Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Neuroscience ; 547: 17-27, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38583506

ABSTRACT

Ghrelin, a hormone secreted by the stomach, binds to the growth hormone secretagogue receptor (GHSR) in various brain regions to produce a number of behavioral effects that include increased feeding motivation. During social defeat stress, ghrelin levels rise in correlation with increased feeding and potentially play a role in attenuating the anxiogenic effects of social defeat. One region implicated in the feeding effects of ghrelin is the ventral tegmental area (VTA), a region implicated in reward seeking behaviors, and linked to social defeat in mice. Here we examined the role of GHSR signaling in the VTA in feeding behavior in mice exposed to social defeat stress. Male C57BL/J6 mice that were socially defeated once daily for 3 weeks ate more, had higher plasma ghrelin level and increased GHSR expression in the VTA compared to non-stressed mice. Socially defeated GHSR KO mice failed to increase their caloric intake in response to this stressor but rescue of GHSR expression in the VTA restored feeding responses. Finally, we pharmacologically blocked VTA GHSR signalling with JMV2959 infused via an indwelling VTA cannula connected to a minipump. Vehicle-treated mice increased their caloric intake during social defeat, but JMV2959-infusions attenuated feeding responses and increased anxiety-like behaviors. The data suggest that GHSR signalling in the VTA is critical for the increases in appetite observed during chronic social defeat stress. Furthermore, these data support the idea that GHSR signaling in the VTA may also have anxiolytic effects, and blocking GHSR in this region may result in an anxiety-like phenotype.


Subject(s)
Feeding Behavior , Ghrelin , Mice, Inbred C57BL , Mice, Knockout , Receptors, Ghrelin , Social Defeat , Stress, Psychological , Ventral Tegmental Area , Animals , Ventral Tegmental Area/metabolism , Receptors, Ghrelin/metabolism , Receptors, Ghrelin/genetics , Male , Stress, Psychological/metabolism , Mice , Feeding Behavior/physiology , Ghrelin/metabolism , Signal Transduction/physiology , Anxiety/metabolism
2.
Lupus Sci Med ; 9(1)2022 07.
Article in English | MEDLINE | ID: mdl-35820718

ABSTRACT

BACKGROUND: Reports of clinical improvement following mesenchymal stromal cell (MSC) infusions in refractory lupus patients at a single centre in China led us to perform an explorative phase I trial of umbilical cord derived MSCs in patients refractory to 6 months of immunosuppressive therapy. METHODS: Six women with a SLEDAI >6, having failed standard of care therapy, received one intravenous infusion of 1×106 MSCs/kg of body weight. They maintained their current immunosuppressives, but their physician was allowed to adjust corticosteroids initially for symptom management. The clinical endpoint was an SRI of 4 with no new British Isles Lupus Activity Guide (BILAG) As and no increase in Physician Global Assessment score of >0.3 with tapering of prednisone to 10 mg or less by 20 weeks. RESULTS: Of six patients, five (83.3%; 95% CI 35.9% to 99.6%) achieved the clinical endpoint of an SRI of 4. Adverse events were minimal. Mechanistic studies revealed significant reductions in CD27IgD double negative B cells, switched memory B cells and activated naïve B cells, with increased transitional B cells in the five patients who met the endpoint. There was a trend towards decreased autoantibody levels in specific patients. Two patients had increases in their Helios+Treg cells, but no other significant T cell changes were noted. GARP-TGFß complexes were significantly increased following the MSC infusions. The B cell changes and the GARP-TGFß increases significantly correlated with changes in SLEDAI scores. CONCLUSION: This phase 1 trial suggests that umbilical cord (UC) MSC infusions are very safe and may have efficacy in lupus. The B cell and GARP-TGFß changes provide novel insight into mechanisms by which MSCs may impact disease. TRIAL REGISTRATION NUMBER: NCT03171194.


