD8+ T cells
- Author
-
Leeana D. Peters, Wen-I Yeh, Juan M. Arnoletti, Matthew E. Brown, Amanda L. Posgai, Clayton E. Mathews, and Todd M. Brusko
- Subjects
CD8 T cell ,T cell receptor knockout ,type 1 diabetes ,gene editing ,autoimmunity ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The autoimmune pathogenesis of type 1 diabetes (T1D) involves cellular infiltration from innate and adaptive immune subsets into the islets of Langerhans within the pancreas; however, the direct cytotoxic killing of insulin-producing β-cells is thought to be mediated primarily by antigen-specific CD8+ T cells. Despite this direct pathogenic role, key aspects of their receptor specificity and function remain uncharacterized, in part, due to their low precursor frequency in peripheral blood. The concept of engineering human T cell specificity, using T cell receptor (TCR) and chimeric antigen receptor (CAR)-based approaches, has been demonstrated to improve adoptive cell therapies for cancer, but has yet to be extensively employed for modeling and treating autoimmunity. To address this limitation, we sought to combine targeted genome editing of the endogenous TCRα chain gene (TRAC) via CRISPR/Cas9 in combination with lentiviral vector (LV)-mediated TCR gene transfer into primary human CD8+ T cells. We observed that knockout (KO) of endogenous TRAC enhanced de novo TCR pairing, which permitted increased peptide:MHC-dextramer staining. Moreover, TRAC KO and TCR gene transfer increased markers of activation and effector function following activation, including granzyme B and interferon-γ production. Importantly, we observed increased cytotoxicity toward an HLA-A*0201+ human β-cell line by HLA-A*02:01 restricted CD8+ T cells engineered to recognize islet-specific glucose-6-phosphatase catalytic subunit (IGRP). These data support the notion of altering the specificity of primary human T cells for mechanistic analyses of autoreactive antigen-specific CD8+ T cells and are expected to facilitate downstream cellular therapeutics to achieve tolerance induction through the generation of antigen-specific regulatory T cells.
- Published
- 2023
- Full Text
- View/download PDF
5. Human CD4+CD25+CD226- Tregs Demonstrate Increased Purity, Lineage Stability, and Suppressive Capacity Versus CD4+CD25+CD127lo/- Tregs for Adoptive Cell Therapy
- Author
-
Matthew E. Brown, Leeana D. Peters, Seif R. Hanbali, Juan M. Arnoletti, Lindsey K. Sachs, Kayla Q. Nguyen, Emma B. Carpenter, Howard R. Seay, Christopher A. Fuhrman, Amanda L. Posgai, Melanie R. Shapiro, and Todd M. Brusko
- Subjects
CD226 ,Treg ,lineage stability ,suppressive function ,autoimmune disease ,adoptive cell therapy ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Regulatory T cell (Treg) adoptive cell therapy (ACT) represents an emerging strategy for restoring immune tolerance in autoimmune diseases. Tregs are commonly purified using a CD4+CD25+CD127lo/- gating strategy, which yields a mixed population: 1) cells expressing the transcription factors, FOXP3 and Helios, that canonically define lineage stable thymic Tregs and 2) unstable FOXP3+Helios- Tregs. Our prior work identified the autoimmune disease risk-associated locus and costimulatory molecule, CD226, as being highly expressed not only on effector T cells but also, interferon-γ (IFN-γ) producing peripheral Tregs (pTreg). Thus, we sought to determine whether isolating Tregs with a CD4+CD25+CD226- strategy yields a population with increased purity and suppressive capacity relative to CD4+CD25+CD127lo/- cells. After 14d of culture, expanded CD4+CD25+CD226- cells displayed a decreased proportion of pTregs relative to CD4+CD25+CD127lo/- cells, as measured by FOXP3+Helios- expression and the epigenetic signature at the FOXP3 Treg-specific demethylated region (TSDR). Furthermore, CD226- Tregs exhibited decreased production of the effector cytokines, IFN-γ, TNF, and IL-17A, along with increased expression of the immunoregulatory cytokine, TGF-β1. Lastly, CD226- Tregs demonstrated increased in vitro suppressive capacity as compared to their CD127lo/- counterparts. These data suggest that the exclusion of CD226-expressing cells during Treg sorting yields a population with increased purity, lineage stability, and suppressive capabilities, which may benefit Treg ACT for the treatment of autoimmune diseases.
- Published
- 2022
- Full Text
- View/download PDF
6. The Immunoregulatory Role of the Signal Regulatory Protein Family and CD47 Signaling Pathway in Type 1 Diabetes
- Author
-
Robert C. Sharp, Matthew E. Brown, Melanie R. Shapiro, Amanda L. Posgai, and Todd M. Brusko
- Subjects
CD47 ,SIRPG ,SIRPA ,SIRPB1 ,type 1 diabetes ,signal regulatory protein ,Immunologic diseases. Allergy ,RC581-607 - Abstract
BackgroundThe pathogenesis of type 1 diabetes (T1D) involves complex genetic susceptibility that impacts pathways regulating host immunity and the target of autoimmune attack, insulin-producing pancreatic β-cells. Interactions between risk variants and environmental factors result in significant heterogeneity in clinical presentation among those who develop T1D. Although genetic risk is dominated by the human leukocyte antigen (HLA) class II and insulin (INS) gene loci, nearly 150 additional risk variants are significantly associated with the disease, including polymorphisms in immune checkpoint molecules, such as SIRPG.Scope of ReviewIn this review, we summarize the literature related to the T1D-associated risk variants in SIRPG, which include a protein-coding variant (rs6043409, G>A; A263V) and an intronic polymorphism (rs2281808, C>T), and their potential impacts on the immunoregulatory signal regulatory protein (SIRP) family:CD47 signaling axis. We discuss how dysregulated expression or function of SIRPs and CD47 in antigen-presenting cells (APCs), T cells, natural killer (NK) cells, and pancreatic β-cells could potentially promote T1D development.Major ConclusionsWe propose a hypothesis, supported by emerging genetic and functional immune studies, which states a loss of proper SIRP:CD47 signaling may result in increased lymphocyte activation and cytotoxicity and enhanced β-cell destruction. Thus, we present several novel therapeutic strategies for modulation of SIRPs and CD47 to intervene in T1D.
- Published
- 2021
- Full Text
- View/download PDF
7. ACE2 chromogenic immunostaining protocol optimized for formalin-fixed paraffin-embedded human tissue sections
- Author
-
Marda Jorgensen, Paul Joseph, Amanda L. Posgai, Richard S. Vander Heide, Irina Kusmartseva, and Mark A. Atkinson
- Subjects
Health Sciences ,Immunology ,Microbiology ,Microscopy ,Antibody ,Science (General) ,Q1-390 - Abstract
Summary: Angiotensin-converting enzyme 2 (ACE2) is a key cellular entry factor for severe acute respiratory syndrome coronavirus 2. Hence, identifying cell types that express ACE2 is important for understanding the pathophysiology of coronavirus disease 2019. We performed extensive testing of multiple primary antibodies across various human tissue types. Here, we describe an optimized protocol for immunostaining of ACE2 in formalin-fixed paraffin-embedded human pancreas, small intestine, and kidney tissue sections obtained from organ donors and autopsies.For complete details on the use and execution of this protocol, please refer to Kusmartseva et al. (2020).
- Published
- 2021
- Full Text
- View/download PDF
8. Immunomodulatory Dual-Sized Microparticle System Conditions Human Antigen Presenting Cells Into a Tolerogenic Phenotype In Vitro and Inhibits Type 1 Diabetes-Specific Autoreactive T Cell Responses
- Author
-
Maigan A. Brusko, Joshua M. Stewart, Amanda L. Posgai, Clive H. Wasserfall, Mark A. Atkinson, Todd M. Brusko, and Benjamin G. Keselowsky
- Subjects
poly-lactic-co-glycolic acid ,microparticle ,autoimmunity ,immunoregulation ,combination therapy ,type 1 diabetes ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Current monotherapeutic agents fail to restore tolerance to self-antigens in autoimmune individuals without systemic immunosuppression. We hypothesized that a combinatorial drug formulation delivered by a poly-lactic-co-glycolic acid (PLGA) dual-sized microparticle (dMP) system would facilitate tunable drug delivery to elicit immune tolerance. Specifically, we utilized 30 µm MPs to provide local sustained release of granulocyte-macrophage colony-stimulating factor (GM-CSF) and transforming growth factor β1 (TGF-β1) along with 1 µm MPs to facilitate phagocytic uptake of encapsulated antigen and 1α,25(OH)2 Vitamin D3 (VD3) followed by tolerogenic antigen presentation. We previously demonstrated the dMP system ameliorated type 1 diabetes (T1D) and experimental autoimmune encephalomyelitis (EAE) in murine models. Here, we investigated the system’s capacity to impact human cell activity in vitro to advance clinical translation. dMP treatment directly reduced T cell proliferation and inflammatory cytokine production. dMP delivery to monocytes and monocyte-derived dendritic cells (DCs) increased their expression of surface and intracellular anti-inflammatory mediators. In co-culture, dMP-treated DCs (dMP-DCs) reduced allogeneic T cell receptor (TCR) signaling and proliferation, while increasing PD-1 expression, IL-10 production, and regulatory T cell (Treg) frequency. To model antigen-specific activation and downstream function, we co-cultured TCR-engineered autoreactive T cell “avatars,” with dMP-DCs or control DCs followed by β-cell line (ßlox5) target cells. For G6PC2-specific CD8+ avatars (clone 32), dMP-DC exposure reduced Granzyme B and dampened cytotoxicity. GAD65-reactive CD4+ avatars (clone 4.13) exhibited an anergic/exhausted phenotype with dMP-DC presence. Collectively, these data suggest this dMP formulation conditions human antigen presenting cells toward a tolerogenic phenotype, inducing regulatory and suppressive T cell responses.
- Published
- 2020
- Full Text
- View/download PDF
9. CD226 Deletion Reduces Type 1 Diabetes in the NOD Mouse by Impairing Thymocyte Development and Peripheral T Cell Activation
- Author
-
Melanie R. Shapiro, Wen-I Yeh, Joshua R. Longfield, John Gallagher, Caridad M. Infante, Sarah Wellford, Amanda L. Posgai, Mark A. Atkinson, Martha Campbell-Thompson, Scott M. Lieberman, David V. Serreze, Aron M. Geurts, Yi-Guang Chen, and Todd M. Brusko
- Subjects
type 1 diabetes ,Sjögren’s disease ,costimulatory receptors ,CD8+ T cells ,CD226 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The costimulatory molecule CD226 is highly expressed on effector/memory T cells and natural killer cells. Costimulatory signals received by T cells can impact both central and peripheral tolerance mechanisms. Genetic polymorphisms in CD226 have been associated with susceptibility to type 1 diabetes and other autoimmune diseases. We hypothesized that genetic deletion of Cd226 in the non-obese diabetic (NOD) mouse would impact type 1 diabetes incidence by altering T cell activation. CD226 knockout (KO) NOD mice displayed decreased disease incidence and insulitis in comparison to wild-type (WT) controls. Although female CD226 KO mice had similar levels of sialoadenitis as WT controls, male CD226 KO mice showed protection from dacryoadenitis. Moreover, CD226 KO T cells were less capable of adoptively transferring disease compared to WT NOD T cells. Of note, CD226 KO mice demonstrated increased CD8+ single positive (SP) thymocytes, leading to increased numbers of CD8+ T cells in the spleen. Decreased percentages of memory CD8+CD44+CD62L– T cells were observed in the pancreatic lymph nodes of CD226 KO mice. Intriguingly, CD8+ T cells in CD226 KO mice showed decreased islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-tetramer and CD5 staining, suggesting reduced T cell receptor affinity for this immunodominant antigen. These data support an important role for CD226 in type 1 diabetes development by modulating thymic T cell selection as well as impacting peripheral memory/effector CD8+ T cell activation and function.
- Published
- 2020
- Full Text
- View/download PDF
10. Human Regulatory T Cells From Umbilical Cord Blood Display Increased Repertoire Diversity and Lineage Stability Relative to Adult Peripheral Blood
- Author
-
Keshav Motwani, Leeana D. Peters, Willem H. Vliegen, Ahmed Gomaa El-sayed, Howard R. Seay, M. Cecilia Lopez, Henry V. Baker, Amanda L. Posgai, Maigan A. Brusko, Daniel J. Perry, Rhonda Bacher, Joseph Larkin, Michael J. Haller, and Todd M. Brusko
- Subjects
cord blood ,peripheral blood ,regulatory T cells ,Tregs ,adoptive cell therapy ,scRNA-seq ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The human T lymphocyte compartment is highly dynamic over the course of a lifetime. Of the many changes, perhaps most notable is the transition from a predominantly naïve T cell state at birth to the acquisition of antigen-experienced memory and effector subsets following environmental exposures. These phenotypic changes, including the induction of T cell exhaustion and senescence, have the potential to negatively impact efficacy of adoptive T cell therapies (ACT). When considering ACT with CD4+CD25+CD127–/lo regulatory T cells (Tregs) for the induction of immune tolerance, we previously reported ex vivo expanded umbilical cord blood (CB) Tregs remained more naïve, suppressed responder T cells equivalently, and exhibited a more diverse T cell receptor (TCR) repertoire compared to expanded adult peripheral blood (APB) Tregs. Herein, we hypothesized that upon further characterization, we would observe increased lineage heterogeneity and phenotypic diversity in APB Tregs that might negatively impact lineage stability, engraftment capacity, and the potential for Tregs to home to sites of tissue inflammation following ACT. We compared the phenotypic profiles of human Tregs isolated from CB versus the more traditional source, APB. We conducted analysis of fresh and ex vivo expanded Treg subsets at both the single cell (scRNA-seq and flow cytometry) and bulk (microarray and cytokine profiling) levels. Single cell transcriptional profiles of pre-expansion APB Tregs highlighted a cluster of cells that showed increased expression of genes associated with effector and pro-inflammatory phenotypes (CCL5, GZMK, CXCR3, LYAR, and NKG7) with low expression of Treg markers (FOXP3 and IKZF2). CB Tregs were more diverse in TCR repertoire and homogenous in phenotype, and contained fewer effector-like cells in contrast with APB Tregs. Interestingly, expression of canonical Treg markers, such as FOXP3, TIGIT, and IKZF2, were increased in CB CD4+CD127+ conventional T cells (Tconv) compared to APB Tconv, post-expansion, implying perinatal T cells may adopt a default regulatory program. Collectively, these data identify surface markers (namely CXCR3) that could be depleted to improve purity and stability of APB Tregs, and support the use of expanded CB Tregs as a potentially optimal ACT modality for the treatment of autoimmune and inflammatory diseases.