Subject(s)
Lupus Erythematosus, Systemic , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Female , Humans , Lupus Erythematosus, Systemic/drug therapy , Mesenchymal Stem Cell Transplantation/adverse effects , Transforming Growth Factor beta , Umbilical Cord
3.
Am J Transplant ; 22(2): 386-401, 2022 02.
Article in English | MEDLINE | ID: mdl-34714588

ABSTRACT

Early insults associated with cardiac transplantation increase the immunogenicity of donor microvascular endothelial cells (ECs), which interact with recipient alloreactive memory T cells and promote responses leading to allograft rejection. Thus, modulating EC immunogenicity could potentially alter T cell responses. Recent studies have shown modulating mitochondrial fusion/fission alters immune cell phenotype. Here, we assess whether modulating mitochondrial fusion/fission reduces EC immunogenicity and alters EC-T cell interactions. By knocking down DRP1, a mitochondrial fission protein, or by using the small molecules M1, a fusion promoter, and Mdivi1, a fission inhibitor, we demonstrate that promoting mitochondrial fusion reduced EC immunogenicity to allogeneic CD8+ T cells, shown by decreased T cell cytotoxic proteins, decreased EC VCAM-1, MHC-I expression, and increased PD-L1 expression. Co-cultured T cells also displayed decreased memory frequencies and Ki-67 proliferative index. For in vivo significance, we used a novel murine brain-dead donor transplant model. Balb/c hearts pretreated with M1/Mdivi1 after brain-death induction were heterotopically transplanted into C57BL/6 recipients. We demonstrate that, in line with our in vitro studies, M1/Mdivi1 pretreatment protected cardiac allografts from injury, decreased infiltrating T cell production of cytotoxic proteins, and prolonged allograft survival. Collectively, our data show promoting mitochondrial fusion in donor ECs mitigates recipient T cell responses and leads to significantly improved cardiac transplant survival.


Subject(s)
Heart Transplantation , Mitochondrial Dynamics , Animals , CD8-Positive T-Lymphocytes , Endothelial Cells , Graft Rejection/etiology , Graft Rejection/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
Front Psychiatry ; 12: 618279, 2021.
Article in English | MEDLINE | ID: mdl-33658952

ABSTRACT

Background: A growing body of research has shown that consumption of probiotics can improve symptoms associated with mood and anxiety disorders through activity of the gut-brain axis. However, the effects of probiotics have yet to be tested in a clinical sample of treatment-naïve patients diagnosed with Major Depressive Disorder (MDD). The aim of this 8-week, open-label pilot study is to examine changes in depressive symptoms before and after the introduction of a probiotic supplement in 10 treatment-naïve MDD patients and to provide data on the feasibility of conducting a larger double-blind, randomized, placebo-controlled trial in the same patient population. Here we report on the clinical outcome measures of the study. Methods: Participants recruited from the community in Kingston, Ontario, Canada consumed a probiotic supplement containing Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 (CEREBIOME®) at a dose of 3 × 109 CFU once per day for 8 weeks. Clinical symptoms of depression were measured using a validated battery of clinical scales and self-report questionnaires (CAN-BIND protocol). Data was collected at baseline, week 4, and week 8. Results: Significant improvements in affective clinical symptoms were observed at week 4 and were sustained at week 8. Significant improvements in subjective sleep quality were observed by week 8. No side effects or adverse effects associated with the probiotic supplement were observed. Conclusions: The findings from this study support the existing evidence in this emerging field for probiotics having a role in alleviating symptoms of depression in treatment-naïve, moderately depressed patients and indicate that the probiotic supplement is safe and well-tolerated in this population. However, further comprehensive studies are required to draw conclusions.

5.
Am J Transplant ; 21(6): 2067-2078, 2021 06.
Article in English | MEDLINE | ID: mdl-33210808

ABSTRACT

Complement is known to play a role in ischemia and reperfusion injury (IRI). A general paradigm is that complement is activated by self-reactive natural IgM antibodies (nAbs), after they engage postischemic neoepitopes. However, a role for nAbs in lung transplantation (LTx) has not been explored. Using mouse models of LTx, we investigated the role of two postischemic neoepitopes, modified annexin IV (B4) and a subset of phospholipids (C2), in LTx. Antibody deficient Rag1-/- recipient mice were protected from LTx IRI. Reconstitution with either B4 or C2nAb restored IRI, with C2 significantly more effective than B4 nAb. Based on these information, we developed/characterized a novel complement inhibitor composed of single-chain antibody (scFv) derived from the C2 nAb linked to Crry (C2scFv-Crry), a murine inhibitor of C3 activation. Using an allogeneic LTx, in which recipients contain a full nAb repertoire, C2scFv-Crry targeted to the LTx, inhibited IRI, and delayed acute rejection. Finally, we demonstrate the expression of the C2 neoepitope in human donor lungs, highlighting the translational potential of this approach.


Subject(s)
Lung Injury , Lung Transplantation , Reperfusion Injury , Transplants , Animals , Complement Inactivating Agents , Humans , Immunoglobulin M , Lung Transplantation/adverse effects , Mice , Reperfusion Injury/prevention & control
6.
BMC Psychiatry ; 20(1): 299, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32539741

ABSTRACT

BACKGROUND: The Gut-Brain-Axis is a bidirectional signaling pathway between the gastrointestinal (GI) tract and the brain. The hundreds of trillions of microorganisms populating the gastrointestinal tract are thought to modulate this connection, and have far reaching effects on the immune system, central and autonomic nervous systems, and GI functioning. These interactions Diagnostic and statistical manual of mental disorders have also been linked to various psychiatric illnesses such as depression, anxiety, substance abuse, autism spectrum disorder, and eating disorders. It is hypothesized that techniques aimed at strengthening and repopulating the gut microbiome, such as Fecal Microbiota Transplant (FMT), may be useful in the prevention and treatment of psychiatric illnesses. METHODS: A systematic search of five databases was conducted using key terms related to FMT and psychiatric illnesses. All results were then evaluated based on specific eligibility criteria. RESULTS: Twenty-one studies met the eligibility criteria and were analysed for reported changes in mood and behavioural measures indicative of psychiatric wellbeing. The studies included were either entirely clinical (n = 8), preclinical with human donors (n = 9), or entirely preclinical (n = 11). All studies found a decrease in depressive and anxiety-like symptoms and behaviours resulting from the transplantation of healthy microbiota. The inverse was also found, with the transmission of depressive and anxiety-like symptoms and behaviours resulting from the transplantation of microbiota from psychiatrically ill donors to healthy recipients. CONCLUSION: There appears to be strong evidence for the treatment and transmission of psychiatric illnesses through FMT. Further research with larger sample sizes and stronger scientific design is warranted in order to fully determine the efficacy and safety of this potential treatment. Registered on PROSPERO, IRD: CRD42019126795.


Subject(s)
Autism Spectrum Disorder , Gastrointestinal Microbiome , Microbiota , Anxiety , Fecal Microbiota Transplantation , Humans
7.
Neuropsychobiology ; 79(1): 108-116, 2020.
Article in English | MEDLINE | ID: mdl-30759442

ABSTRACT

BACKGROUND: A growing body of evidence has linked mental health outcomes to the gut microbiome. This has led to the investigation of the GI tract as a target for novel treatments and interventions for depression, including probiotic supplementation. Our recent pilot study provided the first evidence of probiotics improving symptoms of depression in treatment-naive depressed patients. To further support and expand upon this evidence, data from the pilot study were used to plan a 16-week, double-blind, randomized, placebo-controlled trial to assess the effects of probiotics on depression. Here, we report the protocol for this trial. METHODS: Participants diagnosed with depression will orally consume a probiotic supplement containing Lactobacillus helveticus and Bifidobacterium longum or placebo once daily. Participants will undergo assessments measuring clinical outcomes using a battery of validated clinical scales and questionnaires. Sleep architecture and quality will be measured using polysomnography. Neuroimaging data will be collected using magnetic resonance imaging to examine functional and structural neurophysiological changes. Molecular data will be collected from blood, stool, and urine samples to examine cytokine levels and explore potential genes and proteins that may predict outcomes in depression. RESULTS: We expect results to replicate and expand on our pilot data demonstrating that probiotics may be effective in alleviating symptoms of depression, and to find biomarkers that will predict these outcomes. CONCLUSIONS: The findings from this study will add to the growing body of research in this emerging field, which eventually may provide evidence for probiotics having a role in alleviating symptoms of depression.