- Published
- 2020
- Full Text
- View/download PDF
11. Expansion of Human Tregs from Cryopreserved Umbilical Cord Blood for GMP-Compliant Autologous Adoptive Cell Transfer Therapy
- Author
-
Howard R. Seay, Amy L. Putnam, Judit Cserny, Amanda L. Posgai, Emma H. Rosenau, John R. Wingard, Kate F. Girard, Morey Kraus, Angela P. Lares, Heather L. Brown, Katherine S. Brown, Kristi T. Balavage, Leeana D. Peters, Ashley N. Bushdorf, Mark A. Atkinson, Jeffrey A. Bluestone, Michael J. Haller, and Todd M. Brusko
- Subjects
autoimmunity ,cord blood ,adoptive cell transfer ,regulatory T cell ,type 1 diabetes ,T cell receptor ,good manufacturing practices ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Umbilical cord blood is a traditional and convenient source of cells for hematopoietic stem cell transplantation. Thymic regulatory T cells (Tregs) are also present in cord blood, and there is growing interest in the use of autologous Tregs to provide a low-risk, fully human leukocyte antigen (HLA)-matched cell product for treating autoimmune diseases, such as type 1 diabetes. Here, we describe a good manufacturing practice (GMP)-compatible Treg expansion protocol using fluorescence-activated cell sorting, resulting in a mean 2,092-fold expansion of Tregs over a 16-day culture for a median yield of 1.26 × 109 Tregs from single-donor cryopreserved units. The resulting Tregs passed prior clinical trial release criteria for Treg purity and sterility, including additional rigorous assessments of FOXP3 and Helios expression and epigenetic analysis of the FOXP3 Treg-specific demethylated region (TSDR). Compared with expanded adult peripheral blood Tregs, expanded cord blood Tregs remained more naive, as assessed by continued expression of CD45RA, produced reduced IFN-γ following activation, and effectively inhibited responder T cell proliferation. Immunosequencing of the T cell receptor revealed a remarkably diverse receptor repertoire within cord blood Tregs that was maintained following in vitro expansion. These data support the feasibility of generating GMP-compliant Tregs from cord blood for adoptive cell transfer therapies and highlight potential advantages in terms of safety, phenotypic stability, autoantigen specificity, and tissue distribution.
- Published
- 2017
- Full Text
- View/download PDF
12. Avidity and Bystander Suppressive Capacity of Human Regulatory T Cells Expressing De Novo Autoreactive T-Cell Receptors in Type 1 Diabetes
- Author
-
Wen-I Yeh, Howard R. Seay, Brittney Newby, Amanda L. Posgai, Filipa Botelho Moniz, Aaron Michels, Clayton E. Mathews, Jeffrey A. Bluestone, and Todd M. Brusko
- Subjects
type 1 diabetes ,regulatory T cells ,T cell receptor ,avidity ,suppression mechanisms ,adoptive cellular therapies ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The ability to alter antigen specificity by T-cell receptor (TCR) or chimeric antigen receptor (CAR) gene transfer has facilitated personalized cellular immune therapies in cancer. Inversely, this approach can be harnessed in autoimmune settings to attenuate inflammation by redirecting the specificity of regulatory T cells (Tregs). Herein, we demonstrate efficient protocols for lentiviral gene transfer of TCRs that recognize type 1 diabetes-related autoantigens with the goal of tissue-targeted induction of antigen-specific tolerance to halt β-cell destruction. We generated human Tregs expressing a high-affinity GAD555–567-reactive TCR (clone R164), as well as the lower affinity clone 4.13 specific for the same peptide. We demonstrated that de novo Treg avatars potently suppress antigen-specific and bystander responder T-cell (Tresp) proliferation in vitro in a process that requires Treg activation (P
- Published
- 2017
- Full Text
- View/download PDF
13. Sulfatide Preserves Insulin Crystals Not by Being Integrated in the Lattice but by Stabilizing Their Surface
- Author
-
Karsten Buschard, Austin W. Bracey, Daniel L. McElroy, Andrew T. Magis, Thomas Osterbye, Mark A. Atkinson, Kate M. Bailey, Amanda L. Posgai, and David A. Ostrov
- Subjects
Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Background. Sulfatide is known to chaperone insulin crystallization within the pancreatic beta cell, but it is not known if this results from sulfatide being integrated inside the crystal structure or by binding the surface of the crystal. With this study, we aimed to characterize the molecular mechanisms underlying the integral role for sulfatide in stabilizing insulin crystals prior to exocytosis. Methods. We cocrystallized human insulin in the presence of sulfatide and solved the structure by molecular replacement. Results. The crystal structure of insulin crystallized in the presence of sulfatide does not reveal ordered occupancy representing sulfatide in the crystal lattice, suggesting that sulfatide does not permeate the crystal lattice but exerts its stabilizing effect by alternative interactions such as on the external surface of insulin crystals. Conclusions. Sulfatide is known to stabilize insulin crystals, and we demonstrate here that in beta cells sulfatide is likely coating insulin crystals. However, there is no evidence for sulfatide to be built into the crystal lattice.
- Published
- 2016
- Full Text
- View/download PDF
14. Divergent metabolic phenotypes in two genetic syndromes of low insulin secretion
- Author
-
Jaime Guevara-Aguirre, Arlan L. Rosenbloom, Alexandra Guevara, Mark A. Atkinson, MacKenzie D. Williams, Enrique Terán, Amanda L. Posgai, Carolina Guevara, Verónica Rosado, Antonio W.D. Gavilanes, Clive H. Wasserfall, Kindergeneeskunde, RS: GROW - R4 - Reproductive and Perinatal Medicine, RS: MHeNs - R3 - Neuroscience, and MUMC+: MA Medische Staf Kindergeneeskunde (9)
- Subjects
Endocrinology ,Endocrinology, Diabetes and Metabolism ,Internal Medicine ,General Medicine - Abstract
AIMS: We examined the effect of growth hormone (GH) counter-regulation on carbohydrate metabolism in individuals with life-long diminished insulin secretion (DIS).METHODS: Adults homozygous for the E180 splice site mutation of GHR [Laron syndrome (LS)], adults with a gain-of-function mutation in CDKN1c [Guevara-Rosenbloom syndrome (GRS)], and controls were evaluated for body composition, leptin, total and high molecular weight (HMW) adiponectin, insulin-like growth factor (IGF) axis molecules, and a 5-hour oral glucose tolerance test (OGTT), with measurements of glucose, insulin, glucagon, ghrelin, pancreatic polypeptide, gastric inhibitory peptide, glucagon-like peptide-1, peptide YY, and islet amyloid polypeptide (IAPP).RESULTS: Both syndromic cohorts displayed DIS during OGTT. LS subjects had higher serum concentrations of total and HMW adiponectin, and lower levels of IGF-I, IGF-II, and IGF-Binding Protein-3 than individuals in other study groups. Furthermore, they displayed normal glycemic responses during OGTT with the lowest IAPP secretion. In contrast, individuals with GRS had higher levels of protein glycation, deficient glucose control during OGTT, and increased secretion of IAPP.CONCLUSIONS: A distinct metabolic phenotype depending on GH counter-regulatory status, associates with diabetes development and excess glucose-induced IAPP secretion.
- Published
- 2023
- Full Text
- View/download PDF
15. Biomaterials-based nanoparticles conjugated to regulatory T cells provide a modular system for localized delivery of pharmacotherapeutic agents
- Author
-
Gregory P. Marshall, Judit Cserny, Chun‐Wei Wang, Benjamin Looney, Amanda L. Posgai, Rhonda Bacher, Benjamin Keselowsky, and Todd M. Brusko
- Subjects
Biomaterials ,Mice ,Mice, Inbred NOD ,Metals and Alloys ,Biomedical Engineering ,Ceramics and Composites ,Humans ,Animals ,Interleukin-2 ,Female ,Biocompatible Materials ,T-Lymphocytes, Regulatory ,Diabetes Mellitus, Experimental - Abstract
Type 1 diabetes (T1D) presents with two therapeutic challenges: the need to correct underlying autoimmunity and restore β-cell mass. We harnessed the unique capacity of regulatory T cells (Tregs) and the T cell receptor (TCR) to direct tolerance induction along with tissue-localized delivery of therapeutic agents to restore endogenous β-cell function. Specifically, we designed a combinatorial therapy involving biomaterials-based poly(lactic-co-glycolic acid) nanoparticles co-loaded with the Treg growth factor, IL-2, and the β-cell regenerative agent, harmine (a tyrosine-regulated kinase 1A [DYRK1A] inhibitor), conjugated to the surface of Tregs. We observed continuous elution of IL-2 and harmine from nanoparticles for at least 7 days in vitro. When conjugated to primary human Tregs, IL-2 nanoparticles provided sufficient IL-2 receptor signaling to support STAT5 phosphorylation for sustained phenotypic stability and viability in culture. Inclusion of poly-L-lysine (PLL) during nanoparticle-cell coupling dramatically increased conjugation efficiency, providing sufficient IL-2 to support in vitro proliferation of IL-2-dependent CTLL-2 cells and primary murine Tregs. In 12-week-old female non-obese diabetic mice, adoptive transfer of IL-2/harmine nanoparticle-conjugated NOD.BDC2.5 Tregs, which express an islet antigen-specific TCR, significantly prevented diabetes demonstrating preserved in vivo viability. These data provide the preclinical basis to develop a biomaterials-optimized cellular therapy to restore immune tolerance and promote β-cell proliferation in T1D through receptor-targeted drug delivery within pancreatic islets.
- Published
- 2022
16. The Women’s Leadership Gap in Diabetes: A Call for Equity and Excellence
- Author
-
Jennifer Maizel, Mark A. Atkinson, Jessica L. Dunne, Linda A. DiMeglio, and Amanda L. Posgai
- Subjects
American diabetes association ,Advanced and Specialized Nursing ,Research design ,medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,media_common.quotation_subject ,Women in Diabetes Leadership ,MEDLINE ,Equity (finance) ,Editorial board ,Primary care ,Diabetes education ,medicine.disease ,Clinical diabetes ,Excellence ,Diabetes mellitus ,Family medicine ,Health care ,Internal Medicine ,medicine ,business ,media_common - Abstract
OBJECTIVE Women are broadly underrepresented in scientific leadership positions and their accomplishments are not provided equal recognition compared with those of men, but the imbalance in the field of diabetes is unknown. Hence, we analyzed multiple aspects of historical and present-day female representation in the diabetes field. RESEARCH DESIGN AND METHODS We quantified gender representation at annual American Diabetes Association (ADA) meetings; editorial board service positions for ADA and the European Association for the Study of Diabetes (EASD) journals; principal investigators for ADA, JDRF, and National Institutes of Health National Institute of Diabetes and Digestive and Kidney Diseases P30 grant funding; and ADA, JDRF, and EASD award recipients. RESULTS There are many women in the field of diabetes: registration for the ADA Scientific Sessions has been 43% female since 2016, and for over five decades, women comprised 83% of ADA Presidents of Health Care and Education. Yet, only 9% of ADA Presidents of Medicine and Science have been women. Women were well represented on editorial boards for journals focused on diabetes education (Diabetes Spectrum, 89% female) and primary care (Clinical Diabetes, 49% female) but not for the more academically targeted Diabetes Care (34% female), Diabetes (21% female), and Diabetologia (30% female). Only one-third of ADA Pathway to Stop Diabetes and JDRF grants have been awarded to women, and females only lead 2 of 18 (11%) of the P30-supported Diabetes Research Centers. Finally, only 2–12% of major ADA, JDRF, and EASD awards were given to women, without significant change over time. CONCLUSIONS Despite increasing recognition of gender imbalance in research and medicine, many disparities in the field of diabetes persist. We call for decreasing barriers for advancement of female investigators and creating environments that promote their retention and equitable recognition for their contributions to the field.