Subject(s)
Depressive Disorder, Major/diet therapy , Probiotics/pharmacology , Adolescent , Adult , Aged , Biomarkers , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Double-Blind Method , Female , Humans , Male , Middle Aged , Probiotics/administration & dosage , Research Design , Young Adult
8.
JCI Insight ; 4(7)2019 04 04.
Article in English | MEDLINE | ID: mdl-30944260

ABSTRACT

Multiple myeloma (MM) is an incurable plasma cell malignancy with frequent treatment failures and relapses, suggesting the existence of pathogenic myeloma stem/progenitor populations. However, the identity of MM stem cells remains elusive. We used a murine model of MM with transgenic overexpression of the unfolded protein response sensor X-box binding protein 1 (XBP1s) in the B cell compartment to define MM stem cells. We herein report that a post-germinal center, pre-plasma cell population significantly expands as MM develops. This population has the following characteristics: (a) cell surface phenotype of B220+CD19+IgM-IgD-CD138-CD80+sIgG-AA4.1+FSChi; (b) high expression levels of Pax5 and Bcl6 with intermediate levels of Blimp1 and XBP1s; (c) increased expression of aldehyde dehydrogenase, Notch1, and c-Kit; and (d) ability to efficiently reconstitute antibody-producing capacity in B cell-deficient mice in vivo. We thus have defined a plasma cell progenitor population that resembles myeloma stem cells in mice. These results provide potentially novel insights into MM stem cell biology and may contribute to the development of novel stem cell-targeted therapies for the eradication of MM.


Subject(s)
B-Lymphocytes/pathology , Multiple Myeloma/pathology , Neoplastic Stem Cells/pathology , X-Box Binding Protein 1/metabolism , Animals , Cell Separation , Disease Models, Animal , Flow Cytometry , Humans , Lymphopoiesis , Mice , Mice, Transgenic , Multiple Myeloma/genetics , Primary Cell Culture , Tumor Cells, Cultured , X-Box Binding Protein 1/genetics
9.
Cancer Res ; 79(6): 1178-1190, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30674536

ABSTRACT

Activated regulatory T (Treg) cells express the surface receptor glycoprotein-A repetitions predominant (GARP), which binds and activates latent TGFß. How GARP modulates Treg function in inflammation and cancer remains unclear. Here we demonstrate that loss of GARP in Treg cells leads to spontaneous inflammation with highly activated CD4+ and CD8+ T cells and development of enteritis. Treg cells lacking GARP were unable to suppress pathogenic T-cell responses in multiple models of inflammation, including T-cell transfer colitis. GARP-/- Treg cells were significantly reduced in the gut and exhibited a reduction in CD103 expression, a colon-specific migratory marker. In the colitis-associated colon cancer model, GARP on Treg cells dampened immune surveillance, and mice with GARP-/- Treg cells exhibited improved antitumor immunity. Thus, GARP empowers the functionality of Treg cells and their tissue-specific accumulation, highlighting the importance of cell surface TGFß in Treg function and GARP as a potential therapeutic target for colorectal cancer therapy.Significance: These findings uncover functions of membrane-bound TGFß and GARP that tune the activity of Treg cells, highlighting a potential treatment strategy in autoimmune diseases and cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colitis/immunology , Colonic Neoplasms/immunology , Immune Tolerance/immunology , Inflammation/immunology , Membrane Proteins/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
10.
JCI Insight ; 3(7)2018 04 05.
Article in English | MEDLINE | ID: mdl-29618665

ABSTRACT

GARP, a cell surface docking receptor for binding and activating latent TGF-ß, is highly expressed by platelets and activated Tregs. While GARP is implicated in immune invasion in cancer, the roles of the GARP-TGF-ß axis in systemic autoimmune diseases are unknown. Although B cells do not express GARP at baseline, we found that the GARP-TGF-ß complex is induced on activated human and mouse B cells by ligands for multiple TLRs, including TLR4, TLR7, and TLR9. GARP overexpression on B cells inhibited their proliferation, induced IgA class-switching, and dampened T cell-independent antibody production. In contrast, B cell-specific deletion of GARP-encoding gene Lrrc32 in mice led to development of systemic autoimmune diseases spontaneously as well as worsening of pristane-induced lupus-like disease. Canonical TGF-ß signaling more readily upregulates GARP in Peyer patch B cells than in splenic B cells. Furthermore, we demonstrated that B cells are required for the induction of oral tolerance of T cell-dependent antigens via GARP. Our studies reveal for the first time to our knowledge that cell surface GARP-TGF-ß is an important checkpoint for regulating B cell peripheral tolerance, highlighting a mechanism of autoimmune disease pathogenesis.


Subject(s)
B-Lymphocytes/immunology , Immune Tolerance/immunology , Membrane Proteins/metabolism , Transforming Growth Factor beta/metabolism , Adjuvants, Immunologic/administration & dosage , Animals , Autoimmune Diseases/immunology , B-Lymphocytes/metabolism , Bone Marrow Transplantation , Cells, Cultured , Female , Gene Knock-In Techniques , Healthy Volunteers , Humans , Immune Tolerance/drug effects , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Transgenic , Primary Cell Culture , Signal Transduction/immunology , Transforming Growth Factor beta/immunology , Transplantation Chimera
11.
Health Promot Chronic Dis Prev Can ; 38(2): 37-54, 2018 Feb.
Article in English, French | MEDLINE | ID: mdl-29443484

ABSTRACT

INTRODUCTION: This systematic review identified population-representative youth surveys containing questions on self-reported child maltreatment. Data quality and ethical issues pertinent to maltreatment data collection were also examined. METHODS: A search was conducted of relevant online databases for articles published from January 2000 through March 2016 reporting on population-representative data measuring child maltreatment. Inclusion criteria were established a priori; two reviewers independently assessed articles to ensure that the criteria were met and to verify the accuracy of extracted information. RESULTS: A total of 73 articles reporting on 71 surveys met the inclusion criteria. A variety of strategies to ensure accurate information and to mitigate survey participants' distress were reported. CONCLUSION: The extent to which efforts have been undertaken to measure the prevalence of child maltreatment reflects its perceived importance across the world. Data on child maltreatment can be effectively collected from youth, although our knowledge of best practices related to ethics and data quality is incomplete.


INTRODUCTION: Cette revue systématique a permis de recenser des enquêtes sur les jeunes représentatives de la population et comportant des questions sur la maltraitance envers les enfants déclarée par ces derniers. Nous avons également examiné la qualité des données et les questions d'ordre éthique pertinentes pour la collecte de données sur la maltraitance. MÉTHODOLOGIE: Nous avons effectué une recherche dans diverses bases de données en ligne pour sélectionner les articles publiés entre janvier 2000 et mars 2016 qui contenaient des données représentatives de la population mesurant la maltraitance envers les enfants. Les critères d'inclusion ont été établis a priori et deux examinateurs ont évalué indépendamment l'un de l'autre les articles pour s'assurer que les critères étaient respectés et pour vérifier l'exactitude des données extraites. RÉSULTATS: Au total, 73 articles portant sur 71 enquêtes ont répondu aux critères d'inclusion. Nous avons relevé diverses stratégies visant à assurer l'exactitude des renseignements et à atténuer la détresse des participants à l'enquête. CONCLUSION: L'ampleur des efforts déployés pour mesurer la prévalence de la maltraitance envers les enfants est le reflet de l'importance accordée à cette dernière à l'échelle de la planète. Malgré des connaissances incomplètes en matière de pratiques exemplaires liées à l'éthique et de qualité des données, il est possible de recueillir efficacement auprès des jeunes eux-mêmes des données sur la maltraitance dont ils font l'objet.


Subject(s)
Child Abuse/ethics , Child Abuse/statistics & numerical data , Self Report , Adolescent , Canada , Child , Databases, Factual , Female , Humans , Male , Surveys and Questionnaires
12.
J Hematol Oncol ; 11(1): 24, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29458436

ABSTRACT

GARP (glycoprotein-A repetitions predominant) is a type I transmembrane cell surface docking receptor for latent transforming growth factor-ß (TGF-ß) that is abundantly expressed on regulatory T lymphocytes and platelets. GARP regulates the availability of membrane-bound latent TGF-ß and modulates its activation. For this reason, GARP expression on immune and non-immune cells is involved in maintaining peripheral tolerance. It plays an important role in preventing inflammatory diseases such as allergy and graft versus host disease (GvHD). GARP is also frequently hijacked by cancer cells to promote oncogenesis. This review summarizes the most important features of GARP biology described to date including gene regulation, protein expression and mechanism in activating latent TGF-ß, and the function of GARP in regulatory T cell biology and peripheral tolerance, as well as GARP's increasingly recognized roles in platelet-mediated cancer immune evasion. The promise for GARP-targeted strategy as a novel immunotherapy of cancer is also highlighted.