- Published
- 2021
17. Image-Based Machine Learning Algorithms for Disease Characterization in the Human Type 1 Diabetes Pancreas
- Author
-
Clive Wasserfall, John S. Kaddis, Bart O. Roep, Stephan Speier, Shweta Kulkarni, Irina Kusmartseva, Xiaohan Tang, Mark A. Atkinson, Desmond A. Schatz, Martha Campbell-Thompson, Michael J. Haller, and Amanda L. Posgai
- Subjects
0301 basic medicine ,Adolescent ,030209 endocrinology & metabolism ,Disease ,Machine learning ,computer.software_genre ,Pathology and Forensic Medicine ,Machine Learning ,03 medical and health sciences ,0302 clinical medicine ,Diabetes mellitus ,Image Processing, Computer-Assisted ,medicine ,Acinar cell ,Humans ,Insulin ,Endocrine system ,Pancreas ,Autoantibodies ,Pancreatic duct ,Type 1 diabetes ,business.industry ,Case-control study ,Regular Article ,medicine.disease ,Tissue Donors ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,medicine.anatomical_structure ,Case-Control Studies ,Female ,Artificial intelligence ,business ,computer ,Algorithm ,Algorithms - Abstract
Emerging data suggest that type 1 diabetes affects not only the β-cell-containing islets of Langerhans, but also the surrounding exocrine compartment. Using digital pathology, machine learning algorithms were applied to high-resolution, whole-slide images of human pancreata to determine whether the tissue composition in individuals with or at risk for type 1 diabetes differs from those without diabetes. Transplant-grade pancreata from organ donors were evaluated from 16 nondiabetic autoantibody-negative controls, 8 nondiabetic autoantibody-positive subjects with increased type 1 diabetes risk, and 19 persons with type 1 diabetes (0 to 12 years' duration). HALO image analysis algorithms were implemented to compare architecture of the main pancreatic duct as well as cell size, density, and area of acinar, endocrine, ductal, and other nonendocrine, nonexocrine tissues. Type 1 diabetes was found to affect exocrine area, acinar cell density, and size, whereas the type of difference correlated with the presence or absence of insulin-positive cells remaining in the pancreas. These changes were not observed before disease onset, as indicated by modeling cross-sectional data from pancreata of autoantibody-positive subjects and those diagnosed with type 1 diabetes. These data provide novel insights into anatomic differences in type 1 diabetes pancreata and demonstrate that machine learning can be adapted for the evaluation of disease processes from cross-sectional data sets.
- Published
- 2021
- Full Text
- View/download PDF
18. How Do We Move Type 1 Diabetes Immunotherapies Forward During the Current COVID-19 Pandemic?
- Author
-
Desmond A. Schatz, Michael J. Haller, Laura M. Jacobsen, and Amanda L. Posgai
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,Endocrinology, Diabetes and Metabolism ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,medicine.medical_treatment ,030209 endocrinology & metabolism ,Antibodies, Monoclonal, Humanized ,Risk Assessment ,03 medical and health sciences ,Clinical Trials, Phase II as Topic ,0302 clinical medicine ,Diabetes mellitus ,Pandemic ,Internal Medicine ,medicine ,Humans ,Significant risk ,Intensive care medicine ,Type 1 diabetes ,Leukemia ,SARS-CoV-2 ,business.industry ,Antibodies, Monoclonal ,COVID-19 ,Immunotherapy ,Inflammatory Bowel Diseases ,medicine.disease ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,Increased risk ,Clinical Trials, Phase III as Topic ,Disease Progression ,Tumor Necrosis Factor Inhibitors ,business ,Immunosuppressive Agents - Abstract
Research-based immunotherapy trials seeking to prevent or reverse a number of autoimmune diseases, including type 1 diabetes, have seen near universal suspension due to the coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Diabetes and hyperglycemia are now appreciated as significant risk factors for COVID-19 morbidity and mortality; however, the vast majority of studies have reported on adults. Recent data in children and adolescents with type 1 diabetes suggest no increased risk of COVID-19. Even with immense appreciation for COVID-19 morbidity and mortality, we believe compelling arguments exist to carefully and thoughtfully resume certain type 1 diabetes phase 2–3 immunotherapy trials. In this Perspective, we consider the experience of trials that never halted or have resumed in the oncology and rheumatology fields, and advocate for staged type 1 diabetes immunotherapy trial resumption. With this, we present recommendations to achieve equipoise and mitigate risks for SARS-CoV-2 infection in the weeks surrounding infusion. Given the fact that the COVID-19 pandemic is expected to persist for some time, it is in the best interest of our patients that we find ways to safely move our field forward.
- Published
- 2021
- Full Text
- View/download PDF
19. Low-Dose ATG/GCSF in Established Type 1 Diabetes: A Five-Year Follow-up Report
- Author
-
Stephen E. Gitelman, Desmond A. Schatz, Mark A. Atkinson, Jasmine A Mack, Laura M. Jacobsen, Brittany S. Bruggeman, Amanda L. Posgai, Peter A. Gottlieb, Michael J. Haller, Andrea Lin, Matthew J. Gurka, Todd M. Brusko, Clayton E. Mathews, and Clive Wasserfall
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Clinical Trials and Supportive Activities ,Pilot Projects ,030209 endocrinology & metabolism ,Placebo ,Medical and Health Sciences ,Gastroenterology ,Endocrinology & Metabolism ,03 medical and health sciences ,0302 clinical medicine ,Clinical Research ,Internal medicine ,Granulocyte Colony-Stimulating Factor ,Diabetes Mellitus ,Internal Medicine ,medicine ,Clinical endpoint ,Humans ,Metabolic and endocrine ,Antilymphocyte Serum ,Type 1 diabetes ,C-Peptide ,business.industry ,Diabetes ,Low dose ,Area under the curve ,Five year follow up ,medicine.disease ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,Area Under Curve ,Immunology and Transplantation ,business ,Type 1 - Abstract
Previously, we demonstrated low-dose anti-thymocyte globulin (ATG) and granulocyte colony stimulating factor (GCSF) immunotherapy preserved C-peptide for two years in a pilot study of subjects with established type 1 diabetes (n=25). Herein, we evaluated the long-term outcomes of ATG/GCSF in study participants with five years of available follow-up data (n=15). The primary endpoint was area under the curve (AUC) C-peptide during a two-hour mixed-meal tolerance test (MMTT). After five years, there were no statistically significant differences in AUC C-peptide when comparing subjects who received ATG/GCSF versus placebo (p = 0.41). A modeling framework based on mean trajectories in C-peptide AUC over five years, accounting for differing trends between groups, was applied to re-categorize responders (n=9) and non-responders (n=7). ATG/GCSF reponders demonstrated nearly unchanged HbA1c over five years [mean (95% CI) adjusted change = 0.29% (-0.69%, 1.27%)], but the study was not powered for comparisons against non-responders 1.75% (-0.57%, 4.06%) and placebo 1.44% (0.21%, 2.66%). These data underscore the importance of long-term follow up in previous and ongoing phase 2 trials of low-dose ATG in recent-onset type 1 diabetes.
- Published
- 2021
- Full Text
- View/download PDF
20. Exocrine Pancreatic Enzymes Are a Serological Biomarker for Type 1 Diabetes Staging and Pancreas Size
- Author
-
Desmond A. Schatz, Clive Wasserfall, James J. Ross, Todd M. Brusko, Rhonda Bacher, Mark A. Atkinson, Xiaoru Dong, Kieran M. Mcgrail, Amanda L. Posgai, Martha Campbell-Thompson, Andrew Muir, Michael J. Haller, Xia Li, and Daniel J. Perry
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Magnetic Resonance Spectroscopy ,Trypsinogen ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Gastroenterology ,Serology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Diabetes mellitus ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Humans ,Amylase ,Lipase ,Pancreas ,Type 1 diabetes ,biology ,business.industry ,medicine.disease ,Diabetes Mellitus, Type 1 ,Logistic Models ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Linear Models ,biology.protein ,Biomarker (medicine) ,Immunology and Transplantation ,business - Abstract
Exocrine pancreas abnormalities are increasingly recognized as features of type 1 diabetes. We previously reported reduced serum trypsinogen levels and in a separate study, smaller pancreata at and prior to disease onset. We hypothesized that three pancreas enzymes (amylase, lipase and trypsinogen) might serve as serological biomarkers of pancreas volume and risk for type 1 diabetes. Amylase, lipase, and trypsinogen were measured from two independent cohorts, together comprising 800 serum samples from single-autoantibody positive (1AAb+) and multiple-AAb+ (≥2AAb+) subjects, individuals with recent-onset or established type 1 diabetes, their AAb negative (AAb-) first-degree relatives, and AAb- controls. Lipase and trypsinogen were significantly reduced in ≥2AAb+, recent-onset, and established type 1 diabetes subjects versus controls and 1AAb+, while amylase was reduced only in established type 1 diabetes. Logistic regression models demonstrated trypsinogen plus lipase (AUROC=81.4%) performed equivalently to all three enzymes (AUROC=81.4%) in categorizing ≥2AAb+ versus 1AAb+ subjects. For Cohort 2 (n=246), linear regression demonstrated lipase and trypsinogen levels could individually and collectively serve as indicators of BMI-normalized relative pancreas volume (RPVBMI, PBMI and may improve disease staging in pre-type 1 diabetes.
- Published
- 2021
- Full Text
- View/download PDF
21. Deletion of CD226 in Foxp3+ T cells Reduces Diabetes Incidence in Non-Obese Diabetic Mice by Improving Regulatory T Cell Stability and Function
- Author
-
Puchong Thirawatananond, Matthew E. Brown, Lindsey K. Sachs, Juan M. Arnoletti, Wen-I Yeh, Amanda L. Posgai, Melanie R. Shapiro, Yi-Guang Chen, and Todd M. Brusko
- Abstract
Co-stimulation serves as a critical checkpoint for T cell development and activation, and several genetic variants affecting co-stimulatory pathways confer risk for autoimmune diseases. A single nucleotide polymorphism in CD226 (rs763361; G307S) has been shown to increase susceptibility to type 1 diabetes, multiple sclerosis, and rheumatoid arthritis. CD226 competes with the co-inhibitory receptor TIGIT (T cell immunoreceptor with Ig and ITIM domains) to bind CD155 to amplify TCR signaling. We previously found that Cd226 knockout protected non-obese diabetic (NOD) mice from disease, but the impact of CD226 signaling on individual immune subsets remained unclear. We focused on regulatory T cells (Tregs) as a population of interest, as prior reports demonstrated that human CD226+ Tregs exhibit reduced FOXP3+Helios+ purity and suppressive function following expansion. Hence, we hypothesized that global deletion of Cd226 would increase Treg stability and accordingly, Treg-specific Cd226 deletion would inhibit diabetes in NOD mice. Indeed, crossing the NOD.Cd226-/- and NOD.Foxp3-GFP-Cre.R26-loxP-STOP-loxP-YFP Treg-fate tracking strains resulted in increased Treg induction and decreased FoxP3-deficient “ex-Tregs” in the pancreatic lymph nodes. We generated a Treg-conditional knockout (TregΔCd226) strain and found that female TregΔCd226 mice had decreased insulitis and diabetes incidence compared to TregWT mice. Additionally, we observed increased TIGIT expression on Tregs and conventional CD4+ T cells within the pancreas of TregΔCd226 versus TregWT mice. These findings demonstrate that an imbalance of CD226/TIGIT signaling may contribute to Treg destabilization in the NOD mouse and highlight the potential for therapeutic targeting of this pathway to prevent or reverse autoimmunity.
- Published
- 2022
- Full Text
- View/download PDF
22. Insulin-like Growth Factor-1 Synergizes with IL-2 to Induce Homeostatic Proliferation of Regulatory T cells
- Author
-
Melanie R. Shapiro, Leeana D. Peters, Matthew E. Brown, Cecilia Cabello-Kindelan, Amanda L. Posgai, Allison L. Bayer, and Todd M. Brusko
- Abstract
IL-2 has been proposed to restore tolerance via regulatory T cell (Treg) expansion in autoimmunity, yet off-target effects necessitate identification of a combinatorial approach. We recently reported reduced levels of immunoregulatory insulin-like growth factor-1 (IGF1) during type 1 diabetes (T1D) progression. Thus, we hypothesized that IGF1 would synergize with IL-2 to expand Tregs. We observed IGF1R was elevated on murine memory and human naïve Treg subsets. IL-2 and IGF1 promoted murine PI3K/Akt and human STAT5 signaling in Tregs. IL-2 and IGF1 treatment expanded Tregs beyond either agent alone in NOD mice. Incubation of naïve human CD4+T cells with IL-2 and IGF1 enhanced Treg proliferationin vitro, without the need for T cell receptor ligation. This synergism was attributed to increased high-affinity IL-2Rα expression on naïve Tregs, in contrast to intermediate-affinity IL-2Rβ and IL-2Rγ subunit enhancement on naïve conventional T cells (Tconv). We then demonstrated that IGF1 and IL-2 or the IL2Rγ-chain-dependent cytokine, IL-7, can be used to induce proliferation of genetically-engineered naïve Treg or Tconv cells, respectively. These data support the potential use of IGF1 in combination with common γ-chain cytokines to drive T cell expansions bothin vitroandin vivofor cellular therapeutics and genetic modifications.