Subject(s)
Inflammation/immunology , Membrane Proteins/immunology , Neoplasms/immunology , Transforming Growth Factor beta/immunology , Animals , Blood Platelets/immunology , Blood Platelets/pathology , Gene Expression Regulation, Neoplastic , Humans , Immune Tolerance , Inflammation/complications , Inflammation/genetics , Inflammation/pathology , Membrane Proteins/analysis , Membrane Proteins/genetics , Neoplasms/complications , Neoplasms/genetics , Neoplasms/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/analysis
13.
Front Psychiatry ; 8: 190, 2017.
Article in English | MEDLINE | ID: mdl-29033860

ABSTRACT

BACKGROUND: Research has demonstrated a reduction in olfactory functioning in patients with schizophrenia. This research has led to examination of olfactory functioning in other mental disorders, such as depression. There is a great deal of variation in the results generated from such research, and it remains unclear as to how olfactory functioning is associated with or impacted by depression. METHOD: The current review examined the literature in accordance with PRISMA guidelines in order to generate a better understanding of this relationship and to identify if and what aspects of olfactory processing are altered. Through examination of the available literature from the databases PubMed, Ovid Medline, CINAL, and PsychINFO, 15 manuscripts were selected to determine if there was a difference in olfactory processing-specifically central and peripheral processing-between depressed individuals and non-depressed controls. RESULTS: The comparison of the 15 studies showed that the majority of studies (9/15, 60%) found a difference in overall olfactory functioning between depressed individuals and non-depressed controls (p < 0.05). LIMITATIONS: There is still a lack of definitive conclusions due to variation of which olfactory process was altered. CONCLUSION: Given the differences in the methodology and design of these studies, a possible solution that could eliminate the lack of clarity and reduce variation would be to adhere to a single, thorough methodology that examines and separates central and peripheral olfactory processing. Future research employing a uniform and validated methodology could provide more definitive conclusions as to how and if olfactory functioning is related depression.

14.
J Biol Chem ; 292(44): 18091-18097, 2017 11 03.
Article in English | MEDLINE | ID: mdl-28912269

ABSTRACT

Glycoprotein A repetitions predominant (GARP) (encoded by the Lrrc32 gene) plays important roles in cell-surface docking and activation of TGFß. However, GARP's role in organ development in mammalian systems is unclear. To determine the function of GARP in vivo, we generated a GARP KO mouse model. Unexpectedly, the GARP KO mice died within 24 h after birth and exhibited defective palatogenesis without apparent abnormalities in other major organs. Furthermore, we observed decreased apoptosis and SMAD2 phosphorylation in the medial edge epithelial cells of the palatal shelf of GARP KO embryos at embryonic day 14.5 (E14.5), indicating a defect in the TGFß signaling pathway in the GARP-null developing palates. Of note, the failure to develop the secondary palate and concurrent reduction of SMAD phosphorylation without other defects in GARP KO mice phenocopied TGFß3 KO mice, although GARP has not been suggested previously to interact with TGFß3. We found that GARP and TGFß3 co-localize in medial edge epithelial cells at E14.5. In vitro studies confirmed that GARP and TGFß3 directly interact and that GARP is indispensable for the surface expression of membrane-associated latent TGFß3. Our findings indicate that GARP is essential for normal morphogenesis of the palate and demonstrate that GARP plays a crucial role in regulating TGFß3 signaling during embryogenesis. In conclusion, we have uncovered a novel function of GARP in positively regulating TGFß3 activation and function.


Subject(s)
Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Organogenesis , Palate/metabolism , Protein Processing, Post-Translational , Smad2 Protein/metabolism , Transforming Growth Factor beta3/agonists , Animals , Animals, Newborn , Apoptosis , Cleft Palate/embryology , Cleft Palate/metabolism , Cleft Palate/pathology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Gene Knock-In Techniques , HEK293 Cells , Heterozygote , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice, Knockout , Palate/abnormalities , Palate/embryology , Palate/pathology , Phosphorylation , Pregnancy , Protein Multimerization , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism
15.
Sci Immunol ; 2(11)2017 May 05.
Article in English | MEDLINE | ID: mdl-28763790