- Published
- 2022
- Full Text
- View/download PDF
23. Exocrine and Endocrine Inflammation Increases Cellular Replication in the Pancreatic Duct Compartment in Type 1 Diabetes
- Author
-
Shweta Kulkarni, Amanda L Posgai, Irina Kusmartseva, Clive H Wasserfall, Mark A Atkinson, and Alexandra E Butler
- Subjects
Endocrinology, Diabetes and Metabolism - Abstract
Context We recently demonstrated increased cellular proliferation in the pancreatic ductal gland (PDG) compartment of organ donors with type 1 diabetes, suggesting that PDGs may harbor progenitor cells capable of pancreatic regeneration. Objective We evaluated the impact of diabetes and pancreatic inflammation on PDG and interlobular duct (ILD) cellular proliferation and profiles. Methods Endocrine hormone expression (insulin, glucagon, somatostatin, pancreatic polypeptide) and proliferating Ki67+ cells were localized within the PDG and ILD compartments by multicolor immunohistochemistry in cross-sections from the head, body, and tail regions of pancreata from those with (n = 31) or without type 1 diabetes (n = 43). Whole-slide scanned images were analyzed using digital pathology. Results Type 1 diabetes donors with insulitis or histologically identified pancreatitis had increased cellular replication in the ILD and PDG compartments. Interestingly, while cellular proliferation within the pancreatic ductal tree was significantly increased in type 1 diabetes (PDG mean = 3.36%, SEM = 1.06; ILD mean = 2.78%, SEM = 0.97) vs nondiabetes(ND) subjects without pancreatic inflammation (PDG mean = 1.18%, SEM = 0.42; ILD mean = 0.74%, SEM = 0.15, P Conclusion These data suggest that increased pancreatic ductal cell replication is associated with sustained pancreatic inflammation; however, as replicating cells were hormone-negative, PDGs do not appear to represent a compelling endogenous source of hormone-positive endocrine cells.
- Published
- 2022
24. Erratum. The Women’s Leadership Gap in Diabetes: A Call for Equity and Excellence. Diabetes 2021;70:1623–1633
- Author
-
Jessica L. Dunne, Jennifer L. Maizel, Amanda L. Posgai, Mark A. Atkinson, and Linda A. DiMeglio
- Subjects
Male ,Leadership ,National Institutes of Health (U.S.) ,Endocrinology, Diabetes and Metabolism ,Internal Medicine ,Awards and Prizes ,Diabetes Mellitus ,Humans ,Female ,Erratum ,Societies, Medical ,United States - Abstract
Women are broadly underrepresented in scientific leadership positions and their accomplishments are not provided equal recognition compared with those of men, but the imbalance in the field of diabetes is unknown. Hence, we analyzed multiple aspects of historical and present-day female representation in the diabetes field.We quantified gender representation at annual American Diabetes Association (ADA) meetings; editorial board service positions for ADA and the European Association for the Study of Diabetes (EASD) journals; principal investigators for ADA, JDRF, and National Institutes of Health National Institute of Diabetes and Digestive and Kidney Diseases P30 grant funding; and ADA, JDRF, and EASD award recipients. There are many women in the field of diabetes: registration for the ADA Scientific Sessions has been 43% female since 2016, and for over five decades, women comprised 83% of ADA Presidents of Health Care and Education. Yet, only 9% of ADA Presidents of Medicine and Science have been women. Women were well represented on editorial boards for journals focused on diabetes education (Diabetes Spectrum, 89% female) and primary care (Clinical Diabetes, 49% female) but not for the more academically targeted Diabetes Care (34% female), Diabetes (21% female), and Diabetologia (30% female). Only one-third of ADA Pathway to Stop Diabetes and JDRF grants have been awarded to women, and females only lead 2 of 18 (11%) of the P30-supported Diabetes Research Centers. Finally, only 2-12% of major ADA, JDRF, and EASD awards were given to women, without significant change over time. Despite increasing recognition of gender imbalance in research and medicine, many disparities in the field of diabetes persist. We call for decreasing barriers for advancement of female investigators and creating environments that promote their retention and equitable recognition for their contributions to the field.
- Published
- 2022
25. Human CD4
- Author
-
Matthew E, Brown, Leeana D, Peters, Seif R, Hanbali, Juan M, Arnoletti, Lindsey K, Sachs, Kayla Q, Nguyen, Emma B, Carpenter, Howard R, Seay, Christopher A, Fuhrman, Amanda L, Posgai, Melanie R, Shapiro, and Todd M, Brusko
- Subjects
Interferon-gamma ,Cell- and Tissue-Based Therapy ,Cytokines ,Humans ,Forkhead Transcription Factors ,T-Lymphocytes, Regulatory ,Autoimmune Diseases - Abstract
Regulatory T cell (Treg) adoptive cell therapy (ACT) represents an emerging strategy for restoring immune tolerance in autoimmune diseases. Tregs are commonly purified using a CD4
- Published
- 2022
26. Immune, endothelial and neuronal network map in human lymph node and spleen
- Author
-
Seth Currlin, Harry S. Nick, Marda Jorgensen, Jerelyn A. Nick, Maigan A. Brusko, Hunter Hakimian, Jesus Penaloza-Aponte, Natalie Rodriguez, Miguel Medina-Serpas, Mingder Yang, Robert P. Seifert, Irina Kusmartseva, Todd M. Brusko, Kevin Otto, Amanda L. Posgai, Clive H. Wasserfall, and Mark A. Atkinson
- Subjects
Pathology ,medicine.medical_specialty ,medicine.anatomical_structure ,Tunica externa ,Podoplanin ,Tyrosine hydroxylase ,medicine ,Cholinergic ,Spleen ,Lymph ,Biology ,Choline acetyltransferase ,Lymph node - Abstract
SummaryThe spleen and lymph node represent important hubs for both innate and adaptive immunity1,2. Herein, we map immune, endothelial, and neuronal cell networks within these tissues from “normal”/non-diseased organ donors, collected through the NIH Human BioMolecular Atlas Program (HuBMAP)3, using highly multiplexed CODEX (CO-Detection by indEXing) imaging and 3D light sheet microscopy of cleared tissues. Building on prior reports4–6, we observed the lymph node subcapsular sinus expressing podoplanin, smooth muscle actin, and LYVE1. In the spleen, LYVE1 was expressed by littoral cells lining venous sinusoids, whereas podoplanin was restricted to arteries and trabeculae. 3D visualization of perivascular innervation revealed a subset of axonal processes expressing choline acetyl transferase in both tissues, in contrast with prior literature on human spleen7. We further report our novel observations regarding the distinct localization of GAP43 and β3-tubulin within the vascular anatomy of both lymph node and spleen, with Coronin-1A+ cells forming a dense cluster around β3-tubulin positive GAP43 low/negative segments of large vessels in spleen. These data provide an unprecedented 2D and 3D visualization of cellular networks within secondary lymphoid tissues, laying the groundwork for future disease-specific and system-wide studies of neural regulation of immunity in human lymphatics.
- Published
- 2021
- Full Text
- View/download PDF
27. Monogenic Diabetes and Integrated Stress Response Genes Display Altered Gene Expression in Type 1 Diabetes
- Author
-
Richard A. Oram, Helmut Hiller, Joeseph J. Lebowitz, Habibeh Khoshbouei, Irina Kusmarteva, Mark A. Atkinson, Bernd Bodenmiller, Desmond A. Schatz, Dawn E. Beachy, Amanda L. Posgai, Douglas R. Miller, Clive Wasserfall, Justin Mason, Andrew T. Hattersley, Laura M. Jacobsen, Stefanie Engler, Harry S. Nick, and University of Zurich
- Subjects
endocrine system ,endocrine system diseases ,Endocrinology, Diabetes and Metabolism ,610 Medicine & health ,Biology ,Transcriptome ,Diabetes mellitus ,Gene expression ,Internal Medicine ,medicine ,Integrated stress response ,Humans ,Gene ,Pancreas ,Autoantibodies ,Retrospective Studies ,Genetics ,Type 1 diabetes ,Autoantibody ,Genetics/Genomes/Proteomics/Metabolomics ,medicine.disease ,2712 Endocrinology, Diabetes and Metabolism ,Real-time polymerase chain reaction ,Diabetes Mellitus, Type 1 ,Gene Expression Regulation ,2724 Internal Medicine ,Mutation ,11493 Department of Quantitative Biomedicine - Abstract
Type 1 diabetes (T1D) has a multifactorial autoimmune etiology, involving environmental prompts and polygenic predisposition. We hypothesized that pancreata from individuals with and at risk for T1D would exhibit dysregulated expression of genes associated with monogenic forms of diabetes caused by nonredundant single-gene mutations. Using a "monogenetic transcriptomic strategy," we measured the expression of these genes in human T1D, autoantibody-positive (autoantibody+), and control pancreas tissues with real-time quantitative PCR in accordance with the Minimum Information for Publication of Quantitative Real-Time PCR Experiments (MIQE) guidelines. Gene and protein expression was visualized in situ with use of immunofluorescence, RNAscope, and confocal microscopy. Two dozen monogenic diabetes genes showed altered expression in human pancreata from individuals with T1D versus unaffected control subjects. Six of these genes also saw dysregulation in pancreata from autoantibody+ individuals at increased risk for T1D. As a subset of these genes are related to cellular stress responses, we measured integrated stress response (ISR) genes and identified 20 with altered expression in T1D pancreata, including three of the four eIF2α-dependent kinases. Equally intriguing, we observed significant repression of the three arms of the ISR in autoantibody+ pancreata. Collectively, these efforts suggest monogenic diabetes and ISR genes are dysregulated early in the T1D disease process and likely contribute to the disorder's pathogenesis., Diabetes, 70 (8)
- Published
- 2021
- Full Text
- View/download PDF
28. Altered cellular localisation and expression, together with unconventional protein trafficking, of prion protein, PrP(C), in type 1 diabetes
- Author
-
Amanda L. Posgai, Harry S. Nick, Eduardo Candelario-Jalil, Clive Wasserfall, Mark A. Atkinson, Desmond A. Schatz, Dawn E. Beachy, Irina Kusmartseva, Changjun Yang, and Helmut Hiller
- Subjects
Gene isoform ,Adult ,Male ,Stress-Induced-Phosphoprotein 1 ,Adolescent ,Endocrinology, Diabetes and Metabolism ,animal diseases ,Insulin Antibodies ,Scrapie ,Endoplasmic Reticulum ,Real-Time Polymerase Chain Reaction ,Article ,Prion Proteins ,PRNP ,symbols.namesake ,Young Adult ,mental disorders ,Internal Medicine ,Humans ,PrPC Proteins ,RNA, Messenger ,Child ,Pancreas ,Heat-Shock Proteins ,Autoantibodies ,chemistry.chemical_classification ,Chemistry ,Endoplasmic reticulum ,Cell Membrane ,Golgi apparatus ,Immunohistochemistry ,CD56 Antigen ,Tissue Donors ,Cell biology ,nervous system diseases ,Protein Transport ,Diabetes Mellitus, Type 1 ,Gene Expression Regulation ,symbols ,Neural cell adhesion molecule ,Female ,Glycoprotein - Abstract
AIMS/HYPOTHESIS: Normal cellular prion protein (PrP(C)) is a conserved mammalian glycoprotein found on the outer plasma membrane leaflet through a glycophosphatidylinositol anchor. Although PrP(C) is expressed by a wide range of tissues throughout the body, the complete repertoire of its functions has not been fully determined. The misfolded pathogenic isoform PrP(Sc) (the scrapie form of PrP) is a causative agent of neurodegenerative prion diseases. The aim of this study is to evaluate PrP(C) localisation, expression and trafficking in pancreases from organ donors with and without type 1 diabetes and to infer PrP(C) function through studies on interacting protein partners. METHODS: In order to evaluate localisation and trafficking of PrP(C) in the human pancreas, 12 non-diabetic, 12 type 1 diabetic and 12 autoantibody-positive organ donor tissue samples were analysed using immunofluorescence analysis. Furthermore, total RNA was isolated from 29 non-diabetic, 29 type 1 diabetic and 24 autoantibody-positive donors to estimate PrP(C) expression in the human pancreas. Additionally, we performed PrP(C)-specific immunoblot analysis on total pancreatic protein from non-diabetic and type 1 diabetic organ donors to test whether changes in PrP(C) mRNA levels leads to a concomitant increase in PrP(C) protein levels in human pancreases. RESULTS: In non-diabetic and type 1 diabetic pancreases (the latter displaying both insulin-positive [INS(+)] and -negative [INS(−)] islets), we found PrP(C) in islets co-registering with beta cells in all INS(+) islets and, strikingly, unexpected activation of PrP(C) in alpha cells within diabetic INS(−) islets. We found PrP(C) localised to the plasma membrane and endoplasmic reticulum (ER) but not the Golgi, defining two cellular pools and an unconventional protein trafficking mechanism bypassing the Golgi. We demonstrate PrP(C) co-registration with established protein partners, neural cell adhesion molecule 1 (NCAM1) and stress-inducible phosphoprotein 1 (STI1; encoded by STIP1) on the plasma membrane and ER, respectively, linking PrP(C) function with cyto-protection, signalling, differentiation and morphogenesis. We demonstrate that both PRNP (encoding PrP(C)) and STIP1 gene expression are dramatically altered in type 1 diabetic and autoantibody-positive pancreases. CONCLUSIONS/INTERPRETATION: As the first study to address PrP(C) expression in non-diabetic and type 1 diabetic human pancreas, we provide new insights for PrP(C) in the pathogenesis of type 1 diabetes. We evaluated the cell-type specific expression of PrP(C) in the human pancreas and discovered possible connections with potential interacting proteins that we speculate might address mechanisms relevant to the role of PrP(C) in the human pancreas.
- Published
- 2021
29. Temporal Analysis of Amylase Expression in Control, Autoantibody-Positive, and Type 1 Diabetes Pancreatic Tissues
- Author
-
Helmut Hiller, Martha Campbell-Thompson, Clive Wasserfall, Irina Kusmartseva, Maria Beery, Stephen Selman, Amanda L. Posgai, Michael J. Haller, Desmond A. Schatz, Myriam Padilla, Harry S. Nick, and Mark A. Atkinson
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Time Factors ,Adolescent ,Trypsinogen ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Enteroendocrine cell ,Pathophysiology ,Glucagon ,Gene Expression Regulation, Enzymologic ,Young Adult ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pregnancy ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Humans ,Endocrine system ,Amylase ,Child ,Pancreas ,Aged ,Autoantibodies ,Type 1 diabetes ,biology ,Infant, Newborn ,Infant ,Middle Aged ,medicine.disease ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,Endocrinology ,chemistry ,Case-Control Studies ,Child, Preschool ,Amylases ,biology.protein ,Female ,Hormone - Abstract
Within the human pancreas, exocrine and endocrine cells control secretion of digestive enzymes and production of hormones to maintain metabolic homeostasis, respectively. While the vast majority of type 1 diabetes research efforts have focused on endocrine function and autoimmunity, recent studies identified a series of unique features (e.g., reduced weight and volume, increased density of leukocytes) within the exocrine pancreas in this disease, but the mechanisms underlying these aberrancies are unknown. Therefore, we histologically assessed amylase, insulin, glucagon, lipase, and/or trypsinogen in 78 organ donor pancreata from birth through adulthood in control subjects and those at various stages of type 1 diabetes. While amylase-positive (AMY(+)) acinar cells were detectable in pancreata from all study groups, tissues from individuals >2 years of age contained clusters of acinar cells devoid of amylase (AMY(−)). A majority of these AMY(−) cell clusters localized proximal to islets (i.e., peri-islet). Additionally, most AMY(−) clusters were positive for the exocrine enzymes lipase and trypsinogen. Interestingly, type 1 diabetes pancreata displayed significant reductions in the frequency of these AMY(−) cell clusters. These results support a contribution of the islet-acinar axis in pancreatic development and underscore a potential role for the exocrine pancreas in the pathogenesis of type 1 diabetes.
- Published
- 2019
- Full Text
- View/download PDF
30. De-coding genetic risk variants in type 1 diabetes
- Author
-
Robert C. Sharp, Leeana D. Peters, Puchong Thirawatananond, Melanie R. Shapiro, Daniel J. Perry, Amanda L. Posgai, Todd M. Brusko, and Similoluwa Ogundare
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Genotype ,Immunology ,Single-nucleotide polymorphism ,Disease ,Computational biology ,Human leukocyte antigen ,Biology ,Genome ,Polymorphism, Single Nucleotide ,Article ,PTPN22 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,Molecular genetics ,medicine ,Genetic predisposition ,Immunology and Allergy ,Animals ,Genetic Predisposition to Disease ,Gene ,Cell Biology ,030104 developmental biology ,Diabetes Mellitus, Type 1 ,030215 immunology - Abstract
The conceptual basis for a genetic predisposition underlying the risk for developing type 1 diabetes (T1D) predates modern human molecular genetics. Over half of the genetic risk has been attributed to the human leukocyte antigen (HLA) class II gene region and to the insulin (INS) gene locus - both thought to confer direction of autoreactivity and tissue specificity. Notwithstanding, questions still remain regarding the functional contributions of a vast array of minor polygenic risk variants scattered throughout the genome that likely influence disease heterogeneity and clinical outcomes. Herein, we summarize the available literature related to the T1D-associated coding variants defined at the time of this review, for the genes PTPN22, IFIH1, SH2B3, CD226, TYK2, FUT2, SIRPG, CTLA4, CTSH and UBASH3A. Data from genotype-selected human cohorts are summarized, and studies from the non-obese diabetic (NOD) mouse are presented to describe the functional impact of these variants in relation to innate and adaptive immunity as well as to β-cell fragility, with expression profiles in tissues and peripheral blood highlighted. The contribution of each variant to progression through T1D staging, including environmental interactions, are discussed with consideration of how their respective protein products may serve as attractive targets for precision medicine-based therapeutics to prevent or suspend the development of T1D.
- Published
- 2021
31. Genetic Composition and Autoantibody Titers Model the Probability of Detecting C-Peptide Following Type 1 Diabetes Diagnosis
- Author
-
C Ramsey Grace, Mark A. Atkinson, Srikar Chamala, Daniel J. Perry, Desmond A. Schatz, MacKenzie D. Williams, Andrew Muir, Kieran M. Mcgrail, Amanda L. Posgai, Rhonda Bacher, Michael J. Haller, Clive Wasserfall, and Todd M. Brusko
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Zinc Transporter 8 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Humans ,Receptor-Like Protein Tyrosine Phosphatases, Class 8 ,Generalized estimating equation ,Autoantibodies ,geography ,Type 1 diabetes ,geography.geographical_feature_category ,C-Peptide ,business.industry ,C-peptide ,Autoantibody ,Islet ,medicine.disease ,Cross-Sectional Studies ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,chemistry ,Cohort ,business - Abstract
We and others previously demonstrated that a type 1 diabetes genetic risk score (GRS) improves the ability to predict disease progression and onset in at-risk subjects with islet autoantibodies. Here, we hypothesized that GRS and islet autoantibodies, combined with age at onset and disease duration, could serve as markers of residual β-cell function following type 1 diabetes diagnosis. Generalized estimating equations were used to investigate whether GRS along with insulinoma-associated protein-2 autoantibody (IA–2A), zinc transporter 8 autoantibody (ZnT8A), and GAD autoantibody (GADA) titers were predictive of C-peptide detection in a largely cross-sectional cohort of 401 subjects with type 1 diabetes (median duration 4.5 years [range 0–60]). Indeed, a combined model with incorporation of disease duration, age at onset, GRS, and titers of IA–2A, ZnT8A, and GADA provided superior capacity to predict C-peptide detection (quasi-likelihood information criterion [QIC] = 334.6) compared with the capacity of disease duration, age at onset, and GRS as the sole parameters (QIC = 359.2). These findings support the need for longitudinal validation of our combinatorial model. The ability to project the rate and extent of decline in residual C-peptide production for individuals with type 1 diabetes could critically inform enrollment and benchmarking for clinical trials where investigators are seeking to preserve or restore endogenous β-cell function.
- Published
- 2021
- Full Text
- View/download PDF
32. Limited Extent and Consequences of Pancreatic SARS-CoV-2 Infection
- Author
-
Travis Dawson, Raveen Rathnasinghe, Alejandro Caicedo, Jingjin Qui, Mark A. Atkinson, Seunghee Kim-Schulze, Adeeb H. Rahman, Geoffrey Kelly, Teresa Aydillo, Michael Schotsaert, Verena van der Heide, Amanda L. Posgai, Daniel Geanon, Irina Kusmartseva, Darwin D'Souza, Brian Lee, Brad Rosenberg, Julia K. Panzer, Diana Handler, Sonia Jangra, Dirk Homann, Phillip Cohen, Randy Albrecht, Sadaf Aslam, and Adolfo García-Sastre
- Subjects
Oncology ,History ,medicine.medical_specialty ,Diabetes risk ,Polymers and Plastics ,Coronavirus disease 2019 (COVID-19) ,SARS-CoV-2 ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Center of excellence ,COVID-19 ,Influenza research ,Viral infection ,General Biochemistry, Genetics and Molecular Biology ,Industrial and Manufacturing Engineering ,Insulin-Secreting Cells ,Internal medicine ,Diabetes Mellitus ,medicine ,Humans ,Business and International Management ,business ,Pancreas ,Viral load - Abstract
Concerns that infection with SARS-CoV-2, the etiological agent of COVID-19, may cause new-onset diabetes persist amidst an evolving research landscape, and precise risk assessment is hampered by at times conflicting evidence. Here, leveraging comprehensive single-cell analyses of in vitro SARS-CoV-2-infected human pancreatic islets, we demonstrate that productive infection is strictly dependent on the SARS-CoV-2 entry receptor ACE2 and targets all pancreatic cell types. Importantly, the infection remains highly circumscribed, largely non-cytopathic, and despite high viral burden in infected subsets, promotes only modest cellular perturbations and inflammatory responses. Similar experimental outcomes are also observed after islet infection with endemic coronaviruses. Thus, the limits of pancreatic SARS-CoV-2 infection, even under in vitro conditions of enhanced virus exposure, do not support the proposition that in vivo targeting of beta cells by SARS-CoV-2 precipitates new-onset diabetes. If restricted pancreatic damage accrued by COVID-19 increases cumulative diabetes risk, however, remains to be evaluated. Funding: These efforts were supported by JDRF 3-PDF-2018-575-A-N (V.v.d.H.); NIH/NIDDK R01DK12392, NIH/NIAID P01AI042288 and NIH/NIAID U54AI142766-S1 (M.A.A.); NIH/NIAID Center of Excellence for Influenza Research and Response/Center for Research for Influenza Pathogenesis and Transmission contract # 75N93019R00028, NIH/NIAID U19AI135972 (supplement), Defense Advanced Research Projects Agency HR0011-19-2-0020, JPB Foundation, and Open Philanthropy Project # 2020-215611 (5384), Anonymous (A.G.-S.); NIH/NIAID R01AI151029 and NIA/NIAID U01AI150748 (B.R.R.); NIH/NIDDK R01DK130425 (M.S.); and NIH/NIAID R01AI134971, NIH/NIDDK U01DK123716, NIH/NIDDK U01DK104162, NIH/NIDDK P30DK020541 and NIH/NIDDK R01DK130425 (D.H.). Funding: The AG-S laboratory has received research support from Pfizer, Senhwa Biosciences, Kenall Manufacturing, Avimex, Johnson & Johnson, Dynavax, 7Hills Pharma, Pharmamar, ImmunityBio, Accurius, Nanocomposix, Hexamer, N-fold LLC, Model Medicines and Merck, outside of the reported work. Declaration of Interests: AG-S has consulting agreements for the following companies involving cash and/or stock: Vivaldi Biosciences, Contrafect, 7Hills Pharma, Avimex, Vaxalto, Pagoda, Accurius, Esperovax, Farmak, Applied Biological Laboratories and Pfizer, outside of the reported work. AG-S is inventor on patents and patent applications on the use of antivirals and vaccines for the treatment and prevention of virus infections and cancer, owned by the Icahn School of Medicine at Mount Sinai, New York, outside of the reported work. All other authors declare no conflict of interest. Ethics Approval Statement: Our study is considered “not human subjects research” since all donor islet preparations were provided as de-identified tissue specimens by a commercial purveyor
- Published
- 2021
- Full Text
- View/download PDF
33. Response to Comment on Dunne et al. The Women’s Leadership Gap in Diabetes: A Call for Equity and Excellence. Diabetes Care 2021;44:1734–1743
- Author
-
Jessica L. Dunne, Jennifer L. Maizel, Amanda L. Posgai, Mark A. Atkinson, and Linda A. DiMeglio
- Subjects
Advanced and Specialized Nursing ,Endocrinology, Diabetes and Metabolism ,Internal Medicine - Published
- 2022
- Full Text
- View/download PDF
34. Combination treatment with antigen-specific dual-sized microparticle system plus anti-CD3 immunotherapy fails to synergize to improve late-stage type 1 diabetes prevention in non-obese diabetic mice
- Author
-
Joshua M. Stewart, Michael J. Haller, Amanda L. Posgai, J J Leon, and Benjamin G. Keselowsky
- Subjects
CD86 ,Regulatory T cell ,business.industry ,medicine.medical_treatment ,Biomedical Engineering ,Nod ,Immunotherapy ,Dendritic Cells ,medicine.disease_cause ,Article ,Autoimmunity ,Proinflammatory cytokine ,Diabetes Mellitus, Experimental ,Biomaterials ,Mice ,medicine.anatomical_structure ,Immune system ,Diabetes Mellitus, Type 1 ,Downregulation and upregulation ,Mice, Inbred NOD ,Immunology ,medicine ,Animals ,business - Abstract
Type 1 diabetes (T1D) pathophysiology, while incompletely understood, has in part been attributed to aberrant presentation of self-antigen plus proinflammatory co-stimulation by professional antigen-presenting cells (APCs). Therapies targeting dendritic cells (DCs) offer an avenue to restore antigen-specific tolerance by promoting presentation of self-antigen in an anti-inflammatory or suppressive context. Here, we describe a subcutaneously administered, dual-sized biodegradable microparticle (MP) platform that includes phagocytosable (~1 μm) and non-phagocytosable (~30 μm) MPs to deliver pro-tolerogenic factors both intra- and extracellularly, as well as the T1D-associated autoantigen, insulin, to DCs for amelioration of autoimmunity. This MP platform resulted in increased recruitment of DCs, suppressive skewing of DC phenotype with diminished expression of CD86 and MHC-II, increased regulatory T cell (Treg) frequency, and upregulated expression of the checkpoint inhibitor programmed cell death protein 1 (PD-1) on T cells. When administered concomitantly with anti-CD3 antibody, which provides transient T cell depletion while preserving Treg populations, in 12-week-old non-obese diabetic (NOD) mice, regulatory immune populations persisted out to 20 weeks of age; however, combination anti-CD3 + dual sized MP (dMP) therapy failed to synergistically inhibit diabetes onset.