ABSTRACT

Cancer-associated thrombocytosis has long been linked to poor clinical outcome, but the underlying mechanism is enigmatic. We hypothesized that platelets promote malignancy and resistance to therapy by dampening host immunity. We show that genetic targeting of platelets enhances adoptive T cell therapy of cancer. An unbiased biochemical and structural biology approach established transforming growth factor ß (TGFß) and lactate as major platelet-derived soluble factors to obliterate CD4+ and CD8+ T cell functions. Moreover, we found that platelets are the dominant source of functional TGFß systemically as well as in the tumor microenvironment through constitutive expression of the TGFß-docking receptor glycoprotein A repetitions predominant (GARP) rather than secretion of TGFß per se. Platelet-specific deletion of the GARP-encoding gene Lrrc32 blunted TGFß activity at the tumor site and potentiated protective immunity against both melanoma and colon cancer. Last, this study shows that T cell therapy of cancer can be substantially improved by concurrent treatment with readily available antiplatelet agents. We conclude that platelets constrain T cell immunity through a GARP-TGFß axis and suggest a combination of immunotherapy and platelet inhibitors as a therapeutic strategy against cancer.

16.
Cancer Immunol Res ; 5(8): 666-675, 2017 08.
Article in English | MEDLINE | ID: mdl-28642246

ABSTRACT

The endoplasmic reticulum (ER) is an energy-sensing organelle with intimate ties to programming cell activation and metabolic fate. T-cell receptor (TCR) activation represents a form of acute cell stress and induces mobilization of ER Ca2+ stores. The role of the ER in programming T-cell activation and metabolic fate remains largely undefined. Gp96 is an ER protein with functions as a molecular chaperone and Ca2+ buffering protein. We hypothesized that the ER stress response may be important for CD4+ T-cell activation and that gp96 may be integral to this process. To test our hypothesis, we utilized genetic deletion of the gp96 gene Hsp90b1 in a CD4+ T cell-specific manner. We show that gp96-deficient CD4+ T cells cannot undergo activation-induced glycolysis due to defective Ca2+ mobilization upon TCR engagement. We found that activating naïve CD4+ T cells while inhibiting ER Ca2+ exchange, through pharmacological blockade of the ER Ca2+ channel inositol trisphosphate receptor (IP3R), led to a reduction in cytosolic Ca2+ content and generated a pool of CD62Lhigh/CD44low CD4+ T cells compared with wild-type (WT) matched controls. In vivo IP3R-inhibited CD4+ T cells exhibited elevated tumor control above WT T cells. Together, these data show that ER-modulated cytosolic Ca2+ plays a role in defining CD4+ T-cell phenotype and function. Factors associated with the ER stress response are suitable targets for T cell-based immunotherapies. Cancer Immunol Res; 5(8); 666-75. ©2017 AACR.


Subject(s)
Endoplasmic Reticulum Stress/immunology , Membrane Glycoproteins/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , CD4-Positive T-Lymphocytes/immunology , Calcium/metabolism , Endoplasmic Reticulum Stress/genetics , Glycolysis , Humans , Hyaluronan Receptors/immunology , Inositol 1,4,5-Trisphosphate Receptors/immunology , L-Selectin/immunology , Lymphocyte Activation/immunology , Membrane Glycoproteins/genetics , Neoplasms/pathology , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics
17.
Ann Gen Psychiatry ; 16: 18, 2017.
Article in English | MEDLINE | ID: mdl-28286538

ABSTRACT

[This corrects the article DOI: 10.1186/s12991-017-0138-2.].

18.
Ann Gen Psychiatry ; 16: 14, 2017.
Article in English | MEDLINE | ID: mdl-28239408

ABSTRACT

BACKGROUND: Patients suffering from depression experience significant mood, anxiety, and cognitive symptoms. Currently, most antidepressants work by altering neurotransmitter activity in the brain to improve these symptoms. However, in the last decade, research has revealed an extensive bidirectional communication network between the gastrointestinal tract and the central nervous system, referred to as the "gut-brain axis." Advances in this field have linked psychiatric disorders to changes in the microbiome, making it a potential target for novel antidepressant treatments. The aim of this review is to analyze the current body of research assessing the effects of probiotics, on symptoms of depression in humans. METHODS: A systematic search of five databases was performed and study selection was completed using the preferred reporting items for systematic reviews and meta-analyses process. RESULTS: Ten studies met criteria and were analyzed for effects on mood, anxiety, and cognition. Five studies assessed mood symptoms, seven studies assessed anxiety symptoms, and three studies assessed cognition. The majority of the studies found positive results on all measures of depressive symptoms; however, the strain of probiotic, the dosing, and duration of treatment varied widely and no studies assessed sleep. CONCLUSION: The evidence for probiotics alleviating depressive symptoms is compelling but additional double-blind randomized control trials in clinical populations are warranted to further assess efficacy.

19.
J Hematol Oncol ; 8: 77, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26108343

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is a hematologic malignancy that is characterized by the proliferation of abnormal bone marrow plasma cells (BMPC) and overproduction of immunoglobulin or light chains with evidence of end-organ damage such as bone damage, anemia, hypercalcemia, and renal dysfunction. The pathogenesis of MM is closely linked to dysregulated unfolded protein response (UPR) in the endoplasmic reticulum (ER). Constitutive activation of UPR in mice, as demonstrated by transgenic expression of a master UPR transcription factor XBP1s (a UPR-specific splice variant of X-box binding protein 1), causes myeloma. grp94 (gp96) is a key downstream chaperone in the ER that mediates the UPR as a part of the protein quality control mechanism in the secretory pathway. Our recent study has shown that the persistence of plasma cells as well as the development of myeloma in XBP1s-transgenic mice is critically dependent on grp94. However, the role of grp94 in the initiation and progression of human MM is still unknown. METHODS: The expression level of grp94 in BMPCs was measured by flow cytometry, real-time RT-PCR, and Western blot analysis. We compared the expression levels of grp94 in BMPCs in a spectrum of patients including MM, monoclonal gammopathy of undetermined significance (MGUS), smoldering MM (SMM), as well as non-plasma cell disorders (NPC). RESULTS: We found that grp94 was highly expressed in malignant plasma cells in patients with MM, but not in BMPCs in patients with MGUS/SMM and NPC. The expression level of grp94 correlated significantly with CD138 expression level. We also found that the grp94 expression level in BMPCs from International Staging System (ISS) stage III MM patients is higher than those in ISS stage I/II MM patients. CONCLUSIONS: grp94 is highly expressed in BMPCs in MM, which correlates with the advanced stage of this disease. Our data demonstrated that grp94 is a novel diagnostic and prognostic biomarker. It also positioned grp94 as a promising therapeutic target for MM.


Subject(s)
Biomarkers, Tumor/metabolism , Endoplasmic Reticulum/immunology , Multiple Myeloma/immunology , Plasma Cells/metabolism , Animals , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Humans , Mice , Multiple Myeloma/pathology
20.
Cancer Res ; 74(2): 446-59, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24322981

ABSTRACT

Macrophages are important drivers in the development of inflammation-associated colon cancers, but the mechanistic underpinnings for their contributions are not fully understood. Furthermore, Toll-like receptors have been implicated in colon cancer, but their relevant cellular sites of action are obscure. In this study, we show that the endoplasmic reticulum chaperone gp96 is essential in tumor-associated macrophages (TAM) to license their contributions to inflammatory colon tumorigenesis. Mice where gp96 was genetically deleted in a macrophage-specific manner exhibited reduced colitis and inflammation-associated colon tumorigenesis. Attenuation of colon cancer in these mice correlated strikingly with reduced mutation rates of ß-catenin, increased efficiency of the DNA repair machinery, and reduced expression of proinflammatory cytokines, including interleukin (IL)-17 and IL-23 in the tumor microenvironment. The genotoxic nature of TAM-associated inflammation was evident by increased expression of genes in the DNA repair pathway. Our work deepens understanding of how TAM promote oncogenesis by altering the molecular oncogenic program within epithelial cells, and it identifies gp96 as a lynchpin chaperone needed in TAM to license their function and impact on expression of critical inflammatory cytokines in colon tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Macrophages/metabolism , Membrane Glycoproteins/physiology , Animals , Bone Marrow Cells/cytology , Colitis/metabolism , Colon/pathology , Crosses, Genetic , Cytokines/metabolism , DNA Repair , Disease Progression , Gene Deletion , Inflammation , Interleukin-17/metabolism , Interleukin-23/metabolism , Male , Mice , Mice, Knockout , Mucous Membrane/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...