- Published
- 2020
35. ACE2 and SARS-CoV-2 Expression in the Normal and COVID-19 Pancreas
- Author
-
Changjun Yang, Fahim Syed, Harry S. Nick, Jack L. Harbert, Marda Jorgensen, Sirlene Cechin, Richard E. Lloyd, Mark A. Atkinson, Eduardo Candelario-Jalil, Verena van der Heide, Vander Heide Rs, Amanda L. Posgai, Wenting Wu, Carmella Evans-Molina, Tang X, Irina Kusmartseva, Martha Campbell-Thompson, Alberto Pugliese, Dirk Homann, and Paul N. Joseph
- Subjects
Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,In silico ,Enteroendocrine cell ,medicine.disease ,Blot ,medicine.anatomical_structure ,Diabetes mellitus ,medicine ,Respiratory system ,Pancreas ,business ,Ex vivo ,Fluorescence in situ hybridization - Abstract
SUMMARYDiabetes is associated with increased mortality from Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). Given literature suggesting a potential association between SARS-CoV-2 infection and diabetes induction, we examined pancreatic expression of the key molecule for SARS-CoV-2 infection of cells, angiotensin-converting enzyme-2 (ACE2). Specifically, we analyzed five public scRNAseq pancreas datasets and performed fluorescencein situhybridization, Western blotting, and immunolocalization for ACE2 with extensive reagent validation on normal human pancreatic tissues across the lifespan, as well as those from coronavirus disease 2019 (COVID-19) patients. Thesein silicoandex vivoanalyses demonstrated pancreatic expression of ACE2 is prominent in pancreatic ductal epithelium and the microvasculature, with rare endocrine cell expression of this molecule. Pancreata from COVID-19 patients demonstrated multiple thrombotic lesions with SARS-CoV-2 nucleocapsid protein expression primarily limited to ducts. SARS-CoV-2 infection of pancreatic endocrine cells, via ACE2, appears an unlikely central pathogenic feature of COVID-19 as it relates to diabetes.
- Published
- 2020
- Full Text
- View/download PDF
36. 1663-P: Gene Editing of the Human T-Cell Receptor Locus Enables Optimized In Vitro Testing of Autoreactive T-Cell Function in Type 1 Diabetes
- Author
-
Amanda L. Posgai, Leeana D. Peters, and Todd M. Brusko
- Subjects
Endocrinology, Diabetes and Metabolism ,T cell ,medicine.medical_treatment ,T-cell receptor ,Immunotherapy ,Biology ,Cell biology ,Cell therapy ,medicine.anatomical_structure ,Antigen ,Downregulation and upregulation ,Internal Medicine ,medicine ,IL-2 receptor ,CD8 - Abstract
Gene editing of the human T cell receptor locus enables optimized in vitro testing of autoreactive T cell function in type 1 diabetes. The generation of antigen specific T cells and subsequent implementation as “living drugs” has garnered great interest in the immunotherapy space. One of the main caveats to this approach is the potential for heterologous chain pairing between the α- and β-chains of the endogenous and de-novo T cell receptors (TCRs). The consequences of chain mispairing can range from reduced antigen specific functionality to off-target autoimmune reactivity. The latter is an important consideration when designing therapies for autoimmune and inflammatory diseases, specifically with regards to the introduction of regulatory T cells (Treg) adoptive cell therapy. To date, infusions of polyclonal Tregs have demonstrated safety but have failed to reverse established type 1 diabetes (T1D). This demonstrates the need for the generation of safe and effective antigen-specific Tregs capable of recognizing β-cells in T1D. We have previously shown the ability to generate GAD-specific Tregs which maintain lineage stability and suppressive phenotype. To extend this work, we deleted the endogenous TCR α-chain gene (TRAC) via CRISPR/cas9 in CD8+T cells. We achieved >90% editing efficiency through ribonucleoprotein (RNP) complex mediated single guide RNA delivery. Moreover, we found TRAC-KO CD8+ T cells displayed increased peptide MHC-dextramer staining, CD69 and CD25 upregulation after stimulation with cognate antigen, and increased antigen-specific killing of β-cell lines when compared to mock guide treated T cells. We believe this workflow can be applied to generate antigen specific Tregs, which we expect to provide increased suppressive capacity. Hence, we present a TCR-gene editing protocol in primary human T cells that results in efficient deletion of the endogenous TCR α-chain and robust enhancement of antigen-specific T cell function. Disclosure L.D. Peters: None. A.L. Posgai: None. T.M. Brusko: Board Member; Self; OneVax, LLC. Consultant; Self; Caladrius Biosciences, Inc., Sanofi-Aventis. Speaker’s Bureau; Self; CSL Behring. Funding National Institutes of Health (P01AI42288, R01DK106191, T325T32DK108736-03HIRN, UC4 P0063595)
- Published
- 2020
- Full Text
- View/download PDF
37. Histological validation of a type 1 diabetes clinical diagnostic model for classification of diabetes
- Author
-
Martha Campbell-Thompson, Clive Wasserfall, Maria Beery, Richard A. Oram, Daniel J. Perry, Todd M. Brusko, Srikar Chamala, Irina Kusmartseva, Sarah J. Richardson, Angus G. Jones, Desmond A. Schatz, Christine Flaxman, Seth A. Sharp, Anita L. Lynam, Lauric A. Ferrat, Beverley M. Shields, Laura M. Jacobsen, Alice L. J. Carr, Mark A. Atkinson, and Amanda L. Posgai
- Subjects
Adult ,Male ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Type 2 diabetes ,Zinc Transporter 8 ,Article ,Body Mass Index ,Diagnosis, Differential ,03 medical and health sciences ,Islets of Langerhans ,Young Adult ,0302 clinical medicine ,Endocrinology ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,Diabetes Mellitus ,Medicine ,Humans ,Insulin ,Genetic Predisposition to Disease ,030212 general & internal medicine ,Age of Onset ,Pancreas ,Autoantibodies ,Type 1 diabetes ,Receiver operating characteristic ,C-Peptide ,business.industry ,Surrogate endpoint ,Area under the curve ,Reproducibility of Results ,Middle Aged ,medicine.disease ,Diabetes Mellitus, Type 1 ,Diabetes Mellitus, Type 2 ,Biomarker (medicine) ,Histopathology ,Female ,business - Abstract
AIMS Misclassification of diabetes is common due to an overlap in the clinical features of type 1 and type 2 diabetes. Combined diagnostic models incorporating clinical and biomarker information have recently been developed that can aid classification, but they have not been validated using pancreatic pathology. We evaluated a clinical diagnostic model against histologically defined type 1 diabetes. METHODS We classified cases from the Network for Pancreatic Organ donors with Diabetes (nPOD) biobank as type 1 (n = 111) or non-type 1 (n = 42) diabetes using histopathology. Type 1 diabetes was defined by lobular loss of insulin-containing islets along with multiple insulin-deficient islets. We assessed the discriminative performance of previously described type 1 diabetes diagnostic models, based on clinical features (age at diagnosis, BMI) and biomarker data [autoantibodies, type 1 diabetes genetic risk score (T1D-GRS)], and singular features for identifying type 1 diabetes by the area under the curve of the receiver operator characteristic (AUC-ROC). RESULTS Diagnostic models validated well against histologically defined type 1 diabetes. The model combining clinical features, islet autoantibodies and T1D-GRS was strongly discriminative of type 1 diabetes, and performed better than clinical features alone (AUC-ROC 0.97 vs. 0.95; P = 0.03). Histological classification of type 1 diabetes was concordant with serum C-peptide [median
- Published
- 2020
38. Innate Inflammation Drives NK Cell Activation to Impair Treg Activity
- Author
-
Wen-I Yeh, Mark A. Atkinson, Michael J. Haller, Andrew Muir, Scott E. Stimpson, Amanda L. Posgai, Joseph W. Dean, Leeana D. Peters, Todd M. Brusko, Clayton E. Mathews, Christopher Fuhrman, Howard R. Seay, Daniel J. Perry, Brittney N. Newby, and Maigan A. Brusko
- Subjects
0301 basic medicine ,Adult ,Cytotoxicity, Immunologic ,Male ,Adolescent ,Immunology ,Quantitative Trait Loci ,chemical and pharmacologic phenomena ,Biology ,Lymphocyte Activation ,Models, Biological ,T-Lymphocytes, Regulatory ,Article ,Immunophenotyping ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Immune system ,TIGIT ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,IL-2 receptor ,Lymphocyte Count ,Child ,Cells, Cultured ,030203 arthritis & rheumatology ,Inflammation ,FOXP3 ,Middle Aged ,NKG2D ,Immunity, Innate ,Killer Cells, Natural ,CTL ,030104 developmental biology ,Diabetes Mellitus, Type 1 ,Phenotype ,Interleukin 12 ,Cytokines ,Female ,Disease Susceptibility ,Biomarkers ,T-Lymphocytes, Cytotoxic - Abstract
IL-12 and IL-18 synergize to promote T(H)1 responses and have been implicated as accelerators of autoimmune pathogenesis in type 1 diabetes (T1D). We investigated the influence of these cytokines on immune cells involved in human T1D progression: natural killer (NK) cells, regulatory T cells (Tregs), and cytotoxic T lymphocytes (CTL). NK cells from T1D patients exhibited higher surface CD226 versus controls and lower CD25 compared to first-degree relatives and controls. Changes in NK cell phenotype towards terminal differentiation were associated with cytomegalovirus (CMV) seropositivity, while possession of IL18RAP, IFIH1, and IL2RA T1D-risk variants impacted NK cell activation as evaluated by immuno-expression quantitative trait loci (eQTL) analyses. IL-12 and IL-18 stimulated NK cells from healthy donors exhibited enhanced specific killing of myelogenous K562 target cells. Moreover, activated NK cell increased expression of NKG2A, NKG2D, CD226, TIGIT and CD25, which enabled competition for IL-2 upon co-culture with Tregs, resulting in Treg downregulation of FOXP3, production of IFNγ, and loss of suppressive function. We generated islet-autoreactive CTL “avatars”, which upon exposure to IL-12 and IL-18, upregulated IFNγ and Granzyme-B leading to increased lymphocytotoxicity of a human β-cell line in vitro. These results support a model for T1D pathogenesis wherein IL-12 and IL-18 synergistically enhance CTL and NK cell cytotoxic activity and disrupt immunoregulation by Tregs.
- Published
- 2020
39. Multiplexing DNA methylation markers to detect circulating cell-free DNA derived from human pancreatic β cells
- Author
-
Saar Hashavia, Ori Fridlich, Ruth Shemer, Yuval Dor, Tal Oron, Benjamin Glaser, Desmond A. Schatz, Daniel Neiman, Cate Speake, Olle Korsgren, Gun Forsander, Oskar Skog, David Gillis, Dan Cohen, Mark A. Atkinson, Floris Levy-Khademi, Frida Sundberg, Carla J. Greenbaum, Jennifer Hosford, Dan Arbel, A. M. James Shapiro, Sheina Piyanzin, Aviad Zick, Amanda L. Posgai, and Joshua Moss
- Subjects
0301 basic medicine ,Adult ,Male ,Adolescent ,medicine.medical_treatment ,Cell ,03 medical and health sciences ,chemistry.chemical_compound ,Young Adult ,0302 clinical medicine ,Insulin-Secreting Cells ,medicine ,Humans ,Insulin ,Epigenetics ,Child ,Cell damage ,Aged ,Chemistry ,General Medicine ,DNA Methylation ,Middle Aged ,medicine.disease ,Molecular biology ,Circulating Cell-Free DNA ,Transplantation ,030104 developmental biology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 1 ,030220 oncology & carcinogenesis ,Child, Preschool ,DNA methylation ,Female ,Cell-Free Nucleic Acids ,DNA ,Biomarkers ,Research Article - Abstract
It has been proposed that unmethylated insulin promoter fragments in plasma derive exclusively from β cells, reflect their recent demise, and can be used to assess β cell damage in type 1 diabetes. Herein we describe an ultrasensitive assay for detection of a β cell–specific DNA methylation signature, by simultaneous assessment of 6 DNA methylation markers, that identifies β cell DNA in mixtures containing as little as 0.03% β cell DNA (less than 1 β cell genome equivalent). Based on this assay, plasma from nondiabetic individuals (N = 218, aged 4–78 years) contained on average only 1 β cell genome equivalent/mL. As expected, cell-free DNA (cfDNA) from β cells was significantly elevated in islet transplant recipients shortly after transplantation. We also detected β cell cfDNA in a patient with KATP congenital hyperinsulinism, in which substantial β cell turnover is thought to occur. Strikingly, in contrast to previous reports, we observed no elevation of β cell–derived cfDNA in autoantibody-positive subjects at risk for type 1 diabetes (N = 32), individuals with recent-onset type 1 diabetes (4 months, N = 38). We discuss the utility of sensitive β cell cfDNA analysis and potential explanations for the lack of a β cell cfDNA signal in type 1 diabetes.
- Published
- 2020
40. ACE2 and SARS-CoV-2 Expression in the Normal and COVID-19 Pancreas
- Author
-
Eduardo Candelario-Jalil, Amanda L. Posgai, Dirk Homann, Irina Kusmartseva, Marda Jorgensen, Richard E. Lloyd, Jack L. Harbert, Wenting Wu, Farooq Syed, Mark A. Atkinson, Verena van der Heide, Carmella Evans-Molina, Tang X, Alberto Pugliese, Martha Campbell-Thompson, Richard S. Vander Heide, Harry S. Nick, Changjun Yang, Paul N. Joseph, and Sirlene Cechin
- Subjects
Oncology ,Kidney ,medicine.medical_specialty ,Type 1 diabetes ,business.industry ,CD34 ,Enteroendocrine cell ,Type 2 diabetes ,medicine.disease ,Institutional review board ,medicine.anatomical_structure ,Internal medicine ,Diabetes mellitus ,medicine ,Pancreas ,business - Abstract
Diabetes is associated with increased mortality from Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). Given literature suggesting a potential association between SARS-CoV-2 infection and diabetes induction, we examined pancreatic expression of the key molecule for SARS-CoV-2 infection of cells, angiotensin-converting enzyme-2 (ACE2). Specifically, we analyzed five public scRNAseq pancreas datasets and performed fluorescence in situ hybridization, Western blotting, and immunolocalization for ACE2 with extensive reagent validation on normal human pancreatic tissues across the lifespan, as well as those from coronavirus disease 2019 (COVID-19) patients. These in silico and ex vivo analyses demonstrated pancreatic expression of ACE2 is prominent in pancreatic ductal epithelium and the microvasculature, with rare endocrine cell expression of this molecule. Pancreata from COVID-19 patients demonstrated multiple thrombotic lesions with SARS-CoV-2 nucleocapsid protein expression primarily limited to ducts. SARS-CoV-2 infection of pancreatic endocrine cells, via ACE2, appears an unlikely central pathogenic feature of COVID-19 as it relates to diabetes. Funding: These efforts were supported by NIH P01 AI42288 and UC4 DK108132 (MAA), JDRF (MAA), NIH R01 DK122160 (MCT), NIH R01 AI134971 (DH), NIH P30 DK020541 (D.H.), JDRF 3-PDF-2018-575-A-N (VvdH), R01 DK093954 (CEM); VA Merit Award I01BX001733 (CEM), Imaging Core of NIH/NIDDK P30 DK097512 (CEM), gifts from the Sigma Beta Sorority, the Ball Brothers Foundation, and the George and Frances Ball Foundation (CEM), the Network for Pancreatic Organ donors with Diabetes (nPOD; RRID:SCR_014641) (5-SRA-2018-557-Q-R) and The Leona M. & Harry B. Helmsley Charitable Trust (2018PG-T1D053). Conflict of Interest: The authors declare no relevant conflicts of interest exist. Ethical Approval: Transplant-quality pancreas, duodenum, and kidney were recovered by JDRF nPOD (www.jdrfnpod.com) from 36 COVID-19 negative organ donors without diabetes (Table S2) according to established protocols and procedures (Campbell-Thompson et al., 2012), as approved by the University of Florida Institutional Review Board (201400486), the United Network for Organ Sharing (UNOS), and according to federal guidelines with informed consent obtained from each donor’s legal representative.
- Published
- 2020
- Full Text
- View/download PDF
41. Clinical Applications of Regulatory T cells in Adoptive Cell Therapies
- Author
-
Gregory P Marshall, Todd M. Brusko, Ahmed Gomaa El-Sayed, Howard R. Seay, Wen-I Yeh, Judit Cserny, Amanda L. Posgai, and Daniel J. Perry
- Subjects
medicine.anatomical_structure ,Cell ,medicine ,Cancer research ,chemical and pharmacologic phenomena ,Biology ,General Economics, Econometrics and Finance ,Article - Abstract
Interest in adoptive T-cell therapies has been ignited by the recent clinical success of genetically-modified T cells in the cancer immunotherapy space. In addition to immune targeting for malignancies, this approach is now being explored for the establishment of immune tolerance with regulatory T cells (Tregs). Herein, we will summarize the basic science and clinical results emanating from trials directed at inducing durable immune regulation through administration of Tregs. We will discuss some of the current challenges facing the field in terms of maximizing cell purity, stability and expansion capacity, while also achieving feasibility and GMP production. Indeed, recent advances in methodologies for Treg isolation, expansion, and optimal source materials represent important strides toward these considerations. Finally, we will review the emerging genetic and biomaterial-based approaches on the horizon for directing Treg specificity to augment tissue-targeting and regenerative medicine.
- Published
- 2018
42. Insulin-Like Growth Factor Dysregulation Both Preceding and Following Type 1 Diabetes Diagnosis
- Author
-
Andrew Muir, Amanda L. Posgai, Cate Speake, Todd M. Brusko, Sean M. Mcgrail, Melanie R. Shapiro, Desmond A. Schatz, Mark A. Atkinson, Clive Wasserfall, Rhonda Bacher, William Hagopian, Michael J. Haller, Matthias von Herrath, and Johnna D. Wesley
- Subjects
0301 basic medicine ,Adult ,Male ,endocrine system ,medicine.medical_specialty ,Time Factors ,Adolescent ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,030209 endocrinology & metabolism ,Disease ,Carbohydrate metabolism ,Pathophysiology ,Cohort Studies ,Prediabetic State ,03 medical and health sciences ,Insulin-like growth factor ,Young Adult ,0302 clinical medicine ,Insulin-Like Growth Factor II ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Humans ,Insulin-Like Growth Factor I ,Child ,Autoantibodies ,Type 1 diabetes ,business.industry ,Autoantibody ,medicine.disease ,Insulin-Like Growth Factor Binding Proteins ,030104 developmental biology ,Endocrinology ,Cross-Sectional Studies ,Diabetes Mellitus, Type 1 ,Cohort ,Disease Progression ,Female ,business ,Biomarkers ,hormones, hormone substitutes, and hormone antagonists ,Cohort study - Abstract
Insulin-like growth factors (IGFs), specifically IGF1 and IGF2, promote glucose metabolism, with their availability regulated by IGF-binding proteins (IGFBPs). We hypothesized that IGF1 and IGF2 levels, or their bioavailability, are reduced during type 1 diabetes development. Total serum IGF1, IGF2, and IGFBP1–7 levels were measured in an age-matched, cross-sectional cohort at varying stages of progression to type 1 diabetes. IGF1 and IGF2 levels were significantly lower in autoantibody (AAb)+ compared with AAb− relatives of subjects with type 1 diabetes. Most high-affinity IGFBPs were unchanged in individuals with pre–type 1 diabetes, suggesting that total IGF levels may reflect bioactivity. We also measured serum IGFs from a cohort of fasted subjects with type 1 diabetes. IGF1 levels significantly decreased with disease duration, in parallel with declining β-cell function. Additionally, plasma IGF levels were assessed in an AAb+ cohort monthly for a year. IGF1 and IGF2 showed longitudinal stability in single AAb+ subjects, but IGF1 levels decreased over time in subjects with multiple AAb and those who progressed to type 1 diabetes, particularly postdiagnosis. In sum, IGFs are dysregulated both before and after the clinical diagnosis of type 1 diabetes and may serve as novel biomarkers to improve disease prediction.
- Published
- 2019
43. Human Regulatory T Cells From Umbilical Cord Blood Display Increased Repertoire Diversity and Lineage Stability Relative to Adult Peripheral Blood
- Author
-
Keshav Motwani, Leeana D. Peters, Willem H. Vliegen, Ahmed Gomaa El-sayed, Howard R. Seay, M. Cecilia Lopez, Henry V. Baker, Amanda L. Posgai, Maigan A. Brusko, Daniel J. Perry, Rhonda Bacher, Joseph Larkin, Michael J. Haller, and Todd M. Brusko
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,Adult ,immunosequencing ,Naive T cell ,T cell ,Immunology ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Tregs ,Biology ,Lymphocyte Activation ,Immunotherapy, Adoptive ,T-Lymphocytes, Regulatory ,CCL5 ,regulatory T cells ,Immune tolerance ,03 medical and health sciences ,0302 clinical medicine ,TIGIT ,scRNA-seq ,medicine ,Immunology and Allergy ,Humans ,Cell Lineage ,IL-2 receptor ,RNA-Seq ,Interleukin-7 receptor ,Original Research ,FOXP3 ,hemic and immune systems ,adoptive cell therapy ,peripheral blood ,Fetal Blood ,030104 developmental biology ,medicine.anatomical_structure ,Phenotype ,cord blood ,lcsh:RC581-607 ,030215 immunology - Abstract
The human T lymphocyte compartment is highly dynamic over the course of a lifetime. Of the many changes, perhaps most notable is the transition from a predominantly naïve T cell state at birth to the acquisition of antigen-experienced memory and effector subsets following environmental exposures. These phenotypic changes, including the induction of T cell exhaustion and senescence, have the potential to negatively impact efficacy of adoptive T cell therapies (ACT). When considering ACT with CD4+CD25+CD127–/lo regulatory T cells (Tregs) for the induction of immune tolerance, we previously reported ex vivo expanded umbilical cord blood (CB) Tregs remained more naïve, suppressed responder T cells equivalently, and exhibited a more diverse T cell receptor (TCR) repertoire compared to expanded adult peripheral blood (APB) Tregs. Herein, we hypothesized that upon further characterization, we would observe increased lineage heterogeneity and phenotypic diversity in APB Tregs that might negatively impact lineage stability, engraftment capacity, and the potential for Tregs to home to sites of tissue inflammation following ACT. We compared the phenotypic profiles of human Tregs isolated from CB versus the more traditional source, APB. We conducted analysis of fresh and ex vivo expanded Treg subsets at both the single cell (scRNA-seq and flow cytometry) and bulk (microarray and cytokine profiling) levels. Single cell transcriptional profiles of pre-expansion APB Tregs highlighted a cluster of cells that showed increased expression of genes associated with effector and pro-inflammatory phenotypes (CCL5, GZMK, CXCR3, LYAR, and NKG7) with low expression of Treg markers (FOXP3 and IKZF2). CB Tregs were more diverse in TCR repertoire and homogenous in phenotype, and contained fewer effector-like cells in contrast with APB Tregs. Interestingly, expression of canonical Treg markers, such as FOXP3, TIGIT, and IKZF2, were increased in CB CD4+CD127+ conventional T cells (Tconv) compared to APB Tconv, post-expansion, implying perinatal T cells may adopt a default regulatory program. Collectively, these data identify surface markers (namely CXCR3) that could be depleted to improve purity and stability of APB Tregs, and support the use of expanded CB Tregs as a potentially optimal ACT modality for the treatment of autoimmune and inflammatory diseases.
- Published
- 2019
44. Islet-immune interactions in type 1 diabetes: the nexus of beta cell destruction
- Author
-
Leeana D. Peters, Amanda L. Posgai, and Todd M. Brusko
- Subjects
0301 basic medicine ,Immunology ,Inflammation ,Autoimmunity ,Disease ,Immune receptor ,Biology ,medicine.disease_cause ,Immune tolerance ,03 medical and health sciences ,Islets of Langerhans ,0302 clinical medicine ,Immune system ,Insulin-Secreting Cells ,medicine ,Immune Tolerance ,Immunology and Allergy ,Animals ,Humans ,Insulin ,Pancreas ,geography ,geography.geographical_feature_category ,Islet ,030104 developmental biology ,Diabetes Mellitus, Type 1 ,Immunology of Diabetes Society Review Series: Insights into Pathogenesis of Type 1 Diabetes Series Editors: F. Susan Wong and Timothy I. Tree ,Beta cell ,medicine.symptom ,Neuroscience ,030215 immunology - Abstract
Summary Recent studies in Type 1 Diabetes (T1D) support an emerging model of disease pathogenesis that involves intrinsic β-cell fragility combined with defects in both innate and adaptive immune cell regulation. This combination of defects induces systematic changes leading to organ-level atrophy and dysfunction of both the endocrine and exocrine portions of the pancreas, ultimately culminating in insulin deficiency and β-cell destruction. In this review, we discuss the animal model data and human tissue studies that have informed our current understanding of the cross-talk that occurs between β-cells, the resident stroma, and immune cells that potentiate T1D. Specifically, we will review the cellular and molecular signatures emerging from studies on tissues derived from organ procurement programs, focusing on in situ defects occurring within the T1D islet microenvironment, many of which are not yet detectable by standard peripheral blood biomarkers. In addition to improved access to organ donor tissues, various methodological advances, including immune receptor repertoire sequencing and single-cell molecular profiling, are poised to improve our understanding of antigen-specific autoimmunity during disease development. Collectively, the knowledge gains from these studies at the islet–immune interface are enhancing our understanding of T1D heterogeneity, likely to be an essential component for instructing future efforts to develop targeted interventions to restore immune tolerance and preserve β-cell mass and function.
- Published
- 2019
45. 1366-P: Effect of Substance Use on Type 1 Diabetes (T1D) Pancreas Histopathology
- Author
-
Amanda L. Posgai, Brittany S. Bruggeman, Laura M. Jacobsen, Desmond A. Schatz, Mark A. Atkinson, and Martha Campbell-Thompson
- Subjects
endocrine system ,medicine.medical_specialty ,endocrine system diseases ,business.industry ,Endocrinology, Diabetes and Metabolism ,medicine.disease ,Gastroenterology ,medicine.anatomical_structure ,Fibrosis ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Acinar cell ,Acute pancreatitis ,Pancreatitis ,Histopathology ,Pancreas ,business ,Insulitis - Abstract
Recent efforts addressing the pathogenesis of T1D have benefited greatly from access to transplant-quality human pancreas samples obtained from organ donors. While alcohol use is linked to pancreatitis clinically, the effect of alcohol and other illicit substances on pancreatic histopathology in T1D is not known. Hence, we determined the influence of alcohol, marijuana, tobacco, cocaine, and other controlled substances in T1D pancreata. In total, 140 T1D organ donor pancreata [median age at T1D onset=11 (range 0-36) years, duration=12 (0-84) years, 47% female] were recovered through the Network for Pancreatic Organ donors with Diabetes (nPOD) program. Paraffin sections were stained and reviewed for insulin positivity, insulitis (≥6 CD3+ cells adjacent/within the islet in ≥3 islets/section), amyloid staining, acute (acinar cell loss, mononuclear cell infiltrate, fat necrosis, and hemorrhage) and chronic pancreatitis (acinar cell loss, mononuclear cell infiltrate, and fibrosis) as well as chronic exocrine changes (acinar atrophy, fibrosis, fatty infiltration, or periductal fibrosis). Terminal hospital records were reviewed for a history of alcohol, tobacco, marijuana, and any illicit substance use. Of 140 T1D donors, 51% had a history of alcohol use, 40% tobacco use, 29% marijuana, and 20% other illicit substances. Urine drug screening on admission was available for 74 donors with 36 positive (49%). Alcohol use was associated with histopathological features of acute pancreatitis (p Disclosure B.S. Bruggeman: None. M. Campbell-Thompson: None. A.L. Posgai: None. M.A. Atkinson: None. D. Schatz: None. L.M. Jacobsen: None. Funding JDRF
- Published
- 2019
- Full Text
- View/download PDF
46. The challenge of modulating beta-cell autoimmunity in type 1 diabetes
- Author
-
Daniel C S Wheeler, Bart O. Roep, Mark A. Atkinson, Amanda L. Posgai, and Mark Peakman
- Subjects
medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,media_common.quotation_subject ,030209 endocrinology & metabolism ,Disease ,medicine.disease_cause ,Article ,Autoimmunity ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Optimism ,Diabetes mellitus ,Internal Medicine ,Medicine ,030212 general & internal medicine ,Intensive care medicine ,media_common ,Autoimmune disease ,Type 1 diabetes ,business.industry ,medicine.disease ,3. Good health ,Clinical trial ,business ,Hindsight bias - Abstract
With the conceptual advance some four decades ago that type 1 diabetes (T1D) represents an autoimmune disease, hope emerged that immune-based therapies would quickly evolve as a means to prevent and reverse the disorder. However, despite dozens of clinical trials seeking to achieve that purpose, the goal remains unfulfilled, at least in a pragmatic form. With the benefit of hindsight, multiple reasons are likely to account for this unfortunate situation, and several stand out: failure to appreciate disease heterogeneity; inappropriate utilization of and insight from rodent models of disease; inadequacies in addressing the immunologic and metabolic contributions to the disease; suboptimal trial designs; and lack of a clear understanding of the disorder’s pathogenesis. This review conveys how recent knowledge gains in these areas, combined with efforts related to disease staging and emerging mechanistic data from clinical trials, provide cautious optimism that an immune-based means to prevent the loss of β-cells in T1D will emerge into clinical practice.
- Published
- 2019
47. Expansion of Human Tregs from Cryopreserved Umbilical Cord Blood for GMP-Compliant Autologous Adoptive Cell Transfer Therapy
- Author
-
Ashley N. Bushdorf, Todd M. Brusko, Katherine S. Brown, Jeffrey A. Bluestone, Leeana D. Peters, Judit Cserny, Angela P. Lares, Howard R. Seay, Morey Kraus, Mark A. Atkinson, John R. Wingard, Kate Falcon Girard, Heather L. Brown, Amy L. Putnam, Kristi T. Balavage, Michael J. Haller, Emma Rosenau, and Amanda L. Posgai
- Subjects
0301 basic medicine ,Adoptive cell transfer ,lcsh:QH426-470 ,Regulatory T cell ,type 1 diabetes ,medicine.medical_treatment ,Stem Cell Research - Umbilical Cord Blood/ Placenta - Human ,chemical and pharmacologic phenomena ,Human leukocyte antigen ,Hematopoietic stem cell transplantation ,Umbilical cord ,Autoimmune Disease ,Vaccine Related ,03 medical and health sciences ,0302 clinical medicine ,Stem Cell Research - Nonembryonic - Human ,regulatory T cell ,Genetics ,Medicine ,lcsh:QH573-671 ,Molecular Biology ,adoptive cell transfer ,Transplantation ,5.2 Cellular and gene therapies ,lcsh:Cytology ,business.industry ,Prevention ,Inflammatory and immune system ,T-cell receptor ,autoimmunity ,FOXP3 ,hemic and immune systems ,Stem Cell Research ,3. Good health ,lcsh:Genetics ,030104 developmental biology ,medicine.anatomical_structure ,good manufacturing practices ,Cord blood ,Immunology ,cord blood ,Molecular Medicine ,Immunization ,Original Article ,Development of treatments and therapeutic interventions ,T cell receptor ,business ,030215 immunology ,Stem Cell Research - Umbilical Cord Blood/ Placenta - Abstract
Umbilical cord blood is a traditional and convenient source of cells for hematopoietic stem cell transplantation. Thymic regulatory Tcells (Tregs) are also present in cord blood, and there is growing interest in the use of autologous Tregs to provide a low-risk, fully human leukocyte antigen (HLA)-matched cell product for treating autoimmune diseases, such as type 1 diabetes. Here, we describe a good manufacturing practice (GMP)-compatible Treg expansion protocol using fluorescence-activated cell sorting, resulting in a mean 2,092-fold expansion of Tregs over a 16-day culture for a median yield of 1.26× 109 Tregs from single-donor cryopreserved units. The resulting Tregs passed prior clinical trial release criteria for Treg purity and sterility, including additional rigorous assessments of FOXP3 and Helios expression and epigenetic analysis of the FOXP3 Treg-specific demethylated region (TSDR). Compared with expanded adult peripheral blood Tregs, expanded cord blood Tregs remained more naive, as assessed by continued expression of CD45RA, produced reduced IFN-γ following activation, and effectively inhibited responder Tcell proliferation. Immunosequencing of the Tcell receptor revealed a remarkably diverse receptor repertoire within cord blood Tregs that was maintained following invitro expansion. These data support the feasibility of generating GMP-compliant Tregs from cord blood for adoptive cell transfer therapies and highlight potential advantages in terms of safety, phenotypic stability, autoantigen specificity, and tissue distribution.
- Published
- 2016
48. Insulitis in Autoantibody-Positive Pancreatic Donor With History of Gestational Diabetes Mellitus
- Author
-
Amanda L. Posgai, Jessica Jackson, Irina Kusmartseva, and Martha Campbell-Thompson
- Subjects
0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,030209 endocrinology & metabolism ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,law ,Diabetes mellitus ,Internal Medicine ,medicine ,Advanced and Specialized Nursing ,Past medical history ,business.industry ,Emergency department ,medicine.disease ,Intensive care unit ,3. Good health ,Surgery ,Gestational diabetes ,030104 developmental biology ,medicine.anatomical_structure ,Hemodialysis ,Clinical Images in Diabetes ,Pancreas ,business ,Insulitis - Abstract
The patient (nPOD 6310) was a 28-year-old female, 32 days postoperative from a cesarean delivery. Her past medical history was significant for chronic hypertension, severe preeclampsia, acute renal failure, and gestational diabetes mellitus (GDM). She was brought to the emergency department after collapsing, with total downtime of 20–30 min. On arrival, she was admitted to the intensive care unit and found to be in hypertensive crisis. Her hospital course was complicated by renal failure requiring four rounds of hemodialysis, hypertension requiring multiple agents for control, and a degree of hyperglycemia requiring insulin. Computed tomography of the head demonstrated anoxic encephalopathy with multiple infarcts within the brain. The patient was declared brain-dead and, with authorization obtained from next of kin, became an organ donor. The pancreas, along with blood, was recovered for research through the Network for Pancreatic Organ …
- Published
- 2017
- Full Text
- View/download PDF
49. Expression of SARS-CoV-2 Entry Factors in the Pancreas of Normal Organ Donors and Individuals with COVID-19
- Author
-
Eduardo Candelario-Jalil, Irina Kusmartseva, Verena van der Heide, Harry S. Nick, Richard S. Vander Heide, Farooq Syed, Mark A. Atkinson, Carmella Evans-Molina, Tang X, Dirk Homann, John David Paulsen, Martha Campbell-Thompson, Changjun Yang, Richard E. Lloyd, Wenting Wu, Amanda L. Posgai, Jack L. Harbert, Alberto Pugliese, Sirlene Cechin, Paul N. Joseph, and Marda Jorgensen
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,insulin ,Physiology ,type 1 diabetes ,viruses ,ACE2 ,Gene Expression ,Enteroendocrine cell ,Type 2 diabetes ,03 medical and health sciences ,0302 clinical medicine ,Short Article ,Diabetes mellitus ,Gene expression ,medicine ,Humans ,pancreas ,Molecular Biology ,TMPRSS2 ,Type 1 diabetes ,medicine.diagnostic_test ,islet ,business.industry ,SARS-CoV-2 ,Serine Endopeptidases ,COVID-19 ,Cell Biology ,Virus Internalization ,medicine.disease ,Tissue Donors ,Blot ,030104 developmental biology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 2 ,type 2 diabetes ,CD34 ,Angiotensin-Converting Enzyme 2 ,Pancreas ,business ,030217 neurology & neurosurgery ,Fluorescence in situ hybridization - Abstract
Diabetes is associated with increased mortality from severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Given literature suggesting a potential association between SARS-CoV-2 infection and diabetes induction, we examined pancreatic expression of angiotensin-converting enzyme 2 (ACE2), the key entry factor for SARS-CoV-2 infection. Specifically, we analyzed five public scRNA-seq pancreas datasets and performed fluorescence in situ hybridization, western blotting, and immunolocalization for ACE2 with extensive reagent validation on normal human pancreatic tissues across the lifespan, as well as those from coronavirus disease 2019 (COVID-19) cases. These in silico and ex vivo analyses demonstrated prominent expression of ACE2 in pancreatic ductal epithelium and microvasculature, but we found rare endocrine cell expression at the mRNA level. Pancreata from individuals with COVID-19 demonstrated multiple thrombotic lesions with SARS-CoV-2 nucleocapsid protein expression that was primarily limited to ducts. These results suggest SARS-CoV-2 infection of pancreatic endocrine cells, via ACE2, is an unlikely central pathogenic feature of COVID-19-related diabetes., Graphical Abstract, Highlights • ACE2 mRNA and protein are expressed in human pancreatic ducts and microvasculature • ACE2 mRNA was rarely detected and at low levels in human pancreatic endocrine cells • Pancreatic ACE2 protein expression changes across the lifespan and correlates with BMI • SARS-CoV-2 NP was detected in ducts, but not endocrine cells, of COVID-19 pancreata, Kusmartseva et al. demonstrate preferential ACE2 expression in pancreatic microvascular and ductal structures, suggesting these constitute a more likely target than islet endocrine cells in SARS-CoV-2 infection. This notion was supported by detection of SARS-CoV-2 nucleocapsid protein in ductal epithelium, but not endocrine cells, of pancreata from individuals with COVID-19.
- Published
- 2020
50. Immune Mechanisms and Pathways Targeted in Type 1 Diabetes
- Author
-
Laura M. Jacobsen, Michael J. Haller, Amanda L. Posgai, Todd M. Brusko, Brittney N. Newby, and Daniel J. Perry
- Subjects
0301 basic medicine ,endocrine system diseases ,T-Lymphocytes ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Autoimmunity ,030209 endocrinology & metabolism ,Context (language use) ,medicine.disease_cause ,Bioinformatics ,Article ,03 medical and health sciences ,0302 clinical medicine ,Diabetes mellitus ,Internal Medicine ,Humans ,Medicine ,Immune mechanisms ,Glycemic ,Type 1 diabetes ,business.industry ,nutritional and metabolic diseases ,Immunotherapy ,medicine.disease ,Primary Prevention ,Clinical trial ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,business ,Biomarkers - Abstract
PURPOSE OF REVIEW: The immunosuppressive agent cyclosporine was first reported to lower daily insulin dose and improve glycemic control in patients with new-onset type 1 diabetes (T1D) in 1984. While renal toxicity limited cyclosporine’s extended use, this observation ignited collaborative efforts to identify immunotherapeutic agents capable of safely preserving β cells in patients with or at risk for T1D. RECENT FINDINGS: Advances in T1D prediction and early diagnosis, together with expanded knowledge of the disease mechanisms, have facilitated trials targeting specific immune cell subsets, autoantigens, and pathways. In addition, clinical responder and non-responder subsets have been defined through the use of metabolic and immunological readouts. SUMMARY: Herein, we review emerging T1D biomarkers within the context of recent and ongoing T1D immunotherapy trials. We also discuss responder/non-responder analyses in an effort to identify therapeutic mechanisms, define actionable pathways, and guide subject selection, drug dosing, and tailored combination drug therapy for future T1D trials.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.